Chitin and Chitosan - Proceedings of The Second International Conference
Chitin and Chitosan - Proceedings of The Second International Conference
Chitin and Chitosan - Proceedings of The Second International Conference
Chitosan
Wiley Series
in
Renewable Resources
Series Editor:
Christian V. Stevens, Faculty of Bioscience Engineering, Ghent University, Belgium
Biofuels
Wim Soetaert, Erik Vandamme
Aqueous Pretreatment of Plant Biomass for Biological and Chemical Conversion to Fuels and
Chemicals
Charles E. Wyman
Lignin and Lignans as Renewable Raw Materials: Chemistry, Technology and Applications
Francisco G. Calvo‐Flores, Jose A. Dobado, Joaquín Isac‐García, Francisco J. Martín‐Martínez
Sustainability Assessment of Renewables‐Based Products: Methods and Case Studies
Jo Dewulf, Steven De Meester, Rodrigo A. F. Alvarenga
Fuels, Chemicals and Materials from the Oceans and Aquatic Sources
Francesca M. Kerton, Ning Yan
Bio‐Based Solvents
François Jérôme and Rafael Luque
Forthcoming Titles:
The Chemical Biology of Plant Biostimulants
Danny Geelen, Lin Xu
Biorefinery of Inorganics: Recovering Mineral Nutrients from Biomass and Organic Waste
Erik Meers, Gerard Velthof
CARMEN G. BOERIU
Wageningen Food & Biobased Research
Wageningen
The Netherlands
This edition first published 2020
© 2020 John Wiley & Sons Ltd
All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted,
in any form or by any means, electronic, mechanical, photocopying, recording or otherwise, except as
permitted by law. Advice on how to obtain permission to reuse material from this title is available at
http://www.wiley.com/go/permissions.
The right of Lambertus A.M. van den Broek and Carmen G. Boeriu to identified as the authors of the editorial
material in this work has been asserted in accordance with law.
Registered Offices
John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
John Wiley & Sons Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK
Editorial Office
The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK
For details of our global editorial offices, customer services, and more information about Wiley products visit us
at www.wiley.com.
Wiley also publishes its books in a variety of electronic formats and by print‐on‐demand. Some content that
appears in standard print versions of this book may not be available in other formats.
ISBN: 9781119450436
10 9 8 7 6 5 4 3 2 1
Contents
5 Chitosan‐Based Hydrogels 97
Zhengke Wang, Ling Yang, and Wen Fang
5.1 Introduction 97
5.2 Chitosan‐Based Multilayered Hydrogels 98
5.2.1 Periodic Precipitation 99
5.2.2 Alternating Process 100
5.2.3 Induced by Electrical Signals 100
5.2.4 Layer‐by‐Layer (LbL) Assembly 101
5.2.5 Sequential Curing 101
5.3 Chitin/Chitosan Physical Hydrogels Based on Alkali/Urea Solvent System 103
5.3.1 Chitin Hydrogels Based on Alkali/Urea Solvent System 104
5.3.2 Chitosan Hydrogels Based on Alkali/Urea Solvent System 104
5.4 Chitosan‐Based Injectable Hydrogels 108
5.4.1 Physical Association Networks 108
5.4.2 Chemical Association Networks 110
5.4.3 Double‐Network Hydrogels 116
5.5 Chitosan‐Based Self‐Healing Hydrogels 119
5.5.1 Physical Interactions 119
5.5.2 Dynamic Chemical Bonds 121
5.6 Chitosan‐Based Shape Memory Hydrogels 125
5.6.1 Water‐/Solvent‐Triggered Shape Recovery 126
5.6.2 pH‐triggered Shape Recovery 126
5.6.3 Ultrasound Triggered Shape Recovery 126
5.6.4 Self‐Actuated Shape Memory Hydrogels 127
5.6.5 Chitosan‐Based Hydrogels with Triple Shape Memory Effect 127
5.7 Superabsorbent Chitosan‐Based Hydrogels 131
5.7.1 Cross‐Linked Chitosan‐Based Hydrogels 132
5.7.2 Hydrogels by Graft Copolymerization 133
5.7.3 Chitosan‐Based Composite Hydrogels 134
5.7.4 Pure Chitosan‐Based Materials 135
5.8 Outlook 136
References136
6.5 Outlook 164
Acknowledgement164
References164
11.5 Outlook 276
Acknowledgment276
References276
Index491
List of Contributors
Els D’Hondt VITO (Flemish Institute for Technological Research), Mol, Belgium
Liyou Dong Food & Health Research, Wageningen Food & Biobased Research,
Wageningen, The Netherlands; Food Chemistry, Wageningen University, Wageningen, The
Netherlands
Hermann Ehrlich Institute of Electronics and Sensor Materials, TU Bergakademie‐
Freiberg, Freiberg, Germany
Vincent G.H. Eijsink Faculty of Chemistry, Biotechnology, and Food Science, The
Norwegian University of Life Sciences (NMBU), Ås, Norway
Kathy Elst VITO (Flemish Institute for Technological Research), Mol, Belgium
Wen Fang Institute of Biomedical Macromolecules, Department of Polymer Science
and Engineering, Zhejiang University, Hangzhou, China
Maria Emiliana Fortuna ‘Petru Poni’ Institute of Macromolecular Chemistry,
Romanian Academy, Iași, Romania
Coen Govers Food & Health Research, Wageningen Food & Biobased Research,
Wageningen, The Netherlands
Natalia Gutowska Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Karolina Gzyra‐Jagieła Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Tom Hager German Institutes of Textile and Fiber Research, Denkendorf, Germany
Thomas Hahn Fraunhofer Institute of Interfacial Engineering and Biotechnology,
Stuttgart, Germany
Valeria Harabagiu ‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian
Academy, Iași, Romania
Antoine Hubert Ynsect, Évry, France
Andra Cristina Humelnicu ‘Petru Poni’ Institute of Macromolecular Chemistry,
Romanian Academy, Iași, Romania
Maria Ignat ‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian Academy,
Iași, Romania
Teofil Jesionowski Institute of Chemical Technology and Engineering, Faculty of
Chemical Technology, Poznan University of Technology, Poznan, Poland
Yvonne Joseph Institute of Electronics and Sensor Materials, TU Bergakademie‐
Freiberg, Freiberg, Germany
Malgorzata Kaisler Bioprocess Engineering Group, Wageningen University,
Wageningen, The Netherlands; Wageningen Food & Biobased Research, Wageningen, The
Netherlands
Christine Klinger Institute of Physical Chemistry, TU Bergakademie‐Freiberg,
Freiberg, Germany
List of Contributors xix
Cristiane Krause Santin Food and Chemical Engineering, Polytechnic School, Unisinos
University, São Leopoldo, RS, Brazil; itt CHIP – Unisinos Semiconductor Institute, São
Leopoldo, RS, Brazil
Magdalena Kucharska Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Liziane Dantas Lacerda Food and Chemical Engineering, Polytechnic School, Unisinos
University, São Leopoldo, RS, Brazil
Guilherme Lopes Batista itt CHIP – Unisinos Semiconductor Institute, São Leopoldo,
RS, Brazil
Longina Madej‐Kiełbik The Institute of Security Technologies “MORATEX”, Lodz,
Poland
Sophanit Mekasha Faculty of Chemistry, Biotechnology, and Food Science, The
Norwegian University of Life Sciences (NMBU), Ås, Norway
Bruno M. Moerschbacher University of Münster, Institute for Biology and
Biotechnology of Plants, Münster, Germany
Anna Niehues University of Münster, Institute for Biology and Biotechnology of Plants,
Münster, Germany
Monika Owczarek Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Xenia Patras ‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian Academy,
Iași, Romania
Bożenna Pe ̨czek Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Cristian Peptu ‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian Academy,
Iași, Romania
Iaroslav Petrenko Institute of Experimental Physics, TU Bergakademie‐Freiberg,
Freiberg, Germany
Razvan Rotaru ‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian
Academy, Iași, Romania
Petrisor Samoila ‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian
Academy, Iași, Romania
Monika Sikora Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Lise Soetemans VITO (Flemish Institute for Technological Research), Mol, Belgium
Daiana de Souza Food and Chemical Engineering, Polytechnic School, Unisinos
University, São Leopoldo, RS, Brazil
Thomas Stegmaier German Institutes of Textile and Fiber Research, Denkendorf,
Germany
Marcin H. Struszczyk The Institute of Security Technologies “MORATEX”, Lodz,
Poland
xx List of Contributors
Renewable resources, their use and modification are involved in a multitude of important
processes with a major influence on our everyday lives. Applications can be found in the
energy sector; paints and coatings; and the chemical, pharmaceutical, and textile industry,
to name but a few.
The area interconnects several scientific disciplines (agriculture, biochemistry, chemis-
try, technology, environmental sciences, forestry), which makes it very difficult to have an
expert view on the complicated interaction. Therefore, the idea to create a series of scien-
tific books, focusing on specific topics concerning renewable resources, has been very
opportune and can help to clarify some of the underlying connections in this area.
In a very fast‐changing world, trends are not only characteristic of fashion and political
standpoints; science too is not free from hypes and buzzwords. The use of renewable
resources is again more important nowadays; however, it is not part of a hype or a fashion.
As the lively discussions among scientists continue about how many years we will still be
able to use fossil fuels – opinions ranging from 50 to 500 years – they do agree that the
reserve is limited, and that it is essential not only to search for new energy carriers but also
for new material sources.
In this respect, the field of renewable resources is a crucial area in the search for alterna-
tives for fossil‐based raw materials and energy. In the field of energy supply, biomass‐ and
renewables‐based resources will be part of the solution alongside other alternatives such as
solar energy, wind energy, hydraulic power, hydrogen technology and nuclear energy. In
the field of material sciences, the impact of renewable resources will probably be even big-
ger. Integral utilisation of crops and the use of waste streams in certain industries will grow
in importance, leading to a more sustainable way of producing materials. Although our
society was much more (almost exclusively) based on renewable resources centuries ago,
this disappeared in the Western world in the nineteenth century. Now it is time to focus
again on this field of research. However, it should not mean a ‘retour à la nature’, but
should be a multidisciplinary effort on a highly technological level to perform research
towards new opportunities, to develop new crops and products from renewable resources.
This will be essential to guarantee an acceptable level of comfort for the growing number
of people living on our planet. It is ‘the’ challenge for the coming generations of scientists
to develop more sustainable ways to create prosperity and to fight poverty and hunger in
the world. A global approach is certainly favoured.
xxii Series Preface
This challenge can only be dealt with if scientists are attracted to this area and are recog-
nised for their efforts in this interdisciplinary field. It is, therefore, also essential that con-
sumers recognise the fate of renewable resources in a number of products.
Furthermore, scientists do need to communicate and discuss the relevance of their work.
The use and modification of renewable resources may not follow the path of the genetic
engineering concept in view of consumer acceptance in Europe. Related to this aspect, the
series will certainly help to increase the visibility of the importance of renewable resources.
Being convinced of the value of the renewables approach for the industrial world, as well
as for developing countries, I was myself delighted to collaborate on this series of books
focusing on the different aspects of renewable resources. I hope that readers become aware
of the complexity, the interaction and interconnections, and the challenges of this field, and
that they will help to communicate on the importance of renewable resources.
I certainly want to thank the people of Wiley’s Chichester office, especially David
Hughes, Jenny Cossham and Lyn Roberts, in seeing the need for such a series of books on
renewable resources, for initiating and supporting it, and for helping to carry the project to
the end.
Last, but not least, I want to thank my family, especially my wife Hilde and children
Paulien and Pieter‐Jan, for their patience, and for giving me the time to work on the series
when other activities seemed to be more inviting.
Christian V. Stevens,
Faculty of Bioscience Engineering
Ghent University, Belgium
Series Editor, ‘Renewable Resources’
June 2005
Preface
Chitin was reported for the first time about 200 years ago, in extracts of mushrooms and
insects. About 40 years later, chitosan was obtained from chitin by acid treatment. These
polysaccharides are among the most abundant natural biopolymers in the world. They are,
for example, present in crustaceans, insects and fungi. Just before World War II, there was
a huge interest in the applications of these polysaccharides as a bioplastic. However, the
simultaneous upcoming of synthetic polymers and the exponential increase in high‐
performance synthetic polymers, which outperformed their natural counterparts, resulted
in a decrease of interest in chitin/chitosan materials. In the 1970s, large‐scale production
of chitin and chitosan from the shells of marine organisms started, owing to the develop-
ment of aquaculture and the enactment of severe environmental regulations to decrease
the amount of shellfish dumping in the oceans. Nowadays there is a need to be less
dependent on fossil resources. The transition to a biobased economy and the increasing
societal demand for more green and environmentally friendly products urge us to look for
chemicals, materials and fuels based on renewable resources. The enormous potential of
chitin and chitosan on account of their abundance, unique properties and numerous appli-
cations makes them interesting biomass resources. This book, Chitin and Chitosan:
Properties and Applications, shows the state‐of‐the‐art and future perspectives of chitin
and chitosan materials and applications. The book presents the most recent developments
in the science and technology of all related fields, from extraction and characterisation to
modification, material synthesis and end‐user applications. This book comprises 19 chapters
that deal with most topics related to chitin and chitosan polymers and materials.
In Chapters 1–4, the sources of chitin and chitosan are described and how these biopoly-
mers can be isolated. Next to the isolation, the analysis of the biopolymers is described.
The different sources and/or isolation methods can result in different structures and proper-
ties. In Chapter 5–7, hydrogels, health effects and the anti‐microbial effects of chitin and
chitosan are discussed. To improve or to modify the properties, enzymes and chemical
reactions can be applied to customise these biopolymers, as shown in Chapters 8–10. The
applications of chitin and chitosan in drug delivery, medical devices, agriculture, food,
packaging, horticulture, textile, water purification and sensors are discussed in more detail
in Chapters 11–18. And finally, Chapter 19 is devoted to the market and regulation of chitin
and chitosan.
xxiv Preface
These topics have never been addressed previously in a single book. Books, book chap-
ters and reviews have been dedicated to the specific fields of application of chitin and chi-
tosan materials. This book presents an overview of the latest scientific and technological
advances in almost all areas of application, and show the great potential of chitin and chi-
tosan as materials of the future. We hope that the reader will be inspired by the examples
given of these biopolymers in different areas. We are confident that chitin and chitosan will
become major renewable resources in the biobased circular economy.
This book should be useful for scholars and those in academia, such as undergraduate
and postgraduate students in the areas of agriculture, polymer and material sciences,
biobased economy and life sciences. In addition, we hope this book will aid researchers
and specialists from industry in the field of (bio)polymers, packaging, biomedical applica-
tions, water treatment, textiles, sensors, and agriculture and food – as well as regional and
national policy‐makers.
The input is from well‐known experts from all over the world. We would like to express
our great gratitude to all chapter authors of this book, who have made excellent contribu-
tions. In addition, we would like to thank Sarah Higginbotham, Emma Strickland and
Lesley Jebaraj from Wiley for all their help.
Chitin is a natural biomolecule that was reported for the first time in 1811 by the French
professor Henri Braconnot as fungine [1] and in 1823 by Antoine Odier as chitin. Chitin
consists of large, crystalline nitrogen‐containing polysaccharides made of chains of a mod-
ified glucose monosaccharide, being N‐acetylglucosamine. It is ubiquitously present in the
world and has even been reported to be one of the most abundant biomolecules on earth,
with an estimated annual production of 1011–1014 tons [2, 3]. Chitin serves as template for
biomineralization such as calcification and silicification, providing preferential sites for
nucleation, and controlling the location and orientation of mineral phases [4, 5]. This phe-
nomenon explains the presence of chitin in solid structures in a variety of biomass such as
cell walls of fungi and diatoms and in exoskeletons of Crustaceans. Chitin is present in
diverse structures in at least 19 animal phyla besides its presence in bacteria, fungi, and
algae [5].
Chitosan is mainly known as a partially deacetylated derivative of chitin that is more
water soluble than chitin, and as such is easier to process. For this reason, chitosan—and,
in some cases, even more preferably, the relatively small sized (1–10 kDa) chitosan
oligomers—are the molecules that are envisioned for multiple applications such as agricul-
ture; water and wastewater treatment; food and beverages; chemicals; feed; cosmetics; and
personal care [6, 7]. In addition, chitosan oligomers have been reported as being bioactive
[8], offering potential for application in, for instance, wound dressing and cosmetics.
Although chitin and chitosan are versatile and promising biomaterials [9], the extraction
and purification of chitin and its conversion to chitosan (oligomers) require several process
steps, and these have been mentioned as bottlenecks that hinder the wider use of the under-
spent chitin in the world.
This chapter intends to provide more information related to (1) the structure of chitin, (2)
sources of chitin and chitosan, and (3) their extraction and purification, as well as (4) the
conversion of chitin into chitosan. The further conversion of chitosan to chitosan oligomers
is the subject of Chapter 3.
Algae Crustaceans
Chitin
OH OH OH OH
H O H O H H O H H O H
H O H H O H
H H H H
OH OH OH OH
O H
O O
H HN H H NH H NH
NH2
O O O
H3C H3C H3C
n
Deacetylation
Chitosan
Fungi OH OH Mollusks
OH OH
H O H O H H O H H O H
H O H H O H
H H H H
OH OH OH OH
O H
O O
H NH2 H H NH H NH2
NH2
O
H 3C
n
Figure 1.1 Chemical structure of chitin and chitosan and some examples of species that contain chitin.
(a) (b)
Sheet 1
H C
H C H C
H C
H C H C O
H C O O
H C
HO O O
H C O O
H C HO
O O O O
O O
OH O
O NH OCH
O
O NH OCH NH OCH O
O O O HO
O OH
HO O NH CH
OH NH OCH NH CH HO
NH OCH O NH CH
O H C NH HO HO
HO
OH NH CH HO HO OH
NH CH H CO NH O OH OH
HO OH
HO OH
OH O
O OH OH
O OH O
OH
O O
O O O
O O O
OH O O O O
O O O
O O O
O NH OCH
O
OH NH OCH NH OCH O
OH NH OCH O O HO
NH OCH H C O HO NH CH
O NH HO NH CH HO
OH NH CH NH CH
HO NH CH O HO
H CO NH HO OH
HO OH HO
HO O OH OH
OH O OH
O O OH OH OH
OH O
O O O O O
O
OH O
O
O O O O
O O O O
O OH O O
O OH NH OCH O NH OCH
NH OCH O NH OCH NH OCH O
O H C NH HO O HO
OH NH CH HO O NH CH
HO NH O NH CH HO
CH H CO NH NH CH
HO HO
HO OH HO OH
OH O
O OH HO
O
OH
OH OH
OH OH
O O OH O
O O
OH O O
O O O
O OH O O O O
O OH O O O
O NH OCH O O
O NH OCH H C NH HO NH OCH
OH NH OCH O
HO NH CH O O NH OCH HO
NH CH H CO NH O HO NH
HO HO CH
HO OH NH CH NH CH
OH O HO
O HO OH
O OH OH HO
OH HO
OH
HO O OH
CH OH OH HO
HO HO
HO CH HO
Chain 1 n HO HO
HO
n
n n
Figure 1.2 Schematic representation of (a) α‐form and (b) β‐form of chitin.
Sources of Chitin and Chitosan and their Isolation 5
for solvents and a higher reactivity. It is proven to be soluble in formic acid and can be
swollen in water [15]. This chitin form is present in the squid pen, in the tubes of pogo-
nophoran and vestimentiferan worms, and in monocrystalline spines excreted by diatoms
such as Thalassiosira fluviatilis [7]. Although squid and tubes of Tevnia jerichonana both
contain β‐chitin, their crystallinity differs. This implies that the crystallinity also depends
on the source. Chitin obtained from squid pens is semi‐crystalline, and chitin from T. jeri-
chonana is almost complete crystalline [7, 8, 16].
The third formation, γ‐chitin, is less common. It is considered to be a mixture or inter-
mediate form of α‐ and β‐chitin with both parallel and antiparallel arrangements [16]. More
specifically, every third chitin chain has the opposite direction to the two preceding chitin
sheets [13, 15]. Very few studies have been carried out on γ‐chitin, and it has been s uggested
that γ‐chitin may be a distorted version of the other two instead of a true third polymorphic
form.
(Continued )
Note: *not provided how it is measured; ^based on the weight of the organic cuticle, others were measured based on weight differences of the raw materials and that of the sample
obtained after acid and alkaline treatments2, crude ash3 based on acid detergent fiber, minus present amino acids.
chitin‐rich pen that represents a very minor fraction of the whole squid. Chitin may be
involved in the formation of skeletons in calcifying marine sponges [28]. Sponges are
described more in detail in Chapter 4. Within the Cnidarian taxa, skeletons often contain,
besides chitin, calcium‐based minerals. Black corals form an exception and have a unique
halogenated scleroprotein named antipathin associated with chitin [22]. Lophophorates
(marine and freshwater Octopoda, Phoronida, Brachiopoda) have exoskeletons, named
tubes, that consists of chitin [18]. Chitin is the most important ultrastructural compound of
fungal cell walls, where it is embedded in the amorphous matrix and provides the frame-
work of the cell wall morphology [55]. It exists in the spores, mycelia, and stalks, and has
only been detected as α‐chitin [55]. Its amount ranges from 2% to 50% (w/w) on dry cell
wall base, whereby the lowest value corresponds to yeasts [56] and the highest to
Euascomycetes [55]. Depending on the class of fungi, the cell wall can also contain glu-
cans, mannans, as well as chitosan. As the cell wall is only a part of the fungal biomass, the
overall chitin (plus chitosan) yield is lower, and values have been reported (glucosamine on
dry matter base) of 8–16% (w/w) for Aspergillus niger and Mucor rouxii mycelia [48] and
12% (w/w) for Agaricus bisporus stalks [47].
In the case of algae, since 1965, diatoms such as Thalassiosira were reported to secrete
β‐chitin ropes that span between two recently divided daughter cells to keep them together,
creating flexible cell chains that float in the water [27, 50]. However, chitin has also been
shown to be present in diatoms in other forms—for instance, in the siliceous shell. In calci-
fied coralline algae such as Clathromorphum compactum, chitin has been reported to be
present that strengthens the skeleton and protects the algae from ocean acidification and
grazing in shallow waters [28]. The presence of chitin was also demonstrated in the cell
walls of the green algae Pithophora oedogonia [30] and Chlorella vulgaris [29]. Quantitative
data on the chitin content in the algae, however, are scarce. The fact that chitin in algae is
plant‐based can be an advantage for some applications.
degradation of chitin in nature. Further, these fast‐growing bacteria, or the isolated enzymes,
offer a tool for biological chitosan production [3] (see Section 1.4).
Chitin
Pre- Post-
containing treatment Deproteination Demineralisation Decoloration treatment Chitin
biomass
Figure 1.3 Processes involved in the isolation and purification of chitin from biomass.
1.3.1.1 Pre‐Treatment
The pretreatment step groups all activities that are required to prepare the biomass for chi-
tin extraction. This may comprise the removal of soft tissues by scraping, boiling, and
pressing. For other organisms, boiling can be a part of a hygienization step. The biomass is
mostly dried and reduced in size. However, for fermentation purposes, for example, drying
is not necessary. Examples of pre‐treatments are given in Table 1.2.
1.3.1.2 Demineralization
When a high amount of minerals is present, demineralization is advisable. For example, the
exoskeleton of crustaceans can contain more than 50% (w/w) of CaCO3 to enhance its
strength [38, 52]. Two approaches have been reported—chemical and biological deminer-
alization. Chemical demineralization uses acids such as HCl, HNO3, H2SO4, CH3COOH,
and HCOOH [60]. Among these acids, HCl is the most commonly used reagent for remov-
ing the mineral constituents [40]. Biological demineralization is based on acid‐producing
biological processes using bacteria [3, 39, 40] or enzymes such as Alcalase® [40]. The acids
12 Chitin and Chitosan: Properties and Applications
formed (such as lactic acid) react with calcium carbonate, resulting in a precipitate that can
be removed by washing. Khanafari et al. stated that fermentation is less likely to change the
physicochemical characteristics of the chitin chain [65]. Table 1.3 summarizes some dem-
ineralization procedures as examples.
1.3.1.3 Deproteination
Chitin is traditionally viewed as a chain that is embedded in a protein‐matrix. During the
purification step, complete removal of proteins is advisable when applications in medical,
food, or feed are envisioned since proteins can be allergenic [10]. The proteins are bound
by multiple hydrogen bridges, and the ‘free’ amino groups of the chitin may covalently be
bound to the proteins [8, 51]. Therefore, extreme process parameters are often required for
the deproteination. Chemical deproteination is commonly performed with sodium hydrox-
ide as a preferential reagent. The effectiveness of alkali deproteination depends on the
process temperature, the alkali concentration, as well as the alkali/biomass ratio [26].
During deproteination, chitin may undergo some changes such as partial deacetylation and
hydrolysis of the polymer [10]. Also, biological deproteination using (1) proteases (such as
pepsin, papain, and trypsin) secreted by proteolytic bacteria in the fermentation medium or
(2) isolated proteases (crude or purified) has been reported [39, 40]. As the conditions are
less harsh as compared to chemical deproteination, small peptides and amino acids remain
attached to the chitin chain after enzymatic hydrolysis [10], and the deacetylation degree is
altered less (less increase) [72]. Some examples are summarized in Table 1.4.
Sources of Chitin and Chitosan and their Isolation 13
c omplete within 15 min at ambient temperature [60]. Younes et al. performed a two‐level
fractional factorial design and concluded that the number of batches and the concentration
of HCl are the most influential parameters for the demineralization efficiency. As tempera-
ture was not found to be a significant parameter, lower temperatures are preferred to reduce
chain degradation and chitin deacetylation. The best demineralization for shrimp was
found when using lower temperatures and longer reaction times [71]. Tolaimate et al. con-
firmed that the number of repeats (a multi process) had a positive influence, but did not find
a relation between higher acid concentration and longer reaction time in their settings [74].
Biological demineralization by the use of fermentation, for instance, solid‐state fermenta-
tion with organic acid–producing bacteria, received more attention recently as being a
more eco‐friendly, safer, and more technologically flexible alternative to chemical demin-
eralization. Arbia et al. and Kaur et al. gave an overview of the most common bacteria
utilized [39, 40]. Lactic acid–producing bacteria were usually selected, for example,
Lactobacillus spp. B2 or L. plantorum A3. Other acid‐producing microorganisms that were
tested include Bacillus subtilis, Pseudomonas aeruginosa K‐187, and Serratia marcescens
FS‐3. Since the acid also lowered the pH, which activated proteases [10], demineralization
and deproteination occurred partially simultaneously. Khanafari et al. proved that deminer-
alization using fermentation could be evenly effective if not better than chemical deminer-
alization. They also stated that fermentation was less likely to change the physicochemical
characteristics of the chitin chain [65].
Sources of Chitin and Chitosan and their Isolation 15
have been studied [61]. Although the deproteination levels achieved in such cases are gen-
erally lower than those obtained using alkaline treatments, this alternative has the advan-
tage to produce nutritionally valuable protein hydrolysates in addition to chitin.
In conclusion, the intensity of the demineralization and deproteination steps depends on
the source [74]. It is generally accepted that these steps significantly change the physico-
chemical properties of chitin, for example, the molecular weight and acetylation degree
[40, 60]. Both the degree of crystallinity and the acetylation degree of chitin depend on the
source and the method of purification [62]. Most researchers favor performing the demin-
eralization first, followed by deproteination [74]. However, it is considered that the order
of these two phases in interchangeable depending on the source [61, 79].
(Continued)
Chitosan
H O H O H H O H H O H H O H O H
H O H H O H
H O H H O H H O H O
H H H
OH H H H H H H H
H OH OH OH OH OH OH OH
O O O H
O O O
H HN H H NH H NH H NH NH NH
NH H NH H H
O O O O
H C H C H C H C
n n
CH3
R1
O O– O
O
OH
NH
R NH + OH– ⇋ R ⇋ R OH
+ NHR1–
O
NH O HO
1
R OH O
1 HO O
R
NH
OH
O
O CH3
+ n
NH2R1 R
R O–
follows a pseudo‐first‐order kinetic during the initial period (first hour) when the concen-
tration of alkaline is high [76, 93].
Two traditional deacetylation procedures described by Broussignac [98] and Kurita et al.
[99] were compared by Tolaimate et al. using α‐chitin and β‐chitin from different sources.
The Broussignac procedure is based on an anhydrous reaction medium composed of 50
w/w% potassium hydroxide, 25 w/w% 96% (v/v) ethanol, and 25 w/w% monoethylene
glycol. For the Kurita procedure, an aqueous sodium hydroxide solution is used. Tolaimate
et al. showed a benefit of the Kurita approach for the deacetylation of β‐chitin, but not for
α‐chitin. For α‐chitin, the method of Broussignac proved to be more efficient, explained by
the higher solubility of NaOH in aqueous media as compared to the solubility of KOH in
the anhydrous media. The dielectric constant of NaOH in aqueous solution is much higher
and perhaps favors the development of charges (during the deacetylation reaction). They
also found that α‐chitin treated with the Kurita medium required a higher reaction time for
reaching the same DDA as Broussignac, and this happened at the expense of the molecular
weight of chitosan and its quality. They suggested that the difference between α and β chi-
tin can be due to morphological changes over time leading to more accessible amine
groups. For α‐chitin, these changes may be quicker in time for the Broussignac media, and
may thus lead to more efficient deacetylation [74]. Despite these results, the Kurita proce-
dure is considered the most common process, as indicated in Table 1.7. In general, deacety-
lation is performed at high NaOH concentrations (30–60 w%), high temperatures
(80–150°C), and with long reaction times (1–80 hours) [8, 13, 70, 100, 101].
for lobster or red crab chitin [74]. The intensity of the deproteination caused differences
in partial deacetylation (5–20%) [16].
• Example 2: The optimal process parameters for α‐chitin extracted from shrimp waste
were 100°C for 12 hours with 50 w% NaOH in the study by Tokali et al., where a DDA
of 80.06% was obtained [66]. For β‐chitin, 50 w% NaOH at 90°C for 6 hours proved to
be best and a DDA of 95.6% was achieved [70].
• Example 3: Younes et al. investigated the impact of seven factors on the deacetylation of
shrimp chitin. A clear significant effect of temperature and the alkali reagent was
detected (NaOH was much more efficient than KOH). Also, the DDA was significantly
affected by the use of repeats, the reaction time, and the concentration of alkali. The
atmospheric (nitrogen or air) condition and the use of reducing agents (NaBH4), how-
ever, did not have a significant effect on the DDA, but influenced the molecular weight
of chitosan.
c hitosan with a DDA of 71–88% with deacetylase [105]. Besides the necessary pre‐
treatment, deacetylase has a high production cost and requires a long reaction time. This
makes the enzymatic approach less ideal for commercial applications [100].
1.4.2.1 Pretreatment
Fungi are a wet biomass and are typically extensively washed, dried, and refined before
processing. Their low mineral content, on the other hand, does not require a demineraliza-
tion step that is needed in the chitin production from marine resources. Moreover, as some
fungi of particular interest are rich in the deacetylated chitosan, often the isolation of this
particular compound is targeted, leaving the (crude) chitin behind. This not only alters the
details of the extraction step to allow the specific isolation of chitosan, but also avoids the
need for a post‐deacetylation processing step.
(1–4 M), high temperature (80–121°C), and short contact times (15 min–3 hours) [48, 59,
60, 89, 106]. The alkali‐insoluble residue, containing the chitin, chitosan, and beta‐glucan,
is separated and extensively washed till neutral pH before further processing.
fermentation, and its availability does not experience geographic or seasonal limitations.
The better controllable fermentation guarantees a chitin/chitosan final product with more
consistent properties [119]. Fungal mycelium also has some compositional advantages as
compared to crustacean wastes. It contains less inorganic compounds, avoiding the need
for a demineralization treatment in the chitin production [120]. Moreover, whereas c hitosan
Table 1.10 Advantages and disadvantages of three chitin sources (partially based on [58]).
is not native to animal resources, it is present as structural components in the cell wall of
fungi from the Zygomycetes class, such as Mucor, Absidia, and Rhizopus [55]. In addition,
recent observations suggested that non‐Zygomycetes plant and insect pathogenic fungi
also have high proportions of chitosan in the cell walls [121]. These types of fungi allow
the direct production of chitosan, avoiding the need for a deacetylation step to transform
chitin to chitosan. Additionally, fungal chitosan is free of allergenic shrimp protein and has
been reported to have beneficial properties in terms of homogeneous polymer length and
solubility over the current marine source [58]. However, the processes of chitosan produc-
tion from fungi are not scaled up to industrial level [106].
As insect farms are currently being established in Europe with focus on selected insect
species, insects become a potential alternative source for chitin production. Compared to
crustaceans, the insect‐based source (exoskeletons) is more homogeneous (mostly one spe-
cies) and is not seasonally dependent. As with fungi, the mineral content is lower, which
justifies reducing the demineralization step to a minimum. On the other hand, the chitin
production process from insects is less studied, and mostly procedures elaborated for crus-
taceans are applied.
Although commercial facilities for chitin and chitosan production exist (for Crustaceans),
the examples mentioned in the previous sections were mainly performed at lab scale (<1–
40 g). Upscaling to pilot and full scale usually requires modifications to the protocols.
Lopez et al. estimated, based on lab date, the environmental impact of chitin production
processes that involve strong acids and bases at high temperature in multiple process steps
[122]. The chemical extraction of chitin would require, per ton chitin, 72.7 m3 of water,
12231 kWh, 727 kg of NaOH, 2319 kg HCl, and 354 kg NaClO, and would produce 1.8
tons of CO2 and 21.8 tons of wastewater. Switching to enzymatic deproteintion and water
recovery would generate only half of the chitin from the same biomass, but could reduce
the environmental impact per ton to 0.9 m3 of water, 516 kWh, 46 kg of NaOH, 339 kg HCl,
84 kg NaClO, and 3.7 kg of enzymes, and would produce 0.08 tons of CO2 and 0.2 tons of
wastewater. Both approaches were predicted to be profitable. Gómez‐Rios (2017) also
reported on profitable chitosan production from shrimp shell waste, with low volumetric
yields and high water requirements as main drawbacks. Re‐use of water, including enzy-
matic processes and co‐valorization of other compounds, would increase the sustainability
of the process [123].
1.6 Outlook
Chitin is a natural polysaccharide that is omnipresent in nature. Despite the diverse poten-
tial applications of chitin, the compound is underspent. Upscaling of the multistep extrac-
tion process and the production of reproducible chitin and chitosan products in a more
sustainable way are challenges that need to be overcome to increase the commercial pro-
duction activities of crustaceans and extend them to other source such as fungi and insects.
References
1. R. A. A. Muzzarelli, J. Boudrant, D. Meyer, N. Manno, M. Demarchis, and M. G. Paoletti, “Current
views on fungal chitin/chitosan, human chitinases, food preservation, glucans, pectins and inulin: A
tribute to Henri Braconnot, precursor of the carbohydrate polymers science, on the chitin bicentennial,”
Carbohydr. Polym., vol. 87, no. 2, pp. 995–1012, 2012.
Sources of Chitin and Chitosan and their Isolation 29
2. A. Jardine and S. Sayed, “Challenges in the valorisation of chitinous biomass within the biorefinery
concept,” Curr. Opin. Green Sustain. Chem., vol. 2, no. 2016, pp. 34–39, 2016.
3. K. Kaur, V. Dattajirao, V. Shrivastava, and U. Bhardwaj, “Isolation and characterization of
chitosan‐producing bacteria from beaches of Chennai, India,” Enzyme Res., vol. 2012, Article
ID421683, 2012.
4. H. Merzendorfer, “The cellular basis of chitin synthesis in fungi and insects: Common principles and
differences,” Eur. J. Cell Biol., vol. 90, no. 9, pp. 759–769, 2011.
5. H. Ehrlich, "Chitin and collagen as universal and alternative templates in biomineralization," Int.
Geology Rev., vol. 52, no. 7–8, pp. 661–699, 2010.
6. M. Zuber, K. M. Zia, and M. Barikani, “Chitin and chitosan based blends, composites and nanocom-
posites,” In: Thomas S., Visakh P., Mathew A. (eds) Advances in Natural Polymers. Advanced
Structured Materials, vol. 18, Springer, Berlin, Heidelberg, pp. 55–119, 2013.
7. M. Rinaudo, “Chitin and chitosan: Properties and applications,” Prog. Polym. Sci., vol. 31, no. 7,
pp. 603–632, 2006.
8. K. Kurita, “Chitin and chitosan: Functional biopolymers from marine crustaceans,” Mar. Biotechnol.,
vol. 8, no. 3, pp. 203–226, 2006.
9. V. Zargar, M. Asghari, and A. Dashti, “A review on chitin and chitosan polymers: Structure, chemistry,
solubility, derivatives, and applications,” ChemBioEng Rev., vol. 2, no. 3, pp. 204–226, 2015.
10. I. Younes and M. Rinaudo, “Chitin and chitosan preparation from marine sources. Structure, properties
and applications,” Mar. Drugs, vol. 13, no. 3, pp. 1133–1174, 2015.
11. J. Z. Knaul, S. M. Hudson, K. A. M. Creber, and N. Carolina, “Crosslinking of chitosan fibers with
dialdehydes: Proposal of a new reaction mechanism,” J. Polym. Sci. B: Polym. Phys., vol. 37, pp.
1079–1094, 1999
12. M. Zuber and K. M. Zia, Advances in natural polymers, vol. 18, pp. 55–119, 2013.
13. I. Aranaz et al., “Functional characterization of chitin and chitosan,” Curr. Chem. Biol., vol. 3, pp.
203–230, 2009.
14. K. Y. Zhu, H. Merzendorfer, W. Zhang, J. Zhang, and S. Muthukrishnan, “Biosynthesis, turnover, and
functions of chitin in insects,” Annu. Rev. Entomol., vol. 61, no. 1, pp. 177–196, 2016.
15. Y. W. Cho, J. Jang, C. R. Park, and S. W. Ko, “Preparation and solubility in acid and water of partially
deacetylated chitins,” Biomacromolecules, vol. 1, no. 4, pp. 609–614, 2000.
16. K. Kurita, “Controlled functionalization of the polysaccharide chitin,” Prog. Polym. Sci., vol. 26, no.
9, pp. 1921–1971, 2001.
17. A. Einbu, Characterisation of chitin and a study of its acid‐catalysed hydrolysis, Thesis, Norwegian
University of Science and Technology, 2007.
18. L. Hyman, “The occurrence of chitin in the Lophophorate phyla,” Biol. Discov. Woods hole, vol. 24,
no. 5, pp. 362–369, 1958.
19. S. Kaur and G. S. Dhillon, “The versatile biopolymer chitosan: Potential sources, evaluation of
extraction methods and applications,” Crit. Rev. Microbiol., vol. 40, no. 2, pp. 155–175, 2014.
20. M. Poulicek and C. Jeuniaux, “Chitin biodegradation in marine environments: An experimental
approach,” Biochem. Syst. Ecol., vol. 19, no. 5, pp. 385–394, 1991.
21. S. M. Holl, J. Schaefer, W. M. Goldberg, K. J. Kramer, T. D. Morgan, and T. L. Hopkins, “Comparison
of black coral skeleton and insect cuticle by a combination of carbon‐13 NMR and chemical analyses,”
Arch. Biochem. Biophys., vol. 292, no. 1, pp. 107–111, 1992.
22. M. Bo et al., “Isolation and identification of chitin in the black coral Parantipathes larix (Anthozoa:
Cnidaria),” Int. J. Biol. Macromol., vol. 51, no. 1–2, pp. 129–137, 2012.
23. C. Nielsen, “Entoprocta,” Encycl. Life Sci., vol. 13, no. 1997, pp. 1–2, 2001.
24. H. Ehrlich et al., “Identification and first insights into the structure and biosynthesis of chitin from the
freshwater sponge Spongilla lacustris,” J. Struct. Biol., vol. 183, no. 3, pp. 474–483, 2013.
25. E. S. Abdou, K. S. A. Nagy, and M. Z. Elsabee, “Extraction and characterization of chitin and chitosan
from local sources,” Bioresour. Technol., vol. 99, no. 5, pp. 1359–1367, 2008.
30 Chitin and Chitosan: Properties and Applications
47. T. Wu, S. Zivanovic, F. A. Draughon, and C. E. Sams, “Chitin and chitosan‐value‐added products from
mushroom waste,” J. Agric. Food Chem., vol. 52, no. 26, pp. 7905–7910, 2004.
48. T. Wu, S. Zivanovic, F. A. Draughon, W. S. Conway, and C. E. Sams, “Physicochemical properties and
bioactivity of fungal chitin and chitosan,” J. Agric. Food Chem., vol. 53, no. 10, pp. 3888–3894, 2005.
49. A. Zamani, A. Jeihanipour, L. Edebo, C. Niklasson, and M. J. Taherzadeh, “Determination of glucosa-
mine and N‐acetyl glucosamine in fungal cell walls,” J. Agric. Food Chem., vol. 56, no. 18, pp. 8314–
8318, 2008.
50. M. Falk, D. G. Smith, J. McLachlan, and A. G. McInnes, “Studies on chitan (β‐(1‐4)‐linked 2‐aceta-
mido‐2‐deoxy‐D‐glucan) fibers of the diatom Thalassiosira fluviatilis Hustedt,” Can. J. Chem., vol.
44, no. 9096, 1965.
51. R. A. A. Muzzarelli, Chitin Nanostructures in Living Organisms, In: Gupta N. (eds) Chitin. Topics in
Geobiology, vol. 34. Springer, Dordrecht, 2011.
52. M. Kaya, K. Sofi, I. Sargin, and M. Mujtaba, “Changes in physicochemical properties of chitin at
developmental stages (larvae, pupa and adult) of Vespa crabro (wasp),” Carbohydr. Polym., vol. 145,
pp. 64–70, 2016.
53. M. Kaya, T. Baran, S. Erdoʇan, A. Menteş, M. Aşan Özüsaʇlam, and Y. S. Çakmak, “Physicochemical
comparison of chitin and chitosan obtained from larvae and adult Colorado potato beetle (Leptinotarsa
decemlineata),” Mater. Sci. Eng. C, vol. 45, pp. 72–81, 2014.
54. M. Rhazi, J. Desbrières, A. Tolaimate, A. Alagui, and P. Vottero, “Investigation of different natural
sources of chitin: Influence of the source and deacetylation process on the physicochemical character-
istics of chitosan,” Polym. Int., vol. 49, no. 4, pp. 337–344, 2000.
55. J. Ruiz‐Herrera, Fungal Cell Wall: Structure, Synthesis and Assembly, 2nd edn. CRC Press, Boca
Raton, 2016.
56. B. Aguilar‐Uscanga and J. M. François, “A study of the yeast cell wall composition and structure in
response to growth conditions and mode of cultivation,” Lett. Appl. Microbiol., vol. 37, no. 3, pp.
268–274, 2003.
57. G. S. Dhillon, S. Kaur, S. K. Brar, and M. Verma, “Green synthesis approach: Extraction of chitosan
from fungus mycelia,” Crit. Rev. Biotechnol., vol. 33, no. 4, pp. 379–403, 2013.
58. V. Ghormade, E. K. Pathan, and M. V. Deshpande, “Can fungi compete with marine sources for chi-
tosan production?,” Int. J. Biol. Macromol., vol. 104, pp. 1415–1421, 2017.
59. D. Kafetzopoulos, A. Martinou, and V. Bouriotis, “Bioconversion of chitin to chitosan: Purification
and characterization of chitin deacetylase from Mucor rouxii,” Proc. Natl. Acad. Sci. USA, vol. 90, no.
7, pp. 2564–2568, 1993.
60. A. Percot, C. Viton, and A. Domard, “Optimization of chitin extraction from shrimp shells,”
Biomacromolecules, vol. 4, no. 1, pp. 12–18, 2003.
61. J. A. Vázquez, I. Rodríguez‐Amado, M. I. Montemayor, J. Fraguas, M. P. Del González, and M. A.
Murado, “Chondroitin sulfate, hyaluronic acid and chitin/chitosan production using marine waste
sources: Characteristics, applications and eco‐friendly processes: A review,” Mar. Drugs, vol. 11, no.
3, pp. 747–774, 2013.
62. J. Li, J. Revol, and R. Marchessault, “Effect of degree of deacetylation of chitin on the properties of
chitin crystallites,” J. Appl. Polym. Vol. 65, no. 2, pp. 373–380, 1997.
63. M. H. Mohammed, P. A. Williams, and O. Tverezovskaya, “Extraction of chitin from prawn shells and
conversion to low molecular mass chitosan,” Food Hydrocoll., vol. 31, no. 2, pp. 166–171, 2013.
64. M. Bajaj, A. Freiberg, J. Winter, Y. Xu, and C. Gallert, “Pilot‐scale chitin extraction from shrimp shell
waste by deproteination and decalcification with bacterial enrichment cultures,” Appl. Microbiol.
Biotechnol., vol. 99, no. 22, pp. 9835–9846, 2015.
65. A. Khanafari, R. Marandi, and S. Sanatei, “Recovery of chitin and chitosan from shrimp waste by
chemical and microbial methods,” J. Environ. Heal. Sci. Eng., vol. 5, no. 1, pp. 1–24, 2008.
66. K. Tokatli and A. Demirdoven, “Optimization of chitin and chitosan production from shrimp wastes
and characterization.” J. Food Process. Preserv., vol. 42, no. 2, e13494, 2017.
32 Chitin and Chitosan: Properties and Applications
67. M. Finke and R. Verde, “Estimate of chitin in raw whole insects,” Zoo Biol., vol. 26, no. 2, pp. 201–
214, 2007.
68. S. Liu et al., “Extraction and characterization of chitin from the beetle Holotrichia parallela
Motschulsky,” Molecules, vol. 17, no. 4, pp. 4604–4611, 2012.
69. A. Tolaimate, J. Desbrières, M. Rhazi, A. Alagui, M. Vincendon, and P. Vottero, “On the influence of
deacetylation process on the physicochemical characteristics of chitosan from squid chitin,” Polymer
(Guildf)., vol. 41, no. 7, pp. 2463–2469, 2000.
70. J. Jung and Y. Zhao, “Characteristics of deacetylation and depolymerization of β‐chitin from jumbo
squid (Dosidicus gigas) pens,” Carbohydr. Res., vol. 346, no. 13, pp. 1876–1884, 2011.
71. I. Younes, S. Hajji, M. Rinaudo, M. Chaabouni, K. Jellouli, and M. Nasri, “Optimization of proteins
and minerals removal from shrimp shells to produce highly acetylated chitin,” Int. J. Biol. Macromol.,
vol. 84, pp. 246–253, 2016.
72. L. Manni, O. Ghorbel‐Bellaaj, K. Jellouli, I. Younes, and M. Nasri, “Extraction and characterization of
chitin, chitosan, and protein hydrolysates prepared from shrimp waste by treatment with crude pro-
tease from bacillus cereus SV1,” Appl. Biochem. Biotechnol., vol. 162, no. 2, pp. 345–357, 2010.
73. S. Kumari, P. Rath, A. Sri Hari Kumar, and T. N. Tiwari, “Extraction and characterization of chitin and
chitosan from fishery waste by chemical method,” Environ. Technol. Innov., vol. 3, pp. 77–85, 2015.
74. A. Tolaimate, J. Desbrieres, M. Rhazi, and A. Alagui, “Contribution to the preparation of chitins and
chitosans with controlled physico‐chemical properties,” Polymer (Guildf)., vol. 44, no. 26, pp. 7939–
7952, 2003.
75. A. Waśko, P. Bulak, M. Polak‐Berecka, K. Nowak, C. Polakowski, and A. Bieganowski, “The first
report of the physicochemical structure of chitin isolated from Hermetia illucens,” Int. J. Biol.
Macromol., vol. 92, pp. 316–320, 2016.
76. G. Galed, B. Miralles, I. Paños, A. Santiago, and Á. Heras, “N‐deacetylation and depolymerization
reactions of chitin/chitosan: Influence of the source of chitin,” Carbohydr. Polym., vol. 62, no. 4, pp.
316–320, 2005.
77. D. Gómez‐Ríos, R. Barrera‐Zapata, and R. Ríos‐Estepa, “Comparison of process technologies for
chitosan production from shrimp shell waste: A techno‐economic approach using Aspen Plus®,” Food
Bioprod. Process., vol. 103, pp. 49–57, 2017.
78. K. L. B. Chang and G. Tsai, “Response surface optimization and kinetics of isolating chitin from
pink shrimp (Solenocera melantho) shell waste,” J. Agric. Food Chem., vol. 45, no. 5, pp.
1900–1904,1997.
79. J. Synowiecki and N. A. Al‐khateeb, “Production, properties, and some new applications of chitin and
its derivatives,” Food Chem., vol. 43, no. 2, pp. 145–171, 2003.
80. K. S. Chae, C. S. Shin, and W. S. Shin, “Characteristics of cricket (Gryllus bimaculatus) chitosan and
chitosan‐based nanoparticles,” Food Sci. Biotechnol., vol. 27, no. 3, pp. 1–9, 2018.
81. M. Kaya et al., “Extraction and characterization of chitin and chitosan with antimicrobial and antioxi-
dant activities from cosmopolitan Orthoptera species (Insecta),” Biotechnol. Bioprocess Eng., vol. 20,
no. 1, pp. 168–179, 2015.
82. H. Ai, F. Wang, Q. Yang, F. Zhu, and C. Lei, “Preparation and biological activities of chitosan from the
larvae of housefly, Musca domestica,” Carbohydr. Polym., vol. 72, no. 3, pp. 419–423, 2008.
83. A. T. Paulino, J. I. Simionato, J. C. Garcia, and J. Nozaki, “Characterization of chitosan and chitin
produced from silkworm crysalides,” Carbohydr. Polym., vol. 64, no. 1, pp. 98–103, 2006.
84. C. Song, H. Yu, M. Zhang, Y. Yang, and G. Zhang, “Physicochemical properties and antioxidant activ-
ity of chitosan from the blowfly Chrysomya megacephala larvae,” Int. J. Biol. Macromol., vol. 60, pp.
347–354, 2013.
85. Waśko et al., “The first report of the physicochemical structure of chitin isolated from Hermetia illu-
cens.” Int. J. Biol. Macromol., vol. 92, pp. 316–320, 2016.
86. E. B. Ibitoye, I. H. Lokman, M. N. M. Hezmee, Y. M. Goh, A. B. Z. Zuki, and A. A. Jimoh, “Extraction
and physicochemical characterization of chitin and chitosan isolated from house cricket,” Biomed.
Mater., vol. 13, 025009, 2018.
Sources of Chitin and Chitosan and their Isolation 33
87. F. Di Mario, P. Rapanà, U. Tomati, and E. Galli, “Chitin and chitosan from Basidiomycetes,” Int. J.
Biol. Macromol., vol. 43, no. 1, pp. 8–12, 2008.
88. J. Synowiecki and N. A. Al Khateeb, “Production, properties, and some new applications of chitin
and its derivatives,” Crit. Rev. Food Sci. Nutr., vol. 43, no. 2, pp. 145–171, 2003.
89. A. M. Abdel‐Mohsen et al., “Novel chitin/chitosan‐glucan wound dressing: Isolation, charac-
terization, antibacterial activity and wound healing properties,” Int. J. Pharm., vol. 510, no. 1,
pp. 86–99, 2016.
90. Y. A. Skorik, A. V. Pestov, and Y. G. Yatluk, “Evaluation of various chitin‐glucan derivatives
from Aspergillus niger as transition metal adsorbents,” Bioresour. Technol., vol. 101, no. 6, pp. 1769–
1775, 2010.
91. M. Versali, F. Clerisse, J.‐M. Bruyere, and S. Gautier, “US7556946B2.pdf,” US7556946B2, 2003.
92. C. K. S. Pillai, W. Paul, and C. P. Sharma, “Chitin and chitosan polymers: Chemistry, solubility and
fiber formation,” Prog. Polym. Sci., vol. 34, no. 7, pp. 641–678, 2009.
93. P. Methacanon, M. Prasitsilp, T. Pothsree, and J. Pattaraarchachai, “Heterogeneous N‐deacetylation
of squid chitin in alkaline solution,” vol. 52, pp. 119–123, 2003.
94. M. Bajaj, J. Winter, and C. Gallert, “Effect of deproteination and deacetylation conditions on
viscosity of chitin and chitosan extracted from Crangon crangon shrimp waste,” Biochem. Eng. J.,
vol. 56, no. 1–2, pp. 51–62, 2011.
95. C. M. de Moura, J. M. de Moura, N. M. Soares, and L. A. de A. Pinto, “Evaluation of molar weight
and deacetylation degree of chitosan during chitin deacetylation reaction: Used to produce biofilm,”
Chem. Eng. Process. Process Intensif., vol. 50, no. 4, pp. 351–355, 2011.
96. C. H. Chen, F. Y. Wang, and Z. P. Ou, “Deacetylation of β‐chitin. I. Influence of the deacetylation
conditions,” J. Appl. Polym. Sci., vol. 93, no. 5, pp. 2416–2422, 2004.
97. S. V. Nemtsev, A. I. Gamzazade, S. V. Rogozhin, V. M. Bykova, and V. P. Bykov, “Deacetylation
of chitin under homogenous conditions,” Prikl. Biokhimiya i Mikrobiol., vol. 38, no. 6, pp.
609–615, 2002.
98. P. Broussignac, “Chitosan: A natural polymer not well known by the industry,” Chim. Ind. Genie
Chim., vol. 99, no. 9, p 1241–1247, 1968.
99. K. Kurita, K. Tomita, T. Tada, S. Ishii, S.‐I. Nishimura, K. Shimoda. “Squid chitin as a potential
alternative chitin source: Deacetylation behavior and characteristic properties,” J. Polym. Sci. A, vol.
21, no. 2, pp. 485–491, 1993.
100. C. Liu, G. Wang, W. Sui, L. An, and C. Si, “Preparation and characterization of chitosan by a novel
deacetylation approach using glycerol as green reaction solvent,” ACS Sustain. Chem. Eng., vol. 5,
no. 6, pp. 4690–4698, 2017.
101. T. H. D. Ngo and D. N. Ngo, “Effects of low‐frequency ultrasound on heterogenous deacetylation of
chitin,” Int. J. Biol. Macromol., vol. 104, no. B, pp. 1604–1610, 2016.
102. I. Younes et al., “Use of a fractional factorial design to study the effects of experimental factors on the
chitin deacetylation,” Int. J. Biol. Macromol., vol. 70, pp. 385–390, 2014.
103. B. Focher, P. L. Beltrame, A. Naggi, and G. Torri, “Alkaline N‐deacetylation of chitin enhanced by
flash treatments. Reaction kinetics and structure modifications,” Carbohydr. Polym., vol. 12, no. 4,
pp. 405–418, 1990.
104. Y. Zhao, R. D. Park, and R. A. A. Muzzarelli, “Chitin deacetylases: Properties and applications,” Mar.
Drugs, vol. 8, no. 1, pp. 24–46, 2010.
105. Y. Kim, Y. Zhao, K. Oh, V. Nguyen, and R. Park, “Enzymatic deacetylation of chitin by extracellular
chitin deacetylase from a newly screened Mortierella sp DY‐52,” J. Microbiol. Biotechnol., vol. 18,
no. 4, pp. 759–766, 2008.
106. D. K. Rane and G. D. Hoover, “Production of chitosan by fungi,” Food Microbiol., vol. 7, no. 1,
pp. 11–33, 1993.
107. K. M. Abdel‐Gawad, A. F. Hifney, M. A. Fawzy, and M. Gomaa, “Technology optimization of
chitosan production from Aspergillus niger biomass and its functional activities,” Food Hydrocoll.,
vol. 63, pp. 593–601, 2017.
34 Chitin and Chitosan: Properties and Applications
108. H. Miyoshi, K. Shimura, K. Watanabe, and K. Onodera, “Characterization of some fungal chitosans,”
Biosci. Biotechnol. Biochem., vol. 56, no. 12, pp. 1901–1905, 1992.
109. W. P. Wang, Y. M. Du, and X. Y. Wang, “Physical properties of fungal chitosan,” World J. Microbiol.
Biotechnol., vol. 24, no. 11, pp. 2717–2720, 2008.
110. S. A. White, P. R. Farina, and I. Fulton, “Production and isolation of chitosan from Mucor rouxii,”
Appl. Environ. Microbiol., vol. 38, no. 2, pp. 323–328, 1979.
111. S. Arcidiacono and D. L. Kaplan, “Molecular weight distribution of chitosan isolated from
Mucor rouxii under different culture and processing conditions,” Biotechnol. Bioeng., vol. 39,
pp. 281–286, 1992.
112. J. Synowiecki and N. A. A. Q. Al‐Khateeb, “Mycelia of Mucor rouxii as a source of chitin and
chitosan,” Food Chem., vol. 60, no. 4, pp. 605–610, 1997.
113. S. C. Tan, T. K. Tan, S. M. Wong, and E. Khor, “The chitosan yield of Zygomycetes at their optimum
harvesting time,” Carbohydr. Polym., vol. 30, pp. 239–242, 1996.
114. T. Wu, S. Zivanovic, F. A. Draughon, and C. E. Sams, “Chitin and chitosan—value‐added products
from mushroom waste,” J. Agric. Food Chem., vol. 52, no. 26, pp. 7905–7910, 2004.
115. T. Kleekayai and W. Suntornsuk, “Production and characterization of chitosan obtained from
Rhizopus oryzae grown on potato chip processing waste,” World J. Microbiol. Biotechnol., vol. 27,
no. 5, pp. 1145–1154, 2011.
116. P. Pochanavanich and W. Suntornsuk, “Fungal chitosan production and its characterization,” Lett.
Appl. Microbiol., vol. 35, pp. 17–21, 2002.
117. A. Zamani, L. Edebo, B. Sjostrom, and M. J. Taherzadeh, “Extraction and precipitation of chitosan
from fungal cell wall by dilute sulfuric acid,” Biomacromolecules, vol. 8, no. 12, pp. 3786–3790,
2007.
118. T. Wu, “Production and Characterization of Fungal Chitin and Chitosan,” Grad. Sch., 2004.
119. D. Knorr, M. D. Beaumont, and Y. Pandya, Potential of acid soluble and water soluble chitosan in
biotechnology. In Chitin and chitosan. London, New York: Elsevier Applied Science, 1989.
120. W. L. Teng, E. Khor, T. K. Tan, L. Y. Lim, and S. C. Tan, “Concurrent production of chitin from
shrimp shells and fungi,” Carbohydr. Res., vol. 332, no. 3, pp. 305–316, 2001.
121. P. B. Nahar, V. Ghormade, and M. V. Deshpande, “The extracellular constitutive production of chitin
deacetylase in Metarhizium anisopliae: Possible edge to entomopathogenic fungi in the bio‐control
of insect pest,” J. Invertebr. Pathol., vol. 85, pp. 80–88, 2004.
122. C. Lopes, L. T. Antelo, A. Franco‐Uria, A. A. Alonso, and R. Pérez‐Martin, “Chitin production from
crustacean biomass: Sustainability assessment of chemical and enzymatic processes,” J. Clean.
Prod., vol. 172, pp. 4140–4151, 2018.
123. D. Gómez‐Ríos, R. Barrera‐Zapata, and R. Ríos‐Estepa, “Comparison of process technologies
for chitosan production from shrimp shell waste: A techno‐economic approach using Aspen Plus®,”
Food Bioprod. Process., vol. 103, pp. 49–57, 2017.
124. G. L. Dotto and L. A. Pinto, Frontiers in Biomaterials: Chitosan Based Materials and its Applications,
Bentham Science publishers, Sharjah, 2017.
125. M. Kannan, M. Nesakumari, and K. Rajarathinam, “Production and characterization of mushroom
chitosan under solid‐state fermentation conditions,” Adv. Biol. Res., vol. 4, no. 1, pp. 10–13, 2010.
2
Methods of Isolating Chitin
from Sponges (Porifera)
Sonia Ż ółtowska1,2, Christine Klinger4, Iaroslav Petrenko3,
Marcin Wysokowski1,2, Yvonne Joseph2, Teofil Jesionowski1,
and Hermann Ehrlich2
1
Institute of Chemical Technology and Engineering, Faculty of Chemical Technology,
Poznan University of Technology, Poznan, Poland
2
Institute of Electronics and Sensor Materials, TU Bergakademie‐Freiberg, Freiberg, Germany
3
Institute of Experimental Physics, TU Bergakademie‐Freiberg, Freiberg, Germany
4
Institute of Physical Chemistry, TU Bergakademie‐Freiberg, Freiberg, Germany
2.1 Introduction
Chitin is known to be one of the most abundant biopolymers in nature. It occurs in vari-
ous contexts across a broad range of species, including fungi, yeast, marine protists,
foraminifera [1, 2], diatoms [3, 4], hydroids, coelenterates, brachiopods, polychaetes,
Pogonophora [5], molluscs [6, 9], and crustaceans [10–15], and even in the epidermal
cuticles of fish [16] and the scales of salmon [17]. Interestingly, the existence of chitin in
sponges (phylum Porifera) was poorly studied until 2007. Previously, it was stated that
the presence of chitin in several sponges might be related to ‘contamination’ by a variety
of chitin‐containing micro‐invertebrates harboured by the sponges [18], such as poly-
chaete worms (Haplosyllis basticola) colonising the Ianthella basta demosponge. Later,
the presence of chitin in sponges was considered to be limited only to the gemmules of
freshwater Spongillidae sponges. However, in 2007, for the first time, a team led by
Hermann Ehrlich described the discovery of chitin in the skeletal fibres of the demos-
ponge Verongula gigantea. They showed that chitin isolated from V. gigantea is very
similar in structure to the α‐chitin found in other invertebrates, including arthropods [19].
The authors suggested that the system of chitin production might be an ancestral feature
in Metazoa. This hypothesis indicates that the synthesis of chitin may have evolved in the
Precambrian, much earlier than previously described. In the same year, Ehrlich et al. [20]
published the first report of the presence of chitin in endogenous material within the
skeletal frameworks of the glass sponge Farrea occa. They put forward the hypothesis
that chitin molecules are part of a very old organic template system involved in the
biosilicification process, which was established a long time before the origin of glass
sponges [21].
In 2009, Ehrlich’s team continued systematic studies on the isolation of chitin from
diverse demosponges of the order Verongiida. Their results indicate that the obtained chi-
tin scaffolds closely resemble the shape and morphology of native sponge skeleton for all
tested species [22–24]. The presence of chitin in Mexican Pacific species of the genus
Aplysina and in the sponge Aplysina fistularis was confirmed in 2012 [25] and 2013 [26],
respectively. In 2013, an important discovery was reported by Ehrlich’s team, who
described, for the first time, the isolation of chitin from skeletons of the freshwater dem-
osponges Lubomirskia baicalensis [27] and Spongilla lacustris [28]. The authors sug-
gested that the incorporation of chitin into the proteinaceous skeletons of S. lacustris
appears to be selectively favoured, because the resulting material becomes more rigid.
They also suggested that chitin identified in the holdfast of L. baicalensis plays an impor-
tant role in the adhesion of sponges to the rocky substrate. The presence of chitin in the
heavily mineralised skeleton of the Verongiida demosponge Suberea clavata was con-
firmed in 2017 [29]. In that study, the authors proved that chitin can be an organic tem-
plate for both magnesium‐bearing calcite and amorphous silica, which were shown to be
intercalated in a chitinous matrix. These results imply that chitin is involved in the phe-
nomenon of multiphase biomineralization in Verongiida demosponges (see [21] for
details). Recently, a 3D fibrous chitin scaffold was isolated from another representative of
the order Verongiida, Pseudoceratina purpurea [30]. Intriguingly, in 2018, chitin has also
been discovered in non‐Verongiida marine demosponges from the family Poecilosclerida:
Acarnus wolffgangi, Echinoclathria gibbosa [31], and Mycale euplectellioides [32]. In
contrast to the Verongiida, these demosponges have siliceous spicules in their chitinous
skeletons.
Considering the evolutionary aspect, chitin is hypothesised to have been a major organic
component of prehistoric invertebrates. Interestingly, the oldest chitin found in nature was
also discovered in a fossil of sponge origin. Ehrlich and his co‐authors described the pres-
ence of chitin in 505‐million‐year‐old (Middle Cambrian) Vauxia gracilenta sponge fos-
sils [33]. To date, the chitin found in this sponge is the oldest known chitin, which is
Methods of Isolating Chitin from Sponges (Porifera) 37
Sycon ciliatum
Marine species
Spongilla lacustris
Calcarea Lubomirskia baicalensis
Freshwater species
1 cm
(c) (d)
on
ti
ac
C a p ill a r y
1 cm 10 μm
Figure 2.2 From sponge to chitinous scaffold. Demineralisation of dried Ianthella sp. demosponge (a)
leads to a flat 3D‐structured skeleton (b). Treatment of this skeleton with 5% (w/v) NaOH at 37°C leads
to the isolation of a translucent, pigment‐free chitinous scaffold (c) that resembles the square‐like mesh
architecture of the skeleton. Such meshes (d) are made of tube‐like fibres with diameter around 50 μm
(SEM image e) and exhibit capillary action (arrows) when placed in liquids. Due to this capillary action,
the whole construct can swell and become well visible even in air (c).
matter [59–64]. Demineralisation can be achieved using various acids, most commonly
diluted hydrochloric acid, acetic acid or sulphuric acid (see [57, 65, 66] for examples). To
decompose all of the inorganic salts, the acid intake should be greater than the stoichiometric
quantity of minerals [67, 68]. Due to the heterogeneity of the solid matrix and difficulties
in removing calcium debris, larger volumes or more concentrated acid solutions are
commonly used [56, 65]. Longer demineralisation times – even up to several days – may
cause degradation of the polymer matrix [69]. It has also been reported that the use of high
temperature for demineralisation stimulates the reaction by promoting the penetration of
the solvent into the chitin matrix. Therefore, some attempts have been made to carry out
demineralisation at higher temperatures [70]. It has further been shown that the penetration
of solvent into the chitin matrix is strongly dependent on the particle size. Thus, Marquis‐
Duval reported that the decisive factor in demineralisation is related to the contact area
between the chitin matrix and the solvent [71]. However, it has been shown that high
temperatures, longer incubation times and high acid concentrations affect the final
properties of the isolated chitin. Recently, Kaya et al. proposed to use sodium hypochlorite
(NaClO) pre‐treatment prior to traditional methods of acid‐based demineralisation [72], a
method that appears to be promising in terms of time and energy savings. Also,
ethylenediaminetetraacetic acid (EDTA) is recognised as a useful reagent for the
decalcification of chitin‐based crustacean shells [73].
Industrial deproteinisation is carried out using a base solution such as NaOH or KOH
(see [74, 75] for review). The effectiveness of deproteinisation depends on the ratio of shell
to solution, the temperature and duration of the process, and the concentration of the base.
Industrial methods also include a decolourisation step, which improves the colour of chitin.
For this purpose, hydrogen peroxide or sodium hypochlorite solutions are used [57]. In the
case of fungi which contain no mineral phases within their skeletal structures, only
deproteinisation is necessary [76]. For example, chitin from the cultivated mushroom
Ganoderma lucidum was obtained when the biomass was sonicated and washed with
ethanol, deproteinised with 4 M NaOH at 100°C for 2 h, and then bleached using potassium
permanganate and oxalic acid [41].
However, deproteinisation and demineralisation can also be carried out using
microorganisms and corresponding enzymatic treatment. A comparative study of chemical
and biological methods of chitin extraction was carried out by Khanafari et al. [77]. They
showed that biological treatment produced better results than the chemical method, because
it preserved the structure of chitin. Similarly, a study by Bustos and Healy [78] demonstrated
that chitin obtained by the deproteinisation of shrimp shells with various proteolytic
microorganisms has higher molecular weights than chemically prepared shellfish chitin.
The biological extraction of chitin offers high reproducibility in a shorter time, simpler
manipulation, lower solvent consumption and lower energy input. Diverse alternative
procedures to the chemical extraction of chitin have recently been reported, using such
bacteria as Bacillus subtilis and Lactobacillus plantarum [79], lactic acid fermentation
with Pseudomonas aeruginosa [80] and L. plantarum DSM 20174 [81], Bacillus mojavensis
A21 and Balistes capriscus [82], B. subtilis A26, B. mojavensis A21, Bacillus pumilus A1,
Bacillus amyloliquefaciens An6, Bacillus licheniformis NH1, Bacillus cereus BG1 [83],
Streptomyces griseus [84], Bacillus invictae [85] and Bacillus safensis S406 [86] proteases.
Interestingly, chitin from Portunus segnis and Penaeus kerathurus shells has been isolated
by means of crude digestive alkaline proteases from the viscera of P. segnis [87]. There is
40 Chitin and Chitosan: Properties and Applications
Shell
Chemical Washing Washing Biological
treatment Grinding Drying treatment
Crushing
Demineralisation Penetrated shell waste
HCl Organic acid
(lactic acid)-producing bacteria
Minerals
Minerals
(CaCO3, Ca3(PO4)2)
(CaCO3, Ca3(PO4)2)
Deproteinisation Demineralised shell waste
Base Protease-producing bacteria
Proteins Proteins
Pigments
Bleaching and decolouration Deproteinised shell waste
H2O2 Additional chemical
deproteinization
Pigments Proteins
Chitin
Deacetylation
(NaOH/KOH 30–50%)
80–150 °C
Chitosan
Figure 2.3 Industrial methods of chitin extraction from crustacean shells.
even a report of pilot‐scale chitin extraction from shrimp shell waste with bacterial enrich-
ment cultures [88].
A schematic overview of chemical and biological methods of chitin isolation is pre-
sented in Figure 2.3. Owing to differences in the ultrastructure of the initial material, all
such treatments must be adapted to the specific chitin source, to produce high‐quality chi-
tin, and then chitosan.
24 h, 25°C
15 min, 25°C
48 h, 25°C
14 DAYS, 37°C
V. Alkali etching using
2.5 M NaOH solution, CHITIN MATRIX
without shaking
Figure 2.4 Schematic representation of the slow‐etching approach for chitin isolation from glass sponges.
200 μm
Figure 2.5 Wide field fluorescence microscopy image providing strong evidence for the presence of
chitin (in blue) after Calcofluor‐white staining of an alkali‐resistant siliceous skeleton of Farrea occa glass
sponge.
their biology and the nature of the organic matrix forming their skeleton. The discovery of
chitin in the skeleton of glass sponges suggests that it is silica–chitin composites that play
an important role as a template for skeleton formation, rather than silica–protein scaffolds.
This leads to the hypothesis that chitin molecules are involved in a very old biomineralisation
motif, which was established a long time before the origin of Hexactinellida sponges and
collagen as a structural protein. Therefore, the identification of chitin as a main component
of axial filaments is also especially important from the point of view of evolution and
systematics [106]. It should be noted that the NaOH concentration commonly used for
chitin isolation (2.5 M corresponds to 4% v/v) is well below the critical concentration of
25–30% v/v at which the transformation of β‐chitin into α‐chitin starts to take place [107].
Therefore, the crystal structure of the chitin‐based scaffolds is not influenced by the
extraction procedure, which means that purified chitin scaffolds are obtained without
pigments, proteins, lipids and silica residues and without changing the morphology and
chemistry of the organic matrix [22, 23].
Decalcification Desilicification
Figure 2.6 Scheme of the standard method for chitin isolation from demosponges.
The second step is the decalcification of calcium carbonates, achieved by applying 20%
(v/v) acetic acid for 24 hours at 37°C. In addition to the degradation of calcium carbonates,
further pigments and proteins are dissolved. The decalcification of biominerals was
described in detail by Ehrlich’s team in 2009 [112]. The following equation describes the
reaction of degradation using acetic acid:
After the treatment, the sample has to be neutralised by rinsing with deionised water.
Then the third step, desilicification, can be carried out. As previously noted, beside the
crystalline calcium carbonate, the appearance of amorphous silica in Verongiida sponges
has been detected [113]. For the degradation of this silica, 2.5 M NaOH is applied for
24 hours at 37°C. The alkaline extraction reaction can be described as follows:
As the reaction equation shows, the silica is dissolved by the alkali to form silicic acid.
Furthermore, other alkali‐soluble proteins, lipids and pigments are dissolved. After the
alkaline extraction, the sponge sample is neutralised by rinsing with deionised water. The
alternating treatment, using low‐concentration acetic acid and alkaline solution, is repeated
until the chitin matrix is completely cleaned, demineralised and nearly transparent. Then
the final step is the application of 35% (v/v) hydrogen peroxide to remove the remaining
pigments from the chitin matrix. In the chitin isolation process, the number of alternating
treatments depends on the morphology of the treated sponge species. For example, some
determining factors are the degree of mineralisation and the type of minerals which are
present. Thus, treatment times of up to 4 days are reported [29].
According to the latest findings on chitin isolation from the demosponges Suberea clav-
ata, Pseudoceratina purpurea, Acarnus wolffgangi, Echinoclathria gibbosa and Mycale
euplectellioides, where silica residues as spicules or other incorporated silica particles were
not completely degraded by alkaline extraction, a further step is necessary [29–32]. As is
established for the less mild demineralisation of glass sponges, desilicification is performed
Methods of Isolating Chitin from Sponges (Porifera) 45
(a) (b)
100 μm 100 μm
Figure 2.7 The skeleton of Mycale euplectellioides before (a) and after (b) HF treatment. The siliceous
spicules present in the chitinous skeleton of M. euplectellioides are marked with arrows.
applying low‐concentration (up to 10% (v/v)) hydrofluoric acid for 6–12 hours at room
temperature (Figure 2.7).
optimum temperature was found to be 70°C. Depending on the morphology of the sponges
studied, there is an appreciable difference in the time of treatment and the chemicals used.
For chitin isolation from V. rigida and A. archeri, treatment with acetic acid while apply-
ing ultrasound is essential to obtain a completely demineralised and transparent chitin
matrix (Figure 2.8).
For chitin isolation from A. fistularis, ultrasound‐assisted treatment with alkaline solu-
tion for 1.5 hours is sufficient for complete demineralisation and deproteinisation. Further
acidic extraction applying ultrasound causes damage to the typical chitin fibres. The dam-
age is obvious when the chitin skeleton is viewed with the naked eye, as shown in the fol-
lowing photographs (Figure 2.9).
Altogether, the treatment time is reduced to a maximum of 4 hours and the quantity of
applied chemicals is significantly reduced when the standard method for chitin isolation is
combined with ultrasound. Given the variety of chitin deacetylation methods using ultra-
sound‐assisted alkaline treatment, verification of the obtained chitin is necessary.
(a) (b)
200 μm 200 μm
Figure 2.8 Skeleton isolated from Aplysina archeri marine demosponge (a) with the standard method
and (b) with the ultrasound‐assisted method. The direct comparison shows the ‘defibrillation’ of the chitin
fibres by ultrasound.
(a) (b)
1 cm 1 cm
Figure 2.9 The skeleton isolated from Aplysina fistularis (a) after the first isolation step applying 2.5 M
NaOH and after treatment with 20% (v/v) acetic acid (b). The uncompleted demineralisation at the inter-
mediate stage is visible because of the presence of pigmentation.
Methods of Isolating Chitin from Sponges (Porifera) 47
(a) (b)
1 cm 1 cm
Figure 2.10 (a) Cell‐free but pigmented skeleton of Aplysina aerophoba and (b) the 3D chitinous scaffold
obtained from the same skeleton after complete alkaline and acidic extraction assisted by m icrowaves
(750 W).
(a) (b)
200 μm 10 μm
Figure 2.11 Deformed chitin fibres isolated by the microwave‐assisted method from the skeleton of
Aplysina aerophoba sponge, under different magnifications.
As regards the power applied, it was shown that a power of 400 W provides a milder
procedure in terms of the destructive effect on the structure. As the SEM images show,
microwave‐assisted chitin isolation promotes the breakdown of the characteristic multi‐
layered structure known for A. aerophoba. The isolated chitin scaffolds (Figure 2.11)
promote the typical chitin fibres and flattened chitin layers, which leads to surface
enlargement. Considering the advantages of large surfaces, there arise new potential fields
of application. Despite the breakdown of a great part of the chitin fibres, the isolated matrix
is still capable of liquid adsorption.
Methods of Isolating Chitin from Sponges (Porifera) 49
During the development of the microwave‐assisted method for chitin isolation, tem-
perature measurements were also made. Superheating led to temperatures of up to
118°C. Due to the reported deacetylation of chitin to chitosan at high temperatures, it
was necessary to analyse the chitin using Raman and FTIR spectra, chitinase digestion
and Calcofluor‐white staining. The spectra of the microwave‐assisted isolated chitin
were found to be in agreement with the spectra of chitin isolated using the standard
method. In comparison with a chitosan standard, significant differences were present in
the spectra. Therefore, no deacetylation to chitosan takes place on exposure to micro-
waves during alkaline and acidic extraction. Overall, the microwave‐assisted method
enables chitin isolation in 99.3% of the treatment time of the standard method.
Furthermore, it reduces the quantity of solvents required and offers a new shape of iso-
lated chitin fibres.
20 μm 2 μm
Figure 2.12 SEM images of naturally prefabricated 3D chitin–GeO2 composite under different
magnifications.
In 2015, applying extreme biomimetic conditions (pH 1.5, 80°C), a monolithic silica–
chitin composite was obtained using chitin from A. cauliformis [44]. It was shown that
silica was deposited exclusively in the inside of the tube‐like chitin. Due to the excellent
biocompatibility of both chitin and silica, this is a promising material in the field of tissue
engineering. Furthermore, in the same year, the synthesis of a unique 3D germanium
oxide–chitin composite was reported [47]. The following images (Figure 2.12) show the
germanium oxide nanocrystals on the surface of a fibre of chitin isolated from A. cauli-
formis, viewed by means of scattering electron microscopy with 2000‐ and 6000‐fold
magnification.
After soaking in a solution of germanium tetraethoxide (TEOG), used as the precursor
of germanium oxide, the chitin scaffold was hydrothermally treated at 120°C and pH 14.
The resulting new 3D GeO2–chitin composite exhibited interesting fluorescent and lumi-
nescent properties [47].
The hydrothermal synthesis of a chitin–hematite composite at pH 1.5 and 90°C for 48 hours
is another prime example of the application of sponge chitin in Extreme Biomimetics.
Wysokowski and his team [49, 50] were able to demonstrate the function of chitin as a
carrier and the nucleation of hematite crystals at the surface of chitin nanofibres. This
material was proposed as a promising composite for research on sensors and biocatalysts.
The second of the aforementioned methods for transforming chitin scaffolds of Poriferan
origin into new biocomposites is solvothermal synthesis [52]. The chitin scaffold is treated
together with the reactant at high temperature. This method has been used for the deposi-
tion of polyhedral oligomeric silsesquioxanes (POSS) on the chitin skeleton isolated from
A. cauliformis [46] (Figure 2.13). The deposition increased the hydrophobicity and oleo-
philicity of the material.
Another relatively new approach for the creation of novel bioinspired materials is
bioelectrometallurgy [130]. Because neither the isolated sponge skeleton nor the isolated
chitin scaffolds are electro‐conductive, pre‐treatment of the biopolymers with silver nitrate
and ammonium iron (II) sulphate is an indispensable step for bioelectrometallurgy. The
latter substance reduces the quantity of silver ions deposited on the cell‐free skeleton. The
resulting electro‐conductive matrix is applied as a cathode in a copper sulphate solution.
The electrochemical reaction causes the deposition of copper on the surface of this chitin‐
based cathode [131]. In Figure 2.14, a metallised chitin scaffold isolated from the I. basta
Methods of Isolating Chitin from Sponges (Porifera) 51
(a)
5 mm
(b) (c)
20 μm 2 μm
Figure 2.13 Chitinous scaffold isolated from the demosponge Aplysina cauliformis (a and SEM image b)
was used as a template for the solvothermal synthesis of POSS microcrystals in chloroform at 100°C (SEM
image c).
demosponge (see Figure 2.2) is shown. The large amount of copper and copper oxide
deposited on the chitin scaffold can be clearly seen without further tools. Using scattering
electron microscopy, the complete metallisation of the chitin scaffold is also verified on a
micrometric scale.
(a) (c)
1 mm
(b)
200 μm 1 μm
Figure 2.14 The electrochemical reduction of copper leads to a chitin scaffold completely metallised
with Cu/Cu2O (stereo microscopy images a and b). Nanocrystals of the metal phases (SEM image c)
become tightly covered with nanofibres of sponge chitin and seem to be intercalated within this organic
construct. These investigations resulted in an electro‐conductive material with a number of possible
applications – for example, as a catalyst.
scaffolds for tissue engineering, but these scaffolds have to meet various standards relating
to biocompatibility, biodegradability, appropriate mechanical properties compatible with
the tissue where they will be applied, as well as appropriate architecture. Scaffolds should
possess an interconnected pore structure with high porosity to ensure cellular penetration
and adequate diffusion of nutrients to cells within the construct and to the extra‐cellular
matrix formed by those cells. Another key feature is the pore size of the scaffolds and the
presence of ligands on their surface. The pores should be large enough to allow cells to
migrate into the structure, where they eventually become bound to the ligands within the
scaffold, but also small enough to establish a sufficiently high specific surface area, leading
to a minimal ligand density to allow efficient binding of cells to the scaffold. Other
important factors include the cost of manufacture and the availability of the selected
carriers. Typically, three different groups of biomaterials are used in the fabrication of
scaffolds for tissue engineering: inorganic materials, synthetic polymers and natural poly-
mers [132, 133] (Figure 2.15).
Each of these groups has specific advantages and disadvantages. Therefore, great atten-
tion is paid to the modification of existing carriers as well as the search for and production
of new types of carriers [132].
Particular attention is paid to sponges of the order Verongiida, whose 3D chitinous skel-
etons meet the standards for fabrication of tissue engineering scaffolds [38]. A first attempt
to use 3D chitin scaffolds from Aplysina cavernicola, A. cauliformis, A. fulva and
Aiolochroia crassa as templates for chondrocyte culture in an in vivo environment was
reported by Ehrlich et al. in 2010. The results show that the deposition of proteoglycan‐rich
extracellular matrix and the presence of cartilage‐specific collagen type II on the chitinous
Methods of Isolating Chitin from Sponges (Porifera) 53
Collagen
Gelatin
Proteins Fibrin Hyaluronic acid
Silk fibroin Alginate
Natural polymers Polysaccharides Chitin
Chitosan
Natural composites Chondroitin sulphate
Poly(hydroxyacid)s
Scaffolds of Aliphatic polyesters
Synthetic polymers Hydrogels
tissue engineering Polyurethanes
Polyphosphazenes
Chitin scaffold
Inorganic materials Calcium phosphate
Figure 2.15 Categorisation of scaffolds for tissue engineering based on their source.
(a) (b)
100 μm
100 μm
Figure 2.16 Chitinous scaffold from Ianthella basta as carrier after 2 days of incubation with hMSCs (a).
The distribution of hMSCs within the pores of the chitin carrier is well visible even on day 21 (b).
2.5 Outlook
The newly established methods for the isolation of chitin from sponge skeletons, using
ultrasound or microwaves during alkaline and acidic extraction, offer significant economic
advantages. Even including the pre‐treatment step for extraction of the pharmaceutically
interesting bromotyrosines, which takes one additional day, the ultrasound‐assisted method
of chitin isolation from sponges requires not more than a third of the treatment time of the
standard method. Furthermore, it is possible to reduce the quantities of alkaline solution
and acetic acid used by at least 40%. For A. fistularis, it was possible to isolate pure chitin
without the application of acetic acid; hence, in this case, it is possible to eliminate the
acidic extraction completely when ultrasound is applied. The microwave‐assisted method
of isolation of chitin from sponges also enables a time saving of up to a third of the treatment
time of the standard method, as well as a reduction in the quantity of chemicals used.
These findings open the door to new methods for an industrial approach to chitin
isolation. It is not without significance that marine sponges can be easily farmed in marine
ranching conditions, providing a renewable source of 3D, naturally prefabricated chitin.
Consequently, these methods offer a wide range of opportunities to integrate sponges as a
renewable source for pharmacy and tissue engineering on an industrial scale. Nevertheless,
the fundamental question of the possibility of applying such methods to other chitin sources
remains open. There is a lack of knowledge concerning the mechanism of chitin isolation
using ultrasound or microwaves; such knowledge may help establish the optimum
conditions for the isolation process and avoid the destruction of 3D chitin scaffolds.
Additionally, no research has been done on the possibility of isolating chitin from non‐
Verongiida sponges using treatments of this kind. The potential use of ultrasound and
microwaves in chitin isolation from spiculated sponges also remains unexplored. It seems
that now is the time to develop new and less time‐consuming methods of isolation of both
chitin and secondary metabolites from sponges, to realise the full potential of these
promising marine invertebrates.
Acknowledgment
This work was partially supported by DFG Project HE 394/3‐2 (Germany) and PUT
Research Grant no. 03/32/DSMK/0810 (Poland). M.W. is grateful for financial support
from the Foundation for Polish Science – START 097.2017, and S.Ż .A. for support from
the DAAD and Erasmus Plus programmes.
References
1. H.J. Blumenthal, S. Roseman, J. Bacteriol. 1957, 74, 222–224.
2. M.V. Elorza, H. Rico, R. Sentandreu, J. Gen. Microbiol. 1983, 129, 1577–1582.
3. C.A. Durkin, T. Mock, E.V. Armbrust, Eukaryot. Cell 2009, 8, 1038–1050.
Methods of Isolating Chitin from Sponges (Porifera) 55
4. E. Brunner, P. Richthammer, H. Ehrlich, S. Paasch, P. Simon, S. Ueberlein, K.H. Van Pée, Angew.
Chemie – Int. Ed. 2009, 48, 9724–9727.
5. P.C.J. Brunet, D.B. Carlisle, Nature 1958, 182, 1689.
6. G. Tóth, L. Zechmeister, Nature 1939, 144, 1049.
7. G. Goffinet, C. Jenuiaux, Cah. Biol. Mar. 1979, 20, 341–349.
8. W. Peters, Comp. Biochem. Physiol. Part B Biochem. 1972, 541–544.
9. I.M. Weiss, V. Schönitzer, J. Struct. Biol. 2006, 153, 264–277.
10. S. Liu, J. Sun, L. Yu, C. Zhang, J. Bi, F. Zhu, M. Qu, C. Jiang, Q. Yang, Molecules 2012, 17,
4604–4611.
11. H. Merzendorfer, J. Comp. Physiol. B Biochem. Syst. Environ. Physiol. 2006, 176, 1–15.
12. M.M. Giraud‐Guille, Tissue Cell 1984, 16, 75–92.
13. M.M. Giraud‐Guille, H. Chanzy, R. Vuong, J. Struct. Biol. 1990, 103, 232–240.
14. O.M. Nieto‐Akosta, R.D. Enriques Rodriguez, G.A. Vitivskaya, Appl. Biochem. Microbiol. 1978, 13,
604–607.
15. H.O. Fabritius, C. Sachs, P.R. Triguero, D. Raabe, Adv. Mater. 2009, 21, 391–400.
16. G.P. Wagner, J. Lo, R. Laine, M. Almeder, Experientia 1993, 49, 317–319.
17. J.W. Tang, J. Fernandez, J.J. Sohn, C.T. Amemiya, Curr. Biol. 2015, 25, 897–900.
18. P. Dauby, C. Jeuniaux, Cah. Biol. Mar. 1986, 28, 121–129.
19. H. Ehrlich, M. Malando, K.D. Spindler, C. Eckert, T. Hanke, R. Born, C. Goebel, P. Simon, S.
Heinemann, H. Worch, J. Exp. Zool. B Mol. Dev. Evol. 2007, 308B, 347–356.
20. H. Ehrlich, M. Krautter, T. Hanke, P. Simon, C. Knieb, S. Heinemann, H. Worch, J. Exp. Zool. B Mol.
Dev. Evol. 2007, 308B, 473–483.
21. H. Ehrlich, Int. Geo. Rev. 2010, 52, 661–699.
22. E. Brunner, H. Ehrlich, P. Schupp, R. Hedrich, S. Hunoldt, M. Kammer, S. Machill, S. Paasch, V.V.
Bazhenov, D.V. Kurek, T. Arnold, S. Brockmann, M. Ruhnow, R. Born, J. Struct. Biol. 2009, 168,
539–547.
23. H. Ehrlich, M. Ilan, M. Maldonado, G. Muricy, G. Bavestrello, Z. Kljajic, J.L. Carballo, S. Schiaparelli,
A. Ereskovsky, P. Schupp, R. Born, H. Worch, V.V. Bazhenov, D. Kurek, V. Varlamov, D. Vyalikh, K.
Kummer, V.V. Sivkov, S.L. Molodtsov, H. Meissner, G. Richter, E. Steck, S. Hunoldt, M. Kammer, S.
Paasch, V. Krasokhin, G. Patzke, E. Brunner, Int. J. Biol. Macromol. 2010, 47, 132–140.
24. H. Ehrlich, E. Steck, M. Ilan, M. Maldonado, G. Muricy, G. Bavestrello, Z. Kljajic, J.L. Carballo, S.
Schiaparelli, A. Ereskovsky, P. Schupp, R. Born, H. Worch, V.V. Bazhenov, D. Kurek, V. Varlamov, D.
Vyalikh, K. Kummer, V.V. Sivkov, S.L. Molodtsov, H. Meissner, G. Richter, S. Hunoldt, M. Kammer,
S. Paasch, V. Krasokhin, G. Patzke, E. Brunner, W. Richter, Int. J. Biol. Macromol. 2010, 47,
141–145.
25. J.A. Cruz‐Barraza, J.L. Carballo, A. Rocha‐Olivares, H. Ehrlich, M. Hog, PLoS One 2012, 7, e42049.
26. M. Wysokowski, V.V. Bazhenov, M.V. Tsurkan, R. Galli, A.L. Stelling, H. Stöcker, S. Kaiser, E.
Niederschlag, G. Gärtner, T. Behm, M. Ilan, A.Y. Petrenko, T. Jesionowski, H. Ehrlich, Int. J. Biol.
Macromol. 2013, 62, 94–100.
27. H. Ehrlich, O.V. Kaluzhnaya, M.V. Tsurkan, A. Ereskovsky, K.R. Tabachnick, M. Ilan, A. Stelling, R.
Galli, O.V. Petrova, S.V. Nekipelov, V.N. Sivkov, D. Vyalikh, R. Born, T. Behm, A. Ehrlich, L.I.
Chernogor, S. Belikov, D. Janussen, V.V. Bazhenov, G. Wörheide, Proc. R. Soc. B. 2013, 280,
20130339.
28. H. Ehrlich, O.V. Kaluzhnaya, E. Brunner, M.V. Tsurkan, A. Ereskovsky, M. Ilan, K. R.
Tabachnick, V.V. Bazhenov, S. Paasch, M. Kammer, R. Born, A. Stelling, R. Galli, S. Belikov, O.V.
Petrova, V.V. Sivkov, D. Vyalikh, S. Hunoldt, G. Wörheide, J. Struct. Biol. 2013, 183, 474–483.
29. H. Ehrlich, V.V. Bazhenov, C. Debitus, N. de Voogd, R. Galli, M.V. Tsurkan, M. Wysokowski, H.
Meissner, E. Bulut, M. Kaya, T. Jesionowski, Int. J. Biol. Macromol. 2017, 104, 1706–1712.
30. S. Ż ółtowska‐Aksamitowska, M.V. Tsurkan, S.C. Lim, H. Meissner, K. Tabachnick, L.A. Shaala, D.T.A.
Youssef, V.N. Ivanenko, I. Petrenko, M. Wysokowski, N. Bechmann, Y. Joseph, T. Jesionowski, H.
Ehrlich, Int. J. Biol. Macromol. 2018, 112, 1021–1028.
56 Chitin and Chitosan: Properties and Applications
Pertenko, Z. Kljajić, T. Leisegang, S.L. Molodtsov, D.C. Meyer, T. Jesionowski, H. Ehrlich, RSC Adv.
2014, 4, 61743–61752.
50. M. Wysokowski, I. Petrenko, M. Motylenko, E. Langer, V.V. Bazhenov, R. Galli, A.L. Stelling, Z.
Kljajić, T. Szatkowski, V.Z. Kutsova, D. Stawski, T. Jesionowski, Bioinspired Mater. 2015, 1, 12–22.
51. M. Wysokowski, I. Petrenko, A.L. Stelling, D. Stawski, T. Jesionowski, H. Ehrlich, Polymers 2015, 7,
235–265.
52. M. Wysokowski, T.J. Szalaty, T. Jesionowski, M. Motylenko, D. Rafaja, I. Koltsov, H. Stöcker, V.V.
Bazhenov, H. Ehrlich, A.L. Stelling, J. Beyer, J. Heitmann, S. Petovic, M. Đurovic ́, Mater. Chem. Phys.
2017, 188, 115–124.
53. P.R. Austin, C.J. Brine, J.E. Castle, P. Zikakis, Science 1981, 212, 749–753.
54. S. Islam, M.A.R. Bhuiyan, M.N. Islam, J. Polym. Environ. 2017, 25, 854–866.
55. T. Philibert, B.H. Lee, N. Fabien, Appl. Biochem. Biotechnol. 2017, 181, 1314–1137.
56. S. Kaur, G.S. Dhillon, Crit. Rev. Biotechnol. 2015, 35, 44–61.
57. I. Younes, M. Rinaudo, Mar. Drugs 2015, 13, 1133–1174.
58. T.B. Cahú, S.D. Santos, A. Mendes, C.R. Córdula, S.F. Chavante, L.B. Carvalho, H.B. Nader, R.S.
Bezerra, Process Biochem. 2012, 47, 570–577.
59. R.H. Hackman, Aust. J. Biol. Sci. 1962, 15, 526–537.
60. C.J. Brine, P.R. Austin, Comp. Biochem. Physiol. Part B Biochem. 1981, 69, 283–286.
61. M. Hayes, B. Carney, J. Slater, W. Brück, Biotechnol. J. 2008, 3, 878–889.
62. H. Rasti, K. Parivar, J. Baharara, M. Iranshahi, F. Namvar, IranJ. Pharm. Res. 2017, 16, 366–379.
63. E. Bulut, I. Sargin, O. Arslan, M. Odabasi, B. Akyuz, M. Kaya, Mater. Sci. Eng. C 2017, 70, 552–563.
64. E.B. Ibitoye, I.H. Lokman, M.N.M. Hezmee, Y.M. Goh, A.B.Z. Zuki, A.A. Jimoh, Bioact. Mater. 2018,
13, 025009.
65. W. Arbia, L. Arbia, L. Adour, A. Amrane, Food Technol. Biotechnol. 2013, 51, 12–25.
66. M. Kaya, M. Karaarslan, T. Baran, E. Can, G. Ekemen, B. Bitim, F. Duman, Nat. Prod. Res. 2014, 28,
2186–2190.
67. F. Shahidi, J. Synowiecki, J. Agric. Food Chem. 1991, 39, 1527–1532.
68. E.L. Johnson, Q.P. Peniston, In R.E. Martin, G.J. Flick, C.E. Hebard, D.R. Ward (Eds.), Chemistry &
Biochemistry of Marine Food Products, AVI Publishing Co.: Westport, 1982.
69. N. Okafor, BBA – Mucoproteins Mucopolysaccharides 1965, 101, 193–200.
70. T. Troung, R. Hausler, F. Monette, Rev. Sci. Eau. 2007, 20, 253–262.
71. F.O. Marquis‐Duval, Isolation et valorisation des constituants de la carapace de la crevettenordique,
PhD thesis, Laval University, Quebec, Canada, 2008.
72. M. Kaya, T. Baran, M. Karaarslan, Nat. Prod. Res. 2015, 29, 1477–1480.
73. A.B. Foster, R.H. Hackman, Nature 1957, 180, 40–41.
74. C.Y. Soon, Y.B. Tee, C.H. Tan, A.T. Rosnita, A. Khalina, Int. J. Biol. Macromol. 2018, 108,
135–142.
75. L. Bastiaens, L. Soetemans, E. D’Hondt. Kathy Elst, In L.A.M. van den Broek, C. Boeriu (Eds.),
Chitin and Chitosan: Properties and Applications, Wiley, ISBN: 9781119450436.
76. A. Hassainia, H. Satha, S. Boufi, Int. J. Biol. Macromol. 2017, 117, 1334–1342.
77. A. Khanafari, R. Marandi, S. Sanatei, Iran J. Env. Heal. Sci. Eng. 2008, 5, 1–24.
78. R.O. Bustos, M.G. Healy, In Proceedings of the Second International Symposium on Environmental
Biotechnology; Biotechnology’ 94; Brighton, UK, 1994; pp. 15–25.
79. X. Chen, Q. Jiang, Y. Xu, W. Xia, J. Aquat. Food Prod. Technol. 2017, 26, 1210–1220.
80. F. Sedaghat, M. Yousefzadi, H. Toiserkani, S. Najafipour, Int. J. Biol. Macromol. 2016, 82, 279–283.
81. F.K. Paes da Silva, D.W. Brück, W.M. Brück, FEMS Microbiol. Lett. 2017, 364, 1–6.
82. I. Younes, S. Hajji, V. Frachet, M. Rinaudo, K. Jellouli, M. Nasri, Int. J. Biol. Macromol. 2014, 69,
489–498.
83. S. Hajji, O. Ghorbel‐Bellaaj, I. Younes, K. Jellouli, M. Nasri, Int. J. Biol. Macromol. 2015, 79,
167–173.
58 Chitin and Chitosan: Properties and Applications
84. C. Hongkulsup, V.V. Khutoryanskiy, K. Niranjan, J. Chem. Technol. Biotechnol. 2016, 91,
1250–1256.
85. A. Hammami, M. Hamdi, O. Abdelhedi, M. Jridi, M. Nasri, A. Bayoudh, Int. J. Biol. Macromol.
2017, 96, 272–281.
86. S. Mhamdi, I. Bkhairia, R. Nasri, T. Mechichi, M. Nasri, A.S. Kamoun, Int. J. Biol. Macromol. 2017,
104, 739–747.
87. M. Hamdi, A. Hammami, S. Hajji, M. Jridi, M. Nasri, R. Nasri, Int. J. Biol. Macromol. 2017, 101,
455–463.
88. M. Bajaj, A. Freiberg, J. Winter, Y. Xu, C. Gallert, Appl. Microbiol. Biotechnol. 2015, 99,
9835–9846.
89. R. Jayakumar, H. Tamura, Int. J. Biol. Macromol. 2008, 43, 32–36.
90. S. Kaufmann, I.M. Weiss, M. Tanaka, J. Am. Chem. Soc. 2007, 129, 10807.
91. D. Pisani, W. Pett, M. Dohrmann, R. Feuda, O. Rota‐Stabelli, H. Philippe, N. Lartillot, G. Wörheide,
Proc. Natl. Acad. Sci. 2015, 112, 15402.
92. M. Wysokowski, T. Jesionowski, H. Ehrlich, Am. Mineral. 2018, 103, 665–691.
93. A. Kollikner, Der feinere Bau der Protozoen, Wilhelm Engelmann Leipzig: Leipzig, 1986.
94. W.J. Sollas, Zoology 1888, 25, 1–458.
95. G.C.J. Vosmaer, H.P. Wijsman, In KNAW, Proceedings; Huygens Institute – Royal Netherlands
Academy of Arts and Sciences, Amsterdam, 1905, pp. 15–28.
96. W.J. Schmidt, Zool. Jahrb. Abt. Anat. Ontog. Tiere 1926, 48, 311–364.
97. T.L. Simpson, P.‐F. Langenbruch, L. Scalera‐Liaci, Zoomorphology 1985, 105, 375–382.
98. J.N. Cha, K. Shimizu, Y. Zhou, S.C. Christiansen, B.F. Chmelka, G.D. Stucky, D.E. Morse, Proc.
Natl. Acad. Sci. 1999, 96, 361–365.
99. D.F. Travis, C.J. François, L.C. Bonar, M.J. Glimcher, J. Ultrastructure Res. 1967, 18, 519–550.
100. G. Croce, A. Frache, M. Milanesio, L. Marchese, M. Causà, D. Viterbo, A. Barbaglia, V. Bolis, G.
Bavestrello, C. Cerrano, U. Benatti, M. Pozzolini, M. Giovine, H. Amenitsch, Biophys. J. 2004, 86,
526–534.
101. H. Ehrlich, T. Hanke, P. Simon, C. Goebel, S. Heinemann, R. Born, H. Worch, BIOmaterialien 2005,
6, 297–302.
102. H. Ehrlich, T. Hanke, H. Meissner, G. Richter, R. Born, S. Heinemann, A.V. Ereskovsky, D.D.
Krylova, H. Worch, VDI Berichte 2005, 1920, 163–166.
103. C. Eckert, H. Ehrlich, T. Hanke, S. Heinemann, H. Meissner, H. Worch, Glasschwammkollagen,
Verfahren zu dessen Herstellung und Verwendung, 2005, Patent no. DE 10 2005 041 414.
104. H. Ehrlich, A.V. Ereskovskii, A.L. Drozdov, D.D. Krylova, T. Hanke, H. Meissner, S. Heinemann, H.
Worch, Russ. J. Mar. Biol. 2006, 32, 186–193.
105. H. Ehrlich, H. Worch, In E. Hajdu (Ed.), Porifera Research: Biodiversity, Innovation & Sustainability;
Museu Nacional: Rio de Janiero, 2007, pp. 217–223.
106. H. Ehrlich, D. Janussen, P. Simon, V.V. Bazhenov, N.P. Shapkin, C. Erler, M. Mertig, R. Born, S.
Heinemann, T. Hanke, H. Worch, J.N. Vournakis, J. Nanomater. 2008, Article ID 670235.
107. Y. Noishiki, H. Takami, Y. Nishiyama, M. Wada, O. Shigeru, K. Shigenori, Biomacromolecules 2003,
4, 896–899.
108. J.N.A. Hooper, R.W.M. Van Soest, Systema Porifera: A Guide to the Classification of Sponges;
Kluwer Academic/Plenum Publishers: New York, 2002.
109. C. Morrow, P. Cárdenas, Front. Zool. 2015, 12, 1–27.
110. F. Sandford, Microsc.Res. Tech. 2003, 62, 336–355.
111. H. Ehrlich, P.G. Koutsoukos, K.D. Demadis, O.S. Pokrovsky, Micron 2008, 39, 1062–1091.
112. H. Ehrlich, P.G. Koutsoukos, K.D. Demadis, O.S. Pokrovsky, Micron 2009, 40, 169–193.
113. H. Ehrlich, P. Simon, W. Carrillo‐Cabrera, V.V. Bazhenov, J.P. Botting, M. Ilan, A.V. Ereskovsky, G.
Muricy, H. Worch, A. Mensch, R. Born, A. Springer, K. Kummer, D.V. Vyalikh, S.L. Molodtsov, D.
Kurek, M. Kammer, S. Paasch, E. Brunner, Chem. Mater. 2010, 22, 1462–1471.
Methods of Isolating Chitin from Sponges (Porifera) 59
114. L. de Castro, F.P. Capote, In L. de Castro, F.P. Capote (Eds.), Techniques and Instrumentation in
Analytical Chemistry; Elsevier: Amsterdam, 2007, pp. 1–34.
115. S. Moldoveanu, In L. de Castro, F.P. Capote (Eds.), Techniques and Instrumentation in Analytical
Chemistry; Elsevier: Amsterdam, 2007, pp. 69–97.
116. E. Roselló‐Soto, O. Parniakov, Q. Deng, A. Patras, M. Koubaa, N. Grimi, N. Boussetta, B.K. Tiwari,
E. Vorobiev, N. Lebovka, F.J. Barba, Food Eng. Rev. 2016, 8, 214–234.
117. W.G. Birolli, J.A.D.M. Delezuk, S.P. Campana‐Filho, Appl. Acoust. 2016, 103, 239–242.
118. F.O. Ogutu, J. Food Process. 2015, 6, 446.
119. P. Lidström, J. Tierney, B. Wathey, J. Westman, Tetrahedron 2001, 57, 9225–9283.
120. K.E. Haque, Int. J. Miner. Process. 1999, 57, 1–24.
121. M. Marić, A.N. Grassino, Z. Zhu, F.J. Barba, M. Brnčić, S. RimacBrnčić, Trends Food Sci. Technol.
2018, 76, 28–37.
122. J.L. Shamshina, O. Zavgorodnya, R.D. Rogers, Adv. Biochem.Eng. Biotechnol. 2018, 127–141.
123. A. Sahu, P. Goswami, U. Bora, J. Mater. Sci. Mater. Med. 2009, 20, 171–175.
124. M. Mahdy Samar, M.H. El‐Kalyoubi, M.M. Khalaf, M.M. Abd El‐Razik, Ann. Agric. Sci. 2013, 58,
33–41.
125. T. Lertwattanaseri, N. Ichikawa, T. Mizoguchi, Y. Tanaka, S. Chirachanchai, Carbohydr. Res. 2009,
344, 331–335.
126. R. Zhang, C. Zhou, L. Li, M. Lui, Adv. Mater. Res. 2013, 749, 160–166.
127. C. Peniche, W. Argüelles‐Monal, M. Goycoolea, In M.N. Belgacem, A. Gandini (Eds.), Monomers,
Polymers and Composites from Renewable Resources, Elsevier: Amsterdam, 2008, pp. 517–542.
128. H.C.W. Skinner, H. Ehrlich, In K.K. Turekian, H.D. Holland (Eds.), Treatise on Geochemistry. Vol.
10: Biogeochemistry, 2nd Edition; Elsevier Science: Amsterdam, 2014, pp. 105–162.
129. D. Stawski, S. Rabiej, L. Herczyńska, Z. Draczyński, J. Therm. Anal. Calorim. 2008, 93, 489–494.
130. I. Petrenko, V.V. Bazhenov, A.L. Stelling, V.Z. Kutsova, In H. Ehrlich, (Ed.), Extreme Biomimetics;
Springer‐Verlag: Cham, 2017, pp. 205–221.
131. I. Petrenko, V.V. Bazhenov, R. Galli, M. Wysokowski, J. Fromont, P.J. Schupp, A.L. Stelling, E.
Niederschlag, H. Stöker, V.Z. Kutsova, T. Jesionowski, H. Ehrlich, Int. J. Biol. Macromol. 2017, 104,
1626–1632.
132. B.P. Chan, K.W. Leong, Eur. Spine J. 2008, 17, 467–479.
133. F.J. O’Brien, Mater. Today 2011, 14, 88–95.
3
Physicochemical Properties
of Chitosan and its Degradation
Products
Karolina Gzyra‐Jagieła, Bożenna Pe ̨czek, Maria Wisń iewska‐Wrona,
and Natalia Gutowska
Institute of Biopolymers and Chemical Fibres, Lodz, Poland
Solubility Viscosity
kM a
1
Differential weight fraction
0.8
(1/log(g/mol))
0.6
0.4
0.2
0
3 3.5 4 4.5 5 5.5 6
logM g/mol
Virgin Thermal degradation Thermal–mechanical degradation
Figure 3.2 Distribution of average molar mass (MMD) of microcrystalline chitosan samples after different
degradation processes.
Sample Time of degradation (h) Mn, g/mol Mw, g/mol Decrease of Mw, % Mw/Mn
Chitosan A (DD = 97.7%) Virgin 38 670 97 650 ‐ 2.5
0.5 26 650 105 740 ‐ 4.0
1 23 760 103 400 ‐ 4.4
Chitosan B (DD = 74.4%) Virgin 38 600 120 130 ‐ 3.1
0.5 37 480 115 830 3.6 3.1
1 32 370 108 250 9.89 3.1
two columns were used: PLaquagel–OH MIXED and PLaquagel–OH 40, 300 mm long,
made by Agilent Co. The results of the analysis are shown in Table 3.1 and Figure 3.2.
The thermal degradation results in a distinct drop in the average molar mass. Combining
of the two processes, thermal and mechanical degradation, does not cause any considerable
decrease in the average molar masses in comparison to the thermal treatment. The impact
of the thermal treatment time upon the structure of polymers with varied DD was also
examined. The results are shown in Table 3.2, and Figures 3.3 and 3.4.
It may be noted that the average molar mass dropped in chitosan B with lower DD and
increased for the high‐DD chitosan A; this is probably caused by cross‐linking of the
polymer. Chitosan B has a lower DD, marking a higher content of acetyl groups prone to
the forming of hydrogen bonds and, as a consequence, has cross‐linked polymers. It was
earlier incubated in strong alkaline medium at high temperature for deacetylation.
Physicochemical Properties of Chitosan and its Degradation Products 65
1
Differential weight fraction
0.8
(1/log(g/mol))
0.6
0.4
0.2
0
3 3.5 4 4.5 5 5.5 6
logM g/mol
Figure 3.3 Distribution of average molar mass of degraded microcrystalline chitosan A with high DD
(97.7%).
1
Differential weight fraction
0.8
(1/log(g/mol))
0.6
0.4
0.2
0
3 3.5 4 4.5 5 5.5 6
logM g/mol
Figure 3.4 Distribution of average molar mass of degraded microcrystalline chitosan B with low DD
(74.4%).
Table 3.3 Analytical results of solutions after the degradation of two chitosan grades (universal
calibration).
Sample Time of degradation, h Method of drying Mn, g/mol Mw, g/mol Mw/Mn
Solution chitosan A 0.5 Dryer 1 410 1 430 1.0
(DD 97.7%) Vacuum evaporator 1 350 1 360 1.0
1 Dryer 1 340 1 360 1.0
Vacuum evaporator 1 330 1 330 1.0
Solution chitosan B 0.5 Dryer 1 650 1 870 1.1
(DD 74.4%) Vacuum evaporator 1 720 2 040 1.2
1 Dryer 2 070 2 730 1.3
Vacuum evaporator 2 170 3 070 1.4
The degradation of chitosan resulted in oligomers with average molar mass in the
range of 1400–2000 g/mol. The obtained results do not show any distinct impact of the
drying method. Oligomers with higher molar mass were achieved for chitosan with a
lower DD, that is, chitosan B. Prolonged degradation time caused a distinct increase of
the molar mass only for chitosan B. The preceding is an example of the practical use of
the GPC/SEC method. It is an important method to follow—that is, not only the degrada-
tion of the polymer, but also its molar mass distribution after other treatments and
processing.
lim red
c 0
The average viscometric molar mass is calculated from the Mark–Houwink equation:
k Mv
Physicochemical Properties of Chitosan and its Degradation Products 67
DD 1 A CH3 / A C1
Here, ACH3 = intensity of the CH3 band, and AC1 = intensity of the C1 band.
80,0
75
70
65
60 2551 cm–1
55
50
45
%T
40
35
30
25 2907 cm–1
20
1290 cm–1
15
1429 cm–1
10 1023 cm–1
3451 cm–1 1322 cm–1
5 1626 cm–1
1553 cm–1
3323 cm–1 1072 cm–1
0,0
4000,0 3000 2000 1500 1000 500 400,0
cm–1
Figure 3.5 Background removal from absorption peaks in an IR spectrum of chitosan [28].
DD, %
Divergence in the assessment of DD occurs in the selected method too, depending on the
way in which the tested material is prepared. It is therefore recommended to prepare the
tested material for comparison always by the same technique—for example, in KBr tablets
or in film. No comparison should be made between results of DD obtained from FT‐IR
tests employing different methods of sample preparation.
dissolved in a small amount of 1% (v/v) acetic acid, and then anhydrous acetic acid is
added to have a water content below 10% (v/v) in the solution. The tested chitosan solution
and 1,4‐dioxan are put to a measuring vessel. Peracetic acid with concentration of 70%
(v/v) serves as the titrant. A diagram is drawn up of the relationship emf/volume of the
perchloric acid solution, and the volume is read, which responds to the neutralization of the
NH3 groups. The amount of HClO4 solution needed to obtain neutralization, titer of the
solution, chitosan sample weight, and volume of chitosan solution are all taken into account
in the calculation of DD. The procedure was prepared according to the OECD guidelines
[29]. The potentiometric titration produces reproducible results comparable to those from
the NMR measurements. The drawback of the method is its application, which is limited to
easy dissolvable preparations.
Deacetylation degree, %
are purified, separated to single components, and detected in a measuring cell of the ther-
mal conduction detector (TCD). The applied method enables computer‐controlled optimi-
zation of the combustion, selective separation of the gaseous components in absorption
columns, and a sequential detection of signals in the TCD of high selectivity. Solid sam-
ples are mostly closed in a tin foil and then compacted to tablets in a press. The samples
are burnt in the oven at 1150°C [31]. Other methods that can be used are the Kjeldahl and
Dumas.
2I I2 2e
Iodine is generated on the electrode in the glass titration cell. The generating cell is situ-
ated close to a measurement electrode—a double‐needle platinum electrode—which, by a
voltmeter, monitors the potential of the sample solution in the course of the coulometric
titration. A classical coulometric cell is composed of two parts: an anode chamber and
cathode chamber separated by a diaphragm. The anode chamber contains an anolyte, that
is, a Karl Fischer electrolyte, which makes the oxidation process run. Iodine is generated in
the anolyte as soon as voltage is connected to the electrode. The anolyte contains sulfur
dioxide, imidazole, and iodine salts. Methanol or ethanol is used as solvent. The cathode
holds a catholyte, which is a reagent that enables the entire electrochemical reaction. The
amount of water titrated in the coulometric technique is proportional to the electric charge
(in coulombs) used to generate iodine. Once the entire water content has reacted with
iodine, free iodine emerges in the titrated solution/anolyte. The titration is stopped as soon
as free iodine has been detected in the titrated cell. The apparatus is equipped with an oven
for keeping the sample at suitable temperature. Streaming nitrogen connected to the
apparatus carries the delivered water molecules to the measuring cell. The method is highly
accurate, producing measurement results on the ppm level [34].
Physicochemical Properties of Chitosan and its Degradation Products 73
X d·R K mg /cm 3
Here, d = dilution; R = protein concentration from the calibration curve for bovine serum
albumin; and K = correction coefficient of amino sugars (concentration of amino sugars
determined from the analytical curve prepared for D‐glucosamine)
The protein content in the starting chitosan sample is calculated from the formula:
n
i 1
Xi Vi n
Z 1000 ppm
m
Here, Xi = content of protein in the extract (mg/cm3); Vi = volume of the extract (cm3);
and m = mass of chitosan sample (g).
R
O H2N C COO– O O
H
OH
2 N
OH
CO2, 3 H2O –
O O O
O
C
H R
obtaining oligomers with the preferred structure. The assessment of chitosan molecular
structure actually calls for the solution of two analytical problems:
• Separation of the oligomeric mixture to separate oligomers. The only applicable method
is liquid chromatography HPLC.
• Identification of the oligomers by the offline use of NMR measurements or mass
spectroscopy–type ESI‐MS or MALDI‐TOF MS after the separation of the oligomeric
mixture to more homogeneous fractions.
Due to the cationic nature of the chitosan oligomeric molecules, three HPLC methods
can be employed for the chromatographic separation:
• Partition chromatography with reverse phases: The difference of the chemical affinity
of oligomer molecules to the column filling and eluent (mobile phase) is exploited.
• Ion‐exchange chromatography: Separation is based on the varied cation potential of
oligomers with different polymerization degrees.
• Gel permeation chromatography: The separation proceeds as result of different
hydrodynamic volumes of the oligomers with different polymerization degrees.
It is known from literature, and our own experience, that the partition chromatography
with reverse phases by the use of the aminopropyl column LC‐NH2 (Supelco) gives
difficulties, and does not produce satisfying result. This is due to the partial adsorption of
oligomers on the column filling reflected in non‐reproducible chromatograms with a
changeable profile and ever poorer intensity [49–52].
Heterophase degradation of microcrystalline chitosan (MCCh) in the presence of
cellulolytic enzymes yielded a product with partly reduced molar mass and a blend of
water‐soluble oligomers. It was found that MCCh can be degraded with high yield (ca.
99%), while neutral cellulase proved most effective for the material. Wide‐angle X‐ray
scattering (WAXS) examination of water‐soluble degradation products showed the presence
of barely discernible crystallinity peaks characteristic of chitin and a peak unrelated with
elementary chitin cells, originated from a different type of order. The size of the crystallites
is much smaller than of those in virgin and partly degraded chitosan. A study to assess the
impact of chitosan biodegradation products on the stimulation of the germination power of
the tested plants—radish and lettuce (number of germinated seeds, mass of green germs,
and their length in comparison to reference‐water)—showed that oligomers and partly
degraded MCCh stimulate the germination at a lower concentration (0.001% (w/v)) as
compared to virgin chitosan. D (+) glucosamine hydrochloride used for comparison
revealed phytotoxic action at a concentration of 0.01% (w/v). Among the tested preparations,
partly degraded MCCh and water‐soluble oligomers showed bacteriostatic action against
Escherichia coli and poor bacteriostatic activity against Staphylococcus aureus.
Investigation of the activity against plant viruses and bacteria carried out in an agriculture
research center revealed that chitosan oligomers quite effectively (100%) inhibited the
growth of Lucerne mosaic virus (AIMV) and, to a lesser degree, the growth of tobacco
mosaic virus (TMV). The soluble products of chitosan degradation inhibit the growth of
plant bacteria Clavibacter michiganensis subsp. michganensis, Erwinia carotovora subsp.
carotovora, and E. coli at concentrations of up to 0.5% (w/v). It was found that chitosan
and its degradation products showed activity against pathogens of mushroom plants:
Rhizoctonia solani and Myrothecium roridum. Investigation in the use of chitosan
Physicochemical Properties of Chitosan and its Degradation Products 77
3.3 Outlook
The physicochemical properties of chitosan and its degradation products (molar mass, DD,
etc.) are crucial for their application, making testing of the properties a critical part of the
investigation in that sector. A precise and reliable analysis is a must when it comes to
medical applications bearing hazard to human health and life. Thanks to the development
of new testing techniques, instrumental analysis plays an ever‐growing role in
physicochemical examinations, though experience remains a key factor.
References
1. M. Dasha, F. Chiellini, R.M. Ottenbrite, E. Chiellini: Chitosan—A versatile semi‐synthetic polymer in
biomedical applications, Prog. Polym. Sci., 10.1016/j.progpolymsci.2011.02.001, 36, (981–1014),
(2011).
2. N. Mati‐Baouche, P.H. Elchinger, H. de Baynast, G. Pierre, C. Delattre, P. Michaud: Chitosan as an
adhesive, Eur. Polym. J., 10.1016/j.eurpolymj.2014.09.008, 60, (198–212), (2014).
3. M. Wiśniewska–Wrona, M. Kucharska, M.H. Struszczyk, M. Cichecka, B. Wilbk‐Hałgas, M.
Szymonowicz, D. Paluch, K. Guzińska, Z. Rybak: Hemostatic, resorbable dressing of natural
polymers—Hemoguard, Autex Res. J., 10.1515/aut‐2015‐0051 © AUTEX, 16, (1), (2016).
4. K. Xing, X. Zhu, X. Peng, Sheng Qin: Chitosan antimicrobial and eliciting properties for pest control
in agriculture: A review, Agron. Sustain. Dev., 10.1007/s13593‐014‐0252‐3, 35, (569–588), (2015).
5. R. Singh, K. Shitiz, A. Singh: Chitin and chitosan: Biopolymers for wound management, Int. Wound
J., ISSN 1742‐4801, 10.1111/iwj.12797, 14, (1276–1289), (2017).
6. R.A.A. Muzzarelli: Chitins and chitosans for the repair of wounded skin, nerve, cartilage and bone,
Carbohydrate Polym., 10.1016/j.carbpol.2008.11.002, 76, (167–182), (2009).
78 Chitin and Chitosan: Properties and Applications
7. R. Huang, E. Mendis, N. Rajapakse, S.K. Kim: Strong electronic charge as an important factor for
anticancer activity of chitooligosaccharides (COS), Life Sci., 10.1016/j.lfs.2005.09.039, 78, (2399–
2408), (2006).
8. L. Fernández‐de Castro, M. Mengíbar, A. Sanchez, L. Arroyo, M.C. Villarán, E. Díaz de Apodaca, A.
Heras: Films of chitosan and chitosan‐oligosaccharide neutralized and thermally treated: Effects on its
antibacterial and other activities, LWT–Food Sci. Technol., 10.1016/j.lwt.2016.06.038, 73, (368–374),
(2016).
9. S. Despond, E. Espuche, N. Cartier, A. Domard: Barrier properties of paper–chitosan and paper–chi-
tosan–Carnauba wax films, J. Appl. Polym. Sci., 10.1002/app.21754, 98, (704–710), (2005).
10. W. Xia, P. Liu, J. Zhang, J. Chen: Biological activities of chitosan and chitooligosaccharides, Food
Hydrocolloid., 10.1016/j.foodhyd.2010.03.003, 25, (170–179), (2010).
11. R.A.A. Muzzarelli: Biochemical significance of exogenous chitins and chitosans in animals and
patients, Carbohydrate Polym., 10.1016/0144‐8617(93)90027‐2, 20, (7–16), (1993).
12. S.K. Kim, N. Rajapakse: Enzymatic production and biological activities of chitosan oligosaccharides
(COS): A review, Carbohydrate Polym., 10.1016/j.carbpol.2005.08.012, 62, (357–368), (2005).
13. M.N.V.R. Kumar: A review of chitin and chitosan applications, React. Funct. Polym., 10.1016/
S1381‐5148(00)00038‐9, 46, (1–27), (2000).
14. M. Rinaudo: Chitin and chitosan: Properties and applications, Prog. Polym. Sci., 10.1016/j.progpolym-
sci.2006.06.001, 31, (603–632), (2006).
15. M.A. Barbosa, I.C. Gonçalves, P.M.D. Moreno, R.M. Gonçalves, S.G. Santos, A.P. Pêgo, I.F. Amaral:
2.13 Chitosan, Comprehensive Biomaterials II, 10.1016/B978‐0‐12‐803581‐8.10246‐2, (279–305),
(2017).
16. I. Aranaz, M. Mengíbar, R. Harris, I. Paños, B. Miralles, N. Acosta, G. Galed, Á. Heras: Functional
characterization of chitin and chitosan, Curr. Chem. Biol., 10.2174/2212796810903020203, 3, (203–
230), (2009).
17. P.K. Dutta, M.N.V.R. Kumar, J. Dutta: Chitin and chitosan for versatile applications, J. Macromol. Sci.,
Part C–Polym. Rev., 10.1081/MC‐120006451, 42 (3), (307–354), (2002).
18. M. Lavertu, S. Methot, N. Tran‐Khanh, M.D. Buschmann: High efficiency gene transfer using chi-
tosan/DNA nanoparticles with specific combinations of molecular weight and degree of deacetylation,
Biomaterials, 10.1016/j.biomaterials.2006.04.029, 27, (4815–4824), (2006).
19. H.K. No, S.P. Meyers: Preparation and characterization of chitin and chitosan–A review, J. Aquat.
Food Prod. Technol., 10.1300/J030v04n02_03, 4 (2), (27–52), (1995).
20. S. Podzimek: Light Scattering, Size Exclusion Chromatography and Asymmetric Flow Field Flow
Fractionation: Powerful Tools for the Characterization of Polymers, Proteins and Nanoparticles,
Wiley Book. 10.1002/9780470877975, (2011).
21. M.A. Barbosa, A.P. Pêgo, I.F. Amaral: 2.213—Chitosan, in Comprehensive Biomaterials, P. Ducheyne,
Ed., Elsevier, Oxford, UK, (221–237), (2011).
22. M. Rinaudo, M. Milas, P. Le Dung: Characterization of chitosan. Influence of ionic strength and
degree of acetylation on chain expansion, Int. J. Biol. Macromol., 10.1016/0141‐8130(93)90027‐J, 15,
(281–285), (1993).
23. M.G. Perers: Application and environmental aspects of chitin and chitosan, J. Mat. Sci–Pure Appl.
Chem., Part A, 10.1080/10601329508010276, 32 (4), (629–640), (1995).
24. R. Czechowska‐Biskup, D. Jarosińska, B. Rokita, P. Ulański, J. Rosiak: Determination of degree of
deacetylation of chitosan – comparison of methods, Progress on Chemistry and Application of Chitin
and Its Derivatives, Volume XVII, (2012).
25. M.R. Kasaai: Determination of the degree of N‐acetylation for chitin and chitosan by various NMR
spectroscopy techniques: A review, Carbohydrate Polym., 10.1016/j.carbpol.2009.10.051, 79, (801–
810), (2010).
26. A. Hirai, H. Odani, A. Nakajima: Determination of degree of deacetylation of chitosan by 1H NMR
spectroscopy, Polym Bull., 10.1007/BF00299352, 26, (87–94), (1991).
Physicochemical Properties of Chitosan and its Degradation Products 79
27. K.M. Vårum, M.W. Anthonsen, H. Grasdalen, O. Smidsrød: Determination of the degree of N‐acetyla-
tion and the distribution of N‐acetyl groups in partially N‐deacetylated chitins (chitosans) by high‐field
N.M.R. spectroscopy, Carbohydr Res., 10.1016/0008‐6215(91)84142‐2, 211 (1), (17–23), (1991).
28. J. Burgnerotto, J. Lizardi, F.M. Goycoolea, W. Argüelles‐Mondal, J. Desbriéres, M. Rinaudo: An infra-
red investigation in relation with chitin and chitosan characterization, Polymer, 10.1016/
S0032‐3861(00)00713‐8, 42, (3569–3580), (2001).
29. OECD Guidelines for Testing of Chemicals Test No. 112.
30. Brookfield (https://www.brookfieldengineering.com/‐/media/ametekbrookfield/manuals/obsolete%20
manuals/dial%20m85‐150‐p700.pdf?la=en).
31. A. Ilnicka, J.P. Lukaszewicz: Synthesis of N‐rich microporous carbon materials from chitosan by
alkali activation using Na2CO3, Mat. Sci. Eng. B, 10.1016/j.mseb.2015.08.002, 201, (66–71), (2015).
32. ISO 3451‐1:1997 Plastics – Determination of ash – Part 1: General methods.
33. U.S. Pharmacopeia 38 NF 30.
34. G. Jayme, G. Hahn: Vereinfachte Messung des Wasserrückhaltenvermögen von Zellstoffen, Das Papier,
14, (138–139), (1960).
35. S. Moore: Amino acid analysis: Aqueous dimethyl sulfoxide as solvent for the ninhydrin reaction,
J. Biol. Chem., 243 (23), (6281–6283), (1968).
36. S. Prochazkova, K.M. Vårum, K. Østgaa: Quantitative determination of chitosans by ninhydrin,
Carbohydrate Polymers, 10.1016/S0144‐8617(98)00108‐8, 38, (115–122), (1999).
37. H.K No, N.Y Park, S.H. Lee., S.P. Meyers: Antibacterial activity of chitosans and chitosan oligomers
with different molecular weights, Int. J. Food Microbiol, 10.1016/S0168‐1605(01)00717‐6, 74, (65–72),
(2002).
38. M. Tian, F. Chen, D. Ren, X. Yu, X. Zhang, R. Zhong, Ch. Wan: Preparation of a series of chitooligomers and
their effect on hepatocytes, Carbohydrate Polym., 10.1016/j.carbpol.2009.07.039, 79, (137–144), (2010).
39. V.K. Mourya, N.N. Inamdar, Y.M. Choudhari: Chitooligosaccharides: Synthesis, characterization and
applications, Polym. Sci., 10.1134/S0965545X11070066, 53, (583–612), (2011).
40. G. Lodhi, Y.S. Kim, J.W. Hwang, S.K. Kim, Y.J. Jeon, J.Y. Je, C.B. Ahn, S.H. Moon, B.T. Jeon, P.J.
Park: Chitooligosaccharide and its derivatives: Preparation and biological applications, BioMed Res.
Int., 10.1155/2014/654913, vol. 2014, article ID 654913.
41. B.B. Aam, E.B. Heggset, A.L. Norberg, M. Sorlie, K.M. Varum, V.G.H. Eijsink: Production of chitoo-
ligosaccharides and their potential applications in medicine, Mar. Drugs, 10.3390/md8051482, 8,
(1482–1517), (2010).
42. E.K. Khor, L.Y. Lim: Implantable applications of chitin and chitosan, Biomaterials, 10.1016/
S0142‐9612(03)00026‐7, 24, (2339–2349), (2003).
43. US Patent Nr 5, 482, 843, 1996.
44. H. Struszczyk: Progress on the modification of chitosan, in A. Domard, Ch. Jeunieux, R. Muzzarelli,
G. Roberts, eds.; Advances in Chitin Science; vol. 1; J. Andre Publ., Lyon; 24.
45. A.V. Ilyina, N.Y. Tatarinova: The preparation of low‐molecular‐weight chitosan using chitinolytic
complex from Streptomyces kurssanovii, Process Biochem., 10.1016/S0032‐9592(99)00022‐9, 34;
(875–878), (1999).
46. C. Lee, S.S. Shin, Y.C. Lee: Degradation of chitosan with the aid of lipase from Rhizopus japonicus for
the production of soluble chitosan, IFT Annual Meeting; June 10–14; Dallas; USA; (2000).
47. H. Zhang, S.H. Neau: In vitro degradation of chitosan by a commercial enzyme preparation: Effect of
molecular weight and degree of deacetylation, Biomaterials, 10.1016/S0142‐9612(00)00326‐4, 22;
(1653–1658), (2001).
48. R.A.A. Muzzarelli, W. Xia, M. Tomasetti, P. Ilari: Depolymerization of chitosan and substituted
chitosans with the aid of a wheat germ lipase preparation, Enzyme Microbial Technol., 10.1016/
0141‐0229(94)00015‐J, 17, (541–545), (1995).
49. H. Struszczyk et al. Polish Patent Application P‐351600 – Method to produce modified microcrystal-
line chitosan, (2002).
80 Chitin and Chitosan: Properties and Applications
* This review is dedicated to our friend and teacher Dr. Nour Eddine ’Nouri’ El Gueddari who sadly passed
away in February 2018.
# These authors contributed equally.
describe in more detail the newly emerging mass spectrometric tools for the structural
analysis of partially acetylated chitosan oligomers (paCOS). Quantitative MS sequencing
allows the full structural description of paCOS mixtures, and it is also a prerequisite for
enzymatic / MS fingerprinting analyses of chitosan polymers.
4.1 Introduction
The functional biopolymer chitosan has long been, and far too often still is, hailed as a
cure‐all wonder drug. Not surprisingly, this has raised some unrealistic expectations lead-
ing to disappointment upon failure, or scepticism leading to blunt disbelief and rejection.
However, research done over the past two decades has changed this picture by increasingly
elucidating structure–function relationships of partially acetylated chitosans, in terms of
both their physico‐chemical properties and biological functionalities. Different chitosans
behave differently in aqueous solutions and exhibit different bioactivities. There is not one
cure‐all chitosan; instead, there are many different chitosans, and choosing the right one for
a given task is key to reliable success. Paramount to this transition from the ill‐defined
first‐generation chitosan to well‐defined second‐generation chitosans has been the devel-
opment of reliable methods for the structural characterisation of chitosans. These in turn
have enabled the development of reliable processes for the reproducible production of chi-
tosans with known structural properties and low batch‐to‐batch variabilities. These are now
becoming available in the market in industrially relevant quantities from different suppliers
worldwide, allowing the development of reliable agricultural or biomedical products with
known functionalities.
The structural characteristics which have been the focus of development of second‐
generation chitosans have been DP and FA. For both parameters, a number of analytical
methods have been developed with different advantages and disadvantages, and gold
standards have been established. A number of reviews have been published in the past
describing these, so short summaries will suffice here. It is important to realise that both
DP and FA values are invariably average values as any chitosan sample will be a mixture of
molecules differing in their DP and FA. As a consequence, description of a chitosan sample
should ideally not only indicate these average values, but also their variation within the
sample. While polymer chemistry offers different methods for analysing the ÐDP (‘D
stroke’), describing the FA variability ÐFA is still in its infancy.
A third parameter, namely PA, that is, the distribution of acetylated GlcNAc and deacety-
lated GlcN residues within the chitosan chains, has been discussed controversially. It had
initially been thought that chitosan produced by heterogeneous partial de‐N‐acetylation of
chitin would possess non‐random, block‐wise PA, while chitosans produced by homogene-
ous partial de‐N‐acetylation of chitin or by homogeneous partial re‐N‐acetylation of poly-
glucosamine would possess random PA. Reliably random PA was regarded as a quality
criterion, with some authorities even going as far as defining chitosan as a copolymer of
randomly distributed GlcNAc and GlcN residues. Later, more detailed investigations,
however, suggested that all chitosans available in the market appear to be characterised by
random PA. As a consequence, the possible influence of PA on physico‐chemical properties
and/or biological functionalities is unknown at present. However, recent results showed
that enzymatic rather than chemical modifications of the FA of chitosans can lead to non‐
random PAs. Interestingly, chitosans with either more block‐wise or more regular than
New Developments in the Analysis of Partially Acetylated Chitosan Polymers and Oligomers 83
random PA can be produced using different chitin deacetylases. Functional analyses of first
such chitosans suggest that these structurally novel chitosans are also functionally differ-
ent from conventional chitosans with random PA. PA, thus, emerges as an additional struc-
tural parameter of significant relevance for chitosans’ functionalities. This emerging
insight has been strongly supported by novel analytical methods to characterise the PA of
chitosan polymers.
In parallel to this development, and partially as a consequence of it, the focus of chitosan
research has lately shifted somewhat from polymers to oligomers. This shift was motivated
by the realisation that at least some of the biological activities of chitosan polymers may in
fact be conveyed by their oligomeric breakdown products generated by chitosan‐hydrolysing
enzymes such as chitinases or chitosanases present in the target tissues. In addition, the
shift was clearly favoured by the fact that the structural analysis of chitosan oligomers is
much more straightforward than that of chitosan polymers. It needs to be noted that there
is no clearly defined boundary between chitosan oligomers and chitosan polymers.
Chitosans of DP <10 are clearly oligomers and chitosans of DP >100 are clearly polymers,
and everything in between may be considered large oligomers or, rather, small polymers. A
practical boundary may be a DP of around 20, coinciding cum grano salis with the bound-
ary of analytical techniques best suited for their analysis.
The structural analysis of paCOS is more advanced than that of chitosan polymers,
particularly with the recent developments in mass spectrometry. At the same time, paCOS
analysis is also an integral component of the recently developed enzymatic / MS
fingerprinting analyses of chitosan polymers. Therefore, we will first discuss the structural
analysis of chitosan oligomers followed by that of chitosan polymers.
All commercially available chitosan oligomers and most of the paCOS used for research
are mixtures of oligomers, usually differing in all the three parameters described earlier,
namely DP, FA and PA, rarely in FA and PA only [8]. Therefore, the identification, analysis
and quantification of the paCOS within one sample taking into account DP, FA and PA is the
crucial step to correlate the biological functions of paCOS to their structure. Furthermore,
a detailed analysis of the sample is important to allow batch‐to‐batch comparisons, a con-
ditio sine qua non for reproducible results. To this end, MS is currently seen as the most
suitable method, the gold standard, for a large number of biomolecules. Even in complex
samples, paCOS with different DP and FA can easily be identified due to their different
masses, and PA can be analysed with multiple‐stage mass spectrometry (MSn) experiments.
In these experiments, the paCOS are fragmented in the MS system, and the mass‐to‐charge
ratios of the fragments then allow the deciphering of the sequence of GlcN and GlcNAc
units in the paCOS [3, 9]. A major drawback in MS analysis of paCOS and other com-
pounds is that it yields qualitative but not quantitative data. This situation arises from mul-
tiple problems, for example, that DP, FA and PA are all influencing the ionization efficiency,
that different adducts are formed during ionization, that ion suppression may occur, or that
in‐source fragmentation leads to over‐representation of small and under‐representation of
large paCOS. Furthermore, the distribution of GlcN and GlcNAc units in the oligomer
influences the fragmentation of the paCOS, which is required to elucidate their sequence in
the oligomer, as the glycosidic linkages within the four possible chitosan diads (GlcNAc–
GlcNAc, GlcNAc–GlcN, GlcN–GlcNAc, GlcN–GlcN) are chemically not identical, so that
some fragmentation events will occur more frequently, while some fragments will be
under‐represented [10]. As a consequence, only isolated paCOS can be sequenced without
further precautions. In a mixture of just two paCOS having the same DP and FA, a quantita-
tive statement about the sample composition is not possible using simple MSn, and if too
many paCOS with different PA are in the sample, or if the paCOS are too large, not even the
identification of the PA is possible in this way.
To overcome these drawbacks, several improvements in the analytical process had to be
developed. In a first step, the MS detection was combined with liquid chromatography
(LC) to at least partially separate the paCOS before they are analysed by MS. This
separation can be achieved using hydrophobic chromatography, for example, on a C18
column, or using hydrophilic interaction chromatography (HILIC), for example, on an
amide column [11]. HILIC has been confirmed as the most suitable method for the
separation of complex paCOS mixtures containing oligomers of both low and high DP and
FA [9]. These are separated according to their hydrophilicity, a parameter that is influenced
by both the DP and FA, but very little by the PA. The advantage of a partial or complete
paCOS separation before they are analysed by MS is that problems like ion suppression or
in‐source fragmentation can be easily overcome. Importantly, a complete separation of all
compounds in the LC step is not required.
A further parameter complicating quantitative paCOS analysis is the effect of
their N‐acetylation on ionization and fragmentation. Therefore, quantitative chemical
re‐N‐acetylation of the paCOS mixture using isotopically labelled acetic anhydride is per-
formed before the sample is analysed by MS. The acetic anhydride reacts with the free
amino group of GlcN, resulting in fully acetylated paCOS – in fact, chitin oligomers. To
distinguish between ‘old’, originally present acetyl groups and ‘new’, chemically attached
acetyl groups, deuterated [2H6] acetic anhydride is used in this process, so that the ‘new’
New Developments in the Analysis of Partially Acetylated Chitosan Polymers and Oligomers 85
GlcNAc units are 3 Da heavier than the ‘old’ ones, allowing them to be distinguished in MS
[12]. A further benefit of the re‐N‐acetylation is that the chemical complexity of the sample
is strongly reduced, as all chitosan oligomers are converted to chitin oligomers. As a con-
sequence, HILIC gives access to well‐separated fractions containing chitin oligomers of a
single DP each, but, of course, with different amounts and distributions of isotopically
labelled and non‐labelled acetyl groups within each fraction. Therefore, only a few internal,
double isotopically labelled standards, one for each DP, are sufficient to enable quantitative
analysis, as only the effect of DP on quantification remains, while those of FA and PA are
eliminated. These standards are produced from a series of fully deacetylated GlcN
oligomers with defined DP by chemical re‐N‐acetylation using acetic anhydride, which has
six deuterium atoms and four 13C carbon atoms, leading to GlcNAc units which are 5 Da
heavier than the non‐labelled ones. In the next step, the [2H3]-re‐N‐acetylated paCOS mix-
ture and known amounts of the [13C2;2H3]-labelled internal standards are mixed and ana-
lysed by HILIC‐MS. HILIC separation leads to co‐elution of oligomers having the same
DP, including the internal standard. MS detection is used to characterise different paCOS
in the mixture concerning their DP and FA based on their mass, and the intensity relative to
the internal standards is used for quantification. This process eliminates the influence of
DP and FA on ionization efficiency and fully resolves potential problems of ion suppression
and in‐source fragmentation [7].
Still, the third structural parameter, PA, has to be included in the quantitative analysis. To
this end, the [2H3]‐modified paCOS samples are analysed by MSn, where the compounds
are fragmented in the MS detector and the fragments are used to sequence the oligomers.
Re‐N‐acetylation of the sample eliminates the effect of FA on fragmentation, but a second
modification is required to distinguish between fragments coming from the reducing versus
non‐reducing end of the oligomer. Reduction of the reducing end, coupling the reducing
end to 2‐aminoacridone (AMAC) or exchanging the oxygen in the hydroxyl group of the
reducing end against 18O oxygen are commonly used methods for this step [12–14]. This
change in mass of the reducing end sugar unit allows for the unambiguous allocation of
each fragment to either the reducing or non‐reducing end, a prerequisite for sequencing.
Finally, the quantitative data obtained during MS of the deuterated paCOS mixture
informing about DP and FA of the oligomers is combined with the quantitative pattern
analysis obtained during MSn of the deuterated and 18O‐labelled paCOS mixture. This
method allows to fully and quantitatively analyse complex paCOS mixtures regarding their
DP, FA and PA. One remaining disadvantage of the method is that it is restricted to the
analysis of paCOS with a DP <8 as larger chitin oligomers are not soluble in aqueous
media. This limitation does not appear to significantly impede the analysis of paCOS
mixtures obtained by chemical or enzymatic depolymerisation of chitosan polymers, which
in most cases contain mainly small oligomers [15, 16]. But if a chitosan polymer of low FA
is digested by a chitinase requiring consecutive GlcNAc units for cleavage, or if a chitosan
polymer of high FA is digested by a chitosanase requiring consecutive GlcN units for cleav-
age, large fragments will also occur. As such mixtures might be of special interest when it
comes to their biological activities, overcoming this size limitation is an important task for
future method development.
An alternative or complementing method for PA analysis has recently been described
with the combination of mass spectrometry and infrared (IR) spectroscopy [17]. When
analysing carbohydrates, IR spectroscopy has the advantage of giving information on, for
86 Chitin and Chitosan: Properties and Applications
example, the monosaccharide content, regio‐ and isomer conformations, and the linkage
type between the carbohydrate monomers [18]. Additionally, IR spectroscopy is quantitative,
if a reference standard for each compound is available [19]. By performing IR spectroscopy
inside the MS detector, the advantages of both analytical methods can be combined. MS
detection gives the m/z ratio and enables fragmentation of the compounds, and the IR spec-
tra of the compounds and, even more importantly, of their fragments are used to further
characterise and quantify them. Applying IR spectroscopy to the fragment ions reduces the
number of reference IR spectra needed to identify the compounds to just a few, as all larger
oligomers can be fragmented into smaller ones. In future, it should become possible to use
this method for the analysis of larger oligomers and possibly even polymers that cannot be
analysed by LC–MS alone.
Đ is rarely given in publications reporting biological activities of chitosans (all too often,
not even average MW or DP and average FA values are reported, though such work should
not even be considered for publication in a reputed journal). However, without knowing the
dispersity, it is impossible to judge the relevance of conclusions concerning a possible
influence of MW / DP on a given biological activity. In an extreme (but not at all rare) case,
a sample with a low average MW / DP might consist of a mixture of small oligomers and
some large polymers, in which case clearly the average MW / DP would be strongly
misleading! This problem is probably one of the reasons for the controversy concerning the
antimicrobial activities of chitosan ‘oligomers’ (as is the ambiguous definition of
‘oligomers’).
though little evidence, to believe that chitosan polymer samples can vary strongly in this
respect. It is rather reasonable to assume, as in the case of ÐMW / ÐDP, that ÐFA will have a
strong influence on the physico‐chemical properties and biological functionalities of
chitosan polymer samples. It is, therefore, necessary to develop methods that can reliably
distinguish between intra‐chain PA and inter‐chain ĐFA of chitosan polymers.
As early as 1997, Roberts [29] suggested that the differences observed between the bulk
properties of chitosans produced by different production routes might not be explained by
different intramolecular distributions of acetylated units along the chitosan chains (PA), but
rather by an intermolecular variation in FA between chitosan chains (ĐFA). He showed that
a chitosan sample produced by partial deacetylation under homogeneous conditions had a
low variation in FA, whereas a sample produced under heterogeneous deacetylation
conditions had a broad distribution of FA when separated by a pH‐solubility fractionation
technique. He therefore proposed to characterise chitosan samples in terms of their
dispersity in FA, since this is likely to influence the polymers’ physico‐chemical as well as
biological properties, just like ÐMW / ÐDP.
Later, Ngyuen et al. [30] studied the MW and FA of chitosans after fractionation by semi‐
preparative SEC. For a low‐MW chitosan fraction (prepared by partial nitrous acid
depolymerisation of a commercial high‐molecular‐weight chitosan, whose method of
preparation is not documented), they found intermolecular heterogeneity in FA where the FA
increased with increasing molecular weight (from FA 0.03 at MWn 4 kDa to FA 0.13 at MWn
31 kDa). This dispersity was thought to be caused by recovering the material after
depolymerisation through alkaline precipitation and centrifugation as the solubility of
chitosans depends on both MW and FA.
Recently, the ĐFA of chitosan polymers was investigated by Mnatsakanyan et al. [31]
and by Thevarajah et al. [32]. They used free solution CE and UV absorption detection to
study the electrophoretic mobility distribution of different chitosan samples. They found a
correlation between FA and weight‐average electrophoretic mobility. However, aggrega-
tion of polymers was a problem when using this method. Another challenge for studying
the effect of ĐFA on the electrophoretic mobility of the polymer is the dependence on refer-
ence chitosans with a narrow FA distribution. Such chitosans can, however, be produced by
chemical re‐N‐acetylation of polyglucosamine under homogeneous conditions [29]. The
electrophoretic mobility of chitosan might also be influenced by its PA, but this has not
been investigated, for obvious reasons.
inhibitory acetate allowed the enzymes to continue the deacetylation process, this obser-
vation would appear to suggest that the enzymatic deacetylation had created low‐FA chi-
tosan polymers with a non‐random PA.
In a more systematic study, Niehues and Wattjes et al. [42] recently compared the PA
produced by three different fungal chitin deacetylases originating from Podospora anse-
rina, Pestalotiopsis sp. and Puccinia graminis f. sp. tritici [39, 40, 43]. Using highly acety-
lated chitosan with FA = 0.6 (and random PA, as in the preceding text), they showed that all
enzymes de‐N‐acetylated the substrate to a product with FA = 0.3, but with differences in
PA. These differences were evident both in traditional 13C‐NMR diad frequency analysis
and in newly developed enzymatic fingerprinting approaches. Especially for PA analysis of
chitosan polymers, enzymatic fingerprinting turned out to be a highly valuable tool, able
to overcome some of the drawbacks of 13C‐NMR analysis. The main disadvantage of
NMR, besides the high sample amount required for a single sample and the rather long
measurement time of up to several hours, is that the diad (and even triad) frequencies will
always inform about acetyl distributions on a very short length frame of two (to three)
monosaccharide units. In contrast, MS‐based fingerprinting analysis allows to look at a
high number of different oligomers with a DP of up to 6 if performed quantitatively (and
even higher if only semi‐quantitatively), thus giving better insight into the dispersity within
the sample.
As already mentioned earlier, enzymatic / MS fingerprinting can, in principle, be per-
formed with any chitosanolytic enzyme, as all of them appear to possess some degree of
subsite specificities or preferences. Clearly, the higher the specificity, the more revealing is
the analysis. In this respect, chitinosanase [26], with its absolute specificity for the diad
GlcN–GlcNAc at subsites (‐2) and (‐1), is of particular interest. As already suggested in the
original publication, this specificity leads to the production of oligomeric products that
represent one full GlcNAc‐block of the substrate followed by a full GlcN‐block.
Chitinosanase fingerprinting, thus, allows the determination of GlcNAc‐ and GlcN‐block
size frequencies. Since chitinosanase‐catalysed depolymerisation can be simulated in silico
[25], it is possible to compare the experimentally determined block size frequencies to
theoretical frequencies based on random statistics. In future, the insight into PA gained by
this method will allow studying the mode‐of‐action of chitin deacetylases on polymeric
substrates in more detail. Also, it will open the door towards analysing the influence of PA
on physico‐chemical properties and biological functionalities of partially acetylated
chitosan polymers.
4.4 Outlook
In the past decade, advances in analytical techniques have paved the way for the transition
from yesterday’s ill‐defined first‐generation chitosan (singular) to today’s well‐defined
second‐generation chitosans (plural). The term ‘well-defined’ refers to the two most prom-
inent structural parameters of partially acetylated chitosans, namely DP and FA. These chi-
tosans, which were first available on a small lab scale only, have been used extensively to
elucidate the structure–function relationships of chitosans, in terms of both their physico‐
chemical properties and biological functionalities. As a consequence, second‐generation
chitosans are not only structurally well defined, but their biological activities are also at
92 Chitin and Chitosan: Properties and Applications
least partially known. Such chitosans are now available in the market in industrial quantities
and reliable qualities, thanks to the implementation of strict quality management measures
by some chitosan‐producing companies routinely making use of these analytical tools. This
availability is beginning to be a game changer for the chitosan markets. For the first time,
global market demand exceeds global production of high‐quality chitosans, leading to
increasing prices. And, products based on second‐generation chitosans are appearing in
different markets.
We believe that the new developments described in this chapter, most prominently MS‐
based sequencing and fingerprinting tools, will induce a similar development towards
third‐generation chitosans of the future. These will be well-defined also in terms of their
ĐDP and ĐFA, and their different, random or non‐random PA. Once available on lab scale,
they will be used not only to improve our understanding of molecular structure–function
relationships, but also to help elucidating cellular modes of action. This development is
already ongoing, using fully defined paCOS. We trust that, eventually, selected third‐gen-
eration chitosans will also be mass‐produced, most likely using biotechnological in vitro
biorefinery or in vivo cell factory processes, making them available for new chitosan‐based
products that will be successful in the bio‐economy markets of the future.
References
1. A. Sørbotten, S.J. Horn, V.G.H. Eijsink, and K.M. Vårum (2005). Degradation of chitosans with chi-
tinase B from Serratia marcescens: Production of chito‐oligosaccharides and insight into enzyme pro-
cessivity. FEBS J., 272(2), 538–549. https://doi.org/10.1111/j.1742‐4658.2004.04495.x.
2. K.M. Vårum, M.W. Anthonsen, H. Grasdalen, and O. Smidsrød (1991). 13C‐N.m.r. studies of the
acetylation sequences in partially N‐deacetylated chitins (chitosans). Carbohydrate Res., 217(C), 19–
27. https://doi.org/10.1016/0008‐6215(91)84113‐S.
3. S. Bahrke, J.M. Einarsson, J. Gislason, S. Haebel, M.C. Letzel, J. Peter‐Katalinić, and M.G. Peter
(2002). Sequence analysis of chitooligosaccharides by matrix‐assisted laser desorption ionization post
source decay Mass Spectrometry 1. Biomacromolecules, 3(4), 696–704. https://doi.org/10.1021/
bm020010n.
4. T. Tegl, C.Öhlknecht, R. Vielnascher, A. Rollet, A. Hofinger‐Horvath, P. Kosma, et al. (2016)
Cellobiohydrolase produce different oligosaccharides from chitosan. Biomacromolecules, 17, 2284–
2292, https://doi.org/10.1021/acs.biomac.6b00547.
5. S.N. Hamer, B.M. Moerschbacher, and S. Kolkenbrock (2014). Enzymatic sequencing of partially
acetylated chitosan oligomers. Carbohydrate Res., 392, 16–20. https://doi.org/10.1016/j.
carres.2014.04.006.
6. I. Tsigos, N. Zydowicz, A. Martinou, A. Domard, and V. Bouriotis (1999). Mode of action of chitin
deacetylase from Mucor rouxii on. Enzyme, 705, 698–705.
7. S. Cord‐Landwehr, P. Ihmor, A. Niehues, H. Luftmann, B.M. Moerschbacher, and M. Mormann
(2017). Quantitative mass‐spectrometric sequencing of chitosan oligomers revealing cleavage sites of
chitosan hydrolases. Anal. Chem., 89(5), 2893–2900. https://doi.org/10.1021/acs.analchem.6b04183.
8. Rinaudo, M. (2006). Chitin and chitosan: Properties and applications. Prog. Polym. Sci., 31(7), 603–
632. https://doi.org/10.1016/j.progpolymsci.2006.06.001.
9. M.C. Tang, A. Nisole, C. Dupont, J.N. Pelletier, and K.C. Waldron (2011). Chemical profiling of
the deacetylase activity of acetyl xylan esterase A (AxeA) variants on chito oligosaccharides using
hydrophilic interaction chromatography‐mass spectrometry. J. Biotechnol., 155(2), 257–265. https://
doi.org/10.1016/j.jbiotec.2011.06.041.
New Developments in the Analysis of Partially Acetylated Chitosan Polymers and Oligomers 93
10. B. Vijayakrishnan, A. Issaree, Y.E. Corilo, C.R. Ferreira, M.N. Eberlin, and M.G. Peter (2011).
MSn of the six isomers of (GlcN)2(GlcNAc)2 aminoglucan tetrasaccharides (diacetyl chitotetra-
oses): Rules of fragmentation for the sodiated molecules and application to sequence analysis of
hetero‐chito oligosaccharides. Carbohydrate Polym., 84(2), 713–726. https://doi.org/10.1016/j.
carbpol.2010.04.041.
11. M. Wuhrer, A. R. De Boer, and A.M. Deelder (2009). Structural glycomics using Hydrophilic interac-
tion chromatography (HILIC) with mass spectrometry. Mass Spectrometry Rev., 28(2), 192–206.
https://doi.org/10.1002/mas.20195.
12. S. Haebel, S. Bahrke, and M.G. Peter, (2007). Quantitative sequencing of complex mixtures of hetero-
chito oligosaccharides by vMALDI‐linear ion trap mass spectrometry. Anal. Chem., 79(15), 5557–
5566. https://doi.org/10.1021/ac062254u.
13. D.J. Harvey (2011). Derivatization of carbohydrates for analysis by chromatography; electrophoresis
and mass spectrometry. J. Chromatogr. B: Anal. Technol. Biomed. Life Sci., 879(17–18), 1196–1225.
https://doi.org/10.1016/j.jchromb.2010.11.010.
14. M.C. Ralet, J.C. Cabrera, E. Bonnin, B. Quéméner, P. Hellìn, and J.F. Thibault, (2005). Mapping sugar
beet pectin acetylation pattern. Phytochemistry, 66(15), 1832–1843. https://doi.org/10.1016/j.
phytochem.2005.06.003.
15. S.J. Horn, P. Sikorski, J.B. Cederkvist, G. Vaaje‐Kolstad, M. Sorlie, B. Synstad et al. (2006). Costs and
benefits of processivity in enzymatic degradation of recalcitrant polysaccharides. Proc. Natl. Acad.
Sci., 103(48), 18089–18094. https://doi.org/10.1073/pnas.0608909103.
16. T. Weikert, A. Niehues, S. Cord‐Landwehr, M.J. Hellmann, and B.M. Moerschbacher (2017).
Reassessment of chitosanase substrate specificities and classification. Nat. Commun., 8(1), 1698.
https://doi.org/10.1038/s41467‐017‐01667‐1.
17. J. Wattjes, B. Schindler, S. Trombotto, L. David, B.M. Moerschbacher, and I. Compagnon (2017).
Discrimination of patterns of N‐acetylation in chito oligosaccharides by gas phase IR spectroscopy
integrated to mass spectrometry. Pure Appl. Chem., 89(9), 1349–1357. https://doi.org/10.1515/
pac‐2017‐0110.
18. B. Schindler, G. Laloy‐Borgna, L. Barnes, A.‐R. Allouche, E. Bouju, V. Dugas et al. (2018). Online
separation and identification of isomers using infrared multiple photon dissociation ion spectroscopy
coupled to liquid chromatography: Application to the analysis of disaccharides regio‐isomers
and monosaccharide anomers. Anal. Chem., 90(20), 11741–11745. https://doi.org/10.1021/acs.
analchem.8b02801.
19. B. Schindler, L. Barnes, G. Renois, C. Gray, S. Chambert, S. Fort et al. (2017). Anomeric memory of
the glycosidic bond upon fragmentation and its consequences for carbohydrate sequencing.
Nat. Commun., 8(1), 973. https://doi.org/10.1038/s41467‐017‐01179‐y.
20. J.Z. Knaul, M.R. Kasaai, V.T. Bui, and K.A. Creber (1998). Characterization of deacetylated chitosan
and chitosan molecular weight review. Canadian J. Chem., 76(11), 1699–1706. https://doi.org/10.1139/
cjc‐76‐11‐1699.
21. I. Younes and M. Rinaudo (2015). Chitin and chitosan preparation from marine sources. Structure,
properties and applications. Marine Drugs, 13(3), 1133–1174. https://doi.org/10.3390/
md13031133.
22. M.R. Kasaai (2009). Various methods for determination of the degree of N‐acetylation of chitin and
chitosan: A review. J. Agric. Food Chem., 57(5), 1667–1676. https://doi.org/10.1021/jf803001m.
23. M.R. Kasaai (2010). Determination of the degree of N‐acetylation for chitin and chitosan by various
NMR spectroscopy techniques: A review. Carbohydrate Polym., 79(4), 801–810. https://doi.
org/10.1016/j.carbpol.2009.10.051.
24. J. Kumirska, M. Czerwicka, Z. Kaczyński, A. Bychowska, K. Brzozowski, J. Thöming, and
P. Stepnowski (2010). Application of spectroscopic methods for structural analysis of chitin and
chitosan. Marine Drugs, 8(5), 1567–1636. https://doi.org/10.3390/md8051567.
94 Chitin and Chitosan: Properties and Applications
25. A. Niehues, J. Wattjes, J. Bénéteau, G.R. Rivera‐Rodriguez, and B.M. Moerschbacher (2017).
Chitosananalysis by enzymatic/mass spectrometric fingerprinting and in silicopredictive modeling.
Anal. Chem., 89(22), 12602–12608. https://doi.org/10.1021/acs.analchem.7b04002.
26. M. Kohlhoff, A. Niehues, J. Wattjes, J. Bénéteau, S. Cord‐Landwehr N. E. ElGueddari et al. (2017).
Chitinosanase: A fungal chitosan hydrolyzing enzyme with a new and unusually specific cleavage pat-
tern. Carbohydrate Polym., 174, 1121–1128. https://doi.org/10.1016/j.carbpol.2017.07.001.
27. Z. Han, Y. Zeng, H. Lu, and L. Zhang (2015). Determination of the degree of acetylation and the dis-
tribution of acetyl groups in chitosan by HPLC analysis of nitrous acid degraded and PMP labeled
products. Carbohydrate Res., 413, 75–84. https://doi.org/10.1016/j.carres.2015.03.002.
28. J. Li, L. Chen, Z. Meng, and G. Dou (2018). Development of a mass spectrometry method for the
characterization of a series of chitosan. Int. J. Biol. Macromol., 121, 89–96. https://doi.org/10.1016/j.
ijbiomac.2018.09.194.
29. G.A.F. Roberts, (1997). Chitosan production routes and their role in determining the structure and
properties of the product. In A. Domard, G.A.F. Roberts, and K. Vårum (Eds.), Advances in Chitin
Science (pp. 22–31). Lyon, France: Jaques André Publisher.
30. S. Nguyen, S. Hisiger, M. Jolicoeur, F.M. Winnik, and M.D. Buschmann (2009). Fractionation and
characterization of chitosan by analytical SEC and1H NMR after semi‐preparative SEC. Carbohydrate
Polym., 75(4), 636–645. https://doi.org/10.1016/j.carbpol.2008.09.002.
31. M. Mnatsakanyan, J.J. Thevarajah, R.S. Roi, A. Lauto, M. Gaborieau, and P. Castignolles (2013).
Separation of chitosan by degree of acetylation using simple free solution capillary electrophoresis.
Anal. Bioanal. Chem., 405(21), 6873–6877. https://doi.org/10.1007/s00216‐013‐7126‐4.
32. J.J. Thevarajah, M.P. Van Leeuwen, H. Cottet, P. Castignolles, and M. Gaborieau (2017). Determination
of the distributions of degrees of acetylation of chitosan. Int. J. Biol. Macromol., 95, 40–48. https://doi.
org/10.1016/j.ijbiomac.2016.10.056.
33. K.M. Vårum, M.W. Antohonsen, H. Grasdalen, and O. Smidsrød (1991). Determination of the degree
of N‐acetylation and the distribution of N‐acetyl groups in partially N‐deacetylated chitins (chitosans)
by high‐field n.m.r. spectroscopy. Carbohydrate Res., 211(1), 17–23. https://doi.org/10.1016/0008‐62
15(91)84142‐2.
34. J. Kumirska, M.X. Weinhold, S. Steudte, J. Thöming, K. Brzozowski, and P. Stepnowski (2009).
Determination of the pattern of acetylation of chitosan samples: Comparison of evaluation methods
and some validation parameters. Int. J. Biol. Macromol., 45(1), 56–60. https://doi.org/10.1016/j.
ijbiomac.2009.04.002.
35. M.X. Weinhold, J.C.M. Sauvageau, J. Kumirska, and J. Thöming (2009). Studies on acetylation pat-
terns of different chitosan preparations. Carbohydrate Polym., 78(4), 678–684. https://doi.
org/10.1016/j.carbpol.2009.06.001.
36. F.A. Bovey and P.A. Mirau (1996). NMR of Polymers. San Diego, CA, U.S.A.: Academic Press.
37. N.E. ElGueddari, S. Kolkenbrock, A. Schaaf, N. Chilukoti, F. Brunel, C. Gorzelanny et al. (2014).
Chitin and chitosan modifying enzymes: Versatile novel tools for the analysis of structure‐function
relationship of partially acetylated chitosans. In S.P.C. Filho, M.M. Beppu, and A. Fiamingo (Eds.),
Advances in Chitin Science (Vol. 14, pp. 40–47).
38. A. Martinou, V. Bouriotis, B.T. Stokke, and K.M. Vårum (1998). Mode of action of chitin deacetylase
from Mucor rouxii on partially N‐acetylated chitosans. Carbohydrate Res., 311(1–2), 71–78. https://
doi.org/10.1016/S0008‐6215(98)00183‐9.
39. J. Hoßbach, F. Bußwinkel, A. Kranz, J. Wattjes, S. Cord‐Landwehr, and B.M. Moerschbacher (2018).
A chitin deacetylase of Podospora anserina has two functional chitin binding domains and a unique
mode of action. Carbohydrate Polym., 183(July 2017), 1–10. https://doi.org/10.1016/j.carbpol.
2017.11.015.
40. S. Cord‐Landwehr, R.L.J. Melcher, S. Kolkenbrock, and B.M. Moerschbacher (2016). A chitin dea-
cetylase from the endophytic fungus Pestalotiopsis sp. Efficiently inactivates the elicitor activity of
chitin oligomers in rice cells. Scientific Rep., 6(1), 38018. https://doi.org/10.1038/srep38018.
New Developments in the Analysis of Partially Acetylated Chitosan Polymers and Oligomers 95
41. H. Aragunde, X. Biarnés, and Planas, A. (2018). Substrate recognition and specificity of chitin dea-
cetylases and related family 4 carbohydrate esterases. Int. J. Mol. Sci., 19(2), 412. https://doi.
org/10.3390/ijms19020412.
42. J. Wattjes, A. Niehues, S. Cord-Landwehr, J. Hoßbach, L. David, T. Delair, and B.M. Moerschbacher
(2019). Enzymatic production and enzymatic-mass spectrometric fingerprinting analysis of chitosan
polymers with different non-random patterns of acetylation. J. Am. Chem. Soc., 241, 3137–3145.
https://doi.org/10.1021/jacs.8b12561.
43. S. Naqvi, S. Cord‐Landwehr, R. Singh, F. Bernard, S. Kolkenbrock, N.E. El Gueddari, and B.M.
Moerschbacher (2016). A recombinant fungal chitin deacetylase produces fully defined chitosan oli-
gomers with novel patterns of acetylation. Appl. Environ. Microbiol., 82(22), 6645–6655. https://doi.
org/10.1128/AEM.01961‐16.0004461242.
5
Chitosan‐Based Hydrogels
Zhengke Wang, Ling Yang, and Wen Fang
Institute of Biomedical Macromolecules, Department of Polymer Science and Engineering,
Zhejiang University, Hangzhou, China
Hydrogels are cross‐linked polymeric networks that can retain a moisture environment and
mimic human soft tissues. Chitosan, a linear polysaccharide with pH‐responsive self‐
assembling properties, is an attractive candidate for hydrogel construction owing to the
existence of intrinsic intermolecular and intramolecular hydrogen bonds and versatile reac-
tive amino groups. Recently, a variety of chitosan‐based hydrogels have been developed
via diverse approaches, which could be generally categorized as multilayered hydrogels,
hydrogels based on alkali/urea solvent system, injectable hydrogels, self‐healing hydro-
gels, shape memory hydrogels, and superabsorbent hydrogels. Following a brief introduc-
tion of the chitosan hydrogels, this chapter introduces recent advances in chitosan‐based
hydrogels with emphasis on their preparation, characterization, properties, and possible
applications. In addition, current challenges and future perspectives in this field are also
discussed to suggest a new developing direction.
5.1 Introduction
Hydrogels are chemically or/and physically cross‐linked polymeric networks with hydro-
philic groups that can absorb and retain plenty of liquids in an aqueous environment [1].
They are similar to the human soft tissues (e.g. skin, cartilage, muscle, etc.) due to their
high water content and softness, which enable them to become the best candidates for arti-
ficial tissues [2]. Till date, numerous natural polymers, especially polysaccharides, have
been used to design and prepare eco‐friendly hydrogels owing to their abundant resources,
renewability, low production cost, and biodegradability [3]. Chitosan, the main derivative
II III
30 μm
Figure 5.1 Liesegang ring phenomenon and microstructures of multilayered chitosan‐based hydrogels
formed by periodic precipitation spontaneously. (a) A concentric multilayered hydrogel formed by chi-
tosan acidic solution [12]. (b) Liesegang ring phenomenon occurred in chitosan solution [14]. (c) Confocal
laser scanning microscopy (CLSM) micrographs of a physical chitosan hydrogel obtained at different dis-
tances from the top of the gel [15]. (d) Scanning electron microscope (SEM) images of chitosan honey-
combs formed spontaneously by in situ coprecipitation [16].
100 Chitin and Chitosan: Properties and Applications
(a) (b)
OH OH Electrical Structural
On
O O pKa O O input output
HO O Off
HO O Optical image
~6.3 +
NH2 n NH3 n
High pH Low pH Structural
e– pH Front output
50 μm
H+ SEM image
Cathode
Figure 5.2 Programmable fabrication of chitosan hydrogels with multilayered structures by using on–off
electrical inputs to cue self‐assembly. (a) Neutralization mechanism of the cathodic electrodeposition of
chitosan [23]. (b) Complex self‐assembled structures (chitosan multilayers) within the agarose hydrogel
medium induced by electrical signals and optical/SEM images of the resultant hydrogels [24].
It has been demonstrated that short sequences (minutes) of low‐power (about 1 V) elec-
trical inputs can guide self‐assembly to create complicated soft matters [23]. The multi-
layer formation induced by electrical signals is similar to the hydrogels formed through the
alternating process. Imposing and regulating electrical inputs are much simpler and con-
venient as compared to transferring products among different solutions. Nevertheless, the
shape of the resulting hydrogels in this approach is limited to some extent.
O– Na+ OH OH OH
O
O O O O O
O HO O
HO
OH
O
NH
HO HO
NH
+
NH2
O n O n +
Chitosan + +
Sodium hyaluronate –
– +
+ – +
+ +
+
+ – +
+ – +
– +
Cationic + +
liposome +
Cationic
liposome core Multilayered liposome
– –
– – Hyaluronate
– – – shell
– + –
–
+ –
– – Chitosan
+ shell
–
–
– –
+ +–
– Liposome
– –
– – +
– – –
–
–
(Liposome adsorption) Liposome core
capsosome
Figure 5.3 A liposomal core coated with alternately multilayered sodium hyaluronate and chitosan mate-
rials via layer‐by‐layer (LbL) assembly [26].
superficial layer were rapidly transferred into cylindrical molds and exposed under UV
light to gelate. The inner pore size, swelling ratio, as well as compressive mechanical prop-
erties of this hydrogel exhibited a graded transition from top to bottom.
Unprecedented multilayered hydrogels exhibit sophisticated internal structures and tun-
able properties, which show great potential in various biorelated fields, especially involv-
ing cell bioreactors [28], drug codelivery [29], tissue engineering [14], etc. Nevertheless,
the design and formation of multilayered chitosan‐based hydrogels still remain great chal-
lenges: (i) the relationship between microstructures with microscopic properties is unclear,
especially the tribological properties such as wear resistance [30], which is vital for some
specific biomedical applications such as joint replacement; (ii) the studies of multilayered
hydrogels mainly focused on their preparation, structure, and physical properties, while
lacking feasibility analysis and experimental tests for certain applications; (iii) most
researches fabricated multilayered hydrogels with the same component in each layer [29],
which may cause unalterable structures and the boundary disappearance of two layers
under long treatment duration; and (iv) the mechanical strength of synthesized structural
hydrogels usually cannot be as good as natural tissues. Recently, the 3D printing technique
[31] has shown great advantages in the preparation of soft materials with more complex
patterns and sophisticated capabilities. Thus, the sustained development of novel tech-
niques will promote the construction of multilayered hydrogels to a great extent.
Chitosan‐Based Hydrogels 103
Interface
T
50 μm
100 μm
UV
T
S: G1/CS/PEGDMA
T: G2/CS/PEGDMA
D: G3/CS/PEGDMA/OV-POSS
50 μm
D T
Interface
D
50 μm 100 μm
Figure 5.4 SEM micrographs of the multilayered Gtn–CS hydrogel with graded compositions [27]. G1,
G2, and G3 are referred to the methacrylatedgelatin with 22, 39, and 43% of methacrylation degrees,
respectively. S, T, and D are referred to as superficial, transitional, and deep layers, respectively.
chitosan, and the freezing treatment was crucial [34–36]. In this section, chitin and chitosan
physical hydrogels based on novel alkali/urea solvent system differing from traditional
acidic solvent system are introduced and discussed.
The dissolution process of chitin in NaOH/urea solvents was illustrated in four main
steps: (a) swelling of chitin immersed in NaOH/urea aqueous solution at ambient tempera-
ture; (b) entry of water molecules into the chitin molecular chain assisted by NaOH;
(c) breakage of the inter‐hydrogen and intra‐hydrogen bonds caused by the freezing and
expansion of water at the freezing point; and (d) solubility promotion of chitin chains
(Figure 5.5a–d) [33]. For the purpose of figuring out the conformation of chitin in NaOH/
urea aqueous solution, a variety of measurements were conducted to study the intramolecu-
lar and intermolecular interactions of chitin [37]. The results revealed that chitin presented
an extended worm‐like chain conformation in the NaOH/urea aqueous solution, with a
sheath‐like structure consisting of NaOH and chitin forming a hydrogen‐bonded complex
surrounded by the urea hydrates (Figure 5.5e).
+ + +
+
+
+ + +
(c) (d)
+ +
+ +
+ + +
+
+ + + + +
+ +
+ +
NaOH hydrate Urea hydrate
Chitin chain
Hydrogen bond Water
Figure 5.5 The diagrammatic illustration of the dissolution process of chitin, including: (a) chitin soaked in 8 wt% NaOH/4 wt% urea aqueous solution at ambi-
ent temperature; (b) water molecules entering the chitin molecular chain facilitated by NaOH; (c) water molecules freezing and expanding at the freezing tem-
perature, breaking the inter‐hydrogen and intra‐hydrogen bonds; (d) promoting solubility of the chitin [33]; and (e) a model description of a chitin complex chain
in NaOH/urea aqueous solution [37].
(a)
Step 1
+ +
Step 2
NHCOCH3 CH2OH
O O
NHCOCH3 CH2OH
Physically cross-linked domains
Chitin double-cross-linked hydrogel
Figure 5.6 Preparation and gross mechanical characterization of double‐cross‐linked (DC) chitin hydrogels. (a) Preparation of the DC chitin hydrogels. Step 1:
the chitin chains form covalent cross‐links induced by ECH that produce chemically cross‐linked domains; step 2: physically cross‐linked domains formed through
hydrogen bonding, hydrophobic interactions, and the formation of crystalline hydrates of α‐chitin are intertwined with the chemically cross‐linked domains to
create the DC chitin hydrogels. The red and blue parts represent the chemically and physically cross‐linked domains, respectively. Photographs of the DC chitin
hydrogels under (b) stretching, (c) compression, (d) knotting, and (e) wrapping. The inset shows a traditional Chinese knot [39].
Chitosan‐Based Hydrogels 107
into the mold and immersed in an aqueous solution of H2SO4/ethanol at 40–50°C for 24 h
to neutralize the alkali completely. Finally, the resultant chitosan hydrogels were thor-
oughly washed with tridistilled water to remove any residues. Compared with chitosan
hydrogels formed via the acidic system, chitosan hydrogels via the alkaline system exhib-
ited better mechanical properties, reflected in the high ultimate strength and energy absorp-
tion before breaking [43].
For in‐depth knowledge on the gelation mechanism, Wang et al. introduced a novel,
visualized approach to elaborate the mechanism of the gelation process. Tetraphenylethylene
(TPE), a typical aggregation‐induced emission fluorogen (AIE gens), was employed to
directly observe the transformation of network, since stronger fluorescent emission was
deemed to tighten network formation [44]. It was noted that the formation of chitosan
hydrogel via LiOH/urea aqueous system was unique, with two distinct but integrated
stages: the thermal gelation stage and rinse stage (Figure 5.7). The thermal gelation stage
was driven by the elevated temperature, forming the embryonic structure and crystalline
regions, while the rinse stage was driven by the continuous change of system components,
greatly increasing intermolecular/intramolecular hydrogen bonds.
Various functional hydrogels were fabricated based on the LiOH/urea solvent system
[45–47]. Inspired by the bilayer structures of plant organs, Duan et al. constructed bio‐
hydrogel actuators from chitosan and cellulose/carboxymethylcellulose (CMC) solution in
an alkali/urea aqueous system utilizing ECH as a cross‐linker [46]. Strong electrostatic
attraction and chemical cross‐linking endowed tight adhesion between two layers and
excellent mechanical properties for carrying out rapid, reversible, and repeated self‐rolling
deformation actuated by pH‐triggered swelling/deswelling. The significant difference in
the swelling behavior between the positively charged chitosan and the negatively charged
cellulose/CMC layers generated enough force to actuate the performance of the hydrogels
as soft grippers, smart encapsulators, and bioinspired lenses, revealing potential applica-
tions in biorelated fields.
250 μm 250 μm
250 μm 25 μm
Figure 5.7 Confocal laser scanning fluorescence microscope images of the gelation process of tetraphe-
nylethylene‐chitosan (TPE‐CS). (a) Solution; (b) gel after thermal gelation; (c, d) hydrogel after rinse
procedure—schematic illustration of the formation of junction points of chitosan hydrogel in LiOH–urea
solution; (e) well‐dissolved solution; (f) formation of intermolecular/intramolecular hydrogen bonds of
chitosan, induced by heating; (g) formation of crystalline regions in chitosan gel; and (h) further formation
of hydrogen bonds of chitosan, induced by the removal of LiOH and urea [44].
108 Chitin and Chitosan: Properties and Applications
(a) Fe(III)-catechol
(b)
hydrogel
(coordinative
crosslinking)
Hydrogel
CAT-CHIT Drug loaded hydrogel injection
Healthy cell
Localized treatment Weak gel
Reduced toxicity Fe(III) Cancerous cell High solubility
High tumor suppression Dexorubicin Dead cell
Docetaxal Blood vasculature Chitosan PNIPAAm
Figure 5.8 (a) Schematic representation of the study showing the coordinatively cross‐linked catechol–
derived chitosan hydrogel induced by Fe (III)–catechol interactions for localized delivery of a combination
of drugs as effective anticancer therapy [55]; (b) schematic description on solubility and gelation behavior
of chitosan‐g‐PNIPAAm: copolymers with long side chains are viscous and less soluble at low tempera-
tures, but can form strong hydrogels at high temperatures, whereas copolymers with short side chains are
more soluble at low temperatures. However, they cannot form strong hydrogels at high temperatures [58].
Two different populations of catechols exist in the CAT‐Gel, where a major population
formed monocomplexes with Fe (III) ions and a minor population formed complexes of
higher coordination number with Fe (III) [55]. This combined population of the complexes
contributed to the requisite cohesive interactions needed for in situ gelation, and the revers-
ible nature of coordinate cross‐linking imparts favorable injectability nature to the resulting
CAT‐Gel.
from protonated amino groups to glycerol phosphate occurs, and consequently the forma-
tion of hydrogen bonds and hydrophobic interaction among chitosan chains is facilitated.
(a) O
OH (c) Self-healing chitosan hydrogel
O O
O O
O OH
O 1000
H HO
n
O
90-92 O
Low viscosity
NH2
100 Gʹ
Glycol chitosan DF-PEG Gʺ
10
Gʹ, Gʺ (Pa)
25ºC
1 ~220 s
OH OH 0.1
O O Plenty of time for
O O 0.01 operation at 25ºC
O OH
HO O HO
H
N x NH2 y 0.001
0 200 400 600 800 1000
x+y=n
*
Time (s)
10000 Gʹ
~1.5 kPa Gʺ
1000 Dynamic
(b) Self-healing hydrogel (injectable) 100 self-healing
Gʹ, Gʺ (Pa)
37ºC
10
1
0.1 Rapid gelation in 37ºC
26-gauge needle 0.01
1 cm
0 500 1000 1500
Time (s)
Figure 5.9 Preparation of DF‐PEG–GC injectable hydrogels. (a) Benzaldehydes at both ends of difunc-
tionalized PEG (DF‐PEG) cross‐linking with glycol chitosan to form injectable hydrogel; (b) the injectable
hydrogel can pass through a 26‐gauge (260 μm) needle without clogging; (c) the gelation time and modu-
lus (G’ and G”) of the injectable hydrogels at 25 and 37°C [67].
aqueous milieu and their advantages of greater stability, longer‐term storability, and lower
cytotoxicity, as compared to protein‐based components such as glucose oxidase and conca-
navalin A.
A series pH and glucose dual‐responsive injectable hydrogels were developed by Li
et al. through the cross‐linking of Schiff’s base and phenylboronate ester using phenylbo-
ronic‐modified chitosan (CSPBA) and oxidized dextran (Oxd), and water was the only
by‐product of the two cross‐linking reactions [84]. CSPBA and Oxd were used as starting
materials in this work, they were able to form covalently cross‐linked hydrogels through
imine bond and phenylboronate ester, and encapsulate drugs and cells with pH changing
from slightly acidic to physiological condition (Figure 5.10a). Oxd with aldehyde groups
and vicinal diol were selected as macromolecular cross‐linkers to form hydrogels with
CSPBA containing amino groups and PBAs. Hydrogels can form by the in situ cross‐
linking reaction between CSPBA and Oxd through two kinds of dynamic covalent bonds,
that is, the pH‐responsive imine bond by the reaction of amino between aldehydes, and the
glucose‐responsive phenylboronate ester by the combination of PBA with polyol. The rapid
gelation and biocompatible cross‐linking chemistry are appropriate for the incorporation of
drug molecules and cells by in situ gel formation.
(a)
HO
HO O O
O OH
HO
O
O OH
O NH2 CH2 CH COO O CO CHO
OH + + OHC CH2 CH2
O NH2
OH n n
NH
CO CSPBA PVA OHC-PEO-CHO
B(OH)2
H+/glucose OH–
–
Low pH lable Glucose responsive
covalent linker moiety
– OH
– OH
– N=C C=N
H H O
HO HO H2N –
– NH2 O
B B OH
– O
– OH
O N=C C=N
HO H H O
– –
– – B O
HO O B
OH OH
O N=C C=N
– H
H HO
HO NH2 H2N
Drug – Cell
(b)
OR OR
O O
O O
HO NH n HO NH2 m H O
O N O
R = H or CH2COONa
CM-Chitosan (CMC)
O
O
Oz
O
DA-PEG Hydrogel
(c)
H+ OH– H+ OH–
5 cycles
H O H+ O H+ O
N O N O NH2 O
OH– OH– OHC
Enamine Imine Amine + Aldehyde
Figure 5.10 (a) Illustrative formation of the CSPBA/PVA/OHC–PEO–CHO hydrogel [85]; (b) schematic
representation of DA‐PEG/CMC hydrogels by cross‐linking CMC with DA‐PEG; and (c) pH sensitivity of
the DA‐PEG/CMC hydrogel (upper layer) and its mechanism (lower layer) [85].
114 Chitin and Chitosan: Properties and Applications
(a)
O
CH3
CH2 CH C NH CH
CH2OCH2COOH CH3
H O O (NIPAm)
OH H
APS, 70, 3h
H
H NH2 n
(CMCS-PNIPAm)
(CMCS) O O
Ring opening
O reaction
(GMA)
Photo-irradiation
I2959
(b)
VBL curing Riboflavin (RF)
O
N
NH
RF
N N O
OH
MeGC HO
OH
OH
MeGC
+
pH neutralization
Col II
+
– – –
– + – +
+ +– +–
–
–
CS +
MeGC/CS-L MeGC/CS-H
Figure 5.11 (a) Schematic illustration for synthetic route of CMCS‐PNIPAAm‐GMA hydrogels [93].
(b) Design of visible blue light cross‐linkable chitosan/ECM composite hydrogels. See text for details [95].
116 Chitin and Chitosan: Properties and Applications
which revealing that photopolymerized MeGC hydrogels initiated with riboflavin possessed
the most favorable cell viability and mechanical properties for tissue engineering applica-
tions. This visible light initiating system was further utilized to generate injectable chitosan
hydrogels incorporated with Col II, manifesting increased chondrogenesis for cartilage
repair (Figure 5.11b) [95]. Apart from this, another blue‐light‐mediated cross‐linking
approach based on ruthenium (Ru)‐catalyzed photochemical cross‐linking process, which
could form covalent dityrosine bonds between phenolic groups by blue‐light illumination,
was also applicable for construction of chitosan‐based injectable hydrogels [96].
results in low stability, weak mechanical property, and fast degradation of the gelation
system, while covalently cross‐linked hydrogels are typically embedded with poor tough-
ness and unfavorable cytotoxicity. Thus, each individually fails to recapitulate biological
tissues’ resilience toward repeated loading. Addressing these limitations, double‐network
(DN) hydrogels, a subset of specifically structured interpenetrating networks (IPNs) that
exhibit resistance to mechanical failure, have evolved to produce desirable mechanical
properties for biomedical applications.
A variety of attempts have been made to construct chitosan‐based DN hydrogels [103–
112]. Typically, dynamic covalent bond, integrating both the stability of covalent bond
and the reversibility of noncovalent bond in one system, was employed to build injectable
CEC‐I‐OSA‐I‐ADH DN hydrogels composed of N‐carboxyethyl chitosan (CEC) with
medium molecular weight, adipic acid dihydrazide (ADH), oxidized sodium alginate
(OSA), and CEC with high molecular weight (Figure 5.12a,b) [103]. Imine bonds and
acylhydrazone bonds were two main dynamic covalent bonds that could undergo dynamic
reaction under physiological conditions, and the addition of high‐molecular‐weight CEC
could largely enhance the mechanical properties of the hydrogels. The homogeneously
mixed solution was able to form hydrogel within 20 s at room temperature, and it pos-
sessed the capability to be injected after gelation, since the broken hydrogel pieces
squeezed from the needle can self‐assemble and self‐heal into an integral hydrogel at the
target site, with more uniform distributions of cargos and more controllable placement of
hydrogels in vivo.
Additionally, an efficient approach to construct DN hydrogels is the selective and
sequential combination of physical, chemical, and enzymatic cross‐linking methods
with good compatibility. For instance, bioinspired ultratough hydrogels composed of 3,4‐
dihydroxiphenylalanine amino acid (DOPA)‐functionalized chitosan (DOPA‐CHT) were
generated via the covalent cross‐linking using genipin (GP) as a cytocompatible chemical
cross‐linker along with the noncovalent cross‐linking through coordination bonds among
the catechol groups present in DOPA‐CHT and Fe3+ ions (Figure 5.12c). And the DN
hydrogels were further produced by incorporating a small portion of medium‐molecular‐
weight CHT (MMW‐CHT) in the hydrogel precursor solution containing DOPA‐CHT, GP,
and Fe3+ ions. Except for the potential injectability, the introduction of dynamic ionic
interaction conferred an increased toughness, mechanical properties, and self‐healing
capacity, allowing for the recovery of initial mechanical properties in few minutes [104].
Injectable hydrogels are an important class of materials in light of the drive in the clinic
toward minimally invasive procedures. A variety of approaches have been utilized to
develop chitosan‐based injectable hydrogels, providing opportunities for more versatile
and safe means for constructing the hydrogels with varied and useful properties. Although
a number of chitosan‐based injectable hydrogel systems have proved to be promising can-
didates for biomedical applications, several significant challenges for future work remain
as follows: (i) efficient control of the suitable gelation time and stable gelation behaviors
for different applications; (ii) avoidance of the poor mechanical properties and inferior bulk
stability; (iii) minimization on the cytotoxicity induced by unreacted small molecules,
unfriendly fabrication process, and severe cross‐linking simulation; (iv) maintenance of the
balance between the bulk stability and appropriate degradation behaviors; and (v) explora-
tion on hydrogels with multifunctionality such as self‐healing properties, electroconducti-
bility, magnetism, and shape memory capacities. Therefore, further improvements in
(a) (c)
DN DN
OSA CEC-l-OSA-l-ADH DC DC
CEC+ADH Hydrogel
OH OH
O O OH
O NH
RT, < 20s HO NH HO NH
O
n OH
(PBS 7.0) H N
(b) DOPA-CHT
O
HO OH
O O
O Fe O
OH OH
HO OH
i) O O
H O NH
N
O HH N
GP ii) H OH
H O
CH HO
O O H HH
H
O O HO
– – NH
Cl Cl NH
H H HO
(CEC) Cl3Fe.6H2O H
Fe H HN
O
Cl
–
O
iii) H O
H H
H H HO H N
O
(OSA) HO
OH OH
OH OH H
O O PDMS NH
NH
N
O O iv)
(ADH) HO NH x HO NH y
O
H
NH
NH
C O
Figure 5.12 Synthesis scheme of the CEC‐I‐OSA‐I‐ADH hydrogels. (a) The photographs of before and after gelation of CEC‐I‐OSA‐I‐ADH hydrogel (R = 0.5) in
PBS (pH 7). (b) Chemical structures of the CEC‐I‐OSA‐I‐ADH hydrogel obtained by condensation reaction of the aldehyde groups (from OSA) with the amino
groups (from CEC) and hydrazides (from ADH), resulting in dynamic imine and acylhydrazone bonds, respectively [103]. (c) Schematic representation of the
method developed to fabricate the DC and DN hydrogels. Besides the DOPA‐CHT, GP, and Fe3+ ions, DN hydrogels are also composed of MMW‐CHT. The inset
depicts a picture of the obtained hydrogels (DC and DN showed a similar appearance). Two cross‐linking processes were employed to produce the hydrogels,
namely, a covalent cross‐linking using GP and a physical cross‐linking through coordination bonds in presence of Fe3+ ions. Therefore, DC hydrogels can be
composed of bis‐ and tris‐complexes Fe: (i) DOPA‐CHT or/and covalent bonds between (ii) two DOPA‐CHT chains. Additionally, DN hydrogel can also establish
covalent bonds (iii) between a chain of DOPA‐CHT and a chain of MMW‐CHT or/and (iv) between two chains of MMW‐CHT [104]. More details are presented in
the text.
Chitosan‐Based Hydrogels 119
Figure 5.13 The self‐healing process of a skin‐inspired chitosan hydrogel after being cut into two halves
and then brought together again with intervals of 30 s [118].
120 Chitin and Chitosan: Properties and Applications
Additionally, catechol and ferric ions have also been widely applied in the formation of
self‐healing. Due to the high grafting ratio of catechol, these chitosan derivatives could
form gels even in acidic medium upon the addition of Fe(III) and Cu(II) ions as cross‐
linkers [119]. A novel catechol‐modified N‐(furfural) chitosan‐based hydrogel was formed
by Diels–Alder click reaction and coordination bonds [120], which retains great
mechanical properties as well as excellent self‐healing performance. A mussel‐inspired
DN was synthesized by incorporating a small fraction of chitosan in the former hydrogel
precursor solution containing DOPA‐CHT (a chitosan derivative functionalized with cat-
echol groups), genipin (GP), and Fe3+ ions [104]. The prepared hydrogel was cleaved in
half and then immediately brought into contact. Five minutes after this operation, the DN
product had completely recovered without signs of a cut.
Cut
Chitosan Fe3+ KPS
Free radical
polymerization
Self-healing
Figure 5.14 Photographs demonstrating the electrical conductivity and self‐healing property of the dual‐
network cross‐linked CS‐PAA‐Fe(III) hydrogel [121].
Chitosan‐Based Hydrogels 121
ithout external stimulus, which had potential applications in the field of artificial skin or
w
wearable devices.
(a) (b)
A-1) Injected A-2) 0h 0.75 h 2h
United gel
Figure 5.15 Self‐healing phenomena of the CS‐PEG hydrogel system. (a) Self‐healing process after injec-
tion (blue: self‐healing hydrogel, red: gelatin hydrogel) [128]. (b) Appearance of self‐healing hydrogels
versus time (5 wt% gelatin gel as control) [127].
122 Chitin and Chitosan: Properties and Applications
small bioactive molecules [127], or drugs [129]. Additionally, the gels showed great advan-
tages in 3D cell culture and injection cell therapy [130], which were an excellent material
to encapsulate neural stem cells (NSCs) for rescuing the central nervous system [67].
Further, inorganic particles (e.g. Fe3O4 and SiO2) [131, 132] had been incorporated into this
system to obtain better functions.
Water‐soluble derivatives of chitosan have also been developed to form self‐healing
hydrogels [83, 133]. Further, the addition of natural nontoxic cross‐linking agents to gelate
chitosan became a safer and environmentally friendly way [134, 135]. Chen et al. [74]
formed a bioadhesive and self‐healing hydrogel based on N‐carboxyethyl chitosan and
sodium alginate dialdehyde, with the introduction of carbon quantum dots (C‐dots), which
performed the dual role of bioimaging and reactive oxygen species (ROS) scavenging.
Recently, zinc phthalocyanine tetra‐aldehyde (ZnPcTa), a good photosensitizer and gelator,
has been used to develop self‐healing chitosan‐based hydrogels with porous nanostructures
(Figure 5.16) [136]. The crack (width, 1 mm) of hydrogels made by a razor blade gradually
healed after 10 min and completely disappeared after 15 min. Besides, imine bonds broke
quickly due to the more acidic tumor environment (pH 6.5–6.8), leading to the rapid release
of ZnPcTa for localized photodynamic treatment [137]. The self‐healing chitosan‐based
hydrogels could also be applied as hemostatic materials [138], or utilized in magnetic reso-
nance imaging (MRI) in vivo [139].
CN O
HO OHO O
n HO
+ N NH2 NH2
1 2 O
CHO
O H NO2 O
N
(cut)
N N
N Zn N
O N N
N
O
O CN O
3 OHC
H CN OHC H2N N
H2N OHO
OH O
CHO OH O
n
O HO HO
MW O
HO
10 min
N
N N
N Zn N
N N OH OH OH
O O O O O
N HO
O HO HO HO
NH2 NH2 NH2
O n
OHC
CHO 20 min 30 min
Figure 5.16 Novel self‐healing chitosan hydrogel cross‐linked by zinc phthalocyanine tetra‐aldehyde (ZnPcTa). (a) Chemical structure of in situ formation of
ZnPcTa‐conjugated chitosan through Schiff base reaction [137]. (b) Digital photographs of flexibility and self‐healing process of this hydrogel nanocomposite
[136].
(a) (b)
OCOOH OH OH
O O O O O O O
HO HO HO
N NH2 NH
O
Self-healing
O
O
O
N O
O
OH
O HN O
n
OH
2C
OH
2 H
O
OH C
OH
O HN
O
OH
OH
O OH O
N
O
O O
O
O Acid-resistant
N H2N HN
OH OH OH
O O O O O O O
HO HO HOOCO
Figure 5.17 Chitosan hydrogel with double cross‐linked networks (DN) by combining reversible imine bonds with Diels–Alder (DA) reaction [146].
(a) Chemical structure of the DN hydrogel. (b) Gelation behavior, acid resistance, and self‐healing properties of the DN hydrogel.
Chitosan‐Based Hydrogels 125
at
ge i
A
Shape fixation
o
am
Deformation
on
dr
M
D
Hy
bo
A
Supr
shape mem
nds
A
ar
D
or
l
pramolecu
y
H o s t- g u e s t i n
hydrogels
b on d s
SSMHs
Su
ase
N
nds
fb
ter
h if
a
bo
c ti
Sc
ns
o
nt
Bo d s
le
ro n a a
te ester bon
ov
ic c
Dy n a m
Permanent crosslinks Reversible interactions
Figure 5.18 (a) Mechanism of shape memory hydrogels. (b) Strategies employed to construct shape
memory hydrogels (SMHs) based on reversible interactions [40].
reached the maximum of 97.6% when the PLGA/chitosan ratio was 2.5:1. Besides,
the remote high‐intensity focused ultrasound (HIFU) activation could be utilized to
simultaneously control the shape recovery process and the drug release behavior of
chitosan‐functionalized lysozyme‐loaded PLGA microspheres via variation in irradiation
power and duration time (Figure 5.19). A retarded degradation was observed from the com-
posite microspheres in contrast to the pristine PLGA due to the effective acid‐neutralizing
effect of chitosan on acid metabolites of PLGA. The underlying mechanism for synchro-
nous effects by this indirect trigger was illustrated as that the absorbed ultrasound energy
transformed into heat energy, leading to a rapid rise in temperature above the Ttrans, and thus
enhancing the diffusion coefficient of the SMP, resulting in rapid release of the entrapped
drug molecules.
(a)
Lyz
CTS 0.5% PVA aqueous
Lysozyme solution
aqueous solution
PLGA solution
+CTS
T>Tg
Hot-pressing
Ultrasound Ultrasound
Drug release
Shape recovery and drug release
(b)
0s
Initial shape
30 s 60 s
90 s 120 s
Figure 5.19 Preparation procedures and shape recovery effect of chitosan‐functionalized poly(lactic‐
co‐glycolic acid) (PLGA) microspheres. (a) Schematic illustration of the experimental procedures for
chitosan‐functionalized PLGA microspheres with ultrasound‐modulated shape memory and payload
release effects. (b) Photographs showing the shape recovery effect during four cycles of HIFU irradiation
at 30 s per cycle in panel (b); the sample was immersed in the water bath of the HIFU reactor at 37°C
throughout the examination [154].
(a)
Hand deformation Shape memory
Actuating layer
Self-deformation Shape memory
Memorizing layer
O NH O N O N O N O N
O NH H H H
H
H H H H
HN O N O N O
OH–
HN O N O N O
T>LCST
+ + + +
+ + + +
+ + + + + +
OH NH2 OH NH2
n O O HO O O HO O
= + = HO
O
O O O n
NH3+ NH
O NH2 OH
O
OH
n
= NH = microcrystal = H2O
O
(b)
Initial shape Deformed shape Memorized shape Recovered shape
ºC ºC ºC ºC
NaOH HCl
2.0
Curvature
Curvature
Figure 5.20 Mechanism and shape recovery behavior of self‐deformed hydrogel. (a) Schematic illustra-
tion and mechanism for novel self‐deformed shape memory process. (b) The self‐deformed hydrogel
reversibly self‐deformed between 20 and 45°C. The deformed shape of the hydrogel is partially maintained
in NaOH solution and completely recovered in HCl solution. (c–e) The curvature of a bilayer strip against
time in 45°C KNO3 aqueous solution, 20°C NaOH solution, and 20°C HCl solution [155].
cross‐linked and irreversible network via the free radical polymerization of acrylamide
(AAm). Owning to the excellent chelating capacity of chitosan with various metal cations
such as Fe3+, Ag+, Ni2+, etc., the coordination effects could also stabilize the temporary
shape and mold the hydrogels.
The PAAm/CS‐Oxd hydrogel was authenticated to possess triple shape memory behav-
ior, as shown in Figure 5.21. A hydrogel strip dyed in pink and green was first immersed in
the alkaline buffer solutions (pH 8) to fix temporary shape I based on the formation of
Schiff base bonds (Figure 5.21c). Then the hydrogel strip was deformed to temporary
Oxalate or H2SO4
H+ EDTA
Cu2+ Zn2+ Original shape Temporary Temporary
shape I shape II
Ni2+
OH– Ag+
H+ EDTA
(b)
Oxalate or H2SO4
Ox O
ala Ag+ SO
4 HO
HN
HO H 2N
te H2 O OH O OH
O O O
O OH O O OH O OH NH2 OH NH2
H 2N
HO
H2N HO
n
O
O HO O O HO O PAAm
O O HO O
HO O
NH2 NH n
OH O OH
OH OH
– O
– /Br
ED O O HO HO
H 2N C N
Cl TA HO HN OH
O
O O O O OH O O O
OH O OH O O OH O
O O n
H 2N HO H2N HO
Figure 5.21 The process and mechanism of triple shape memory and shape recovery processes. (a) The shape memory behavior based on the coordination of
chitosan with various metal ions. All the dashed lines indicate that the temporary shape hydrogel will be recovered in the EDTA aqueous solution. (b) Shape
memory behavior based on CS–Ag+ coordination. (c) The process and mechanism of the triple shape memory and shape recovery; the triple shape recovery could
be realized step by step or in one step [156].
Chitosan‐Based Hydrogels 131
shape II and transferred into the AgNO3 aqueous solution to fix temporary shape II via
metal coordination interaction. Finally, an “N”‐shaped hydrogel strip was obtained. After
being sequentially transferred into the aqueous solutions of Ethylenediaminetetraacetic
acid (EDTA) and acetic acid–sodium acetate buffer solutions (pH 3), the complex “N”
temporary shape would be gradually recovered to temporary shape I and then to the origi-
nal straight shape. Moreover, the complex “N” temporary shape could also be directly and
quickly recovered to the starting straight shape when using oxalate as the external stimulus.
This phenomenon was termed as triple shape memory capability ascribed to the dual attrib-
utes of oxalate acid both serving as a moderately strong acid and a good binding agent to
Ag+, causing simultaneous destruction on the two reversible interactions. The programma-
ble triple shape memory effect triggered by two different stimuli step by step, or a single
stimulus in one step, largely expanded the potential applications of reversible interaction‐
induced SMPs.
So far, various chitosan‐based hydrogels with fast shape memory/recovery rates or good
shape fixity/recovery ratios have been constructed. However, it is still challenging to com-
bine all these advantages into one single hydrogel system. Most of the previously reported
SMHs have poor mechanical properties, thus limiting their applications. Besides, most
investigations in this field are focused on dual SMHs. In order to meet the increasing
demands for versatile materials, it is of urgent need to develop more shape memory hydro-
gels with elegant triple/multi shape memory properties. Even though great efforts have
been primarily devoted to the preparation of novel SMHs in the last decades, less attention
has been given to the practical applications of SMHs, especially in the fields of biomedical
applications, such as drug delivery systems, tissue engineering, biosensing, etc. Considering
the excellent attributes of chitosan, SMHs with excellent biocompatibility can be con-
structed and be employed for in vivo studies. It is also quite promising to develop new types
of SMHs with multifunctionality, such as magnetism, luminescence, electrical conductiv-
ity, expansion–contraction, self‐healing properties, etc. Such supramolecular hydrogels
with various functions are expected to be the next generation of smart soft materials that
can function more closely with natural or biological systems.
(a) (b)
1 cm 1 cm
Figure 5.22 Photographs of a superabsorbent hydrogel in (a) dry state and (b) the swollen state [159].
these CSSH into four types according to the components of synthetic systems: (i) cross‐
linked chitosan (or their derivatives) hydrogels without synthetic polymers; (ii) chitosan
hydrogels prepared through graft copolymerization of vinyl monomers, such as AA or
acrylamide (AAm) on the polymer chains; (iii) chitosan‐based composite hydrogels
obtained via the incorporation of inorganic or organic substances; and (iv) pure chitosan
materials fabricated without any additives, such as initiators, vinyl monomers, or
cross‐linkers.
of around 1250 g/g of distilled water and 210 g/g of 0.1% (w/v) sodium chloride solution.
The new material with rapid and high water uptake could play a vital role in applications
that demand the quick absorption and slow release of water, such as agriculture.
The advantage of a chemical approach (cross‐linking through covalent bonds) is that the
hydrogel matrix remains stable over time. However, the agents used in the chemical cross‐
linking are often toxic and the residual cross‐linkers must be removed before using in
biomedical applications. Thus, physical cross‐linking is an efficient route to prepare hydro-
gels without using toxic chemical agents. A novel gellan gum–chitosan superabsorbent
hydrogel was constructed via the ionic bonds between chitosan and gellan gum [164]. The
best water absorption capacity (more than 218 g/g) was obtained and the fertilizer‐release
test showed that almost complete release occurred in approximately 8 h. Additionally, the
hydrogel showed excellent ability of water retention through the water evaporation analy-
sis. Therefore, this material could be applied in environment‐friendly agriculture success-
fully for controlling soil humidity and releasing the fertilizer.
1000
Repulsive
force
800
Repulsive
force
600
Q (g/g)
400
200
0 2 4 6 8 10 12 14
pH
Figure 5.23 Effects of pH of the solution on water absorption capacity of the polyampholyte superabsor-
bents [167].
ammonium chloride (DMDAAC) onto the chain of carboxymethyl chitosan [167]. The
highest swelling ratios in distilled water and normal saline water could be as high as 612
and 81 g/g, respectively. The swelling and shrinking of the products exhibited unique pH‐
dependent properties and showed peaks in swelling capacity at both acidic condition (pH
3.5) and basic condition (pH 9.5) (Figure 5.23), which made them good candidates as
potential drug carriers.
obtained at the optimum condition. Attapulgite contained a lot of cations and they were
easily ionized and dispersed into the superabsorbent network, which enhanced hydrophi-
licity of the composite and made it swell more. Zhang et al. [170] prepared a chitosan‐g‐
poly(acrylic acid)/attapulgite composite hydrogel with water absorbency of 159.6 g/g in
distilled water and 42.3 g/g in 0.9 wt% NaCl solution.
Wang et al. have applied several inorganic clay materials to form superabsorbent chi-
tosan‐based composites. A novel chitosan‐g‐poly(acrylic acid)/vermiculite composite was
prepared with a high water absorbency (585.3 g/g) and fast swelling rate [171]. Another
composite material was synthesized by replacing vermiculite with rectorite. It was found
that introducing 10 wt% of rectorite could enhance water absorbency and water retention
in the same time. The maximum water uptake capacity (415 g/g) was obtained under opti-
mum reaction condition.
optimal conditions were 1812 and 92.7 g/g in distilled water and 0.9 wt% NaCl solution,
respectively [176].
Nowadays, superabsorbent hydrogels formed by various materials and methods have
been applied in extensive technological applications, such as waste‐water treatment, per-
sonal hygiene, and agricultural and biomedical fields. However, some challenges still exist
as follows: (i) various reaction parameters have been investigated in order to form high
performance superabsorbent hydrogels. However, most studies have just drawn qualitative
conclusions, or their quantitative conclusions are not widely applicable. There is still a lack
of understanding regarding the relationship between the network structure and properties
of superabsorbent hydrogels; (ii) hydrogels with superabsorbent characteristics and excel-
lent mechanical properties at the same time are difficult to achieve; (iii) most of the supera-
bsorbent hydrogels absorb much less water in salt medium than in distilled water, which
exhibits poor salt‐resistance; (iv) hydrogels prepared through graft copolymerization of
vinyl monomers show excellent water uptake capacity, but the worse biodegradation prop-
erty; and (v) lowering the final cost production remains as a challenge for applications of
chitosan‐based superabsorbent hydrogels, because chitosan raw materials show higher cost
than other natural polysaccharides (e.g. cellulose and starch). More strategies should be
developed for the formation of environment‐friendly chitosan‐based superabsorbent poly-
mers with other functional properties, such as good mechanical strength, self‐healing, and
responsive property, which have potentials to become substitutes for synthetic commercial
superabsorbent polymers.
5.8 Outlook
Chitosan is chemically similar to glycosaminoglycans, and regarded as an excellent candi-
date matrix for hydrogels, since it is nontoxic, stable, biodegradable, biocompatible, and
can be sterilized. Moreover, the free amino and hydroxyl groups in the polymer chains
endow chitosan with covalent binding sites for versatile small molecules and macromole-
cules. Two completely opposite solvent systems in nature, acidic solution and alkali/urea
solvent, have been utilized to prepare chitosan‐based hydrogels with unique structures and
enhanced mechanical performance. Based on their intrinsic pH‐sensitivity, chitosan‐based
hydrogels that exhibit multifunctional properties, such as injectability, self‐healing, shape
memory, and superabsorbent performance, have been widely synthesized and discussed,
showing great potential in various fields, especially involving biomedical applications.
Further progresses are needed to obtain chitosan‐based hydrogels with other excellent
functions such as being conductive, stimuli‐responsive, luminescent, ultratough, and highly
stretchable.
References
1. Hennink, W.E. and C.F. van Nostrum, Novel crosslinking methods to design hydrogels. Advanced
Drug Delivery Reviews, 2012. 64: p. 223–236.
2. Cao, L. et al., An injectable hydrogel formed by in situ cross‐linking of glycol chitosan and multi‐
benzaldehyde functionalized PEG analogues for cartilage tissue engineering. Journal of Materials
Chemistry B, 2015. 3(7): p. 1268–1280.
3. Racine, L., I. Texier, and R. Auzély‐Velty, Chitosan‐based hydrogels: recent design concepts to tailor
properties and functions. Polymer International, 2017. 66(7): p. 981–998.
4. Rinaudo, M., Chitin and chitosan: Properties and applications. Progress in Polymer Science, 2006.
31(7): p. 603–632.
5. Bhattarai, N., J. Gunn, and M. Zhang, Chitosan‐based hydrogels for controlled, localized drug deliv-
ery. Advanced Drug Delivery Reviews, 2010. 62(1): p. 83–99.
6. Sahoo, D. et al., Chitosan: A new versatile bio‐polymer for various applications. Designed Monomers
and Polymers, 2012. 12(5): p. 377–404.
7. Xu, H. and S. Matysiak, Effect of pH on chitosan hydrogel polymer network structure. Chemical
Communication, 2017. 53(53): p. 7373–7376.
8. Ma, S. et al., Structural hydrogels. Polymer, 2016. 98: p. 516–535.
9. Morrow, B.H., G.F. Payne, and J. Shen, pH‐Responsive self‐assembly of polysaccharide through a
rugged energy landscape. Journal of American Chemical Society, 2015. 137(40): p. 13024–13030.
10. Packter, A., The Liesegang phenomenon II. Addition of protecting agents. Journal of Colloid Science,
1956. 11(1): p. 96–106.
11. Hu, Q.L. et al., Studies on chitosan rods prepared by in situ precipitation method. Chemical Journal of
Chinese Universities‐Chinese, 2003. 24(03): p. 528–531.
12. Nie, J. et al., Orientation in multi‐layer chitosan hydrogel: morphology, mechanism, and design prin-
ciple. Scientific Reports, 2015. 5: p. 7635.
13. Nie, J., Z. Wang, and Q. Hu, Chitosan hydrogel structure modulated by metal ions. Scientific Reports,
2016. 6: p. 36005.
14. Nie, J. et al., Biomimetic multi‐layered hollow chitosan–tripolyphosphate rod with excellent mechani-
cal performance. RSC Advances, 2015. 5(47): p. 37346–37352.
15. Sereni, N. et al., Dynamic structuration of physical chitosan hydrogels. Langmuir, 2017. 33(44):
p. 12697–12707.
16. Ma, H. et al., Formation of multi‐layered chitosan honeycomb spheres via breath‐figure‐like approach
in combination with co‐precipitation processing. Materials Letters, 2018. 211: p. 91–95.
17. Ladet, S., L. David, and A. Domard, Multi‐membrane hydrogels. Nature, 2008. 452(7183): p. 76–79.
18. Li, B. et al., Formation of concentric multilayers in a chitosan hydrogel inspired by Liesegang ring
phenomena. Journal of Biomaterials Science. Polymer Edition, 2011. 22(17): p. 2295–2304.
19. Xiong, Y. et al., Compartmentalized multilayer hydrogel formation using a stimulus‐responsive self‐
assembling polysaccharide. ACS Applied Materials & Interfaces, 2014. 6(4): p. 2948–2957.
20. Duan, J. et al., Versatile fabrication of arbitrarily shaped multi‐membrane hydrogels suitable for bio-
medical applications. Journal of Materials Chemistry B, 2013. 1(4): p. 485–492.
21. Zhao, P. et al., Electrochemical deposition to construct a nature inspired multilayer chitosan/layered
double hydroxides hybrid gel for stimuli responsive release of protein. Journal of Materials Chemistry
B, 2015. 3(38): p. 7577–7584.
22. Yan, K. et al., Coding for hydrogel organization through signal guided self‐assembly. Soft Matter,
2014. 10(3): p. 465–469.
23. Yan, K. et al., Electrical programming of soft matter: Using temporally varying electrical inputs to
spatially control self‐assembly. Biomacromolecules, 2018. 19(2): p. 364–373.
24. Yan, K. et al., Electro‐molecular assembly: Electrical writing of information into an erasable polysac-
charide medium. ACS Applied Materials & Interfaces, 2016. 8(30): p. 19780–19786.
25. Choi, J. et al., Regulation of cell proliferation by multi‐layered phospholipid polymer hydrogel coat-
ings through controlled release of paclitaxel. Biomaterials, 2012. 33(3): p. 954–961.
26. Yoo, C.Y., J.S. Seong, and S.N. Park, Preparation of novel capsosome with liposomal core by layer‐by‐
Layer self‐assembly of sodium hyaluronate and chitosan. Colloids and Surface B‐Biointerfaces, 2016.
144: p. 99–107.
27. Han, F. et al., Photocrosslinked layered gelatin‐chitosan hydrogel with graded compositions for
osteochondral defect repair. Journal of Materials Science‐Materials in Medicine, 2015. 26(4): p. 160.
28. He, M. et al., Fast contact of solid‐liquid interface created high strength multi‐layered cellulose hydro-
gels with controllable size. ACS Applied Materials & Interfaces, 2014. 6(3): p. 1872–1878.
29. Lin, N. et al., Biocompatible double‐membrane hydrogels from cationic cellulose nanocrystals and
anionic alginate as complexing drugs codelivery. ACS Applied Materials & Interfaces, 2016. 8(11):
p. 6880–6889.
30. Faivre, J. et al., Bioinspired microstructures of chitosan hydrogel provide enhanced wear protection.
Soft Matter, 2018. 14(11): p. 2068–2076.
31. He, H. et al., Reversible programing of soft matter with reconfigurable mechanical properties.
Advanced Functional Materials, 2017. 27(13): p. 1605665.
32. Zhou, J.P. and L.N. Zhang, Solubility of cellulose in NaOH urea aqueous solution. Polymer Journal,
2000. 32(10): p. 866–870.
33. Hu, X. et al., Solubility and property of chitin in NaOH/urea aqueous solution. Carbohydrate Polymers,
2007. 70(4): p. 451–458.
34. Fan, M. and Q. Hu, Chitosan‐LiOH‐urea aqueous solution–a novel water‐based system for chitosan
processing. Carbohydrate Polymers, 2009. 344(7): p. 944–947.
35. Fan, M., Q. Hu, and K. Shen, Preparation and structure of chitosan soluble in wide pH range.
Carbohydrate Polymers, 2009. 78(1): p. 66–71.
36. Fan, M. and Q. Hu, Superadsorption of LiOH solution on chitosan as a new type of solvent for chitosan
by freezing/blasting. Carbohydrate Polymers, 2013. 94(1): p. 430–435.
37. Fang, Y. et al., Intermolecular interaction and the extended wormlike chain conformation of chitin in
NaOH/urea aqueous solution. Biomacromolecules, 2015. 16(4): p. 1410–1417.
38. Hu, X. et al., Rheological behaviour of chitin in NaOH/urea aqueous solution. Carbohydrate Polymers,
2011. 83(3): p. 1128–1133.
39. Xu, D. et al., High‐flexibility, high‐toughness double‐cross‐linked chitin hydrogels by sequential
chemical and physical cross‐linkings. Advanced Materials, 2016. 28(28): p. 5844–5849.
40. Lu, W. et al., Supramolecular shape memory hydrogels: A new bridge between stimuli‐responsive
polymers and supramolecular chemistry. Chemical Society Reviews, 2017. 46(5): p. 1284–1294.
41. Xu, H. et al., Green fabrication of amphiphilic quaternized beta‐chitin derivatives with excellent
biocompatibility and antibacterial activities for wound healing. Advanced Materials, 2018. 30(29):
p. 1801100.
42. Sun, Y. et al., High‐strength chitosan hydrogels prepared from LiOH/Urea solvent system. Chemistry
Letters, 2013. 42(8): p. 838–840.
43. Nie, J., Z. Wang, and Q. Hu, Difference between chitosan hydrogels via alkaline and acidic solvent
systems. Scientific Reports, 2016. 6: p. 36053.
44. Wang, Z. et al., Gelation process visualized by aggregation‐induced emission fluorogens. Nature
Communications, 2016. 7: p. 12033.
45. Nie, J. et al., High strength chitosan rod prepared via LiOH/urea solvent through centrifugation
induced orientation processing. RSC Advances, 2015. 5(83): p. 68243–68250.
46. Duan, J. et al., Bilayer hydrogel actuators with tight interfacial adhesion fully constructed from natural
polysaccharides. Soft Matter, 2017. 13(2): p. 345–354.
47. Ding, B. et al., Tough and cell‐compatible chitosan physical hydrogels for mouse bone mesenchymal
stem cells in vitro. ACS Applied Materials & Interfaces, 2016. 8(30): p. 19739–19746.
48. Guvendiren, M., H.D. Lu, and J.A. Burdick, Shear‐thinning hydrogels for biomedical applications.
Soft Matter, 2012. 8(2): p. 260–272.
49. Yu, L. and J. Ding, Injectable hydrogels as unique biomedical materials. Chemical Society Reviews,
2008. 37(8): p. 1473–1481.
50. Overstreet, D.J. et al., Injectable hydrogels. Journal of Polymer Science Part B: Polymer Physics,
2012. 50(13): p. 881–903.
51. Wang, K., G. Buschle‐Diller, and R.D.K. Misra, Chitosan‐based injectable hydrogels for biomedical
applications. Materials Technology, 2016. 30(sup8, part B4): p. 1–9.
52. Ta, H.T., C.R. Dass, and D.E. Dunstan, Injectable chitosan hydrogels for localised cancer therapy.
Journal of Controlled Release, 2008. 126(3): p. 205–216.
53. Fujita, M. et al., Vascularization in vivo caused by the controlled release of fibroblast growth
factor‐2 from an injectable chitosan/non‐anticoagulant heparin hydrogel. Biomaterials, 2004. 25(4):
p. 699–706.
54. Oliveira, M.B., H.X.S. Bastos, and J.F. Mano, Sequentially moldable and bondable four‐dimensional
hydrogels compatible with cell encapsulation. Biomacromolecules, 2018. 19(7): p. 2742–2749
55. Yavvari, P.S. et al., Injectable, self‐healing chimeric catechol‐Fe(III) hydrogel for localized combina-
tion cancer therapy. ACS Biomaterials Science & Engineering, 2017. 3(12): p. 3404–3413.
56. Ji, D.Y. et al., A novel injectable chitosan/polyglutamate polyelectrolyte complex hydrogel with
hydroxyapatite for soft‐tissue augmentation. Carbohydrate Polymers, 2012. 89(4): p. 1123–1130.
57. Chenite, A. et al., Novel injectable neutral solutions of chitosan form biodegradable gels in situ.
Biomaterials, 2000. 21(21): p. 2155–2161.
58. Mellati, A. et al., A biodegradable thermosensitive hydrogel with tuneable properties for mimicking
three‐dimensional microenvironments of stem cells. RSC Advances, 2014. 4(109): p. 63951–63961.
59. Chiu, Y.L. et al., pH‐triggered injectable hydrogels prepared from aqueous N‐palmitoyl chitosan: In
vitro characteristics and in vivo biocompatibility. Biomaterials, 2009. 30(28): p. 4877–4888.
60. Mahanta, A.K., S. Senapati, and P. Maiti, A polyurethane–chitosan brush as an injectable hydrogel for
controlled drug delivery and tissue engineering. Polymer Chemistry, 2017. 8(40): p. 6233–6249.
61. Wei, C.Z. et al., A thermosensitive chitosan‐based hydrogel barrier for post‐operative adhesions’ pre-
vention. Biomaterials, 2009. 30(29): p. 5534–5540.
62. Zhang, E. et al., Thermoresponsive polysaccharide‐based composite hydrogel with antibacterial and
healing‐promoting activities for preventing recurrent adhesion after adhesiolysis. Acta Biomaterialia,
2018. 74: p. 439–453.
63. Chen, J.‐P. and T.‐H. Cheng, Preparation and evaluation of thermo‐reversible copolymer hydrogels
containing chitosan and hyaluronic acid as injectable cell carriers. Polymer, 2009. 50(1):
p. 107–116.
64. Wu, S.W. et al., Strengthening injectable thermo‐sensitive NIPAAm‐g‐chitosan hydrogels using chem-
ical cross‐linking of disulfide bonds as scaffolds for tissue engineering. Carbohydrate Polymers, 2018.
192: p. 308–316.
65. Park, K.M. et al., Thermosensitive chitosan‐pluronic hydrogel as an injectable cell delivery carrier for
cartilage regeneration. Acta Biomaterialia, 2009. 5(6): p. 1956–1965.
66. Ryu, J.H. et al., Catechol‐functionalized chitosan/pluronic hydrogels for tissue adhesives and hemo-
static materials. Biomacromolecules, 2011. 12(7): p. 2653–2659.
67. Tseng, T.C. et al., An injectable, self‐healing hydrogel to repair the central nervous system. Advanced
Materials, 2015. 27(23): p. 3518–3524.
68. Tan, H. et al., Injectable in situ forming biodegradable chitosan‐hyaluronic acid based hydrogels for
cartilage tissue engineering. Biomaterials, 2009. 30(13): p. 2499–2506.
69. Li, L. et al., Biodegradable and injectable in situ cross‐linking chitosan‐hyaluronic acid based hydro-
gels for postoperative adhesion prevention. Biomaterials, 2014. 35(12): p. 3903–3917.
70. Kim, D.Y. et al., Injectable hydrogels prepared from partially oxidized hyaluronate and glycol chitosan
for chondrocyte encapsulation. Carbohydrate Polymers, 2017. 157: p. 1281–1287.
71. Sun, J. et al., Covalently crosslinked hyaluronic acid‐chitosan hydrogel containing dexamethasone as
an injectable scaffold for soft tissue engineering. Journal of Applied Polymer Science, 2013. 129(2): p.
682–688.
72. Shi, J. et al., Schiff based injectable hydrogel for in situ pH‐triggered delivery of doxorubicin for breast
tumor treatment. Polymer Chemistry, 2014. 5(21): p. 6180–6189.
73. Wan, W. et al., BMSCs laden injectable amino‐diethoxypropane modified alginate‐chitosan hydrogel
for hyaline cartilage reconstruction. Journal of Materials Chemistry B, 2015. 3(9): p. 1990–2005.
74. Chen, J. et al., A reloadable self‐healing hydrogel enabling diffusive transport of C‐dots across gel‐gel
interface for scavenging reactive oxygen species. Advanced Healthcare Materials, 2017. 6(21):
p. 1700746.
75. Zhang, H. et al., Delivery of rosiglitazone from an injectable triple interpenetrating network hydrogel
composed of naturally derived materials. Biomaterials, 2011. 32(3): p. 890–898.
76. Xiong, L. et al., An injectable drug‐loaded hydrogel based on a supramolecular polymeric prodrug.
Chemical Communication, 2015. 51(78): p. 14644–14647.
77. Hoque, J. et al., Dual function injectable hydrogel for controlled release of antibiotic and local antibac-
terial therapy. Biomacromolecules, 2018. 19(2): p. 267–278.
78. Lu, S. et al., Injectable and self‐healing carbohydrate‐based hydrogel for cell encapsulation. ACS
Applied Materials & Interfaces, 2015. 7(23): p. 13029–13037.
79. Huang, J. et al., Novel in situ forming hydrogel based on xanthan and chitosan re‐gelifying in liquids
for local drug delivery. Carbohydrate Polymers, 2018. 186: p. 54–63.
80. Song, L. et al., Peritoneal adhesion prevention with a biodegradable and injectable N,O‐carboxymethyl
chitosan‐aldehyde hyaluronic acid hydrogel in a rat repeated‐injury model. Scientific Reports, 2016. 6:
p. 37600.
81. Deng, Y. et al., Injectable in situ cross‐linking chitosan‐hyaluronic acid based hydrogels for abdominal
tissue regeneration. Scientific Reports, 2017. 7(1): p. 2699.
82. Xia, L.Y. et al., Enhanced fluorescence emission and singlet oxygen generation of photosensitizers
embedded in injectable hydrogels for imaging‐guided photodynamic cancer therapy.
Biomacromolecules, 2017. 18(10): p. 3073–3081.
83. Khan, M. et al., Composite hydrogels using bioinspired approach with in situ fast gelation and self‐
healing ability as future injectable biomaterial. ACS Applied Materials & Interfaces, 2018. 10(14):
p. 11950–11960.
84. Li, J. et al., pH and glucose dually responsive injectable hydrogel prepared by in situ crosslinking of
phenylboronic modified chitosan and oxidized dextran. Journal of Polymer Science Part A: Polymer
Chemistry, 2015. 53(10): p. 1235–1244.
85. Zhao, L. et al., pH and glucose dual‐responsive injectable hydrogels with insulin and fibroblasts as
bioactive dressings for diabetic wound healing. ACS Applied Materials & Interfaces, 2017. 9(43):
p. 37563–37574.
86. Huang, J. and X. Jiang, Injectable and degradable pH‐responsive hydrogels via spontaneous amino‐
yne click reaction. ACS Applied Materials & Interfaces, 2018. 10(1): p. 361–370.
87. Gao, X. et al., A water‐soluble photocrosslinkable chitosan derivative prepared by Michael‐addition
reaction as a precursor for injectable hydrogel. Carbohydrate Polymers, 2010. 79(3): p. 507–512.
88. Teng, D.‐y. et al., Synthesis and characterization of in situ cross‐linked hydrogel based on self‐assem-
bly of thiol‐modified chitosan with PEG diacrylate using Michael type addition. Polymer, 2010. 51(3):
p. 639–646.
89. Fan, M. et al., Cytocompatible in situ forming chitosan/hyaluronan hydrogels via a metal‐free click
chemistry for soft tissue engineering. Acta Biomaterialia, 2015. 20: p. 60–68.
90. Li, B. et al., Hydrosoluble, UV‐crosslinkable and injectable chitosan for patterned cell‐laden microgel
and rapid transdermal curing hydrogel in vivo. Acta Biomaterialia, 2015. 22: p. 59–69.
91. Zhong, C. et al., Synthesis, characterization and cytotoxicity of photo‐crosslinked maleic chitosan‐
polyethylene glycol diacrylate hybrid hydrogels. Acta Biomaterialia, 2010. 6(10): p. 3908–3918.
92. Kang, W. et al., Photocrosslinked methacrylated carboxymethyl chitin hydrogels with tunable degra-
dation and mechanical behavior. Carbohydrate Polymers, 2017. 160: p. 18–25.
93. Zhang, L. et al., Cytocompatible injectable carboxymethyl chitosan/N‐isopropylacrylamide hydrogels
for localized drug delivery. Carbohydrate Polymers, 2014. 103: p. 110–118.
94. Hu, J. et al., Visible light crosslinkable chitosan hydrogels for tissue engineering. Acta Biomaterialia,
2012. 8(5): p. 1730–1738.
95. Choi, B. et al., Cartilaginous extracellular matrix‐modified chitosan hydrogels for cartilage tissue engi-
neering. ACS Applied Materials & Interfaces, 2014. 6(22): p. 20110–20121.
96. Lu, M. et al., Fabrication of photo‐crosslinkable glycol chitosan hydrogel as a tissue adhesive.
Carbohydrate Polymers, 2018. 181: p. 668–674.
97. Teixeira, L.S. et al., Enzyme‐catalyzed crosslinkable hydrogels: Emerging strategies for tissue engi-
neering. Biomaterials, 2012. 33(5): p. 1281–1290.
98. Jin, R. et al., Injectable chitosan‐based hydrogels for cartilage tissue engineering. Biomaterials, 2009.
30(13): p. 2544–2551.
99. Jin, R., C. Lin, and A. Cao, Enzyme‐mediated fast injectable hydrogels based on chitosan–glycolic
acid/tyrosine: Preparation, characterization, and chondrocyte culture. Polymer Chemistry, 2014. 5(2):
p. 391–398.
100. Gohil, S.V. et al., Evaluation of enzymatically crosslinked injectable glycol chitosan hydrogel.
Journal of Materials Chemistry B, 2015. 3(27): p. 5511–5522.
101. Wu, L.‐Q., W.E. Bentley, and G.F. Payne, Biofabrication with biopolymers and enzymes: Potential
for constructing scaffolds from soft matter. The International Journal of Artificial Organs, 2011.
34(2): p. 215–224.
102. Yamamoto, H. et al., Insolubilizing and adhesive studies of water‐soluble synthetic model proteins.
International Journal of Biological Macromolecules, 1990. 12(5): p. 305–310.
103. Wei, Z. et al., Novel biocompatible polysaccharide‐based self‐healing hydrogel. Advanced Functional
Materials, 2015. 25(9): p. 1352–1359.
104. Azevedo, S. et al., Bioinspired ultratough hydrogel with fast recovery, self‐healing, injectability and
cytocompatibility. Advanced Materials, 2017. 29(28): p. 1700759
105. Zan, J. et al., Preparation and properties of crosslinked chitosan thermosensitive hydrogel for inject-
able drug delivery systems. Journal of Applied Polymer Science, 2006. 101(3): p. 1892–1898.
106. Chen, C. et al., Preparation and properties of an injectable thermo‐sensitive double crosslinking
hydrogel based on thiolated chitosan/beta‐glycerophosphate. Journal of Materials Science, 2011.
47(5): p. 2509–2517.
107. Moura, M.J. et al., In situ forming chitosan hydrogels prepared via ionic/covalent co‐cross‐linking.
Biomacromolecules, 2011. 12(9): p. 3275–3284.
108. McKay, C.A. et al., An injectable, calcium responsive composite hydrogel for the treatment of acute
spinal cord injury. ACS Applied Materials & Interfaces, 2014. 6(3): p. 1424–1438.
109. Wei, Z. et al., Dual cross‐linked biofunctional and self‐healing networks to generate user‐defined
modular gradient hydrogel constructs. Advanced Healthcare Materials, 2017. 6(16): p. 1700523.
110. Yan,Y. et al., Construction of injectable double‐network hydrogels for cell delivery. Biomacromolecules,
2017. 18(7): p. 2128–2138.
111. Abandansari, H.S. et al., In situ formation of interpenetrating polymer network using sequential thermal
and click crosslinking for enhanced retention of transplanted cells. Biomaterials, 2018. 170: p. 12–25.
112. Latifi, N. et al., A tissue‐mimetic nano‐fibrillar hybrid injectable hydrogel for potential soft tissue
engineering applications. Scientific Reports, 2018. 8(1): p. 1047.
113. Duan, J.‐J. and L.‐N. Zhang, Robust and smart hydrogels based on natural polymers. Chinese Journal
of Polymer Science, 2017. 35(10): p. 1165–1180.
114. Wei, Z. et al., Self‐healing gels based on constitutional dynamic chemistry and their potential applica-
tions. Chemical Society Reviews, 2014. 43(23): p. 8114–8131.
115. Gyarmati, B., B.Á. Szilágyi, and A. Szilágyi, Reversible interactions in self‐healing and shape mem-
ory hydrogels. European Polymer Journal, 2017. 93: p. 642–669.
116. Li, Q. et al., The design, mechanism and biomedical application of self‐healing hydrogels. Chinese
Chemical Letters, 2017. 28(9): p. 1857–1874.
117. Taylor, D.L. and M. In Het Panhuis, Self‐healing hydrogels. Advanced Materials, 2016. 28(41):
p. 9060–9093.
118. Darabi, M.A. et al., Skin‐inspired multifunctional autonomic‐intrinsic conductive self‐healing hydro-
gels with pressure sensitivity, stretchability, and 3D printability. Advanced Materials, 2017. 29(31):
p. 1700533.
119. Yavvari, P.S. and A. Srivastava, Robust, self‐healing hydrogels synthesised from catechol rich
polymers. Journal of Materials Chemistry B, 2015. 3(5): p. 899–910.
120. Li, S. et al., Synthesis and characterization of a novel double cross‐linked hydrogel based on Diels‐
Alder click reaction and coordination bonding. Materials Science & Engineering C‐Materials for
Biological Applications, 2018. 82: p. 299–309.
121. Kang, M. et al., Template method for dual network self‐healing hydrogel with conductive property.
Materials & Design, 2018. 148: p. 96–103.
122. Rodell, C.B. et al., Selective proteolytic degradation of guest–host assembled, injectable hyaluronic
acid hydrogels. ACS Biomaterials Science & Engineering, 2015. 1(4): p. 277–286.
123. Tuncaboylu, D.C. et al., Structure optimization of self‐healing hydrogels formed via hydrophobic
interactions. Polymer, 2012. 53(24): p. 5513–5522.
124. Tuncaboylu, D.C. et al., Autonomic self‐healing in covalently crosslinked hydrogels containing
hydrophobic domains. Polymer, 2013. 54(23): p. 6381–6388.
125. Gulyuz, U. and O. Okay, Self‐healing polyacrylic acid hydrogels. Soft Matter, 2013. 9(43):
p. 10287–10293.
126. Li, Y. et al., Chitosan‐based self‐healing hydrogel for bioapplications. Chinese Chemical Letters,
2017. 28(11): p. 2053–2057.
127. Zhang, Y. et al., Synthesis of multiresponsive and dynamic chitosan‐based hydrogels for controlled
release of bioactive molecules. Biomacromolecules, 2011. 12(8): p. 2894–2901.
128. Yang, B. et al., Facilely prepared inexpensive and biocompatible self‐healing hydrogel: a new inject-
able cell therapy carrier. Polymer Chemistry, 2012. 3(12): p. 3235–3238.
129. Xie, W. et al., Injectable and self‐healing thermosensitive magnetic hydrogel for asynchronous con-
trol release of doxorubicin and docetaxel to treat triple‐negative breast cancer. ACS Applied Materials
& Interfaces, 2017. 9(39): p. 33660–33673.
130. Dong, R. et al., Self‐healing conductive injectable hydrogels with antibacterial activity as cell deliv-
ery carrier for cardiac cell therapy. ACS Applied Materials & Interfaces, 2016. 8(27):
p. 17138–17150.
131. Zhang, Y. et al., A magnetic self‐healing hydrogel. Chemical Communication, 2012. 48(74):
p. 9305–9307.
132. Fu, C. et al., Hierarchically porous chitosan‐PEG‐silica biohybrid: Synthesis and rapid cell adsorp-
tion. Advanced Healthcare Materials, 2013. 2(2): p. 302–305.
133. Liu, H. et al., Self‐healing and injectable polysaccharide hydrogels with tunable mechanical proper-
ties. Cellulose, 2017. 25(1): p. 559–571.
134. Ding, F. et al., Flexible polysaccharide hydrogel with pH‐regulated recovery of self‐healing and
mechanical properties. Macromolecular Materials and Engineering, 2017. 302(11): p. 1700221.
135. Xu, C. et al., Self‐healing chitosan/vanillin hydrogels based on Schiff‐base bond/hydrogen bond
hybrid linkages. Polymer Testing, 2018. 66: p. 155–163.
136. Karimi, A.R. and A. Khodadadi, Mechanically robust 3D nanostructure chitosan‐based hydrogels
with autonomic self‐healing properties. ACS Applied Materials & Interfaces, 2016. 8(40):
p. 27254–27263.
137. Karimi, A.R., A. Khodadadi, and M. Hadizadeh, A nanoporous photosensitizing hydrogel based on
chitosan cross‐linked by zinc phthalocyanine: An injectable and pH‐stimuli responsive system for
effective cancer therapy. RSC Advances, 2016. 6(94): p. 91445–91452.
138. Huang, W. et al., Strong and rapidly self‐healing hydrogels: Potential hemostatic materials. Advanced
Healthcare Materials, 2016. 5(21): p. 2813–2822.
139. Liu, J. et al., Visualization of in situ hydrogels by MRI in vivo. Journal of Materials Chemistry B,
2016. 4(7): p. 1343–1353.
140. Yang, X. et al., Highly efficient self‐healable and dual responsive cellulose‐based hydrogels
for controlled release and 3D cell culture. Advanced Functional Materials, 2017. 27(40):
p. 1703174.
141. Liu, H. et al., Self‐healing polysaccharide hydrogel based on dynamic covalent enamine bonds.
Macromolecular Materials and Engineering, 2016. 301(6): p. 725–732.
142. Collins, J. et al., Doubly dynamic self‐healing materials based on oxime click chemistry and boronic
acids. Macromolecular Rapid Communications, 2017. 38(6): p. 1600760.
143. Cash, J.J. et al., Room‐temperature self‐healing polymers based on dynamic‐covalent boronic esters.
Macromolecules, 2015. 48(7): p. 2098–2106.
144. Matxain, J.M., J.M. Asua, and F. Ruiperez, Design of new disulfide‐based organic compounds for the
improvement of self‐healing materials. Physical Chemistry Chemical Physics, 2016. 18(3):
p. 1758–1770.
145. Shao, C. et al., A self‐healing cellulose nanocrystal‐poly(ethylene glycol) nanocomposite hydrogel
via Diels–Alder click reaction. ACS Sustainable Chemistry & Engineering, 2017. 5(7):
p. 6167–6174.
146. Li, S. et al., Preparation and characterization of acid resistant double cross‐linked hydrogel for poten-
tial biomedical applications. ACS Biomaterials Science & Engineering, 2018. 4(3): p. 872–883.
147. Correia, C.O. and J.F. Mano, Chitosan scaffolds with a shape memory effect induced by hydration.
Journal of Materials Chemistry B, 2014. 2(21): p. 3315–3323.
148. Gao, H.‐L. et al., A shape‐memory scaffold for macroscale assembly of functional nanoscale building
blocks. Materials Horizons, 2014. 1(1): p. 69–73.
149. Correia, C.O., A.J. Leite, and J.F. Mano, Chitosan/bioactive glass nanoparticles scaffolds with shape
memory properties. Carbohydrate Polymers, 2015. 123: p. 39–45.
150. You, J. et al., Quaternized Chitosan/Poly(acrylic acid) Polyelectrolyte complex hydrogels with tough,
self‐recovery, and tunable mechanical properties. Macromolecules, 2016. 49(3): p. 1049–1059.
151. Chen, M.C. et al., The characteristics and in vivo suppression of neointimal formation with sirolimus‐
eluting polymeric stents. Biomaterials, 2009. 30(1): p. 79–88.
152. Zhang, Y. et al., Bio‐inspired layered chitosan/graphene oxide nanocomposite hydrogels with high
strength and pH‐driven shape memory effect. Carbohydrate Polymers, 2017. 177: p. 116–125.
153. Jian, Y.K. et al., Shape memory hydrogels with simultaneously switchable fluorescence behavior.
Macromolecular Rapid Communications, 2018. 39(12): p. 1800130.
154. Bao, M. et al., Ultrasound‐modulated shape memory and payload release effects in a biodegradable
cylindrical rod made of chitosan‐functionalized PLGA microspheres. Biomacromolecules, 2013.
14(6): p. 1971–1979.
155. Wang, L. et al., Actuating and memorizing bilayer hydrogels for a self‐deformed shape memory func-
tion. Chemical Communication, 2018. 54(10): p. 1229–1232.
156. Xiao, H. et al., A multi‐responsive hydrogel with a triple shape memory effect based on reversible
switches. Chemical Communication, 2016. 52(90): p. 13292–13295.
157. Cheng, B. et al., Advances in chitosan‐based superabsorbent hydrogels. RSC Advances, 2017. 7(67):
p. 42036–42046.
158. Guilherme, M.R. et al., Superabsorbent hydrogels based on polysaccharides for application in agri-
culture as soil conditioner and nutrient carrier: A review. European Polymer Journal, 2015. 72:
p. 365–385.
159. He, G. et al., Preparation and properties of quaternary ammonium chitosan‐g‐poly(acrylic acid‐co‐
acrylamide) superabsorbent hydrogels. Reactive and Functional Polymers, 2017. 111: p. 14–21.
160. Bidgoli, H., A. Zamani, and M.J. Taherzadeh, Effect of carboxymethylation conditions on the water‐
binding capacity of chitosan‐based superabsorbents. Carbohydrate Research, 2010. 345(18):
p. 2683–2689.
161. Narayanan, A. and R. Dhamodharan, Super water‐absorbing new material from chitosan, EDTA and
urea. Carbohydrate Polymers, 2015. 134: p. 337–343.
162. Pourjavadi, A., V. Aghajani, and H. Ghasemzadeh, Synthesis, characterization and swelling behavior
of chitosan‐sucrose as a novel full‐polysaccharide superabsorbent hydrogel. Journal of Applied
Polymer Science, 2008. 109(4): p. 2648–2655.
163. Narayanan, A. et al., Super water absorbing polymeric gel from chitosan, citric acid and urea:
Synthesis and mechanism of water absorption. Carbohydrate Polymers, 2018. 191: p. 152–160.
164. Sabadini, R.C., V.C.A. Martins, and A. Pawlicka, Synthesis and characterization of gellan gum:
Chitosan biohydrogels for soil humidity control and fertilizer release. Cellulose, 2015. 22(3): p.
2045–2054.
165. Chen, Y. and H.M. Tan, Crosslinked carboxymethylchitosan‐g‐poly(acrylic acid) copolymer as a
novel superabsorbent polymer. Carbohydrate Research, 2006. 341(7): p. 887–896.
166. Huacai, G., P. Wan, and L. Dengke, Graft copolymerization of chitosan with acrylic acid under micro-
wave irradiation and its water absorbency. Carbohydrate Polymers, 2006. 66(3): p. 372–378.
167. Yu, C. et al., Study of carboxymethyl chitosan based polyampholyte superabsorbent polymer I.
Carbohydrate Polymers, 2010. 81(2): p. 365–371.
168. Mahdavinia, G.R., M.J. Zohuriaan‐Mehr, and A. Pourjavadi, Modified chitosan III, superabsorbency,
salt‐ and pH‐sensitivity of smart ampholytic hydrogels from chitosan‐g‐PAN. Polymers for Advanced
Technologies, 2004. 15(4): p. 173–180.
169. Kabiri, K. et al., Superabsorbent hydrogel composites and nanocomposites: A review. Polymer
Composites, 2011. 32(2): p. 277–289.
170. Zhang, J., Q. Wang, and A. Wang, Synthesis and characterization of chitosan‐g‐poly(acrylic acid)/
attapulgite superabsorbent composites. Carbohydrate Polymers, 2007. 68(2): p. 367–374.
171. Xie, Y. and A. Wang, Study on superabsorbent composites XIX. Synthesis, characterization and
performance of chitosan‐g‐poly (acrylic acid)/vermiculite superabsorbent composites. Journal of
Polymer Research, 2008. 16(2): p. 143–150.
172. Rodrigues, F.H.A. et al., Chitosan‐graft‐poly(acrylic acid)/rice husk ash based superabsorbent
hydrogel composite: preparation and characterization. Journal of Polymer Research, 2012. 19(12): p. 1.
173. Liu, J., Q. Wang, and A. Wang, Synthesis and characterization of chitosan‐g‐poly(acrylic acid)/
sodium humate superabsorbent. Carbohydrate Polymers, 2007. 70(2): p. 166–173.
174. Spagnol, C. et al., Superabsorbent hydrogel composite made of cellulose nanofibrils and chitosan‐
graft‐poly(acrylic acid). Carbohydrate Polymers, 2012. 87(3): p. 2038–2045.
175. Wang, X. et al., Preparation of pure chitosan film using ternary solvents and its super absorbency.
Carbohydrate Polymers, 2016. 153: p. 253–257.
176. Yu, Y. et al., Synthesis and properties of N‐maleyl chitosan‐cross‐linked poly(acrylic acid‐co‐acrylamide)
superabsorbents. Journal of Polymers and the Environment, 2011. 19(4): p. 926–934.
Chitin and chitosan have been recognised for their beneficial health effects since the 1980s
[1]. Over the past few decades, numerous studies and several clinical trials have been per-
formed which demonstrated that these compounds can reduce body weight and cardiovas-
cular disease (CVD), improve wound healing, but can also modulate the immune system
and demonstrate antifungal and antibacterial activity. In particular, weight reduction and
improvement of cardiovascular status are interesting targets as the prevalence of obesity
and CVD are increasing. Both diseases are associated with various pathological disorders,
including diabetes and hypertension and put a strain on healthcare costs and capacity. In
general, lifestyle‐based interventions such as the oral intake of chitin and/or chitosan are
becoming increasingly popular as these are easily integrated into current treatments and
improve the self‐assertiveness of patients. Of interest, but beyond the scope of this chapter,
is the use of chitin‐glucans as supplements in cosmetics [2, 3]. Several studies have dem-
onstrated that chitin‐glucan reduced wrinkling and skin-ageing, suggesting an interaction
between the chitin‐glucan and cells of the epidermis. Although these effects may not be
attributed to chitin alone, this does demonstrate the strong biological potential of chitin on
cells of the human body. In this chapter, scientific literature has been reviewed that demon-
strated the beneficial health effects of chitin and chitosan from an immunomodulatory
point-of-view. First, we provide an overview of in vitro studies that offer in‐depth mecha-
nistic insights, followed by describing preclinical animal studies. Finally, we list various
human intervention trials that most clearly demonstrate the beneficial health effects of
chitin and chitosan. Furthermore, we purposely discriminate between data on chitin and
chitosan as they are chemically distinct and therefore possibly demonstrate unique effects
on health.
Box 6.1 Receptors that have been demonstrated to bind to chitin or chitosan.
TLRs: Toll‐like receptors are expressed on the surface of multiple cells, including that of
immune cells such as macrophages and dendritic cells. TLR2 and TLR4 are shown to bind to
chitin or chitosan, and subsequently activate an innate immune response [8, 9]. In addition to
TLRs expressed on the cell surface, TLR9, which is expressed in the endosomal compart-
ments, was identified to be involved in chitin and chitosan‐mediated production of the anti‐
inflammatory cytokine IL‐10 [10].
Dectin‐1: This is a member of the C‐type lectin receptors that is mainly involved in the
recognition of fungal pathogens. It is also important for the immune response towards bacteria,
viruses and parasites [11, 12] and reported to be able to recognise chitin.
Mannose receptor (MR): MR is an endocytic receptor that also belongs to the C‐type lectin
family. It is predominately expressed on the surface of macrophages and dendritic cells. Chitin
can bind to the mannose receptor and activate the phagocytic process of immune cells [13].
NKR‐P1: This again is a C‐type lectin receptor that is primarily expressed on natural killer
(NK) cells and T cells. This receptor was shown to possess a chitin oligomer binding domain
which leads to the activation of NK cells [14].
FIBCD1: This is a transmembrane endocytic receptor that is expressed primarily by intesti-
nal epithelial cells. It serves as an acetyl binding receptor to recognise chitin [15].
RegIIIγ: This is a secreted C‐type lectin receptor which is expressed by intestinal epithelial
cells. It can recognise N‐acetyl‐glucosamine oligomers [16].
Galectin‐3: This is a member of the galectin family that is abundantly expressed on mac-
rophages. This receptor was shown to have an N‐acetyl‐glucosamine binding domain [17].
NOD2: NOD2 is an intracellular chitin recognition receptor which is primarily expressed
by macrophages and epithelial cells. It was reported to mediate chitin‐induced IL‐10
production [18].
Abbreviations: FIBCD1: Fibrinogen C Domain Containing 1; NKR‐P1: Killer cell lectin‐
like receptor subfamily B, member 1; RegIIIγ: regenerating islet‐derived protein 3 gamma;
TLR: toll‐like receptor; NOD2: nucleotide‐binding oligomerization domain‐like receptors.
of the pro‐inflammatory cytokines IL‐6 and TNFα by peripheral blood mononuclear cells
(PBMCs) which contain a large variety of immune cells dominated by lymphocytes (i.e. T
and B cells) [10]. A similar chitin preparation with a size of 1–10 μm was shown to induce
IFNγ, IL‐12 and TNFα secretion by an immune cell mixture isolated from the spleen [22].
Despite the differences in immune cell responses to chitin, both studies demonstrated that
the responses were dependent on chitin interaction with the mannose receptor, NOD2 and
TLR9, and also that immune responses can be chitin‐concentration‐dependent. Low
concentrations of chitin induced secretion of low levels of TNFα, whereas this exponentially
increased upon stimulation with higher concentrations of chitin. In contrast, all chitin
concentrations induced similar concentrations of IL‐10.
Next to concentration, the size of chitin too was demonstrated to affect immune
responses. Upon exposure to chitin fractions of <40 μm, 40–70 μm or 70–100 μm,
macrophages showed different responses in cytokine production, receptor binding and
downstream signalling pathway activation [23]. The smallest fraction induced both TNFα
and IL‐10 secretion, whereas the intermediate‐sized chitin induced only TNFα secretion
and the large particles did not induce any cytokine production. The varying dependency of
148 Chitin and Chitosan: Properties and Applications
cytokine production on receptor binding adds to the complexity of immune responses and
downstream signalling. TNFα secretion in response to small‐sized chitin proved to be
dependent on dectin‐1 recognition with optional TLR2 and mannose receptor binding, and
relied on downstream NF‐κB and partial Syk signalling. In contrast, TNFα secretion in
response to intermediate‐sized chitin was dectin‐1 and NF‐κB dependent, but also fully
TLR2 dependent and mannose receptor independent. The intermediate‐sized chitin
fragments were also subject to a more in‐depth study which demonstrated that these also
induced IL‐17, IL‐12 and IL‐23 production in a TLR2‐ and MyD88‐dependent manner
[24]. Interestingly, IL‐12, IL‐23 and TNFα production all depended on the initial IL‐17
production. Furthermore, intermediate‐sized chitin also induced, independently of IL‐17,
CCL5 and CXCL2.
Similar to chitin, chitosan has also been shown to exert pro‐ and anti‐inflammatory
effects on immune cells. Chitosan oligomers with a limited polymerisation degree of 3–8
units were shown to activate macrophages [9]. This particular macrophage cell‐line (RAW
264.7 macrophages) demonstrated increased proliferation and production of pro‐inflam-
matory mediators TNFα and nitric oxide. TLR4 was shown to be important for these
responses as an anti‐TLR4 antibody appeared to block the direct interaction between chi-
tosan and macrophages and nitric oxide production. Small fragments of chitosan were
also shown to be most potent in enhancing intra‐epithelial lymphocyte (IEL) cytotoxicity
against YAC‐1 cells ex vivo [25]. As YAC‐1 cells are particularly sensitive to NK cell–
mediated cytotoxicity, these results suggest that chitosan specifically enhanced NK cell
activity in the IEL population. In addition to the direct immunomodulatory effects of
chitosan, a number of studies investigated the modulatory effects of chitosan upon pri-
mary lipopolysaccharides (LPS)‐stimulation. LPS‐stimulated macrophages demonstrated
an increase in IL‐1β production when co‐stimulated with chitosan, but not chitin [26].
This increase in IL‐1β production is both concentration‐ and size‐dependent, with higher‐
concentration and smaller‐sized chitosan inducing a more potent IL‐1β production.
Indeed, chitosan <20 μm proved most potent and >100 μm almost functionally inert,
which is reminiscent of other reported chitin‐size‐dependent effects. In this study, the
authors also demonstrate that chitosan depends on the NLRP3 inflammasome for intracel-
lular signalling.
In a similar study, treatment of LPS‐stimulated macrophages with chitosan reduced the
TNFα, IL‐6 and NO production [27]. Unfortunately, it is unclear whether these experiments
were performed with similar small‐sized chitosan. Another study, in which the production
of pro‐inflammatory mediators following LPS‐stimulation of macrophages was again
measured, demonstrated that chitosan negatively affected the production of nitric oxide,
PGE2, TNFα, IL‐6 and also IL‐1β [28]. Here, the authors again investigated the effect of
chitosan size and demonstrated that the inhibiting effects on these mediators were stronger
for smaller particles. Still, a direct comparison between studies remains difficult as the size
of chitosan in this study is not described in degrees of polymerisation or μm but in kDa.
Also, the authors demonstrated that the inhibiting effects of chitosan are mediated by
reducing intracellular MAP‐kinase activation. Interestingly, Li and colleagues revealed that
LPS‐mediated cytokine production is regulated by the transcription factor NF‐κB, which in
turn is activated by MAP‐kinases [29]. Therefore, it appears that chitosan is inhibiting
LPS‐mediated pro‐inflammatory macrophage responses by reducing intracellular MAP‐
kinase activation, potentially via activation of TLR4.
Beneficial Health Effects of Chitin and Chitosan 149
Initial insights in cellular responses towards chitin and chitosan were acquired using in
vitro studies and some ex vivo studies. These studies have demonstrated that there are a
number of receptors that can recognise chitin and chitosan, which leads to modulating
responses of a number of cell types. It is important to note that the immunomodulatory
effects of chitin and chitosan are highly concentration‐ and size‐dependent. The variation
of chitin and chitosan concentration and size leads to different immune responses. In in
vitro studies, chitin concentration has a positive correlation with its immunomodulatory
activity. In contrast, chitin or chitosan size was inversely linked with its immune activity,
and particles with a size bigger than 100 μm were considered as immunomodulatory inert.
These in vitro studies were in part related to the analysis of in vivo effects of chitin or
chitosan towards fungal lung infection and tumour growth. The in vitro results corroborated
the in vivo data which was encouraging for the potential of translational research to estimate
the potential health effects of chitin and chitosan, which is described in more detail in the
following paragraphs. However, it should be noted that, in in vitro studies, LPS contamination
is commonly present and can strongly interfere with proper interpretation of the
immunomodulatory effects of chitin or chitosan. The chitosan studies especially
demonstrate clearly that the presence of LPS can strongly affect the net result of immune
responses. Nevertheless, many studies did not determine the LPS content during the
preparation of chitin or chitosan products [24, 30]. The presence of LPS in chitin or chitosan
samples does not interfere with readouts when using in vivo models, which is another
advantage beyond testing the full complexity of the body. Therefore, we will focus on the
beneficial health effects and validation of immunomodulatory effects of chitin or chitosan
in in vivo studies following oral intake while also focussing on the used particle sizes and
doses.
No. of subjects
(experimental arm/control);
Products Origin Size/MW DD Product administration subject status Summary of effects References
Chitin microparticles n.d. 1–10 μm n.d. 6.5 mg/day for 16 days; 6 or 7/6 or 7; allergic mice ↓IL‐4, IL‐5, IL‐10 (p<0.05); [31]
oral administration IgE levels (p<0.0005)
↑IFNγ (p<0.05); IgG2a levels
(p<0.05)
LM‐chitosan n.d. <1 kDa 98.5% 16.5 mg/kg twice a day for 14 10–16/10–16; asthmatic ↓TNFα, IL‐4, IL‐5, IL‐13 [32]
days; oral administration mice (p<0.001)
Chitin microparticles n.d. 1000 kDa n.d. 0.2% wt chitin contained diet for 8/8; peanut allergic mice ↓IL‐5, IL‐10, IL‐13 (p<0.05) [33]
6 weeks; oral administration
Chitosan microparticles n.d. 70 kDa 91% 0.2% wt chitin contained diet for 8/8; peanut allergic mice ↓IL‐5, IL‐10, IL‐13 (p<0.05)
6 weeks; oral administration ↓serum IgE (p<0.05) [33]
Chitin nanofibrils Crab 200 μm n.d. 1% (w/v) chitin for 7 days; oral 5/5; IBD mice ↓NF‐κB staining area (p<0.05) [34]
administration ↓MCP‐1 (p<0.05)
Abbreviations: DD: degree of de‐acetylation; IBD: inflammatory bowel disease; IFN: interferon; Ig: immunoglobulin; IL: interleukin; LM: low molecular; MCP‐1: monocyte
chemotactic protein‐1; MW, molecular weight; NF‐κB: nuclear factor kappa light chain enhancer of activated B cells; n.d.: not described; NK, natural killer.
No. of subjects
(experimental arm/control);
Products Origin Size/MW DD Product administration subjects status Summary of effects References
Chitin n.d. n.d. n.d. 50 mg/kg per day for 6 18 (i.p.) or 20 (i.v.)/18 or ↑Serum polymorphonuclear [38]
days at a 2‐day 20; C. albicans infected leucocytes (p<0.01)
interval; intravenous mice ↑Survival time
or intraperitoneal
administration
Chitosan n.d. n.d. n.d. 50 mg/kg per day for 6 18 (i.p.) or 20 (i.v.)/18 or ↑Active oxygen generating cells [38]
days at a 2‐day 20; C. albicans infected (p<0.01)
interval; intravenous mice ↑Survival time
or intraperitoneal
administration
Chitosan oligosaccharides Crab 208 kDa 70–100% 0.5 or 1 mg per day for 5/5; S. aureus infected mice ↑The number of monocytes (p<0.05) [39]
5 and 10 days; ↑IFNγ, IL‐6 (p<0.01)
intraperitoneal ↑Survival time
injection
0.5, 1, 2 mg per day
for 7 days; oral
administration
Chitin Crab n.d. n.d. 4 μg/g or 6 μg/g for 10/10; V. alginolyticus ↑THC, respiratory burst (p<0.05) [40]
once; injection infected shrimp ↑Phagocytosis ability
↑Survival time
Chitosan Crab n.d. n.d. 2 μg/g or 4 μg/g for 10/10; V. alginolyticus ↑THC, respiratory burst (p<0.05) [40]
once; injection infected shrimp ↑Phagocytosis ability
↑Survival time
Chitin Prawn n.d. 26.7% 1% (wt) chitin 50/50; A. invadans infected ↑WBC, RBC, haematocrit, [41]
contained diet for 15 fish lymphocytes, monocytes,
days; oral neutrophils (p<0.05)
administration ↑Phagocytosis activity
↓Mortality
(Continued)
No. of subjects
(experimental arm/control);
Products Origin Size/MW DD Product administration subjects status Summary of effects References
Chitosan Prawn n.d. 58.5% 1% (wt) chitin 50/50; A. invadans infected ↑WBC, RBC, haematocrit, [41]
contained diet for 15 fish lymphocytes, monocytes,
days; oral neutrophils (p<0.05)
administration ↑Phagocytosis activity,
↓Mortality
Chitin microparticles n.d. <40 μm n.d. 100 μg/μL for 2 weeks 5/5; L. major infected mice ↓Parasitic load (p<0.05) [42]
at a 2‐day interval; ↓Lesion formation (p=0.021)
injected at the ↑TNFα, IL‐10 (p<0.001)
infected site
Chitosan microparticles n.d. <40 μm n.d. 100 μg/μL for 2 weeks 5/5; L. major infected mice ↓Parasitic load (p< 0.05) [42]
at a 2‐day interval; ↓Lesion formation (p=0.039)
injected at the ↑TNFα, IL‐10 (n.s.)
infected site
Chitin microparticles n.d. <44 μm n.d. 100 μg/μL for 2 weeks 6/6; L. major infected mice ↓Lesion formation (p=0.023) [43]
at a 2‐day interval; ↓TNFα (p=0.026)
injected at the ↑IFNγ/IL‐5 (p=0.023)
infected site ↑IL‐10
Chitosan n.d. n.d. n.d. 250 mg/kg once a day 8/8; E. papillata infected ↓The number of parasites in the [45]
for 5 days; oral mice developmental stage (p<0.01)
administration ↓oocyst in faeces (p<0.01)
↓NO production; TNFα, TGFβ gene
expression (p<0.001)
↑IL‐4, IL‐10 gene expression
(p<0.001)
Chitosan particles n.d. n.d. 88.5% 500 μg once a week 10/10; H. nana infected ↓Gene expression of iNOS, IFNα, [46]
for 4 weeks; oral mice IFNγ, IL‐9 and TNFα (p< 0.001)
administration ↑The number of mucosal mast cells;
gene expression of IL‐4 (p<0.001)
Abbreviations: DD: degree of de‐acetylation; IFN: interferon; IL: interleukin; iNOS: inducible nitric oxide synthase; i.p.: intraperitoneal injection; i.v.: intravenous injection; MW: molecular
weight; NF‐κB: nuclear factor kappa light chain enhancer of activated B cells; n.d.: not described; NK: natural killer; NO: nitric oxide; n.s.: not significant; RBC: red blood cells; THC:
haemocyte count; TNF: tumour necrosis factor; TGF: transforming growth factor; WBC: white blood cells.
on average 6.5 micrograms per mouse per day. The preventive treatment with chitin signifi-
cantly (p<0.0005) reduced the serum IgE levels and ragweed‐specific IgE levels. In con-
trast, the ragweed‐specific IgG2a levels are significantly (p<0.05) increased. By using
bronchoalveolar lavage (BAL) to harvest cells and cytokines from the lung, it became
evident that oral chitin intervention significantly reduced (p<0.05) the migration of eosino-
phils and lymphocytes to the lung and drastically (p<0.05) altered cytokine profiles from
an IL‐4‐ to an IFNγ‐dominated type. Strikingly, not only did a 13‐day preventive adminis-
tration of chitin drastically alter a Th2‐dominated immune response to a Th1‐dominated
type, but also a curative setting in which chitin was provided on days 12 and 13 signifi-
cantly (p<0.01) decreased total serum IgE levels and immune cell lung infiltrates (p<0.05).
A similar curative allergy model was used to study the immunomodulatory effects of low
molecular (LM) chitosan [32]. In this study, mice were sensitised to ovalbumin (OVA) with
a 12‐day protocol using three i.p. injections with alum as adjuvant, after which allergic
responses were induced by intranasal administration of OVA. The intranasal administration
was performed from day 13 to 27 and coincided with daily oral administration of LM‐chitosan
(16.5 mg/kg) or saline solution as control. Analysis of the allergic responses were focussed
on cytokine production. BAL fluids of mice treated with LM‐chitosan demonstrated a
significant (p<0.001) reduction in IL‐4, IL‐13 and TNFα levels. Furthermore, lung tissue
also revealed a significant reduction of mRNA levels of IL‐4, IL‐5, IL‐13 and TNFα.
Interestingly, no change in IFNγ mRNA levels were observed. In comparison to the previ-
ous study, no chitosan‐mediated skewing of the immune response was observed (i.e. Th2
towards Th1), but LM‐chitosan did induce a strong reduction in allergic responses.
Chitosan and chitin were directly compared as preventive treatment in a peanut allergy
model [33]. This model is, in essence, similar to the earlier described models, but with a
different timeline and location of sensitisation and challenge. The mice were provided with
α‐chitin, β‐chitin or β‐chitosan mixed in food pellets at a concentration of 0.2% (w/w). The
interventions resulted in anaphylaxis reduction when the sensitised mice were challenged
with peanut. To investigate whether the chitin or chitosan indeed affected immune cell
responses, splenocytes were isolated and ex vivo stimulated with peanut extracts. This
revealed that splenocytes from mice treated with α‐chitin, β‐chitin or β‐chitosan were less
sensitive to the peanut extract and produced significantly (p<0.05) less IL‐5, IL‐13 and
IL‐10. Authors also investigated serum peanut–specific IgE levels which were lowered by
α‐chitin and β‐chitosan, but not β‐chitin. Within the group of β‐chitin‐treated mice, there
was a strong division in mice with low and high IgE levels, indicating that β‐chitin has the
potency to lower allergic responses, but is dependent on a parameter beyond the scope of
this study.
Instead of redirecting immune responses towards a Th1 type response, chitin has also
demonstrated the potency to reduce the strength of immune responses. To investigate the
potential of chitin to reduce the inflammation in inflammatory bowel disease (IBD), a
mouse model was used in which ulcerative colitis was induced using dextran sulphate
sodium (DSS) [34]. Regular chitin with an average diameter of 200 μm or specifically
generated chitin nanofibrils with average lengths of 4 nm at a concentration of 0.1% (w/v)
in tap water was provided as a 7‐day intervention before the DSS‐mediated induction of
ulcerative colitis. Analysis of colonic epithelium demonstrated a significant (p<0.05)
reduction in activation of the NF‐κB transcription factor, and serum analysis revealed sig-
nificantly (p<0.05) reduced levels of monocyte chemotactic protein‐1 (MCP‐1), which is
Table 6.3 Anti‐tumour effects of chitin and chitosan.
No. of subjects
(experimental arm/
Products Origin Size/MW DD Product administration control); subjects status Summary of effects References
Chitin and chitosan Crab n.d. n.d. 1, 2, 4% (wt) chitin or 8/8; Colon‐26 tumour‐ ↓Tumour weight (p<0.05) [47]
oligomers chitosan contained diet bearing mice ↑IFNγ, IL‐12p70 (p<0.01)
for 28 days; oral
administration
LM‐chitosan n.d. 21, 46 and 130 kDa n.d. 100 mg/kg or 300 mg/kg 10/10; Sarcoma 180‐ ↓Tumour weight and growth [25]
twice daily for 20 days; bearing mice (p<0.05)
oral administration ↑NK cells activity
LM‐chitosan Shrimp 30 kDa 95% 100 mg/kg or 200 mg/kg per 10/10; Liver tumour ↓Tumour weight (100 mg/kg) [48]
day for 14 days; oral H‐22 bearing mice (p<0.05)
administration ↓Tumour weight (200 mg/kg)
(p<0.01)
↑Leucocytes, monocytes,
neutrophils and lymphocytes
(p<0.05)
↑The number of CD3/CD4 T‐
cells, NK cells (p<0.05)
↑TNFα, IL‐12 (p<0.05)
Abbreviations: CD3: cluster of differentiation 3; CD4: cluster of differentiation 4; DD: degree of de‐acetylation; IFN: interferon; IL: interleukin; MW, molecular weight; n.d.: not
described; NK, natural killer; TNF: tumour necrosis factor.
the key chemokine for the migration and infiltration of monocytes and macrophages. These
effects, however, were only observed following preventive treatment with chitin nanofibrils
and not with regular chitin. Based on the knowledge gained from in vitro analysis, it appears
that the chitin length in in vivo situations as well might be a crucial parameter to reduce
intestinal inflammation.
Chitosan and, in particular, chitin have long been associated with allergenic response.
Recent studies indicate that these correlations are mainly attributable to various proteins
[35], and clinical trials have demonstrated the safety of using chitosan in bandages [36].
The general observation that chitin and chitosan prevent allergenic responses in the lung
and shift Th2 responses towards Th1 substantiates this. In addition, chitin also demonstrated
lowering of intestinal immune responses. Both applications could potentially improve the
quality of life for many patients.
or chitin (1% (w/w)) to treat Aphanomyces invadans infection. Interestingly, in this study,
pretreatment and continued additions of either chitin or chitosan reduced mortality in fish
from 70% to almost the levels of uninfected fish 4 weeks post‐infection. Similar as for
shrimp, chitosan was slightly more effective in reducing the mortality in fish which was
accompanied by an increased level of red and white blood cell counts, among which were
monocytes, lymphocytes and neutrophils, which was already apparent at the first moment
of measuring 1 week after infection.
Studies to investigate anti‐parasitic activity of chitin and chitosan were mostly performed
in a curative setting. In a model to test the effects against Leishmania major infections,
mice were subcutaneously injected with the parasite and treated with local injections of
small‐sized chitin or chitosan (<40 μm) for 2 weeks with 2‐day intervals [42]. The onset of
lesions in the treated groups was delayed and ulcer diameter of the lesions was significantly
smaller (p<0.05). Twelve weeks after infection, the mice were sacrificed and analysis
revealed a significantly (p<0.05) reduced parasitic load in the draining lymph nodes. In
relation to reducing infections, the authors investigated the potential of chitin and chitosan
to induce production of cytokines by lymph node‐derived immune cells. Only chitin
incubation resulted in a significant (p<0.001) increase in both IL‐10 and TNFα production.
Although the production of these cytokines appears contradictive, an activation of Th1
response which is stimulated by TNFα enhanced macrophage activity and increased IL‐10
production critically limited the harmful side effects of inflammatory responses. In a
previous L. major infection mice trial performed by the same group, chitin was shown to
also increase IFNγ and reduce IL‐5 production [43]. These results were in line with an in
vitro study, also performed by this group, which showed that chitin induced the increase of
TNFα production in peritoneal macrophages after L. major infection [44]. Although these
ex vivo cytokine productions do not directly correlate to a lowered burden of infection, it
does appear to substantiate that chitin enhances the immune system’s ability to combat L.
major.
The treatment of the intestinal parasite Eimeria papillata was only tested with chitosan.
Mice were intestinally infected with E. papillata and subsequently treated with a daily dose
of chitosan for 5 days. The infectious grade was determined on day 5 by the oocyte count
in the faeces of the mice and was found to be significantly reduced (p<0.01) because of the
chitosan treatment when compared to non‐treated mice [45]. In line with this finding, the
authors observed a significantly reduced number of parasites per crypt (p<0.01) and a
reversion of the decline in goblet cells in histological sections following chitosan treatment.
Chitosan treatment also reversed other infection‐related immunological changes such as an
increase in the level of oxidative stress, neutrophil infiltration and inflammatory cytokine
gene transcription, and a decrease in anti‐inflammatory cytokine gene transcription. In
contrast, E. papillata infection also reduced the levels of T cells in the villi and IgA levels
in sera and the intestine, but treatment with chitosan did not prevent this.
Interestingly, when compared head‐to‐head based on a reduction of the burden of infec-
tion and accompanying histological and immunological changes, it appears that chitosan is
more effective in reducing the burden of microbial infection whereas chitin appears more
effective against parasites. After all these studies, it remains unclear, however, whether
these chitin‐ and chitosan‐mediated effects are instigated by potentiating the immune sys-
tem or reducing the virulence of microbes and parasites and thereby lowering immune
responses.
Beneficial Health Effects of Chitin and Chitosan 157
A single study has demonstrated effects of chitosan beyond restoring immune parame-
ters towards non‐infected controls. To treat the infection of an intestinal tapeworm
(Hymenolepis nana) mice were pretreated with a weekly dose of 500 μg chitosan for 4
weeks [46]. As observed in the earlier described studies, most parameters, histological and
immune‐related, that were affected by the H. nana infection were restored to or towards the
levels found in non‐treated mice by the pretreatment with chitosan (p<0.01). Similar to the
earlier described study, the authors also observed a reduced load in H. nana adults and
eggs, which altogether again suggests that chitosan reduces the virulence of H. nana.
However, in contrast to the earlier described study, mast cell concentrations were investi-
gated and these were not affected by the H. nana infection but significantly increased by
chitosan beyond levels found in both non‐treated and infected mice. It, therefore, appears
that chitosan pretreatment increases mast cell numbers and, potentially, their activity which
allows a quick and efficient anti‐H. nana response and thereby prevents a full‐scale immune
response resulting in histological damage and production of inflammatory cytokines.
So far, most studies using various microbes or parasites infecting either the skin or
intestine demonstrate that chitin and chitosan can indeed reduce these infections although
the mechanism behind this remains elusive. By reducing the extent of infection, chitin and
chitosan most likely also reduce the inflammatory response, which explains the lowered
cytokine levels and immune cell recruitment. Mast cells appear to play a critical role as
their presence is selectively increased by chitosan in the intestinal infection by H. nana. In
contrast, the production of IL‐5, which is related to mast cell activity in allergenic responses,
is reduced by chitin in ex vivo immune cell stimulations, which demonstrates the necessity
for further research into the anti‐parasitic potential of chitin and chitosan.
chitosan could not be detected in blood or spleen but was detected in the small intestine.
Coincidently, IELs from mice that were orally administered low molecular chitosan, but
again not high molecular chitosan, demonstrate significantly (p<0.05) enhanced ex vivo
cytotoxicity against YAC‐1 cells in a size‐dependent manner. Recently, a group from China
confirmed that low MW water‐soluble chitosan contains anti‐tumour activity and did so in
an H22 liver tumour mice model [48]. The chitosan was orally administrated to mice at 100
or 200 mg/kg for 14 days, starting 12 hours after the inoculation of the tumour cells. The
treatment revealed significant concentration‐dependent (p<0.05) reduction in tumour
growth and spleen weight compared to non‐treated animals. At a cellular level, the number
of leucocytes, lymphocytes, monocytes and also neutrophils were significantly (p<0.05)
increased in the chitosan intervention group in comparison to the untreated group. In par-
ticular, the CD4+ T‐cells, but not CD8+ T‐cells, and NK cell concentrations were signifi-
cantly (p<0.05) increased after chitosan intervention. Finally, also TNFα and IL‐2 serum
levels, cytokines that are critical in anti‐tumour response, were significantly (p<0.05)
increased upon chitosan treatment [48, 49].
These papers demonstrate that low MW chitosan can modulate the immune system and
induce pro‐inflammatory anti‐tumour activities in different tumour‐bearing murine models.
In these studies it is more plausible that the observed immune cell counts and activities are
a direct chitosan‐derived effect, in contrast to the potential physical interference in anti‐
pathogen studies. Therefore, low MW chitosan may be a promising adjuvant for anti‐
tumour therapies.
Abbreviations: DD: degree of de‐acetylation; HDL: high‐density lipoprotein; LDL: low‐density lipoprotein; n.a.: not applicable; n.d.: not described; TG: triglyceride
Chitosan has also been tested in clinical trials for its hypocholesterolaemic potency. A
pilot study was conducted by Rizzo and colleagues to investigate the effect of fungal chi-
tosan on regulation of plasma lipid and lipoprotein in hypertriglyceridaemia patients [52].
Subjects were orally administered 125 mg/day of chitosan tablets for a period of 4 months.
This intervention resulted in a significant decrease in total cholesterol (p=0.019) and tri-
glyceride (TG) levels (p<0.001) and a concomitant significant increase in high‐density
lipoprotein (HDL) cholesterol (p=0.016) and LDL‐2 particles (p<0.0001), a less athero-
genic LDL particle. Total LDL levels did decrease although not significantly (p>0.05). The
observed increase in HDL and lowering of LDL constitutes a beneficial change in plasma
lipid composition and lowering the risk of CVD. The potential of chitosan to reduce the
risk of CVD has also been tested in smokers who are at higher risk of developing this dis-
ease [53]. The study setup included a higher dose of chitosan (1 g/day) for a shorter period
(6 weeks), which also resulted in a significant decrease in total cholesterol (p<0.01) and
LDL cholesterol (p<0.05) levels.
These results for chitosan have led to a European Food Safety Authority (EFSA) claim
on cholesterol lowering, and chitosan is available as a commercial product. So far, how-
ever, the mechanism behind chitosan’s effect remains to be elucidated. Chitosan might bind
bile acids in the intestine, preventing their reuptake and forcing enhanced cholesterol
metabolism to generate bile acids [54]. Other potential mechanisms are the increased
expression of LDL receptors in the liver, which would result in enhanced uptake and break-
down of serum LDL or the inhibition of pancreatic lipase activity to reduce the digestion
and uptake of dietary fats [55]. Depending on the mechanism, and whether the acetylation
level is involved in this, a similar EFSA claim might be possible for chitin, which clearly
demonstrated its potency in clinical trials.
No. of subjects
(experimental arm/
Products Origin MW DD Product administration Study design placebo); subject status Summary of effects References
WSC n.d. 20 kDa 95% 540 mg/day for 4 weeks; Open label 10/n.a.; healthy subjects ↓Plasma glucose (p<0.05) [56]
orally ingested ↓Atherogenic index (p<0.05)
↑HDL cholesterol (p<0.05)
Chitosan n.d. n.d. n.d. 1.5 g/day for 12 weeks; Randomised, 25/26; pre‐diabetic ↓Serum glucose control [57]
orally ingested double‐blind, participants (p=0.03);
placebo‐controlled ↓Plasma level of
haemoglobin level
(p=0.023)
↑Plasma adiponectin
(p=0.013)
Chitosan n.d. n.d. n.d. 2.25 g/day for 3 months; Randomised, 6/6; obese subjects ↓Body weight (p=0.027) [58]
orally ingested double‐blind ↓TG level (p=0.028)
Chitosan Fungi n.d. n.d. 2.5 g/day for 90 days; Single‐blind, 64/32; obese subjects ↓Body weight (p<0.0001) [59]
orally ingested placebo‐controlled, ↓HbA1c level (p=0.0343)
randomised
Water‐soluble Crab 3–5 kDa 70% 37.5 or 150 mg/day for Randomised, 12/n.a.; healthy subjects ↓Dental plaque formation (S. [62]
chitosan 6 weeks; rinsed with double‐blind, mutans) (p=0.014)
mouth rinse solution placebo‐controlled
LMW‐WSC n.d. 5 kDa n.d. 20 mg chitosan solution Randomised, 40/n.a.; denture ↓Formation of erythematous [61]
four times a day for 2 single‐blind stomatitis patients surface area (p<0.001)
weeks; immerse in ↓Number of blastopores and
chitosan solution mycelia of C. albicans
(n.s.)
Chitosan n.d. n.d. n.d. 2% w/v solubility in Randomised, 50/n.a.; healthy subjects ↓Amount of bacteria [63]
saliva; chewed eight double‐blind, (Streptococci) (p<0.05)
gums in total placebo‐controlled
Chitin Prawn 1–20 μm n.d. 2 mg in total; intranasal Randomised, 14/n.a.; healthy ↓IL‐4; IL‐6 [64]
administration double‐blind, participants ↑Leukotriene B4
placebo‐controlled
Abbreviations: DD: degree of de‐acetylation; HbA1c: haemoglobin A1c; HDL: high‐density lipoprotein; IL: interleukin; LDL: low‐density lipoprotein; LMW‐WSC: low‐molecular‐
weight water‐soluble chitosan; MW: molecular weight; n.a.: not applicable; n.d.: not described; n.s.: not significant; TG: triglyceride.
insulin levels were observed, which might have been expected with lowered blood glucose
levels. Another study also designed as randomised, double‐blind and placebo‐controlled
analysed insulin sensitivity, rather than insulin levels, in obese subjects following chitosan
intervention [58]. Subjects orally ingested 2.25 g of chitosan (750 mg before every main
meal) for a period of 3 months, which are conditions similar to the previous study, which
did not yield a change in blood insulin level. As hypothesised by the authors, chitosan
significantly increased insulin sensitivity (p=0.43). At the same time, subjects also lost a
significant amount of body weight through the intake of chitosan (p=0.027), which was
accompanied by a significant decrease in body mass index (BMI) (p=0.028) and waist
circumference (p=0.028). These changes might relate again to the reduction in uptake of
cholesterol and bile acids as described earlier since triglycerides levels were found to be
significantly reduced (p=0.028). This potential relationship was used as the hypothesis to
investigate the effect of chitosan on body weight reduction [59]. Again similar study con-
ditions were used with a 90‐day intervention period and daily 2.5 g intake of chitosan and
a randomised, double‐blind, placebo‐controlled setting. Of note, this study included a
larger amount of subjects (i.e. 96 instead of 12), which increased the significance and
validity of the study. Again in this study, chitosan proved to significantly (p<0.0001)
reduce body weight when compared to the placebo group and again corresponding meas-
urements such as BMI, body fat and visceral fat were also significantly reduced (p<0.001).
Taken together, these clinical studies demonstrate that chitosan has a clear effect on
metabolism that is reflected in cholesterol regulation, body weight, antioxidant capacity
and insulin sensitivity.
The cationic interactions that underlie the binding of bile acids by chitosan might also
make chitosan a suitable bactericidal and fungicidal component [60], in addition to the
other putative mechanisms as described earlier. The positive charge of the amino group can
interact with anionic cell surface components, such as lipopolysaccharides of gram‐
negative bacteria. This interaction was shown to destabilise the integrity of the outer mem-
brane which might sensitise bacteria but cannot directly be described as bactericidal
activity. This aspect of chitosan was utilised in a number of studies investigating the benefi-
cial effect on oral health [61–63]. Chitosan was added to mouthwash or chewing gum and
indeed reduced the plaque index and related oral bacterial levels and even local
inflammation.
In contrast to chitosan, only a single clinical trial was performed with chitin focussing on
non‐CVD end‐points. This study (Table 6.5) did include the analysis of interleukin produc-
tion, which are key mediators of immune cells. This study was designed as a randomised,
double‐blind, placebo‐controlled, crossover study with a 2‐week washout period in between
each intervention [64]. Healthy participants were randomised into two groups and received
either intranasal chitin microparticles (1–20 μm) or placebo. Chitin microparticles demon-
strated a significant reduction in local IL‐4 and IL‐6 levels but an increase in leukotriene
B4 levels and total white blood cell counts. These results are somewhat contradicting as
IL‐6 is also an important pro‐inflammatory marker, preventing a general conclusion on
how chitin microparticles affect the intranasal immune system.
Taken together, it is not possible to draw firm conclusions on chitin‐mediated beneficial
health effects because of the limited number of clinical studies. Although the potential of
chitin in CVD treatment has been demonstrated, it has only been tested in healthy subjects
with relatively low oxidised LDL. More studies are required to potentially acquire an EFSA
Beneficial Health Effects of Chitin and Chitosan 163
status similar to chitosan. The clinical studies towards the effect of chitosan on cholesterol
reduction, glucose control and weight loss were more comprehensive and clearly demon-
strated beneficial health effects.
study compound details should have been described. This, however, is a challenge of the
growing field of studies towards beneficial health effects of food compounds or orally
administered supplements such as protein, dietary fibres, etc. The lack of detailed infor-
mation prevents a direct comparison of results and therefore hampers the analysis of
contradictory results and accumulation of knowledge to unlock the potential of chitin
and chitosan.
6.5 Outlook
The terms ‘chitin’ and ‘chitosan’ cover a wide range of molecules with varying MW, acety-
lation levels and sources and purification methods. All these variables are demonstrated to
impact immune responses. There is a large body of work describing the immunomodula-
tory effects of chitin and chitosan, but often lacks a detailed description of the structural
characteristics of the preparations under investigation, which hampers clear‐cut conclu-
sions of the potential of chitin and chitosan. As for other areas of food and nutrition
research, we are only scraping the surface knowledge of interaction with the immune sys-
tem. Most studies focus on epithelial and macrophage responses which represent important
parts of the innate immune system. Studies towards the adaptive immune system are more
complicated and arguably more receptive to minor differences in chitin and chitosan struc-
ture. To ensure that work benefits the current body of knowledge on immunomodulatory
effects of chitin and chitosan, it is crucial that all parameters as indicated earlier are ana-
lysed and described.
Currently, the beneficial health effects of chitin and chitosan have already been com-
mercialised in various products (e.g. Chitosan®, xtendlifeTM). The main functions of these
products are weight loss and cholesterol reduction. The clinical trials and animal studies
performed to demonstrate other beneficial health effects demonstrate the potency of chitin
and chitosan to be more broadly applied to improve health.
Acknowledgement
This work was supported by the Dutch Ministry of Economic Affairs and Climate Policy
(KB‐23‐001‐015).
References
1. Allan, G. et al., Biomedical applications of chitin and chitosan, in Chitin, Chitosan, and Related
Enzymes. 1984, Elsevier. p. 119–133.
2. Gautier, S. et al., Chitin–glucan, a natural cell scaffold for skin moisturization and rejuvenation. Int. J.
Cosmet. Sci., 2008. 30(6): p. 459–469.
3. Morganti, P. et al., New chitin complexes and their anti‐aging activity from inside out. J. Nutr., Health
& Aging, 2012. 16(3): p. 242–245.
4. Shahabuddin, M. and D.C. Kaslow, Plasmodium: parasite chitinase and its role in malaria transmission.
Exp. Parasitol., 1994. 79(1): p. 85–88.
5. Fuhrman, J.A. and W.F. Piessens, Chitin synthesis and sheath morphogenesis in Brugia malayi
microfilariae. Mol. Biochem. Parasitol., 1985. 17(1): p. 93–104
6. Debono, M. and R.S. Gordee, Antibiotics that inhibit fungal cell wall development. Annu. Rev.
Microbiol., 1994. 48(1): p. 471–497.
Beneficial Health Effects of Chitin and Chitosan 165
7. Komi, D.E.A., L. Sharma, and C.S.D. Cruz, Chitin and its effects on inflammatory and immune
responses. Clin. Rev. Allergy Immunol., 2017: p. 1–11.
8. Bueter, C.L., C.A. Specht, and S.M. Levitz, Innate sensing of chitin and chitosan. PLoS Pathog., 2013.
9(1): p. e1003080.
9. Zhang, P. et al., Toll like receptor 4 (TLR4) mediates the stimulating activities of chitosan
oligosaccharide on macrophages. Int. Immunopharmacol., 2014. 23(1): p. 254–261.
10. Wagener, J. et al., Fungal chitin dampens inflammation through IL‐10 induction mediated by NOD2
and TLR9 activation. PLoS Pathog., 2014. 10(4): p. e1004050
11. Drummond, R.A. and Brown, G.D. The role of Dectin‐1 in the host defence against fungal infections[J].
Curr. Opin. Microbiol., 2011, 14(4): 392–399.
12. Dong, B. et al., A chitin‐like component on sclerotic cells of Fonsecaea pedrosoi inhibits dectin‐1‐
mediated murine Th17 development by masking β‐glucans. PloS One, 2014. 9(12): p. e114113
13. Taylor, P.R., S. Gordon, and L. Martinez‐Pomares, The mannose receptor: linking homeostasis and
immunity through sugar recognition. Trends Immunol., 2005. 26(2): p. 104–110.
14. Kirkham, C.L. and J.R. Carlyle, Complexity and diversity of the NKR‐P1: Clr (Klrb1: Clec2) recogni-
tion systems. Front. Immunol., 2014. 5: p. 214.
15. Schlosser, A. et al., Characterization of FIBCD1 as an acetyl group‐binding receptor that binds chitin.
J. Immunol., 2009. 183(6): p. 3800–3809.
16. Cash, H.L. et al., Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science,
2006. 313(5790): p. 1126–1130.
17. Seetharaman, J. et al., X‐ray crystal structure of the human galectin‐3 carbohydrate recognition domain
at 2.1‐Å resolution. J. Biol. Chem., 1998. 273(21): p. 13047–13052.
18. Rosenstiel, P. et al., A short isoform of NOD2/CARD15, NOD2‐S, is an endogenous inhibitor of
NOD2/receptor‐interacting protein kinase 2‐induced signaling pathways. Proc. Natl. Acad. Sci. USA,
2006. 103(9): p. 3280–3285.
19. Van Dyken, S.J. et al., Chitin activates parallel immune modules that direct distinct inflammatory
responses via innate lymphoid type 2 and γδ T cells. Immunity, 2014. 40(3): p. 414–424.
20. O’Dea, E.M. et al., Eosinophils are recruited in response to chitin exposure and enhance Th2‐mediated
immune pathology in Aspergillus fumigatus infection. Infect. Immun., 2014. 82(8): p. 3199–3205.
21. Wiesner, D.L. et al., Chitin recognition via chitotriosidase promotes pathologic type‐2 helper T cell
responses to cryptococcal infection. PLoS Pathog., 2015. 11(3): p. e1004701.
22. Shibata, Y., W.J. Metzger, and Q.N. Myrvik, Chitin particle‐induced cell‐mediated immunity is
inhibited by soluble mannan: Mannose receptor‐mediated phagocytosis initiates IL‐12 production.
J. Immunol., 1997. 159(5): p. 2462–2467.
23. Da Silva, C.A. et al., Chitin is a size‐dependent regulator of macrophage TNF and IL‐10 production.
J. Immunol., 2009. 182(6): p. 3573–3582.
24. Da Silva, C.A. et al., TLR‐2 and IL‐17A in chitin‐induced macrophage activation and acute inflamma-
tion. J. Immunol., 2008. 181(6): p. 4279–4286.
25. Maeda, Y. and Y. Kimura, Antitumor effects of various low‐molecular‐weight chitosans are due to
increased natural killer activity of intestinal intraepithelial lymphocytes in sarcoma 180–bearing mice.
J. Nutr., 2004. 134(4): p. 945–950.
26. Bueter, C.L. et al., Chitosan but not chitin activates the inflammasome by a mechanism dependent
upon phagocytosis. J. Biol. Chem., 2011. 286(41): p. 35447–35455.
27. Yoon, H.J. et al., Chitosan oligosaccharide (COS) inhibits LPS‐induced inflammatory effects in RAW
264.7 macrophage cells. Biochem. Biophys. Res. Commun., 2007. 358(3): p. 954–959.
28. Pangestuti, R., S.‐S. Bak, and S.‐K. Kim, Attenuation of pro‐inflammatory mediators in LPS‐stimu-
lated BV2 microglia by chitooligosaccharides via the MAPK signaling pathway. Int. J. Biol. Macromol.,
2011. 49(4): p. 599–606.
29. Li, Y. et al., Chitosan oligosaccharides block LPS‐induced O‐GlcNAcylation of NF‐κB and endothelial
inflammatory response. Carbohydrate Polym., 2014. 99: p. 568–578.
166 Chitin and Chitosan: Properties and Applications
30. Shibata, Y. et al., Alveolar macrophage priming by intravenous administration of chitin particles,
polymers of N‐acetyl‐D‐glucosamine, in mice. Infect. Immun., 1997. 65(5): p. 1734–1741.
31. Shibata, Y. et al., Oral administration of chitin down‐regulates serum IgE levels and lung eosinophilia
in the allergic mouse. J. Immunol., 2000. 164(3): p. 1314–1321.
32. Chung, M.J., J.K. Park, and Y.I. Park, Anti‐inflammatory effects of low‐molecular weight chitosan
oligosaccharides in IgE–antigen complex‐stimulated RBL‐2H3 cells and asthma model mice. Int.
Immunopharmacol., 2012. 12(2): p. 453–459.
33. Bae, M.‐J. et al., Oral administration of chitin and chitosan prevents peanut‐induced anaphylaxis in a
murine food allergy model. Int. J. Biol. Macromol., 2013. 61: p. 164–168.
34. Azuma, K. et al., α‐Chitin nanofibrils improve inflammatory and fibrosis responses in inflammatory
bowel disease mice model. Carbohydrate Polym., 2012. 90(1): p. 197–200.
35. Faber, M. A. et al. Shellfish allergens: Tropomyosin and beyond. Allergy, 2017. 72.6: 842–848.
36. Waibel, Kirk H. et al. Safety of chitosan bandages in shellfish allergic patients. Military Med., 2011.
176.10: 1153–1156.
37. Park, B.K. and M.‐M. Kim, Applications of chitin and its derivatives in biological medicine. Int. J.Mol.
Sci., 2010. 11(12): p. 5152–5164.
38. Suzuki, K. et al., Protecting effect of chitin and chitosan on experimentally induced murine candidia-
sis. Microbiol. Immunol., 1984. 28(8): p. 903–912.
39. Moon, J.‐S. et al., The antibacterial and immunostimulative effect of chitosan‐oligosaccharides against
infection by Staphylococcus aureus isolated from bovine mastitis. Appl. Microbiol. Biotechnol., 2007.
75(5): p. 989–998.
40. Wang, S.‐H. and J.‐C. Chen, The protective effect of chitin and chitosan against Vibrio alginolyticus in
white shrimp Litopenaeus vannamei. Fish Shellfish Immunol., 2005. 19(3): p. 191–204.
41. Mari, L.S.S. et al., Protective effect of chitin and chitosan enriched diets on immunity and disease
resistance in Cirrhina mrigala against Aphanomyces invadans. Fish Shellfish Immunol., 2014. 39(2):
p. 378–385
42. Hoseini, M.H.M. et al., Immunotherapeutic effects of chitin in comparison with chitosan against
Leishmania major infection. Parasitol. Int., 2016. 65(2): p. 99–104.
43. Ghotloo, S. et al., Immunomodulatory effects of chitin microparticles on Leishmania major‐infected
BALB/c mice. Parasitol. Int., 2015. 64(2): p. 219–221.
44. Dehghani, F. et al., Immunomodulatory activities of chitin microparticles on Leishmania major‐
infected murine macrophages. Arch.Med. Res., 2011. 42(7): p. 572–576.
45. Abdel‐Latif, M., H.M. Abdel‐Haleem, and A.‐A.S. Abdel‐Baki, Anticoccidial activities of chitosan on
Eimeria papillata‐infected mice. Parasitol. Res., 2016. 115(7): p. 2845–2852.
46. Abdel‐Latif, M. et al., Immunoprotective effect of chitosan particles on Hymenolepis nana‐infected
mice. Scand. J. Immunol., 2017. 86(2): p. 83–90.
47. Masuda, S. et al., Anti‐tumor properties of orally administered glucosamine and N‐acetyl‐D‐glucosamine
oligomers in a mouse model. Carbohydrate Polym., 2014. 111: p. 783–787.
48. Zhao, L. et al., Water‐soluble chitosans shows anti‐cancer effect in mouse H22 liver cancer by enhancing
the immune. Int. J. Clin. Exp. Med., 2016. 9(1): p. 164–171.
49. Seo, W.‐G. et al., Synergistic cooperation between water‐soluble chitosan oligomers and interferon‐γ
for induction of nitric oxide synthesis and tumoricidal activity in murine peritoneal macrophages.
Cancer Lett., 2000. 159(2): p. 189–195.
50. Audrey D. and N. PAqUoT, Managing oxidative stress with a vegetal ingredient, chitin‐glucan. Agro
Food Industry Hi Tech, 2009. 20(4): p. 12–14.
51. Bays, H. et al., Chitin‐glucan fiber effects on oxidized low‐density lipoprotein: A randomized controlled
trial. Eur. J. Clin. Nutr., 2013. 67(1): p. 2–7.
52. Rizzo, M. et al., Effects of chitosan on plasma lipids and lipoproteins: a 4‐month prospective pilot
study. Angiology, 2014. 65(6): p. 538–542.
53. Choi, C.‐R. et al., Chitooligosaccharides decreases plasma lipid levels in healthy men. Int. J. Food Sci.
Nutr., 2012. 63(1): p. 103–106.
Beneficial Health Effects of Chitin and Chitosan 167
54. Xu, G.‐F. et al., Mechanism study of chitosan on lipid metabolism in hyperlipidemic rats. Asia Pacific
J. Clin. Nutr., 2007. 16(S1): p. 313–317.
55. Han, L., Y. Kimura, and H. Okuda, Reduction in fat storage during chitin‐chitosan treatment in mice
fed a high‐fat diet. Int. J. Obes., 1999. 23(2): p. 174.
56. Anraku, M. et al., Antioxidant effects of a dietary supplement: Reduction of indices of oxidative stress
in normal subjects by water‐soluble chitosan. Food Chem. Toxicol., 2009. 47(1): p. 104–109.
57. Kim, H.J. et al., The effects of chitosan oligosaccharide (GO2KA1) supplementation on glucose
control in subjects with prediabetes. Food Funct., 2014. 5(10): p. 2662–2669.
58. Hernández‐González, S.O. et al., Chitosan improves insulin sensitivity as determined by the euglycemic‐
hyperinsulinemic clamp technique in obese subjects. Nutr. Res., 2010. 30(6): p. 392–395.
59. Trivedi, V. et al., Single‐blind, placebo controlled randomised clinical study of chitosan for body
weight reduction. Nutr. J., 2015. 15(1): p. 3.
60. Helander, I. et al., Chitosan disrupts the barrier properties of the outer membrane of Gram‐negative
bacteria. Int. J. Food Microbiol., 2001. 71(2–3): p. 235–244.
61. Atai, Z., M. Atai, and J. Amini, In vivo study of antifungal effects of low‐molecular‐weight chitosan
against Candida albicans. J. Oral Sci., 2017. 59(3): p. 425–430.
62. Bae, K. et al., Effect of water‐soluble reduced chitosan on Streptococcus mutans, plaque regrowth and
biofilm vitality. Clin. Oral Investig., 2006. 10(2): p. 102.
63. Hayashi, Y. et al., Chewing chitosan‐containing gum effectively inhibits the growth of cariogenic bac-
teria. Arch. Oral Biol., 2007. 52(3): p. 290–294.
64. Sigsgaard, T. et al. The change in nasal inflammatory markers after intranasal challenges with particu-
late chitin and lipopolysaccharide: A randomized, double‐blind, placebo‐controlled, crossover study
with a positive control. in International Forum of Allergy & Rhinology. 2015. Wiley Online Library.
65. Alvarez, F.J., The effect of chitin size, shape, source and purification method on immune recognition.
Molecules, 2014. 19(4): p. 4433–4451.
66. Zimmerman, J.R., The characterization of chitin microparticle preparations: Degree of acetylation and
its effect on immunologic response. Conference Proceeding, 2014.
67. Barbosa, J.N. et al., Evaluation of the effect of the degree of acetylation on the inflammatory response
to 3D porous chitosan scaffolds. J. Biomed. Mater. Res. Part A, 2010. 93(1): p. 20–28.
68. Huang, Chun‐Jung et al.Chitin enhances obese inflammation ex vivo. Human Immunol., 2014. 75.1:p.
41–46.
69. Ali, M.F. et al., β‐Glucan–activated human B lymphocytes participate in innate immune responses by
releasing proinflammatory cytokines and stimulating neutrophil chemotaxis. J. Immunol, 2015,
195(11): p. 5318–5326.
7
Antimicrobial Properties of Chitin
and Chitosan
Magdalena Kucharska, Monika Sikora, Kinga Brzoza‐Malczewska,
and Monika Owczarek
Institute of Biopolymers and Chemical Fibres, Lodz, Poland
charged groups of lipopolysaccharides and proteins on the surface of the microbial cells,
which results in disintegration of the cell membrane and damage of the bacterial cell wall.
The pH values below the pKa of chitosan (< 6.3) result in the formation of a polycationic
compound capable of interacting with negatively charged groups on the surface of micro-
bial cells. Chitosan at pH above 7 loses its antimicrobial properties, due to the lack of
protonated amino groups and low solubility. Therefore, in an acidic environment, chitosan
is characterised by the highest solubility and the ability to react with the negatively charged
surface of bacterial cells. All changes associated with the permeability of the cell mem-
brane cause leakage of intracellular substances as well as inactivation and lysis of the cell.
The conducted research also reveals that the complexes of chitosan with some metals
(mainly chitosan‐Ag, chitosan‐Zn and chitosan‐Cu) show a broad antibacterial spectrum
and act more effectively on some Gram‐negative bacteria including Escherichia coli.
Literature reports indicate that the antibacterial activity of chitosan is also influenced by
the ‘age’ of the bacterial cell (its growth phase). Additionally, it is associated with changes
in the membrane potential on the surface of the cells, which increases in the logarithmic
phase of cell growth and gradually decreases in the stationary phase. Therefore, the most
susceptible to the antibacterial action of chitosan are ‘young’ cells (in the early and late
exponential phase), and the least susceptible are the cells already entering the stationary
phase. The mechanism of this bactericidal action is also associated with inactivation of
bacterial enzymes, substitution of metal ions and interaction with teichoic acid on the sur-
face of the bacterial cell [1–3].
The other parameter, temperature, also affects bacterium–chitosan interactions, being
the most optimal in the range of 4–37 °C. Lower or higher temperatures may cause changes
in the cell surface structure and affect cell electronegativity, thus, reducing the number of
free sites for binding with chitosan. The presence of sodium, calcium or magnesium salts
can adversely affect the antimicrobial activity of chitosan, by forming complexes and
reducing the amount of free chitosan and, thus, preventing the interaction between chitosan
and cell wall.
The antibacterial effect of chitosan varies also with its degree of deacetylation and
molecular weight. High‐molecular‐weight chitosan is believed to be capable of forming a
polymeric film with a microbial cell wall, which, among others, prevents the delivery of
nutrients and, hence, the death of bacterial cells. On the other hand, low‐molecular‐weight
chitosan can penetrate into the cells and bind with negatively charged intracellular compo-
nents, for example, phosphate residues of DNA molecules, which results in blocking reac-
tions on the transcriptional level and synthesis of mRNA. The antibacterial effect of
chitosan is associated with the structure of the microbial cell wall. In Gram‐positive bacte-
ria, due to the greater thickness of the cell wall, high‐molecular‐weight chitosan shows
better bactericidal effect, forming a coating encapsulating bacterial cells. Gram‐negative
bacteria have much thinner cell walls, and that is why low‐molecular‐weight chitosan acts
most effectively, penetrating into cells and disintegrating their genetic material [2, 4].
Chitosan is active against both Gram‐negative and Gram‐positive bacteria as well as fila-
mentous fungi and yeasts. Its antifungal activity mainly consists of the inhibition of sporu-
lation and germination of spores. Chitosan oligomers diffuse inside the fungal hyphae and
interfere with the activity of the growth‐promoting enzymes. It is active against a whole
range of microorganisms such as Aspergillus niger, Candida albicans, Candida glabrata,
Fusarium solani (fungi), Staphylococcus aureus, E. coli, Salmonella typhimurium and
Antimicrobial Properties of Chitin and Chitosan 171
produced in the form of a sponge for its utilisation in dressing purposes. According to the
assumption, these dressings can be used in the initial period of wound healing accompa-
nied by inflammation. The obtained wound dressings are characterised by antibacterial
activity against E. coli and S. aureus [17].
Helicobacter pylori is a kind of bacteria that may lead to the development of gastric and
duodenal ulcers, chronic gastritis, gastric cancer, possibly also ischemic heart disease, and
atherosclerotic vascular disease. According to literature, chitosan is highly active against
this bacterium. The research focussed on the determination of the antibacterial activity of
chitosan ascorbate. Chitosan with 60% deacetylation degree prepared from krill chitin was
used to produce this form of polymer. Chitosan ascorbate activity was determined against
17 H. pylori strains. The strains were isolated from the periodontal pockets (7 strains),
atherosclerotic plaque from the carotid and femoral arteries (10 strains). The studies
showed that the flagella of H. pylori strains isolated from the periodontal pockets showed
higher susceptibility to chitosan ascorbate than the strains isolated from atherosclerotic
plaque [18].
In stomatology and treatment of oral cavity diseases, chitosan is used as a viscous, bio-
adhesive antimicrobial, prolonging release of the drug in the treatment of periodontal dis-
ease, as well as for reduction of plaque formation. According to literature, chitosan
ascorbate also shows activity against bacteria isolated from pathological gum pockets, root
canals and abscesses that cause infections in the oral cavity. The experiments included 70
clinical isolates and 4 standard strains. The following bacterial strains were tested:
Aggregatibacter (22 strains), Corynebacterium (3 strains), Capnocytophaga (5 strains),
Campylobacter (19 strains), Eikenella (17 strains), Rothia (4 strains) and 4 standard strains:
E. coli (ATCC 25922), Enterococcus faecalis (ATCC 29212), S. aureus (ATCC 25923) and
P. aeruginosa (ATCC 27853). Chitosan ascorbate showed good activity against microaero-
philic bacteria. The most sensitive to the polymer were strains of Corynebacterium matru-
chotii (100% sensitive) and Aggregatibacter actinomycetemcomitans (72% sensitive). The
Gram‐negative and Gram‐positive bacteria showed slightly less sensitivity (48% and 43%,
respectively), while the least sensitivity exhibited strains were Capnocytophaga gingivalis
and Rothia dentocariosa [19].
A. actinomycetemcomitans plays a key role in the pathogenesis of periodontal diseases:
periodontitis and gingivitis. Kochańska et al. reported that in vitro ascorbic salt of chitosan
shows good activity against these bacteria. The MIC for 77% of A. actinomycetemcomitans
strains is in the range of ≤ 0.06–0.5 mg/mL [20].
An antimicrobial formulation in the form of a thermally sensitive mucoadhesive gel has
been developed for the treatment of oral mucositis. It is a mixture of chitosan trimethylsilyl
or chitosan methyl pyrrolidinone with glycerophosphate [21].
Antibacterial properties of chitosan have also been used to develop a preparation for the
treatment of chronic periodontitis. Chitosan gel with or without metronidazole, as an active
substance, was used to treat this disease. The evaluation of the effectiveness of the devel-
oped product was carried out in clinical tests. They studied the evaluation of the gel’s
impact on clinical parameters such as depth of a gum pocket, clinical attachment level,
gingival recession, plaque index, gingival index and gingival bleeding time index. The
research shows that chitosan alone, as well as in combination with metronidazole, is an
effective preparation for the treatment of periodontitis. Throughout the whole study, no
complications associated with the use of chitosan in patients were observed [22].
Antimicrobial Properties of Chitin and Chitosan 173
type of pathogen are patients with diabetes and endocrinopathies. The clinical form of
Candida is often associated with a predisposing factor, such as AIDS, neutropenia, cancer,
radiotherapy as well as destructive periodontal disease, gingivitis and caries. It is known
that the growth of Candida depends on the availability of sugars, especially glucose.
Ascorbic salt of chitosan was selected for the activity tests against fungi of the Candida
family. The antifungal effect of this form of chitosan on different strains such as C. albi-
cans, C. tropicalis, C. guilliermondii, C. parapsilosis, C. kefyr, C. krusei and the standard
PCM1409 strain has been assessed. Conducted studies demonstrated that in experimental
conditions, chitosan ascorbate showed different antifungal activity. The most sensitive to
the preparation was C. tropicalis strain (MIC ≤ 1 mg/mL, 33% strains), and the least C. guil-
liermondi and C. parapsilosis strains (MIC ≥ 2 mg/mL, 100% strains). Strains belonging to
C. albicans, the dominant species in the study, were less susceptible to chitosan ascorbate
(at a concentration of 0.12–0.5 mg/mL 25% of strains inhibited). It was also found that 31%
of the tested Candida strains were not sensitive to chitosan ascorbate at the concentration
tested (MIC > 2 mg/mL) [30].
The potential biomedical use of chitin includes surgical sutures and wound healing
materials. Chitin has also been tested for its antibacterial and tissue regenerating properties,
which make it useful in the wound healing process and in preparation of materials for
wound treatment, mainly adhesive plasters and dressings. Researchers, after trials on ani-
mals, place great hope in the use of such dressings in wound treatment using chitin with a
molecular weight in the range of 10–50 kDa [31].
shelf life, and maintained freshness by inhibiting the growth of microorganisms responsible
for food spoilage without altering the taste. In addition, antioxidant properties of thyme
extract inhibited lipid peroxidation in fillets. A similar effect may be demonstrated by a
combination of chitosan with essential oils (cinnamon, oregano, tangerine) [35, 36], as well
as chitosan with bamboo vinegar, used to prolong the shelf life of pork [37].
Thanks to modern methods of electrospinning or encapsulation it is possible to exploit
the ability to form nanofibers and nanoparticles (e.g., nanoemulsions) by many natural
polymers (including chitosan) and to use them in the food industry to produce modern,
biologically active and biodegradable food packaging, microbiologically active coatings
and natural preservatives [36, 38]. Studies conducted so far have proved the applicability
of chitosan membranes for coating fresh fruits and vegetables (mainly strawberries, blue-
berries and grapes, fresh cucumbers, tomatoes and carrots), and the addition of herbal
extracts (e.g., rosemary), garlic, nisin, phenolics and flavonoids, or lysozymes and makes
it possible to use these mixtures as antioxidants and natural preservatives protecting against
the development of food spoilage by bacteria and fungi [34, 39, 40]. The use of edible
chitosan films as a matrix for introducing bioactive components could effectively prolong
the shelf life of food products and enrich them with active compounds with a positive effect
on health and food. Actively acting nanoemulsions have a strong antibacterial and antifun-
gal effect, limiting food spoilage [41]. Other literature data indicate positive effect of chi-
tosan on colloidal stabilisation of light beer and wines. Due to its ability to actively
counteract protein coagulation, chitosan can be a natural clarification agent for beers and
wines, and some chitosan derivatives are able to absorb heavy metals from them [42, 32].
The antioxidant properties of chitosan were used, among others, for food preservation,
mainly as an addition to meat and fish. Chitosan acts as a preservative in combination with
other natural antioxidant compounds, for example, with lycopene (a health‐promoting
compound naturally found in tomatoes). It prevents the oxidation of compounds present in
the meat product, limiting its spoiling without changing the taste and nutritional values
and, in addition, it is neutral for the body. In the food industry, the chitosan application
techniques used are usually dipping, spray coating and vacuum packaging of food products
[43, 44].
Chitosan is a strong chelating agent and easily forms complexes with metals. This prop-
erty can be used in design of chitosan–metal nanocomposites with antifungal and antibac-
terial activity. Studies carried out so far prove that chitosan–copper and chitosan–zinc
nanocomposites in vitro strongly inhibit growth of S. aureus, P. aeruginosa and Salmonella
enterica. This feature can be used to limit the growth of these pathogens in the food as well
as cosmetic and textile industries [45].
In the era of propagation of many civilisation diseases, which is often associated with an
improper diet, special attention has to be paid to the consumption of fresh fruits and vegeta-
bles and the beneficial polyunsaturated fatty acids, whose best sources are fresh fish and
seafood. The problem of fresh fish storage is associated with its high content of moisture
and nutrients that enable development of bacterial microflora, which accelerates decompo-
sition of the product, changing its organoleptic and pro‐health properties. Chitosan oligo-
saccharides, which are low‐molecular‐weight, biodegradable products of chitosan, can
prevent these processes. They are a mixture of oligomers that consist of D‐glucosamine
residues connected by β‐1,4 bonds. Due to their low molecular weight, low viscosity and
good solubility in water, they can be applied in food industry as an additive to fish and
176 Chitin and Chitosan: Properties and Applications
seafood. It has been shown that chitosan oligosaccharides have neuroprotective, anticancer,
immunity‐strengthening and cholesterol‐reducing effects. Besides health‐promoting
action, the addition of natural antioxidants and preservatives based on chitosan reduces
economic losses caused by generation of large volume of food waste [46, 47].
modified chitosan proved that they were resistant to high temperatures, humidity and
mechanical washing. Application of modern methods for the production of antibacterial
fabrics for medicine or health care may be a breakthrough in the struggle against spreading
of pathogenic microorganisms, and thus protect society against epidemics threats [49].
hydroxyl radicals was noted for the oligomer with the highest molecular weight. At the
same time, the lower‐molecular‐weight chitosan oligomers showed better antioxidant
activity [84, 85].
So far, a number of compounds is known that induce plant resistance – elicitors natu-
rally occurring in nature as well as synthesised chemical substances. The natural elicitors
are some components of cell membranes of phytopathogenic fungi and plants, such as
glucan, pectin, as well as chitin and chitosan. Large‐scale production of elicitors (induc-
ers) of fungal origin is technically impossible; that is why natural polymers with proper-
ties similar to elicitors from fungi and bacteria and easily available in large quantities are
sought. It should also be noted that, in plant protection against pathogens, the condition
of plants is very important, especially in the initial stages of development. Plants with
stimulated rapid growth easily leave the development stage in which they are very sus-
ceptible to diseases. This effect will be particularly strong if it is supported by the stimu-
lation of immune processes in the plant. Chitosan as a polymer having a positive charge
when applied to the plant from the outside reacts with negatively charged molecules on
the surface of the fungal cell and reacts with specific active areas, mutually chemically
compatible, which causes significant changes in the composition of the membrane func-
tion. Properly selected chitosan also directly affects fungi by inhibiting their growth [68].
Based on the research, it was found that the use of chitosan pentamers on the surface of
soy and maize leaves, affected the net coefficient of photosynthesis one day after appli-
cation [86], which correlated with an increase in stomatal conductance and transpiration
rate. The use of chitosan formulations on leaves had no effect on the concentration of
intercellular CO2 which is necessary for the process of photosynthesis. The foliar appli-
cation of these oligomers also did not negatively affect the height of the corn or soy, the
length of the roots, the surface of the leaves and the total amount of dry matter. Bittelli
et al. suggested that chitosan may be an effective measure to prevent excessive transpira-
tion in order to reduce the need for huge water amounts for irrigation of crops. Researchers
studied the possibility to use chitosan oligomers on the leaf surface for the transpiration
of pepper plants in greenhouse and fields conditions. In both cases, water consumption
was monitored directly and indirectly. Plant biomass and crop yield were also checked to
calculate biomass/water ratio, and the differences in water vapour resistance were ana-
lysed, comparing control and chitosan‐treated plants. Using scanning electron micros-
copy and histochemical analyses, it was shown that the stomata in the leaves treated with
the preparation containing oligomers became closed, which reduces the transpiration
process, providing the leaves with the optimal turgor. The reduction in water consump-
tion for pepper plants after chitosan treatment was estimated at 26–43% compared to the
control, with no changes in the production efficiency and biomass yield [87]. Iriti et al.
revealed some aspects by which chitosan was able to reduce transpiration in bean plants
after application as a foliar spray. The authors have shown that this activity probably
occurred due to the increase in the content of abscisic acid (ABA) in the treated leaves.
Using scanning electron microscopy and histocytochemistry techniques, the authors
showed that after applying the chitosan preparations and increasing the ABA content,
partial closure of stomata occurred, which led to a decrease in the loss of water from the
treated plants [88]. Faoro et al. [58] showed that the use of chitosan as a spray on barley,
locally and systemically reduced infection with the pathogen of powdery mildew
Blumeria graminis f. sp. hordei [89].
180 Chitin and Chitosan: Properties and Applications
(Helianthus annuus L.) in 0.5% (w/v) chitosan solution for 18 hours can increase the mass
of sunflower shoots by 12.9%. It was also found that the emergence of seeds soaked in a
chitosan solution was 16.0% higher in relation to the control seeds soaked in water [94].
Foliar application of chitosan compounds was used in cultivation of tobacco (Nicotiana
tabacum L.), and tobacco seeds were encapsulated in chitosan solution to protect plants
against fungi. Researchers showed that both these ways of using chitosan strengthened the
immune reactions, making them more resistant to Phytophthora parasitica nicotianae [95].
Orlikowski et al. showed that soaking tulip bulbs (Tulipa sp.) in chitosan solution within
30 minutes caused inhibition of fungus F. oxysporum f. sp. tulipae [96, 97]. Chitosan used
in the cultivation of multiflora beans and head lettuce in the form of seed dressing solution
(concentration of 1% (w/v)) effectively protects beans and lettuce against fungal diseases
caused by Bortytis cinerea, F. solani, F. oxysporum f. sp. phaseoli, Fusarium culmorum,
Pythium irregulare and R. solani. Plants grown from dressed seeds were healthier com-
pared to those that were grown from seeds not treated before sowing [98, 99].
7.6 Outlook
The preceding studies show that chitosan is an environmentally safe polymer that stimu-
lates growth and development of certain plant species. It can be used not only for treating
plants by spraying but also for so‐called soil activation. Soil activation is associated with
limiting the number of harmful organisms, but above all with the stimulation of the increase
in the number of chitinolitic microorganisms that inhibit the growth of fungal soil patho-
gens. The results of numerous field trials also confirm that chitosan compounds can be used
to stimulate immune reactions in crop plants. They can be used during cultivation, but also
to treat fruits and vegetables after harvesting in order to improve their health and quality.
The efficacy of chitosan as a preservative during cultivation and storage of fruits and veg-
etables is slightly weaker than fungicides, but the advantages of chitosan such as natural
origin, high biological activity and harmlessness to humans and the environment, makes it
frequently recommended for use in agriculture and horticulture, especially in organic farm-
ing and in the production of healthy food.
References
1. Tsai, G.J. and Su, W.H.: Antibacterial activity of shrimp chitosan against Escherichia coli. J. Food
Protection, 1999, 62(3), 239–243.
2. Kim, J.S. and Shin, D.H.: Inhibitory effect on Streptococcus mutans and mechanical properties of the
chitosan containing composite resin. Korean Acad. Cons. Dentistry, 2013, 2234–7666. doi:10.5395/
rde.2013.38.1.36.
3. Helander, I.M., Nurmiaho‐Lassila, E.‐L., Ahvenainen, R., Rhoades, J., and Roller, S.: Chitosan dis-
rupts the barrier properties of the outer membrane of Gram‐negative bacteria. Int. J. Food Microbiol.,
2001, 71, 235–244.
4. Raafat, D., von Bargen, K., Haas, A., and Sahl, H‐G.: Insights into the mode of action of chitosan as
an antibacterial compound. Appl. Environ. Microbiol., June; 2008, 3764–3773. doi:10.1128/
AEM.00453‐08.
5. Savard, T., Beaulieu, C., Boucher, I., and Champagne, C.P.: Antimicrobial action of hydrolyzed
chitosan against spoilage yeasts and lactic acid bacteria of fermented vegetables. J. Food Protection,
2002, 65(5), 828–833.
182 Chitin and Chitosan: Properties and Applications
6. Costa, E.M., Silva, S., Pina, C., Tavaria, F.K., and Pentado, M.M.: Evaluation and insights into chitosan
antimicrobial activity against anaerobic oral pathogens. Anaerobe, 2012, 18, 305–309. doi:10.1016/j.
anaerobe.2012.04.009.
7. Jumaaa, M., Furkert, F.H., and Muller, B.W.: A new lipid emulsion formulation with high antimicro-
bial efficacy using chitosan. Eur. J. Pharm. Biopharm., 2002, 53, 115–123.
8. Maa, Z., Garrido‐Maestua, A., and Jeonga, K.C.: Application, mode of action, and in vivo activity of
chitosan and its micro and nanoparticles as antimicrobial agents: A review. Carbohydrate Polym.,
2017, 176, 257–265. doi.org/10.1016/j.carbpol.2017.08.082.
9. Kong, M., Chen, X.G., Xing, K., and Park, H.J.: Antimicrobial properties of chitosan and mode of
action: A state of the art review. Int. J. Food Microbiol., 2010, 144, 51–63. doi:10.1016/j.
ijfoodmicro.2010.09.012.
10. Burkatovskaya, M., Tegos, G.P., Swietlik, E., Demidova, T.N., Castano, A.P., and Hamblinabe, M.R.:
Use of chitosan bandage to prevent fatal infections developing from highly contaminated wounds in
mice. Biomaterials, 2006, 27, 4157–4164. doi:10.1016/j.biomaterials.2006.03.028.
11. Inamdar, N. and Mourya, V.: Chitosan and low molecular weight chitosan: Biological and biomedical
applications. Advanced Biomaterials and Biodevices (ed. Tiwari, A. and Nordin, A.N.). John Wiley &
Sons, 2014, 182–242.
12. Niekraszewicz, A.: Chitosan medical dressings. Fibres Text. Eastern Eur., 2005, 13, 6(54), 16–18.
13. http://lunainc.com/wp‐content/uploads/2014/04/Hemostatic‐Agents‐August‐2013.pdf, 30.01.2016.
14. Kucharska, M., Struszczyk, M.H., Niekraszewicz, A., Ciechańska, D., Witczak, E., Tarkowska, S.,
Fortuniak, K., Gulbas‐Diaz, A., Rogaczewska, A., Płoszaj, I., Pluta, A., and Ga ̨siorowski T.:
TROMBOGUARD® – first aid wound dressing. Progress on Chemistry and Application of Chitin and
Its Derivatives, Vol. XVI, 2011, ISSN 1896–5644.
15. Brzoza‐Malczewska, K., Kucharska, M., Wiśniewska‐Wrona, M., Guzińska, K., and Ulacha‐
Bacciarelli, A.: Modified cellulose products for application in hygiene and dressing materials (Part I).
Fibres Text. Eastern Eur., 2015, 23; 3(111), 126–132. doi: 10.5604/12303666.1152554.
16. Brzoza‐Malczewska, K., Kucharska, M., Wiśniewska‐Wrona, M., Kaźmierczak, D., Jóźwicka, J., and
Pałys, B.: Modified cellulosic products for application in hygiene and dressing materials – Part II.
Fibres Text. Eastern Eur., 2015, 23; 5(113), 124–128. doi: 10.5604/12303666.1161768.
17. Wiśniewska‐Wrona, M., Niekraszewicz, A., Struszczyk, H., and Guzińska K.: Estimation of polymer
compositions containing chitosan for veterinary applications. Fibres Text. Eastern Eur., 2002, 3(38),
82–85.
18. Kędzia, A., Kochańska, B., Gębska, A., Wierzbowska, M., and Kufel, A.: Evaluation activities of chi-
tosan ascorbate against rods of Helicobacter pylori isolated from gingival pockets and atherosclerotic
plaques. Progress on Chemistry and Application of Chitin and its Derivatives, 2016, Vol. XXI, ISSN
1896‐5644. doi: 10.15259/PCACD.21.10.
19. Kochańska, B., Kędzia, A., and Gębska, A.: Sensitivity to chitosan ascorbate microaerophilic bacteria
isolated from infections of oral cavity. Progress on Chemistry and Application of Chitin and its
Derivatives, Vol. XXI, 2016, ISSN 1896‐5644. doi:10.15259/PCACD.21.11.
20. Kochańska, B., Kędzia, A., and Wojtasz‐Paja ̨k, A.: The evaluation in vitro susceptibility of
Actinobacillus actinomycetemcomitans. Progress on Chemistry and Application of Chitin and Its
Derivatives, Vol. IX, 2003, ISSN 1896–5644, 2003, 199–203.
21. Rossi, S., Marciello, M., Bonferoni, M.C., Ferrari, F., Sandri, G., Dacarro, C., Grisoli, P., and
Caramella, C.: Thermally sensitive gels based on chitosan derivatives for the treatment of oral mucosi-
tis. Eur. J. Pharm. Biopharm., 2010, Feb; 74(2), 248–254. doi:10.1016/j.ejpb.2009.10.003.
22. Akncbay, H., Senel, S., and Ay, Z.Y.: Application of chitosan gel in the treatment of chronic periodon-
titis. J. Biomed. Mater. Res. Part B: Appl. Biomater., 2007, Feb; 80(2), 290–296. doi: 10.1002/
jbm.b.30596.
23. Decker, E.‐M., Ohle, C., Weiger, R., Wiech, I., and Brecx, M.: A synergistic chlorhexidine/chitosan
combination for improved antiplaque strategies. J. Periodontal Res., 2005; 40, 373–377.
doi:10.1111/j.1600‐0765.2005.00817.x.
Antimicrobial Properties of Chitin and Chitosan 183
24. Franca, J.R., De Luca, M.P., Ribeiro, T.G. et al.: Propolis‐based chitosan varnish: Drug delivery, con-
trolled release and antimicrobial activity against oral pathogen bacteria. BMC Complement. Altern.
Med., 2014, 14:478. doi:10.1186/1472‐6882‐14‐478.
25. Wiśniewska‐Wrona, M., Niekraszewicz, A., and Struszczyk, H.: Some aspects of the chitosan utiliza-
tion in dentistry. Progress on Chemistry and Application of Chitin and Its Derivatives, ISBN
83‐87202‐49‐5, Polish Chitin Society, Vol. VI, 2000, 137–143.
26. Cakmak, A., Cirpanli, Y., Bilensoy, E., Yorganci, K., Caliş, S., Saribaş, Z., and Kaynaroğlu, V.:
Antibacterial activity of triclosan chitosan coated graft on hernia graft infection model. Int. J.
Pharmaceutics, 2009, 381, 214–219. doi:10.1016/j.ijpharm.2009.05.059.
27. Majeti, N.V. and Kumar, R.: A review of chitin and chitosan applications. React. Funct. Polym., 2000,
46, 1–27. doi:10.1016/S1381‐5148(00)00038‐9.
28. Felt, O., Carrel, A., Baehni, P., Buri, P., and Gurny, R.: Chitosan as tear substitute: A wetting agent endowed
with antimicrobial efficacy. J. Ocular Pharmacol. Ther., 2000, 16(3). doi:/10.1089/jop.2000.16.261.
29. Jeon, S.J., Oh, M., Yeo, W‐S., Galvão, K.N., and Jeong, K.C.: Underlying mechanism of antimicrobial
activity of chitosan microparticles and implications for the treatment of infectious diseases. Plos One,
2014, 9(3): e92723. doi:10.1371/journal.pone.0092723.
30. Kochańska, B. and Kędzia, A.: The susceptibility to chitosan ascorbate of yeast like fungi from genus
of Candida isolated from children with diabetes mellitus. Progress on Chemistry and Application of
Chitin and Its Derivatives, Vol. IX, 2003, 193–198, ISSN 1896‐5644.
31. Ciuca, A. and Mihailescu, D.: Chitin depolymerization: Medical applications. Bulletin of the
Transilvania University of Braşov, Series VI, 8(57), No. 2, 2015.
32. Wzorek, W., Bonin, S., and Basiak, A.: Próba zastosowania chitozanu w formie rozpuszczonej do
stabilizacji win. Z ̇ywnos ́c ́, 1(42), 2005.
33. Fernandez‐de Castro, L., Mengíbar, M., Sanchez, A., Arroyo, L., and Villaran, M. C., Díaz de Apodaca
E., Heras A.: Films of chitosan and chitosan‐oligosaccharide neutralized and thermally treated: Effects
on its antibacterial and other activities. LWT – Food Sci. Technol., 2016, 73, 368–374. doi:10.1016/j.
lwt.2015.05.042.
34. Paja ̨k, P., Fortuna, T., and Przetaczek‐Rożnowska, I.: Opakowania jadalne na bazie białek i polisacha-
rydów‐ charakterystyka i zastosowanie. Żywnos ́c ́. Nauka. Technologia. Jakos ́c ́, 2013, 2(87), 5–18.
35. El‐Obeid, T., Yehia, H.M., Sakkas, H., Lambrianidi, L., Tsiraki, M.I., Ioannis, N., and Savvaidis, I.N.:
Shelf‐life of smoked eel fillets treated with chitosan or thyme oil. Int. J. Biol. Macromol., 2018, 114,
578–583. doi:10.1016/j.ijbiomac.2018.03.125.
36. Cui, H., Bai, M., Marwan, M.A. Rashed, Lin L.: The antibacterial activity of clove oil/chitosan nano-
particles embedded gelatin nanofibers against Escherichia coli O157:H7 biofilms on cucumber. Int.
Food Microbiol., 2018, 266, 69–78. doi:10.1016/j.ijfoodmicro.2017.11.019.
37. Zhang, H., He, P., Kang, H., and Li, X.: Antioxidant and antimicrobial effects of edible coating based
on chitosan and bamboo vinegar in ready to cook pork chops. LWT – Food Sci. Technol., 2018, 93,
470–476. doi:10.1016/j.lwt.2018.04.005.
38. Ceylan, Z., UnalSengor, G.F, and Tahsin Yilmaz, M.: Nanoencapsulation of liquid smoke/thymol com-
bination in chitosan nanofibers to delay microbiological spoilage of sea bass (Dicentrarchus labrax)
fillets. J. Food Eng., 2018, 229, 43–49. doi:10.1016/j.jfoodeng.2017.11.038.
39. Song, Z., Li, F., Guan, H., Xu, Y., Fu, Q., and Li, D.: Combination of nisin and ε‐polylysine with chi-
tosan coating inhibits the white blush of fresh‐cut carrots. Food Control, 2017, 74, 34–44. doi:10.1016/j.
foodcont.2016.11.026.
40. Serrano‐Leóna, J.S., Bergamaschia, K.B., Cristiana, M.P. Yoshidab Cristiana, M.P., Saldañaa, E.,
Selanic, M.M., Juan, D., Rios‐Meraa, J.D., Alencara, S.M., and Contreras‐Castillo, C.J.: Chitosan
active films containing agro‐industrial residue extracts for shelf life extension of chicken restructured
product. Food Res. Int., 2018, 108, 93–100. doi:10.1016/j.foodres.2018.03.031.
41. Robledo, N., Vera, P., López, L., Yazdani‐Pedram, M., Ta ̨pia ̨, C., and Abugoch, L.: Thymol nanoemul-
sions incorporated in quinoa protein/chitosan edible films; antifungal effect in cherry tomatoes. Food
Chem., 2018, 246, 211–219. doi:10.1016/j.foodchem.2017.11.032.
184 Chitin and Chitosan: Properties and Applications
42. Wzorek, W., Haberowi, H., and Jędrysiak, P.: Zastosowanie chitozanu w kombinacji z innymi pre-
paratami do stabilizacji piwa jasnego. Żywnos ́c ́, 2(27), 2001.
43. Bagnowska, A., Krala, L., Nowak, A., and Oracz, J.: Właściwości przeciwutleniaja c̨ e chitozanu w
kiełbasach bez dodatku azotanu (III). Żywnos ́c ́. Nauka. Technologia. Jakos ́c ́, 2014, 4(95), 173–187.
doi:10.15193/ZNTJ/2014/95/173‐187.
44. Bonillaa F., Chouljenkoa A., Renesa V., Bechtelc P.J., Kinga J.M., Sathivel S.: Impact of chitosan
application technique on refrigerated catfish fillet Quality. LWT – Food Sci. Technol., 2018, 90,
277–282. doi:10.1016/j.lwt.2017.12.010.
45. Kaur, P., Thakur, R., Barnela, M., Chopra, M., Manujab, A., and Chaudhurya, A.: Synthesis, charac-
terization and in vitro evaluation of cytotoxicity and antimicrobial activity of chitosan–metal nano-
composites. J. Chem. Technol. Biotechnol., 2015;90, 867–873. doi:10.1002/jctb.4383.
46. Jia, S., Liu, X., Huang, Z., Li, Y., Hang, L., and Luo, Y.: Effects of chitosan oligosaccharides on micro-
biota composition of silver carp (Hypophthalmichthys molitrix) determined by culture‐dependent and
independent methods during chilled storage. Int. J. Food Microbiol., 2018, 268, 81–91. doi:10.1016/j.
ijfoodmicro.2018.01.011.
47. Soares, N., Silva, P., Barbosa, C., Pinheiro, R., and Vicente, A.A.: Comparing the effects of glazing and
chitosan‐based coating applied on frozen salmon on its organoleptic and physicochemical characteris-
tics over six‐months storage. J. Food Eng., 2017, 194, 79–86. doi:10.1016/j.jfoodeng.
2016.07.021.
48. Linga, Y., Luoa, Y., Luob, J., Wanga, X., and Sun, R.: Novel antibacterial paper based on quaternized
carboxymethyl chitosan/organic montmorillonite/Ag NP nanocomposites. Ind. Crops Products, 2013,
51, 470–479. doi:10.1016/j.indcrop.2013.09.040.
49. Muxika, A., Etxabide, A., Uranga, J., Guerrero, P., and de la Caba, K.: Chitosan as a bioactive polymer:
Processing, properties and applications. Int. J. Biol. Macromol., 2017, 105, 1358–1368. doi:10.1016/j.
ijbiomac.2017.07.087.
50. Fu, X., Shen, Y., Jiang, X., Huang, D., and Yan, Y.: Chitosan derivatives with dual‐antibacterial func-
tional groups for antimicrobial finishing of cotton fabrics. Carbohydrate Polym., 2011, 85, 221–227.
doi:10.1016/j.carbpol.2011.02.019.
51. Nadaa, A., Al‐Moghazyb, M., Solimanc, A.A.F., Rashwand, G.M.T., Eldawyd, T.H.A., Zassane,
A.A.E., and Saye, G.H.: Pyrazole‐based compounds in chitosan liposomal emulsion for antimicrobial
cotton fabrics. Int. J. Biol. Macromol., 2018, 107, 585–594. doi:10.1016/j.ijbiomac.2017.09.031.
52. Arshad, N., Zia, K.M., Jabeen, F., Anjum, M.N., Akram, N., and Zuber, M.: Synthesis, characteriza-
tion of novel chitosan based water dispersible polyurethanes and their potential deployment as anti-
bacterial textile finish. Int. J. Biol. Macromol., 2018, 111, 485–492. doi:10.1016/j.ijbiomac.
2018.01.032.
53. Placek, M., Dobrowolska, A., Wraga, K., Zawadzińska, A., and Ż urawik, P.: Wykorzystanie chitozanu
w uprawie, przechowalnictwie i ochronie roślin ogrodniczych. PostępyNaukRolniczych, 2009, 3–4,
101–109.
54. Kumaraswamy, R.V., Kumari, S., Choudhary, R.C., Pal, A., Raliya, R., Biswas, P., and Saharan, V.:
Engineered chitosan based nanomaterials: Bioactivities, mechanisms and perspectives in plant
protection and growth. Int. J. Biol. Macromol., 2018, 113, 494–506. doi:10.1016/j.ijbiomac.
2018.02.130.
55. Xing, K., Zhu, X., Peng, X., and Qin, S.: Chitosan antimicrobial and eliciting properties for pest
control in agriculture: A review. Agron. Sustain. Dev., 2015, 35, 569–588. doi:10.1007/
s13593‐014‐0252‐3.
56. Hahn, M.G., Cheong, I‐I., Alba, R., Enkerli, J., and Côte, F.: Oligosaccharide elicitors: Structure and
recognition. Mechanisms of Plant Defense Responses, 1989, 99–116.
57. Sun, B., Zhang, L., Yang, L., Zhang, F., Norse, D., and Zhu, Z.: Agricultural non‐point source pollution
in China: Causes and mitigation measures. Ambio, 2012, 41(4), 370–379. doi: 10.1007/
s13280‐012‐0249‐6.
Antimicrobial Properties of Chitin and Chitosan 185
58. Kempka, J.: Biologiczna ochrona roślin przed chorobami jako element integrowanej ochrony roślin.
Centrum Doradztwa Rolniczego w Brwinowie Oddział w Krakowie, Kraków, 2014.
59. Robak, J., Ostrowska, A., and Adamicki, F.: Effect of pre‐ and postharvest treatments on diseases con-
trol during storage of vegetable. Progr. Plant Protection, 2007, 47(2), 299−305, ISSN 1427‐4337.
60. Włodarek, A., Badełek, E., and Robak, J.: The influence of various products applied during the celeriac
vegetation period on soft root rot. Progr. Plant Protection, 2015, 55(4), 386–390, ISSN 1427‐4337.
doi: 10.14199/ppp‐2015‐065.
61. Salachna P.: Wykorzystanie pochodnych chitozanu w celu stymulacji wzrostu roślin ozdobnych. Ecol.
Eng., 2017, 18(6), 63–68. doi 10.12912/23920629/79426.
62. Wojdyła, A., Orlikowski, L.B., Niekraszewicz, A., and Struszczyk, H.: Effectiveness of chitosan in the
control of Sphaerotheca pannosa var. rosae and Peronospora sparsa on roses and Myrothecium rori-
dum on Dieffenbachia. 1996 in Proceedings of 48th International Symposium on Crop Protection, part
1, 461–464, DriemaandelijksTijdschrift ISSN 0368‐9697, Gent, Belgium.
63. Chandra, S., Chakraborty, N., Dasgupta, A., Sarkar, J., Panda, K., and Acharya, K.: Chitosan nanopar-
ticles: A positive modulator of innate immune responses in plants. Scientific Rep., 2015; 5:15195,
1–14. doi:10.1038/srep15195.
64. Rabea, E.I., El Badawy, M.T., Stevens, C.V., Smagghe, G., and Steurbaut, W.: Chitosan as antimicro-
bial agent: Applications and mode of action. Biomacromolecules, 2003, 4, 1457–1465.
65. Kulikov, S.N., Chirkov, S.N., Ilina, A.V., Lopatin, S.A., and Varlamov, V.P.: Effect of the molecular
weight of chitosan on its antiviral activity in plants. Appl. Biochem. Microbiol., 2006, 42(2),
224–228.
66. Savard, T., Beaulieu, C., Boucher, I., and Champagne, C.P.: Antimicrobial action of hydrolyzed chi-
tosan against spoilage yeasts and lactic acid bacteria of fermented vegetables. J. Food Protection,
2002, 65, 828–833.
67. Pospieszny, H., Mac ḱ owiak, A., and Ż ołobowska, L.: Wpływ pochodnych naturalnych polimerów na
wirusy i bakterie roślinne. Prace Naukowe Inst. Tech. Org. Politech. Wrocławskiej, 472–473, 1995.
68. Struszczyk, H., Niekraszewicz, A., Kucharska, M., Wiśniewska‐Wrona, M., Lisiewski, D. Domard,
W.A., Jeunieux, Ch., Muzzarelli, R., Roberts G.M., eds.: Advances in Chitin Science, Vol. 1, J. Andre
Publ., Lyon, 497.
69. Niekraszewicz, A.,: Raport z Projektu Badawczego KBN nr 3 T09 B 189 08 – Badania wpływu
budowy molekularnej i nadcza ̨steczkowej wybranych form mikrokrytalicznego chitozanu na jego
aktywnośc ́ biologiczna ,̨ 1997.
70. Struszczyk, H., Raport z Projektu Badawczego KBN nr 3 T09 B 074 09‐ Kreowanie bioaktywności
chitozanu i jego pochodnych.
71. Kurzawińska, H., Mazur, S., and Nadziakiewicz, M.: Biological activity of natural compounds applied
for potato protection against early blight (Alternaria spp.). Progress in Plant Protection, 2012, 52(1),
78–81, ISSN 1427‐4337.
72. Struszczyk, H. and Pospieszny, H.: New applications of chitin and its derivatives in plant protection,
W.A.F. Goosen, ed., Applications of Chitin and Chitosan, 1997, Technomic Press, New York.
73. Struszczyk, H. and Pospieszny, H.: Biological activity of selected forms of chitosan, in M. Peter, A.
Domard, R. Muzzarelli, eds., Advances in Chitin Science, 2000, Vol. IV, Universitat Potsdam Publs.,
75–80, Potsdam.
74. Struszczyk, H. and Pospieszny, H.: pat. pol. A 0154, środek do ograniczania infekcji wirusów
roślinnych przenoszonych na drodze mechanicznej, 1990.
75. Struszczyk, H., Niekraszewicz, A., and Pospieszny, H. i in. PL‐186594, Biopreparat do ochrony roślin
przed chorobami grzybów.
76. Struszczyk, H., Niekraszewicz, A., and Pospieszny, H., i in. PL 186593, Biopreparat do ochrony roślin
przed chorobami bakteryjnymi.
77. Struszczyk, H., Niekraszewicz, A., and Pospieszny, H., i in. zgłosz. pat. pol. P‐340131 Biopreparat do
pielęgnacjiroślin, 2000.
186 Chitin and Chitosan: Properties and Applications
78. Kuc ,́ J.: Concepts and direction of induced systemic resistance in plants and its application. Eur. J.
Plant Pathol., 2001, 107, 7–12.
79. Pospieszny, H. and Struszczyk, H.: Stimulation of the seed germination and the plant growth by chi-
tosan. Progress in chemistry and application of chitin and its derivatives. Monograph of Polish Chitin
Society, 1999, Vol. V, 43–48, Łódź. ISSN 1896‐5644.
80. Orlikowski, L.B., Skrzypczak, C., Niekraszewicz, A., and Struszczyk, H.: Effectiveness of micro-
crystalline chitosan in the control of Fusarium Wilt of carnation, Monograph: Progress on
Chemistry and Application of Chitin and Its Derivatives, ed. H. Struszczyk, 1998, Vol. IV, 135,
Łódź. ISSN 1896‐5644.
81. Niekraszewicz, A., Ciechańska, D., Wiśniewska‐Wrona, M., Strobin, G., Pospieszny, H., and
Orlikowski, L.B.: Studies of application possibilities of the products of microcrystalline chitosan bio-
degradation. Polimery, 2007, 52(3), 217–220.
82. Orlikowski, L.B., Niekraszewicz, A., and Wiśniewska–Wrona, M.: Możliwości wykorzystania
kompozycji biopolimerowych jako zapraw nasiennych. Progr. Plant Protection, 2012, 52(4), 1033–1037,
2012.
83. Domard, A., Jeuniaux, Ch., Muzzarelli, R.A.A., and Roberts, G., eds, Advances in Chitin Science,
J. Andre Publ., Vol. I, 1996, Lyon.
84. Sun, T., Zhou, D., Xie, J., and Mao, F.: Preparation of chitosan oligomers and their antioxidant activity.
Chem. Mater. Sci., 2006, 225 (3–4), 451–456.
85. Hadrami, A.E., Adam, L.R., Hadrami, I.E., and Daayf, F.: Chitosan in plant protection. Marine Drugs,
2010, 8, 968–987. doi:10.3390/md8040968.
86. Khan, W., Prithiviraj, B., and Smith, D.L.: Effect of foliar application of chitin and chitosan oligosac-
charides on photosynthesis of maize and soybean. Photosynth. Res., 2002, 40, 621–624.
87. Bittelli, M., Flury, M., Campbell, G.S., and Nichols, E.J.: Reduction of transpiration through foliar
application of chitosan. Agric. Forest Meteorol., 2001, 107, 167–175.
88. Iriti, M., Picchi, V., Rossoni, M., Gomarasca, S., Ludwig, N., Garganoand, M., and Faoro, F.: Chitosan
antitranspirant activity is due to abscisic acid‐dependent stomatal closure. Environ. Exp. Botany, 2009,
66, 493–500, doi:10.1016/j.envexpbot.2009.01.004.
89. Faoro, F., Maffi, D., Cantu, D., and Iriti, M.: Chemical‐induced resistance against powdery mildew in
barley: the effects of chitosan and benzothiadiazole. BioControl, 2008, 53, 387–401.
90. Struszczyk, H., Niekraszewicz, A., and Pospieszny, H., i in, EP 1211939 i PL 189890 B1.
91. Niekraszewicz, A., Wiśniewska‐Wrona, M., Struszczyk, H., Wesołowska, E., and Polka, N.:
Opracowanie kompozycji wybranej formy chitozanu z ligninami jako preparatu do ochrony i stymula-
cji wzrostu roślin, praca w ramach podstawowej działalności statutowej, 2002.
92. Niekraszewicz, A, Wiśniewska‐Wrona, M., Kopania, E., Orlikowski, L., Pospieszny, H., and Krawczyk,
K.: Biopolymer compositions for ecological protection and growth stimulation of plants. Progress on
Chemistry and Application of Chitin and Its Derivatives (PCACD), 2012, Vol. XVII, 145–158.
93. Ohta, K., Taniguchi, A., Konishi, N., and Hosoki, T.: Chitosan treatment affects plant growth and
flower quality in Eustoma grandiflorum. Hort Sci., 1999, 34(2), 233–234.
94. Cho, M.H., No, H.K., and Prinyawiwatkul, W.: Chitosan treatments affect growth and selected quality
of sunflower sprouts. J. Food Sci., 2008, 73(1), 70–77.
95. Falcon, A.B., Cabrera, J.C., Costales, D., Ramirez, M.A., Cabrera, G., Toledo, V., and Martinez‐
Tellez, M.A.: The effect of size and acetylation degree of chitosan derivatives on tobacco plant
protection against Phytophthora parasitica nicotianae. World J. Microbiol. Biotechnol., 2008,
24(1), 103–112.
96. Orlikowski, L. and Skrzypczak, Cz.: Biologiczna ochrona ozdobnych roślin cebulowych przed choro-
bami., Mat. konf. nauk. Techniki szklarniowe i rośliny cebulowe, 2000, 40–42, Skierniewice.
97. Orlikowski, L., Wojdyła, A., and Skrzypczak, Cz.: Elicitory w ochronie roślin przed grzybami choro-
botwórczymi. Mat. symp. nauk. Choroby roślin a środowisko, 1996, 99–106, Poznań.
Antimicrobial Properties of Chitin and Chitosan 187
98. Pięta, D., Pastucha, A., and Struszczyk, H.: Skutecznośc zaprawiania chitozanem nasion fasoli wielok-
wiatowej (Phaseolus coccineus L.) w ograniczaniu jej porażenia przez grzyby, Rocz. AR Pozn. 323
Ogrodn., 2000, 31(1), 409–413.
99. Borkowski, J., Sobolewski, J., and Robak, J.: Wpływ chitozanu na zdrowotnośc ́ i wzrost sałaty odm.
Zimowa Nansena., Mat. konf. nauk., Biologiczne i agrotechniczne kierunki rozwoju warzywnictwa,
2001, 158–159, Skierniewice.
8
Enzymes for Modification of
Chitin and Chitosan
Gustav Vaaje‐Kolstad, Tina Rise Tuveng, Sophanit Mekasha, and
Vincent G.H. Eijsink
Faculty of Chemistry, Biotechnology, and Food Science, The Norwegian University of Life Sciences
(NMBU), Ås, Norway
(CBM) class contains proteins with no enzymatic activity. CBMs are normally covalently
attached to enzymes and their primary function is to promote substrate binding [2].
Chitin
1 2 3
LPMOs Chitinases Deacetylases
AA10 GH18 CE4
AA11 GH19 CE14
AA15
OR
GlcNAc GlcN-GlcNAc (GlcN)2
exo-β-glucosaminidases
GH2
GH9
GH35
GlcNAc-GlcNAc1A
OR
GlcNAc GlcN
Figure 8.1 Schematic illustration of enzymatic pathways for degradation of chitin. The dominant CAZy
families responsible for performing each step in the chitin degradation pathways are indicated next to the
arrows. Chitin‐active lytic polysaccharide monooxygenases (LPMOs) cleave glycosidic bonds in crystal-
line regions of chitin using an oxidative mechanism, producing oxidised CHOS (lactones that are in
equilibrium with their aldonic acids; pathway 1). Chitinases and β‐N‐hexosaminidases catalyse the con-
version of the oxidised products to GlcNAc and the aldonic acid GlcNAc–GlcNAc1A (chitobionic acid;
this compound is relatively resistant to hydrolysis by β‐N‐hexosaminidases [5]). The grey colour of the
aldonic acid product symbolises the low amount of this product compared to native GlcNAc. Chitinases
convert polymeric and oligomeric chitin chains to GlcNAc–GlcNAc (chitobiose), which will be con-
verted to GlcNAc by β‐N‐hexosaminidases (pathway 2). The modification of chitin through deacetylation
of the C‐2 carbon is catalysed by deacetylases and produces chitosan (which may vary in terms of the
degree of deacetylation). Chitosan will then be degraded to mixed dimeric products containing acety-
lated and deacetylated sugars (GlcN–GlcN, GlcN–GlcNAc or GlcNAc–GlcN). Further degradation of
these dimers to either GlcN and/or GlcNAc is catalysed by glucosamine specific exo‐β‐glucosaminidases.
Note that the separation of chitinases and chitosanases is not absolute; many chitinases will also cleave
a wide range of chitosans (e.g. [6]).
Enzymes for Modification of Chitin and Chitosan 191
8.1.1 Chitinases
Chitinases, enzymes that hydrolyse the β-1,4 glycosidic bonds in chitin chains, are found
in GH families 18 and 19 [7]. GH18s have a (β/α)8‐barrel fold [8, 9], while GH19 chitinases
mainly comprise α‐helices [10]. GH18 chitinases occur in a wide variety of organisms and
have been studied more extensively than GH19 chitinases, which were originally discov-
ered in plants, but also occur in actinomycetes and other bacteria. The two enzyme types
use different catalytic mechanisms. GH18s use a substrate‐assisted mechanism that is
retaining [11], the latter meaning that the anomeric configuration is retained. GH19s use a
classical inverting mechanism, that is, the anomeric configuration is inverted [12]. Bond
cleavage by inverting enzymes proceeds through a single displacement mechanism, con-
trary to the double displacement mechanism in retaining enzymes. Both mechanisms use
general acid catalysis and require a pair of carboxylic acids. One carboxylic acid acts as the
catalytic acid, whereas the other acts as a water‐activating base in the inverting mechanism
or as a nucleophile in the retaining mechanism. The retaining mechanism entails the forma-
tion of an intermediate that is hydrolysed by water that is activated by the deprotonated
catalytic acid. Both reactions proceed through oxacarbenium‐ion‐like transition states [13].
GH18 family chitinases use a special version of the retaining mechanism, namely a
substrate‐assisted mechanism (Figure 8.2), where the oxygen of the N‐acetyl group in the
substrate acts as the nucleophile, leading to the formation of an oxazolinium ion intermedi-
ate. In GH18 enzymes, the catalytic acid is a glutamate, which is located in a diagnostic
DXDXE sequence motif that occurs in all active GH18 chitinases [14–16].
Chitinases degrade chitin chains from one of the chain ends (exo‐mechanism) or from a
random point on the chain (endo‐mechanism). In addition, the endo‐ or exo‐activity can be
combined with processivity, meaning that the enzyme stays attached to the substrate after
a successful cleavage. The enzyme thus slides along the chitin chain, making several suc-
cessive cleavages before it detaches from the substrate [12, 17]. Processive and non‐processive
enzymes have been studied in detail, revealing both sequence and structural differences
that underlay processivity and its direction [17–19]. Importantly, variation in these features
is most prominent, and has been studied most extensively, in the GH18 chitinases, which
are more involved in microbial chitin turnover in nature and show more structural variation
compared to GH19 enzymes.
During growth on chitin, the well‐known chitinolytic Gram‐negative bacterium Serratia
marcescens produces mainly three chitinases (SmChiA, SmChiB, and SmChiC), of which
SmChiA and SmChiB are processive exo‐enzymes, while SmChiC is a non‐processive
endo‐enzyme. The structures (Figure 8.3) of these proteins show that SmChiA and SmChiB
have deep active site clefts, contrary to the shallow and open active site in SmChiC. The
deep clefts of the two processive enzymes are defined by several loops, and a small sub‐
domain that occurs only in a subset of GH18 enzymes (including SmChiA and SmChiB, but
not SmChiC) and that has been named the α+β domain [8]. Aromatic residues lining the
surface of the active site cleft form another characteristic feature of processive enzymes.
One of these aromatic amino acids is next to a highly conserved SXGG sequence motif,
which is followed by a Trp in processive enzymes [19]. These aromatic residues likely help
the enzyme to stay attached to the substrate as it moves along the chain. Horn et al. [6] and
Zakariassen et al. [18] mutated several of the aromatic residues in SmChiB and SmChiA,
respectively, to non‐aromatic residues and showed that some of these mutations almost
192 Chitin and Chitosan: Properties and Applications
O O
Asp215 NH
O O
HO
NH
O HO O O
O O
O HO OH
HO O H
NH O O
HO
O O
HO O Tyr214
O O
N H O
Asp140 Asp140
O H O O O H
CH3
O O H OH O H
Asp142 O H
O
Asp142 O
Glu144 Glu144
NH
HO O
Asp215 HO
O
HO
O OH
O O O
OH
HO NH H H
O
O O
O H
O
HO
Tyr214
O
O
Asp140 N H O
O H O O H
Asp140 O CH3
O O H
Asp142 O H Asp142 O
O
O O
Glu144 Glu144
Figure 8.2 The substrate‐assisted mechanism used by GH18 chitinases. Binding of the substrate leads to
distortion of the sugar bound in subsite –1 towards a boat conformation. Simultaneously, Asp142 rotates
and forms hydrogen bonds with the catalytic Glu144 and the acetamido group of the sugar in the –1
subsite. At this point, Glu144 acts as a general acid by protonating the glycosidic oxygen, which supports
leaving group departure (i.e. cleavage of the glycosidic bond) that is further promoted by nucleophilic
attack of the acetamido group on the anomeric carbon, which leads to formation of an oxazolinium ion
intermediate. Glu144 then acts as a general base, activating an incoming water molecule that hydrolyses
the oxazolinium ion. The product is released from the active site and Asp142 rotates back to its original
conformation. Notably, if the water molecule hydrolysing the oxazolinium ion is outcompeted by another
acceptor, transglycosylation will occur instead of hydrolysis [23, 24]. Amino acid numbering is based on
chitinase B (SmChiB) from Serratia marcescens. The figure was obtained from [16] and permission was
granted through International Association of Scientific, Technical & Medical Publishers (STM).
Enzymes for Modification of Chitin and Chitosan 193
(a)
(b)
(c)
Figure 8.3 Structures of Serratia marcescens chitinases. The left figures show (a) SmChiA (PDB id 1CTN),
(b) SmChiB (PDB id 1E15) and (c) SmChiC (PDB id 4AXN) in cartoon representation. The α+β domain
present in SmChiA and SmChiB is shown in pink. Extra domains that promote substrate binding in SmChiA
(FnIII) and SmChiB (CBM5) are shown in green, as is a small β‐hairpin domain in SmChiC possibly aiding
substrate binding. Note that SmChiC has an FnIII and a CBM5/12 domain, but structural data for these
domains are not available. The catalytic Glu is shown as sticks with blue carbons. The figures in the middle
show the chitinases in surface representation, with aromatic residues lining the active site highlighted in
orange and the catalytic Glu in blue. The right figures show the differences in depth of the active site clefts.
The figure was made using PyMol.
abolished processivity while maintaining or even improving catalytic activity towards solu-
ble substrates. Of note, this large and functionally relevant structural variation among the
catalytic domains of GH18 chitinases, is not observed for GH19 chitinases, although some
minor variation in the substrate binding cleft has been observed, which is due to variation
in the lengths of certain loops [20–22] (Figure 8.4).
194 Chitin and Chitosan: Properties and Applications
Glu70
Figure 8.4 The structure of the GH19 chitinase BcChi‐A from Bryum coronatum. Chitotetraose, shown in
stick representation with green‐coloured carbon atoms, can be observed binding to the −2 to +2 subsites.
The catalytic base (Glu70) is shown in stick representation and is coloured with yellow carbon atoms. The
catalytic acid (Glu61) is mutated to alanine in this structure and therefore not shown. A deeper analysis of
the structure of this enzyme and its putative catalytic mechanism can be found in [22].
It is worth noting that chitinases in families GH18 and GH19 also act on chitosan, as
documented in various studies [6, 18, 25–27]. Expectedly, the efficiency of the enzymes
decreases as the degree of acetylation (FA) decreases. Furthermore, due to the substrate‐
assisted nature of the catalytic mechanism, GH18 enzymes only cleave the chitin chain
when an acetylated monosaccharide is placed in the –1 subsite.
Experimental determination of the processivity and the endo‐ or exo‐nature of a chi-
tinase is challenging. For GH18 chitinases, insight can be obtained from studies with highly
acetylated chitosan as shown [17]. When using water‐soluble chitosans, processive SmChiA
and SmChiB mainly produce even‐numbered oligosaccharides, while the endochitinase
SmChiC produces equal amounts of even‐ and odd‐numbered chitooligosaccharides
(CHOS) [17, 26, 28]. The production of even‐numbered oligosaccharides by processive
chitinases is expected, as an N‐acetyl group in subsite –1 is essential for catalysis and the
repetitive unit in chitin and chitosan is a dimer [16]. Initial productive binding of the sub-
strate to the enzyme will yield products of any length. If the enzyme is processive, it will
move by two monosaccharide moieties at a time, until a new productive complex is formed,
meaning that any further products resulting from the same initial enzyme‐substrate asso-
ciation will be even‐numbered. Non‐processive enzymes will detach and rebind in between
each reaction, thus yielding a continuum of product lengths. The exo‐ or endo‐activity of
a chitinase can be determined by measuring the reduction of viscosity during reactions
with chitosan. An endo‐enzyme cutting randomly along the chitosan chain will lead to fast
Enzymes for Modification of Chitin and Chitosan 195
reduction of viscosity [29]. On the contrary, an exo‐enzyme cutting from the chain ends
will lead to slow reduction of viscosity [28]. Further considerations concerning the analysis
of processivity are provided in [29].
8.1.2 β‐N‐acetylhexosaminidases
The most dominant product arising from chitin degradation by GH18 and GH19 chitinases
is (GlcNAc)2. To further convert (GlcNAc)2 to GlcNAc, most chitinolytic enzyme systems
contain a GH20 β‐N‐acetylhexosaminidase (also known as chitobiase), but other enzymes
performing similar reactions are also known (e.g. in families GH3 and GH84). The chito-
biase cleaves off the non‐reducing end sugar of CHOS using a catalytic mechanism similar
to the substrate‐assisted mechanism used by chitinases [11, 30]. Since most chitinases
yield (GlcNAc)2 as their primary product, this also represents the primary substrate for
chitobiases. However, it is well‐known that chitobiases are capable of efficiently removing
GlcNAc residues from the non‐reducing end of longer CHOS [30].
The structure of the chitobiase from S. marcescens (Figure 8.5) reveals the presence of
four domains: I, II, III and IV. Domain III accommodates the catalytic site whereas the
roles of domains I, II and IV are not known. Domain I shares some features with a CBM,
but the chitobiase binds only weakly to chitin. Domains II and IV lack similarity to any
domain/enzyme with a known function [16, 31].
Interestingly, like the GH18s, the catalytic domain of chitobiase (domain III) has a (β/α)8
fold, and catalysis is retaining and substrate‐assisted. Still, the catalytic site is different in
that the catalytic acid (Glu540) is directly adjacent to the aspartate that is co‐responsible
for positioning and activating the N‐acetyl group of the sugar bound in the –1 subsite
(Asp539) (Figure 8.5). Despite these differences, the determination of the complex struc-
ture as depicted in Figure 8.5, showing a substrate spanning the active site [31] was instru-
mental in unravelling a similar mechanism in the GH18 chitinases [11]. The substrate‐binding
site of chitobiase contains aromatic and charged amino acids that are responsible for sub-
strate binding (Figure 8.5).
8.1.3 Exo‐β‐glucosaminidases
Glucosamine‐specific exo‐β‐glucosaminidases (EC 3.2.1.165) are found in families GH2,
GH9 and GH35 [32] and could play a role in the chitosan pathway for chitin degradation
(Figure 8.1, pathway 3). Family GH2 and GH35 enzymes exploit retaining catalytic
mechanisms while family GH9 enzymes use the inverting mechanism. Only a few
exo‐β‐glucosaminidases have been characterised. Among these are bacterial GH2 exo‐β‐
glucosaminidases from Amycolatopsis orientalis named CsxA [33], and a bacterial GH9,
from Photobacterium profundum named PpGlcNase [34], which have been characterised
both functionally and structurally. The structures of these exo‐β‐glucosaminidases show a
narrow substrate‐binding pocket that is shaped by negatively charged residues that are
assumed to interact with the amine group of the substrate. The GH2 exo‐β‐glucosaminidases
consist of an incomplete (β/α)8 barrel and a domain with immunoglobulin‐like topology
consisting of four β‐sandwiches. The GH9 exo‐β‐glucosaminidases consist of an N‐terminal
domain with an FnIII‐type fold and unknown function and a catalytic C‐terminal (α/α)6
barrel. The structures of these enzymes in complex with GlcN revealed distinct interactions
196 Chitin and Chitosan: Properties and Applications
IV
E739 D671
R349 W685
Y669
II
+1
–1
W737
W616
III W639
D539 E540
Figure 8.5 Structure of the chitobiase from Serratia marcescens in complex with (GlcNAc)2. The overall
structure of the chitobiase (shown on the left) shows the presence of four domains; I, II, III and IV. Domain
I, II and III are associated to one another whereas domain IV protrudes relative to the other three domains.
The upper right panel shows the substrate binding pocket (shown as a light grey surface) and (GlcNAc)2
(magenta‐coloured sticks); the sugar bound in the –1 subsite, that is the non‐reducing end of the chitobi-
ose substrate, is buried deep in the protein, whereas the more visible sugar bound in the +1 subsite is
exposed to the solvent. The lower right panel shows details of the interaction between (GlcNAc)2 and cata-
lytic site residues. The (GlcNAc)2 is shown as magenta‐coloured sticks and residues from the chitobiase
are shown as light grey sticks. The catalytic acid (E540) is shown in yellow in all figures. The figure was
obtained from [16] and permission was granted through STM.
of active site residues with the substrate. CsxA (Figure 8.6a) displays a dominance of acidic
residues interacting with the hydroxyl groups and the amino group of the GlcN moiety. In
PpGlcNase, interactions involving aromatic residues seem to play a more prominent role
(Figure 8.6b).
In addition, a fungal GH2 exo‐β‐glucosaminidase from Trichoderma reesei and two
GH35 exo‐β‐glucosaminidases from the hyper‐thermophilic archaea Pyrococcus horiko-
shii and Thermococcus kodakarensis have been functionally characterised. The two
archaeal GH35 enzymes are homodimeric enzymes. All reports on the functionality of
exo‐β‐glucosaminidases belonging to families GH2, GH9 and GH35 conclude that the
enzymes act at the non‐reducing end of CHOS [33–37].
Interestingly, extensive studies of various enzymes from T. kodakarensis have led to the
conclusion that this microbe indeed uses the ’chitosan’ pathway for chitin saccharification
(see Section 8.4.3). In short, the organism degrades chitin to GlcNAc–GlcNAc using a
chitinase and the non‐reducing terminal sugar is then specifically deacetylated by a
Enzymes for Modification of Chitin and Chitosan 197
(a)
D203 E394
O
O H D469
O
W642 O
H
NH O OH
O
NH3
OH
H O
N O
O O O
W204 E541
D469
E394
E541
(b) E555
Q448 (acid)
Y445 N524
D143
(base)
W446
H150 W557
F231 D139
Q161
Y147
Figure 8.6 Structure of the active sites of CsxA (a) and PpGlcNase (b) in complex with GlcN. Panel A
shows the structure of CsxA in a complex with GlcN. The left figure is a schematic illustration of how the
enzyme interacts with GlcN; H‐bonds are indicated by dashed lines. The right‐side figure shows the pocket
accommodating the GlcN; the side chains of three acidic residues are shown in yellow and the grey
dashed lines represent H‐bonds with distances shown in angstrom. Asp469 is the catalytic acid/base and
Glu541 is the catalytic nucleophile. Figure source: [33], permission of use granted by the publisher. Panel
B shows the superimposed structures of PpGlcNase in a complex with GlcN complex (green) and of ligand
free PpGlcNase (magenta). H‐bonds are indicated by dashed lines. Glu555 acts as the catalytic acid (pos-
sibly via a water molecule), and Asp143, with help from Asp139, acts as the catalytic base. The figure has
been obtained from [34] and permission is granted through STM.
8.1.4 Chitosanases
Chitosanases are found in the GH5, GH7, GH8, GH46, GH75 and GH80 families of the
CAZy database [32, 39]. Among these, families GH5, 7 and 8 contain a few chitosanases,
while families GH 46, 75 and 80 exclusively contain chitosanases [39]. GH 5 and 7 chi-
tosanases utilise retaining catalytic mechanisms, while enzymes belonging to families
198 Chitin and Chitosan: Properties and Applications
(a) (b)
Asp55 Asp40 Glu309
Glu122
Glu22
Glu37
Figure 8.7 Structure of family GH46 chitosanases. Panel A shows the structures of GH46 enzymes from
Bacillus sp. MHK1 (left) and from Streptomyces sp. N174 (right); the arrows indicate the substrate binding
clefts. The extra loop protruding from the ’upper’ domain of the MHK1 chitosanase is coloured yellow. See
the text and [46] for more details. Panel B shows the structure of a GH8 chitosanase from Bacillus sp. K17.
This structure comprises a double‐(α/α)6 barrel. The side chains of the catalytic residues are shown in stick
representation and labelled.
these residues is about 10 Å, which is a typical distance for enzymes with an inverting
mechanism [45, 46, 52, 53].
The crystal structure of a family GH8 chitosanase from Bacillus sp. K17 shows an (α/α)6
double barrel and long loops containing small β‐strands and 310 helices that protrude from
the helices and form a substrate binding cleft and catalytic centre on top of the barrel.
Despite structural differences between the GH46 and GH8 enzymes, the distance between
the catalytic residues is similar in all these chitosanases. Similar to the N174 and MK1
chitosanases, the substrate‐binding cleft of the K17 chitosanase contains acidic residues
that are likely to interact with positive charges on the substrate. Aromatic residues with the
potential to interact with the substrate are also present in the active cleft [43].
OH Lactone
O
(a)
O C1-oxidation R
OH H2 O 2 HO
NH
or O
O NH
R HO R O2 + 2H+ + 2e– O
HO O
NH O LPMO O
AA10
OH
O AA11 NH
C4-oxidation ? HO R
AA15
O
O
(b) OH
Figure 8.8 Catalytic centre and reaction mechanism of LPMOs. (a) Reaction mechanism of chitin‐active
LPMOs showing possible C1 and C4 oxidised products (see text for details; C4 oxidation of chitin has not
been observed for chitin‐active LPMOs, but has been documented for other substrates with a β‐1,4 glucose
backbone, such as cellulose). (b) The catalytic centre in LPMOs exemplified by BaLPMO10A from Bacillus
amyloliquefaciens (PDB id 2YOX) showing the T‐shaped coordination (called the histidine brace; [59]) of
the Cu‐ion (brown sphere). The two conserved histidines, coordinating with the Cu‐ion, are shown as
sticks with pink‐coloured carbon atoms. One of these histidines is the N‐terminal residue of the protein.
oxidising, strictly C4 oxidising, or be able to oxidise both the C1 and C4 position [64, 65].
Independent of regioselectivity, LPMOs depend on copper‐reducing equivalents [59, 61] to
reduce this copper and dissolved molecular dioxygen in order to perform catalysis.
However, in a recent publication, Bissaro et al. (2017) [66] questioned the validity of diox-
ygen being the LPMO co‐substrate and provided compelling evidence indicating that
hydrogen peroxide (H2O2) is the true co‐substrate of LPMOs. Notably, LPMOs can them-
selves generate H2O2 from O2 [67].
O HN N+ O HN N+ H O HN N R
H R
– HN – R – H2N
HN
O O O
Asp391 C HN Asp391 HN Asp391 CH3 HN
O
H3 H3C O
Tyr367 Tyr367 Tyr367
Asp275 H OH Asp275 OH Asp275 OH O
O OH OH
–
O O O
H2N Zn H 2N + Zn H2N Zn
+ His326 +
NH2 Asp276 NH2 Asp276 His326 NH2 Asp276 His326
HN Arg364 His330 HN Arg364 His330 HN Arg364 His330
Figure 8.9 Reaction mechanism of CE4 deacetylases. The figure shows a proposed acid/base reaction
mechanism for CE4 deacetylases that remove N‐acetyl groups. The amino acid numbering is based on
SpPgdA. See the main text for a detailed description of the catalytic mechanism. The figure was obtained
from [76]. Copyright (2005) National Academy of Sciences, U.S.A.
(CHOS). The CE4 family also contains enzymes capable of removing O‐linked acetyl
groups from acetyl xylan [69] and several family members are known to act both on xylan
(O‐deacetylation) and chitin (N‐deacetylation [70–73]). The activity and structural features
of CE4 deacetylases are discussed in more detail in the following text.
The first deacetylase acting on chitin was found in extracts from the fungus Mucor rouxii
[74]. However, a few years earlier, Araki and co‐workers described an enzyme from
Bacillus cereus that deacetylates GlcNAc units in peptidoglycan [75]. Both these enzymes
are today classified in the carbohydrate esterase family 4. CE4 deacetylases removing the
N‐acetyl group from GlcNAc units share five conserved sequence motifs: motif 1, T(F/x)
DD; motif 2, H(S/T)xxH; motif 3, R(P/x)PY; motif 4, (Dxx)D(W/Y); motif 5, LxH [76].
Blair et al. (2005) [76] first proposed a catalytic mechanism for family CE4 deacetylases,
which, notably, depends on a bound metal ion, preferably zinc or cobalt [76–78]. They sug-
gested a general acid/base reaction mechanism based on an extensive structural analysis
(Figure 8.9). In this catalytic cycle, the catalytic base (the first Asp in motif 1) activates a
metal‐bound water molecule, which subsequently performs a nucleophilic attack on carbon
in the scissile C‐N bond creating a tetrahedral oxyanion intermediate. The metal ion and
backbone nitrogen of tyrosine in motif 3 stabilises the negative charge on the carbonyl
oxygen. The catalytic acid (His in motif 5) protonates the nitrogen in the substrate generat-
ing a free amine on the deacetylated product and leads to release of acetate [76, 78].
Motif 1, 2, and 5 are highly conserved between different deacetylases, while motif 3 and
4 display more sequence variation (Figure 8.10). The Asp‐His‐His metal binding triad is
located in motif 1 and 2 (Figure 8.10 and Figure 8.11, panel A). Motif 3 and 4 form one side
of the active site groove each (Figure 8.11, panel A [76, 78]).
As shown in Figure 8.10, some CE4 members have big insertions, representing loops
that are located close to the active site (Figure 8.11, panel B). Andrés et al. (2014) [78]
proposed a ’subsite capping model’ involving six loops (indicated in Figure 8.10) that each
cap parts of the active site cleft. Such loops would contribute to substrate specificity and
could endorse the deacetylase with sequence specificity because they could define which
substrates bind to the enzymes and which GlcNAc unit in the substrate becomes deacety-
lated. The family CE4 representative, VcCDA, from Vibrio cholera, is special in that these
loops are particularly long and form a buried active site (Figure 8.11, panel B). It was fur-
ther suggested that these loops may rearrange depending on the length of the substrate [78].
202 Chitin and Chitosan: Properties and Applications
Figure 8.10 Structure‐based sequence alignment of CE4 enzymes. The five conserved sequence motifs
are indicated with dark purple background. The yellow asterisks indicate the metal binding triad, while a
red triangle and circle indicate the catalytic base and acid, respectively. The deacetylases included in the
alignment are: SpPgdA, peptidoglycan deacetylase from Streptococcus pneumoniae [77]; AnCDA, chitin
deacetylase from Aspergillus nidulans [73]; SlCE4, acetyl xylan deacetylase from Streptomyces lividans
[77]; BsPdaA, peptidoglycan deacetylase from Bacillus subtilis [84]; ClCDA, chitin deacetylase from
Colletotrichum lindemuthianum [80]; VcCDA, chitin deacetylase from Vibrio cholerae [78]. Loop number-
ing and colouring was taken from [78]. The alignment was prepared using PyMod 1.0 [85].
Indeed, VcCDA is a highly specific enzyme that is restricted to deacetylate the GlcNAc
next to the non‐reducing end in CHOS. It is important to note that most other deacetylases
in this family, including the other structurally characterised ones (Figure 8.10), have shorter
loops and, hence, more open active sites (Figure 8.11, panel C). These enzymes generally
show less specificity compared to VcCDA, deacetylating a variety of substrates at several
positions [73, 76, 79–81]. Hence, CE4 deacetylases are generally considered to have broad
substrate specificity [70, 73]. For example, a deacetylase from Aspergillus nidulans
(AnCDA), having an open active site, is able to deacetylate all GlcNAc units in a chitohexa-
ose and also shows activity towards acetyl xylan [73].
The GlcNAc residue to be deacetylated binds in subsite 0, and apart from the interac-
tions between the enzyme and substrate in subsite 0, little experimental data for enzyme‐
substrate interactions of other subsites exist. The crystal structure of VcCDA in complex
with (GlcNAc)2 and (GlcNAc)3 was determined by Andrés et al. (2014) [78]. The dimer
occupies subsite 0 and –1, while the trimer occupies subsite –1 to +1, with the non‐reducing
end in subsite –1 in both cases. VcCDA makes several interactions with the sugar in subsite –1,
and binding of a sugar in subsite +1 requires rearrangement of several loops to allow its
binding. As pointed out above and illustrated in Figure 8.11, the structure of VcCDA is very
Enzymes for Modification of Chitin and Chitosan 203
(a) (b)
MT3
MT4 MT2
MT5
MT1
(c) (d)
W148
H173
D32
D33 Y123
H82
H86
Figure 8.11 Structure of CE4 deacetylases. (a) ClCDA in cartoon representation zooming in on the
active site. The side chains of the most important residues from the five catalytically important sequence
motifs are shown as sticks. The metal ion (blue sphere) is coordinated in an octahedral fashion (black
dashed lines) by the metal binding triad, a water molecule (red sphere), and an acetate ion (sticks with
pink carbons). (b) Surface representation of VcCDA (PDB id 4NY2) highlighting the loops that are pro-
posed to be involved in the substrate‐capping model. (c) Surface representation of ClCDA (PDB id
2IW0), showing the open active site. The loops are coloured according to the colour scheme used in
Figure 8.10: loop 1 in yellow, loop 2 in blue, loop 3 in red, loop 4 in orange, loop 5 in green and loop 6
in black. The metal ion is shown as a grey sphere. (d) The active site of ArCE4A in cartoon representation
with the GlcNAc2 substrate (yellow‐coloured carbon) bound in the active site. The active site residues
involved in substrate binding and catalysis are shown in stick representation with grey‐coloured carbon
atoms. Black dashed lines indicate hydrogen bonds. The figure was made using PyMol [86].
different compared to other CE4 enzymes with a known structure. Blair et al. (2006) [80]
performed a docking of (GlcNAc)3 bound in subsite –1 to +1 in ClCDA, which indicated
that there are no interactions between the enzyme and sugar bound in subsite –1.
This is different from what Andrés et al. (2014) [78] found for VcCDA. Recently, the
structure of ArCE4A was determined in complex with (GlcNAc)2 occupying subsite
204 Chitin and Chitosan: Properties and Applications
0 and +1 (Figure 8.11D [82]). Interestingly, the authors used (GlcNAc)4 as substrate in the
crystallisation, however, no interactions between the enzyme and substrate was observed
beyond subsite 0 and +1, similar to the findings in [83].
From an applied point of view, utilisation of deacetylases for production of chitosans
and CHOS with a defined degree of polymerisation (PA) is of great interest since the PA
together with the FA influences the biochemical properties of chitosan and CHOS [87, 88].
In this context, VcCDA and NodB from Rhizobium sp. GRH2 have been utilised to pro-
duce CHOS that are deacetylated specifically at the non‐reducing end and the neighbour-
ing sugar unit [88]. Recently, a fungal deacetylase from Puccinia graminis was shown to
deacetylate all GlcNAc units in different CHOS, except the two at the non‐reducing end
[89], thus providing another type of specificity. Importantly, Hembach et al. have recently
demonstrated the production of a series of CHOS with defined sequences by using com-
binations of deacetylases with varying specificities [81]. Notably, the use of deacetylases
to directly convert non‐soluble polymeric chitin into soluble polymeric chitosan has been
regularly presented as an attractive biocatalytic scenario, but has not yet been
accomplished.
sequences [99]. Many chitinases, chitosanases and LPMOs contain one or more additional
domains and the various domain compositions can be extracted from Pfam.
The most common domains appended to these enzyme types are non‐catalytic domains
known as CBMs. CBMs contribute to substrate binding and make sure that the enzyme
does not wander too far off from its substrate [90, 100, 100]. The positive impact of CBMs
on overall enzyme efficiency is well documented in literature.
Other domains often found include the fibronectin type III (FnIII) domain, an evolution-
ary conserved domain found in many extracellular enzymes [102, 103]. This domain is
found in chitinases, chitosanases, LPMOs and many other GHs (Figure 8.12), but its role
is not always well established and may relate both to substrate‐binding and positioning of
the other domains [98, 99, 104].
Interestingly, a wide variety of non‐catalytic and catalytic domains may be appended to
chitinases, chitosanases and LPMOs and the function of some of these domains is unclear.
For example, the RicinB lectin occurring in some chitosanases was originally found in
seeds of castor bean, Ricinus communis. RicinB lectin domains are found in a wide variety
of CAZymes and also in proteases and are known to bind mono‐sugars, lactose and galac-
tose [105–107]. The F5/F8 type domains seen in some chitosanases, also known as discoi-
din domains, are found in many blood coagulation factors [108]. DUF 2272 and 4157 are
domains of unknown function.
The domain architectures shown in Figure 8.12 also demonstrate that several GH
domains may occur in the same protein, which is not uncommon. Only a few such enzymes
have been studied, but the general impression is that such multi‐catalytic enzymes may be
quite effective [109, 110]. Furthermore, Figure 8.12 shows that GH domains may also be
coupled to other catalytic domains that are not GHs or LPMOs. The PD1 (Polysaccharide
deacetylase 1) domain is a family CE4 CAZyme containing the NodB domain and likely
responsible for deacetylation of CHOS. The GT2 (glycosyltransferase family 2) domain
likely synthesizes glycosidic bonds and family GT2 members include cellulose, chitin and
CHOS synthases [111]. PGB (peptidoglycan binding) domains occur in many enzymes
involved in bacterial cell‐wall degradation [112, 113]. CHAP (cysteine, histidine‐dependent
amidohydrolases/peptidases) domains are found in several amidase families and are
assumed to hydrolyse peptidoglycans [114, 115]. Most of the enzymes having complex
domain architectures discussed in this section have not been biochemically characterised,
which implies that enzyme diversity available for chitin conversion and modification still
remains a large untapped resource with a great future potential.
(a)
(b)
(c)
(d)
Figure 8.12 Schematic illustration of modular architectures of chitinolytic enzymes. GH18 chitinases
(red; a), GH19 chitinases (orange; b), GH8, GH46 and GH75 chitosanases (black; c) and AA10 LPMOs
(green; d). The figure was made by gathering data from the Pfam database. Pfam accessions numbers:
GH18, PF00704; GH8, PF01270; GH46, PF01374, GH75, PF07335, AA10 and PF03067. The numbers
shown in parenthesis indicate the number of sequences found in the Pfam database with the shown modu-
lar architecture. See text for further details.
Enzymes for Modification of Chitin and Chitosan 207
while the acidic mammalian chitinase has gained attention due to its possible link to the
pathophysiology of asthma [119]. In fungi, chitinases are postulated to have a wide variety
of functions including degradation of exogenous chitin for nutrition, remodelling of the
(own) fungal cell wall (which contains chitin), and defence against other fungi and arthro-
pods [120]. Fungal chitinases have also been proposed to act as virulence factors in insect
pathogenic fungi [121]. The number of chitinases encoded by fungi varies from 1 to over
30, making it an extensive task to determine the exact role of each chitinase [122, 123]. In
plants, chitinases, primarily of the GH19 type, are important in the defence against fungal
attacks [124, 125]. Bacteria generally use chitinases to degrade chitin for utilising it as a
nutrient source [126, 127]. However, there are indications that bacterial chitinases have
additional roles, based on putative activities on non‐chitin substrates such as glycoproteins
[128, 129]. Of note, the Listeria monocytogenes chitinase, ChiA, has been shown to con-
tribute to suppression of host innate immunity [130].
Chitobiases have several biological roles, dependent on the organism and even the cell
type. In addition to participating in chitin catabolism, several additional biological func-
tions of bacterial chitobiases have been proposed. For example, a chitobiase in Escherichia
coli contributes to cell wall recycling by hydrolysing the β‐1,4‐linkage between GlcNAc
and anhydro‐N‐acetylmuramic acid [131]. In the biofilm‐forming bacterium
Actinobacillus actinomycetemcomitans, a chitobiase is important for detachment of cells
from the biofilm in order to enable spreading of the biofilm to other surfaces [132]. In
fungi, chitobiases have several biological roles, especially in controlling the composition
of chitin in the cell wall. Chitobiase activity is also important for nutrient release during
the saprophytic and mycoparasitic growth phases of fungi and may be involved in insect
pathogenesis [132].
Chitosanases are found in bacteria, fungi and plants, having different biological func-
tions [133]. Some organisms are thought to secrete chitosanases to degrade chitosan and
utilise this as a nutrition source [134]. Saito et al. (2009) [135] demonstrated that a GH46
chitosanase from Amycolatopsis sp. CsO‐2 has antifungal activity against Rhizopus oryzae
and could thus be part of a defence system. Chitosanases may also have a role in protection
against the well‐known antimicrobial activity of chitosan and CHOS [136].
Chitin‐active LPMOs are mostly involved in the process of chitin catabolism (i.e.
making crystalline chitin more amenable to enzymatic depolymerisation), but some are
(also) suggested to act as virulence factors. Listeria monocytogenes possesses two
chitinases and an LPMO is possibly involved in the pathogenesis of this bacterium
[57, 137]. In V. cholerae, an LPMO termed GbpA binds mucin, and thereby enhances
bacterial colonisation of the intestine [138]. Mucin consists of glycoproteins, which are
glycosylated with different carbohydrates including GlcNAc [139]; it is not yet known
whether the LPMO domain of GbpA acts on the mucins, but its activity on chitin has
been proven [5]. Another example of an LPMO potentially involved in bacterial viru-
lence is SmLPMO10A from S. marcescens. Kawada et al. (2008) [140] showed that
knocking out this protein significantly decreased adhesion of the bacterium to colonic
epithelial cells, and suggested that SmLPMO10A and similar proteins are involved in
bacterial adhesion to such cells.
Deacetylases in the CE4 family are believed to have a role in pathogenesis of Gram‐positive
bacteria, since they, by modifying the peptidoglycan layer, make the bacteria less sus-
ceptible to the host innate immune system [141, 142]. The CE4 peptidoglycan deacetylase
208 Chitin and Chitosan: Properties and Applications
–1 +1
Chitobiase
Chi
–1 B ChiA
+1 +1
–1
CBM
5/12 CBP21 Fnlll
NR R
–1 +1
GlcNAc
ChiC
GlcNAcA
Figure 8.13 The chitinolytic machinery of Serratia marcescens. The chitinases SmChiA, SmChiB, and
SmChiC hydrolyse glycosidic bonds in different fashions. SmLPMO10A (named CBP21 in the figure) is an
LPMO making oxidative cuts in the crystalline parts of the chitin. The chitobiase degrades soluble products
generated by the chitinases and LPMO into monomers. Note that SmChiC has an FnIII and a CBM5/12
domain and that the chitobiase is a four‐domain protein; see text for details. Figure is obtained from [16]
and permission is granted through STM.
despite these findings, the exact secretion mechanism and the role of the holin/endopepti-
dase system remain unclear. Interestingly, the genes encoding SmLPMO10A and SmChiB
are localised in a gene locus dedicated to chitin metabolism. This gene locus also contains
the holin and endopeptidase genes, a LysR transcriptional regulator involved in regulation
of chitinase expression and two spanin genes [164, 165]. The chitobiase, whose expression
is induced by the presence of (GlcNAc)2, is located in the periplasm [166], although small
amounts of the enzyme have been detected in culture supernatants [167]. Figure 8.14 shows
a proposed model for the chitin utilisation pathway of S. marcescens. Generally, the
Chitin
O
OH
NH H
H O
O HO O
HO O
O
NH
OH
O n
SmChi18A, B- & C
Extracellular
SmLPMO10A
O
OH OH
NH O
O
HO HO OH Chitobiase HO
HO O HO OH
O NH
NH
OH
O O
O OH
OH
O
Periplasm
NH
O
HO OH Chitobiase HO
HO OH
HO
HO O NH
O
NH
OH O
O
HO O OH
P
O
putative
HO O
O phoshpo- OH P
O NH chitobiase O O
HO OH HO
HO
Cytoplasm
HO O HO OH O
O NH
NH HO
HO O
OH NH
O O
O
GlcNAc
kinase
Aminosugar
catabolism
Figure 8.14 Catabolic pathway for chitin utilisation in Serratia marcescens. Dashed arrows indicate
minor pathways. It should be noted that the oxidised CHOS formed by the activity of the LPMO and
SmLPMO10A are not shown in this figure. The catabolic fate of oxidised CHOS (i.e. aldonic acids) is not
known. For a detailed discussion of chitin catabolism by S. marcescens, see [167].
Enzymes for Modification of Chitin and Chitosan 211
ChiA
ChiB
CusD
CusC
GH20
TonB- CusS
ExbBD IM
complex transp.
P P P
NagK
GlcNAc GlcNAc-6P GlcN-6P Fru-6P
Figure 8.15 Proposed pathway for chitin utilisation in Flavobacterium johnsoniae. The secreted multi‐
domain ChiA converts chitin to CHOS. ChiB can also degrade CHOS and chitin, although not as efficiently
as ChiA. The SusC/D‐like pairs (here called CusC/D) capture and import CHOS to the periplasm. The
chitobiase (GH20) converts CHOS to monomers before translocation of the monomers to the cytoplasm
where they are converted to fructose‐6‐phosphate by NagK, A and B. Proteins not encoded by the PUL are
coloured grey. This figure has been obtained from [110], an original BioMed Central (BMC) publication.
hydrolyses the GlcN–GlcNAc dimer. Finally, Tk‐Dac deacetylates the GlcNAc monomer
to generate GlcN [38].
O O
OH OH
H O HO NH O H Tk-ChiA O HO NH
O HO OH
O O
HO HO
NH O O
NH
OH n OH
O O
Tk-Dac
O O
OH OH
O NH O NH
HO OH Tk-Glm HO
HO + HO OH
HO OH HO O
O HO
NH2 NH2 O
OH OH
Tk-Dac
Figure 8.16 The chitinolytic pathway of Thermococcus kodakarensis. Tk‐ChiA converts chitin to
(GlcNAc)2, which is deacetylated on the non‐reducing end by Tk‐Dac. Tk‐GlmA then hydrolyses the
GlcN–GlcNAc dimer and Tk‐Dac finally deacetylates GlcNAc to produce GlcN.
Chitinases in fungi are sometimes divided into three subgroups, A, B and C, based on their
modular structures. Group A contains chitinases without extra domains, group B contains
chitinases linked to CBM1s, while group C comprises chitinases with CBM18s and LysM
(CBM 50) domains [120, 181]. Of note, chitin is an important part of the fungal cell wall,
which also contains β‐1,3‐glycan and a manno‐protein layer [182].
As mentioned in Section 8.3, fungal chitinases are involved in cell wall remodelling and
in degradation of exogenous chitin, and an often raised question is how the fungi discrimi-
nate between self and non‐self chitin. It has been proposed that different chitinases are
specialised to do different jobs. Gruber and Seidl‐Seiboth (2012) [122] hypothesised that
differentiation between self and non‐self is regulated by substrate accessibility [120, 152].
The high number of chitinases makes genetic assessment of chitinase functionality a chal-
lenging task. It has been shown that double or multiple knock‐outs are needed to get a
phenotypic effect indicating possible redundancy among these many fungal chitinases
[123, 183].
Some fungi contain chitin‐active LPMOs [184], which likely play a role in chitin conver-
sion, but whose roles in chitin degradation and interplay with chitinases have not yet been
assessed. Several fungal chitin deacetylases have also been characterised (see Section 8.1.6)
and are believed to be involved in cell wall remodelling during growth [185] or to protect
hyphae during host invasion (plant pathogenic fungi [186]).
cocktails tailored for industrial use, which could entail a need for using non‐natural pro-
cessing conditions, such as high temperature, low pH and a high dry matter content.
All recalcitrant polysaccharide‐containing biomasses require pretreatment in order to
increase the surface area of the biomass and thereby obtain efficient enzymatic saccharifi-
cation. Chitin‐containing biomass is usually deproteinised and demineralised in order to
isolate the chitin (Aye and Stevens, [208] 2004). Depending on the application and particu-
larly if the goal is to produce chitosan, an extra alkaline pretreatment step is needed [195].
Mechanical size‐reduction of the chitin particles has proven to be successful for further
improvement of degradability [56].
Subsequent enzymatic hydrolysis of the purified chitin or chitosan will generate the
desirable oligomeric or monomeric products. So far, enzymatic saccharification of chitin
and chitosan has received little attention and has been mainly focused on bacterial enzymes.
There are no commercial products available for efficient saccharification of chitin or chi-
tosan. In order to obtain efficient and low‐cost enzymatic conversion of chitin, optimisation
of enzyme mixtures and characterisation of more chitin‐targeting enzymes seems to be
required, as shown and discussed by Mekasha et al. (2017) [209].
It is important to note that low‐molecular‐weight chitin and chitosan, that is, CHOS,
have received attention in different fields due to reported bioactivities. CHOS with distinct
physiochemical properties such as molecular weight, FA and acetylation pattern have poten-
tial in different applications in medicine and agriculture [87]. Further research on the enzy-
matic conversion of chitin‐rich biomass should consider the production of CHOS and full
saccharification will not always be the key target of a chitin conversion process. The direct
production of well‐defined CHOS from chitin by a combination of enzymatic deacetyla-
tion and (controlled) hydrolysis is an attractive scenario for valorisation of chitin‐rich bio-
mass [87, 210], which, however, has not yet been achieved.
Enzymatic conversion of chitin has not been explored in the same scale as cellulose. In
order to put the efforts of chitin biorefining in perspective, it is useful to briefly review the
status of the cellulose biorefining field, which resembles chitin biorefining in many ways
(abundant feedstock, recalcitrant and insoluble, and valuable for industrial use, with one
bottleneck being the efficiency and cost of enzyme cocktails).
In recent years, efficient processes for the conversion of lignocellulosic biomass have
been developed, and these are currently being used commercially. Industrial conversion of
lignocellulosic biomass to streams of mono‐sugars (primarily cellulose‐derived glucose)
involves a substrate pretreatment step that alters structural features of lignocellulose, breaks
the ‘lignin‐barrier’ and promotes substrate accessibility for subsequent enzymatic hydroly-
sis. The pretreated material is then hydrolysed using either cell‐free enzyme systems or
microbes. The resulting sugars are used in different applications, one of the best known
being the production of ethanol [211–213]. The term ’consolidated bioprocessing’ is used
when enzymatic depolymerisation and fermentation of the solubilised sugars are performed
simultaneously, meaning that the pretreated feedstock is directly converted to the final
product, for example ethanol, in a one‐pot reaction.
Enzymatic hydrolysis of lignocellulosic biomass is commonly performed using complex
enzyme mixtures, which may be crude or composed of purified enzymes. Cellulolytic
enzymes from fungal strains such as T. reesei show high hydrolytic potential on ligno-
cellulosic biomass and are among the best known cellulases, which were prominent in
early commercial enzyme cocktails [214]. Today’s enzyme cocktails are so efficient that
216 Chitin and Chitosan: Properties and Applications
commercial production of lignocellulosic ethanol is feasible, with one cause being the
inclusion of LPMOs [215–218].
An important lesson to be learned from the field of cellulose biorefining is that fungal
enzymes dominate. Essentially all commercially available enzyme cocktails developed for
degradation of cellulose‐containing biomass are based on fungal enzymes. Interestingly,
few fungal chitin‐degrading enzymes (and chitin‐degrading fungi) have been characterised
in detail. If fungal chitin‐degrading enzymes show characteristics similar to fungal cellu-
lose degrading enzymes, they may represent a great potential for future chitin biorefining.
It is important to note the potential of chitin‐degrading enzymes from thermophilic
archae, as reviewed by Egorova and Antranikian (2005) [219] and described in more detail
earlier, for T. kodakarensis (Section 8.3). Such enzymatic systems have superior properties
due to the extreme stability of enzymes that allow chitin conversion at temperatures where
potentially contaminating bacteria do not grow. Furthermore, enzymes from thermophiles
are often also tolerant to extreme pressure and pH, which may be beneficial for industrial
processes.
8.7 Outlook
In conclusion, while today we know a lot about enzymes for the modification of chitin and
chitosan, the industrial use of these enzymes for biorefining of chitin‐rich biomass is still
in its infancy. Importantly, next to the development of efficient enzymatic processes for
controlled conversion of chitin or chitosan to monomeric sugars or CHOS, truly ’green’
processes for chitin biorefining would also require the development of enzymatic or micro-
bial pretreatment techniques. Such techniques should replace the current chemical tech-
nologies that are needed to convert the original complex feedstock into reasonably clean
chitin or chitosan, which can then be used for further enzymatic valorisation.
References
1. Lombard, V., Bernard, T., Rancurel, C., Brumer, H., Coutinho, P.M., and Henrissat, B. 2010. A hierar-
chical classification of polysaccharide lyases for glycogenomics. Biochem. J., 432, 437–444.
2. Cantarel, B.L., Coutinho, P.M., Rancurel, C., Bernard, T., Lombard, V., and Henrissat, B. 2009. The
Carbohydrate‐Active EnZymes database (CAZy): An expert resource for Glycogenomics. Nucl. Acids
Res., 37, D233–D238.
3. Levasseur, A., Drula, E., Lombard, V., Coutinho, P.M., and Henrissat, B. 2013. Expansion of the enzy-
matic repertoire of the CAZy database to integrate auxiliary redox enzymes. Biotechnol. Biofuels, 6,
41.
4. Vaaje‐Kolstad, G., Westereng, B., Horn, S. J., Liu, Z., Zhai, H., Sørlie, M., and Eijsink, V. G. H. 2010.
An oxidative enzyme boosting the enzymatic conversion of recalcitrant polysaccharides. Science, 330,
219–222.
5. Loose, J. S., Forsberg, Z., Fraaije, M. W., Eijsink, V. G. H., and Vaaje‐Kolstad, G. 2014. A rapid quan-
titative activity assay shows that the Vibrio cholerae colonization factor GbpA is an active lytic poly-
saccharide monooxygenase. FEBS Lett., 588, 3435–3440.
6. Horn, S. J., Sikorski, P., Cederkvist, J. B., Vaaje‐Kolstad, G., Sørlie, M., Synstad, B., Vriend, G.,
Vårum, K. M., and Eijsink, V. G. H. 2006. Costs and benefits of processivity in enzymatic degradation
of recalcitrant polysaccharides. Proc. Natl. Acad. Sci., USA, 103, 18089–18094.
7. Henrissat, B. and Bairoch, A. 1993. New families in the classification of glycosyl hydrolases based on
amino acid sequence similarities. Biochem. J., 293, 781–788.
Enzymes for Modification of Chitin and Chitosan 217
8. Perrakis, A., Tews, I., Dauter, Z., Oppenheim, A. B., Chet, I., Wilson, K. S., and Vorgias, C. E. 1994.
Crystal structure of a bacterial chitinase at 2.3 Å resolution. Structure, 2, 1169–1180.
9. van Scheltinga, A. C. T., Kalk, K. H., Beintema, J. J., and Dijkstra, B. W. 1994. Crystal structures of
hevamine, a plant defence protein with chitinase and lysozyme activity, and its complex with an inhibi-
tor. Structure, 2, 1181–1189.
10. Hart, P. J., Pfluger, H. D., Monzingo, A. F., Hollis, T., and Robertus, J. D. 1995. The refined crystal
structure of an endochitinase from Hordeum vulgare L. seeds at 1.8 Å resolution. J. Mol. Biol., 248,
402–413.
11. Tews, I., van Scheltinga, A. C. T., Perrakis, A., Wilson, K. S. and Dijkstra, B. W. 1997. Substrate‐
assisted catalysis unifies two families of chitinolytic enzymes. J. Am. Chem. Soc., 119, 7954–7959.
12. Davies, G. J. and Henrissat, B. 1995. Structures and mechanisms of glycosyl hydrolases. Structure, 3,
853–859.
13. Rye, C. S. and Withers, S. G. 2000. Glycosidase mechanisms. Curr. Opin. Chem. Biol., 4, 573–580.
14. van Aalten, D. M. F., Komander, D., Synstad, B., Gåseidnes, S., Peter, M. G., and Eijsink, V. G. H.
2001. Structural insights into the catalytic mechanism of a family 18 exo‐chitinase. Proc. Natl. Acad.
Sci., USA, 98, 8979–8984.
15. Gloster, T. M. and Davies, G. J. 2010. Glycosidase inhibition: Assessing mimicry of the transition
state. Org. Biomol. Chem., 8, 305–320.
16. Vaaje‐Kolstad, G., Horn, S. J., Sørlie, M., and Eijsink, V. G. H. 2013. The chitinolytic machinery of
Serratia marcescens – a model system for enzymatic degradation of recalcitrant polysaccharides.
FEBS J., 280, 3028–3049.
17. Horn, S. J., Sørbotten, A., Synstad, B., Sikorski, P., Sørlie, M., Vårum, K. M., and Eijsink, V. G. H.
2006. Endo/exo mechanism and processivity of family 18 chitinases produced by Serratia marcescens.
FEBS J., 273, 491–503.
18. Zakariassen, H., Aam, B. B., Horn, S. J., Vårum, K. M., Sørlie, M., and Eijsink, V. G. H. 2009. Aromatic
residues in the catalytic center of chitinase A from Serratia marcescens affect processivity, enzyme
activity, and biomass converting efficiency. J. Biol. Chem., 284, 10610–10617.
19. Payne, C. M., Baban, J., Horn, S. J., Backe, P. H., Arvai, A. S., Dalhus, B., Bjørås, M., Eijsink, V. G.
H., Sørlie, M., Beckham, G. T., and Vaaje‐Kolstad, G. 2012. Hallmarks of processivity in glycoside
hydrolases from crystallographic and computational studies of the Serratia marcescens chitinases. J.
Biol. Chem., 287, 36322–36330.
20. Hoell, I. A., Dalhus, B., Heggset, E. B., Aspmo, S. I., and Eijsink, V. G. H. 2006. Crystal structure and
enzymatic properties of a bacterial family 19 chitinase reveal differences from plant enzymes. FEBS
J., 273, 4889–4900.
21. Ohnuma, T., Numata, T., Osawa, T., Inanaga, H., Okazaki, Y., Shinya, S., Kondo, K., Fukuda, T., and
Fukamizo, T. 2012. Crystal structure and chitin oligosaccharide‐binding mode of a “loopful” family
GH19 chitinase from rye, Secale cereale, seeds. FEBS J., 279, 3639–3651.
22. Ohnuma, T., Umemoto, N., Nagata, T., Shinya, S., Numata, T., Taira, T., and Fukamizo, T. 2014.
Crystal structure of a ’loopless’ GH19 chitinase in complex with chitin tetrasaccharide spanning the
catalytic center. Biochim. Biophys. Acta, 1844, 793–802.
23. Williams, S. J. and Withers, S. G. 2000. Glycosyl fluorides in enzymatic reactions. Carbohydr. Res.,
327, 27–46.
24. Zakariassen, H., Hansen, M. C., Joranli, M., Eijsink, V. G. H., and Sørlie, M. 2011. Mutational effects
on transglycosylating activity of family 18 chitinases and construction of a hypertransglycosylating
mutant. Biochemistry, 50, 5693–5703.
25. Heggset, E. B., Tuveng, T. R., Hoell, I. A., Liu, Z., Eijsink, V. G. H., and Vårum, K. M. 2012.
Mode of action of a family 75 chitosanase from Streptomyces avermitilis. Biomacromolecules, 13,
1733–41.
26. Sørbotten, A., Horn, S. J., Eijsink, V. G. H., and Vårum, K. M. 2005. Degradation of chitosans with
chitinase B from Serratia marcescens. FEBS J., 272, 538–549.
218 Chitin and Chitosan: Properties and Applications
27. Ohnuma, T., Numata, T., Osawa, T., Mizuhara, M., Varum, K. M., and Fukamizo, T. 2011. Crystal
structure and mode of action of a class V chitinase from Nicotiana tabacum. Plant Mol. Biol., 75,
291–304.
28. Sikorski, P., Sørbotten, A., Horn, S. J., Eijsink, V. G. H., and Vårum, K. M. 2006. Serratia marcescens
chitinases with tunnel‐shaped substrate‐binding grooves show endo activity and different degrees of
processivity during enzymatic hydrolysis of chitosan. Biochemistry, 45, 9566–9574.
29. Horn, S. J., Sørlie, M., Vårum, K. M., Väljamäe, P., and Eijsink, V. G. H. 2012. Measuring processivity.
Methods Enzymol., 510, 69–95.
30. Drouillard, S., Armand, S., Davies, G. J., Vorgias, E. C., and Henrissat, B. 1997. Serratia marcescens
chitobiase is a retaining glycosidase utilizing substrate acetamido group participation. Biochem. J.,
328, 945–949.
31. Tews, I., Perrakis, A., Oppenheim, A., Dauter, Z., Wilson, K. S., and Vorgias, C. E. 1996. Bacterial
chitobiase structure provides insight into catalytic mechanism and the basis of Tay–Sachs disease. Nat.
Struct. Mol. Biol., 3, 638–648.
32. Lombard, V., Golaconda Ramulu, H., Drula, E., Coutinho, P. M., and Henrissat, B. 2014. The carbo-
hydrate‐active enzymes database (CAZy) in 2013. Nucl. Acids Res., 42, D490–D495.
33. van Bueren, A. L., Ghinet, M. G., Gregg, K., Fleury, A., Brzezinski, R., and Boraston, A. B. 2009. The
structural basis of substrate recognition in an exo‐β‐d‐Glucosaminidase involved in chitosan hydroly-
sis. J. Mol. Biol., 385, 131–139.
34. Honda, Y., Arai, S., Suzuki, K., Kitaoka, M., and Fushinobu, S. 2016. The crystal structure of an invert-
ing glycoside hydrolase family 9 exo‐β‐D‐glucosaminidase and the design of glycosynthase. Biochem.
J., 473, 463–472.
35. Nogawa, M., Takahashi, H., Kashiwagi, A., Ohshima, K., Okada, H., and Morikawa, Y. 1998.
Purification and characterization of Exo‐β‐d‐Glucosaminidase from a cellulolytic fungus, Trichoderma
reesei PC‐3‐7. Appl. Environ. Microbiol., 64, 890–895.
36. Tanaka, T., Fukui, T., Atomi, H., and Imanaka, T. 2003. Characterization of an exo‐β‐D‐glucosamini-
dase involved in a novel chitinolytic pathway from the hyperthermophilic Archaeon Thermococcus
kodakarensis KOD1. J. Bacteriol., 185, 5175–5181.
37. Liu, B., Li, Z., Hong, Y., Ni, J., Sheng, D., and Shen, Y. 2006. Cloning, expression and characterization
of a thermostable exo‐β‐D‐glucosaminidase from the hyperthermophilic archaeon Pyrococcus horiko-
shii. Biotechnol. Lett., 28, 1655–1660.
38. Tanaka, T., Fukui, T., Fujiwara, S., Atomi, H., and Imanaka, T. 2004. Concerted action of diacetylchi-
tobiose deacetylase and exo‐β‐D‐glucosaminidase in a novel chitinolytic pathway in the hyperthermo-
philic archaeon Thermococcus kodakarensis KOD1. J. Biol. Chem., 279, 30021–30027.
39. Eijsink, V. G. H., Hoell, I., and Vaaje‐Kolstad, G. 2010. Structure and function of enzymes acting on
chitin and chitosan. Biotechnol. Genet. Eng. Rev., 27, 331–366.
40. Aspeborg, H., Coutinho, P. M., Wang, Y., Brumer, H., and Henrissat, B. 2012. Evolution, substrate
specificity and subfamily classification of glycoside hydrolase family 5 (GH5). BMC Evol. Biol.,
12, 1.
41. Wang, S. L., Peng, J. H., Liang, T. W., and Liu, K. C. 2008. Purification and characterization of a chi-
tosanase from Serratia marcescens TKU011. Carbohydr. Res., 343, 1316–1323.
42. Wang, S. L., Tseng, W. N.,and Liang, T. W. 2011. Biodegradation of shellfish wastes and production
of chitosanases by a squid pen‐assimilating bacterium, Acinetobacter calcoaceticus TKU024.
Biodegradation, 22, 939–948.
43. Adachi, W., Sakihama, Y., Shimizu, S., Sunami, T., Fukazawa, T., Suzuki, M., Yatsunami, R.,
Nakamura, S., and Takénaka, A. 2004. Crystal structure of family GH‐8 chitosanase with subclass II
specificity from Bacillus sp. K17. J. Mol. Biol., 343, 785–795.
44. Boucher, I., Dupuy, A., Vidal, P., Neugebauer, W. A., and Brzezinski, R. 1992. Purification and
characterization of a chitosanase from Streptomyces N174. Appl. Microbiol. Biotechnol., 38,
188–193.
Enzymes for Modification of Chitin and Chitosan 219
45. Marcotte, E., Hart, P. J., Boucher, I., Brzezinski, R., and Robertus, J. D. 1993. Crystallization of a
chitosanase from Streptomyces N174. J. Mol. Biol., 232, 995–996.
46. Saito, J. I., Kita, A., Higuchi, Y., Nagata, Y., Ando, A., and Miki, K. 1999. Crystal structure of chitosan-
ase from Bacillus circulans MH‐K1 at 1.6‐Å resolution and its substrate recognition mechanism.
J. Biol. Chem., 274, 30818–30825.
47. Fukamizo, T., Honda, Y., Goto, S., Boucher, I., and Brzezinski, R. 1995. Reaction mechanism of chi-
tosanase from Streptomyces sp. N174. Biochem. J., 311, 377–383.
48. Izume, M., Nagae, S., Kawagishi, H., Mitsutomi, M., and Ohtakara, A. 1992. Action pattern of Bacillus
sp. No. 7‐M chitosanase on partially N‐acetylated chitosan. Biosci. Biotechnol. Biochem., 56,
448–453.
49. Mitsutomi, M., Ueda, M., Arai, M., Ando, A., and Watanabe, T. 1996. Action patterns of microbial
chitinases and chitosanases on partially N‐acetylated chitosan. Chitin Enzymol., 2, 273–284.
50. Heggset, E. B., Dybvik, A. I., Hoell, I. A., Norberg, A. L., Sørlie, M., Eijsink, V. G. H., and Vårum, K.
M. 2010. Degradation of chitosans with a family 46 chitosanase from Streptomyces coelicolor A3(2).
Biomacromolecules, 11, 2487–2497.
51. Pechsrichuang, P., Lorentzen, S. B., Aam, B. B., Tuveng, T. R., Hamre, A. G., Eijsink, V. G. H., and
Yamabhai, M. 2018. Bioconversion of chitosan into chito‐oligosaccharides (CHOS) using family 46
chitosanase from Bacillus subtilis (BsCsn46A). Carbohydr. Polym., 186, 420–428.
52. Wang, Q., Graham, R. W., Trimbur, D., Warren, R. A. J., and Withers, S. G. 1994. Changing enzymic
reaction mechanisms by mutagenesis: Conversion of a retaining glucosidase to an inverting enzyme. J.
Am. Chem. S, 116, 11594–11595.
53. Mccarter, J. D. and Withers, G. S. 1994. Mechanisms of enzymatic glycoside hydrolysis. Curr. Opin.
Struc. Biol., 4, 885–892.
54. Vaaje‐Kolstad, G., Horn, S. J., van Aalten, D. M. F., Synstad, B., and Eijsink, V. G. H. 2005. The non‐
catalytic chitin‐binding protein CBP21 from Serratia marcescens is essential for chitin degradation. J.
Biol. Chem., 280, 28492–28497.
55. Horn, S. J., Vaaje‐Kolstad, G., Westereng, B., and Eijsink, V. G. H. 2012. Novel enzymes for the deg-
radation of cellulose. Biotechnol. Biofuels, 5, 45.
56. Nakagawa, Y. S., Eijsink, V. G. H., Totani, K., and Vaaje‐Kolstad, G. 2013. Conversion of α‐chitin
substrates with varying particle size and crystallinity reveals substrate preferences of the chitinases and
lytic polysaccharide monooxygenase of Serratia marcescens. J. Agric. Food Chem., 61,
11061–11066.
57. Paspaliari, D. K., Loose, J. S. M., Larsen, M. H., and Vaaje‐Kolstad, G. 2015. Listeria monocytogenes
has a functional chitinolytic system and an active lytic polysaccharide monooxygenase. FEBS J., 282,
921–936.
58. Nakagawa, Y. S., Kudo, M., Loose, J. S., Ishikawa, T., Totani, K., Eijsink, V. G. H., and Vaaje‐Kolstad,
G. 2015. A small lytic polysaccharide monooxygenase from Streptomyces griseus targeting α‐and β‐
chitin. FEBS J., 282, 1065–1079.
59. Quinlan, R. J., Sweeney, M. D., Leggio, L. L., Otten, H., Poulsen, J.‐C. N., Johansen, K. S., Krogh, K.
B., Jørgensen, C. I., Tovborg, M., and Anthonsen, A. 2011. Insights into the oxidative degradation of
cellulose by a copper metalloenzyme that exploits biomass components. Proc. Natl. Acad. Sci., USA,
108, 15079–15084.
60. Forsberg, Z., Vaaje‐Kolstad, G., Westereng, B., Bunæs, A. C., Stenstrøm, Y., Mackenzie, A., Sørlie, M.,
Horn, S. J., and Eijsink, V. G. H. 2011. Cleavage of cellulose by a CBM33 protein. Protein Sci., 20,
1479–1483.
61. Phillips, C. M., Beeson IV, W. T., Cate, J. H., and Marletta, M. A. 2011. Cellobiose dehydrogenase and
a copper‐dependent polysaccharide monooxygenase potentiate cellulose degradation by Neurospora
crassa. ACS Chem. Biol., 6, 1399–1406.
62. Westereng, B., Ishida, T., Vaaje‐Kolstad, G., Wu, M., Eijsink, V. G. H., Igarashi, K., Samejima, M.,
Ståhlberg, J., Horn, S. J., and Sandgren, M. 2011. The putative endoglucanase PcGH61D from
220 Chitin and Chitosan: Properties and Applications
80. Blair, D. E., Hekmat, O., Schüttelkopf, A. W., Shrestha, B., Tokuyasu, K., Withers, S. G., and van
Aalten, D. M. F. 2006. Structure and mechanism of chitin deacetylase from the fungal pathogen
Colletotrichum lindemuthianum. Biochemistry, 45, 9416–9426.
81. Hembach, L., Cord‐Landwehr, S., and Moerschbacher, B. M. 2017. Enzymatic production of all four-
teen partially acetylated chitosan tetramers using different chitin deacetylases acting in forward or
reverse mode. Sci. Rep., 7, 17692.
82. Tuveng, T. R., Rothweiler, U., Udatha, G., Vaaje‐Kolstad, G., Smalås, A., and Eijsink, V. G. H. 2017.
Structure and function of a CE4 deacetylase isolated from a marine environment. PLoS One, 12,
e0187544.
83. Blair, D. E., Hekmat, O., Schüttelkopf, A. W., Shrestha, B., Tokuyasu, K., Withers, S. G., and van
Aalten, D. M. F. 2006. Structure and mechanism of chitin deacetylase from the fungal pathogen
Colletotrichum lindemuthianum. Biochemistry, 45, 9416–9426.
84. Blair, D. E. and van Aalten, D. M. F. 2004. Structures of Bacillus subtilis PdaA, a family 4 carbohy-
drate esterase, and a complex with N‐acetyl‐glucosamine. FEBS Lett., 570, 13–9.
85. Bramucci, E., Paiardini, A., Bossa, F., and Pascarella, S. 2012. PyMod: Sequence similarity searches,
multiple sequence‐structure alignments, and homology modeling within PyMOL. BMC Bioinformatics,
13, S2.
86. Schrödinger, L. L. C. 2015. The PyMOL Molecular Graphics System Version 1.8.
87. Aam, B.B., Heggset, E.B., Norberg, A. L., Sørlie, M., Vårum, K. M., and Eijsink, V. G. H. 2010.
Production of chitooligosaccharides and their potential applications in medicine. Marine Drugs, 8,
1482–1517.
88. Hamer, S. N., Cord‐Landwehr, S., Biarnes, X., Planas, A., Waegeman, H., Moerschbacher, B. M.,
and Kolkenbrock, S. 2015. Enzymatic production of defined chitosan oligomers with a specific
pattern of acetylation using a combination of chitin oligosaccharide deacetylases. Sci. Rep., 5,
8716.
89. Naqvi, S., Cord‐Landwehr, S., Singh, R., Bernard, F., Kolkenbrock, S., El Gueddari, N. E.,
and Moerschbacher, B. M. 2016. A recombinant fungal chitin deacetylase produces fully
defined chitosan oligomers with novel patterns of acetylation. Appl. Environ. Microbiol., 82,
6645–6655.
90. Boraston, A. B., Bolam, D. N., Gilbert, H. J., and Davies, G. J. 2004. Carbohydrate‐binding modules:
Fine‐tuning polysaccharide recognition. Biochem. J., 382, 769–781.
91. Várnai, A., Siika‐Aho, M., and Viikari, L. 2013. Carbohydrate‐binding modules (CBMs) revisited:
Reduced amount of water counterbalances the need for CBMs. Biotechnol. Biofuels, 6, 30.
92. Buist, G., Steen, A., Kok, J., and Kuipers, O. P. 2008. LysM, a widely distributed protein motif for
binding to (peptido)glycans. Mol. Microbiol., 68, 838–847.
93. Akcapinar, G. B., Kappel, L., Sezerman, O. U., and Seidl‐Seiboth, V. 2015. Molecular diversity of
LysM carbohydrate‐binding motifs in fungi. Curr. Genet., 61, 103–113.
94. Akagi, K.‐I., Watanabe, J., Hara, M., Kezuka, Y., Chikaishi, E., Yamaguchi, T., Akutsu, H., Nonaka, T.,
Watanabe, T., and Ikegami, T. 2006. Identification of the substrate interaction region of the chitin‐bind-
ing domain of Streptomyces griseus chitinase C. J. Biochem., 139, 483–493.
95. Viegas, A., Brás, N. F., Cerqueira, N. M., Fernandes, P. A., Prates, J. A., Fontes, C. M., Bruix, M.,
Romao, M. J., Carvalho, A. L., and Ramos, M. J. 2008. Molecular determinants of ligand specificity
in family 11 carbohydrate binding modules–an NMR, X‐ray crystallography and computational chem-
istry approach. FEBS J., 275, 2524–2535.
96. Katouno, F., Taguchi, M., Sakurai, K., Uchiyama, T., Nikaidou, N., Nonaka, T., Sugiyama, J., and
Watanabe, T. 2004. Importance of exposed aromatic residues in chitinase B from Serratia marcescens
2170 for crystalline chitin hydrolysis. J. Biochem., 136, 163–168.
97. Uchiyama, T., Katouno, F., Nikaidou, N., Nonaka, T., Sugiyama, J., and Watanabe, T. 2001. Roles of
the exposed aromatic residues in crystalline chitin hydrolysis by chitinase A from Serratia marcescens
2170. J. Biol. Chem., 276, 41343–41349.
222 Chitin and Chitosan: Properties and Applications
98. Uchiyama, T., Katouno, F., Nikaidou, N., Nonaka, T., Sugiyama, J., and Watanabe, T. 2001. Roles of
the exposed aromatic residues in crystalline chitin hydrolysis by chitinase A from Serratia marces-
cens 2170. J. Biol. Chem., 276, 41343–41349.
99. Finn, R. D., Coggill, P., Eberhardt, R. Y., Eddy, S. R., Mistry, J., Mitchell, A. L., Potter, S. C., Punta,
M., Qureshi, M., and Sangrador‐Vegas, A. 2016. The Pfam protein families database: Towards a
more sustainable future. Nucl. Acids Res., 44, D279–D285.
100. Hashimoto, H. 2006. Recent structural studies of carbohydrate‐binding modules. Cell Mol. Life Sci.,
63, 2954–2967.
101. Talamantes, D., Biabini, N., Dang, H., Abdoun, K., and Berlemont, R. 2016. Natural diversity of cel-
lulases, xylanases, and chitinases in bacteria. Biotechnol. Biofuels, 9, 133.
102. Bateman, A. and Chothia, C. 1996. Fibronectin type III domains in yeast detected by a hidden Markov
model. Curr. Biol., 6, 1544–1547.
103. Despres, J., Forano, E., Lepercq, P., Comtet‐Marre, S., Jubelin, G., Yeoman, C. J., Miller, M. E. B.,
Fields, C. J., Terrapon, N., and Bourvellec, C. 2016. Unraveling the pectinolytic function of
Bacteroides xylanisolvens using a RNA‐seq approach and mutagenesis. BMC Genomics, 17, 1.
104. Watanabe, T., Ito, Y., Yamada, T., Hashimoto, M., Sekine, S. and Tanaka, H. 1994. The roles of the
C‐terminal domain and type III domains of chitinase A1 from Bacillus circulans WL‐12 in chitin
degradation. J. Bacteriol., 176, 4465–4472.
105. Hazes, B. 1996. The (QxW)3 domain: A flexible lectin scaffold. Protein Sci., 5, 1490–1501.
106. Hazes, B. and Read, R. J. 1995. A mosquitocidal toxin with a ricin‐like cell‐binding domain. Nat.
Struct. Biol., 2, 358–359.
107. Hirabayashi, J., Dutta, S. K.,and Kasai, K. 1998. Novel galactose‐binding proteins in Annelida.
Characterization of 29‐kDa tandem repeat‐type lectins from the earthworm Lumbricus terrestris.
J. Biol. Chem., 273, 14450–14460.
108. Kane, W. H. and Davie, E. W. 1986. Cloning of a cDNA coding for human factor V, a blood coagula-
tion factor homologous to factor VIII and ceruloplasmin. Proc. Natl. Acad. Sci. U. S. A., 83,
6800–4.
109. Brunecky, R., Alahuhta, M., Xu, Q., Donohoe, B. S., Crowley, M. F., Kataeva, I. A., Yang, S. J.,
Resch, M. G., Adams, M. W. W., and Lunin, V. V. 2013. Revealing nature’s cellulase diversity: The
digestion mechanism of Caldicellulosiruptor bescii CelA. Science, 342.
110. Larsbrink, J., Zhu, Y., Kharade, S. S., Kwiatkowski, K. J., Eijsink, V. G. H., Koropatkin, N. M.,
Mcbride, M. J., and Pope, P. B. 2016. A polysaccharide utilization locus from Flavobacterium john-
soniae enables conversion of recalcitrant chitin. Biotechnol. Biofuels, 9, 260.
111. Coutinho, P. M., Deleury, E., Davies, G. J., and Henrissat, B. 2003. An evolving hierarchical family
classification for glycosyltransferases. J. Mol. Biol., 328, 307–317.
112. Dideberg, O., Charlier, P., Dive, G., Joris, B., Frère, J. M., and Ghuysen, J. M. 1982. Structure of a
Zn2+‐containing D‐alanyl‐D‐alanine‐cleaving carboxypeptidase at 2.5 Å resolution. Nature, 299,
469–470.
113. Krogh, S., Jorgensen, S., and Devine, K. 1998. Lysis genes of the Bacillus subtilis defective prophage
PBSX. J. Bacteriol., 180, 2110–2117.
114. Bateman, A. and Rawlings, N. D. 2003. The CHAP domain: A large family of amidases including
GSP amidase and peptidoglycan hydrolases. Trends Biochem. Sci., 28, 234–7.
115. Rigden, D. J., Jedrzejas, M. J., and Galperin, M. Y. 2003. Amidase domains from bacterial and phage
autolysins define a family of gamma‐D,L‐glutamate‐specific amidohydrolases. Trends Biochem. Sci.,
28, 230–234.
116. Hollak, C. E., van Weely, S., van Oers, M. H., and Aerts, J. M. 1994. Marked elevation of plasma
chitotriosidase activity. A novel hallmark of Gaucher disease. J. Clin. Invest., 93, 1288–1292.
117. Boot, R. G., Blommaart, E. F. C., Swart, E., Ghauharali‐van der Vlugt, K., Bijl, N., Moe, C., Place,
A. and Aerts, J. M. F. G. 2001. Identification of a novel acidic mammalian chitinase distinct from
chitotriosidase. J. Biol. Chem., 276, 6770–6778.
Enzymes for Modification of Chitin and Chitosan 223
118. van Eijk, M., van Roomen, C. P., Renkema, G. H., Bussink, A. P., Andrews, L., Blommaart,
E. F., Sugar, A., Verhoeven, A. J., Boot, R. G., and Aerts, J. M. 2005. Characterization of
human phagocyte‐derived chitotriosidase, a component of innate immunity. Int. Immunol.,
17, 1505–1512.
119. Zhu, Z., Zheng, T., Homer, R. J., Kim, Y.‐K., Chen, N. Y., Cohn, L., Hamid, Q., and Elias, J. A. 2004.
Acidic mammalian chitinase in asthmatic Th2 inflammation and IL‐13 pathway activation. Science,
304, 1678–1682.
120. Seidl, V. 2008. Chitinases of filamentous fungi: A large group of diverse proteins with multiple physi-
ological functions. Fungal Biol. Rev., 22, 36–42.
121. Huang, Z., Hao, Y., Gao, T., Huang, Y., Ren, S., and Keyhani, N. O. 2016. The Ifchit1 chitinase gene
acts as a critical virulence factor in the insect pathogenic fungus Isaria fumosorosea. Appl. Microbiol.
Biotechnol., 100, 5491–5503.
122. Gruber, S. and Seidl‐Seiboth, V. 2012. Self versus non‐self: Fungal cell wall degradation in
Trichoderma. Microbiology, 158, 26–34.
123. Langner, T. and Göhre, V. 2016. Fungal chitinases: Function, regulation, and potential roles in plant/
pathogen interactions. Current Genetics, 62, 243–254.
124. Broglie, K. and Chet, I. 1991. Transgenic plants with enhanced resistance to the fungal pathogen
Rhizoctonia solani. Science, 254, 1194–1197.
125. Collinge, D. B., Kragh, K. M., Mikkelsen, J. D., Nielsen, K. K., Rasmussen, U., and Vad, K. 1993.
Plant chitinases. Plant J., 3, 31–40.
126. Watanabe, T., Kimura, K., Sumiya, T., Nikaidou, N., Suzuki, K., Suzuki, M., Taiyoji, M., Ferrer, S.,
and Regue, M. 1997. Genetic analysis of the chitinase system of Serratia marcescens 2170. J.
Bacteriol., 179, 7111–7117.
127. Orikoshi, H., Nakayama, S., Miyamoto, K., Hanato, C., Yasuda, M., Inamori, Y. and Tsujibo, H. 2005.
Roles of four chitinases (ChiA, ChiB, ChiC, and ChiD) in the chitin degradation system of marine
bacterium Alteromonas sp. strain O‐7. Appl. Environ. Microbiol., 71, 1811–1815.
128. Adrangi, S. and Faramarzi, M. A. 2013. From bacteria to human: A journey into the world of chi-
tinases. Biotechnol. Adv., 31, 1786–1795.
129. Frederiksen, R. F., Paspaliari, D. K., Larsen, T., Storgaard, B. G., Larsen, M. H., Ingmer, H., Palcic,
M. M., and Leisner, J. J. 2013. Bacterial chitinases and chitin‐binding proteins as virulence factors.
Microbiology, 159, 833–847.
130. Chaudhuri, S., Gantner, B. N., Ye, R. D., Cianciotto, N. P., and Freitag, N. E. 2013. The Listeria
monocytogenes ChiA chitinase enhances virulence through suppression of host innate immunity.
mBio, 4, e00617–12.
131. Cheng, Q., Li, H., Merdek, K., and Park, J. T. 2000. Molecular characterization of the β‐N‐
Acetylglucosaminidase of Escherichia coli and its role in cell wall recycling. J. Bacteriol., 182,
4836–4840.
132. Slámová, K., Bojarová, P., Petrásková, L., and Křen, V. 2010. β‐N‐Acetylhexosaminidase: What’s in
a name? Biotechnol. Adv., 28, 682–693.
133. Thadathil, N. and Velappan, S. P. 2014. Recent developments in chitosanase research and its biotech-
nological applications: A review. Food Chem., 150, 392–399.
134. Viens, P., Dubeau, M.‐P., Kimura, A., Desaki, Y., Shinya, T., Shibuya, N., Saito, A., and Brzezinski,
R. 2015. Uptake of chitosan‐derived D‐glucosamine oligosaccharides in Streptomyces coelicolor
A3(2). FEMS Microbiol. Lett., 362, fnv048.
135. Saito, A., Ooya, T., Miyatsuchi, D., Fuchigami, H., Terakado, K., Nakayama, S.‐Y., Watanabe, T.,
Nagata, Y., and Ando, A. 2009. Molecular characterization and antifungal activity of a family 46
chitosanase from Amycolatopsis sp. CsO‐2. FEMS Microbiol. Lett., 293, 79–84.
136. Ghinet, M. G., Roy, S., Poulin‐Laprade, D., Lacombe‐Harvey, M.‐È., Morosoli, R. and Brzezinski, R.
2010. Chitosanase from Streptomyces coelicolor A3 (2): Biochemical properties and role in protec-
tion against antibacterial effect of chitosan. Biochem. Cell Biol., 88, 907–916.
224 Chitin and Chitosan: Properties and Applications
137. Chaudhuri, S., Bruno, J. C., Alonzo, F., Xayarath, B., Cianciotto, N. P., and Freitag, N. E. 2010.
Contribution of chitinases to Listeria monocytogenes pathogenesis. Appl. Environ. Microbiol., 76,
7302–7305.
138. Bhowmick, R., Ghosal, A., Das, B., Koley, H., Saha, D. R., Ganguly, S., Nandy, R. K., Bhadra, R. K.,
and Chatterjee, N. S. 2008. Intestinal adherence of Vibrio cholerae involves a coordinated interaction
between colonization factor GbpA and mucin. Infect. Immun., 76, 4968–4977.
139. Barchi, J. J. 2013. Mucin‐type glycopeptide structure in solution: Past, present, and future.
Biopolymers, 99, 713–723.
140. Kawada, M., Chen, C. C., Arihiro, A., Nagatani, K., Watanabe, T., and Mizoguchi, E. 2008. Chitinase
3‐like‐1 enhances bacterial adhesion to colonic epithelial cells through the interaction with bacterial
chitin‐binding protein. Lab Invest, 88, 883–895.
141. Boneca, I. G. 2005. The role of peptidoglycan in pathogenesis. Curr. Opin. Microbiol., 8, 46–53.
142. Zhao, Y., Park, R. D., and Muzzarelli, R. A. 2010. Chitin deacetylases: Properties and applications.
Marine Drugs, 8, 24–46.
143. Vollmer, W. and Tomasz, A. 2000. The pgdA gene encodes for a peptidoglycan N‐acetylglucosamine
deacetylase in Streptococcus pneumoniae. J. Biol. Chem., 275, 20496–20501.
144. Vollmer, W. and Tomasz, A. 2002. Peptidoglycan N‐acetylglucosamine deacetylase, a putative viru-
lence factor in Streptococcus pneumoniae. Infect. Immun., 70, 7176–7178.
145. El Gueddari, N. E., Rauchhaus, U., Moerschbacher, B. M., and Deising, H. B. 2002. Developmentally
regulated conversion of surface‐exposed chitin to chitosan in cell walls of plant pathogenic fungi.
New Phytol., 156, 103–112.
146. Cord‐Landwehr, S., Melcher, R. L. J., Kolkenbrock, S., and Moerschbacher, B. M. 2016. A chitin
deacetylase from the endophytic fungus Pestalotiopsis sp. efficiently inactivates the elicitor activity
of chitin oligomers in rice cells. Sci. Rep., 6, 38018.
147. Christodoulidou, A., Bouriotis, V., and Thireos, G. 1996. Two sporulation‐specific chitin deacetylase‐
encoding genes are required for the ascospore wall rigidity of Saccharomyces cerevisiae. J. Biol.
Chem., 271, 31420–31425.
148. Christodoulidou, A., Briza, P., Ellinger, A., and Bouriotis, V. 1999. Yeast ascospore wall assembly
requires two chitin deacetylase isozymes. FEBS Lett., 460, 275–279.
149. Hirano, T., Kadokura, K., Ikegami, T., Shigeta, Y., Kumaki, Y., Hakamata, W., Oku, T., and Nishio, T.
2009. Heterodisaccharide 4‐O‐(N‐acetyl‐β‐D‐glucosaminyl)‐D‐glucosamine is a specific inducer of
chitinolytic enzyme production in Vibrios harboring chitin oligosaccharide deacetylase genes.
Glycobiology, 19, 1046–1053.
150. Li, X., Wang, L.‐X., Wang, X., and Roseman, S. 2007. The chitin catabolic cascade in the marine
bacterium Vibrio cholerae: Characterization of a unique chitin oligosaccharide deacetylase.
Glycobiology, 17, 1377–1387.
151. Guillén, D., Sánchez, S., and Rodríguez‐Sanoja, R. 2010. Carbohydrate‐binding domains: Multiplicity
of biological roles. Appl. Microbiol. Biotechnol., 85, 1241–1249.
152. van den Burg, H. A., Harrison, S. J., Joosten, M. H., Vervoort, J. and de Wit, P. J. 2006. Cladosporium
fulvum Avr4 protects fungal cell walls against hydrolysis by plant chitinases accumulating during
infection. Mol. Plant‐Microbe Interact., 19, 1420–1430.
153. van Esse, H. P., Bolton, M. D., Stergiopoulos, I., de Wit, P. J., and Thomma, B. P. 2007. The chitin‐
binding Cladosporium fulvum effector protein Avr4 is a virulence factor. Mol. Plant‐Microbe
Interact., 20, 1092–1101.
154. de Jonge, R., van Esse, H. P., Kombrink, A., Shinya, T., Desaki, Y., Bours, R., van der Krol, S.,
Shibuya, N., Joosten, M. H. A. J., and Thomma, B. P. H. J. 2010. Conserved fungal LysM effector
Ecp6 prevents chitin‐triggered immunity in plants. Science, 329, 953–955.
155. Bai, Y., Eijsink, V. G. H., Kielak, A. M., van Veen, J. A. and de Boer, W. 2016. Genomic comparison
of chitinolytic enzyme systems from terrestrial and aquatic bacteria. Environ. Microbiol., 18, 38–49.
Enzymes for Modification of Chitin and Chitosan 225
156. van Aalten, D. M. F., Synstad, B., Brurberg, M. B., Hough, E., Riise, B. W., Eijsink, V. G. H. and
Wierenga, R. K. 2000. Structure of a two‐domain chitotriosidase from Serratia marcescens at 1.9‐Å
resolution. Proc. Natl. Acad. Sci., USA, 97, 5842–5847.
157. Hult, E.‐L., Katouno, F., Uchiyama, T., Watanabe, T. and Sugiyama, J. 2005. Molecular directionality
in crystalline β‐chitin: Hydrolysis by chitinases A and B from Serratia marcescens 2170. Biochem. J.,
388, 851–856.
158. Igarashi, K., Uchihashi, T., Uchiyama, T., Sugimoto, H., Wada, M., Suzuki, K., Sakuda, S., Ando, T.,
Watanabe, T., and Samejima, M. 2014. Two‐way traffic of glycoside hydrolase family 18 processive
chitinases on crystalline chitin. Nat. Commun., 5, 3975.
159. Horn, S. J., Sørlie, M., Vaaje‐Kolstad, G., Norberg, A. L., Synstad, B., Vårum, K. M., and Eijsink, V.
G. H. 2006. Comparative studies of chitinases A, B and C from Serratia marcescens. Biocatal.
Biotransformation, 24, 39–53.
160. Suzuki, K., Taiyoji, M., Sugawara, N., Nikaidou, N., Henrissat, B. and Watanabe, T. 1999. The third
chitinase gene (chiC) of Serratia marcescens 2170 and the relationship of its product to other bacte-
rial chitinases. Biochem. J., 343, 587.
161. Brurberg, M. B., Eijsink, V. G. H., and Nes, I. F. 1994. Characterization of a chitinase gene (chiA)
from Serratia marcescens BJL200 and one‐step purification of the gene product. FEMS Microbiol.
Lett., 124, 399–404.
162. Brurberg, M. B., Eijsink, V. G. H., Haandrikman, A. J., Venema, G. and Nes, I. F. 1995. Chitinase B
from Serratia marcescens BJL200 is exported to the periplasm without processing. Microbiology,
141, 123–131.
163. Suzuki, K., Suzuki, M., Taiyoji, M., Nikaidou, N., and Watanabe, T. 1998. Chitin binding protein
(CBP21) in the culture supernatant of Serratia marcescens 2170. Biosci. Biotechnol. Biochem., 62,
128–135.
164. Hamilton, J. J., Marlow, V. L., Owen, R. A., Costa Mde, A., Guo, M., Buchanan, G., Chandra, G.,
Trost, M., Coulthurst, S. J., Palmer, T., Stanley‐Wall, N. R., and Sargent, F. 2014. A holin and an
endopeptidase are essential for chitinolytic protein secretion in Serratia marcescens. J. Cell Biol.,
207, 615–26.
165. Suzuki, K., Uchiyama, T., Suzuki, M., Nikaidou, N., Regue, M. and Watanabe, T. 2001. LysR‐type
transcriptional regulator ChiR is essential for production of all chitinases and a chitin‐binding protein,
CBP21, in Serratia marcescens 2170. Biosci. Biotechnol. Biochem., 65, 338–347.
166. Tuveng, T. R., Hagen, L. H., Mekasha, S., Frank, J., Arntzen, M. Ø., Vaaje‐Kolstad, G., and Eijsink,
V. G. H. 2017. Genomic, proteomic and biochemical analysis of the chitinolytic machinery of
Serratia marcescens BJL200. Biochim. Biophys. Acta (BBA)‐Proteins Proteom., 1865, 414–421.
167. Toratani, T., Shoji, T., Ikehara, T., Suzuki, K., and Watanabe, T. 2008. The importance of chitobiase
and N‐acetylglucosamine (GlcNAc) uptake in N, N′‐diacetylchitobiose [(GlcNAc) 2] utilization by
Serratia marcescens 2170. Microbiology, 154, 1326–1332.
168. Hunt, D. E., Gevers, D., Vahora, N. M.,and Polz, M. F. 2008. Conservation of the chitin utilization
pathway in the Vibrionaceae. Appl. Environ. Microbiol., 74, 44–51.
169. Nazari, B., Kobayashi, M., Saito, A., Hassaninasab, A., Miyashita, K., and Fujii, T. 2013. Chitin‐
induced gene expression in secondary metabolic pathways of Streptomyces coelicolor A3(2) grown
in soil. Appl. Environ. Microbiol., 79, 707–713.
170. Thomas, F., Hehemann, J.‐H., Rebuffet, E., Czjzek, M., and Michel, G. 2011. Environmental and gut
Bacteroidetes: The food connection. Front. Microbiol., 2.
171. Martens, E. C., Koropatkin, N. M., Smith, T. J., and Gordon, J. I. 2009. Complex glycan catabolism
by the Human gut microbiota: The Bacteroidetes Sus‐like paradigm. J. Biol. Chem., 284,
24673–24677.
172. D’elia, J. N. and Salyers, A. A. 1996. Effect of regulatory protein levels on utilization of starch by
Bacteroides thetaiotaomicron. J. Bacteriol., 178, 7180–7186.
226 Chitin and Chitosan: Properties and Applications
173. Reeves, A. R., Wang, G., and Salyers, A. A. 1997. Characterization of four outer membrane pro-
teins that play a role in utilization of starch by Bacteroides thetaiotaomicron. J. Bacteriol., 179,
643–649.
174. Terrapon, N., Lombard, V., Gilbert, H. J., and Henrissat, B. 2015. Automatic prediction of polysac-
charide utilization loci in Bacteroidetes species. Bioinformatics, 31, 647–655.
175. Hemsworth, G. R., Déjean, G., Davies, G. J., and Brumer, H. 2016. Learning from microbial strate-
gies for polysaccharide degradation. Biochem. Soc. Trans., 44, 94–108.
176. Tanaka, T., Fujiwara, S., Nishikori, S., Fukui, T., Takagi, M. and Imanaka, T. 1999. A unique chitinase
with dual active sites and triple substrate binding sites from the hyperthermophilic archaeon
Pyrococcus kodakaraensis KOD1. Appl. Environ. Microbiol., 65, 5338–5344.
177. Tanaka, T., Fukui, T. and Imanaka, T. 2001. Different cleavage specificities of the dual catalytic
domains in chitinase from the hyperthermophilic archaeon Thermococcus kodakaraensis KOD1.
J. Biol. Chem., 276, 35629–35635.
178. Hanazono, Y., Takeda, K., Niwa, S., Hibi, M., Takahashi, N., Kanai, T., Atomi, H. and Miki, K. 2016.
Crystal structures of chitin binding domains of chitinase from Thermococcus kodakarensis KOD1.
FEBS Lett., 590, 298–304.
179. St Leger, R. J., Cooper, R. M. and Charnley, A. K. 1986. Cuticle‐degrading enzymes of
entomopathogenic fungi: Regulation of production of chitinolytic enzymes. Microbiology,
132, 1509–1517.
180. Xiao, G., Ying, S.‐H., Zheng, P., Wang, Z.‐L., Zhang, S., Xie, X.‐Q., Shang, Y., St. Leger, R. J., Zhao,
G.‐P., Wang, C., and Feng, M.‐G. 2012. Genomic perspectives on the evolution of fungal entomopath-
ogenicity in Beauveria bassiana. Sci. Rep., 2, 483.
181. Seidl, V., Huemer, B., Seiboth, B. and Kubicek, C. P. 2005. A complete survey of Trichoderma chi-
tinases reveals three distinct subgroups of family 18 chitinases. FEBS J., 272, 5923–5939.
182. Bowman, S. M. and Free, S. J. 2006. The structure and synthesis of the fungal cell wall. Bioessays,
28, 799–808.
183. Alcazar‐Fuoli, L., Clavaud, C., Lamarre, C., Aimanianda, V., Seidl‐Seiboth, V., Mellado, E. and
Latgé, J.‐P. 2011. Functional analysis of the fungal/plant class chitinase family in Aspergillus fumiga-
tus. Fungal Genet. Biol., 48, 418–429.
184. Hemsworth, G. R., Henrissat, B., Davies, G. J. and Walton, P. H. 2014. Discovery and characteriza-
tion of a new family of lytic polysaccharide monooxygenases. Nat. Chem. Biol., 10, 122–126.
185. Geoghegan, I. A. and Gurr, S. J. 2017. Investigating chitin deacetylation and chitosan hydrolysis dur-
ing vegetative growth in Magnaporthe oryzae. Cell Microbiol, 19.
186. El Gueddari, N. E., Rauchhaus, U., Moerschbacher, B. M. and Deising, H. B. 2002. Developmentally
regulated conversion of surface‐exposed chitin to chitosan in cell walls of plant pathogenic fungi.
New Phytol., 156, 103–112.
187. Jayakumar, R., Prabaharan, M., Sudheesh Kumar, P. T., Nair, S. V. and Tamura, H. 2011. Biomaterials
based on chitin and chitosan in wound dressing applications. Biotechnol. Adv., 29, 322–337.
188. Felt, O., Buri, P. and Gurny, R. 1998. Chitosan: A unique polysaccharide for drug delivery. Drug
Develop. Ind. Pharm., 24, 979–993.
189. Freier, T., Montenegro, R., Koh, H. S. and Shoichet, M. S. 2005. Chitin‐based tubes for tissue engi-
neering in the nervous system. Biomaterials, 26, 4624–4632.
190. Suh, J. K. F. and Matthew, H. W. T. 2000. Application of chitosan‐based polysaccharide biomaterials
in cartilage tissue engineering: A review. Biomaterials, 21, 2589–2598.
191. Mohire, N. C. and Yadav, A. V. 2010. Chitosan‐based polyherbal toothpaste: As novel oral hygiene
product. Indian J. Dent. Res., 21, 380.
192. Dutta, P. K., Dutta, J. and Tripathi, V. S. 2004. Chitin and chitosan: Chemistry, properties and applica-
tions. J. Sci. Ind. Res. India, 63, 20–31.
193. Kumar, M. N. V. R. 2000. A review of chitin and chitosan applications. React. Funct. Polym., 46,
1–27.
Enzymes for Modification of Chitin and Chitosan 227
194. Miretzky, P. and Cirelli, A. F. 2009. Hg(II) removal from water by chitosan and chitosan derivatives:
A review. J. Hazard Mater., 167, 10–23.
195. Peter, M. G. 1995. Applications and environmental aspects of chitin and chitosan. J. Macromol. Sci.
Pure, 32, 629–640.
196. Aider, M. 2010. Chitosan application for active bio‐based films production and potential in the food
industry: Review. LWT‐Food Sci. Technol., 43, 837–842.
197. Hungerford, D. S. and Jones, L. C. 2003. Glucosamine and chondroitin sulfate are effective in the
management of osteoarthritis. J. Arthroplasty, 18, 5–9.
198. Towheed, T. E. 2003. Current status of glucosamine therapy in osteoarthritis. Arthrit. Care Res., 49,
601–604.
199. Katiyar, D., Hemantaranjan, A., Singh, B., and Bhanu, N. A. 2014. A future perspective in crop pro-
tection: Chitosan and its oligosaccharides. Adv. Plants Agric. Res., 1.
200. Kong, M., Chen, X. G., Xing, K., and Park, H. J. 2010. Antimicrobial properties of chitosan and mode
of action: A state of the art review. Int. J. Food Microbiol., 144, 51–63.
201. Wendland, J., Schaub, Y., and Walther, A. 2009. N‐acetylglucosamine utilization by Saccharomyces
cerevisiae based on expression of Candida albicans NAG genes. Appl. Environ. Microbiol., 75,
5840–5845.
202. Yabushita, M. 2016. A Study on Catalytic Conversion of Non‐Food Biomass into Chemicals:
Fusion of Chemical Sciences and Engineering. Doctoral dissertation. Springer, ISBN
978‐981‐10‐0331‐8.
203. Oranusi, N. A. and Trinci, A. P. 1985. Growth of bacteria on chitin, fungal cell walls and fungal bio-
mass, and the effect of extracellular enzymes produced by these cultures on the antifungal activity of
amphotericin B. Microbios, 43, 17–30.
204. Vega, K. and Kalkum, M. 2012. Chitin, chitinase responses, and invasive fungal infections. Int. J.
Microbiol., 2012, 920459.
205. Melchers, L. S. and Stuiver, M. H. 2000. Novel genes for disease‐resistance breeding. Curr. Opin.
Plant Biol., 3, 147–152.
206. Grover, A. 2012. Plant chitinases: Genetic diversity and physiological roles. Crit. Rev. Plant Sci., 31,
57–73.
207. Singh, G., Bhalla, A., Singh Bhatti, J., Chandel, S., Rajput, A., Abdullah, A., Andrabi, W., and Kaur,
P. 2014. Potential of chitinases as a biopesticide against agriculturally harmful fungi and insects. Res.
Rev.: J. Microbiol. Biotechnol., 3, 27–32.
208. Aye, K. N. and Stevens, W. F. 2004. Improved chitin production by pretreatment of shrimp shells.
J. Chem. Tech. Biotechnol., 79, 421–425.
209. Mekasha, S., Byman, I. R., Lynch, C., Toupalová, H., Andě r a, L., Næs, T., Vaaje‐Kolstad, G.,
and Eijsink, V. G. H. 2017. Development of enzyme cocktails for complete saccharification
of chitin using mono‐component enzymes from Serratia marcescens. Process Biochem., 56,
132–138.
210. Younes, I. and Rinaudo, M. 2015. Chitin and chitosan preparation from marine sources. Structure,
properties and applications. Mar. Drugs, 13, 1133–1174.
211. Alvira, P., Tomás‐Pejó, E., Ballesteros, M., and Negro, M. J. 2010. Pretreatment technologies for an
efficient bioethanol production process based on enzymatic hydrolysis: A review. Bioresource
Technol., 101, 4851–4861.
212. Sánchez, Ó. J. and Cardona, C. A. 2008. Trends in biotechnological production of fuel ethanol from
different feedstocks. Bioresource Technol., 99, 5270–5295.
213. Jørgensen, H., Kristensen, J. B., and Felby, C. 2007. Enzymatic conversion of lignocellulose into
fermentable sugars: Challenges and opportunities. Biofuels Bioprod. Bior., 1, 119–134.
214. Herpoël‐Gimbert, I., Margeot, A., Dolla, A., Jan, G., Mollé, D., Lignon, S., Mathis, H., Sigoillot, J.
C., MonoT, F., and Asther, M. 2008. Comparative secretome analyses of two Trichoderma reesei
RUT‐C30 and CL847 hypersecretory strains. Biotechnol. Biofuels, 1, 1.
228 Chitin and Chitosan: Properties and Applications
215. Müller, G., Várnai, A., Johansen, K. S., Eijsink, V. G. H., and Horn, S. J. 2015. Harnessing the poten-
tial of LPMO‐containing cellulase cocktails poses new demands on processing conditions. Biotechnol.
Biofuels, 8, 187.
216. Fushinobu, S. 2014. Metalloproteins: A new face for biomass breakdown. Nat. Chem. Biol., 10,
88–89.
217. Harris, P. V., Xu, F., Kreel, N. E., Kang, C., and Fukuyama, S. 2014. New enzyme insights drive
advances in commercial ethanol production. Curr. Opin. Chem. Biol., 19, 162–170.
218. Johansen, K. S. 2016. Lytic Polysaccharide Monooxygenases: The microbial power tool for lignocel-
lulose degradation. Trends Plant Sci., 21, 926–936.
219. Egorova, K. and Antranikian, G. 2005. Industrial relevance of thermophilic Archaea. Curr. Opin.
Microbiol., 8, 649–655.
9
Chitin and Chitosan as Sources
of Bio‐Based Building Blocks
and Chemicals
Malgorzata Kaisler1,2, Lambertus A.M. van den Broek2, and
Carmen G. Boeriu2
Bioprocess Engineering Group, Wageningen University, Wageningen, The Netherlands
1
2
Wageningen Food & Biobased Research, Wageningen, The Netherlands
Chitin and chitosan polymers are a valuable source of functional chemicals and materials.
Chemical and/or enzymatic depolymerisation processes have been developed for the
production of chitooligosaccharides (COS), N‐acetylglucosamine (GlcNAc) and
glucosamine (GlcN), which have a wide variety of applications. New technologies are now
emerging to convert chitin and its derivatives into platform chemicals. Chemical liquefaction
of chitin can lead to bulk chemicals like acetic acid and platform chemicals like
hydroxymethylfurfural (HMF) and amine‐containing monomers for polymers, in low
yield. The monomers GlcNAc and GlcN can be converted into N‐containing HMF
derivatives, opening a pathway for furan‐based monomers for polyamides. Selective
catalytic oxidation of GlcN results in the production of D‐glucosaminic acid (DGA). This
acid is a valuable building block for the synthesis of various amino acids for biomedical
applications and bio‐based chiral polyamides. Further technological improvements are
necessary to increase the selectivity and efficiency of reactions, particularly for the
conversion of polymeric chitin and chitosan into building blocks.
9.1 Introduction
In the past decades, significant technological developments have been achieved in
the utilisation of lignocellulosic biomass as an alternative source to fossil fuels for the
production of chemicals and materials for the food, feed and chemical industry. The
production of building blocks for materials and chemicals from renewable biological
resources, such as plants, microorganisms and animals creates the opportunity to reduce
the emission of greenhouse gas and reduce our dependence on coal, oil and gas, and
constitutes as such the fundamental concept of bio‐based economy [1, 2]. Recent devel-
opments and regulations significantly enlarge the scope of bio‐based economy towards
a zero‐waste circular economy that extends the biomass source towards industrial
side streams and promotes the recycling of plastics and biodegradable materials in the
process cycle.
In this context, biomass side stream (biowastes) generated by different industries are
considered an attractive renewable biological resource for chemicals, building blocks and
materials. The recovery of valuable components such as biopolymers, fats and fine
chemicals, pigments and pharmaceuticals from biowastes decreases landfilling of wastes
and protects the environment. Moreover, conversion of cheap biowastes to added‐value
products can bring additional economic benefit [3]. Recovered biopolymers like proteins,
lignin, starch and chitin are of high importance, since they can be used directly as feed or
food or can be applied as a resource of building blocks for the production of non‐food bio‐
based products.
Chitin, one of the most abundant biopolymers on earth, is generated in huge amounts as
a by‐product (biowaste) from the food and chemical industry. Industrial sources of chitin
and chitosan are the side streams of the shell food industry, insect farms, mushroom
cultivation and the fungal fermentation industry (Figure 9.1).
So far, the seafood industry generates the biggest amounts of chitin‐containing biomass
(6–8 million tons/year produced globally) comprising the shells from crustaceans (e.g.
prawns, shrimps, crabs, lobsters) [4]. This biomass is often considered as a waste and is just
dumped in the sea or landfilled without any processing [5]. However, such a landfilling
leads to serious environmental pollution of coastal areas. Thus, in the 1970s, the USA
introduced strict environmental regulations and banned the dumping of waste shells without
any processing [6], which directly resulted in a significant increase of chitin utilisation. In
some countries, for example, in Australia, disposal of crustacean shells is costly and can be
up to €130 per tonne [5]. Crustacean shells from the food industry are processed by drying
and desizing (i.e. crushing and milling) and sold as a low‐value fertiliser at a much lower
price (€44/tonne) as compared to the price of dried crustacean shells (€85–102/tonne) [5].
However, chitin‐containing biomass has a much higher potential than only being a low‐
value fertiliser. A small part of chitin‐rich streams is used though for the extraction and
valorisation of chitin and chitosan, chitin’s most valuable derivative.
Besides chitin, chitin‐containing biomass comprises proteins, fats, glucans and pigments
(e.g. astaxanthin) (Figure 9.1). All the components can be separated in a biorefinery concept
using chemical and/or enzymatic extraction methods. Apart from food, the refined
components can be used as animal feed (e.g. proteins) or can be further converted into
valuable chemicals or materials. Lipids can be used in the production of high‐value oil,
biodiesel and animal feed [5]. Calcium carbonate (CaCO3) is applied in the pharmaceutical,
Cephalopods Crustaceans Insects Fungi
agricultural, construction and paper industries [5]. Astaxanthin, a carotenoid with antioxi-
dant properties, is widely used in aquaculture feeds, food, cosmetic, pharmaceutical and
medical applications [7]. Glucans recovered from fungi have potential application in food
and feed, medicine and pharmacy [8].
The first step of chitin valorisation is its extraction. Traditionally, chitin is extracted by
the removal of proteins (deproteinisation) with alkali treatment (e.g. with NaOH) and
removal of CaCO3 (demineralisation) with acidic treatment (e.g. HCl) [9, 10]. After
extraction, chitin is often decoloured with acetone or an organic acid solvent mixture [11].
This conventional fractionation process for chitin extraction is, however, destructive and
has a high environmental impact due to the use of high amounts of corrosive acids and
alkaline reagents and large wastewater streams. In the past years, research focused on the
development of more efficient and greener alternative routes based on solvent extraction
with ionic liquids (ILs) [12–14] and biotechnological conversion using microorganisms or
isolated proteolytic enzymes [10, 15].
The production of chitin is less than 100,000 metric tons per year [16], but reliable data
are difficult to be found in open literature. Almost all commercial chitin is produced from
crustacean shells, but in the past years new chitins from insects, fungal mycelia and
mushrooms have emerged in the market. Due to its intrinsic functional properties, chitin
has many industrial applications: in agriculture, for controlling plant diseases and as plant
growth regulators, in the food and drink industry for clarification of fruit juices, in water
treatment, as well as in the cosmetic, pharmaceutical and biomedical industry [10, 17].
However, an important application of chitin is as a source for the production of
glucosamine (GlcN), N‐acetylglucosamine (GlcNAc), chitosan and COS. In the past years,
novel promising routes have been demonstrated on a laboratory scale for the conversion of
chitin into bulk chemicals like acetic acid, platform chemicals like 5‐HMF and levulinic
acid (LA), and amine‐containing monomers for polymers. An overview of the chemicals
that can be produced from chitin and chitin’s monomers GlcN and GlcNAc is shown in
Figure 9.2.
Many reviews have been dedicated in the past years to the use of chitin as a renewable
source of chemicals and materials, promoting the concepts of ’shell biorefinery’ as a major
technology platform of the Blue Biotechnology and Circular Bioeconomy roadmaps [1, 3,
5, 15, 18–21]. In this chapter, we will discuss only the latest development in the production
of high‐value carbohydrates and bio‐based building blocks derived from chitin.
Polymer
HO OH H2 N OH
NH2
OR OR
GlcN
H C O
Chemical or
C OH Enzymatic
s
O
NH H C O
HO O O
O
C
Chemical or H2N OH
O O HO
NH
NH
HO O Enzymatic OH
OH
n O
O O
HO HO
COS, 2<n<20 OH n
HO OH
NH2 O
OH
Chitin O
O HO
GlcNAc HN
O
O
O O
Pyrolysis OH
OH HO
O Hydrogenolysis 3,6-anhydro-GNF O
Liquefaction Hydrogenation O
O HN
5-HMF Chemical HO
LA O OH
O HO
3,6-anhydro-MNF
O O N OH OH
O O H
O OH OH
HO 5-FMF H2N OH
HN
O ADS OH NH2
AS
3A5AF
s
Polymer
Figure 9.2 A schematic representation of chitin conversion into chitosan, chitooligosaccharides (COS), N‐acetylglucosamine (GlcNAc), glucosamine (GlcN)
and various platform chemicals and building blocks for polymers. 3,6‐anhydro‐GNF is 2‐acetamido‐3,6‐anhydro‐2‐deoxy‐D‐glucofuranose; 3,6‐anhydro‐MNF is
3,6‐anhydro‐2‐deoxy‐D‐mannofuranose; 3A5AF is 3‐acetamido‐5‐acetylfuran; 5‐FMF is 5‐[(formyloxy)methyl]furfural; 5‐HMF is hydroxymethylfurfural; ADS is
2‐acetamido‐2‐deoxy‐D‐sorbitol; AS is 2‐amino‐2‐deoxy‐D‐sorbitol; DGA is D‐glucosaminic acid; HAc is acetic acid; and LA is levulinic acid.
234 Chitin and Chitosan: Properties and Applications
c oncentration of hydroxide, temperature and reaction time strongly influence the molecular
properties of the chitosan product, in particular the DA and the molecular weight (Mw)
[24]. Although effective, chemical deacetylation has a number of drawbacks, of which high
energy consumption, use of corrosive chemicals, large streams of polluting wastes and
large variation in product properties are most relevant. Enzymatic chitin deacetylation
using chitin deacetylases of fungal [22, 25–28], bacterial [29, 30] and insect origin [24, 31]
has been proposed as a green and environment‐friendly alternative for chitosan production
that offers the possibility to obtain well‐defined chitosan through a controlled, mild and
non‐destructive process [24]. However, the efficiency of all tested chitin deacetylases on
polymeric, crystalline chitin is very low, and less than 10% of the N‐acetylglucosamine
residues are deacetylated rapidly [22, 25, 26, 28–30, 32]. Based on these results, it was
concluded that pretreatment of chitin is required in order to increase the accessibility of the
acetyl groups for deacetylation. Indeed, low‐Mw, partly deacetylated chitin/chitosan are
rapidly deacetylated by chitin deacetylases from Mucor rouxii [22], Absidia coerulea [32]
and Podospora anserina [33] to degrees of deacetylation of up to 90–97 mol%.
Next to chitosan, large amounts of acetic acid (HAc in Figure 9.1), a valuable bulk
chemical, are also produced, irrespective of the method used for chitin deacetylation.
There are three characteristics, which make chitosan an interesting industrial
polysaccharide: (i) it behaves like a polyelectrolyte with positive charge density at low pH,
(ii) is the only known high‐Mw cationic natural biopolymer with unique and exceptional
properties and (iii) it is and is often claimed to be GRAS (generally recognised as safe)
[34]. In addition, chitosan is biocompatible, bioabsorbable and nontoxic, and has a plethora
of bioactive properties, like antioxidative and antimicrobial activities, among others. These
characteristics enable chitosan to have applications in various fields. For example, chitosan
is used in medicine (as an antibacterial lining for bandages and wound dressings),
agriculture (for coating seeds to enhance disease resistance), food industry (as food
preservative, active edible packaging, dietary fibre), in water treatment as a bioflocculant,
in textiles and bioprinting, in cosmetics and the pharmaceutical industry for drug delivery
[10, 11, 35]. Chitosan is also used as a dietary supplement for weight loss since it was
shown to bind fat and hence should prevent fat digestion in the human body [34].
Chitosan prices depend on the quality of the product given mainly by chitosan’s degree
of deacetylation (DDA), Mw, water solubility, viscosity and residual protein, pigment and
mineral [36]. The demand for chitosan is increasing every year. In 2012, the global market
for chitosan was estimated at 21,400 metric tons, while in 2015, 0.2 million tons were
produced. The global chitosan market size is expected to reach €2.2 million by 2022 from
€1.1 million in 2015, with a CAGR of 18% (https://www.alliedmarketresearch.com).
low viscosity, low Mw, short chain lengths and water solubility make COS more suitable
for some industrial applications than chitin and chitosan [11, 40]. Due to their biological
activities, COS have high potential to be used on an industrial scale [40–42], and, at the
moment, different companies produce COS on a commercial scale [40].
COS can be prepared by chemical, physical and enzymatic treatments. Chemical
methods consist mainly of acid hydrolysis although oxidative reductive agents also can be
used. The most commonly applied chemical hydrolysis is the use of concentrated HCl at
80°C for 1–2 h. Other acids that can be used are nitrous acid, hydrofluoric acid and
phosphoric acid. As oxidative reductive agents, hydrogen peroxide, ozone and persulphate
can be used [42, 43]. A drawback of chemical hydrolysis is low yield, (detrimental)
secondary products can be formed and in general small size COS with a DP of 1–4 are
produced [41]. The mechanism of chemical hydrolysis is described in detail in [37].
Physical hydrolysis of chitin and chitosan can be performed by sonication, microwave
and irradiation. These high‐energy‐impact treatments result in cleavage of the O‐glycosidic
linkages [42]. Sonication can be performed in combination with acids to obtain COS [44,
45]. If sonication was used without acid, then only degradation of chitosan into lower Mw
was observed [46, 47]. Gamma and electron‐beam irradiation also resulted only in
degradation of chitosan into lower Mw [48]. In addition, gamma irradiation is more
effective in aqueous solution than in the solid state [49]. Production of COS from chitin and
chitosan in electron‐beam plasma gases has been reported [50]. Microwave irradiation is
performed in the presence of chemicals like acid or salts to obtain COS [51, 52].
Enzymatic methods are another option to produce COS in a more controlled way. Chitin
and/or chitosan can be converted to COS with glycoside hydrolases like chitinases or
chitosanases [53]. Depending on the structure of the starting material (DP, Mw, PA and
DA) enzymes can cleave the glycosidic linkages or modify the starting material (e.g.
deacetylation). Depending on the specificity of the enzymes, specific oligosaccharides can
be released. The characteristics of these enzymes are described in more detail in [54]. As
mentioned earlier, different methods or combinations hereof can be used to obtain COS. In
general, chemical and enzymatic treatments are used. Acetic acid is the main by‐product of
COS production processes.
Hydrolysis is traditionally performed with chemical methods using a strong acid as cata-
lyst (e.g. HCl) which breaks the glycosidic bonds in chitin and chitosan polymers [11].
Depending on process conditions, either GlcN or GlcNAc are produced. GlcN is usually
obtained from chitin with concentrated HCl (20–37%) at 100°C [64]. GlcNAc is obtained
by acid hydrolysis at lower temperature, for example, 40–80°C [65] or by re‐acetylation of
GlcN with acetic anhydride [66]. The chemical production of GlcNAc from chitin is esti-
mated to be economically feasible, but there are some drawbacks, including low yield
(below 65%), low specificity of the chemical catalyst, high operational costs and genera-
tion of large streams of acidic wastes [18].
Mechanocatalytic conversion of chitin to GlcNAc has recently been reported [67]. This
process consists of two steps: (i) impregnation of chitin with catalytic amounts of H2SO4,
and (ii) ball‐milling of acid‐impregnated chitin for 6 h, at 500 rpm, under solvent‐free con-
ditions, which results in total dissolution of chitin and the formation of low‐Mw chitin
oligosaccharides, with DP2 to DP5 and some longer chitin oligomers. Further hydrolysis
of the mixture at high temperature for a short time results in 53% GlcNAc, while
methanolysis produces the 1‐methyl ether derivative of GlcNAc, in 70% yield [67]. The
1‐O‐methyl‐N‐acetylglucosamine formed is a functional building block for biodegradable
polyesters and polyamides [67].
Following long and intensive research, enzymatic methods are now confirmed to be a
viable green alternative to acid depolymerisation for the production of GlcNAc and
GlcN from chitin and/or chitosan. Using chitin‐ and chitosan‐specific enzymes of fun-
gal and/or bacterial origin, chitin and chitosan can both be converted to GlcN, while
only chitin could be used to produce GlcNAc as a major product, in a selective way.
Figure 9.3 illustrates two major enzymatic pathways occurring in nature, which have
also been partly translated into laboratory or industrial processes for the production of
GlcNAc.
The chitinolytic pathway (route 1 in Figure 9.3) is based on the synergistic action of
three groups of chitin‐hydrolysing enzymes: chitinases, N‐acetylglucosaminidases
(NAGase) and lytic polysaccharide monooxygenases (LPMOs). Chitinases cleave
β‐1,4‐glycosidic linkages within chitin polymer chains and intermediate chitin oligo-
saccharides to generate chitobiose, (GlcNAc)2, which is finally degraded to GlcNAc by
Chitin GlcN
GlcNase
(2)
Figure 9.3 Enzymatic degradation of chitin via chitinolytic pathway (1) and chitosanolytic pathway (2).
LPMO, lytic polysaccharide monooxygenase; NAGase, N‐acetylglucosaminidase; GlcNase, glucosami-
nidase. *Some chitinases are able to degrade chitosan.
Chitin and Chitosan as Sources of Bio‐Based Building Blocks and Chemicals 237
of chitin by dry ball mill grinding prior to reaction significantly increased the yield of
3A5AF to 28.5 wt% [84].
9.4 Outlook
Valorisation of chitin and its derivatives is an important aspect of the bio‐based circular
economy in general and of the blue bioeconomy in particular, which recognises the need to
maximise the enormous economic potential presented by the ocean while preserving it.
Production of chitin, chitosan and chitin monomers is already demonstrated and applied on
an industrial scale, and chitin‐derived products are largely utilised. Recent studies
demonstrated the feasibility of converting chitin, chitosan and their monomers, that is,
glucosamine and N‐acetylglucosamine, into platform chemicals of high relevance for the
chemical and polymer industry. Further technological improvements are necessary to
increase the selectivity and efficiency of the reactions, particularly for the conversion of
polymeric chitin and chitosan into building blocks. Also, a better understanding of the
relationship between structural and molecular properties of chitin and chitosan (e.g.
crystallinity, Mw, DA, PA, etc.) and their reactivity and process efficiency is necessary. The
240 Chitin and Chitosan: Properties and Applications
Acknowledgement
Malgorzata Kaisler received funding from the Netherlands Organisation for Scientific
Research (NWO) in the framework of the TASC Technology Area BIOMASS.
References
1. EuropaBio, Building a Bio‐based Economy for Europe in 2020. 2011, European Association for
Bioindustries: Brussels.
2. McCormick, K. and N. Kautto, The bioeconomy in Europe: An overview. Sustainability, 2013. 5(6): p.
2589–2608.
3. Jardine, A. and S. Sayed, Challenges in the valorisation of chitinous biomass within the biorefinery
concept. Curr. Opin. Green Sustainable Chem., 2016. 2: p. 34–39.
4. Halder, S.K. and C.K. Mondal, Microbial valorization of chitinous bioresources for chitin
extraction and production of chito‐oligomers and N‐acetylglucosamine: Trends, perspectives
and prospects, in Microbial Biotechnology, J.K. Patra, G. Das, and H.S. Shin, Editors. 2018,
Springer. p. 69–107.
5. Yan, N. and X. Chen, Don’t waste seafood waste. Nature, 2015. 524(7564): p. 155–157.
6. Ahmed, S. and S. Ikram, Chitin and chitosan: History, composition and properties, in Chitosan:
Derivatives, Composites and Applications, S. Ahmed and S. Ikram, Editors. 2017, Scrivener Publishing
LLC: Beverley, USA. p. 1–24.
7. Prameela, K. et al., Next generation nutraceutical from shrimp waste: The convergence of applications
with extraction methods. Food Chem., 2017. 237: p. 121–132.
8. Zhu, F.M., B. Du, and B.J. Xu, A critical review on production and industrial applications of beta‐glucans.
Food Hydrocolloid., 2016. 52: p. 275–288.
9. Arbia, W. et al., Chitin extraction from crustacean shells using biological methods – A review. Food
Technol. Biotechnol., 2013. 51(1): p. 12–25.
10. Younes, I. and M. Rinaudo, Chitin and chitosan preparation from marine sources. Structure, properties
and applications. Marine Drugs, 2015. 13(3): p. 1133–1174.
11. Hamed, I., F. Özogul, and J.M. Regenstein, Industrial applications of crustacean by‐products (chitin,
chitosan, and chitooligosaccharides): A review. Trends Food Sci. Technol., 2016. 48: p. 40–50.
12. Qin, Y. et al., Dissolution or extraction of crustacean shells using ionic liquids to obtain high molecular
weight purified chitin and direct production of chitin films and fibers. Green Chem., 2010. 12(6): p.
968–971.
13. Setoguchi, T. et al., Facile production of chitin from crab shells using ionic liquid and citric acid. Int.
J. Biol. Macromol., 2012. 50(3): p. 861–864.
14. Xie, H., S. Zhang, and S. Li, Chitin and chitosan dissolved in ionic liquids as reversible sorbents of
CO2. Green Chem., 2006. 8(7): p. 630–633.
15. Chen, X., H. Yang, and N. Yan, Shell biorefinery: Dream or reality? Chem. – A Eur. J., 2016. 22(38):
p. 13402–13421.
16. Peter, M.G., On the Abundance of Chitin in the Biosphere and Its Importance. 2016: European Chitin
Society Newsletter.
17. Rinaudo, M., Chitin and chitosan: Properties and applications. Progr. Polym. Sci., 2006. 31(7):
p. 603–632.
Chitin and Chitosan as Sources of Bio‐Based Building Blocks and Chemicals 241
18. Chen, J.‐K., C.‐R. Shen, and C.‐L. Liu, N‐Acetylglucosamine: Production and applications. Marine
Drugs, 2010. 8(9): p. 2493.
19. Day, J. et al., Blue Biotechnology, in Commonwealth Blue Economy Report Series. 2016: London.
20. Kerton, F.M. et al., Green chemistry and the ocean‐based biorefinery. Green Chem., 2013. 15(4):
p. 860–871.
21. Nisticò, R., Aquatic‐derived biomaterials for a sustainable future: A European opportunity. Resources,
2017. 6(4): p. 65.
22. Martinou, A. et al., Mode of action of chitin deacetylase from Mucor rouxii on partiality N‐acetylated
chitosans. Carbohydr. Res., 1998. 311(1–2): p. 71–78.
23. Mao, X.Z. et al., Comprehensive utilization of shrimp waste based on biotechnological methods: A
review. J. Clean. Prod., 2017. 143: p. 814–823.
24. Younes, I. et al., Use of a fractional factorial design to study the effects of experimental factors on the
chitin deacetylation. Int. J. Biol. Macromol., 2014. 70: p. 385–390.
25. Alfonso, C. et al., Purification of a heat‐stable chitin deacetylase from Aspergillus nidulans and its role
in cell‐wall degradation. Curr. Microbiol., 1995. 30(1): p. 49–54.
26. Araki, Y. and E. Ito, Pathway of chitosan formation in Mucor rouxii – Enzymatic deacetylation of
chitin. Eur. J. Biochem., 1975. 55(1): p. 71–78.
27. Chambon, R. et al., High yield production of Rhizobium NodB chitin deacetylase and its use for in
vitro synthesis of lipo‐chitinoligosaccharide precursors. Carbohydr. Res., 2017. 442: p. 25–30.
28. Martinou, A., D. Kafetzopoulos, and V. Bouriotis, Isolation of chitin deacetylase from Mucor rouxii by
immunoaffintiy chromatography. J. Chromatogr., 1993. 644(1): p. 35–41.
29. Kadokura, K. et al., Purification and characterization of Vibrio parahaemolyticus extracellular chi-
tinase and chitin oligosaccharide deacetylase involved in the production of heterodisaccharide from
chitin. Appl. Microbiol. Biotechnol., 2007. 75(2): p. 357–365.
30. Li, X.B. et al., The chitin catabolic cascade in the marine bacterium Vibrio cholerae: Characterization
of a unique chitin oligosaccharide deacetylase. Glycobiology, 2007. 17(12): p. 1377–1387.
31. Dixit, R. et al., Domain organization and phylogenetic analysis of proteins from the chitin deacetylase
gene family of Tribolium castaneum and three other species of insects. Insect Biochem. Mol. Biol.,
2008. 38(4): p. 440–451.
32. Win, N.N. and W.F. Stevens, Shrimp chitin as substrate for fungal chitin deacetylase. Appl. Microbiol.
Biotechnol., 2001. 57(3): p. 334–341.
33. Hoßbach, J. et al., A chitin deacetylase of Podospora anserina has two functional chitin binding
domains and a unique mode of action. Carbohydr. Polym., 2018. 183: p. 1–10.
34. Bellich, B. et al., “The Good, the bad and the ugly” of chitosans. Marine Drugs, 2016. 14(5): p. 31.
35. van den Broek, L.A.M. et al., Chitosan films and blends for packaging material. Carbohydr. Polym.,
2015. 116: p. 237–242.
36. Gomez‐Rios, D., R. Barrera‐Zapata, and R. Rios‐Estepa, Comparison of process technologies for chi-
tosan production from shrimp shell waste: A techno‐economic approach using Aspen Plus (R). Food
Bioprod. Process., 2017. 103: p. 49–57.
37. Mourya, V.K., N.N. Inamdar, and Y.M. Choudhari, Chitooligosaccharides: Synthesis, characterization
and applications. Polym. Sci. Series A, 2011. 53(7): p. 583–612.
38. Jeon, Y.‐J. and S.‐K. Kim, Continuous production of chitooligosaccharides using a dual reactor sys-
tem. Proc. Biochem., 2000. 35(6): p. 623–632.
39. Cord‐Landwehr, S. et al., New Developments in the Analysis of Partially Acetylated Chitosan Polymers
and Oligomers, in Chitin and Chitosan: Properties and Applications, L.A.M. van de Broek and C.G.
Boeriu, Editors. 2019, Wiley.
40. Liaqat, F. and R. Eltem, Chitooligosaccharides and their biological activities: A comprehensive review.
Carbohydr. Polym., 2018. 184: p. 243–259.
41. Zou, P. et al., Advances in characterisation and biological activities of chitosan and chitosan oligosac-
charides. Food Chem., 2016. 190: p. 1174–1181.
242 Chitin and Chitosan: Properties and Applications
42. Muanprasat, C. and V. Chatsudthipong, Chitosan oligosaccharide: Biological activities and potential
therapeutic applications. Pharmacol. Ther., 2017. 170: p. 80–97.
43. Lodhi, G. et al., Chitooligosaccharide and its derivatives: Preparation and biological applications.
BioMed Res. Int., 2014. 2014: p. 13.
44. Takahashi, Y., F. Miki, and K. Nagase, Effect of sonolysis on acid degradation of chitin to form oligo-
saccharides. Bull. Chem. Soc. Jpn., 1995. 68(7): p. 1851–1857.
45. Savitri, E. et al., Degradation of chitosan by sonication in very‐low‐concentration acetic acid. Polym.
Degrad. Stab., 2014. 110: p. 344–352.
46. Kasaai, M.R., J. Arul, and G. Charlet, Fragmentation of chitosan by ultrasonic irradiation. Ultrason.
Sonochem., 2008. 15(6): p. 1001–1008.
47. Tang, E.S.K., M. Huang, and L.Y. Lim, Ultrasonication of chitosan and chitosan nanoparticles. Int. J.
Pharm., 2003. 265(1): p. 103–114.
48. Gryczka, U. et al., The mechanism of chitosan degradation by gamma and e‐beam irradiation. Radiat.
Phys. Chem., 2009. 78(7): p. 543–548.
49. Tahtat, D. et al., Effect of molecular weight on radiation chemical degradation yield of chain scission
of γ‐irradiated chitosan in solid state and in aqueous solution. Radiat. Phys. Chem., 2012. 81(6):
p. 659–665.
50. Vasil’eva, T.M., S.A. Lopatin, and V.P. Varlamov, Production of the low‐molecular‐weight chitin and
chitosan forms in electron‐beam plasma. High Energy Chem., 2016. 50(2): p. 150–154.
51. Xing, R. et al., Monomer composition of chitooligosaccharides obtained by different degradation
methods and their effects on immunomodulatory activities. Carbohydr. Polym., 2017. 157:
p. 1288–1297.
52. Xing, R.E. et al., Salt‐assisted acid hydrolysis of chitosan to oligomers under microwave irradiation.
Carbohydr. Res., 2005. 340(13): p. 2150–2153.
53. Aam, B.B. et al., Production of chitooligosaccharides and their potential applications in medicine.
Marine Drugs, 2010. 8(5): p. 1482.
54. Vaaja‐Kolsted, G. et al., Enzymes for modification of chitin and chitosan, in Chitin and Chitosan:
Properties and Applications, L.A.M. van de Broek and C.G. Boeriu, Editors. 2019, Wiley.
55. Salvatore, S. et al., A pilot study of N‐acetyl glucosamine, a nutritional substrate for glycosaminogly-
can synthesis, in paediatric chronic inflammatory bowel disease. Aliment. Pharmacol. Ther., 2000.
14(12): p. 1567–1579.
56. Kubomura, D. et al., Evaluation of the chondroprotective action of N‐acetylglucosamine in a rat exper-
imental osteoarthritis model. Exp. Ther. Med., 2017. 14(4): p. 3137–3144.
57. Bissett, D.L., Glucosamine: An ingredient with skin and other benefits. J. Cosmet. Dermatol., 2006.
5(4): p. 309–315.
58. Riordan, N.H., Skin Treatment System. 1999, US005866142A1.
59. Xu, Q., J. Liu, and Z. Yuan, The Use of N‐acetylglucosamine as Additive in Beer and Beer Comprising
N‐acetylglucosamine. 2004, WO2004085603A1.
60. Xu, Q., J. Liu, and Z. Yuan, The Use of N‐acetyl‐D‐glucosamine as Additive in Milk Products. 2004,
WO2004085604A1.
61. Xu, Q., J. Liu, and Z. Yuan, The Use of N‐acetylglucosamine in Wine and Wine Comprising N‐acetyl-
glucosamine. 2004, WO2004093556A1.
62. Inokuma, K., M. Takano, and K. Hoshino, Direct ethanol production from N‐acetylglucosamine and
chitin substrates by Mucor species. Biochem. Eng. J., 2013. 72: p. 24–32.
63. van Putten, R.J. et al., Hydroxymethylfurfural, a versatile platform chemical made from renewable
resources. Chem. Rev., 2013. 113(3): p. 1499–1597.
64. Mojarrad, J.S. et al., Preparation of glucosamine from exoskeleton of shrimp and predicting produc-
tion yield by response surface methodology. J. Agric. Food Chem., 2007. 55(6): p. 2246–2250.
65. Fan, W. et al., Glucosamine and Method of Making Glucosamine from Microbial Biomass. 2006,
Cargill, Incorporated (Wayzata, MN, US).
Chitin and Chitosan as Sources of Bio‐Based Building Blocks and Chemicals 243
66. Roseman, S. and J. Ludowieg, N‐Acetylation of the hexosamines. J. Am. Chem. Soc., 1954. 76(1):
p. 301–302.
67. Yabushita, M. et al., Catalytic depolymerization of chitin with retention of N‐acetyl group.
Chemsuschem, 2015. 8(22): p. 3760–3763.
68. Hartl, L., S. Zach, and V. Seidl‐Seiboth, Fungal chitinases: diversity, mechanistic properties and bio-
technological potential. Appl. Microbiol. Biotechnol., 2012. 93(2): p. 533–543.
69. Beier, S. and S. Bertilsson, Bacterial chitin degradation‐mechanisms and ecophysiological strategies.
Front. Microbiol., 2013. 4: p. 149–149.
70. Vaaje‐Kolstad, G. et al., An oxidative enzyme boosting the enzymatic conversion of recalcitrant poly-
saccharides. Science, 2010. 330(6001): p. 219–222.
71. Ohtakara, A., H. Matsunaga, and M. Mitsutomi, Action pattern of Streptomyces griseus chitinase on
partially N‐acetylated chitosan. Agric. Biol. Chem., 1990. 54(12): p. 3191–3199.
72. Sikorski, P. et al., Development and application of a model for chitosan hydrolysis by a family 18
chitinase. Biopolymers, 2005. 77(5): p. 273–285.
73. Sorbotten, A. et al., Degradation of chitosans with chitinase B from Serratia marcescens ‐ Production
of chito‐oligosaccharides and insight into enzyme processivity. Febs J., 2005. 272(2): p. 538–549.
74. Krolicka, M. et al., Chitinase Chi1 from Myceliophthora thermophila C1, a thermostable enzyme for
chitin and chitosan depolymerization. J. Agric. Food Chem., 2018. 66(7): p. 1658–1669.
75. Pichyangkura, R. et al., Quantitative production of 2‐acetamido‐2‐deoxy‐D‐glucose from crystalline
chitin by bacterial chitinase. Carbohydr. Res., 2002. 337(6): p. 557–559.
76. Sashiwa, H. et al., Production of N‐acetyl‐D‐glucosamine from alpha‐chitin by crude enzymes from
Aeromonas hydrophila H‐2330. Carbohydr. Res., 2002. 337(8): p. 761–763.
77. Binod, P. et al., Fungal biosynthesis of endochitinase and chitobiase in solid state fermentation and
their application for the production of N‐acetyl‐D‐glucosamine from colloidal chitin. Bioresour.
Technol., 2007. 98(14): p. 2742–2748.
78. Jaworska, M.M., T. Kozlecki, and A. Gorak, Review of the application of ionic liquids as solvents for
chitin. J. Polym. Eng., 2012. 32(2): p. 67–69.
79. Villa‐Lerma, G. et al., Enzymatic hydrolysis of chitin pretreated by rapid depressurization from super-
critical 1,1,1,2‐tetrafluoroethane toward highly acetylated oligosaccharides. Bioresour. Technol., 2016.
209: p. 180–186.
80. Dziril, M. et al., Chitin oligomers and monomers production by coupling gamma radiation and enzy-
matic hydrolysis. J. Ind. Eng. Chem., 2015. 26: p. 396–401.
81. Roy, I., K. Mondal, and M.N. Gupta, Accelerating enzymatic hydrolysis of chitin by microwave pre-
treatment. Biotechnol. Prog., 2003. 19(6): p. 1648–1653.
82. Wei, G.G. et al., Enzymatic production of N‐acetyl‐D‐glucosamine from crayfish shell wastes pre-
treated via high pressure homogenization. Carbohydr. Polym., 2017. 171: p. 236–241.
83. Nakagawa, Y.S. et al., Development of innovative technologies to decrease the environmental burdens
associated with using chitin as a biomass resource: Mechanochemical grinding and enzymatic degra-
dation. Carbohydr. Polym., 2011. 83(4): p. 1843–1849.
84. Chen, X. et al., Effect of treatment methods on chitin structure and its transformation into nitrogen‐
containing chemicals. Chempluschem, 2015. 80(10): p. 1565–1572.
85. Osada, M. et al., Effect of sub‐ and supercritical water pretreatment on enzymatic degradation of chi-
tin. Carbohydr. Polym., 2012. 88(1): p. 308–312.
86. Kaisler, M., Efficient production of N‐acetylglucosamine with chitinolytic enzymes from
Myceliophthora thermophila C1, in Bioprocess Engineering. 2019, Wageningen University:
Wageningen, NL. p. 156.
87. Mekasha, S. et al., Development of enzyme cocktails for complete saccharification of chitin using
mono‐component enzymes from Serratia marcescens. Process Biochem., 2017. 56: p. 132–138.
88. Mascal, M. and E.B. Nikitin, Dramatic advancements in the saccharide to 5‐(chloromethyl) furfural
conversion reaction. Chemsuschem, 2009. 2(9): p. 859–861.
244 Chitin and Chitosan: Properties and Applications
89. Omari, K.W., J.E. Besaw, and F.M. Kerton, Hydrolysis of chitosan to yield levulinic acid
and 5‐hydroxymethylfurfural in water under microwave irradiation. Green Chem., 2012. 14(5):
p. 1480–1487.
90. Lee, S.B. and G.T. Jeong, Catalytic conversion of chitosan to 5‐hydroxymethylfurfural under low
temperature hydrothermal process. Appl. Biochem. Biotechnol., 2015. 176(4): p. 1151–1161.
91. Li, M. et al., Efficient conversion of chitosan into 5‐hydroxymethylfurfural via hydrothermal synthe-
sis in ionic liquids aqueous solution. Polym. Degrad. Stab., 2015. 121: p. 331–339.
92. Wang, Y.X. et al., Direct conversion of chitin biomass to 5‐hydroxymethylfurfural in concentrated
ZnCl2 aqueous solution. Bioresour. Technol., 2013. 143: p. 384–390.
93. Chen, X. et al., Direct conversion of chitin into a N‐containing furan derivative. Green Chem., 2014.
16(4): p. 2204–2212.
94. Drover, M.W. et al., Formation of a renewable amide, 3‐acetamido‐5‐acetylfuran, via direct conver-
sion of N‐acetyl‐D‐glucosamine. Rsc Adv., 2012. 2(11): p. 4642–4644.
95. Omari, K.W., L. Dodot, and F.M. Kerton, A simple one‐pot dehydration process to convert N‐acetyl‐
D‐glucosamine into a nitrogen‐containing compound, 3‐acetamido‐5‐acetylfuran. Chemsuschem,
2012. 5(9): p. 1767–1772.
96. Bueno, M. et al., Synthesis of stereoregular poylgluconamides from D‐glucose and D‐glucosamine.
J. Polym. Sci. Part A‐Polym. Chem., 1995. 33(2): p. 299–305.
97. Mirescu, A. and U. Prusse, A new environmental friendly method for the preparation of sugar acids
via catalytic oxidation on gold catalysts. Appl. Catal. B‐Environ., 2007. 70(1‐4): p. 644–652.
98. Gu, W.X. and W.S. Xia, Catalytic synthesis of D‐glucosaminic acid from D‐glucosamine on active
charcoal‐supported Pd‐Bi catalysts. J. Carbohydr. Chem., 2006. 25(4): p. 297–301.
99. Pezzotti, F., H. Therisod, and M. Therisod, Enzymatic synthesis of D‐glucosaminic acid from
D‐glucosamine. Carbohydr. Res., 2005. 340(1): p. 139–141.
100. Moonmangmee, D. et al., D‐hexosaminate production by oxidative fermentation. Appl. Microbiol.
Biotechnol., 2004. 66(3): p. 253–258.
101. Bobbink, F.D. et al., Conversion of chitin derived N‐acetyl‐D‐glucosamine (NAG) into polyols over
transition metal catalysts and hydrogen in water. Green Chem., 2015. 17(2): p. 1024–1031.
102. Kobayashi, H., K. Techikawara, and A. Fukuoka, Hydrolytic hydrogenation of chitin to amino sugar
alcohol. Green Chem., 2017. 19(14): p. 3350–3356.
103. Osada, M. et al., Non‐catalytic synthesis of Chromogen I and III from N‐acetyl‐D‐glucosamine in
high‐temperature water. Green Chem., 2013. 15(10): p. 2960–2966.
10
Chemical and Enzymatic
Modification of Chitosan
to Produce New Functional
Materials with Improved
Properties
Carmen G. Boeriu and Lambertus A.M. van den Broek
Wageningen Food & Biobased Research, Wageningen, The Netherlands
Chitin and chitosan are natural biopolymers with applications in different areas. However,
to broaden its application area, research is needed to impart new functionality, develop new
and better materials and to explore the whole potential of chitin/chitosan derivatives. These
derivatives can be made by chemical and enzymatic synthesis. Here, the main classes of
chitosan derivatives obtained by using green and sustainable conversion routes and as
much as possible using green and/or bio‐based chemicals for modification and their poten-
tial areas for application are discussed.
10.1 Introduction
Chitin and chitosan are natural biopolymers with unique structures and properties. Both
polymers are aminated linear polysaccharides with N‐acetylamide and free amino groups
at the C2 position of the β‐glucose unit, that are connected through rigid β‐1,4‐glycosidic
linkages. The β‐1,4‐configuration of the N‐acetylglucosamine (GlcNAc) and glucosamine
(GlcN) building blocks results in a rigid configuration of the linear polymer chains that are
associated through intra‐ and inter‐molecular hydrogen bonds into linear aggregates with
high crystallinity [1]. Chitin and chitosan differ from each other by the content of acety-
lated and free amino groups, expressed as degree of acetylation (DA) and the distribution
of the acetyl group along the polymer chain, referred to as pattern of acetylation (PA), both
having a strong influence on the polymers’ properties. The distribution of the GlcNAc and
GlcN residues in the chitosan chain can be block‐wise or random, depending on the condi-
tions of the deacetylation process [1]. In nature, chitin is most probably fully acetylated,
but during the extraction process some deacetylation of the GlcNAc units occurs and the
isolated (commercial) chitin is partly deacetylated. It has been shown that even low dea-
cetylation of chitin, for example, 10–15% free amino groups, can induce bioactivities and
increase solubility and reactivity. Therefore, technologies have been developed to deacety-
late chitin to obtain chitosan, a family of deacetylated chitin derivatives with degree of
deacetylation (DDA) ranging from 50% to 99.8% and molecular weight between 5 × 104
and 2.5 × 106 g/mol [2, 3]. The high density of glucosamine residues in chitosan has tre-
mendous effects on its properties. The polymer is positively charged in acidic solution, due
to the protonation of the free amine groups (pKa~ 6.3), which increases chitosan’s solubil-
ity by increasing both the polarity and extent of electrostatic repulsion. Water‐soluble salts
of chitosan with formate, acetate, lactate, malate, citrate, tartrate, glycolate and other weak
organic acids are used for chitosan processing and the production of biomedical devices.
The cationic sites along the polymer chain are responsible for biological activity, for exam-
ple, antibacterial and antifungal [4, 5], and hydrogel formation through electrostatic asso-
ciation with anionic biopolymers and synthetic polymers [6]. This makes chitosan a
valuable component in food, feed, agrochemical, cosmetics, biomedical and pharmaceuti-
cal applications. Chitosan also shows pH‐dependent metal chelating properties that form
the basis for its utilisation in the removal of heavy metals from polluted water [7, 8] or as
support for metal catalysts in heterogeneous synthetic reactions [9] and antimicrobial coat-
ings and fibres [10–13], for example. This unique assembly of multiple and versatile func-
tional activities associated with biocompatibility, biodegradability and lack of toxicity
distinguishes chitosan from all other commercial natural and synthetic polymers.
The potential for application of native chitosan in different areas has been proven, but
research has also shown the need to enhance its properties, impart new functionality,
develop new and better materials and explore the whole potential of the compounds. There
is a huge interest in modification of chitosan, illustrated by the high number of research
papers published each year: a search in the web of science (https://login.webofknwoledge.
com/) for ’chitosan AND modification’ gave 598 hits in 2018. Numerous comprehensive
reviews, books and book chapters addressing chitosan modification are available. In
Figure 10.1, we show an overview of the most important modification strategies applied for
chitosan derivatisation and application areas.
In this chapter, we discuss the main classes of chitosan derivatives obtained by using
green and sustainable conversion routes and as much as possible using green and/or bio‐
based chemicals for modification and their potential areas for application.
Figure 10.1 Derivatisation of chitosan and fields of application of the modified biopolymers. Dashed arrows indicate the most reactive groups prone to
modification.
248 Chitin and Chitosan: Properties and Applications
number of chemical reactions (Figure 10.2). Highly cationic and quaternised chitosan, ani-
onic chitosan with carboxyl or sulphate groups, arylated, nonpolar or hydrophobic chitosan
derivatives have all been obtained by chemocatalytic or mild and selective enzymatic con-
version. Some examples are given in Figure 10.2. Recent developments will be discussed
in the following text.
O O R
O
HO
NH O O
HO
OH O R NH2
O O R N-acyl chitosan O-acyl chitosan
HO OH OH O
NH O O +
O O N
R + O O
O HO
R HO NH
N-, O O
NH2 O-A HO
Hydroxyalkyl chitosan cyla O
on
tion + NH2
N
lati
HO OH
lky
O O O
O O
HO O HO
O NH
N-,
O NH2
HO O Chitosan
NH2 O Re
HO d
O-carboxymethyl-chitosan N-carboxymethyl-chitosan
ion am uct
at ina ive
ulf
OH
Arylatio
S tio
n O
n
OSO3Na O O
tio
HO
NH
ida
O O HO
n
OH
Ox
HO
NH2 O OH HO OH
HO
Chitosan sulfate O O
COOH
HO O Sugar-linked chitosan
O NH n
O
HO R
OH O
6-Carboxychitosan HO
OH
Arylchitosan
Figure 10.2 Functional chitosan derivatives obtained by chemical or enzymatic modification of amine or primary hydroxyl groups.
250 Chitin and Chitosan: Properties and Applications
10.2.2 Hydroxyalkylchitosans
Hydroxyalkylchitosans – that is hydroxyethyl chitosan, hydroxypropyl chitosan and glycol
chitosan – are obtained by reacting with alkyl epoxides (e.g. ethylene oxide, propylene
oxide, butylene oxide) and glycidol, which is a common procedure for modification of
polysaccharides, including cellulose and starch. Selective synthesis of N‐hydroxyalkyl or
O‐hydroxyalkyl derivatives can be controlled by the reaction conditions, particularly the
process temperature and catalyst used. Hydroxyalkylchitosans are water soluble and show
antimicrobial properties and have potential application as a temperature‐sensitive injecta-
ble carrier for cells [27] and as self‐assembled nanoparticles for drug delivery [28–30].
(a)
C C C C C C C C C C C C
C
C +
C
C
(b)
C C C C C C C C C C
(c)
C C C C C C C C C C C C C C C
Figure 10.3 Schematic illustration of (a) grafting through, (b) grafting to, and (c) grafting from. Encircled
C is a chitosan monomer (e.g. N‐acetyl glucosamine, glucosamine or chitosan molecule); X is the initiating
functionality; tail is a co‐polymer; star is another co‐polymer added to the tail.
both acid and alkaline conditions, even at low substitution degrees. These phenolic chi-
tosans show antioxidant and antimicrobial activity, and have a rheological behaviour char-
acteristic of associating water‐soluble polymers [3].
other two methods are used. In the case of ’grafting to’, end‐group functionalised co‐polymers
react with complementary functional groups on the chitosan chain (Figure 10.3b).
Telechelic polymers, polyethylene glycol and polydimethylsiloxane are some examples
that are usually utilised for grafting to chitosan. ’Grafting from’ is often based on the intro-
duction of a reactive group, usually referred to as initiator, on the chitosan backbone medi-
ated by the ’grafting to’ method. The initiator, that can be an acrylate or vinyl, is used to
covalently link co‐polymers to the chitosan chains (Figure 10.3c.) [46]. The efficiency and
percentage of grafting is dependent on the type of monomer and initiator concentration and
also reaction temperature and time.
Techniques that can be used for grafting are chemical [47–52] including grafting through
living polymerisation [48, 53], irradiation [47, 48, 51, 52], photochemical [48], plasma‐
induced [48, 50], and enzymatic synthesis [47, 48, 50, 51, 54]. Chemical grafting can be
performed by free radical and ionic approach. Free radicals are produced from initiators
and transferred to the chitosan backbone which will form the graft co‐polymer. Examples
of initiators are ammonium persulphate, potassium persulphate, ceric ammonium nitrate,
thiocarbonate–potassium bromate, potassium diperiodatocuprate (III), 2,2’‐azobisisobu-
tyronitrile and ferrous ammonium sulphate (FAS) [51]. Ionic mode grafting can be per-
formed by cationic and anionic mechanisms using alkali metal suspensions in a Lewis base
liquid, organometallic compounds, sodium naphthalenide and sodium‐ammonia as initia-
tors [48]. A drawback of the free radicals and ionic approach is that the polymerisation
process is less controlled, resulting in high polydispersity of the grafted co‐polymers.
’Living’ polymerisation, a process defined by Szwarc [55], results in grafted co‐polymers
with regulated molecular weights and low polydispersity. In case of living polymerisation,
controlled free radical polymerisation can be performed with atom transfer radical
polymerisation (ATRP). The advantage of ATRP is twofold due to the following principles.
First, the initiation reaction is fast enough in order to have a continuous concentration of
growing polymer chains. Second, the graft co‐polymers are able to grow due to the equilibrium
between the radicals formed and graft co‐polymers synthesised [56].
In addition, click chemistry can be used to combine/conjugate different polymeric chains
to synthesise grafted co‐polymers with different structures such as linear, branched, comb‐
shaped and star‐shaped co‐polymers [57].
Photochemical grafting is initiated by chromophores that absorb light and as a conse-
quence dissociate into reactive radicals. Photosensitisers can be used to accelerate the pro-
cess. Irradiation‐induced grafting can be performed by microwave and γ‐irradiation such as
60
Co γ‐irradiation. Plasma‐induced grafting depends on electron‐induced excitation, ioni-
sation and dissociation; the released energy, like in irradiation, is able to form radicals.
Enzymatic conjugation is another possibility to graft chitosan to biomacromolecules like
peptides and proteins. Tyrosinase, transglutaminase, laccase and horseradish peroxidase
have been used for the synthesis of chitosan‐protein bioconjugates with a wide range of
proteins and peptides, such as, for example, gelatin, sericin‐peptides and collagen peptides,
among others [58–62]. These chitosan‐protein and chitosan‐peptide conjugates offer inter-
esting mechanical and rheological properties, comparable with that of natural glycopro-
teins, and are suitable for biomedical and industrial applications. The grafted chitosan
co‐polymers may form hydrogels and also display various biological properties. Some
examples of graft co‐polymers of chitosan and their potential application are listed in
Table 10.1.
254 Chitin and Chitosan: Properties and Applications
10.5 Outlook
Decades of research on chitosan and its modified derivatives have shown, beyond any
doubt, the huge application potential for use in various fields including medical, pharma-
ceutical, cosmetics, bio‐related science and technology, food industry, agriculture, and
environmental protection. Viable technologies and processing routes have been developed
for the synthesis of chitosan derivatives and chitosan‐based materials, and a number of
them are already reaching the consumer in various end‐use products. Now, when the
world is facing the transition from the petrochemical economy to a sustainable bio‐based
circular economy, there are many opportunities for chitosan‐derived chemicals and mate-
rials to be recognised as a major bioresource for bioactives and biomaterials and to expand
their market share. Chitosan and its derivatives are very useful since they can be made
available in a variety of morphologies including fibres, films, hydrogels, membranes,
nanoparticles, microparticles and with various functional properties and bioactivities.
Nevertheless, scientific and technological developments are still needed to develop
greener and more sustainable routes for chitosan production and modification, addressing
not only process efficiency but also the source of substrates, catalysts and solvents, the
carbon and energy balance, the environmental impacts and the process and product costs.
Standardisation of methods for chemical, structural and functional characterisation of chi-
tosan and chitosan derivatives is needed to allow objective tools and criteria to compare
different materials.
Chemical and Enzymatic Modification of Chitosan to Produce New Functional Materials 255
References
1. Prashanth, K.V.H. and R.N. Tharanathan, Chitin/chitosan: Modifications and their unlimited applica-
tion potential – an overview. Trends Food Sci. Technol., 2007. 18(3): p. 117–131.
2. Hejazi, R. and M. Amiji, Chitosan‐based gastrointestinal delivery systems. J. Control. Release, 2003.
89(2): p. 151–165.
3. Mourya, V.K. and N.N. Inamdar, Chitosan‐modifications and applications: Opportunities galore.
React. Funct. Polym., 2008. 68(6): p. 1013–1051.
4. Younes, I. and M. Rinaudo, Chitin and chitosan preparation from marine sources. Structure, properties
and applications. Marine Drugs, 2015. 13(3): p. 1133–1174.
5. Younes, I. et al., Influence of acetylation degree and molecular weight of homogeneous chitosans on
antibacterial and antifungal activities. Int. J Food Microbiol., 2014. 185: p. 57–63.
6. Bhattarai, N., J. Gunn, and M. Zhang, Chitosan‐based hydrogels for controlled, localized drug deliv-
ery. Adv. Drug Delivery Rev., 2010. 62(1): p. 83–99.
7. Sun, Y. et al., Characterization and flocculation evaluation of a novel carboxylated chitosan modified
flocculant by UV initiated polymerization. Carbohydr. Polym., 2019. 208: p. 213–220.
8. Sun, Y.J. et al., Performance evaluation of chitosan‐based flocculants with good pH resistance and high
heavy metals removal capacity. Sep. Purif. Technol., 2019. 215: p. 208–216.
9. Todea, A. et al., Increase of stability of oleate hydratase by appropriate immobilization technique and
conditions. J. Mol. Catal. B: Enzym., 2015. 119: p. 40–47.
10. van den Broek, L.A.M. et al., Chitosan films and blends for packaging material. Carbohydr. Polym.,
2015. 116: p. 237–242.
11. Emam, H.E., Antimicrobial cellulosic textiles based on organic compounds. 3 Biotech, 2019. 9:29.
https://doi.org/10.1007/s13205‐018‐1562‐y.
12. Mondal, M.I.H. and J. Saha, Antimicrobial, UV resistant and thermal comfort properties of chitosan‐
and aloe vera‐modified cotton woven fabric. J. Polym. Environ., 2019. 27(2): p. 405–420.
13. Xu, Q.B. et al., One‐pot fabrication of durable antibacterial cotton fabric coated with silver nanoparti-
cles via carboxymethyl chitosan as a binder and stabilizer. Carbohydr. Polym., 2019. 204: p. 42–49.
14. Shariatinia, Z., Carboxymethyl chitosan: Properties and biomedical applications. Int. J. Biol.
Macromol., 2018. 120: p. 1406–1419.
15. Liu, X.F. et al., Antibacterial action of chitosan and carboxymethylated chitosan. J. Appl. Polym. Sci.,
2001. 79(7): p. 1324–1335.
16. Kim, C.H. and K.S. Choi, Synthesis and properties of carboxyalkyl chitosan derivatives. J. Ind. Eng.
Chem., 1998. 4(1): p. 19–25.
17. Rinaudo, M. et al., Substituent distribution on O, N‐carboxymethylchitosan by H‐1‐NMR and C‐13‐
NMR. Int. J. Biol. Macromol., 1992. 14(3): p. 122–128.
18. Lin, Y.W., Q. Chen, and H.B. Luo, Preparation and characterization of N‐(2‐carboxybenzyl) chitosan
as a potential pH‐sensitive hydrogel for drug delivery. Carbohydr. Res., 2007. 342(1): p. 87–95.
19. Skorik, Y.A. et al., Synthesis of N‐succinyl‐ and N‐glutaryl‐chitosan derivatives and their antioxidant,
antiplatelet, and anticoagulant activity. Carbohydr. Polym., 2017. 166: p. 166–172.
20. Petrova, V.A. et al., Preparation of N‐succinyl‐chitin nanoparticles and their applications in otoneuro-
logical pathology. Int. J. Biol. Macromol., 2018. 120: p. 1023–1029.
21. da Silva, S.B. et al., Water‐soluble chitosan derivatives and pH‐responsive hydrogels by selective C‐6
oxidation mediated by TEMPO‐laccase redox system. Carbohydr. Polym., 2018. 186: p. 299–309.
22. Dimassi, S. et al., Sulfonated and sulfated chitosan derivatives for biomedical applications: A review.
Carbohydr. Polym., 2018. 202: p. 382–396.
23. Jayakumar, R. et al., Sulfated chitin and chitosan as novel biomaterials. Int. J. Biol. Macromol., 2007.
40(3): p. 175–181.
24. Je, J.Y., P.J. Park, and S.K. Kim, Prolyl endopeptidase inhibitory activity of chitosan sulfates with dif-
ferent degree of deacetylation. Carbohydr. Polym., 2005. 60(4): p. 553–556.
256 Chitin and Chitosan: Properties and Applications
25. Hirano, S. et al., Effect of sulfated derivatives of chitosan and some blood coagulant factors. Carbohydr.
Res., 1985. 137(MAR): p. 205–215.
26. Zhang, C. et al., Synthesis and characterization of water‐soluble O‐succinyl‐chitosan. Eur. Polym. J.,
2003. 39(8): p. 1629–1634.
27. Peng, Y.F. et al., Preparation and antimicrobial activity of hydroxypropyl chitosan. Carbohydr. Res.,
2005. 340(11): p. 1846–1851.
28. Kim, J.H. et al., Hydrophobically modified glycol chitosan nanoparticles as carriers for paclitaxel
(Reprinted from Journal of Controlled Release, vol 109, p. 1, 2005). J. Control. Release, 2006. 111(1–2):
p. 228–234.
29. Park, J.H. et al., Self‐assembled nanoparticles based on glycol chitosan bearing hydrophobic moieties
as carriers for doxorubicin: In vivo biodistribution and anti‐tumor activity. Biomaterials, 2006. 27(1):
p. 119–126.
30. Park, J.H. et al., Preparation and characterization of self‐assembled nanoparticles based on glycol
chitosan bearing adriamycin. Colloid. Polym. Sci., 2006. 284: p. 763–770.
31. Naskar, S., S. Sharma, and K. Kuotsu, Chitosan‐based nanoparticles: An overview of biomedical appli-
cations and its preparation. J. Drug Delivery Sci. Technol., 2019. 49: p. 66–81.
32. Je, J.Y. and S.K. Kim, Water‐soluble chitosan derivatives as a BACE1 inhibitor. Bioorg. Med. Chem.,
2005. 13(23): p. 6551–6555.
33. Lee, J.K., H.S. Lim, and J.H. Kim, Cytotoxic activity of aminoderivatized cationic chitosan deriva-
tives. Bioorg. Med. Chem. Lett., 2002. 12(20): p. 2949–2951.
34. Holappa, J. et al., Synthesis and characterization of chitosan N‐betainates having various degrees of
substitution. Macromolecules, 2004. 37(8): p. 2784–2789.
35. Gao, Y. et al., Chitosan N‐betainates/DNA self‐assembly nanoparticles for gene delivery: In vitro
uptake and transfection efficiency. Int. J. Pharm., 2009. 371(1–2): p. 156–162.
36. Aberg, C.M., T.H. Chen, and G.F. Payne, Renewable resources and enzymatic processes to create
functional polymers: Adapting materials and reactions from food processing. J. Polym. Environ., 2002.
10(3): p. 77–84.
37. Boeriu, C.G. et al., New aryl‐chitosan derivatives with strong antioxidant properties by chemoenzy-
matic modification, Communication at the 17th Polymers and Organic Chemistry Conference, Palavas‐
les‐Flots, France, 2018.
38. Chen, T.H. et al., Enzymatic grafting of hexyloxyphenol onto chitosan to alter surface and rheological
properties. Biotechnol. Bioeng., 2000. 70(5): p. 564–573.
39. Kumar, G., P.J. Smith, and G.F. Payne, Enzymatic grafting of a natural product onto chitosan to confer
water solubility under basic conditions. Biotechnol. Bioeng., 1999. 63(2): p. 154–165.
40. Li, K. et al., Antibacterial activity and mechanism of a laccase‐catalyzed chitosan–gallic acid deriva-
tive against Escherichia coli and Staphylococcus aureus. Food Control, 2019. 96: p. 234–243.
41. Vachoud, L. et al., Peroxidase catalyzed grafting of gallate esters onto the polysaccharide chitosan.
Enzyme Microbial Technol., 2001. 29(6–7): p. 380–385.
42. Donati, I. et al., Tailor‐made alginate bearing galactose moieties on mannuronic residues: Selective
modification achieved by a chemoenzymatic strategy. Biomacromolecules, 2005. 6(1): p. 88–98.
43. Donati, I. et al., The aggregation of pig articular chondrocyte and synthesis of extracellular matrix by
a lactose‐modified chitosan. Biomaterials, 2005. 26(9): p. 987–998.
44. Morimoto, M. et al., Biological activities of carbohydrate‐branched chitosan derivatives.
Biomacromolecules, 2001. 2(4): p. 1133–1136.
45. Sashiwa, H. and Y. Shigemasa, Chemical modification of chitin and chitosan – 2: preparation and
water soluble property of N‐acylated or N‐alkylated partially deacetylated chitins. Carbohydr. Polym.,
1999. 39(2): p. 127–138.
46. Minko, S., Grafting on Solid Surfaces: “Grafting to” and “grafting from” methods, in Polymer Surfaces
and Interfaces: Characterization, Modification and Applications. 2008, Springer Berlin Heidelberg:
Berlin, Heidelberg. p. 215–234.
Chemical and Enzymatic Modification of Chitosan to Produce New Functional Materials 257
47. Alves, N.M. and J.F. Mano, Chitosan derivatives obtained by chemical modifications for biomedical
and environmental applications. Int. J. Biol. Macromol., 2008. 43(5): p. 401–414.
48. Bhattacharya, A. and B.N. Misra, Grafting: A versatile means to modify polymers: Techniques, factors
and applications. Prog. Polym. Sci., 2004. 29(8): p. 767–814.
49. Thakur, V.K. and M.K. Thakur, Recent advances in graft copolymerization and applications of chi-
tosan: A review. ACS Sustain. Chem. Eng., 2014. 2(12): p. 2637–2652.
50. Shahid ul, I. and B.S. Butola, Recent advances in chitosan polysaccharide and its derivatives in antimi-
crobial modification of textile materials. Int. J. Biol. Macromol., 2019. 121: p. 905–912.
51. Jayakumar, R. et al., Graft copolymerized chitosan—present status and applications. Carbohydr.
Polym., 2005. 62(2): p. 142–158.
52. Zohuriaan‐Mehr, M.J., Advances in chitin and chitosan modification through graft copolymerization:
A comprehensive review. Iranian Polym. J., 2005. 14(3): p. 235–265.
53. Bhavsar, C. et al., Functionalized and graft copolymers of chitosan and its pharmaceutical applica-
tions. Exp. Opin. Drug Delivery, 2017. 14(10): p. 1189–1204.
54. Hu, W.Q. et al., Modification of chitosan grafted with collagen peptide by enzyme crosslinking.
Carbohydr. Polym., 2019. 206: p. 468–475.
55. Szwarc, M., Living polymers. Nature, 1956. 178(4543): p. 1168–1169.
56. Xu, F.J. and W.T. Yang, Polymer vectors via controlled/living radical polymerization for gene delivery.
Prog. Polym. Sci., 2011. 36(9): p. 1099–1131.
57. Fournier, D., R. Hoogenboom, and U.S. Schubert, Clicking polymers: A straightforward approach to
novel macromolecular architectures. Chem. Soc. Rev., 2007. 36(8): p. 1369–1380.
58. Mittal, H. et al., Recent progress in the structural modification of chitosan for applications in diversi-
fied biomedical fields. Eur. Polym. J., 2018. 109: p. 402–434.
59. Anghileri, A. et al., Tyrosinase‐catalyzed grafting of sericin peptides onto chitosan and production of
protein‐polysaccharide bioconjugates. J. Biotechnol., 2007. 127(3): p. 508–519.
60. Chen, T.H. et al., In vitro protein‐polysaccharide conjugation: Tyrosinase‐catalyzed conjugation of
gelatin and chitosan. Biopolymers, 2002. 64(6): p. 292–302.
61. Hu, W. et al., Modification of chitosan grafted with collagen peptide by enzyme crosslinking.
Carbohydr. Polym., 2019. 206: p. 468–475.
62. Payne, G.F., R.H. Chen, and H.D. Embree, In vitro biochemical coupling to create protein‐
polysaccharide conjugates. Abstracts of Papers of the American Chemical Society, 2002. 223:
p. D91–D91.
63. Chen, Y. et al., Preparation of porous carboxymethyl chitosan grafted poly(acrylic acid)superabsorbent
by solvent precipitation and its application as a hemostatic wound dressing. Mater. Sci. Eng.: C, 2016.
63: p. 18–29.
64. Ibrahim, A.G. et al., Chitosan‐g‐maleic acid for effective removal of copper and nickel ions from their
solutions. Int. J. Biol. Macromol., 2019. 121: p. 1287–1294.
65. Saber‐Samandari, S. and S. Saber‐Samandari, Biocompatible nanocomposite scaffolds based on
copolymer‐grafted chitosan for bone tissue engineering with drug delivery capability. Mater. Sci. Eng.
C‐Mater. Biol. Appl., 2017. 75: p. 721–732.
66. Wang, M.X. et al., Fabrication of bifunctional chitosan‐based flocculants: Characterization, assess-
ment of flocculation, and sterilization performance. Materials, 2018. 11(10): p. 20.
67. Samzadeh‐Kermani, A. and S. Miri, Synthesis, characterization and bactericidal property
of chitosan‐graft‐polyaniline/montmorillonite/ZnO nanocomposite. Korean J. Chem. Eng., 2015.
32(6): p. 1137–1141.
68. Adeyemi, S.A. et al., Synthesis and in vitro characterization of a pH‐responsive chitosan‐ polyethylen-
imine nanosystem for the delivery of therapeutic proteins. J. Drug Delivery Sci. Technol., 2017. 39: p.
266–276.
69. Patil, A.S. et al., Biocompatible tumor micro‐environment responsive CS‐g‐PNIPAAm co‐polymeric
nanoparticles for targeted Oxaliplatin delivery. J. Polym. Res., 2018. 25(3): p. 13.
258 Chitin and Chitosan: Properties and Applications
70. Yao, F.L. et al., Synthesis and characterization of chitosan grafted oligo(L‐lactic acid). Macromol.
Biosci., 2003. 3(11): p. 653–656.
71. Yao, F.L. et al., A study on cytocompatible poly(chitosan‐g‐L‐lactic acid). Polymer, 2003. 44(21):
p. 6435–6441.
72. Shi, Z. et al., Effects of graft copolymer of chitosan and salicylic acid on reducing rot of postharvest
fruit and retarding cell wall degradation in grapefruit during storage. Food Chem., 2019. 283:
p. 92–100.
73. Xu, W.Y. et al., Preparation and characterization of C‐phycocyanin peptide grafted N‐succinyl chitosan
by enzyme method. Int. J. Biol. Macromol., 2018. 113: p. 841–848.
74. Han, Y.F. et al., Preparation and characterization of chitosan‐based nanoparticles as protein delivery
system. Adv. Polym. Technol., 2018. 37(4): p. 1214–1220.
75. Zou, Y.J., S.S. He, and J.Z. Du, epsilon‐Poly(L‐lysine)‐based Hydrogels with Fast‐acting and
Prolonged Antibacterial Activities. Chinese J. Polym. Sci., 2018. 36(11): p. 1239–1250.
76. Timur, M. and A. Paşa, Synthesis, characterization, swelling, and metal uptake studies of aryl cross‐
linked chitosan hydrogels. ACS Omega, 2018. 3(12): p. 17416–17424.
77. Rokhade, A.P. et al., Semi‐interpenetrating polymer network microspheres of gelatin and sodium car-
boxymethyl cellulose for controlled release of ketorolac tromethamine. Carbohydr. Polym., 2006.
65(3): p. 243–252.
78. Rokhade, A.P. et al., Novel interpenetrating polymer network microspheres of chitosan and methylcel-
lulose for controlled release of theophylline. Carbohydr. Polym., 2007. 69(4): p. 678–687.
79. Kim, S.J. et al., Thermal characteristics of polyelectrolyte complexes composed of chitosan and hya-
luronic acid. J. Macromol. Sci.‐Pure Appl. Chem., 2003. A40(8): p. 807–815.
80. Kim, S.J. et al., Swelling kinetics of interpenetrating polymer hydrogels composed of poly(vinyl alco-
hol)/chitosan. J. Macromol. Sci.‐Pure Appl. Chem., 2003. A40(5): p. 501–510.
81. Kim, S.J. et al., Water sorption of poly(vinyl alcohol)/poly(diallyldimethylammonium chloride) inter-
penetrating polymer network hydrogels. J. Appl. Polym. Sci., 2003. 90(5): p. 1389–1392.
82. Kim, S.J. et al., Electrical response characterization of chitosan/polyacrylonitrile hydrogel in NaCl
solutions. J. Appl. Polym. Sci., 2003. 90(1): p. 91–96.
83. Kim, S.J. et al., Thermal characterizations of semi‐interpenetrating polymer networks composed
of poly(ethylene oxide) and poly(N‐isopropylacrylamide). J. Appl. Polym. Sci., 2003. 90(14):
p. 3922–3927.
84. Kim, S.J. et al., Thermal characteristics of interpenetrating polymer networks composed of poly(vinyl
alcohol) and poly(N‐isopropylacrylamide). J. Appl. Polym. Sci., 2003. 90(3): p. 881–885.
85. Kim, S.J. et al., Water and temperature response of Semi‐IPN hydrogels composed of chitosan and
polyacrylonitrile. J. Appl. Polym. Sci., 2003. 88(12): p. 2721–2724.
86. Kim, S.J. et al., Electrical sensitive behavior of a polyelectrolyte complex composed of chitosan/hya-
luronic acid. Solid State Ionics, 2003. 164(3–4): p. 199–204.
87. Kim, S.E. et al., Porous chitosan scaffold containing microspheres loaded with transforming
growth factor‐beta 1: Implications for cartilage tissue engineering. J. Control. Release, 2003. 91(3):
p. 365–374.
11
Chitosan‐Based Drug
Delivery Systems
Cristian Peptu1, Andra Cristina Humelnicu1, Razvan Rotaru1, Maria
Emiliana Fortuna1, Xenia Patras1, Mirela Teodorescu1, Bogdan Ionel
Tamba2, and Valeria Harabagiu1
1
‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian Academy,
Iași, Romania
2
A&B Pharm Corporation, Roman, Neamț, Romania
Drug delivery systems (DDSs) represent one of the most active research topics with a
history as long as more than 50 years, and it is an increasing actual interest focused on
sustained/controlled and targeted pharmaceutical formulations able to provide increased
compliance of the patients, personalized, and more efficient therapeutic effects. DDSs of
different shapes ranging from microparticles to nanoparticles (NPs), from hydrogels to
scaffolds, and membranes containing the drugs embedded into organic, inorganic,
polymeric matrices or combinations between them were investigated, a few of them being
already commercialized.
Chitosan and chitosan‐modified polymers are one of the most studied materials of natural
origin in their use as matrices for the preparation of DDSs. This chapter describes the unique
properties of chitosan/modified chitosan as cationic polymers in providing increased bioa-
vailability and higher therapeutic efficiency of various anionic drugs/genetic materials or of
hydrophobic/hydrophilic drugs as a result of their higher bioadhesion to different tissues/
mucosa, transfection properties, efflux pump inhibition or permeation across biological bar-
riers (oral, nasal, and vaginal mucosa or skin, ophthalmic, gastrointestinal, and blood–brain
barriers (BBB)). Recent and representative results on the preparation and applications of
chitosan/chitosan derivatives–based DDSs are also reviewed.
11.1 Introduction
Drug delivery systems (DDSs) could be defined as engineered devices aimed to provide
sustained, controlled, and/or targeted release of therapeutic agents. These systems are
composed of the therapeutic agent embedded into a matrix that could be an organic/
inorganic compound or a polymer. The main advantages of controlled‐release systems are
(i) the reduction of the drug side effects by maintaining their concentration in the therapeutic
range between the minimum effective and the toxic concentrations, (ii) increased
compliance of the patients by diminishing the number of administrations, and (iii) reduced
amounts of therapeutic agents. However, one should also mention the drawbacks of such
systems linked to the low biocompatibility and toxicity of the matrix or of its biodegradation
compounds, any discomfort of the patient due to the administration method (e.g., surgery
for implantation) or a higher cost as compared to the traditional drug administration as
solutions and ointments.
Three steps in the development of DDSs were identified by Park 2014. Thus, during a
30‐year period, in the second part of the twentieth century (1950–1980), the first generation
of pharmaceutical formulations of sustained release was commercialized for oral and
transdermal delivery systems, the physicochemical properties of controlled DDSs were
established, and the mechanisms of drug release (dissolution, diffusion, osmosis, and ion‐
exchange) were identified as a function of drug/matrix nature and of the administration
procedure. That time, macroscopic and microscopic polymeric DDSs were mainly
developed.
The second generation of DDSs (1980–2010) started with the development of the zero‐
order release kinetics formulations, considered to be the only systems able to provide a
steady blood concentration of the drug. However, the thinking “flatter is better” [1] was
proved to be inappropriate, as an efficient but variable drug concentration between the
lower and higher limits of the therapeutic range is registered for many delivery systems, on
one hand, and zero‐order release kinetic does not always warrant a constant blood
concentration. This is especially the case of the oral drug administration since a decrease
of the drug absorption was observed when DDS moves from upper small intestine to the
colon. Moreover, for some drugs—nitroglycerin, pituitary gland hormones, and insulin—a
constant blood concentration is even not desired. Thus, other release kinetics was proposed
for both biodegradable and nonbiodegradable matrices [2]. Drug conditioning into
cyclodextrin macrocycles and sustained drug release from the obtained inclusion complexes
were also investigated [3–9]. An important research effort was done to elaborate stimuli‐
responsive [10–14] and targeted DDSs [15], part of them being prepared by using
nanotechnology concepts, but only a few formulations achieved the clinical application
maturity due to the difficulties in overcoming the biological barriers, a challenging issue to
be solved during the next period.
Although a pioneering report on NPs designed for drug delivery appeared many years
ago [16], the miniaturization of DDSs at nanoscale (the third generation) was boosted by
introducing the nanoscience and nanotechnology program in USA, the beginning of 1990s,
since a growing interest in nanomedicine was registered everywhere in the world, generating
valuable results on liposomal formulations, polymer–drug conjugates [17–21], or
poly(ethylene glycol) ((PEG)ylated) NPs [22, 23] showing long circulation in the body
[24]. The enhanced permeation and retention effect by the conjugates accumulated in
Chitosan‐Based Drug Delivery Systems 261
tumor due to the “leaky” vasculature [25] was developed and the effects of passive
(determined by NP physicochemical properties, dimension, and shape) and active targeting
(generated by affinity ligands linked to NP surface) were compared [26].
A recent paper presenting very interesting views of medical specialists from Harvard
Medical School [27] considers the evolution of the DDSs from macroscopic (1960s) to
microscopic (1980s) and nanoscopic formulations (1990s). The advantages and
disadvantages of each type of drug delivery formulations are described, and the classical
and new mechanisms and mathematical models for the drug release are also compared.
Several macro‐ and microscaled DDSs that were already commercialized and of targeted
nanoscaled‐drug delivery formulations which are in the phases of preclinical and clinical
testing are presented in Table 11.1 (entrees 1–8, 9–11, and 12–18, respectively). More
information on commercialized DDSs was published by Kearney and Mooney (2013) [28].
Many research efforts were focused on the development of preparation methods, such
as ProLease technique to obtain injectable microparticles with prolonged release [29],
nanoprecipitation [30, 31] and nanoemulsion [32, 33] or microfluidics [34] to allow the
nanoentrapment of hydrophobic and hydrophilic drugs, the last mentioned technique
offering the advantages of a better control of NP dimensions and a higher content of
encapsulated drug. Drug delivery NPs containing two drugs dispersed in polymeric layers
of different structures were also prepared through layer‐by‐layer deposition, a method that
allows the release rate control of each drug [35].
When choosing a polymer to be used as a matrix in the preparation of DDSs, one should
consider its physicochemical (chemical structure/functionality, molecular weight,
solubility, degradability, etc.) and biological properties as well as the drug properties and
mechanism of action. This chapter concentrates on chitosan‐based DDSs considering the
biological properties of this biopolymer that generate unique end‐use properties of
pharmaceutical formulations containing either pristine chitosan, chemically/physically
modified chitosan or combinations of chitosan with other polymers (Figure 11.1). As very
rich information focusing on chitosan‐based DDSs is available in review articles [56–64],
only the most recent and representative results on different formulations shapes (micro/
NPs, gels, membranes, nanofibers, etc.) and their specific applications are discussed here.
No. DDSs type: release mechanism Matrix* Active principle Reference Commercial trademark: therapeutic
application
1 Reservoir: diffusion Silicone Anesthetic gases [36] First report on macroscaled DDS
2 Implant: diffusion Progestin levonorgestrel [37] Norplant: contraceptive drug
Progesterone
3 Implant: reservoir diffusion PEVA Pilocarpine [38] Ocusert: intraocular antiglaucoma drug
4 Implant: reservoir diffusion Progesterone [38, 39] Progestasert: intrauterine contraceptive drug
5 Skin patch: zero‐order kinetics Scopolamine [40] Transderm‐Scop: drug treating motion sickness
6 Oral delivery DDSs: pulsatile Polymeric capsules with drilled Salbutamol, nifedipine, [41] Different commercialized drugs
release, osmotic control surface membrane hydromorphone, verapamil,
paliperidone
7 Polymeric tablet: swelling, HPMC Diclofenac, molsidomine, [42] Geomatrix: platform for different drugs
gelling, erosion zileuton, nisoldipine, etc.
8 Disk‐like brain implant Poly(carboxyphenoxy Bis(2‐chloroethyl) nitrosourea [43] Gliadel: treatment of brain cancer
propane)—sebacic acid
9 Depot microparticles PLGA Luteinizing hormone‐releasing [44] Decapentyl LP or Lupron Depot: treatment of
hormone prostate cancer
10 Injectable microspheres PLGA Risperidone [45] Risperdal Consta: antipsychotic drug
11 Microparticles Poly(butylene terephthalate) Interferon‐α2b [46] Locteron: (treatment of hepatitis C
12 Micelle formulation PEG–PLA Paclitaxel (treatment of different [47] Genexol‐PM: antitumor
tumors)
13 Drug‐loaded NPs Poly(L‐glutamic acid) Paclitaxel [15] Xyotax: ovarian and lung cancer
14 The systems are in different Cyclodextrin‐ polymer conjugate Camptothecin [48–50] CRLX‐101 or IT‐101
15 phases of preclinical and PEG–PLA Docetaxel [51] BIND‐014: prostate, non‐small lung, cervical,
clinical studies. head and neck cancers
16 Adamantine‐PEG and siRNA [52] CALAA‐01: siRNA delivery
adamantine‐PEG conjugated
with human transferrin
17 PLGA and PEG–PLA Nicotine antigen, T‐helper cell [53, 54] SEL‐068: antismoking vaccine (first NP for
peptide, and adjuvant vaccine formulation)
Under clinical trials at SELECTABIO: www.
selectabio.com
18 Poly(isohexyl cyanoacrylate) Doxorubicin [55] Livatag: treatment of hepatocellular carcinoma
Note: *PEVA = poly(ethylene‐co‐vinyl acetate); HPMC = hydroxypropyl methylcellulose; PLGA = poly(lactic acid‐co‐glycolic acid) copolymers; PEG-PLA = poly(ethylene glycol -co- lactic acid)
copolymers; and NPs = nanoparticles; DDSs = drug delivery systems.
inorganic compounds, metallic or metal oxide NPs, or other polymers [58, 59, 67]. Chitosan
used in the formulation of DDSs is mainly based on its cationic nature that provides useful
properties such as interactions with anionic drugs, mucoadhesion, increased permeation,
and transfection or inhibition of the pump efflux to the system [68].
CHITIN
CH3
OH O
Crustaceans O HO NH
O O O
HO
O
NH
O
Squid Deacetylation OH
CH3
CH3
OH O
O HO NH
O O
Fungi O
HO O
NH2
Insects OH
DRUG DELIVERY
SYSTEMS
Sustained release
(antibiotics; anti-inflammatory Modulated release Targeted release
Gene therapy
drugs, growth factors, (insulin) (tumoral cells)
steroids, diuretics)
Figure 11.1 Properties of chitosan/modified chitosan and their applications in the preparation of drug
delivery systems.
vectors [83]. However, the transport capacity of chitosan is lower as compared to the viral
vectors. Branched structures [84, 85], thiolated chitosan [86, 87], thiolated/PEGylated chi-
tosan able to stabilize the supramolecular complexes [88], poly(L‐lysine)‐grafted chitosan
[89], and chitosan/cyclodextrin NPs [90] were found to possess improved transfection
properties.
Chitosan‐Based Drug Delivery Systems 265
natural protection of certain biological barriers. The intracellular delivery of the drug
molecules must defeat the cellular membrane and ensure transcellular transport, while the
passage through a tissue may occur either by defeating the cellular membrane or by pas-
sage through the intercellular junctions (paracellular transport). Some drugs may penetrate
the biological membranes by passive diffusion and without energy consumption, while
other drugs require an energetically activated mechanism. The drug delivery devices are
aimed to facilitate the drug penetration and, among these devices, the nanoparticulate car-
riers are proved to be a very effective tool for various types of biological membranes.
Chitosan‐based nanocarriers raised an enormous interest due to the above-mentioned
outstanding properties promoting their use for the preparation of DDSs across various
biological membranes.
r eservoir for hydrophilic proteins. The addition of chitosan increased the average NPs diam-
eter from approx. 60 to 70 nm. However, fluorescence analysis of the penetration efficiency
clearly demonstrated that chitosan‐conjugated nanocarriers were superior to bare nanocar-
rier, in spite of the size difference. The hydrophilic nature of chitosan is limiting its use as a
carrier for hydrophobic drugs. Recent studies [109] focused on the use of cyclodextrin–
warfarin complexes in combination with chitosan NPs for transdermal delivery. Thus, it was
demonstrated that cyclodextrin increases the drug amount to be loaded in the chitosan NPs
and the overall transdermal delivery of warfarin. Moreover, cyclodextrins were proved to act
as skin penetration enhancers [110]. Tacrolimus (FK506), a potent macrolide immunosup-
pressive agent, was delivered using NPs (average size of 110 nm) based on a combination of
chitosan and nicotinamide, a water‐soluble derivative of vitamin B3, proved to enhance the
solubility of some poorly water‐soluble molecules [111]. The abovementioned studies
revealed that the drug‐complex loaded particles were more effective than neat drug‐complex
in delivery at the level of both superficial and deeper layers of the skin. Recent studies, [112]
by using confocal laser scanning microscopy, demonstrated that the curcumin‐loaded
chitosan NPs, having different average diameters ranging from 70 to 190 nm, get accumu-
lated in the hair follicles where they act as reservoirs for the drug delivery.
effective in drug delivery via the nasal mucosal barrier. The successful nasal delivery of
insulin‐mediated by PEG‐graft‐chitosan nanogels with average diameters from 175 to
280 nm showed that, besides the size of the nanocarriers, the surface charge is also very
important for successful drug delivery [113]. Chitosan particulate carriers were also found
effective for delivery of vaccines via the nasal pathway, as previously reviewed [114]. The
modifications of chitosan, as in N‐trimethyl chitosan (TMC) or mono‐N‐carboxymethyl
chitosan (MCC), were tested in view of the performance in nasal delivery of tetanus toxoid
antigen [115]. The TMC‐based particles had positive surface charge, while MCC had
negatively charged surface. The delivery performance of the tetanus toxoid antigen was
found slightly better in the case of TMC NPs characterized by smaller average diameters,
a feature that compensates for the lack in positive charges for the enhancement of the
delivery through the nasal mucosa. The performance of chitosan NPs can be further
enhanced by combination with other delivery strategies. In a recent study, chitosan was
conjugated with monophosphoryl lipid A as a novel formulation for use as a mucosal
adjuvant to generate protective immune responses against Mycobacterium tuberculosis
infection [116].
attacks, good contact with the intestinal epithelium due to high surface/volume ratio, and
appropriate size to favor the trans‐ and paracellular passage. The chitosan‐based particles
were proven to be able to disrupt the epithelial tight junctions due to its positively charged
nature. However, the low pH in the stomach induces changes of the chitosan nanoparticulate
carriers, and, as a consequence, chitosan has been modified or used in combination with
other polymers. Thus, a succinic anhydride derivative (2‐dodecenyl succinic anhydride)
was used to modify the chitosan amino groups [117]. The obtained product, chitosan with
increased hydrophobicity, was further used to prepare insulin‐loaded NPs, and the authors
observed that the release at intestinal pH is slightly reduced as compared with unmodified
chitosan but the protection against acid pH was significantly improved. In a recent study,
polymeric NPs based on quaternary ammonium‐chitosan and S‐protected thiolated
derivative were prepared by cross‐linking low‐molecular‐weight hyaluronic acid [118]. It
was shown that S‐protected thiolated chitosan derivative has better mucoadhesion, probably
because of the cleavage of the S–S bonds and exchange reactions with the thiol components
of the mucus. The increased adhesion was considered responsible for prolonging the drug
residence (a model drug for insulin) at the absorption site, and enhancing the drug
bioavailability. In another study, chitosan was combined with cyclodextrins in order to
enhance the intestinal delivery of peptide glutathione [100]. Transport studies performed in
the frog intestine model confirmed that both types of NPs could induce permeabilization of
the intestinal epithelia; however, the cyclodextrin‐based NPs exhibited absorption‐
enhancing properties in all segments of the duodenum, whereas chitosan‐only NPs effect
was restricted to the first segment of the duodenum.
the drugs may be delivered to the anterior or to the posterior sides of the eye. The noninvasive
drug delivery via the anterior side of the eye, topical instillation, is the most commonly
used pathway, [122] and must defeat the lacrimal clearance. The posterior eye treatment
may use the periocular pathway which must pass several barriers (ocular tissues)—
episclera, sclera, choroid, Bruch’s membrane, and retinal pigment epithelium—to reach the
retina and vitreous humor [123]. Besides the tissues bypass, there is also a dynamic barrier
consisting of blood flow, lymphatic drainage, fluid flow from the intraocular drainage
systems, and enzymatic degradation. A more direct drug delivery pathway may be used
when large amounts of drugs are needed in the posterior eye segment, and that is the
intravitreal injection. However, potential side effects such as retinal detachment, cataract,
endophthalmitis, etc., may prevent its usage. Nevertheless, intravitreal administration is the
preferred drug delivery route in treating diseases of the posterior segment, achieving the
highest intraocular bioavailability [124].
Topical administration of drugs using chitosan NPs (around 400 nm diameter) suggests
that they may penetrate into the corneal and conjunctival epithelia as demonstrated by
confocal laser scanning microscopy studies [125]. The prepared chitosan NPs were able to
interact and remain associated to the ocular mucosa for extended periods of time, thus
enabling the local drug delivery. Chitosan was also combined with cyclodextrins in order
to improve the drug delivery capacity. Thus, the naringenin‐loaded sulfobutylether‐β‐
cyclodextrin/chitosan NPs were shown to have better ability to prolong the ocular mucosa
residence time than the naringenin suspension [126]. Also, chitosan was used together with
alginate in the preparation of NPs by polyanion–polycation complexation. Thiolated
chitosan–alginate NPs were showed to have improved mucoadhesive properties when
tested on corneal tissue [127]. Hyaluronic‐acid‐modified lipid–polymer hybrid NPs were
designed to improve the ocular bioavailability of the strongly hydrophilic moxifloxacin
hydrochloride [128]. The proposed ocular drug delivery platform could effectively cross
the ocular tissues through both para‐ and transcellular routes. The authors propose that the
internal core of the lipid–polymer hybrid NPs could transiently open the intercellular tight
junctions by altering the levels of junction protein. Next, the external lipid layer possessed
biomimetic advantages and could significantly prolong precorneal retention. Furthermore,
the surface‐modified hyaluronic acid in the structure could readily target the CD44
receptors on epithelial cells and promote endocytosis.
The chitosan‐based NP systems were not only tested for topical delivery of adrenaline
[129] but also for intravitreal injection administration of a model drug, cefuroxime.
Chitosan–gelatin nanocapsules prepared by double cross‐linking in double emulsion were
tested for intraocular drug delivery via intravitreal injection [130]. This experimental study
demonstrated the ability of fluorescent chitosan‐based nanocarriers to penetrate ocular
tissues close to the administration site (intravitreal injection), and especially their tendency
to migrate deeply in the retina at time intervals of 72 hours. Moreover, after initial burst
release, the NPs continue to release the drug up to 96 hours, while the drug administered as
the solution is removed rapidly by the intravitreal clearance system.
most of the drugs. It is built of endothelial cells and makes part of the capillary walls. In
normal capillary vessels, the endothelial cells are connected by more loose junctions but in
the case of the BBB capillaries, the endothelial cells are connected through tight junctions
which are regulating the diffusion processes from the blood to the brain. The delivery of
therapeutic agents to the brain may be performed by invasive and noninvasive methods.
The invasive methods are consisting of direct injection of drugs or drug delivery devices at
CNS level or by temporary physical disruption of BBB using ultrasounds, osmotic shocks,
etc. The noninvasive methods consist of using chemical BBB penetration enhancers (e.g.,
lipid nanocarriers or lipid conjugation of drugs), biological enhancers (drug conjugation
with biologically active ligands that insure the transport across BBB), and the delivery via
nasal mucosa (the olfactory and trigeminal nervous tract shortcut). The delivery via nasal
mucosa pathway was previously detailed and, in the following text, only the delivery across
BBB using chitosan colloidal carriers will be discussed. The mechanisms of breaching
through BBB for CNS‐targeted drug delivery may consider several strategies [131]. One
possibility is that the NPs may be retained in the BBB capillaries and from there they may
create a local concentration gradient that might enhance the drug transport across the
barrier. Also, a certain disruption of the BBB by affecting the epithelial cells membrane of
tight junctions may enhance the NPs permeation through the BBB. Another strategy may
consist of surface masking of the NPs in order to be transported through the BBB cell via
activated transport mechanisms.
The studies of Yemisci et al. (2015) [132] employed the avidin–biotin coupling technol-
ogy to design chitosan nanospheres conjugated with PEG and bearing the OX26 monoclonal
antibody whose affinity for the transferrin receptor may trigger receptor‐mediated transport
across the BBB. The NPs having a diameter of approximately 200 nm were observed in the
lumen of the blood vessels and also in the brain interstitium, thus demonstrating the success
of the delivery strategy. Other studies showed that the results obtained by this strategy are
highly dependent on the degree of surface functionalization [133]. Thus, NPs having too
many interacting sites may remain bound to BBB and have reduced drug delivery efficiency.
A similar approach used nanoprobes comprising an iron oxide NP coated with a PEGylated
chitosan‐branched copolymer, to which a targeting ligand, chlorotoxin, and a near‐infrared
fluorophore, Cy.5.5, were conjugated [134]. In this design, chitosan was used as a linker to
anchor the polymer to the iron oxide surface through electrostatic interaction and physical
adsorption. Also, the chitosan’s role was to stabilize the nanostructure by alleviating the
need for cross‐linking agents and to provide sites for subsequent conjugation of ligands
without the need for further chemical modification. Moreover, the bound PEGylated chi-
tosan also acts as a sterically stabilizing corona, preventing particle aggregation under physi-
ologic conditions. More recent studies employing the inhibition of BBB cellular uptake by
chlorpromazine clearly demonstrated that the transferrin receptor antibody‐conjugated NPs
were preferentially crossing BBB via the receptor‐mediated endocytosis pathway [135].
(Continued)
11.4.1 Hydrogels
Hydrogels are cross‐linked, three‐dimensional polymeric materials prepared by chemical
and physical methods [137]. They were intensively studied as carriers for bioactive
compounds (low molecular drugs, nucleic acids, proteins) or cells. In situ gelling can be
realized by solvent exchange, UV‐irradiation, ionic cross‐linking, pH and temperature
changes [138–142], by using the reaction of thiolated chitosan with oxidizing agents, [143]
or the dynamic imino–chitosan chemistry [144, 145].
One of the most studied systems is represented by thermosensitive hydrogels [146–151].
Recent studies have described the sustained release of doxorubicin [152] and dopamine/
metronidazole [153] from injectable hydrogels in the treatment of liver cancer and
Parkinson’s disease, respectively. In another study, different hydrophilic gel‐forming
matrix tablets based on chitosan and anionic polymers for sustained oral release of sodium
valproate and valproic acid were evaluated [154].
11.4.2 Micro/NPs
Chitosan‐based micro/nanoparticulate systems were extensively investigated as DDSs.
Therefore, for the development of micro/NP forms, researchers used various methods such
as emulsion cross‐linking [207], spray drying [182], precipitation [178, 179], coacervation
[208], ionic gelation [180], sieving [183], or reverse micellar technique [209]. Among
drug‐loading methods, another important factor to be considered is the size of the particles,
which has to be in accordance with the administration route [186, 210].
Recent papers demonstrated significantly enhanced drug delivery from chitosan NPs
after conjugation with folic acid [184], grafting with silica [185], obtaining ZnO‐based
nanocomposites [189] or introducing the magnetic properties of Fe3O4 [188].
11.4.3 Nanofibers
Based on the large values of their surface/volume ratio and porosity, the nanofibrous mate-
rials obtained by electrospinning of chitosan, chitosan derivatives, or combinations with
other polymers [211–214] were shown to possess higher drug‐loading capacity as com-
pared to similar materials but of other morphologies, enhanced drug transport properties as
well as cell attachment properties [215]. To improve the ability of chitosan nanofibers for
drug and gene delivery, their functionalization [216] or the preparation of composite
nanofibers were also proposed [175, 217, 218].
specific uses, they were charged with antibiotics, anti‐inflammatory, analgesic drugs,
proteins, growth factors, nucleic acids, or cells [110, 223, 224].
11.5 Outlook
As a result of high impact on human health and well‐being, DDSs represent one of the most
studied topics at frontiers between chemistry, biology, medicine, and bioengineering.
During more than 50 years of fundamental and applied research, DDSs preparation
procedures, as well as their shapes and administration routes, were continuously diversified.
Based on fundamental insights into the release mechanisms correlated with the biological
mechanisms of drug action, the performance of DDSs evolved from sustained release and
long in vivo circulation to controlled and targeted delivery.
Chitosan is one of the most studied biopolymers for application as a carrier in DDSs. The
interest in this biopolymer resides in its physicochemical characteristics (easily to be modulated
by changing the molecular weight, the degree of deacetylation or by chemical and physical
modifications of chitosan with organic/inorganic compounds or other biopolymers) and benefi-
cial biological properties mainly based on its cationic chemical structure (biodegradation, inter-
action with anionic drugs/tissue components, adhesion to different mucosa, inhibition of pump
efflux or promoting increased permeation of different drugs and transfection of nucleic acids
and proteins). Moreover, chitosan and its metabolic degradation products are non‐toxic. Thus,
hydrogels, micro/NPs, nanofibers or scaffolds, and membranes containing chitosan‐based
carriers for DDSs addressed to various diseases and able to surpass skin, mucosa, gastrointesti-
nal, ophthalmic, and blood–brain biological barriers were prepared and tested.
However, there are still challenges in the development and commercialization of efficient
DDSs related to analytical methods to evaluate in vivo the biocompatibility/toxicity of
complex drug formulations or to mathematical models meant to correlate the in vitro
release to the in vivo pharmacokinetic profile of specific drugs. With the increasing
incidence of diabetic and oncological diseases, patients and medical doctors are expecting
innovative solutions from researchers and bioengineers for strict “on–off” release
mechanism, for modulated insulin delivery, efficient tumor‐targeting systems, and long‐
term depot formulations for proteins and peptides without important burst effect.
Acknowledgment
The financial support of European Social Fund for Regional Development, Competitiveness
Operational Programme Axis 1—Project “Petru Poni Institute of Macromolecular
Chemistry—Interdisciplinary Pol for Smart Specialization through Research and
Innovation and Technology Transfer in Bio(nano)polymeric Materials and (Eco)
Technology,” InoMatPol (ID P_36_570, Contract 142/10.10.2016, cod MySMIS: 107464),
is gratefully acknowledged.
References
1. Park, K. (2014). The controlled drug delivery systems: Past forward and future back. Journal of
Controlled Release 190 (9):3–8
2. Fu, Y. and Kao, W.J. (2010). Drug release kinetics and transport mechanisms of non‐degradable and
degradable polymeric delivery systems. Expert Opinion on Drug Delivery 7:429–444.
Chitosan‐Based Drug Delivery Systems 277
3. Uekama, K., Hirayama, F., and Irie, T. (1998). Cyclodextrin drug carrier systems. Chemical Reviews
98:2045–2076.
4. Spulber, M., Pinteala, M., Fifere, A., Harabagiu, V., and Simionescu, B.C. (2008a). Inclusion com-
plexes of 5‐flucytosine with β‐cyclodextrin and hydroxypropyl‐β‐cyclodextrin: Characterization in
aqueous solution and in solid state. Journal of Inclusion Phenomena and Macrocyclic Chemistry
62(1–2):117–125.
5. Spulber, M., Pinteala, M., Fifere, A., Moldoveanu, C., Mangalagiu, I., Harabagiu, V., and Simionescu,
B.C. (2008). Water soluble complexes of methyl β‐cyclodextrin and sulconazole nitrate. Journal of
Inclusion Phenomena and Macrocyclic Chemistry 62(1–2):135–142.
6. Fifere, A, Budtova, T., Tarabukina, E., Pinteala, M., Peptu, C., Harabagiu, V., and Simionescu, B.C.
(2009). Inclusion complexes of γ‐cyclodextrin and carboxyl‐modified γ‐cyclodextrin with C60:
Synthesis, characterization and controlled release application via microgels. Journal of Inclusion
Phenomena and Macrocyclic Chemistry 64:83–94.
7. Miron, L., Mares, M., Nastasa, V., Spulber, M., Pinteala, M., Fifere, A, Harabagiu V., and Simionescu,
B.C. (2009). Water soluble sulconazole/β‐cyclodextrin complex: Physico‐chemical characterization
and preliminary pharmacological studies. Journal of Inclusion Phenomena and Macrocyclic Chemistry
63:159–162.
8. Fifere, A., Spulber, M., Marangoci, N., Fifere, N., Pinteala, M., Harabagiu, V., and Simionescu, B.C.
(2011). Semi‐empirical PM3 study on complexation of β‐cyclodextrin with 5‐flucytosine. Cellulose
Chemistry and Technology 45:149–156.
9. Neamtu, A., Marangoci, N., and Harabagiu, V. (2013). β‐cyclodextrin/propiconazole complexes
probed by constraint free and biased molecular dynamics simulations. Revista de Chimie
64(5):502–508.
10. Fundueanu, G., Constantin, M., Oanea, I., Harabagiu, V., Ascenzi, P., and Simionescu, B.C. (2012).
Prediction of the appropriate size of drug molecules that could be released by a pulsatile mechanism
from pH/thermoresponsive microspheres obtained from preformed polymers. Acta Biomaterialia
8(3):1281–1289.
11. Asmarandei, I., Fundueanu, G., Cristea, M., Harabagiu, V., and Constantin, M. (2013). Thermo‐ and
pH‐sensitive interpenetrating poly(N‐isopropylacrylamide)/carboxymethyl pullulan network for drug
delivery. Journal of Polymer Research 20(11):293–305.
12. Fundueanu, G., Constantin, M., Asmarandei, I., Harabagiu, V., Ascenzi, P., and Simionescu, B.C.
(2013). The thermosensitivity of pH/thermoresponsive microspheres activated by the electrostatic
interaction of pH‐sensitive units with a bioactive compound. Journal of Biomedical Materials
Research – Part A 101 A(6):1661–1669.
13. Constantin, M., Bucatariu, S., Harabagiu, V., Popescu, I., Ascenzi, P., and Fundueanu, G. (2014).
Poly(N‐isopropylacrylamide‐co‐methacrylic acid) pH/thermo‐responsive porous hydrogels as self‐
regulated drug delivery system. European Journal of Pharmaceutical Sciences 62:86–95.
14. Filippov, A., Tarabukina, E., Simonova, M., Kirila, T., Fundueanu, G., Harabagiu, V., Constantin, M.,
and Popescu, I. (2015). Synthesis and investigation of double stimuli behavior of N‐isopropylacryla-
mide and maleic acid copolymer in solutions. Journal of Macromolecular Science Part B: Physics
54(9):1105–1121.
15. Galic, V.L., Wright, J.D., Lewin, S.N., and Herzog, T.J. (2011). Paclitaxel poliglumex for ovarian
cancer. Expert Opinion on Investigational Drugs 20:813–821.
16. Kreuter, J. and Speiser, P. (1976). In vitro studies of poly(methylmethacrylate) adjuvants. Journal of
Pharmaceutical Science 65:1624–1627.
17. Langer, R. (1990). New methods of drug delivery. Science 249:1527–1533.
18. LaVan, D.A., McGuire, T., and Langer, R. (2003). Small‐scale systems for in vivo drug delivery.
Nature Biotechnology 21:1184–1191.
19. Wagner, V., Dullaart, A., Bock, A.K., Zweck, A. (2006) The emerging nanomedicine landscape. Nature
Biotechnology 24:1211–1217.
278 Chitin and Chitosan: Properties and Applications
20. Farokhzad, O.C. and Langer, R. (2009). Impact of nanotechnology on drug delivery. ACS Nano
3:16–20.
21. Shi, J., Xiao, Z., Kamaly, N., and Farokhzad, O.C. (2011). Self‐assembled targeted nanoparticles:
Evolution of technologies and bench to bedside translation. Accounts of Chemical Research
44:1123–1134.
22. Torchilin, V.P. (1994). Immunoliposomes and PEGylated immunoliposomes: Possible use for t argeted
delivery of imaging agents. ImmunoMethods 4:244–258.
23. Gabizon, A., Shmeeda, H., and Barenholz, Y. (2003). Pharmacokinetics of PEGylated liposomal
Doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics 42:419–436.
24. Gref, R., Minamitake, Y., Peracchia, M. T., Trubetskoy, V., Torchilin, V., and Langer, R. (1994).
Biodegradable long‐circulating polymeric nanospheres. Science 263:1600–1603.
25. Maeda, H. (2010). Tumor‐selective delivery of macromolecular drugs via the EPR effect: Background
and future prospects. Bioconjugate Chemistry 21:797–802.
26. Bertrand, N., Wu, J., Xu, X., Kamaly, N., and Farokhzad, O.C. (2014). Cancer nanotechnology: The
impact of passive and active targeting in the era of modern cancer biology. Advanced Drug Delivery
Reviews 66:2–25.
27. Kamaly, N., Basit Yameen, B., Wu, J., and Farokhzad, O.C. (2016). Degradable controlled‐release
polymers and polymeric nanoparticles: Mechanisms of controlling drug release. Chemical Review
116:2602–2663.
28. Kearney, C.J. and Mooney, D.J. (2013). Macroscale delivery systems for molecular and cellular
payloads. Nature Materials 12:1004–1017.
29. Gombotz, W.R., Healy, M.S., and Brown, L. (1991). Very low temperature casting of controlled release
microspheres. U.S. Patent US5019400 A, filed 01 May, 1989, issued 28 May, 1991.
30. Quintanar‐Guerrero, D., Allemann, E., Fessi, H., and Doelker, E. (1998). Preparation techniques and
mechanisms of formation of biodegradable nanoparticles from preformed polymers. Drug Development
and Industrial Pharmacy 24:1113–1128.
31. Farokhzad, O.C., Cheng, J.J., Teply, B.A., Sherifi, I., Jon, S., Kantoff, P.W., Richie, J.P., and Langer,
R. (2006). Targeted nanoparticle‐aptamer bioconjugates for cancer chemotherapy in vivo. Proceedings
of the National Academy of Sciences of the United States of America 103:6315–6320.
32. Anton, N., Benoit, J.P., and Saulnier, P. (2008). Design and production of nanoparticles formulated
from nano‐emulsion templates – a review. Journal of Controlled Release 128:185–199.
33. Avgoustakis, K. (2004) PEGylated poly(lactide) and poly(lactide‐coglycolide) nanoparticles:
Preparation, properties and possible applications in drug delivery. Current Drug Delivery 1:321–333.
34. Wang, H., Liu, K., Chen, K.J., Lu, Y., Wang, S., Lin, W.Y., Guo, F., Kamei, K., Chen, Y.C., Ohashi, M.,
Wang, M., Garcia, M.A., Zhao, X.Z., Shen, C.K., and Tseng, H.R. (2010). A rapid pathway toward a
superb gene delivery system: Programming structural and functional diversity into a supramolecular
nanoparticle library. ACS Nano 4:6235–6243.
35. Zamani, M., Prabhakaran, M.P., and Ramakrishna, S. (2013). Advances in drug delivery via electro-
spun and electrosprayed nanomaterials. International Journal of Nanomedicine 8:2997–3017.
36. Folkman, J., Long, D.M. Jr., and Rosenbaum, R. (1966). Silicone rubber: A new diffusion property
useful for general anesthesia. Science 154:148–149.
37. Cullins, V.E. (1992). Injectable and implantable contraceptives. Current Opinion in Obstetrics and
Gynecology 4:536–543.
38. Zaffaroni, A. (1981). Systems for controlled drug delivery. Medical Research Review 1:373–386.
39. Pollack, I.P., Quigley, H.A., and Harbin, T.S. (1976). The Ocusert pilocarpine system: Advantages and
disadvantages. The Southern Medical Journal 69:1296–1298.
40. Apfel, C.C., Zhang, K., George, E., Shi, S., Jalota, L., Hornuss, C., Fero, K.E., Heidrich, F., Pergolizzi,
J.V., Cakmakkaya, O.S., and Kranke, P. (2010). Transdermal scopolamine for the prevention of post-
operative nausea and vomiting: A systematic review and meta‐analysis. Clinical Therapeutics
32:1987–2002.
Chitosan‐Based Drug Delivery Systems 279
41. Sareen, R., Jain, N., and Kumar, D. (2012). An insight to osmotic drug delivery. Current Drug Delivery
9:285–296.
42. Moodley, K., Pillay, V., Choonara, Y.E., du Toit, L.C., Ndesendo, V.M., Kumar, P., Cooppan, S., and
Bawa, P. (2012). Oral drug delivery systems comprising altered geometric configurations for con-
trolled drug delivery. International Journal of Molecular Sciences 13:18–43.
43. Brem, H. and Gabikian, P. (2001). Biodegradable polymer implants to treat brain tumors. Journal of
Controlled Release 74:63–67.
44. Okada, H. and Toguchi, H. (1995). Biodegradable microspheres in drug delivery. Critical Reviews in
Therapeutic Drug Carrier Systems 12:1–99.
45. (2012). Second‐generation long‐acting injectable antipsychotic agents: An overview. Drug and
Therapeutics Bulletin 50:102–105.
46. De Leede, L.G., Humphries, J.E., Bechet, A.C., Van Hoogdalem E.J., Verrijk R., and Spencer D.G. (2008).
Novel controlled‐release Lemna‐derived IFN‐alpha2b (Locteron): Pharmacokinetics, pharmacodynamics,
and tolerability in a phase I clinical trial. Journal of Interferon and Cytokine Research 28:113–122.
47. Oerlemans, C., Bult, W., Bos, M., Storm, G., Nijsen, J.F., and Hennink, W.E. (2010). Polymeric
micelles in anticancer therapy: Targeting, imaging and triggered release. Pharmaceutical Research
27:2569–2589.
48. Conley, S.J., Baker, T.L., Burnett, J.P., Theisen, R.L., Lazarus, D., Peters, C.G., Clouthier, S.G.,
Eliasof, S., and Wicha, M.S. (2015). CRLX101, an investigational camptothecin‐containing nanopar-
ticle‐drug conjugate, targets cancer stem cells and impedes resistance to antiangiogenic therapy in
mouse models of breast cancer. Breast Cancer Research and Treatment 150:559–567.
49. Weiss, G.J., Chao, J., Neidhart, J.D. Ramanathan, R.K., Bassett, D., Neidhart, J.A., Choi, C.H.J.,
Chow, W., Chung, V., Forman, S.J., Garmey, E., Hwang, J., Kalinoski, D.L., Koczywas, M., Longmate,
J., Melton, R.J., Morgan, R., Oliver, J., Peterkin, J.J., Ryan, J.L., Schluep, T., Synold, T.W., Twardowski,
P., Davis, M.E., and Yen, Y. (2013). First‐in‐human phase 1/2a trial of CRLX101, a cyclodextrin‐con-
taining polymer‐camptothecin nanopharmaceutical in patients with advanced solid tumor malignan-
cies. Investigational New Drugs 31:986–1000.
50. Gaur, S., Wang, Y., Kretzner, L., Chen, L., Yen, T., Wu, X., Yuan, Y.C., Davis, M., and Yen, Y. (2014).
Pharmacodynamic and pharmacogenomic study of the nanoparticle conjugate of camptothecin
CRLX101 for the treatment of cancer. Nanomedicine 10:1477–1486.
51. Sanna, V., Pala, N., and Sechi, M. (2014). Targeted therapy using nanotechnology: Focus on cancer.
International Journal of Nanomedicine 9:467–483.
52. Davis, M.E. (2009). The first targeted delivery of siRNA in humans via a self‐assembling, cyclodextrin
polymer‐based nanoparticle: From concept to clinic. Molecular Pharmaceutics 6:659–668.
53. Ilyinskii, P.O., Roy, C.J., O’Neil, C.P., Browning, E.A., Pittet, L.A., Altreuter, D.H., Alexis, F., Tonti,
E., Shi, J., Basto, P.A., Iannacone, M., Radovic‐Moreno, A.F., Langer, R.S., Farokhzad, O.C., von
Andrian, U.H., Johnston, L.P., and Kishimoto, T.K. (2014). Adjuvant‐carrying synthetic vaccine parti-
cles augment the immune response to encapsulated antigen and exhibit strong local immune activation
without inducing systemic cytokine release. Vaccine 32:2882–2895.
54. Desai, R.J. and Bergman, J. (2015). Effects of the nanoparticle‐based vaccine, SEL‐068, on nicotine
discrimination in squirrel monkeys. Neuropsychopharmacology 40:2207–2216.
55. Reddy, L.H. and Couvreur, P. (2011). Nanotechnology for therapy and imaging of liver diseases.
Journal of Hepatology 55:1461–1466.
56. Patel, M.P., Patel, R.R., and Patel, J.K. (2010). Chitosan mediated drug delivery system: A review.
Journal of Pharmacy and Pharmaceutical Sciences 13(3):536–557.
57. Hu, L., Sun, Y., and Wu, Y. (2013). Advances in chitosan‐based drug delivery vehicles. Nanoscale
5(8):3103–3111.
58. Elgadir, M.A., Uddin, M.S., Ferdosh, S., Adam, A., Chowdhury, A.J.K., and Sarker, M.Z.I. (2015).
Impact of chitosan composites and chitosan nanoparticle composites on various drug delivery s ystems:
A review. Journal of Food and Drug Analysis 23(4):619–629.
280 Chitin and Chitosan: Properties and Applications
59. Saikia, C., Gogoi, P., and Maji, T.K. (2015). Chitosan: A promising biopolymer in drug delivery appli-
cations. Journal of Molecular and Genetic Medicine S4:6–10.
60. Wu, W., Wu, Z., Yu, T., Jiang C., Kim W.‐S. (2015). Recent progress on magnetic iron oxide nanopar-
ticles: Synthesis, surface functional strategies and biomedical applications. Science and Technology of
Advanced Materials 16(2):023501–023543.
61. Wang, J., Wang, L., Yu, H., Abdin, Z., Chen, Y., Chen, Q., Zhou, W., Zhang, H., and Chen, X. (2016).
Recent progress on synthesis, property and application of modified chitosan: An overview. International
Journal of Biological Macromolecules 88(7):333–344.
62. Usman, A., Zia, K.M., Zuber, M., Tabasum, S., Rehman, S., and Zia, F. (2016). Chitin and chitosan‐
based polyurethanes: A review of recent advances and prospective biomedical applications.
International Journal of Biological Macromolecules 85:630–645.
63. Lv, D., Hu, Z., Lu, L., Lu, H., and Xu, X. (2017). Three‐dimensional cell culture: A powerful tool in
tumor research and drug discovery (Review). Oncology Letters 14:6999–7010.
64. Hussein‐Al‐Ali, S.H., Kura, A., Hussein, M.Z., and Fakurazi, S. (2018). Preparation of chitosan nano-
particles as a drug delivery system for perindopril erbumine. Polymer Composites 39(2):544–552.
65. Wedmore, I., McManus, J.G., Pusateri, A.E., and Holcomb, J.B. (2006). A special report on the chi-
tosan‐based hemostatic dressing: Experience in current combat operations. The Journal of Trauma:
Injury, Infection, and Critical Care 60(3):655–658.
66. Funkhouser, J.D. and Aronson, N.N., Jr. (2007). Chitinase family GH18: Evolutionary insights from
the genomic history of a diverse protein family. BMC Evolutionary Biology 7: 96–112.
67. Luo, Y. and Wang, Q. (2014). Recent development of chitosan‐based polyelectrolyte complexes with
natural polysaccharides for drug delivery. International Journal of Biological Macromolecules
64:353–367.
68. Ngo, D.‐H., Vo, T.‐S., Ngo, D.‐N., Kang, K.‐H., Je, J.‐Y., Pham, H.N.‐D., Byun, H.‐G., and Kim, S.‐K.
(2015). Biological effects of chitosan and its derivatives. Food Hydrocolloids 51:200–216.
69. Bhise, K., Dhumal, R., Paradkar, A., and Kadam, S. (2008). Effect of drying methods on swelling,
erosion and drug release from chitosan‐naproxen sodium complexes. AAPS PharmSciTech 9:1–12.
70. Sun, W., Mao, S., Wang, Y., Junyaprasert, V.B., Zhang, T., Na, L., and Wang, J. (2010). Bioadhesion
and oral absorption of enoxaparin nanocomplexes. International Journal of Pharmaceutics
386:275–281.
71. Li, L. Wang L., Li, J., Jiang, S., Wang, Y., Zhang, X., Jiaojiao Ding, J., Yu T., Mao, S. (2014). Insights
into the mechanisms of chitosan‐anionic polymers‐based matrix tablets for extended drug release.
International Journal of Pharmaceutics 476(1–2):253–265.
72. Shavi, G., Nayak, U., Reddy, M., Karthik, A., Deshpande, P., Kumar, A., and Udupa, N. (2011).
Sustained release optimized formulation of anastrozole‐loaded chitosan microspheres: In vitro and in
vivo evaluation. Journal of Materials Science: Materials in Medicine 22:865–878.
73. Sandri, G., Rossi, S., Bonferoni, M.C., Ferrari, F., Mori, M., and Caramella C. (2012). The role of
chitosan as a mucoadhesive agent in mucosal drug delivery. Journal of Drug Delivery Science and
Technology 22(4):275–284.
74. Grabovac, V., Guggi, D., and Bernkop‐Schnürch, A. (2005). Comparison of the mucoadhesive proper-
ties of various polymers. Advanced Drug Delivery Reviews 57:1713–1723.
75. Jintapattanakit, A., Junyaprasert, V.B., and Kissel, T. (2009). The role of mucoadhesion of trimethyl
chitosan and PEGylated trimethyl chitosan nanocomplexes in insulin uptake. Journal of Pharmaceutical
Science 98:4818–4830.
76. Werle, M. and Bernkop‐Schnürch, A. (2008). Thiolated chitosans: Useful excipients for oral drug
delivery. Journal Pharmacy and Pharmacology 60:273–281.
77. Schattling, P., Taipaleenmäki, E., Zhang, Y., and Städler, B. (2017). A polymer chemistry point of view
on mucoadhesion and mucopenetration. Macromolecular Bioscience. 17(9):1700060–1700080.
78. Moreno, J.A.S., Mendes, A.C., Stephansen, K., Engwer, C., Goycoolea, F.M., Boisen, A., Nielsen,
L.H., and Chronakis I.S. (2018). Development of electrosprayed mucoadhesive chitosan microparti-
cles. Carbohydrate Polymers 190:240–247.
Chitosan‐Based Drug Delivery Systems 281
79. Yin, H., Kanasty, R.L., Eltoukhy, A.A., Vegas, A.J., Dorkin, J.R., and Anderson, D.G. (2014). Non‐
viral vectors for gene‐based therapy. Nature Reviews. Genetics 15:541–555.
80. Fernández Fernández, E., Santos‐Carballal, B., Weber, W.M., and Goycoolea, F.M. (2016). Chitosan
as a non‐viral co‐transfection system in a cystic fibrosis cell line. International Journal of Pharmaceutics
502(102):1–9.
81. Layek, B. and Singh, J. (2017). Chitosan for DNA and gene therapy. In: Chitosan Based Biomaterials
(eds. Jennings, J.A. and Bumgardner, J.D.), Vol. 2.
82. Cupino, T.L., Watson B.A., Cupino, A.C., Oda, K., Ghamsary, M.G., Soriano, S., and Kirsch, W.M.
(2018). Stability and bioactivity of chitosan as a transfection agent in primary human cell cultures: A
case for chitosan‐only controls. Carbohydrate Polymers 180:376–384.
83. Mao, S., Sun, W., and Kissel, T. (2010). Chitosan‐based formulations for delivery of DNA and siRNA.
Advanced Drug Delivery Reviews 62:12–27.
84. Malmo, J., Vrum, K.M., and Strand, S.P. (2011). Effect of chitosan chain architecture on gene delivery:
Comparison of self‐branched and linear chitosans. Biomacromolecules 12:721–729.
85. Třmmeraas, K., Strand, S.P., Christensen, B.E., Smidsrřd, O., and Vrum, K.M. (2011). Preparation and
characterization of branched chitosans. Carbohydrate Polymers 83:1558–1564.
86. Varkouhi, A.K., Verheul, R.J., Schiffelers, R.M., Lammers, T., Storm, G., and Hennink, W.E. (2010).
Gene silencing activity of siRNA polyplexes based on thiolated N,N,N‐trimethylated chitosan.
Bioconjugate Chemistry 21:2339–2346.
87. Oliveira, A.V. Silva, A.P., Bitoque, D.B., Silva, G.A., and Rosa da Cost, A.M. (2013). Transfection
efficiency of chitosan and thiolated chitosan in retinal pigment epithelium cells: A comparative study.
Journal of Pharmaceutical and Biological Sciences 5: 111–118.
88. Malhotra, M., Lane, C., Tomaro‐Duchesneau, C., Saha, S., and Prakash, S. (2011). A novel method for
synthesizing PEGylated chitosan nanoparticles: Strategy, preparation, and in vitro analysis.
International Journal of Nanomedicine 6:485–494.
89. Yu, H., Chen, X., Lu, T., Sun, J., Tian, H., Hu, J., Wang, Y., Zhang, P., and Jing, X. (2007). Poly(L‐
lysine)‐graft‐chitosan copolymers: Synthesis, characterization, and gene transfection effect.
Biomacromolecules 8:1425–1435.
90. Teijeiro‐Osorio, D., Remuńán‐López, C., and Alonso, M.J. (2009) Chitosan/cyclodextrin nanoparti-
cles can efficiently transfect the airway epithelium in vitro. European Journal of Pharmaceutics and
Biopharmaceutics 71:257–263.
91. Főger F., Malaivijitnond S., Wannaprasert T., Huck C., Bernkop‐Schnűrch A., and Werle M. (2008).
Effect of oral paclitaxel in presence of a thiolated polymer on absorption and tumor growth in rats.
Journal of Drug Targeting 16(2):149–155.
92. Mandracchia, D., Trapani, A., Tripodo, G., Perrone, M.G., Giammona, G., Trapani, G., and Colabufo
N.A. (2017). In vitro evaluation of glycol chitosan based formulations as oral delivery systems for
efflux pump inhibition. Carbohydrate Polymers 166:73–82.
93. Palmberger T.F., Hombach J., and Bernkop‐Schnűrch A. (2008). Thiolated chitosan: Development and
in vitro evaluation of an oral delivery system for acyclovir. International Journal of Pharmaceutics
348(1–2):79–85.
94. Werle M. and Hoffer M. (2006) Glutathione and thiolated chitosan inhibit multidrug resistance P‐
glycoprotein activity in excised small intestine. Journal of Controlled Release 111(1–2):41–46.
95. Rosenthal, R., Gűnzel, D., Finger, C., Krug, S.M., Richter, J.F., Schulzke, J.‐D., Fromma, M., and
Amasheh, S. (2012). The effect of chitosan on transcellular and paracellular mechanisms in the intes-
tinal epithelial barrier. Biomaterials 33:2791–2800.
96. Benediktsdóttir, B.E., Baldursson, O., and Másson M. (2014). Challenges in evaluation of chitosan and
trimethylated chitosan (TMC) as mucosal permeation enhancers: From synthesis to in vitro applica-
tion. Journal of Controlled Release 173:18–31.
97. Shah, P., Jogani, V., Mishra, P., Mishra, A.K., Bagchi, T., and Misra, A. (2007). Modulation of ganci-
clovir intestinal absorption in presence of absorption enhancers. Journal of Pharmaceutical Science
96:2710–2722.
282 Chitin and Chitosan: Properties and Applications
98. Langoth, N., Kahlbacher, H., Schöffmann, G., Schmerold, I., Schuh, M., Franz, S., Kurka, P., and
Bernkop‐Schnürch, A. (2006). Thiolated chitosans: Design and in vivo evaluation of a mucoadhesive
buccal peptide drug delivery system. Pharmaceutical Research 23:573–579.
99. Na, L., Wang, J., Wang, L., and Mao, S. (2013). A novel permeation enhancer: N‐succinyl chitosan
on the intranasal absorption of isosorbide dinitrate in rats. European Journal of Pharmaceutical
Sciences 48:301–306.
100. Trapani, A., Lopedota, A., Franco, M., Cioffi N., Ieva, E., Garcia‐Fuentes, M., and Alonso, M.J.
(2010). A comparative study of chitosan and chitosan/cyclodextrin nanoparticles as potential carriers
for the oral delivery of small peptides. European Journal of Pharmaceutics and Biopharmaceutics
75(1):26–32.
101. du Toit, T., Malan, M.M., Lemmer, H.J.R., Chrisna Gouws, C., Aucamp, M.E., Breytenbach, W.J.,
and Hamman, J.H. (2016). Combining chemical permeation enhancers for synergistic effects.
European Journal of Drug Metabolism and Pharmacokinetics 41:575–586.
102. Alonso, M.J. and Csaba, N.S. (eds.) (2012) Nanostructured Biomaterials for Overcoming Biological
Barriers, RSC Publishing, Cambridge.
103. Lademann, J.M., Patzelt, A., Richter, H., Antoniou, C., Sterry, W., and Knorr, F. (2009) Determination
of the cuticula thickness of human and porcine hairs and their potential influence on the penetration
of nanoparticles into the hair follicles., Journal of Biomedical Optics 14(2): 021014, https://doi.
org/10.1117/1.3078813.
104. Prow, T.W., Grice, J.E., Lin, L.L., Faye, R., Butler, M., Becker, W., Wurm, E.M.T., Yoong, C.,
Robertson, T.A., Soyer, H.P., and Roberts, M.S. (2011). Nanoparticles and microparticles for skin
drug delivery. Advanced Drug Delivery Review 63:470–491.
105. Hafner, A., Lovric, J., Pepic, I., and Filipovic‐Grcic, J. (2011). Lecithin/chitosan nanoparticles for
transdermal delivery of melatonin. Journal of Microencapsulation 28(8):807–815.
106. Smith, J., Wood, E., and Dornish, M. (2004). Effect of chitosan on epithelial cell tight junctions.
Pharmaceutical Research 21:43–49.
107. Sapra, B., Jain, S., and Tiwary, A.K. (2009). Effect of Asparagus racemosus extract on transdermal
delivery of carvedilol: A mechanistic study. AAPS PharmSciTech 10:199–210.
108. Choi, W.I., Lee, J.H., Kim, J.Y., Kim, J.C., Kim, Y.H., and Tae, G. (2012). Efficient skin permeation
of soluble proteins via flexible and functional nano‐carrier. Journal Control Release
157(2):272–278.
109. Khalil, S.K., El‐Feky, G.S., El‐Banna, S.T., and Khalil, W.A. (2012). Preparation and evaluation of
warfarin‐β‐cyclodextrin loaded chitosan nanoparticles for transdermal delivery. Carbohydrate
Polymers 90(3):1244–1253.
110. Peptu, C., Rotaru, R., Ignat, L., Humelnicu, A.C., Harabagiu, V., Peptu, C.A., Leon, M.M., Mitu, F.,
Cojocaru, E., Boca, A., and Tamba, B.I. (2015). Nanotechnology approaches for pain therapy through
transdermal drug delivery. Current Pharmaceutical Design 21(42):6125–6139.
111. Yu, K., Wang, Y., Wan, T., Zhai, Y., Cao, S., Ruan, W., Wu, C., and Xu, Y. (2018). Tacrolimus nano-
particles based on chitosan combined with nicotinamide: Enhancing percutaneous delivery and treat-
ment efficacy for atopic dermatitis and reducing dose. International Journal of Nanomedicine
13:129–142.
112. Abdel‐Hafez, S.M., Hathout, R.M., and Sammour, O.A. (2018). Tracking the transdermal penetration
pathways of optimized curcumin‐loaded chitosan nanoparticles via confocal laser scanning micros-
copy. International Journal of Biological Macromolecules 108:753–764.
113. Zhang, X., Zhang, H., Wu, Z., Wang, Z., Niu, H., and Li, C. (2008). Nasal absorption enhancement
of insulin using PEG‐grafted chitosan nanoparticles. European Journal of Pharmaceutics and
Biopharmaceutics 68(3):526–534.
114. Csaba, N., Garcia‐Fuentes, M., and Alonso, M.J. (2009). Nanoparticles for nasal vaccination.
Advanced Drug Delivery Reviews 61:140–157.
Chitosan‐Based Drug Delivery Systems 283
115. Sayin, B., Somavarapu. S., Li. X.W., Thanov. M., Sesardic. D., Alpar, H.O., Senel, S. (2008).
Mono‐N‐carboxymethyl chitosan (MCC) and N‐trimethyl‐chitosan (TMC) nanoparticles for non‐
invasive vaccine delivery. International Journal of Pharmaceutics 340(1–2):139–140.
116. Ahmed, M., Jiao, H., Domingo‐Gonzalez, R., Das, S., Griffiths, K.L., Rangel‐Moreno, J., Nagarajan,
U.M., and Khader, S.A. (2017). Rationalized design of a mucosal vaccine protects against
Mycobacterium tuberculosis challenge in mice. Journal of Leukocyte Biology 101:1373–1381.
117. Rekha, M.R. and Sharma, C.P. (2009). Synthesis and evaluation of lauryl succinyl chitosan particles
towards oral insulin delivery and absorption. Journal Control Release 135(2):144–151.
118. Fabiano, A., Piras, A.M., Uccello‐Barretta, G., Balzano, F., Cesari A., Testai, L., Citi, V., and Zambito,
Y. (2018). Impact of mucoadhesive polymeric nanoparticulate systems on oral bioavailability of a
macromolecular model drug. European Journal of Pharmaceutics and Biopharmaceutics
130:281–289.
119. Meng, J., Sturgis, T.F., and Yovan, B.B.C. (2011). Engineering tenofovir loaded chitosan nanoparti-
cles to maximize microbicide mucoadhesive. European Journal of Pharmaceutics and
Biopharmaceutics 44(1–2):57–67.
120. Sekar, V., Rajendran, K., Vallinayagam, S., Deepak, V., and Mahadevan, S. (2018). Synthesis and
characterization of chitosan ascorbate nanoparticles for therapeutic inhibition for cervical cancer and
their in silico modeling. Journal of Industrial and Engineering Chemistry 115:185–195.
121. Lupo, N., Fodor, B., Muhammad, I., Yaqoob, M., Matuszczak, B., and Bernkop‐Schnürch, A. (2017).
Entirely S‐protected chitosan: A promising mucoadhesive excipient for metronidazole vaginal tab-
lets. Acta Biomaterialia 64:106–115.
122. Patel, A., Cholkar, K., Agrahari, V., and Mitra, A. K. (2013). Ocular drug delivery systems: An over-
view. World Journal of Pharmacology 2(2):47–64.
123. Ranta, V.‐P., Mannermaa, E., Lummepuro, K., Subrizi, A., Laukkanen, A., Antopolsky, M.,
Murtomaki, L., Hornof, M., and Urtti, A. (2010). Barrier analysis of periocular drug delivery to the
posterior segment. Journal of Controlled Release 148(1):42–48.
124. Ying G.S. (ed.) (2012). Age Related Macular Degradation–The Recent Advances in Basic Research
and Clinical Care, InTech Publishers, Shanghai.
125. De Campos, A.M., Diebold, Y., Carvallo, E.L.S., Sanchez, A., and Alonso, M.J. (2004). Chitosan
nanoparticles as new ocular drug delivery systems: In vitro stability, in vivo fate and cellular toxicity.
Pharmaceutical Research 21(5):803–810.
126. Zhang, P., Liu, X., Hu, W., Bai, Y., and Zhang, L. (2016). Preparation and evaluation of naringenin‐
loaded sulfobutylether‐β‐cyclodextrin/chitosan nanoparticles for ocular drug delivery. Carbohydrate
Polymers 149:224–230.
127. Zhu, X., Su, M., Tang, S., Wang, L., Liang, X., Meng, F., Hong, Y., and Xu, Z. (2012). Synthesis of
thiolated chitosan and preparation nanoparticles with sodium alginate for ocular delivery. Molecular
Vision 18:1973–1982.
128. Liu, D., Lian, Y., Fang, Q., Liu L., Zhang, J., and Li J. (2018). Hyaluronic‐acid‐modified lipid‐
polymer hybrid nanoparticles as an efficient ocular delivery platform for moxifloxacin hydrochloride.
International Journal of Biological Macromolecules 116:1026–1036.
129. Peptu, C., Popa, M., Tataru, G., Perichaud, A., and Antimisiaris, S. (2010). Double crosslinked
chitosan‐gelatin particulate systems for controlled drug release. Journal of Bioactive and Compatible
Polymers 25(1):98–116.
130. Andrei, G., Peptu, C.A., Popa, M., Desbrieres, J., Peptu, C., Gardikiotis, F., Costuleanu, M., Costin,
D., Dupin, J.C., Uhart, A., and Tamba, B.I. (2015). Formulation and evaluation of cefuroxim loaded
submicron particles for ophthalmic delivery. International Journal of Pharmaceutics
493(1–2):16–29.
131. Pakl, G. and Palit, P.(2012). Use of liposomes and nanoparticles for brain drug targeting. Journal of
Pharmaceutical and Scientific Innovation 1(4):11–13.
284 Chitin and Chitosan: Properties and Applications
132. Yemisci, M., Caban, S., Gursoy‐Ozdemir, Y., Lule, S., Novoa‐Carballal, R., Riguera, R., Fernandez‐
Megia, E., Andrieux, K., Couvreur, P., Capan, Y., and Dalkara, T. (2015). Systemically administered
brain‐targeted nanoparticles transport peptides across the blood–brain barrier and provide neuropro-
tection. Journal of Cerebral Blood Flow and Metabolism 35:3469–3475.
133. Wiley, D.T., Webster, P., Gale, A., and Davis, M.E. (2013). Avidity‐tuned nanoparticles reach the
brain. Proceedings of the National Academy of Sciences 110 (21):8662–8667.
134. Veiseh, O., Sun, C., Fang, C., Bhattarai, N., Gunn, J., Kievit, F., Du, K., Pullar, B., Lee, D., Ellenboge,
R., Olson, J., and Zhang, M. (2009). Specific targeting of brain tumors with an optical/magnetic reso-
nance imaging nanoprobe across the blood–brain barrier. Cancer Research 69(15):6200–6207.
135. Sahin, A., Yoyen‐Ermis, D., Caban‐Toktas, S., Horzum, U., Aktas, Y., Couvreur, P., Esendagli, G., and
Capan, Y. (2017). Evaluation of brain‐targeted chitosan nanoparticles through blood–brain b arrier
cerebral microvessel endothelial cells. Journal of Microencapsulation, 34(7):659–666.
136. Ali, A. and Ahmed, S. (2018). A review on chitosan and its nanocomposites in drug delivery.
International Journal of Biological Macromolecules 109:273–286.
137. Hennink, W.E. and van Nostrum, C.F. (2012). Novel crosslinking methods to design hydrogels.
Advanced Drug Delivery Reviews 64:223–236.
138. Assad, E., Maire, M., and Lerouge, S. (2015). Injectable thermosensitive chitosan hydrogels with con-
trolled gelation kinetics and enhanced mechanical resistance. Carbohydrate Polymers 130:87–96.
139. Zhou, H.Y, Jiang, L.J., Cao, P.P., Li, J.B., and Ken, X.G. (2015). Glycerophosphate‐based c hitosan
thermosensitive hydrogels and their biomedical applications. Carbohydrate Polymers
117:524–536.
140. Huang, C.‐L, Chen, Y.‐B, Lo, Y.‐L, and Lin Y.H. (2016). Development of chitosan/β‐glycerophosphate/
glycerol hydrogel as a thermosensitive coupling agent. Carbohydrate Polymers 147:409–414.
141. Liu, L., Gao, Q., Lu, X., and Zhou, H. (2016). In situ forming hydrogels based on chitosan for drug
delivery and tissue regeneration. Asian Journal of Pharmaceutical Sciences 11:673–683.
142. Deng, A., Kang, X., Zhang, J., Yang, Y., Yang, S. (2017). Enhanced gelation of chitosan/β‐sodium
glycerophosphate thermosensitive hydrogel with sodium bicarbonate and biocompatibility evaluated.
Materials Science and Engineering 78:1147–1154.
143. Sakloetsakun, D., Hombach, J., and Bernkop‐Schnürch, A. (2009). In situ gelling properties of chitosan‐
thioglycolic acid conjugate in the presence of oxidizing agents. Biomaterials 30:6151–6157.
144. Ailincai, D., Tartau‐Mititelu, L., and Marin, L. (2018). Drug delivery systems based on biocompatible
imino‐chitosan hydrogels for local anticancer therapy. Drug Delivery 25:1080–1090.
145. Olaru, A.‐M., Marin, L., Morariu, S., Pricope, G., Pinteala, M., and Tartau‐Mititelu L. (2018).
Biocompatible chitosan based hydrogels for potential application in local tumor therapy. Carbohydrate
Polymers 179:59–70.
146. Zhang D, Sun P, Li P, Xue, A., Zhang, X., Zhang, H., and Jin, X. (2013). A magnetic chitosan hydro-
gel for sustained and prolonged delivery of Bacillus Calmette‐Guérin in the treatment of bladder
cancer. Biomaterials 34:10258–10266.
147. Supper, S., Anton, N., Boisclair, J., Seidel N, Riemenschnitter, M., Curdy, C., and Vandamme, T.
(2014). Chitosan/glucose 1‐phosphate as new stable in situ forming depot system for controlled drug
delivery. European Journal of Pharmaceutics and Biopharmaceutics 88:361–373.
148. Jiang, Y., Meng, X., Wu, Z., and Qi, X. (2016). Modified chitosan thermosensitive hydrogel enables
sustained and efficient anti‐tumor therapy via intratumoral injection. Carbohydrate Polymers
144:245–253.
149. Wang, T., Khen, L., Shen, T., and Wu, D. (2016). Preparation and properties of a novel thermo‐
sensitive hydrogel based on chitosan/hydroxypropyl methylcellulose/glycerol. International Journal
of Biological Macromolecules 93:775–782.
150. Constantin, M., Bucatariu, S.‐M., Doroftei, F., and Fundueanu, G., (2017). Smart materials based on
chitosan microspheres embedded in thermosensitive hydrogel for controlled delivery of drugs.
Carbohydrate Polymers 157:493–502.
Chitosan‐Based Drug Delivery Systems 285
151. Cheng, N.‐C., Lin, Q.‐J., Ling, T.Y., and Young, T.‐H. (2017). Sustained release of adipose‐derived
stem cells by thermosensitive chitosan/gelatin hydrogel for therapeutic angiogenesis. Acta
Biomaterialia 51:258–267.
152. Abou Taleb, M.F., Alkahtani, A., and Mohamed, S.K. (2015). Radiation synthesis and characteriza-
tion of sodium alginate/chitosan/hydroxyapatite nanocomposite hydrogels: A drug delivery system
for liver cancer. Polymer Bulletin 72(4):725–742.
153. Ren, Y., Zhao, X., Liang, X., Ma, P.X., and Guo, B. (2017). Injectable hydrogel based on quaternized
chitosan, gelatin and dopamine as localized drug delivery system to treat Parkinson’s disease.
International Journal of Biological Macromolecules 105:1079–1087.
154. Shao, Y., Li, L., Gu, X., Wang, L., and Mao, S. (2015). Evaluation of chitosan‐anionic polymers based
tablets for extended‐release of highly water‐soluble drugs. Asian Journal of Pharmaceutical Sciences
10(1):24–30.
155. Xu, J., Strandman, S., Zhu, J.X.X., Barralet, J., and Cerruti, M. (2015). Genipin‐crosslinked catechol‐
chitosan mucoadhesive hydrogels for buccal drug delivery. Biomaterials 37:395–404.
156. Kono, H. and Teshirogi, T. (2015). Cyclodextrin‐grafted chitosan hydrogels for controlled drug deliv-
ery. International Journal of Biological Macromolecules 72:299–308.
157. Xu, J., Tam, M., Samaei, S., Lerouge, S., Barralet, J., Stevenson, M.M., and Cerruti, M. (2017).
Mucoadhesive chitosan hydrogels as rectal drug delivery vessels to treat ulcerative colitis. Acta
Biomaterialia 48:247–257.
158. Cheng, Y.H., Tsai, T.H., Jhan, Y.Y., Chiu, A.W.H., Tsai, K.L., Chien, C.S., Chiou, S.H., and Liu,
C.J.L. (2016). Thermosensitive chitosan‐based hydrogel as a topical ocular drug delivery system of
latanoprost for glaucoma treatment. Carbohydrate Polymers 144:390–399.
159. Tang, Q., Luo, C., Lu, B., Fu, Q., Yin, H., Qin, Z., Lyu, D., Zhang, L., Fang, Z., Zhu, Y., and Yao, K.
(2017). Thermosensitive chitosan‐based hydrogels releasing stromal cell derived factor‐1 alpha
recruit MSC for corneal epithelium regeneration. Acta Biomaterialia 61:101–113.
160. Lajud, S.A., Nagda, D.A., Qiao, P., Tanaka, N., Civantos, A., Gu, R., Cheng, Z., Tsourkas, A.,
O’Malley, B.W., and Li, D. (2015). A novel chitosan‐hydrogel‐based nanoparticle delivery system for
local inner ear application. Otology & Neurotology 36(2):341–347.
161. Chen, H., Xing, X., Tan, H., Jia, Y., Zhou, T., Chen, Y., Ling, Z., and Hu, X. (2017). Covalently anti-
bacterial alginate‐chitosan hydrogel dressing integrated gelatin microspheres containing tetracycline
hydrochloride for wound healing. Materials Science and Engineering: C 70(1):287–295.
162. Yadollahi, M., Farhoudian, S., and Namazi, H. (2015). One‐pot synthesis of antibacterial chitosan/
silver bio‐nanocomposite hydrogel beads as drug delivery systems. International Journal of Biological
Macromolecules 79:37–43.
163. Yadollahi, M., Farhoudian, S., Barkhordari S., Gholamali, I., Farhadnejad, H., and Motasadizadeh, H.
(2016). Facile synthesis of chitosan/ZnO bio‐nanocomposite hydrogel beads as drug delivery sys-
tems. International Journal of Biological Macromolecules 82:273–278.
164. Pathania, D., Gupta, D., Kothiyal, N.C., Sharma G., Eldesoky G.E., and Naushad, M. (2016).
Preparation of a novel chitosan‐g‐poly(acrylamide)/Zn nanocomposite hydrogel and its applications
for controlled drug delivery of ofloxacin. International Journal of Biological Macromolecules
84:340–348.
165. Zhang, L., Wang, L., Guo, B., and Ma, P.X. (2014). Cytocompatible injectable carboxymethyl
chitosan/N‐isopropylacrylamide hydrogels for localized drug delivery. Carbohydrate Polymers
103:110–118.
166. Gao, J., Liu, R., Wu, J., Liu, Z., Li, J., Zhou, J., Hao, T., Wang, Y., Du, Z., Duan, C., and Wang, C.
(2012). The use of chitosan based hydrogel for enhancing the therapeutic benefits of adipose‐derived
MSCs for acute kidney injury. Biomaterials 33(14):3673–3681.
167. Unsoy, G., Khodadust, R., Yalcin, S., Mutlu, P., Gunduz, U. (2014). Synthesis of Doxorubicin loaded
magnetic chitosan nanoparticles for pH responsive targeted drug delivery. European Journal of
Pharmaceutical Sciences 62:243–250.
286 Chitin and Chitosan: Properties and Applications
168. Wang, T., Hou, J., Su, C., Zhao, L., and Shi, Y. (2017). Hyaluronic acid‐coated chitosan nanoparticles
induce ROS‐mediated tumor cell apoptosis and enhance antitumor efficiency by targeted drug deliv-
ery via CD44. Journal of Nanobiotechnology 15(1):7–18.
169. Khdair, A., Hamad, I., Alkhatib, H., Bustanji, Y., Mohammad, M., Tayem, R., and Aiedeh, K. (2016).
Modified‐chitosan nanoparticles: Novel drug delivery systems improve oral bioavailability of doxo-
rubicin. European Journal of Pharmaceutical Sciences 93:38–44.
170. El‐Feky, G.S., Sharaf, S.S., El Shafei, A., and Hegazy, A.A. (2017). Using chitosan nanoparticles as
drug carriers for the development of a silver sulfadiazine wound dressing. Carbohydrate Polymers
158:11–19.
171. Mahdavinia, G.R., Etemadi, H., and Soleymani, F. (2015). Magnetic/pH‐responsive beads based on
caboxymethyl chitosan and κ‐carrageenan and controlled drug release. Carbohydrate Polymers
128:112–121.
172. Cerchiara, T., Abruzzo, A., Ñahui Palomino, R.A., Vitali, B., De Rose, R., Chidichimo, G., Ceseracciu,
L., Athanassiou, A., Saladini, B., Dalena, F., Bigucci, F., and Luppi, B. (2017). Spanish Broom
(Spartium junceum L.) fibers impregnated with vancomycin‐loaded chitosan nanoparticles as new
antibacterial wound dressing: Preparation, characterization and antibacterial activity. European
Journal of Pharmaceutical Sciences 99:105–112.
173. Morgado, P.I., Miguel, S.P., Correia, I.J., and Aguiar‐Ricardo, A. (2017). Ibuprofen loaded PVA/chi-
tosan membranes: A highly efficient strategy towards an improved skin wound healing. Carbohydrate
Polymers 159:136–145.
174. Siafaka, P.I., Zisi, A.P., Exindari, M.K., Karantas, I.D., and Bikiaris, D.N. (2016). Porous dressings of
modified chitosan with poly(2‐hydroxyethyl acrylate) for topical wound delivery of levofloxacin.
Carbohydrate Polymers 143:90–99.
175. Song, J., Remmers, S.J.A., Shao, J., Kolwijck, E., Walboomers, X.F., Jansen, J.A., Leeuwenburgh,
S.C.G., and Yang F. (2016). Antibacterial effects of electrospun chitosan/poly(ethylene oxide) nanofi-
brous membranes loaded with chlorhexidine and silver. Nanomedicine: Nanotechnology, Biology and
Medicine 12(5):1357–1364.
176. Tang, C., Guan, Y.‐X., Yao, S.‐J., and Zhu, Z.‐Q. (2014). Preparation of ibuprofen‐loaded chitosan
films for oral mucosal drug delivery using supercritical solution impregnation. International Journal
of Pharmaceutics 473(1–2):434–441.
177. Shariatinia, Z. and Zahraee, Z. (2017). Controlled release of metformin from chitosan‐based nano-
composite films containing mesoporous MCM‐41 nanoparticles as novel drug delivery systems.
Journal of Colloid and Interface Science 501:60–76
178. Ding, Y., Shen, S.Z., Sun, H., Sun, K., Liu, F., Qi, Y., and Yan, J. (2015). Design and construction of
polymerized‐chitosan coated Fe3O4 magnetic nanoparticles and its application for hydrophobic drug
delivery. Materials Science and Engineering: C 48:487–498.
179. Lin, J., Li, Y., Li, Y., Wu, H., Yu, F., Zhou, S., Xie, L., Luo, F., Lin, C. and Hou, Z. (2015). Drug/
dye‐Loaded, multifunctional PEG‐chitosan‐iron oxide nanocomposites for methotraxate synergisti-
cally self‐targeted cancer therapy and dual model imaging. ACS Applied Materials & Interfaces
7(22):11908–11920.
180. Erel, G., Kotmakçı, M., Akbaba, H., Sözer Karadağlı, S., and Kantarcı, A.G. (2016).
Nanoencapsulated chitosan nanoparticles in emulsion‐based oral delivery system: In vitro and in
vivo evaluation of insulin loaded formulation. Journal of Drug Delivery Science and Technology
36:161–167.
181. Zu, Y., Zhang, Y., Wang, W., Zhao, X., Han, X., Wang, K., and Ge, Y. (2016). Preparation and in vitro/
in vivo evaluation of resveratrol‐loaded carboxymethyl chitosan nanoparticles. Drug Delivery
23(3):971–981.
182. Garg, T., Rath, G., and Goyal, A.K. (2016). Inhalable chitosan nanoparticles as antitubercular drug
carriers for an effective treatment of tuberculosis. Artificial Cells, Nanomedicine, and Biotechnology
44(3):997–1001.
Chitosan‐Based Drug Delivery Systems 287
183. Zhang, Y., Dong, K., Wang, F., Wang, H., Wang, J., Jiang, Z., and Diao, S. (2018). Three dimensional
macroporous hydroxyapatite/chitosan foam‐supported polymer micelles for enhanced oral delivery
of poorly soluble drugs. Colloids and Surfaces B: Biointerfaces 170:497–504.
184. Song, H., Su, C., Cui, W., Zhu, B., Liu, L., Chen, Z., and Zhao, L. (2013). Folic acid‐chitosan conju-
gated nanoparticles for improving tumor‐targeted drug delivery. BioMed Research International
2013:1–6.
185. Sun, L., Wang, Y., Jiang, T., Zheng, X., Zhang, J., Sun, J., Sun, C., and Wang, S. (2012). Novel chi-
tosan‐functionalized spherical nanosilica matrix as an oral sustained drug delivery system for poorly
water‐soluble drug Carvedilol. ACS Applied Materials & Interfaces 5(1):103–113.
186. Cánepa, C., Imperiale, J.C., Berini, C.A., Lewicki, M., Sosnik, A., and Biglione, M.M. (2017).
Development of a drug delivery system based on chitosan nanoparticles for oral administration of
interferon‐α. Biomacromolecules, 18(10):3302–3309.
187. Katas, H., Raja, M.A.G., and Lam, K.L. (2013). Development of chitosan nanoparticles as a stable
drug delivery system for protein/siRNA. International Journal of Biomaterials 2013:1–9.
188. Prabha, G. and Raj, V. (2016). Preparation and characterization of polymer nanocomposites coated
magnetic nanoparticles for drug delivery applications. Journal of Magnetism and Magnetic Materials
408:26–34.
189. Vasile, B.S., Oprea, O., Voicu, G., Ficai, A., Andronescu, E., Teodorescu, A., and Holban, A. (2014).
Synthesis and characterization of a novel controlled release zinc oxide/gentamicin‐chitosan composite
with potential applications in wound care. International Journal of Pharmaceutics 463(2):
161–169.
190. Chen, C., Yao, W., Sun, W., Guo, T., Lv, H., Wang, X., Ying, H., Wang, Y., and Wang, P. (2018). A
self‐targeting and controllable drug delivery system by fabricating with multi‐stimuli responsive chi-
tosan‐based thin film layer on mesoporous silica nanoparticles. International Journal of Biological
Macromolecules. https://doi.org/10.1016/j.ijbiomac.2018.09.058
191. Oliveira, P.M., Matos, B.N., Pereira, P.A.T., Gratieri, T., Faccioli, L.H., Cunha‐Filho, M.S.S., and
Gelfuso, G.M. (2017). Microparticles prepared with 50–190 kDa chitosan as promising nontoxic car-
riers for pulmonary delivery of isoniazid. Carbohydrate Polymers 174:421–431.
192. Irani, M., Sadeghi, G.M.M., and Haririan, I. (2017). A novel biocompatible drug delivery system of
chitosan/temozolomide nanoparticles loaded PCL‐PU nanofibers for sustained delivery of temozolo-
mide. International Journal of Biological Macromolecules 97:744–751
193. Hadjianfar, M., Semnani, D., and Varshosaz, J. (2018). Polycaprolactone/chitosan blend nanofibers
loaded by 5‐fluorouracil: An approach to anticancer drug delivery system. Polymers Advanced
Technologies 2018:1–10.
194. Sedghi, R., Shaabani, A., Mohammadi, Z., Samadi, F.Y., and Isaei, E. (2017). Biocompatible electro-
spinning chitosan nanofibers: A novel delivery system with superior local cancer therapy.
Carbohydrate Polymers 159:1–10.
195. Mendes, A.C., Shekarforoush, E., Engwer, C., Beeren, S.R., Gorzelanny, C., Goycoolea, F.M., and
Chronakis, I.S. (2016). Co‐assembly of chitosan and phospholipids into hybrid hydrogels. Pure and
Applied Chemistry 88(9):905–916.
196. Shekarforoush, E., Ajalloueian, F., Zeng, G., Mendes, A.C., and Chronakis, I.S. (2018). Electrospun
xanthan gum‐chitosan nanofibers as delivery carrier of hydrophobic bioactives. Materials Letters
228:322–326.
197. Ardeshirzadeh, B., Anaraki, N.A., Irani, M., Rad, L.R., and Shamshiri, S. (2015). Controlled release
of doxorubicin from electrospun PEO/chitosan/graphene oxide nanocomposite nanofibrous scaf-
folds. Materials Science and Engineering C 48:384–390
198. Karri, V.V.S.R., Kuppusamy, G., Talluri, S.V., Mannemala, S.S., Kollipara, R., Wadhwani, A.D.,
Mulukutla, S., Raju, K.R.S., and Malayandi, R. (2016). Curcumin loaded chitosan nanoparticles
impregnated into collagen‐alginate scaffolds for diabetic wound healing. International Journal of
Biological Macromolecules 93(B):1519–1529.
288 Chitin and Chitosan: Properties and Applications
199. Li, H., Ji, Q., Chen, X., Sun, Y., Xu, Q., Deng, P., Hu, F., and Yang, J. (2016). Accelerated bony defect
healing based on chitosan thermosensitive hydrogel scaffolds embedded with chitosan nanoparticles
for the delivery of BMP2 plasmid DNA. Journal of Biomedical Materials Research Part A
105(1):265–273.
200. Saber‐Samandari, S. and Saber‐Samandari, S. (2017). Biocompatible nanocomposite scaffolds based
on copolymer‐grafted chitosan for bone tissue engineering with drug delivery capability. Materials
Science and Engineering: C 75:721–732.
201. Zhang, Y.‐G., Zhu, Y.‐J., Chen, F., Sun, T.‐W. (2017). A novel composite scaffold comprising ultra-
long hydroxyapatite microtubes and chitosan: Preparation and application in drug delivery. Journal
of Materials Chemistry B 5(21):3898–3906.
202. Fan, M., Ma, Y., Tan, H., Jia, Y., Zou, S., Guo, S., Zhao, M., Huang, H., Ling, Z., Chen, Y., Hu, X.
(2017). Covalent and injectable chitosan‐chondroitin sulfate hydrogels embedded with chitosan
microspheres for drug delivery and tissue engineering, Materials Science and Engineering: C
71:67–74.
203. Poornima, B. and Korrapati, P.S. (2017). Fabrication of chitosan‐polycaprolactone composite nanofi-
brous scaffold for simultaneous delivery of ferulic acid and resveratrol. Carbohydrate Polymers
157:1741–1749.
204. Paredes, M., Pulcinelli, S.H., Peniche, C., Gonçalves, V., and Santilli, C.V. (2014). Chitosan/(ureasil‐
PEO hybrid) blend for drug delivery. Journal of Sol‐Gel Science and Technology 72(2):233–238.
205. Can, A., Erdal, M., Güngör, S., and Özsoy, Y. (2013). Optimization and characterization of chitosan
films for transdermal delivery of ondansetron. Molecules 18(5):5455–5471.
206. Anjum, S., Arora, A., Alam, M.S., and Gupta B. (2016). Development of antimicrobial and scar preven-
tive chitosan hydrogel wound dressings. International Journal of Pharmaceutics 508(1–2):92–101.
207. Wu, H., Xu, Y., Liu, G., Ling, J., Dash, B.C., Ruan, J., and Zang, C. (2014). Emulsion crosslinked
chitosan/nanohydroxyapatite microspheres for controlled release of alendronate. Journal of Material
Science. Materials in Medicine 25(12):2649–2658.
208. Battaglia, L., D’Addino, I., Peira, E., Trotta, M., and Gallarate, M. (2012). Solid lipid nanoparticles
prepared by coacervation method as vehicles for ocular cyclosporine. Journal of Drug Delivery
Science and Technology 22(2):125–130.
209. Sheikholeslami, Z.S., Salimi‐Kenari, H., Imani, M., Atai, M., and Nodehi, A. (2017). Exploring the
effect of formulation parameters on the particle size of carboxymethyl chitosan nanoparticles pre-
pared via reverse micellar crosslinking. Journal of Microencapsulation 34(3):270–279.
210. Lai, P., Daear, W., Löbenberg, R., and Prenner, E.J. (2014). Overview of the preparation of organic
polymeric nanoparticles for drug delivery based on gelatine, chitosan, poly(d,l‐lactide‐co‐glycolic
acid) and polyalkylcyanoacrylate. Colloids and Surfaces B: Biointerfaces 118:154–163.
211. Pillay, V., Dott, C., Choonara, Y.E., Tyagi, C., Tomar, L., Kumar, P., du Toit, L.C., and Ndesendo,
V.M.K. (2013). A review of the effect of processing variables on the fabrication of electrospun
nanofibers for drug delivery applications. Journal of Nanomaterials 2013:1–22.
212. Sridhar, R., Lakshminarayanan, R., Madhaiyan, K., Barathi, V.A., Lim, K.H.C., and Ramakrishna, S.
(2015). Electrosprayed nanoparticles and electrospun nanofibers based on natural materials:
Applications in tissue regeneration, drug delivery and pharmaceuticals. Chemical Society Reviews
44:790–814.
213. Khalf, A. and Madihally, S.V. (2017). Recent advances in multiaxial electrospinning for drug d elivery.
European Journal of Pharmaceutics and Biopharmaceutics 112:1–17.
214. Rijal, N.P., Adhikari, U., and Bhattarai, N. (2017). Production of electrospun chitosan for biomedical
applications. In Chitosan Based Biomaterials (eds. Jennings J.A. and Bumgardner J.D.), Vol. 1,
211–237.
215. Hu, X., Liu, S., Zhou, G., Huang, Y., Xie, Z., and Jing, X. (2014). Electrospinning of polymeric
nanofibers for drug delivery applications. Journal of Controlled Release 185:12–21.
216. Yoo, H.S., Kim, T.G., Park, T.G. (2009) Surface‐functionalized electrospun nanofibers for tissue
engineering and drug delivery. Advanced Drug Delivery 61:1033–1042.
Chitosan‐Based Drug Delivery Systems 289
217. Chen, M., Gao, S., Dong, M., Song, J., Yang, C., Howard, K.A., Kjems, J., and Besenbacher, F.
(2012). Chitosan/siRNA Nanoparticles Encapsulated in PLGA Nanofibers for siRNA Delivery. ACS
Nano 6(6):4835–4844.
218. Monteiro, N., Martins, M., Martins, A., Fonseca, N.A., Moreira, J.N., Reis, R.L., and Neves, N.M.
(2015). Antibacterial activity of chitosan nanofiber meshes with liposomes immobilized releasing
gentamicin. Acta Biomaterialia 18:196–205.
219. Racine, L., Costa, G., Bayma‐Pecit, E., Texier, I., and Auzély‐Velty, R. (2017). Design of interpene-
trating chitosan and poly(ethylene glycol) sponges for potential drug delivery applications.
Carbohydrate Polymers 170:166–175.
220. Biondi, M., Ungaro, F., Quaglia, F., and Netti, P. A. (2008). Controlled drug delivery in tissue
engineering. Advanced Drug Delivery Reviews 60(2):229–242.
221. Dorati, R., DeTrizio, A., Modena, T., Conti, B., Benazzo, F., Gastaldi, G., and Genta, I. (2017).
Biodegradable scaffolds for bone regeneration combined with drug‐delivery systems in osteomyelitis
therapy. Pharmaceuticals 10(4):96.
222. Vunain, E., Mishra, A.K., and Mamba, B.B. (2017). Fundamentals of chitosan for biomedical appli-
cations. Chitosan Based Biomaterials (eds. Jennings, J.A. and Bumgardner, J.D.), Vol. 1, 3–30.
223. Mucha, M., Michalak, I., Balcerzak, J., and Tylman, M. (2012). Chitosan scaffolds, films and micro-
granules for medical application – Preparation and drug release studies. Polimery 10:714.
224. Prabaharan, M. and Sivashankari, P.R. (2016). Prospects of bioactive chitosan‐based scaffolds in tis-
sue engineering and regenerative medicine. In: Chitin and Chitosan for Regenerative Medicine (ed.
Dutta P.). Springer Series on Polymer and Composite Materials. Springer, New Delhi.
12
The Application of Chitin and its
Derivatives for the Design
of Advanced Medical Devices
Marcin H. Struszczyk, Longina Madej‐Kiełbik, and Dorota Zielińska
The Institute of Security Technologies “MORATEX”, Lodz, Poland
The two most important European Union (EU) regulations that specify the possibility of
chitin and chitosan medical application are as follows:
• Medical Devices Directive (MDD) – Directive 93/42/EEC (MEDDEV);
• EU Regulation 2017/745 on medical devices, amending Directive 2001/83/EC,
Regulation (EC) No 178/2002 and Regulation (EC) No 1223/2009, and repealing
Council Directives 90/385/EEC and 93/42/EEC, which will be implementing a new
regulation of the medical devices in 2020.
chitinous raw materials consists of several verification and validation steps. The general
limitations for selecting the most proper raw sources are:
• Detailed characterisation of raw materials (chemical, physical verification) for the deter-
mination of the biopolymers degree of purity as well as determination of the characteristic
of the initial raw sources as an input for ensuring the reproducibility of the manufacture;
• Verification of their biocompatibility regarding the future application and design attrib-
utes of the medical device;
• Determination of the biodegradation susceptibility or selection of the biopolymers
characteristic related to the required biodegradation period;
• Verification of the extent and effect of leakage of the substances (such as low‐molecular‐
weight fractions, proteins, minerals, etc.);
• Determination of the effect of the fabrication process including the sterilization with
suitable selection of the optimal sterilization agent (irradiation, steam or ethylene oxide
(EO)) on the biopolymer characteristic (degradation, chemical modification, changes in
crystallinity, etc.) and performance (changes in insolubility or solubility);
• Determination of the possible effects of the wound dressing performance due to the
interaction of the biopolymer and other constituents, processing aids, etc.;
• Verification of the wound dressing biocompatibility and determination of the constitu-
ent effects, risk of cross‐contamination, as well as manufacture process (including
sterilization);
• Verification of the microbiological and particulate contamination levels, both of the
raw materials and the final wound dressing (before sterilization) due to the risk of
pyrogenicity [21].
The crucial limitations in applying chitin and its derivatives for designing the wound
dressing are:
• Acquisition of reproducible sources of raw chitin (animals) as a starting point for the
separation of high‐quality biopolymer;
• Absence of the European (EN) standards describing the essential requirements for
chitin‐originated raw materials applicable for medical devices;
• Restricted access for the reproducible, ultra‐pure biopolymer manufactured in controlled
environments (reduction of the microbiological contamination) with verifiable
purification steps;
• Obligation to meet requirements of EN ISO 22442 standards regarding the documented
validation of the elimination of animal originated pathogens as well as measurable
determination of the risk of pathogens transfer to human in consideration to the process
of biopolymer separation as well as main process of the wound dressing manufacture;
• Necessity to use sterile wound dressing in relation to risk of modification of its perfor-
mance and safety by action of the sterilization agent (mainly EO or irradiation) [21,22].
• ASTM F2103‐18 (Guide for Characterization and Testing of Chitosan Salts as Starting
Materials Intended for Use in Biomedical and Tissue‐Engineered Medical Product
Applications);
• ASTM F2260‐03 (Test Method for Determining Degree of Deacetylation in Chitosan
Salts by Proton Nuclear Magnetic Resonance (1H NMR) Spectroscopy);
• ASTM F2602‐13 (Test Method for Determining the Molar Mass of Chitosan and
Chitosan Salts by Size Exclusion Chromatography with Multi‐angle Light Scattering
Detection (SEC‐MALS)).
The most applicable analytical tests to verify the quality of chitosan that is most often used
for designing the wound dressings are focused on (but not limited to) the determination of:
• Moisture;
• Residual substances (ash, protein, dyes, heavy metal) content;
• Degree of insolubility;
• Solution turbidity;
• Viscosity;
• Degree of deacetylation (DD);
• Crystallinity;
• Molecular mass and degree of polymerization.
The quality control should consider the identification of the usable form of the biopoly-
mer (powder, flakes, etc.) and colour [22].
Moreover, the crucial aspect for the verification of the safety of chitinous biopolymers is
the determination of microbiological contamination (bioburden ‐ aerobic bacteria and
fungi content) affecting the potential risk of the pyrogenicity of the final wound dressing.
In some critical applications, such as implants, the endotoxin level should also be
determined using the selected methodology, since chitosan as a polyaminosaccharide
induces an increase of the endotoxins level. An outline (flow chart) of the chitosan purity
determination process is shown in detail in [23].
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 295
sterilization yielded great darkening of the colour of chitosan powder with the colora-
tion deepening (yellow to brown) by an increase in the temperature and prolongation of
heat exposure [24]. Steam sterilization of chitosan flakes dispersed in water remained
the molecular weight of the biopolymer [28]. However, the changes induced by dry
steam did not significantly affect sterilized chitosan under anoxic conditions [24].
c. EO – EO sterilization of chitinous biopolymers or wound dressing (in the usable form
of a film) containing these biopolymers produces by‐products and cross‐linking result-
ing in reduction of mechanical strength [26]. Additionally, EO is irritating to human
body and is mutagenic, has fetotoxic and teratogenic behaviour, can adversely affect
testicular function and shows systemic toxicity. After exposure to EO, several adverse
systemic effects were detected, such as the induction of various types of neoplastic
changes including leukaemia, brain tumours and mammary tumours [29]. The risk of
the adverse accumulation of EO (after sterilization) increases with the porosity of the
materials, and affects cytotoxicity, irritation, mutagenicity and genotoxicity potential.
According to Standard EN‐ISO 10993‐7 (Biological evaluation of medical devices – Part
7: Ethylene oxide sterilization residuals), the EO residue reacts easily with water (such
as hydrogels), resulting in ethylene glycol formation. In some cases, ethylene chloro-
hydrin is also a co‐product of the reaction of EO residues. It shows high irritation affin-
ity, acute toxicity as well as weak mutagenic and carcinogenic potential. The study by
França et al. [30] shows that EO exposure on three types of chitosan with different
degree of N‐deacetylation, and carboxyethyl chitosan resulted in unexpected changes
in the chemical structure of the biopolymers, nonetheless without any negative effect
on the biocompatibility and toxicity. Additionally, EO exposure led to a minor change
in the crystallinity and network structure of chitosan powder. EO sterilization of medi-
cal devices based on chitinous biopolymers is limited to the dry state due to the possible
derivatisation of chitinous biopolymers if the wet state underwent sterilization [28].
This research underlined the risks associated with toxicity, flammability and aspects of
material contamination with EO residues.
d. Irradiation (γ‐irradiation or accelerated electrons) – γ radiation is a rapid, residue‐free
method that can be monitored by dosimetry. However, γ radiation cannot be used to
sterilize some types of polymers because of their inherent sensitivities or susceptibilities.
Sterilization of chitosan in the film form by γ‐irradiation with irradiation dose higher
than 10 kGy resulted in biopolymer chain degradation, deterioration of the mechanical
properties as well as decrease in the susceptibility to swell. On the other hand, realisa-
tion of the process under anoxic conditions significantly reduces negative alteration,
indicating that the irradiation process formed free radicals and accelerated the process of
biopolymer chains degradation. The standard γ irradiation sterilization dose of 25 kGy is
used in the industry for most medical devices that come in contact with skin or tissue.
This dose is considered sufficient for gaining a minimum of 10−6 sterility assurance level
(1 in 1,000,000 of medical devices can be unsterile after the sterilization process).
Each sterilization agent induces changes in the biopolymer structure, and initiates deg-
radation, cross‐linking and/or chemical modification affecting the performance of the final
product, as well as its safety. The determination of the effect of sterilization on biopolymers
from the possible interaction of the constituents and processing aids is the main aspect of
the process of proper selection of the biopolymer parameters, as well as in designing the
wound dressing.
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 297
Wound dressing Manufacturer Main constituent Usable form and main performance
Celox™ Gauze Medtrade Products Ltd. Chitosan • 5’ Z‐fold gauze or gauze 10′ roll impregnated by haemostatic granules
• Bleeding control
• Chosen by multiple NATO forces
• CE‐certified
Celox RAPID Gauze • 5’ Z‐Fold gauze impregnated by haemostatic granules
• Works with just 60 seconds compression
• Bleeding control
• CE‐certified
Celox™ Haemostatic • Granules are available in a 15 g sachet
Granules • Bleeding control
• Moulding to the shape of the wound
• Registered as class III CE‐marked medical device
Celox™‐A • A unique applicator pre‐packed with 6 g of Celox™ granules
• CE‐certified
HemConChitoGauze® Tricol Biomedical, Inc. Chitosan • 10 cm × 3.5 m Z‐fold gauze coated by chitosan
PRO • Antibacterial properties
• Stops severe bleeding
• Reduces blood loss
• CE‐certified
ChitoGauze® XR PRO • 7.6 cm × 3.7 m Z‐fold gauze coated by chitosan with x‐ray detectable element
• Antibacterial properties
• Stops severe bleeding
• Reduces blood loss
• CE‐certified
HemCon Patch® PRO • 2.5 cm × 7.5 cm haemostatic patch
• Antibacterial properties
• Severe bleeding control
ChitoDot® • ∅1,9 cm a double‐sided haemostatic dressing
• Antibacterial properties
• Bleeding control
HemCon® Strip PRO • Haemostatic bandage
• Antibacterial properties
• Bleeding control
(Continued )
Wound dressing Manufacturer Main constituent Usable form and main performance
Chitoderm® Plus Trusetal Chitosan • Foam, 10 cm × 12 cm
Verbandstoffwerk • Super absorber with antibacterial action
GmbH • Promotes wound healing
• Inhibits the growth of bacteria
• Rapid reduction of pathogenic organisms
ExcelArrest® XT Hemostasis, LLC. Carboxymethylated • Foam in three various sizes: 5 cm × 5 cm, 5 cm × 10 cm, 10 cm × 10 cm
chitosan • Moderate to severe bleeding control
• CE‐certified
• FDA‐cleared
KytoCel® Aspen Medical Chitosan • Gelling fibre dressing available in six sizes: 5 cm × 5 cm, 10 cm × 10 cm,
15 cm × 15 cm, 4 cm × 10 cm, 4 cm × 20 cm, 4 cm × 30 cm
• Minor bleeding control
HEMO‐ Coreleader Biotech Chitosan • Woven into a strong haemostatic gauze in three various sizes:
FiberKompresse Co. Ltd. 7.5 cm × 300 cm, 10 cm × 200 cm, 10 cm × 300 cm
• Control of severe bleeding
• Biocompatibility
• High tensile strength
• CE‐certified
∘∘ Heavy metals (Marine sources of chitosan can contain heavy metal contaminants
such as lead and mercury, and these should be analysed. Mercury has been desig-
nated as a hazardous material that can damage the central nervous system, kidney and
liver. Therefore, properties of chitosan should be evaluated using test methods that
are appropriate to ensure safety and efficacy.)
∘∘ Microbiological burden (bacteria, yeast and mould; the presence of bacteria may also
contribute to the presence of endotoxins)
• Reproducibility in the manufacture of the compound
• Compatibility with the essential requirements for medical devices and the European
Standards (e.g. EN ISO 22442‐1/2/3 Standards)
• Sterilization of medical devices containing chitin or its derivatives – most critical pro-
cesses may affect their biocompatibility [21].
The safety of chitosan in biomedical applications should be determined according to
current guidelines, such as ISO 10993 and ASTM F748 standards.
The performance assessment should review analytical performance data and, where
appropriate, clinical performance data in the form of any:
• Literature
• Performance study reports
• Experience gained by routine diagnostic testing.
This is to establish that medical devices achieve their intended performance under normal
conditions of use, and that known and foreseeable risks, and any undesirable effects, are
minimised and acceptable when weighed against the benefits of the intended performance.
In recent years, growing scientific attention to wound dressing based on chitin and its
derivatives, including chitosan, is observed. The Scopus database shows how many
publications (keywords: ‘chitosan’ and ‘dressing’) appeared since 2010 (Figure 12.1).
Since 2010, the number of documents has doubled. This shows that a large number of
research groups are dedicated to producing new and improved wound dressings. Most
papers are connected with materials science and engineering (Figure 12.2).
Some new ideas and concepts of dressing based on chitosan and its derivatives are
discussed in the following text.
The gel‐e® [49] products are made from modified chitosan with natural fatty acids (corn
and sunflower oil). It is capable of rapidly self‐assembling into robust physical seals
designed to stop bleeding. It is available in four forms: Vascular gel‐e®, Nuisance bleed
gel‐e®, Chronic wound gel‐e® (films, bandages, gels) and Surgical gel‐e® (gels, foams,
sponges). All these products are not yet commercially available, but at the stage of
preclinical and clinical testing. Only Vascular gel‐e® has been cleared by the FDA for use
in vascular access procedures.
Sarhan et al. developed biocompatible, antimicrobial, nanofibrous wound dressing based
on fabricated honey, poly(vinyl alcohol) and chitosan nanofibres. A preliminary in vivo
study proved the biocompatibility and showed that developed nanofibre mats enhanced the
wound healing process [50].
Lu et al. incorporated Ag/ZnO nanocomposites into a chitosan sponge, and then it was
exposed to a lyophilisation process [51]. The results showed high porosity and swelling, as
well as enhanced blood clotting and antibacterial activity. Cytocompatibility test evaluated
in vitro demonstrated a very low toxic nature of the composite dressing, and in vivo
evaluation in mice showed that the developed composite dressing enhances wound healing
200
180 180
160 163
147
140
Published documents
120
107
100
96
81 83
80
72
60
40
20
0
2010 2011 2012 2013 2014 2015 2016 2017
Figure 12.1 Published documents on the application of chitin and its derivatives, including chitosan, for
designing wound dressing, as listed in the Scopus database (Scopus; 2018.02.15).
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 303
Pharmacology,
Toxicology and
Pharmaceutics
7%
Materials Science
23%
Medicine
10%
Engineering
Chemical 15%
Engineering
10%
Chemistry
Biochemistry, 12%
Genetics and
Molecular Biology
11%
Figure 12.2 Published documents on the application of chitin and its derivatives as listed in the Scopus
database, divided into science areas (Scopus; 2018.02.15).
and promotes re‐epithelialisation and collagen deposition. Authors suggested that there
was a possibility of using this novel chitosan–AgZnO composite dressing for wound care
application.
According to the literature [52], chitosan dressing with silver can be used as a burn
dressing. It offered an optimal environment for moist wound management. Moreover, it did
not demonstrate shrinkage or disintegration as compared to the widely used hydrofibre
dressings. Studies by Alemdaroğlu et al. [53], related to the development of chitosan gel
formulation containing epidermal growth factor, suggested a potential opportunity to use
that material for healing second‐degree burn wounds (the studies were carried out on rats).
Chilarski et al. [54] suggested a potential opportunity to use the chitin derivative
dibutyrylchitin for the treatment of burn wounds, postoperative/posttraumatic wounds and
various other conditions causing skin/epidermis loss.
Chitosan–sulphadiazine membrane has been developed by Dragostin et al. [55]. The
authors prepared six chitosan–sulphonamide derivatives. The results demonstrated an
improved healing effect and enhanced epithelialisation of chitosan–sulphonamide
derivatives as compared to untreated chitosan.
N,N,N‐trimethyl chitosan was deposited onto polypropylene and polylactide nonwovens
[56]. The results showed that modified material exhibited good antibacterial properties
against Gram‐positive bacteria Staphylococcus aureus.
The research carried out by Zhou et al. indicated that N,N,N‐trimethyl chitosan fibres
have the potential to be used as wound dressing materials [57]. The results showed that the
obtained fibres had higher absorption capacity, higher antibacterial performance compared
to chitosan fibres, and were non‐toxic to mouse embryo fibroblasts.
304 Chitin and Chitosan: Properties and Applications
Chitosan was used by Dumont et al. for coating alginate fibres [58]. The obtained fibres
contained about 10% (v/v) of chitosan by an external coating. The result showed that
chitosan‐coated alginate fibres presented an antibacterial activity against Gram‐negative
Escherichia coli sp., and more interestingly against Gram‐positive Staphylococcus genus.
Moreover, it was demonstrated that antibacterial properties did not change after steriliza-
tion process by beta‐irradiation.
Alginate fibres with chitosan sheath were prepared by Chang et al. with the electrospinning
method [59]. Obtained fibres exhibited a core–sheath structure, and the average diameter
of the fibres ranged from 600 to 900 nm, depending on the electrospinning parameters.
They suggested that that core–sheath electrospun fibre had a potential for biomedical
applications.
The research carried out by Chen et al. [60] was focused on electrospun nanofibre mats
of pectinate, alginate and chitosan. They showed that all nanofibre mats had comparable
mechanical strength and vapour permeability. However, pectinate nanofibre mats could
absorb more exudates in less time and suppress the growth of bacteria more effectively, so
the authors suggested that pectinate nanofibre mats might be a superior wound dressing as
compared to the alginate and chitosan nanofibre mats.
Nanofibre mats were also developed by Kohsari et al. [61]. The chitosan–PEO nanofibre
mats were prepared by the electrospinning technique. Silver nanoparticles were added in
two different weight percentages. The results showed that the obtained 21anofibers
exhibited 100% antibacterial performance against both the Gram‐positive S. aureus and
Gram‐negative E. coli bacteria.
In another study [62], chitosan membranes were prepared as potential coating for tita-
nium implants. A surface roughness and fibronectin adsorption increase was observed with
increased DD. The authors concluded that there was a possibility of preparing chitosan
membranes by selecting from chitosan types with a wide range of DD, thereby displaying
that different bioactivity would be a useful advancement in the coating of titanium for
orthopaedic applications.
Composite wound dressing based on the chitosan‐oxidised Bletilla striata polysaccharide
and chitosan‐Ag, both cross‐linked with genipin, were successfully fabricated for wound
repair applications by Ding et al. [63]. The in vivo studies indicated that wound dressing
significantly accelerated the healing rate of cutaneous wounds in mice, and it had great
potential in wound dressing applications.
The aim of the research of Saporito et al. was to develop sponge‐like dressings based on
chitosan and chondroitin sulphate or hyaluronic acid [64]. It was prepared by lyophilisation
and loaded with tranexamic acid. The in vitro and ex vivo studies showed that dressings
were biocompatible and able to sustain cells. The result suggested that these dressings are
effective tools to enhance haemostasis and healing in bleeding wounds.
The surface of nonwoven chitosan was modified by the succinyl groups, carboxymethyl
groups and quaternary ammonium groups by Yan et al. [65]. The results demonstrated that
the modified nonwoven exhibited better haemostatic property than gauze and initial
nonwoven, especially, the nonwoven with succinyl groups had an excellent haemostatic
effect, compared to other nonwoven samples.
The research carried by Zielińska et al. [66] described two usable forms of a topical
haemostatic agent: chitosan/alginate lyophilised foam and chitosan/alginate impregnated
gauze. The presented forms of the topical haemostatic agent, being the main part of the
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 305
Subsequent researches have shown that chitosan derivatives might be used in the preparation
of influenza vaccines [76,77]. The research carried by Frohbergh et al. [78] described
nanofibrous scaffolds from chitosan for bone tissue engineering applications. Scientists
have developed a one‐step technology to generate electrospun and hydroxyapatite‐
containing fibrous chitosan scaffolds that are subsequently cross‐linked with genipin as
potential substitutes for periosteum. The authors proposed that electrospun cross‐linked
hydroxyapatite‐containing chitosan nanofibrous scaffolds subsequently cross‐linked with
genipin were potential candidates for non‐weight‐bearing bone tissue engineering – that is,
for cranial and maxillofacial reconstruction.
The smartness of chitin nanofibril/chitosan glycolate‐based preparations, a spray
(Chit‐A), a gel (Chit‐B) and a gauze (Chit‐C), in healing cutaneous lesions was assessed
macroscopically and by light microscopy immunohistochemistry [79]. Those evaluations
were compared to the results obtained using a laser co‐treatment. The wound repair pro-
vided by those preparations was clearly evident even without the synergistic effect of the
laser co‐treatment.
Chitosan derivatives with quaternary ammonium groups showed very good performance
against bacteria and fungi. The explanation that the cytoplasmic membrane of bacterial
cells is the target of these cationic polymers is generally accepted [80]. The photo‐cross‐
linked electrospun mats containing quaternary chitosan (QCS) were capable of inhibiting
the growth of both Gram‐positive and Gram‐negative bacteria [81]. The results indicate
that the cross‐linked QCS/poly(vinyl alcohol) (PVA) electrospun mats are advantageous
material applicable for wound dressing. Likewise, the photo‐cross‐linked electrospun
nanofibrous QCS/PVA products exhibited optimal bactericidal activity towards the Gram‐
negative E. coli and Gram‐positive S. aureus bacteria [81].
In vivo studies of a bioactive dressing containing silver sulphadiazine (AgSD) as anti‐
infective drug and alpha tocopherol (αTph) as antioxidant agent, and intended to be loaded
with autologous platelet lysate in the treatment of chronic skin wounds, were performed by
Bonferoni et al. [82]. As both AgSD and αTph are hardly soluble, nanocarriers such as
polymeric micelles (AgSD‐Mic) and nanoemulsions (αTphNE), based on an amphiphilic
salt of chitosan with oleic acid, were previously developed and characterised to improve
their dispersion in aqueous environment. The results confirmed the positive encapsulation
of poorly soluble hydrophobic molecules in chitosan oleate. The authors suggested that the
bioactive wound dressing can be a suitable and versatile support of the haemoderivative for
the treatment of wounds. Moreover, the effect on wound healing was dose‐dependent, mak-
ing it a flexible tool in wound treatment.
The design of the new usable form of the chitinous biopolymers applicable for innova-
tive wound dressing is supported by initiation activity focused on understanding the prob-
lem to be solved with strict interconnection of the identified problems with the hazards in
the risk analysis (the risk identification).
The initiation design phase is helpful at defining and making assumptions of the concept
to be developed, by the ideation of the models and prototypes that would be verified and
validated during the testing phase of the design process. Creating the design of a usable
form of the biopolymers included three phases: initialisation, main design process and the
post‐design processes (collecting end‐user feedback) [66].
The initialisation is related to the problem identification by the design team, mostly by
deeply empathising with the end‐users (practitioners and patients). As a result of this phase,
initial assumptions for the raw materials, technology (to be developed) and design of the
final wound dressing should be defined. The main document – the research programme
based on the initial risk analysis – is also the main output of the initialisation phase. As a
minimum, the research programme should cover testing methods for risk reduction to an
acceptable level, as well as define the optimal level of the parameters describing each
functionality of the wound dressing to be designed [66].
The development phase consists of the verification and validation stages of the designed
models and the resulting prototypes. During the main development activities, the models
and prototypes of the usable forms of the topical haemostatic agents are verified and
validated. The primary verification activities are usually focused on testing the usable
properties that define the functionalities of the designed medical devices, and then
verification of the biocompatibility, stability as well as validation stages, including process
validation, sterilization validation and, finally, the clinical study [66].
12.6 Outlook
Chitin and its derivatives, including chitosan – the chitinous polymer most commonly used
for designing wound dressings – are promising biopolymers in this kind of applications due
to the documented wide‐range bioactivity and biocompatibility. The haemostatic, wound‐
healing acceleration, high gelling affinity and various usable forms are the main indicators
for selecting chitinous biopolymers for medical applications. However, the critical disad-
vantage of the above‐mentioned polymers is the absence of reproducibility of the initial
parameters because of the specificity of the natural origins. The elimination of this aspect
should allow for enhancing the application of chitinous biopolymers in medical fields.
Acknowledgements
This research was supported by the National Science Centre for Research and Development
under the research project No. DOB‐BIO6/19/98/2014 ‘The dressing set for protecting the
injuries which occur at the duty activities of uniformed forces’.
References
1. Blumenthal, H. J. and Roseman, S. Quantitative estimation of chitin in fungi. J Bacteriol. [Internet].
1957;74(2):222–224. Available from: http://www.ncbi.nlm.nih.gov/pubmed/13475224
2. Wagner, G. P., Lo, J., Laine, R., and Almeder, M. Chitin in the epidermal cuticle of a vertebrate
(Paralipophrys trigloides, Blenniidae, Teleostei). Experientia [Internet]. 1993;49(4):317–319. Available
from: http://link.springer.com/10.1007/BF01923410
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 309
3. Khoushab, F. and Yamabhai, M. Chitin research revisited [Internet]. Vol. 8, Mar. Drugs. Molecular
Diversity Preservation International; 2010:1988–2012. Available from: http://www.mdpi.com/
1660‐3397/8/7/1988
4. Nishiyama, A., Tsuji, S., Yamashita, M., Henriksen, R. A., Myrvik, Q. N., and Shibata, Y. Phagocytosis
of N‐acetyl‐d‐glucosamine particles, a Th1 adjuvant, by RAW 264.7 cells results in MAPK activation
and TNF‐alpha, but not IL‐10, production. Cell Immunol. [Internet]. 2006;239(2):103–112. Available
from: http://www.ncbi.nlm.nih.gov/pubmed/16781693
5. Nishiyama, A., Shinohara, T., Pantuso, T., Tsuji, S., Yamashita, M., Shinohara, S. et al. Depletion of
cellular cholesterol enhances macrophage MAPK activation by chitin microparticles but not by heat‐
killed Mycobacterium bovis BCG. Am. J. Physiol. Cell. Physiol. [Internet]. 2008;295(2):C341–349.
Available from: http://www.ncbi.nlm.nih.gov/pubmed/18524942
6. Kogiso, M., Nishiyama, A., Shinohara, T., Nakamura, M., Mizoguchi, E., Misawa, Y. et al. Chitin
particles induce size‐dependent but carbohydrate‐independent innate eosinophilia. J. Leukoc. Biol.
[Internet]. 2011;90(1):167–176. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21447645
7. Reese, T. A., Liang, H. E., Tager, A. M., Luster, A. D., Van Rooijen, N., Voehringer, D. et al. Chitin
induces accumulation in tissue of innate immune cells associated with allergy. Nature [Internet].
2007;447(7140):92–96. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17450126
8. Shibata, Y., Honda, I., Justice, J. P., Van Scott, M. R., Nakamura, R. M., and Myrvik, Q. N. Th1
Adjuvant N‐acetyl‐D‐glucosamine polymer up‐regulates Th1 immunity but down‐regulates Th2
immunity against a Mycobacterial protein (MPB‐59) in interleukin‐10‐knockout and wild‐type
mice. Infect. Immun. [Internet]. 2001;69(10):6123–6130. Available from: http://www.ncbi.nlm.nih.
gov/pubmed/11553551
9. Shibata, Y., Foster, L. A., Metzger, W. J., and Myrvik, Q. N. Alveolar macrophage priming by intrave-
nous administration of chitin particles, polymers of N‐acetyl‐D‐glucosamine, in mice. Infect. Immun.
[Internet]. 1997;65(5):1734–1741. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9125555
10. Shibata, Y., Metzger, W. J., and Myrvik, Q. N. Chitin particle‐induced cell‐mediated immunity is
inhibited by soluble mannan: mannose receptor‐mediated phagocytosis initiates IL‐12 production.
J. Immunol. [Internet]. 1997;159(5):2462–2467. Available from: http://www.ncbi.nlm.nih.gov/
pubmed/9278339
11. Da Silva, C. A., Hartl, D., Liu, W., Lee, C. G., and Elias, J. A. TLR‐2 and IL‐17A in chitin‐induced
macrophage activation and acute inflammation. J. Immunol. [Internet]. 2008;181(6):4279–86. Available
from: http://www.ncbi.nlm.nih.gov/pubmed/18768886
12. Da Silva, C. A., Pochard, P., Lee, C. G., and Elias, J. A. Chitin particles are multifaceted immune
adjuvants. Am. J. Respir. Crit. Care Med. [Internet]. 2010;182(12):1482–91. Available from: http://
www.ncbi.nlm.nih.gov/pubmed/20656945
13. Koller, B., Müller‐Wiefel, A. S., Rupec, R., Korting, H. C., and Ruzicka, T. Chitin modulates innate
immune responses of keratinocytes. Schauber J, editor. PLoS One [Internet]. 2011;6(2):e16594.
Available from: http://www.ncbi.nlm.nih.gov/pubmed/21383982
14. Suzuki, K., Okawa, Y., Suzuki, S., and Suzuki, M. Candidacidal effect of peritoneal exudate cells in
mice administered with chitin or chitosan. Microbiol. Immunol. [Internet]. 1987;31(4):375–379.
Available from: http://doi.wiley.com/10.1111/j.1348‐0421.1987.tb03098.x
15. Suzuki, K., Okawa, Y., Hashimoto, K., Suzuki, S., and Suzuki, M. Protecting effect of chitin and
chitosan on experimentally induced Murine Candidiasis. Microbiol. Immunol. [Internet].
1984;28(8):903–912. Available from: http://doi.wiley.com/10.1111/j.1348‐0421.1984.tb00746.x
16. Rementería, A., Abaitua, F., García‐Tobalina, R., Hernando, F., Pontón, J., and Sevilla, M. J.
Resistance to candidiasis and macrophage activity in chitin‐treated mice. FEMS Immunol. Med.
Microbiol. [Internet]. 1997;19(3):223–230. Available from: https://academic.oup.com/femspd/
article‐lookup/doi/10.1111/j.1574‐695X.1997.tb01091.x
17. Cartier, A., Lehrer, S. B., Horth‐Susin, L., Swanson, M., Neis, B., Howse D. et al. Prevalence of crab
asthma in crab plant workers in Newfoundland and Labrador. Int. J. Circumpolar Health [Internet].
2004;63 Suppl. 2:333–336. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15736679
310 Chitin and Chitosan: Properties and Applications
18. Vo, T.‐S., Ngo, D.‐H., Ta Q Van, Wijesekara, I., Kong, C.‐S., and Kim, S‐K. Protective effect of chitin
oligosaccharides against lipopolysaccharide‐induced inflammatory response in BV‐2 microglia. Cell
Immunol. [Internet]. 2012;277(1–2):14–21. Available from: https://www.sciencedirect.com/science/
article/pii/S0008874912001177
19. Nagatani, K., Wang, S., Llado, V., Lau, C. W., Li, Z., Mizoguchi, A. et al. Chitin microparticles for the
control of intestinal inflammation. Inflamm. Bowel Dis. [Internet]. 2012;18(9):1698–1710. Available
from: http://www.ncbi.nlm.nih.gov/pubmed/22241684
20. Da Silva, C. A., Chalouni, C., Williams, A., Hartl, D., Lee, C. G., and Elias, J. A. Chitin is a size‐
dependent regulator of macrophage TNF and IL‐10 production. J. Immunol. [Internet]. 2009
Mar;182(6):3573–3582. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19265136
21. Struszczyk, M. H. and Struszczyk, K. J. Medical applications of chitin and its derivatives. Polish Chitin
Soc. Monogr XII [Internet]. 2007; Available from: http://ptchit.lodz.pl/PTChit/12_18.pdf
22. Struszczyk, M. H. 12. Global requirements for medical applications of chitin and its derivatives. 2006;
Polish Chitin Society, Monograph XI.
23. Kardas, I., Struszczyk, M. H., Kucharska, M., van den Broek, L. A. M., van Dam, J. E. G., and
Ciechańska, D. Chitin and chitosan as functional biopolymers for industrial applications. In: The
European Polysaccharide Network of Excellence (EPNOE): Research Initiatives and Results [Internet].
Vienna: Springer Vienna; 2012. p. 329–373. Available from: http://link.springer.com/10.1007/978‐3‐7
091‐0421‐7_11
24. Lim, L.‐Y., Khor, E., and Ling, C.‐E. Effects of dry heat and saturated steam on the physical properties
of chitosan. J. Biomed. Mater. Res. [Internet]. 1999;48(2):111–116. Available from: http://doi.wiley.com
/10.1002/%28SICI%291097‐4636%281999%2948%3A2%3C111%3A%3AAID‐JBM3%3E3.0.
CO%3B2‐W
25. Furuta, M., Oka, M., and Hayashi, T. Radiation sterilization of enzyme hybrids with biodegradable
polymers. Radiat. Phys. Chem. [Internet]. 2002 Mar;63(3–6):323–325. Available from: https://www.
sciencedirect.com/science/article/pii/S0969806X01005205?via%3Dihub
26. Marreco, P. R., Moreira, P. da L., Genari, S. C., and Moraes, A. M. Effects of different sterilization
methods on the morphology, mechanical properties, and cytotoxicity of chitosan membranes used as
wound dressings. J. Biomed. Mater. Res. [Internet]. 2004;71B(2):268–277. Available from: http://doi.
wiley.com/10.1002/jbm.b.30081
27. Jarry, C., Chaput, C., Chenite, A., Renaud, M. A., Buschmann, M., and Leroux, J. C. Effects of steam
sterilization on thermogelling chitosan‐based gels. J. Biomed. Mater. Res. [Internet].
2001;58(1):127–135. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11153009
28. San Juan, A., Montembault, A., Gillet, D., Say, J. P., Rouif, S., Bouet, T. et al. Degradation of chitosan‐
based materials after different sterilization treatments. In: IOP Conference Series: Materials Science
and Engineering. IOP Publishing; 2012. p. 12007.
29. Steenland, K., Stayner, L., Greife, A., Halperin, W., Hayes, R., Hornung, R. et al. Mortality among
workers exposed to ethylene oxide. N. Engl. J. Med. [Internet]. 1991;324(20):1402–1427. Available
from: http://www.nejm.org/doi/abs/10.1056/NEJM199105163242004
30. França, R., Mbeh, D. A., Samani, T. D., Le Tien, C., Mateescu, M. A., Yahia, L. et al. The effect of
ethylene oxide sterilization on the surface chemistry and in vitro cytotoxicity of several kinds of chi-
tosan. J. Biomed. Mater. Res. ‐ Part B Appl. Biomater. [Internet]. 2013;101(8):1444–1455. Available
from: http://doi.wiley.com/10.1002/jbm.b.32964
31. Products Celox Hemostats Celox [Internet]. [cited 2018 Feb 22]. Available from: http://www.
celoxmedical.com/products/
32. Products Archive – Tricol Biomedical [Internet]. [cited 2018 Feb 22]. Available from: https://www.
tricolbiomedical.com/welcome/shop/
33. Algi‐Plaster [Internet]. [cited 2018 Feb 22]. Available from: http://www.coreleaderbio.com/product_
view.php?cid=796&id
34. Military / Governmental | Core Scientific Creations [Internet]. [cited 2018 Feb 22]. Available from:
http://www.woundclot.com/?product=woundclot‐trauma‐ce‐iib
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 311
35. KytoCel – Aspen Medical [Internet]. [cited 2018 Feb 22]. Available from: http://www.aspenmedicaleurope.
com/specialist_wound_car/kytocel/
36. ExcelArrest XT Topical Hemostatic Patch [Internet]. [cited 2018 Feb 22]. Available from: http://www.
hemostasisllc.com/excelarrest‐techinfo.html
37. chitoderm®plus – Trusetal [Internet]. [cited 2018 Feb 22]. Available from: http://www.tshs.eu/en/
hydroactive‐bandages/chitosan‐wound‐dressing/chitodermr‐plus.html
38. Scion BioMedical‘s Topical Hemostasis [Internet]. [cited 2018 Feb 22]. Available from: http://
scionbiomed.com/topical‐hemostasis/
39. About Us | Syvek [Internet]. [cited 2018 Feb 22]. Available from: http://syvek.com/about‐us/
40. Emergency Bleeding Control Dressing, Stop Haemorrhage [Internet]. [cited 2018 Feb 22]. Available
from: http://www.axiobio.com/emergency/
41. Products | LifeScience Plus | BloodSTOP [Internet]. [cited 2018 Feb 22]. Available from: http://www.
lifescienceplus.com/products/
42. CHITOSAM – SAM Medical [Internet]. [cited 2018 Feb 22]. Available from: https://www.sammedical.
com/products/chitosam
43. Ishihara, M., Nakanishi, K., Ono, K., Sato, M., Kikuchi, M., Saito, Y. et al. Photocrosslinkable chitosan
as a dressing for wound occlusion and accelerator in healing process. Biomaterials [Internet].
2002;23(3):833–840. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11771703
44. Younes, I. and Rinaudo, M. Chitin and chitosan preparation from marine sources. Structure, properties
and applications. Mar. Drugs [Internet]. 2015 Mar;13(12):1133–1174. Available from: http://www.
mdpi.com/1660‐3397/13/3/1133
45. Jayakumar, R., Menon, D., Manzoor, K., Nair, S. V., and Tamura, H. Biomedical applications of chitin
and chitosan based nanomaterials – A short review. Carbohydr. Polym. [Internet]. 2010;82(2): 227–
232. Available from: https://www.sciencedirect.com/science/article/pii/S0144861710003589
46. Anitha, A., Sowmya, S., Kumar, P. T. S., Deepthi, S., Chennazhi, K. P., Ehrlich, H. et al. Chitin and
chitosan in selected biomedical applications. Prog. Polym. Sci. 2014;39(9):1644–1667.
47. Dutta, P. K., Rinki, K., and Dutta, J. Chitosan: A promising biomaterial for tissue engineering
scaffolds. Adv. Polym. Sci. [Internet]. 2011;244(1):45–80. Available from: http://link.springer.com/
10.1007/12_2011_112
48. Croisier, F. and Jérôme, C. Chitosan‐based biomaterials for tissue engineering. Eur. Polym. J.
[Internet]. 2013;49(4):780–792. Available from: https://www.sciencedirect.com/science/article/pii/
S0014305712004181
49. gel‐e life sciences technology [Internet]. [cited 2018 Feb 22]. Available from: http://www.gel‐e.co/
tech.html
50. Sarhan, W. A., Azzazy, H. M. E., and El‐Sherbiny, I. M. Honey/chitosan nanofiber wound dressing
enriched with Allium sativum and Cleome droserifolia: Enhanced antimicrobial and wound healing
activity. ACS Appl. Mater. Interfaces [Internet]. 2016 Mar;8(10):6379–6390. Available from: http://
www.ncbi.nlm.nih.gov/pubmed/26909753
51. Lu, Z., Gao, J., He, Q., Wu, J., Liang, D., Yang, H. et al. Enhanced antibacterial and wound healing
activities of microporous chitosan‐Ag/ZnO composite dressing. Carbohydr. Polym. [Internet].
2017;156:460–469. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27842847
52. Massand, S., Cheema, F., Brown, S., Davis, W. J., Burkey, B., and Glat, P. M. The use of a chitosan
dressing with silver in the management of paediatric burn wounds: A pilot study. J. Wound Care
[Internet]. 2017;26(sup4):S26‐‐S30. Available from: http://www.ncbi.nlm.nih.gov/pubmed/
28379104
53. Alemdaroǧlu, C., Deǧim, Z., Çelebi, N., Zor, F., Öztürk, S., and Erdoǧan, D. An investigation on burn
wound healing in rats with chitosan gel formulation containing epidermal growth factor. Burns
[Internet]. 2006;32(3):319–27. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16527411
54. Chilarski, A., Krucinska, I., Kiekens, P., Blasinska, A., Schoukens, G., Cislo, R. et al. Fibres & textiles
in Eastern Europe. [Internet]. Vol. 15, Fibres Text. East. Europe. Instytut Włókien Chemicznych; 2007.
p. 77–81. Available from: https://biblio.ugent.be/publication/744697
312 Chitin and Chitosan: Properties and Applications
55. Dragostin, O. M., Samal, S. K., Dash, M., Lupascu, F., Pânzariu, A., Tuchilus, C. et al. New antimicro-
bial chitosan derivatives for wound dressing applications. Carbohydr. Polym. [Internet]. 2016;141:28–
40. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26876993
56. Stawski, D., Sahariah, P., Hjálmarsdóttir, M., Wojciechowska, D., and Puchalski Michałand Másson,
M. N,N,N‐trimethyl chitosan as an efficient antibacterial agent for polypropylene and polylactide non-
wovens. J. Text Inst. [Internet]. 2017;108(6):1041–1049. Available from: https://www.tandfonline.
com/doi/full/10.1080/00405000.2016.1218594
57. Zhou, Z., Yan, D., Cheng, X., Kong, M., Liu, Y., Feng, C. et al. Biomaterials based on N,N,N‐trimethyl
chitosan fibers in wound dressing applications. Int. J. Biol. Macromol. [Internet]. 2016;89:
471–476. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26893050
58. Dumont, M., Villet, R., Guirand, M., Montembault, A., Delair, T., Lack, S. et al. Processing and anti-
bacterial properties of chitosan‐coated alginate fibers. Carbohydr. Polym. 2017;190:31–42.
59. Chang, J.‐J., Lee, Y.‐H., Wu, M.‐H., Yang, M.‐C., and Chien, C.‐T. Preparation of electrospun alginate
fibers with chitosan sheath. Carbohydr. Polym. 2012;87(3):2357–2361.
60. Chen, S., Cui, S., Hu, J., Zhou, Y., and Liu, Y. Pectinate nanofiber mat with high absorbency and
antibacterial activity: A potential superior wound dressing to alginate and chitosan nanofiber mats.
Carbohydr. Polym. [Internet]. 2017;174:591–600. Available from: http://www.ncbi.nlm.nih.gov/
pubmed/28821109
61. Kohsari, I., Shariatinia, Z., and Mahdi, S. Antibacterial electrospun chitosan – polyethylene oxide
nanocomposite mats containing bioactive silver nanoparticles. Carbohydr. Polym. [Internet].
2016;140:287–298. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26876856
62. Lieder, R., Darai, M., Thor, M. B., Ng, C. H., Einarsson, J. M., Gudmundsson, S. et al. In vitro bioactiv-
ity of different degree of deacetylation chitosan, a potential coating material for titanium implants. J.
Biomed. Mater. Res. – Part A [Internet]. 2012;100 A(12):3392–3399. Available from: http://www.ncbi.
nlm.nih.gov/pubmed/22767519
63. Ding, L., Shan, X., Zhao, X., Zha, H., Chen, X., Wang, J. et al. Spongy bilayer dressing composed of
chitosan–Ag nanoparticles and chitosan–Bletilla striata polysaccharide for wound healing applica-
tions. Carbohydr. Polym. [Internet]. 2017;157:1538–1547. Available from: http://www.ncbi.nlm.nih.
gov/pubmed/27987866
64. Saporito, F., Sandri, G., Rossi, S., Bonferoni, M. C., Riva, F., Malavasi, L. et al. Freeze dried chitosan
acetate dressings with glycosaminoglycans and traxenamic acid. Carbohydr. Polym. 2018;184:408–417.
65. Yan, D., Hu, S., Zhou, Z., Zeenat, S., Cheng, F., Li, Y. et al. Different chemical groups modification on
the surface of chitosan nonwoven dressing and the hemostatic properties. Int. J. Biol. Macromol.
2018;107:463–469.
66. Zielińska, D., Struszczyk, M., Madej‐Kiełbik, L., Chmal‐Fudali, E., Kucharska, M., Wiśniewska‐
Wrona, M. et al. Design of new‐generation usable forms of topical haemostatic agents containing
chitosan. Molecules [Internet]. 2017;22(12):2240. Available from: http://www.ncbi.nlm.nih.gov/
pubmed/29244748
67. Kamel, N. A., El‐messieh, S. L. A., and Saleh, N. M. Chitosan/banana peel powder nanocomposites for
wound dressing application: Preparation and characterization. Mater. Sci. Eng. C. 2017;72:543–550.
68. Kang, P. L., Chang, S. J., Manousakas, I., Lee, C. W., Yao, C. H., Lin, F. H. et al. Development and
assessment of hemostasis chitosan dressings. Carbohydr. Polym. [Internet]. 2011;85(3):565–570.
Available from: https://www.sciencedirect.com/science/article/pii/S0144861711001962
69. Chan, L. W., Kim, C. H., Wang, X., Pun, S. H., White, N. J., and Kim, T. H. PolySTAT‐modified chi-
tosan gauzes for improved hemostasis in external hemorrhage. Acta Biomater. 2016;31:178–185.
70. Dash, M., Chiellini, F., Ottenbrite, R. M., and Chiellini, E. Chitosan—A versatile semi‐synthetic poly-
mer in biomedical applications. Prog. Polym. Sci. 2011;36(8):981–1014.
71. Seol, Y. J., Lee, J. Y., Park, Y. J., Lee, Y. M., Young‐Ku, Rhyu, I. C. et al. Chitosan sponges as tissue
engineering scaffolds for bone formation. Biotechnol. Lett. [Internet]. 2004;26(13):1037–1041.
Available from: http://www.ncbi.nlm.nih.gov/pubmed/15218375
The Application of Chitin and its Derivatives for the Design of Advanced Medical Devices 313
72. Costa‐Pinto, A. R., Reis, R. L., and Neves, N. M. Scaffolds based bone tissue engineering: The role of
chitosan. Tissue Eng. Part B Rev. [Internet]. 2011;17(5):331–347. Available from: http://www.ncbi.
nlm.nih.gov/pubmed/21810029
73. Arpornmaeklong, P., Pripatnanont, P., and Suwatwirote, N. Properties of chitosan–collagen sponges
and osteogenic differentiation of rat‐bone‐marrow stromal cells. Int. J. Oral Maxillofac Surg.
2008;37(4):357–366.
74. Kumar, P. T. S., Srinivasan, S., Lakshmanan, V.‐K., Tamura, H., Nair, S. V., and Jayakumar, R.
Synthesis, characterization and cytocompatibility studies of α‐chitin hydrogel/nano hydroxyapatite
composite scaffolds. Int. J. Biol. Macromol. 2011;49(1):20–31.
75. Kumar, P. T. S., Srinivasan, S., Lakshmanan, V.‐K., Tamura, H., Nair, S. V., and Jayakumar, R. β‐Chitin
hydrogel/nano hydroxyapatite composite scaffolds for tissue engineering applications. Carbohydr.
Polym. 2011;85(3):584–591.
76. Svindland, S. C., Jul‐Larsen, Å., Pathirana, R., Andersen, S., Madhun, A., Montomoli, E. et al. The
mucosal and systemic immune responses elicited by a chitosan‐adjuvanted intranasal influenza H5N1
vaccine. Influenza Other Respi. Viruses [Internet]. 2012 Mar;6(2):90–100. Available from: http://www.
ncbi.nlm.nih.gov/pubmed/21749672
77. Jabbal‐Gill, I., Watts, P., and Smith, A. Chitosan‐based delivery systems for mucosal vaccines
[Internet]. Vol. 9, Exp. Opin. Drug Del. 2012 [cited 2018 Feb 22]. p. 1051–1067. Available from: http://
www.ncbi.nlm.nih.gov/pubmed/22708875
78. Frohbergh, M. E., Katsman, A., Botta, G. P., Lazarovici, P., Schauer, C. L., Wegst, U. G. K. et al.
Electrospun hydroxyapatite‐containing chitosan nanofibers crosslinked with genipin for bone tissue
engineering. Biomaterials. 2012;33(36):9167–9178.
79. Mattioli‐Belmonte, M., Zizzi, A., Lucarini, G., Giantomassi, F., Biagini, G., Tucci, G. et al. Chitin
nanofibrils linked to chitosan glycolate as spray, gel, and gauze preparations for wound repair.
J. Bioact. Compat Polym. [Internet]. 2007;22(5):525–538. Available from: http://journals.sagepub.
com/doi/10.1177/0883911507082157
80. Tashiro, T. Antibacterial and bacterium adsorbing macromolecules. Macromol. Mater. Eng. [Internet].
2001;286(2):63–87. Available from: http://doi.wiley.com/10.1002/1439‐2054%2820010201%29286
%3A2%3C63%3A%3AAID‐MAME63%3E3.0.CO%3B2‐H
81. Ignatova, M., Starbova, K., Markova, N., Manolova, N., and Rashkov, I. Electrospun nano‐fibre mats
with antibacterial properties from quaternised chitosan and poly(vinyl alcohol). Carbohydr. Res.
[Internet]. 2006;341(12):2098–2107. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16750180
82. Bonferoni, M., Sandri, G., Rossi, S., Dellera, E., Invernizzi, A., Boselli, C. et al. Association of alpha
tocopherol and Ag sulfadiazine chitosan oleate nanocarriers in bioactive dressings supporting platelet
lysate application to skin wounds. Mar. Drugs [Internet]. 2018;16(2):56. Available from: http://www.
ncbi.nlm.nih.gov/pubmed/29425164
83. Kucharska, M., Struszczyk, M. H., Cichecka, M., Brzoza‐Malczewska, K., and Wiśniewska‐Wrona,
M. Prototypes of primary wound dressing of fibrous and Quasi‐Fibrous structure in terms of safety of
their usage. Fibres Text East Eur. [Internet]. 2012;96(6 B):142–148. Available from: http://yadda.icm.
edu.pl/baztech/element/bwmeta1.element.baztech‐9b28fc50‐2dae‐4ebd‐9e86‐a6b695270ed8
84. Kucharska, M., Wiśniewska‐Wrona, M., Pałys, B., Struszczyk, M., Cichecka, M., Karuga‐
Kuźniewska, E. et al. Biological dressings based on natural folymers. Fibres Text East Eur [Internet].
2016;24(6(120)):170–174. Available from: http://220.indexcopernicus.com/abstracted.
php?level=5&ICID=1221753
85. Struszczyk, M. H., Wilbik‐Hałgas, B., Miklas, M., Cichecka, M., Kucharska, M., Wiśniewska‐Wrona,
M., and Brzoza‐Malczewska, K. Estimation of the performance stability of the newly developed topi-
cal haemostatic agents based on the chitosan/alginate fibrids. Text Res. J. [Internet]. 2017;87(7): 780–
789. Available from: http://journals.sagepub.com/doi/10.1177/0040517516639828
13
Food Applications of Chitosan
and its Derivatives
Suse Botelho da Silva, Daiana de Souza, and Liziane Dantas Lacerda
Food and Chemical Engineering, Polytechnic School, Unisinos University, São Leopoldo, RS, Brazil
This chapter presents the current and potential applications of chitosan and its derivatives
in food processing. Chitosan is a polysaccharide obtained from partial or full deacetylation
of chitin. Having highly reactive amino and hydroxyl groups, its structure imparts
antioxidant, antimicrobial, and chelating properties to this polymer. These characteristics
are associated with good film‐forming ability, biodegradability, and nontoxicity of chitosan,
and have allowed the development of a wide range of food applications. In this chapter,
recent works are presented as examples of different ways in which chitosan can be applied,
focusing on uses as a food additive and functional ingredient, material for active packaging,
enzyme immobilization, encapsulation of probiotics and bioactive compounds, and for
removal of toxic substances from food. Research on new chitosan derivatives, combination
with other materials, and the advances made by nanotechnology are reflected in these
applications and have allowed for improved materials and high‐performance processes.
13.1 Introduction
Chitosan is a polymer of D‐glucosamine and N‐acetyl‐glucosamine linked through β‐(1–4)
glycosidic linkages, obtained from partial or full deacetylation of chitin with a molecular
weight typically ranging between 16 and 1,000 kDa or even more [1]. Oligomers obtained
from the depolymerization of chitosan, having a degree of polymerization less than 20
(MW < 4 kDa), are recognized as chitooligosaccharides (COS) [2, 3]. Except for cellulose,
chitin is the most abundant polysaccharide in nature, being present in the exoskeleton of
crustaceans and insects, and also in mollusks, annelids, fungi, algae, and protozoa.
Commercially available chitosan is obtained from crustacean chitin processing waste from the
food industry in countries such as USA, Japan, India, and Brazil [4, 5]. The industrial produc-
tion of chitosan contributes, therefore, to minimizing waste generation and to produce a
high‐value added material for the food industry itself [2].
Biodegradability, biocompatibility, and nontoxicity are characteristics attributed to
chitosan; in addition, this polysaccharide has a GRAS (generally recognized as safe) status
by the FDA (Food and Drug Administration) [6, 7]. Chitosan is also approved by the
European authorities and by the International Organization of Vine and Wine to be used as
a fining agent and antimicrobial in wines [8]. Recently, the toxicity of shrimp and fungal
chitosan and nanostructures (5–13 nm) obtained hereof was assessed by brine shrimp and
rat bioassays [9]. The brine shrimp bioassay technique showed that no toxic compounds
were detected for both chitosan and nano‐chitosan tested at concentrations up to 15,000 ppm.
Adding chitosan and nano‐chitosan in the rat diet (100 and 200 mg kg−1 BW rat) showed
no significant changes in blood biochemical profile, oxidative stress parameters, and in the
histopathology of liver, kidney, and stomach, when compared with the control group.
Chitosan has a good film‐forming ability and exhibits a significant pH‐sensitive behavior
due to the presence of amino groups in its structure. At pH lower than 6.2, the protonation
of amino groups occurs, imparting a cationic character to chitosan, that allows its solubility
[4]. Apart from highly reactive amino groups in C‐2, chitosan also has hydroxyl groups
(primary hydroxyl at C‐6 and secondary hydroxyl at C‐3) that are also easily modified [5].
This peculiar structure makes chitosan a remarkable material to be used as an antioxidant,
antimicrobial, or adsorbent agent, and for encapsulation or immobilization of bioactive
compounds in food processing and packaging applications. Even so, in recent years, modi-
fied derivatives from chitosan such as grafted and cross‐linked structures have been devel-
oped for improving its properties [5, 6, 10].
This chapter will present the current and potential applications of chitosan and its
derivatives in the food processing and food packaging area. These applications involve the
use of chitosan as an antioxidant, antimicrobial, or stabilizing agent in product formulations
and in active packaging; as a nutritional ingredient in functional foods; as a chelating or
adsorbent material for metals or toxic substances; and also as a material for immobilization
of enzymes and for encapsulating of microorganisms and other bioactive compounds.
Target food
Chitosan or chitosan derivative Concentration Food deterioration reaction Main result Reference
Chitooligosaccharides (COS) 0.5% (w/w) Fish, meat, Maillard reaction Inhibition of the formation of AGEs in the [21]
added with transglutaminase sausage (harmful advanced product. Cross‐linking of proteins and
(TGase) glycation end glycosylation of glutamine induced by TGase
TGase/COS 1:1 products—AGEsa) and COS resulted in steric hindrance that
inhibited glycation.
Chitosan (Mw 650.00 kDa, 1 g/100 g total weight Bread Microbial growth Chitosan and COS showed an inhibitory effect [22]
DD 30.8%) basis against Bacillus cereus and Rhizopus sp.
COS (Mw 13 KDa, DD 54.8%) growth and delayed ropiness and fruity odor
in bread stored under 30°C for 4 days. Better
results were obtained with COS, resulting in a
shelf life extension of the bread in comparison
with the control.
Chitosan (Mw 32 kDa, DD 1% (w/w) chitosan + Minced Microbial growth Chitosan could control Listeria monocytogenes [23]
90.4%) from Aspergillus LBE at percentages catfish (pathogenic bacteria) and lipid oxidation in fish mince, and its
niger and Goji berry (Lycium of 0.2 and 0.4% and lipid oxidation efficacy was higher when LBE was added at
barbarum) extract (LBE) (w/w) 0.2 and 0.4% (w/w). The sensorial assessment
of treated minces indicated better acceptance
of samples with chitosan and LBE.
Chitosan (not defined Mw 0.2 g/L Wine Microbial growth Chitosan induced a reduction of viability of [8]
and DD) (acetic acid bacteria) acetic acid bacteria immediately after its
addition, showing the similar effectiveness of
SO2 in wine preservation.
Chitosans (MW 310 kDa, DD 100 mg chitosan/kg Mayonnaise Lipid oxidation Retarding of lipid oxidation of the mayonnaise [24]
77.7%) and (MW 123 kDa, product for 63 days under accelerated storage.
DD = 83.2%) Improvement of the sensorial attributes (taste
and odor) compared to the control.
Chitosan (Mw 160 kDa, 95% 0.5% (w/v) chitosan, Stewed pork Microbial growth, lipid Reduce the values of pH, lipid oxidation markers [25]
DD) and aqueous extract 5% (v/v) extract (dipping oxidation, (TVB‐N, peroxide value, TBA value) and total
GOG (ginger, onion, and GOG solution) discoloration, and bacterial count during 12 days of refrigerated
garlic) flavor deterioration storage. Reduction of off‐flavors and better
acceptance than control.
Note: aAGEs: advanced glycation end products; COS: chitooligosaccharides; DD: degree of deacetylation; LBE: Lycium barbarum extract (Goji berry) extract; TBA: 2‐thiobarbituric acid;
TGase: transglutaminase; TVB‐N: total volatile basic nitrogen.
13.2.1 Antioxidant
Antioxidants are compounds capable of retarding or inhibiting oxidative processes in
food, and thereby they can stabilize the color and flavor of food and protect sensitive
nutrients from oxidation. Most antioxidants currently used in the food industry are low‐
molecular‐weight compounds, which may eventually have their efficiency reduced by
processes such as heating or leaching [11]. Antioxidant polymers such as chitosan, on the
other hand, tend to be more resistant to this type of degradation or loss [11], although
they are not considered a practical antioxidant ingredient due to their typical low
solubility [10].
The antioxidant property of chitosan is attributed to its ability to chelate metal ions and
to act on free radicals [12]. In both, this activity is due to the presence of amino group and
hydroxyl groups in the chitosan structure that imparts a strong hydrogen‐donating ability
of this polymer [13, 14]. The chelating activity involves binding of metal ions with the
hydroxyl group at the C6 and with the amino at the C2 of chitosan [13, 15] preventing
metal ions from initiating lipid peroxidation. Similarly, it occurs with the free radicals
which react with active hydrogen atoms of the hydroxyl or amino groups of chitosan to
form a very stable macromolecular radical and, consequently, block the advance of the
oxidative reactions [14]. The antioxidant activity of chitosan is therefore related to the
structural characteristics of this polymer, including its molecular weight, deacetylation
degree, and origin [6].
Antioxidant activity increases with the deacetylation degree and decreases with the
molecular weight, in both cases due to the increase of its solubility [13]. The lower the
molecular weight, the more flexible the chain, so that amino and hydroxyl groups have less
impediment to react with free radicals or to chelate metals, and chitosan exhibits a higher
antioxidant activity [6]. The number of free amino groups (deacetylation degree) itself also
exerts great importance on the antioxidant activity of chitosan, as shown by Yen et al. [13]
working with crab chitosan with different deacetylation degrees (DD). These authors
showed that chitosan with a higher DD had more amino groups to inhibit the oxidation both
via radicals scavenging and metal chelating.
In order to improve the antioxidant activity, especially for chitosan of medium and
high molecular weights, several chemical and enzymatic modification can be per-
formed. These modifications aim to increase the solubility of chitosan and/or improve
the free radical scavenging activity of the structure and conjugation of small antioxi-
dant molecules [6]. Grafting chitosan with flavonoids and phenolic acids using oxida-
tive enzymes is a nontoxic and eco‐friendly alternative for functionalization chitosan
to improve its antioxidant activity [10, 11, 16–18]. For instance, using this approach,
Božič et al. [10] functionalized chitosan with caffeic acid (CA) and gallic acid (GA)
using laccase from Trametes versicolor. In this reaction, the oxidative enzyme con-
verted phenols in reactive o‐quinones which reacted with primary amino groups of
chitosan producing phenolic acid chitosan derivatives with higher antioxidant activity
compared to untreated chitosan. Since the solubility and the protonation of amino
groups of chitosan are dependent on pH, derivatives produced under different pHs
undergo different functionalization mechanisms and intensity of antioxidant activity,
as shown by the authors. The maximum activity was found for chitosan modified at
pH 4.5 [10].
Food Applications of Chitosan and its Derivatives 319
13.2.2 Antimicrobial
The electrostatic interaction between the polycationic structure of chitosan and the predomi-
nantly anionic components (lipopolysaccharides, peptidoglycan, and teichoic acid) of the bac-
terial surface is the main factor responsible for its antimicrobial activity [6]. Chitosan
antimicrobial activity is, therefore, influenced by the deacetylation degree, and also by origin
and degree of polymerization of the polymer, and by the environmental conditions, especially
pH [6, 19, 20]. The higher the degree of deacetylation and the lower the pH, the greater the
number of amino groups likely to be positively charged to interact with the cell surface of the
microorganisms [6, 20]. The degree of polymerization of chitosan affects the antioxidant activ-
ity of different ways, depending on whether the bacteria are Gram‐positive or ‐negative [19].
Recently, Verlee et al. [1] clarified some aspects regarding the antioxidant activity of
chitosan and highlighted the type of microorganism also as an important factor to understand
the mechanism behind the antimicrobial activity of chitosan. According to these authors,
there are four types of microorganisms regarding its susceptibility towards chitosan and,
consequently, four different mechanisms of action: Gram‐positive bacteria, Gram‐negative
bacteria, chitosan‐sensitive fungi, and chitosan‐resistant fungi.
In Gram‐positive bacteria, the main mechanism of action of chitosan is through
electrostatic interaction with teichoic acids in the peptidoglycan layer, leading to disruption
of different functions and resulting in cell death [1]. In this case, the antimicrobial activity
tends to enhance on increasing the molecular weight of chitosan, suggesting that high‐
molecular‐weight chitosan forms films around the cell that inhibit the absorption of
nutrients [1, 19]. In Gram‐negative bacteria, two different mechanisms of action of chitosan
are suggested: chelation with divalent cations under acidic pH decreasing the stability of
the membrane and the nutrient uptake; and, electrostatic interactions with the
lipopolysaccharide at the outer membrane enabling chitosan to penetrate towards the inner
membrane, resulting in cell leakage [1]. In this case, low‐molecular‐weight chitosan
penetrates more easily into Gram‐negative bacteria, binding to DNA, preventing
transcription and translation, and causing disturbances in cellular metabolism [19]. In
chitosan‐sensitive fungi, chitosan affects the cell membrane via electrostatic interactions
with the negatively charged phospholipids, causing the disruption of the membrane, and
further penetrates in the cell and inhibits DNA/RNA synthesis [1]. For chitosan‐resistant
fungi, chitosan is unable to permeabilize the cell membrane which has a high amount of
unsaturated fatty acids and remains at the outer surface [1].
The chitosan modifications that increase amino groups and decrease the molecular
weight frequently enhance its antimicrobial activity [6]. Grafting chitosan with groups with
antimicrobial properties, such as phenolic compounds, also enhance the antimicrobial
activity of the polymer as demonstrated by Božič et al. [10]. These authors functionalized
chitosan with CA and GA and reported an increase in activity against Escherichia coli and
Listeria monocytogenes compared to untreated chitosan.
crystals; and control the release of flavors [26]. Among them, chitosan is considered as an
excellent emulsifier capable of stabilizing oil‐in‐water (O/W) emulsions, which is very
useful in the development of products as dairy desserts, ice creams, and sauces [27]. This
property results from the protonation of the amino groups of chitosan in acidic medium,
which confers an amphiphilic character to the molecule, allowing the adsorption at the oil/
water interfaces and the formation of the emulsion by the decrease of the interfacial tension
[6]. In this sense, the emulsifying ability of chitosan is highly dependent on its molecular
weight and degree of deacetylation [6].
Souza Soares et al. [27] evaluated the performance of chitosan as a thickener and
emulsion stabilizer. O/W emulsions were prepared from 0.1% (m/v) chitosan dispersions
in lactic acid solutions (pH 3.0, 3.5, or 4.0) with Tween 20 (1% v/v) and sunflower oil (9:1).
Emulsions of lactic acid solutions with Tween 20 and sunflower oil was used as a control.
The results showed chitosan as an effective thickener agent even at only 0.1% (m/v),
improving both the size distribution and average size of droplets in emulsions. The surfaces
of oil droplets in emulsions containing chitosan showed highly positive ζ‐potential values,
which is important to sustain the emulsion stability. The average diameter and the ζ‐
potential values did not show significant differences after storage at 25°C for 7 days [27].
Another important aspect of this work is that chitosan has been dispersed in lactic acid
rather than commonly used acetic acid. This new approach would allow producing chitosan
emulsions without the off‐flavor characteristic of acetic acid.
cells and organs against these deleterious effects; however, an imbalance between the
antioxidant protection system and ROS leads to oxidative stress, which is associated with
various chronic diseases [34].
Halder et al. [32] evaluated the bioactivity of COS from fermented shrimp shell hydro-
lysates and chitosan (60% deacetylated) in male albino rats, regarding hypocholester-
olemic, antioxidant, and prebiotic effects. The levels of triglycerides, total cholesterol
(TC), low‐density lipoprotein cholesterol (LDL cholesterol), and atherogenic index (TC/
high‐density lipoprotein cholesterol) were significantly lower in animals fed on chitosan or
COS compared to animals that received the same cholesterol‐rich feeding but did not
receive these chitosan supplements. The antioxidant activity and the resistance to lipid
peroxidation were higher due to COS than to chitosan feeding. COS also selectively stimu-
lated the growth of health beneficial microbes (Bifidobacterium spp. and Lactobacillus
spp.) and inhibited pathogenic bacteria, demonstrating, therefore, prebiotic effect. The
same observation was not found for chitosan.
Long et al. [35] performed a study where orally COS (500 mg kg−1) was given to normal
mice and colitis‐treated mice. The results showed that COS could act as prebiotics by
increasing the levels of Bacteroidetes and Actinobacteria phyla, and common health‐
promoting bacteria as Lactobacillus spp. and Bifidobacterium spp. and inhibiting the
growth of Firmicutes and Proteobacteria phyla, and potential pathogens in both normal and
colitic mice. Additionally, the oral intake of COS induced the enhancement of the
concentrations of short‐chain fatty acids (SCFAs) in the colon. These findings suggest that
COS administration have beneficial effects on the intestinal tract and can protect against
the development of colitis [35].
Regarding chitosan health claims, the European Food Safety Authority (EFSA) Panel on
Dietetic Products, Nutrition and Allergies provided a scientific opinion in 2011 [36].
According to them, there is sufficient evidence between cause and effect to attribute to
chitosan the claim of Maintenance of normal blood LDL‐cholesterol concentrations,
informing that 3 g of chitosan should be consumed to obtain the claimed effect. Nevertheless,
for claims of reduction in body weight, reduction of intestinal transit time, and reduction of
inflammation, the Panel concluded that a cause‐and‐effect relationship has not been
established respecting to the consumption of chitosan.
comparable to those values found in high‐density polyethylene films. The values of the
percentage of elongation are the main concern, which are far from the desirable values
found for synthetic polymers [39].
Blending chitosan with other polymers had been a strategy to obtain chitosan‐based
films and coatings with enhanced mechanical and physical properties [42]. For blending
chitosan, some methods are available such as solvent blending, extrusion blending, and
reactive extrusion blending, and are reviewed in detail by van den Broek et al. [61]. These
authors emphasize that for increasing the antimicrobial effect of chitosan and improving
the mechanical properties of the material at the same time, a good interfacial adhesion
between chitosan and the polymer matrix is needed. In this sense, the reactive extrusion
blending at which the chitosan and polymer matrix will be covalently connected is a better
option of the blending method. Another advantage of the reactive extrusion is that it is
possible to direct the orientation of functional groups of chitosan to the outside of the
polymeric matrix, improving in this way the antimicrobial activity of the film [61].
The study of combinations of polysaccharides with other materials to improve the barrier
and mechanical properties is a field with continuous advancements and may allow
biopolymer matrices to replace synthetic polymers [39]. Indeed, the high hydrophilicity of
the polysaccharides is one of their major limitations, because as soon as the films enter in
contact with water, they undergo changes in their physical aspect and properties, which
also compromise their acceptance. Moreover, the disintegration of the films could also hap-
pen. For example, the incorporation of the hydrophobic poly(ε‐caprolactone) to the chi-
tosan matrix and the use of glacial acetic acid may be useful tools to prevent undesirable
swelling in contact with food [62].
Recently, the application of nanocomposite technology has been proven as a promising
option to improve the mechanical properties of chitosan films [44, 63, 64]. With the aim
focused on the development of a composite‐improved film, chitosan was incorporated with
montmorillonite (MMT). Film thickness, water vapor permeability, and mechanical and
optical properties were investigated. MMT could enhance water vapor permeability and
mechanical properties of the chitosan‐based films [63]. In another study [64], chitosan/
nanoclay nanocomposite active films containing three different levels of sodium MMT
(1, 3, and 5% w/w based on chitosan) and Silybum marianum L. extract (SME) (0.5, 1, and
1.5% v/v) were prepared. The results showed that water vapor permeability and solubility
of films reduced significantly by incorporation of MMT and SME. The tensile properties
of the prepared nanocomposites indicated an improvement in the mechanical properties in
comparison to control chitosan film, which can be explained by the formation of exfoliated
structure. Improved elastic modulus clearly demonstrates the reinforcing effects of the
nanoparticles on the films, which could occur as a result of strong hydrogen bonding
between MMT and chitosan [64].
A sodium lactate–loaded chitosan–polyvinyl alcohol/montmorillonite (NaL‐CS/PVA/
MMT) barrier film with antibacterial activity was developed by the coating method [65].
An intercalated structure was achieved for the CS/PVA/MMT film and the interfacial
interactions among CS, PVA, and MMT were intermolecular hydrogen bonds. An
appropriate increase of MMT contents (15 wt% and below) could achieve a remarkable
enhancement in TS and Young’s modulus, meanwhile, the water vapor, oxygen, and carbon
dioxide barrier properties of the films were also significantly improved. The NaL‐CS/PVA/
MMT film exhibited good antibacterial activity against E. coli and well‐controlled release
324 Chitin and Chitosan: Properties and Applications
Film‐forming Molecules or
solution extracts tested Concentrations Antioxidant activity Antimicrobial activity against Effects on film properties Reference
Chitosan Eucalyptus globulus 0, 1, 2, 3, and Significantly enhanced Escherichia coli, Staphylococcus Significantly decreases water [67]
(2% w/v) (EG) essential oil 4% (v/v) with increasing EG aureus, Pseudomonas solubility
essential oil aeruginosa, Candida albicans,
concentration and Candida parapsilosis;
enhanced with increasing EG
essential oil concentration
Chitosan Spirulina extract (SE) 0, 2.5, 5, 10, 15 Significantly enhanced E. coli, S. aureus, P. aeruginosa, The incorporation of SE into [59]
(2% w/v) and 20% (w/v) with increasing SE L. monocytogenes, crab chitosan film led to
concentration Salmonella typhimurium, considerable improvement
Bacillus subtilis, B. cereus; in mechanical and barrier
films exhibited concentration‐ properties of the resulting
dependent antimicrobial film.
activities against bacteria.
Chitosan Turmeric ethanol 67% (v/v) ‐ TEE incorporated chitosan film The addition of turmeric to [68]
(2% w/v) extract (TEE) reduced the counts of S. chitosan film significantly
aureus and Salmonella sp. increased the TS of the film
significantly, compared to and improved the
native chitosan film during ultraviolet‐visible light
the 3 h exposure period. barrier of the film.
Chitosan Epigallocatechin 1% (w/v) EGCG grafted‐chitosan The antibacterial activity of the EGCG‐grafted chitosan [69]
(1% w/v) gallate (EGCG) showed higher EGCG‐grafted chitosan was showed higher antioxidant
antioxidant activity increased compared to pure activity than blank
than native chitosan. EGCG or native chitosan chitosan.
against S. aureus and
Pseudomonas sp.
Chitosan Ziziphora 0 and 1% v/w, The highest antioxidant S. aureus, B. subtilis, B. cereus, ZEO and GSE reduces [70]
(2% clinopodioides separately and activity was found in L. monocytogenes, S. swelling index, TS,
w/v)— essential oil (ZEO) in chitosan enriched with typhimurium, E. coli puncture force, and
Gelatin Ethanolic grape seed combination 1% (v/w) ZEO in O157:H7 puncture deformation of
(3% w/v) extract (GSE) combination with 1% chitosan and gelatin films.
(v/w) GSE
(Continued )
Film‐forming Molecules or
solution extracts tested Concentrations Antioxidant activity Antimicrobial activity against Effects on film properties Reference
Chitosan Thinned young 0.25, 0.50, 0.75, The antioxidant activity YAP‐chitosan films have A significant increase in the [52]
(2% w/v) apple polyphenols and 1.0% of 1.0% (w/v) YAP‐ concentration‐dependent thickness, density, swelling
(YAP) (w/v) chitosan films antimicrobial activities against degree, solubility, and
increased nearly bacteria (E. coli, L. opacity of chitosan film,
threefold compared to monocytogenes, and S. aureus) but the water content,
the control films. and molds (Colletotrichum water vapor permeability,
fructicola, Botryosphaerial and mechanical properties
dothidea, and Alternaria of the film were decreased.
tenuissima) but no activity
against yeasts (Saccharomyces
cerevisiae, baker’s yeast, and
tropical candida)
Chitosan Ellagic acid (EA) 0.5, 1.0, 2.5 and Concerning the chitosan/ Chitosan/EA films totally These set of films display high [56]
(1% w/v) 5.0% (weight EA films, DPPH a
inhibited the growth of both mechanical properties,
percentage of scavenging activity Gram‐positive (S. aureus) and thermal stability up to 215
chitosan) increased from 0.0 to Gram‐negative (P. aeruginosa) and 220°C, UVA‐ and
about 28% with the food pathogenic bacteria due UVB‐barrier properties,
increase of EA content to chitosan antimicrobial moderate water vapor
from 0.5 to 5.0 wt% effectiveness. permeability.
Chitosan Honeysuckle flower 5, 10, 20, and The incorporation of HFE Film solution created a larger Compared with the chitosan [51]
(4% w/v) extract (HFE) 30% (weight enhanced the inhibitory zone (127.36 mm ) 2
control film, the chitosan/
percentage of antioxidant activity of against E. coli compared with HFE films had darker
chitosan) the films. 36.77 mm without HFE.
2
appearance, higher water
solubility, and lower TS
and elongation at break.
Chitosan Apple peel (APP) 0.25, 0.50, 0.75, Incorporating of APP E. coli, B. cereus, S. aureus, and Thickness, density, swelling [46]
(2% w/v) and 1.0% significantly improved S. typhimurium; films degree, solubility, and
(w/v) the antioxidant activity exhibited concentration‐ opacity of film were
of chitosan‐based films, dependent antimicrobial significantly increased, but
and the antioxidant activities against bacteria. the water content and
activity was enhanced vapor permeability were
with increased decreased.
concentration of APP.
Note: aDPPH: 1,1‐diphenyl‐2‐picrylhydrazyl radical; bTS: is tensile strengths; cEAB: elongations at break.
Application
Film‐forming solution Food tested technique Molecules or extracts tested Concentration Main result Reference
Chitosan (1.5% w/v) Fresh red drum Coating Grape seed extract (GS) tea 0.2% w/v of each Both treatments extend the shelf [72]
(Sciaenops polyphenols (TP) extract, separately life of fish by 6–8 days compared
ocellatus) fillets with the control group.
Chitosan (1% w/v) Eggs Coating Lysozyme (L) 0, 10, 20, and 60% 20% w/v L–chitosan coating [73]
(dry weight maintained the internal quality
lysozyme/dry of eggs 3 weeks longer than
weight chitosan) observed for uncoated control
Chitosan (2% w/v) Lean pork slices Coating Clove oil (CO)— 0.05% v/v (CO) Chitosan + CO and [74]
ethylenediaminetetraacetate 10 mM (E) Chitosan + CO + E coatings have
(E) a good potential for
antimicrobial purposes in
refrigerated pork as active
packaging materials.
Chitosan (1.5% w/v) Strawberry Coating Nisin (NS) natamycin (NT) 1% w/v of each, Antimicrobial agents incorporated [75]
pomegranate extract (PE) separately in chitosan coating improved the
grape seed extract quality and extended the shelf
life of fresh strawberry up to 30
days
Chitosan (1% w/v), Shrimp Coating ‐ ‐ A shelf life of <14 days was [76]
gelatin (3% w/v), observed for the coated shrimp,
70:30 as compared to only <8 days for
the control.
Chitosan (1% w/v) Barracuda fish Film Ginger essential oil (GEO) 0.1, 0.2, 0.3% v/v Sensorially, chitosan‐GEO film [41]
wrapping wrapped sample was acceptable
till the end of storage for 20 days
as compared to 12 days for
unwrapped control and fish
steak packed in EVOH film.
Chitosan (2% w/v) Shrimp Film Z. clinopodioides essential oil 1% v/v (ZEO and The group treated with chitosan-ZEO [48]
wrapping (ZEO) pomegranate peel PPE); 1% w/v 1% + PPE 1% + CN 1% had the
extract (PPE) cellulose (CN), separately best antibacterial effectiveness
nanoparticle (CN) and in and also the highest organoleptic
combination scores after 11 days.
(Continued)
Application
Film‐forming solution Food tested technique Molecules or extracts tested Concentration Main result Reference
Chitosan (1.25% w/v) Salmon (Salmo Film Grape seed extract and With and without This study reports the beneficial [81]
salar) wrapping carvacrol microcapsules GSECM effects of the chitosan film with
(GSECM) GSECM on salmon samples
stored at 5°C, with the samples
remaining acceptable between
the fourth and seventh days of
storage.
Chitosan (1% w/v) Minced beef Film Kombucha tea (KT) 1, 2, and 3% (KT) The shelf life of stored minced beef [55]
wrapping (w/w) packaged in chitosan/KT can be
extended up to 6 days. The
Staphylococcus growth counts
were also inhibited by packaging
in chitosan containing KT.
Chitosan (2% w/v) Cherry tomato fruit Film Titanium dioxide (TiO2) 1% The quality changes of climacteric [50]
wrapping nanopowders (T) fruit are known to be triggered
and regulated by ethylene;
hence, the chitosan‐T
nanocomposite film which
exhibited ethylene
photodegradation could delay
the ripening process and extend
the storage life of the tomato
fruit
Nanomontmorillonite‐ Minced camel’s Film Z. clinopodioides essential oil 0.5, 1, and 2% w/v An increase in the shelf life of [47]
chitosan (MMT meat wrapping (ZEO) Ficus carica extract (ZEO) alone or in minced camel meat by active
0.1% w/v chitosan (FCE) combination of Chitosan‐MMT films was
2% w/v) 1% v/v (FCE) observed.
showed that chitosan is considered an excellent supporting matrix for the immobilization
of pectinolytic enzymes such as polygalacturonase pectin lyase, and pectin methylesterase
[89]. Khoshnevisan et al. [90] provided a detailed overview of the applications and recent
advances of immobilization of cellulase onto different functionalized magnetic nanoparticles
(MNPs). For instance, the synthesis of chitosan‐MNPs was carried out by adsorption or
covalent binding after activating the hydroxyl groups of chitosan with carbodiimide,
methanediimine, or cyanuric chloride.
Some recent studies focus on using carboxypeptidase A (CPA) insoluble derivatives for
producing hydrolysates of food proteins mainly from milk and by‐products for obtaining
food preparations with low allergenicity and standardized for phenylketonuric consumers
[91]. Manzo et al. [91] developed strategies for effective multipoint covalent immobilization
of CPA metalloenzyme on chitosan beads by N‐alkylation. Urrutia et al. [92] reported a
complete analysis of the use of hydrophobic chitosan as a support for the immobilization
of lipases and their further application in the selective hydrolysis of fish oil. According to
these authors, the length of alkyl chains linked to chitosan is a critical factor in the final
behavior of immobilized lipases, mainly affecting biocatalyst activity and stability.
Chitosan and modified chitosan have a strong affinity towards immobilization of biomol-
ecules, which have demonstrated also a huge potential in providing a platform for the fabri-
cation of biosensors. A biosensor is an analytical device that uses specific biochemical
reactions mediated by a biological recognition element (an enzyme) immobilized onto a
signal transducer. Quantification limit (LOQ), low limit of detection (LOD), response time,
repeatability, reproducibility, and accuracy are other factors responsible for the characteriza-
tion of biosensor performance [93, 94]. Neusakumar et al. [95] reported a lactic acid biosen-
sor by immobilizing lactate dehydrogenase (LDH) with chitosan onto ZnO nanorods. The
one‐dimensional ZnO nanorods exhibit high surface area and act as an immobilization
matrix for LDH. This lactate detecting biosensor was shown to have an LOD of 4.73 nmol L−1,
LOQ of 15.75 nmol L−1, the response time of < 1 s, sensitivity of 1.832 μA μmol−1 L, and
linearity of 0.2–0.8 μmol L−1. This sensor was stable up to 23 days under dry conditions at
room temperature.
of amine groups on D‐glucosamine units [4, 5]. This characteristic leads to an interesting
property to chitosan, which is dependent on the protonation of amino groups and results
in an excellent mechanism which can be used to release the encapsulated active com-
pounds as pH changes [4, 99].
Several works are found in the literature describing the preparation of encapsulating
systems made only or in combination with chitosan. These systems are obtained using
primarily mechanical techniques as emulsification, spray drying, spray chilling, fluidized‐
bed coating, centrifugal extrusion, electrospinning/electrospraying, pressure extrusion, and
hot‐melt extrusion, or essentially chemical techniques as ionic gelation, simple or complex
coacervation, and coating with solvent evaporation [97–101]. The more advanced
procedures involve the addition of a cross‐linker (tripolyphosphate, transglutaminase, or
genipin) during the last step of coacervation [96]. Even using merely mechanical techniques
to produce the beads, chemical modifications of the polymer may eventually be induced, or
further steps may be added for that purpose. This is especially true for charged polymers or
having chemically active functional groups.
In ionic gelation, a hydrocolloid with a bioactive compound are dripped or atomized into
an ionic solution under constant stirring, obtaining spherical gel structures [99]. For
chitosan‐based systems, typically chitosan is dissolved in diluted acetic (or other) acid
solution in order to induce the protonation of amino groups and exhibit its cationic character
followed by dripping in an anionic solution. In coacervation, the formation of complex
structures results from liquid–liquid phase separation from an initial suspension followed
by deposition and solidification of the dense coacervate phase surrounding the core material
[26, 100]. The simple coacervation result from the presence of only one polymer or
hydrocolloid in the initial suspension, while for complex coacervation it is necessary to
have two or even more hydrocolloids and polymers in an initial suspension mixture [96].
The formation of the coacervate phase is due to changes in the physical parameters
(temperature, pH, or composition) of the initial suspension leading to the separation of
incompatible polymer [100].
Chitosan is an extremely useful material in a chemical encapsulation process due to its
hydroxyl and amino‐reactive groups which can be used for modification of this polymer
and for obtaining complex structures [5, 102]. In particular, the protonation of the amino
groups of chitosan in low pH imparts a polycationic character to the polymer and allows
the formation of polyelectrolyte complexes of chitosan (PEC) prepared with natural,
naturally modified or synthetic polyanions [103]. In ionic gelation, the electrostatic
interaction occurs between the amine group of chitosan and a negatively charged group of
polyanion such as tripolyphosphate [104]. In these approaches, not only electrostatic
interactions between chitosan and anionic polymers are involved but also hydrogen bond-
ing and hydrophobic interactions [6].
Among the bioactive compounds which exhibit huge potential for use in nutraceutical
encapsulation systems are polyphenols and vitamins. Polyphenols are beneficial to human
health, however, they are highly unstable to conventional food process conditions and
frequently have an unpleasant taste [105]. So, encapsulation is an effective tool for
preserving the stability and ensuring bioactivity and bioavailability to these compounds, as
demonstrated by several authors [98, 105–110]. Vitamins, especially those that have
antioxidant potential as vitamin C or vitamin E, are susceptible to degradation and can have
their bioavailability limited by low absorption and degradation during delivery [111].
334 Chitin and Chitosan: Properties and Applications
Liposoluble vitamins are not readily soluble in water, which also restricts their use in some
food matrices. Encapsulating systems have been investigated and proposed to improve the
solubility, stability, and bioavailability of vitamins, but also to enhance proprieties as
antioxidant activity [112]. Several works involving chitosan‐based systems to encapsulate
polyphenols and vitamins have been published in recent years, showing the emergence of
this research area. Some examples of these works are shown in Table 13.4.
Even more than for bioactive chemicals, encapsulation systems are crucial for probiotics,
in order to ensure their viability during food processing and storage, as well for delivering
the microorganisms through the gastrointestinal tract [113, 114]. Probiotics are defined as
live microorganisms that when administered in adequate amounts in food confer a health
benefit on the host [115]. Thus, metabolically stable and active cells and ensured high
viability rates are the requirements to consider microorganisms as functional probiotics
[100]. In this sense, encapsulating systems capable to protect microorganisms in acid
gastric condition and their release under intestine conditions are particularly desired.
Several works have demonstrated the ability of chitosan in increasing the viability of
encapsulating microorganisms in food matrices under storage or under gastrointestinal
conditions [116–122]. Frequently in these works, the microorganism is initially encapsulated
using alginate, pectin, or another hydrocolloid, followed by application of chitosan as a
coating. Bepeyeva et al. [119] prepared gel beads by extrusion of amidated pectin in cal-
cium chloride solution, coated with or without chitosan. The authors reported the protec-
tive effect of chitosan in this encapsulating system. Capsules with chitosan provided
improved protection to Lactobacillus casei in simulated gastric juice resulting in high lev-
els of viable bacteria released in simulated intestinal juice [119]. Brinques and Ayub [116]
encapsulated Lactobacillus plantarum BL011 in beads of sodium alginate coated with chi-
tosan and verified that the viability of the encapsulated cells under refrigerated storage was
greatly enhanced compared to unencapsulated microorganisms. However, the viability was
drastically reduced in the simulated gastric medium [116]. These results demonstrate that
the effectiveness of encapsulation and the viability of microorganism depend on the encap-
sulation materials used beyond the selected strain [101]. Details of encapsulating tech-
nique, materials, capsule dimension, and main results for these works are shown in
Table 13.5.
Probiotic Encapsulating Encapsulation Capsule diameter Food Matrix Main Results Reference
microorganisms materials techniques
Lactobacillus casei Calcium alginate, Ionic gelation 71.5 ± 4.9 μm for uncoated Ice cream Chitosan and poly L‐lysine coatings [118]
ATCC 39392 and wheat, rice, and mediated by capsules, 94.58 ± 2.28 μm significantly improved the
Bifidobacterium high‐amylose corn emulsion and 95.6 ± 3.2 μm for encapsulated probiotic survival
adolescentis ATCC (Hylon VII) starches, followed by chitosan and poly L‐lysine in ice cream after 100 days of
15703 chitosan, and poly coating coated capsules, storage at ‐30°C.Encapsulated
L‐lysine respectively probiotic showed no significant
effect on typical sensorial
characteristics of ice cream.
L. casei NCIMB Pectin and chitosan Ionic gelation 2.57 ± 0.02 to 3.50 ± 0.05 mm No food Coating with chitosan effectively [119]
30185 (PXN37) mediated by for uncoated capsules and matrix protected the bacterial cells in
extrusion 0.236 ± 0.061 mm for tested pectin capsules from the acid in
followed by coating thickness the simulating gastric juice, while
coating capsules without chitosan
coating had limited protection.
Lactobacillus Chitosan, Ionic gelation Microparticles (by nozzle‐ No food The capsules were stable at the [120]
rhamnosus GG carboxymethyl‐ mediated by spraying): 5 μm matrix gastric pH conditions (pH 2.4)
LMG 18243 cellulose, and extrusion (syringe macroparticles (obtained tested and swell considerably at higher
genipin or nozzle sprayer) with a syringe): 2 mm pHs, close to intestine pH
followed by (pH 7.4).
coating
Lactobacillus Sodium alginate, Ionic gelation 1,302–1,335 μm No food Survival of encapsulated [121]
plantarum LAB12 xanthan gum, and mediated by matrix microorganism at pH 1.8 with
chitosanβ‐ extrusion tested facilitated release at pH
cyclodextrin as a followed by 6.8.Minimum loss of the bacterial
supplement coating viability at 75 and 90°C and
survival over cold storage (4°C)
for 4 weeks.Cholesterol‐lowering
effects of β‐cyclodextrin‐
supplemented beads were
demonstrated and
cytocompatibility was assessed
in vitro, confirmed nontoxicity.
are strongly toxic to humans and animals. These toxins can contaminate cereal grains and
other feedstuffs generating global concern [124]. When animals ingest contaminated feed,
mycotoxins are metabolized and transferred to food products, such as milk or meat, thereby
becoming a risk to human health [127]. There are more than 300 mycotoxins identified
worldwide, and the most monitored have been aflatoxin, ochratoxins, zearalenone,
trichothecenes, and fumonisins [124].
Some studies report the use of chitosan as an adsorbent to sequester mycotoxins.
Experimental data are frequently tested to fit Langmuir, Freundlich, and Hill models.
Quintela et al. [128] reported that chitosan could efficiently bind ochratoxin in wine and
improve wine safety. Zhao et al. [124] investigated the capability of chitosan salts and
cross‐linked chitosan beads for the adsorption of multiple mycotoxins including aflatoxin
B1 (AFB1), ochratoxin A (OTA), zearalenone, fumonisin B1 (FB1), deoxynivalenol
(DON), and T‐2 (T2) toxin. Among the tested adsorbents, cross‐linked chitosan–
glutaraldehyde complex presented the highest adsorption capability for AFB1 (73%),
OTA (97%), ZEN (94%), and FB1 (99%), but no obvious adsorption for DON and T2
(< 30%). Luo et al. [129] prepared a magnetic chitosan adsorbent combining Fe3O4 par-
ticles and chitosan using Triton X‐100 as an emulsifier and evaluated the effective
absorption of patulin, a mycotoxin produced by certain fungi on harvested fruits. These
authors reported patulin adsorption in kiwi fruit juice without any impact on the flavor
and quality of treated juice [129].
Besides mycotoxins, heavy metals are also of great concern for food safety. Lead (Pb) is
one of the most toxic heavy metals which is the main source of contamination for humans
being found in water and food. Abou El Fadl [130] studied the use of natural alginate and
modified alginate/chitosan beads synthesized by graft copolymerization with acrylic acid
by gamma radiation as sorbents. The graft copolymer showed a greater ability to absorb Pb
ions compared to ungrafted beads.
In addition to the removal of toxic compounds in foods, some studies have focused on
the use of chitosan as a chelating agent for compounds associated with food spoilage and/
or have nutritional or health concerns, and also as a technological adjunct. The International
Union of Pure and Applied Chemistry (IUPAC) provides a detailed description of chela-
tion: it involves the creation or even the presence of two or more separate coordinate
bonds between a polydentate (multiple bonded) ligand and a single central atom. In the
majority of cases, the ligands are organic‐based compounds (namely chelants, chelators,
chelating agents)[126]. Examples of chitosan as chelating agent are discussed in the
following text.
Since ferrous ions are the most effective prooxidants in the food system, the high ferrous
ion chelating abilities of chitosan would be beneficial if they were formulated into food.
Fungal chitosans were prepared by alkaline N‐deacetylation of crude chitin, which was
obtained from air‐dried shiitake stripes. At 1.0 mg/mL, chelating abilities of chitosan on
ferrous ions were superior to 85% [131]. Zhu et al. [132] evaluated the antimicrobial, anti-
oxidant, and darkening inhibitory activities of Maillard Reaction Products (MRPs) pre-
pared from chitosan and xylose, and their effects on the preservation and quality of
semi‐dried noodles. The chitosan–xylose mixture in this study showed better antimicrobial
and antioxidant activities than chitosan alone. Additionally, MRPs exhibited copper‐
chelating property and good inhibitory effect on polyphenol oxidase, which is recognized
as the main factor leading to the darkening of Chinese and Japanese noodles [132].
Food Applications of Chitosan and its Derivatives 339
Lamas et al. [133] employed four different strategies to obtain egg‐derived products in form of
sticks with a reduced cholesterol content (use of β‐cyclodextrins, extraction of egg‐yolk granules,
bioconversion of cholesterol by cholesterol oxidase, and use of chitosan as a chelating agent).
Considering all the results obtained, egg‐derived yolk that was chelated by chitosan contained the
best balance out of the four methodologies employed, considering price, the ease of industrial
application, and the minor changes in color and consumer acceptability. The final reduction of
cholesterol content using this method was about 50%. Do Amaral [134] investigated the fat
absorption capacity of chitosan in low‐fat fresh pork sausage. The reduction of fat content to
levels of 5% was positively achieved with the incorporation of chitosan. The sensorial analysis
showed that panelists did not detect any significant difference in taste and unfavorable effect on
the sausage appearance as a consequence of chitosan addition and variation of fat [134].
Chitosan can also be used as an effective coagulating agent of suspended particles in
juices since the amino groups became protonated under acidic conditions, and chitosan
exhibits a typical behavior of a polyelectrolyte [135]. In this sense, clarification conditions
of apple juice were optimized using response surface methodology and also compared to
the traditional method of clarification during storage. Clarification of apple juice by using
chitosan (191.58 mg/100 mL) was suitable and easily applicable compared to the traditional
clarification method. Nevertheless, the high cost of chitosan may seem like a disadvantage
as compared to the traditional clarification agents [135].
13.8 Outlook
The development of applications for chitosan and its derivatives in the food area has been
extensively carried out in recent years, motivated by the investigation of new routes for
chitosan modification and preparation of blends, and also for the development of
nanotechnology. Although chitosan already exhibits widely recognized antimicrobial and
antioxidant properties, mainly due to the presence of reactive amino and hydroxyl groups,
these same groups have been the starting point for modification reactions, blending
processes, and obtaining complex structures with enhanced functional properties.
The encapsulation systems for bioactive compounds and active packaging have shown
great growth in recent years. Chitosan is a polymer that presents great compatibility with
several antimicrobial and antioxidant agents and can act as support for these compounds.
The application of nanotechnology to the production of chitosan nanocomposites is a very
promising area, which can solve some bottlenecks still present in packaging technology,
related to the mechanical and barrier properties of chitosan‐based films. In vitro assays for
evaluation of microbial or antioxidant properties or to test the efficiency of encapsulation
and delivering systems are very useful, but they need to be complemented with test
applications in food processing and under storage conditions. When applicable, delivering
through the gastrointestinal tract should be evaluated, as well as the bioactivity and
bioavailability of encapsulated compounds, even in simulated conditions.
The wide dissemination of chitosan applications by the food industry depends, in many
cases, on safety assessment and regulation. In addition, it is important to keep in mind that
the adoption of new technologies in substitution as conventional, needs besides the
comparable or better efficiency and environmental safety, also of economic viability. To
this end, the study and development of alternative routes and the optimization of processes
are increasingly necessary.
340 Chitin and Chitosan: Properties and Applications
References
1. Verlee, A., Mincke, S., and Stevens, C. V. (2017) Recent developments in antibacterial and antifungal
chitosan and its derivatives. Carbohydr. Polym., 164, 268–283.
2. Hamed, I., Özogul, F., and Regenstein, J.M. (2016) Industrial applications of crustacean by‐products
(chitin, chitosan, and chitooligosaccharides): A review. Trends Food Sci. Technol., 48, 40–50.
3. Lodhi, G., Kim, Y.‐S., Hwang, J.‐W., Kim, S.‐K., Jeon, Y.‐J., Je, J.‐Y., Ahn, C.‐B., Moon, S.‐H., Jeon,
B.‐T., and Park, P.‐J. (2014) Chitooligosaccharide and its derivatives: Preparation and biological appli-
cations. Biomed Res. Int., 2014, 654913.
4. Kardas, I., Struszczyk, M.H., Kucharska, M., Broek, L.A.M., van Dam, J.E.G., and Ciechańska, D.
(2013) Chitin and chitosan as functional biopolymers for industrial applications, in The European
Polysaccharide Network of Excellence (EPNOE): Research Initiatives and Results, Springer, pp.
329–373.
5. Botelho da Silva, S., Krolicka, M., van den Broek, L.A.M., Frissen, A.E., and Boeriu, C.G. (2018)
Water‐soluble chitosan derivatives and pH‐responsive hydrogels by selective C‐6 oxidation mediated
by TEMPO‐laccase redox system. Carbohydr. Polym., 186, 299–309.
6. Luo, Y., and Wang, Q. (2013) Recent advances of chitosan and its derivatives for novel applications in
food science. J. Food Process. Beverages, 1 (1), 1–13.
7. Gibson, G.R., Hutkins, R., Sanders, M.E., Prescott, S.L., Reimer, R.A., Salminen, S.J., Scott, K.,
Stanton, C., Swanson, K.S., Cani, P.D., Verbeke, K., and Reid, G. (2017) Expert consensus document:
The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on
the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol., 14 (8), 491.
8. Valera, M.J., Sainz, F., Mas, A., and Torija, M.J. (2017) Effect of chitosan and SO2 on viability of
Acetobacter strains in wine. Int. J. Food Microbiol., 246, 1–4.
9. Darwesh, O.M., Sultan, Y.Y., Seif, M.M., and Marrez, D.A. (2018) Bio‐evaluation of crustacean and
fungal nano‐chitosan for applying as food ingredient. Toxicol. Rep., 5, 348–356.
10. Božič, M., Gorgieva, S., and Kokol, V. (2012) Laccase‐mediated functionalization of chitosan by
caffeic and gallic acids for modulating antioxidant and antimicrobial properties. Carbohydr. Polym., 87
(4), 2388–2398.
11. Hashemi Gahruie, H., and Niakousari, M. (2017) Antioxidant, antimicrobial, cell viability and
enzymatic inhibitory of antioxidant polymers as biological macromolecules. Int. J. Biol. Macromol.,
104, 606–617.
12. Xie, W., Xu, P., and Liu, Q. (2001) Antioxidant activity of water‐soluble chitosan derivatives. Bioorg.
Med. Chem. Lett., 11 (13), 1699–1701.
13. Yen, M.‐T., Yang, J.‐H., and Mau, J.‐L. (2008) Antioxidant properties of chitosan from crab shells.
Carbohydr. Polym., 74 (4), 840–844.
14. Vinsova, J. and Vavrikova, E. (2011) Chitosan derivatives with antimicrobial, antitumour and antioxi-
dant activities – a review. Curr. Pharm. Des., 17 (32), 3596–3607.
15. Inoue, K., Baba, Y., Yoshizuka, K., Noguchi, H., and Yoshizaki, M. (1988) Selectivity series in the
adsorption of metal ions on resin prepared by crosslinking copper (II)‐complexed chitosan. Chemistry
Lett., 1281–1284.
16. Aljawish, A., Chevalot, I., Piffaut, B., Rondeau‐Mouro, C., Girardin, M., Jasniewski, J., Scher, J., and
Muniglia, L. (2012) Functionalization of chitosan by laccase‐catalyzed oxidation of ferulic acid and
ethyl ferulate under heterogeneous reaction conditions. Carbohydr. Polym., 87 (1), 537–544.
17. Aljawish, A., Chevalot, I., Jasniewski, J., Revol‐Junelles, A.‐M., Scher, J., and Muniglia, L. (2014)
Laccase‐catalysed functionalisation of chitosan by ferulic acid and ethyl ferulate: Evaluation of phys-
icochemical and biofunctional properties. Food Chem., 161, 279–287.
18. Rocasalbas, G., Francesko, A., Touriño, S., Fernández‐Francos, X., Guebitz, G.M., and Tzanov, T.
(2013) Laccase‐assisted formation of bioactive chitosan/gelatin hydrogel stabilized with plant poly-
phenols. Carbohydr. Polym., 92 (2), 989–996.
Food Applications of Chitosan and its Derivatives 341
19. Dutta, P.K., Tripathi, S., Mehrotra, G.K., and Dutta, J. (2009) Perspectives for chitosan based antimi-
crobial films in food applications. Food Chem., 114 (4), 1173–1182.
20. Kong, M., Chen, X.G., Xing, K., and Park, H.J. (2010) Antimicrobial properties of chitosan and mode
of action: A state of the art review. Int. J. Food Microbiol., 144 (1), 51–63.
21. Wang, J., Zou, L., Yuan, F., Lv, L., Tian, S., Li, Z., and Lin, H. (2018) Inhibition of advanced glycation
endproducts during fish sausage preparation by transglutaminase and chitosan oligosaccharides
induced enzymatic glycosylation. Food Funct., 9 (1), 253–262.
22. Rakkhumkaew, N. and Pengsuk, C. (2018) Chitosan and chitooligosaccharides from shrimp shell
waste: Characterization, antimicrobial and shelf life extension in bread. Food Sci. Biotechnol., 1–8.
23. Alsaggaf, M.S., Moussa, S.H., Elguindy, N.M., and Tayel, A.A. (2017) Fungal chitosan and Lycium
barbarum extract as anti‐Listeria and quality preservatives in minced catfish. Int. J. Biol. Macromol.,
104, 854–861.
24. Garcia, M., Silva, Y., and Casariego, A. (2014) Development of a mayonnaise with chitosan as natural
antioxidant. Emirates J. Food Agric., 26 (10), 835.
25. Cao, Y., Gu, W., Zhang, J., Chu, Y., Ye, X., Hu, Y., and Chen, J. (2013) Effects of chitosan, aqueous
extract of ginger, onion and garlic on quality and shelf life of stewed‐pork during refrigerated storage.
Food Chem., 141 (3), 1655–1660.
26. Gao, Z., Fang, Y., Cao, Y., Liao, H., Nishinari, K., and Phillips, G.O. (2017) Hydrocolloid‐food com-
ponent interactions. Food Hydrocoll., 68, 149–156.
27. de Souza Soares, L., de Faria, J.T., Amorim, M.L., de Araújo, J.M., Minim, L.A., dos Reis Coimbra,
J.S., de Carvalho Teixeira, A.V.N., and de Oliveira, E.B. (2017) Rheological and physicochemical
studies on emulsions formulated with chitosan previously dispersed in aqueous solutions of lactic acid.
Food Biophys., 12 (1), 109–118.
28. Harikrishnan, R., Kim, J.‐S., Balasundaram, C., and Heo, M.‐S. (2012) Dietary supplementation with
chitin and chitosan on haematology and innate immune response in Epinephelus bruneus against
Philasterides dicentrarchi. Exp. Parasitol., 131 (1), 116–124.
29. Hayes, M. and Tiwari, B. (2015) Bioactive carbohydrates and peptides in foods: An overview of
sources, downstream processing steps and associated bioactivities. Int. J. Mol. Sci., 16 (9),
22485–22508.
30. Patti, A.M., Katsiki, N., Nikolic, D., Al‐Rasadi, K., and Rizzo, M. (2015) Nutraceuticals in lipid‐
lowering treatment. Angiology, 66 (5), 416–421.
31. Prashanth, K. V, Harish Tharanathan, R.N., Harish Prashanth, K.V., and Tharanathan, R.N. (2007)
Chitin/chitosan: Modifications and their unlimited application potential—an overview. Trends Food
Sci. Technol., 18 (3), 117–131.
32. Halder, S.K., Adak, A., Maity, C., Jana, A., Das, A., Paul, T., Ghosh, K., Mohapatra, P.K. Das, Pati,
B.R., and Mondal, K.C. (2013) Exploitation of fermented shrimp‐shells hydrolysate as functional
food: Assessment of antioxidant, hypocholesterolemic and prebiotic activities. Indian J. Exp. Biol., 51
(11), 924–934.
33. Gibson, G.R. and Roberfroid, M.B. (1995) Dietary modulation of the human colonic microbiota:
Introducing the concept of prebiotics. J. Nutr., 125 (6), 1401–1412.
34. Fu, L., Wang, A., Lyv, F., Lai, G., Zhang, H., Yu, J., Lin, C.‐T., Yu, A., and Su, W. (2018) Electrochemical
antioxidant screening based on a chitosan hydrogel. Bioelectrochemistry, 121, 7–10.
35. Long, T., Yu, Z.‐J., Wang, J., Liu, J., and He, B.‐S. (2018) Orally administered chitooligosaccharides
modulate colon microbiota in normal and colitis mice. Int. J. Pharmacol., 14 (2), 291–300.
36. Authority, E.F.S. (2011) Scientific opinion on the substantiation of health claims related to chitosan
and reduction in body weight (ID 679, 1499), maintenance of normal blood LDL‐cholesterol concen-
trations (ID 4663), reduction of intestinal transit time (ID 4664) and reduction of inflammation (ID
1985) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFSA J., 9 (6), 1–21.
37. Realini, C.E., and Marcos, B. (2014) Active and intelligent packaging systems for a modern society.
Meat Sci., 98 (3), 404–419.
342 Chitin and Chitosan: Properties and Applications
38. Sharma, C., Dhiman, R., Rokana, N., and Panwar, H. (2017) Nanotechnology: An untapped resource
for food packaging. Frontiers Microbiol., 8 (September), Doi:103389/fmicb.2017.01735
39. Cazón, P., Velazquez, G., Ramírez, J.A., and Vázquez, M. (2017) Polysaccharide‐based films and coat-
ings for food packaging: A review. Food Hydrocoll., 68, 136–148.
40. Bourtoom, T. (2008) Review article edible films and coatings: Characteristics and properties. Int. Food
Res. J., 15 (3), 237–248.
41. Remya, S., Mohan, C.O., Bindu, J., Sivaraman, G.K., Venkateshwarlu, G., and Ravishankar, C.N.
(2016) Effect of chitosan based active packaging film on the keeping quality of chilled stored barra-
cuda fish. J. Food Sci. Technol., 53 (1), 685–693.
42. Elsabee, M.Z. and Abdou, E.S. (2013) Chitosan based edible films and coatings: A review. Mater. Sci.
Eng. C, 33 (4), 1819–1841.
43. Kurek, M., Guinault, A., Voilley, A., Galić, K., and Debeaufort, F. (2014) Effect of relative humidity
on carvacrol release and permeation properties of chitosan based films and coatings. Food Chem., 144,
9–17.
44. Yu, Z., Li, B., Chu, J., and Zhang, P. (2018) Silica in situ enhanced PVA/chitosan biodegradable films
for food packages. Carbohydr. Polym., 184, 214–220.
45. Jagadish, R.S., Raj, B., Parameswara, P., and Somashekar, R. (2010) Effect of glycerol on structure‐
property relations in chitosan/poly(ethylene oxide) blended films investigated using wide‐angle X‐ray
diffraction. Polym. Int., 59 (7), 931–936.
46. Riaz, A., Lei, S., Akhtar, H.M.S., Wan, P., Chen, D., Jabbar, S., Abid, M., Hashim, M.M., and Zeng, X.
(2018) Preparation and characterization of chitosan‐based antimicrobial active food packaging film
incorporated with apple peel polyphenols. Int. J. Biol. Macromol., 114, 547–555.
47. Khezrian, A. and Shahbazi, Y. (2018) Application of nanocomposite chitosan and carboxymethyl
cellulose films containing natural preservative compounds in minced camel’s meat. Int. J. Biol.
Macromol., 106, 1146–1158.
48. Mohebi, E. and Shahbazi, Y. (2017) Application of chitosan and gelatin based active packaging films
for peeled shrimp preservation: A novel functional wrapping design. LWT – Food Sci. Technol., 76,
108–116.
49. Kaya, M., Khadem, S., Cakmak, Y.S., Mujtaba, M., Ilk, S., Akyuz, L., Salaberria, A.M., Labidi, J.,
Abdulqadir, A.H., and Deligöz, E. (2018) Antioxidative and antimicrobial edible chitosan films
blended with stem, leaf and seed extracts of Pistacia terebinthus for active food packaging. RSC Adv.,
8 (8), 3941–3950.
50. Kaewklin, P., Siripatrawan, U., Suwanagul, A., and Lee, Y.S. (2018) Active packaging from chitosan‐
titanium dioxide nanocomposite film for prolonging storage life of tomato fruit. Int. J. Biol. Macromol.,
112, 523–529.
51. Wang, L., Wang, Q., Tong, J., and Zhou, J. (2017) Physicochemical properties of chitosan films
incorporated with honeysuckle flower extract for active food packaging. J. Food Process Eng., 40 (1),
1–8.
52. Sun, L., Sun, J., Chen, L., Niu, P., Yang, X., and Guo, Y. (2017) Preparation and characterization of
chitosan film incorporated with thinned young apple polyphenols as an active packaging material.
Carbohydr. Polym., 163, 81–91.
53. Souza, V.G.L., Fernando, A.L., Pires, J.R.A., Rodrigues, P.F., Lopes, A.A.S., and Fernandes, F.M.B.
(2017) Physical properties of chitosan films incorporated with natural antioxidants. Ind. Crops Prod.,
107, 565–572.
54. Benbettaïeb, N., Tanner, C., Cayot, P., and Karbowiak, T. (2018) Impact of functional properties and
release kinetics on antioxidant activity of biopolymer active films and coatings. Food Chem., 242,
369–377.
55. Ashrafi, A., Jokar, M., and Mohammadi Nafchi, A. (2018) Preparation and characterization of
biocomposite film based on chitosan and kombucha tea as active food packaging. Int. J. Biol.
Macromol., 108, 444–454.
Food Applications of Chitosan and its Derivatives 343
56. Vilela, C., Pinto, R.J.B., Coelho, J., Domingues, M.R.M., Daina, S., Sadocco, P., Santos, S.A.O., and
Freire, C.S.R. (2017) Bioactive chitosan/ellagic acid films with UV‐light protection for active food
packaging. Food Hydrocoll., 73, 120–128.
57. Liu, J., Liu, S., Wu, Q., Gu, Y., Kan, J., and Jin, C. (2017) Effect of protocatechuic acid incorporation
on the physical, mechanical, structural and antioxidant properties of chitosan film. Food Hydrocoll.,
73, 90–100.
58. Cui, H., Yuan, L., and Lin, L. (2017) Novel chitosan film embedded with liposome‐encapsulated phage
for biocontrol of Escherichia coli O157:H7 in beef. Carbohydr. Polym., 177, 156–164.
59. Balti, R., Ben, M., Sayari, N., Yacoubi, L., Rabaoui, L., Brodu, N., and Massé, A. (2017) Development
and characterization of bioactive edible films from spider crab (Maja crispata) chitosan incorporated
with Spirulina extract. Int. J. Biol. Macromol., 105, 1464–1472.
60. Kim, K.M., Son, J.H., Kim, S.‐K., Weller, C.L., and Hanna, M.A. (2006) Properties of chitosan films
as a function of pH and solvent type. J. Food Sci., 71 (3), E119–E124.
61. van den Broek, L.A.M., Knoop, R.J.I., Kappen, F.H.J., and Boeriu, C.G. (2015) Chitosan films and
blends for packaging material. Carbohydr. Polym., 116, 237–242.
62. Seal, B.L., Otero, T.C., and Panitch, A. (2001) Polymeric biomaterials for tissue and organ regenera-
tion. Mater. Sci. Eng., 34 (4–5), 147–230.
63. Qin, Y.Y., Zhang, Z.H., Li, L., Yuan, M.L., Fan, J., and Zhao, T.R. (2015) Physio‐mechanical properties
of an active chitosan film incorporated with montmorillonite and natural antioxidants extracted from
pomegranate rind. J. Food Sci. Technol., 52 (3), 1471–1479.
64. Beigzadeh Ghelejlu, S., Esmaiili, M., and Almasi, H. (2016) Characterization of chitosan‐nanoclay
bionanocomposite active films containing milk thistle extract. Int. J. Biol. Macromol., 86, 613–621.
65. Zhang, L., Wang, H., Jin, C., Zhang, R., Li, L., Li, X., and Jiang, S. (2017) Sodium lactate loaded
chitosan‐polyvinyl alcohol/montmorillonite composite film towards active food packaging. Innov.
Food Sci. Emerg. Technol., 42, 101–108.
66. Mata, M.A.L., Ruiz, S., José, C., Ornelas, D.J., Lizette, C., Toro, D., Enrique, S., Ríos, M., Silva, N.P.,
Luis, B., Chávez, A.C., and Burruel, S.E. (2018) Mechanical, barrier and antioxidant properties of
chitosan films incorporating cinnamaldehyde. J. Polym. Environ., 26 (2), 452–461.
67. Hafsa, J., Smach, M. ali, Ben Khedher, M.R., Charfeddine, B., Limem, K., Majdoub, H., and Rouatbi,
S. (2016) Physical, antioxidant and antimicrobial properties of chitosan films containing Eucalyptus
globulus essential oil. LWT – Food Sci. Technol., 68, 356–364.
68. Kalaycıoğlu, Z., Torlak, E., Akın‐Evingür, G., Özen, İ., and Erim, F.B. (2017) Antimicrobial and phys-
ical properties of chitosan films incorporated with turmeric extract. Int. J. Biol. Macromol., 101,
882–888.
69. Moreno‐vásquez, M.J., Valenzuela‐buitimea, E.L., Sánchez‐valdes, S., Rosas‐burgos, E.C., and Oca,
V.M. (2017) Functionalization of chitosan by a free radical reaction: Characterization, antioxidant and
antibacterial potential. 155, 117–127.
70. Shahbazi, Y. (2017) The properties of chitosan and gelatin films incorporated with ethanolic red grape
seed extract and Ziziphora clinopodioides essential oil as biodegradable materials for active food pack-
aging. Int. J. Biol. Macromol., 99, 746–753.
71. Ramziia, S., Ma, H., Yao, Y., Wei, K., and Huang, Y. (2018) Enhanced antioxidant activity of fish
gelatin–chitosan edible films incorporated with procyanidin. J. Appl. Polym. Sci., 135 (10).
72. Li, T., Li, J., Hu, W., and Li, X. (2013) Quality enhancement in refrigerated red drum (Sciaenops ocel-
latus) fillets using chitosan coatings containing natural preservatives. Food Chem., 138 (2–3),
821–826.
73. Yuceer, M. and Caner, C. (2014) Antimicrobial lysozyme‐chitosan coatings affect functional proper-
ties and shelf life of chicken eggs during storage. J. Sci. Food Agric., 94 (1), 153–162.
74. He, S., Yang, Q., Ren, X., Zi, J., Lu, S., Wang, S., Zhang, Y., and Wang, Y. (2014) Antimicrobial effi-
ciency of chitosan solutions and coatings incorporated with clove oil and/or ethylenediaminetetraace-
tate. J. Food Saf., 34 (4), 345–352.
344 Chitin and Chitosan: Properties and Applications
75. Duran, M., Aday, M.S., Zorba, N.N.D., Temizkan, R., Büyükcan, M.B., and Caner, C. (2016) Potential
of antimicrobial active packaging “containing natamycin, nisin, pomegranate and grape seed extract in
chitosan coating” to extend shelf life of fresh strawberry. Food Bioprod. Process., 98, 354–363.
76. Farajzadeh, F., Motamedzadegan, A., Shahidi, S.A., and Hamzeh, S. (2016) The effect of chitosan‐
gelatin coating on the quality of shrimp (Litopenaeus vannamei) under refrigerated condition. Food
Control, 67, 163–170.
77. Sangsuwan, J., Rattanapanone, N., and Pongsirikul, I. (2015) Development of active chitosan films
incorporating potassium sorbate or vanillin to extend the shelf life of butter cake. Int. J. Food Sci.
Technol., 50 (2), 323–330.
78. Higueras, L., López‐Carballo, G., Gavara, R., and Hernández‐Muñoz, P. (2014) Reversible covalent
immobilization of cinnamaldehyde on chitosan films via Schiff base formation and their application in
active food packaging. Food Bioprocess Technol., 8 (3), 526–538.
79. Lo’ay, A.A. and Dawood, H.D. (2017) Active chitosan/PVA with ascorbic acid and berry quality of
“Superior seedless” grapes. Sci. Hortic. (Amsterdam)., 224, 286–292.
80. Frazão, G.G.S., Blank, A.F., and de Aquino Santana, L.C.L. (2017) Optimisation of edible chitosan
coatings formulations incorporating Myrcia ovata Cambessedes essential oil with antimicrobial
potential against foodborne bacteria and natural microflora of Mangaba fruits. LWT – Food Sci.
Technol., 79, 1–10.
81. Alves, V.L.C.D., Rico, B.P.M., Cruz, R.M.S., Vicente, A.A., Khmelinskii, I., and Vieira, M.C. (2018)
Preparation and characterization of a chitosan film with grape seed extract‐carvacrol microcapsules
and its effect on the shelf‐life of refrigerated salmon (Salmo salar). LWT – Food Sci. Technol., 89,
525–534.
82. Sheldon, R.A. and van Pelt, S. (2013) Enzyme immobilisation in biocatalysis: Why, what and how.
Chem. Soc. Rev., 42 (15), 6223–6235.
83. Liu, D.‐M., Chen, J., and Shi, Y.‐P. (2018) Advances on methods and easy separated support materials
for enzymes immobilization. TrAC Trends Anal. Chem., 102, 332–342.
84. Younes, I., and Rinaudo, M. (2015) Chitin and chitosan preparation from marine sources. Structure,
properties and applications. Mar. Drugs, 13 (3), 1133–1174.
85. Ricardi, N.C., de Menezes, E.W., Valmir Benvenutti, E., da Natividade Schöffer, J., Hackenhaar, C.R.,
Hertz, P.F., and Costa, T.M.H. (2018) Highly stable novel silica/chitosan support for β‐galactosidase
immobilization for application in dairy technology. Food Chem., 246, 343–350.
86. Demir, T. and Önal, S. (2018) Bioaffinity immobilization and characterization of α‐galactosidase on
aminophenyl boronic acid derivatized chitosan and Sepabeads EC‐EA. LWT – Food Sci. Technol., 90,
547–555.
87. Liu, D.M., Chen, J., and Shi, Y.P. (2017) α‐Glucosidase immobilization on chitosan‐enriched magnetic
composites for enzyme inhibitors screening. Int. J. Biol. Macromol., 105, 308–316.
88. Graebin, N.G., de Andrades, D., Bonin, M.C., Rodrigues, R.C., and Ayub, M.A.Z. (2016)
Dextransucrase immobilized on activated‐chitosan particles as a novel biocatalyst. J. Mol. Catal. B
Enzym., 133, S143–S149.
89. Irshad, M., Murtza, A., Zafar, M., Bhatti, K.H., Rehman, A., and Anwar, Z. (2017) Chitosan‐immobi-
lized pectinolytics with novel catalytic features and fruit juice clarification potentialities. Int. J. Biol.
Macromol., 104, 242–250.
90. Khoshnevisan, K., Vakhshiteh, F., Barkhi, M., Baharifar, H., Poor‐Akbar, E., Zari, N., Stamatis, H.,
and Bordbar, A.K. (2017) Immobilization of cellulase enzyme onto magnetic nanoparticles:
Applications and recent advances. Mol. Catal., 442, 66–73.
91. Manzo, R.M., Ceruti, R.J., Bonazza, H.L., Adriano, W.S., Sihufe, G.A., and Mammarella, E.J. (2018)
Immobilization of carboxypeptidase A into modified chitosan matrixes by covalent attachment. Appl.
Biochem. Biotechnol., 185, 1029–1043.
92. Urrutia, P., Arrieta, R., Alvarez, L., Cardenas, C., Mesa, M., and Wilson, L. (2018) Immobilization of
lipases in hydrophobic chitosan for selective hydrolysis of fish oil: The impact of support functionali-
zation on lipase activity, selectivity and stability. Int. J. Biol. Macromol., 108, 674–686.
Food Applications of Chitosan and its Derivatives 345
93. Rathee, K., Dhull, V., Dhull, R., and Singh, S. (2016) Biosensors based on electrochemical lactate
detection: A comprehensive review. Biochem. Biophys. Reports, 5, 35–54.
94. Shukla, S.K., Mishra, A.K., Arotiba, O.A., and Mamba, B.B. (2013) Chitosan‐based nanomaterials:
A state‐of‐the‐art review. Int. J. Biol. Macromol., 59, 46–58.
95. Nesakumar, N., Thandavan, K., Sethuraman, S., Krishnan, U.M., and Rayappan, J.B.B. (2014) An
electrochemical biosensor with nanointerface for lactate detection based on lactate dehydrogenase
immobilized on zinc oxide nanorods. J. Colloid Interface Sci., 414, 90–96.
96. Dordevic, V., Balanc, B., Belscak‐Cvitanovic, A., Levic, S., Trifkovic, K., Kalusevic, A., Kostic, I.,
Komes, D., Bugarski, B., and Nedovic, V. (2014) Trends in encapsulation technologies for delivery of
food bioactive compounds. Food Eng. Rev., 7 (4), 452–490.
97. Speranza, B., Petruzzi, L., Bevilacqua, A., Gallo, M., Campaniello, D., Sinigaglia, M., and Corbo,
M.R. (2017) Encapsulation of active compounds in fruit and vegetable juice processing: Current state
and perspectives. J. Food Sci., 82 (6), 1291–1301.
98. Vieira da Silva, B., Barreira, J.C.M., and Oliveira, M.B.P.P. (2016) Natural phytochemicals and pro-
biotics as bioactive ingredients for functional foods: Extraction, biochemistry and protected‐delivery
technologies. Trends Food Sci. Technol., 50, 144–158.
99. Kurozawa, L.E. and Hubinger, M.D. (2017) Hydrophilic food compounds encapsulation by ionic
gelation. Curr. Opin. Food Sci., 15, 50–55.
100. Coghetto, C.C., Brinques, G.B., and Ayub, M.A.Z. (2016) Probiotics production and alternative
encapsulation methodologies to improve their viabilities under adverse environmental conditions. Int.
J. Food Sci. Nutr., 7486, 1–15.
101. Šipailienė, A. and Petraitytė, S. (2018) Encapsulation of probiotics: Proper selection of the probiotic
strain and the influence of encapsulation technology and materials on the viability of encapsulated
microorganisms. Probiotics Antimicrob. Proteins, 10 (1), 1–10.
102. Chen, H., Song, Y., Liu, N., Wan, H., Shu, G., and Liao, N. (2015) Effect of complexation conditions
on microcapsulation of Lactobacillus acidophilus in xanthan‐chitosan polyelectrolyte complex gels.
Acta Sci. Pol. Technol. Aliment., 14 (3), 207–213.
103. Krayukhina, M.A., Samoilova, N.A., and Yamskov, I.A. (2008) Polyelectrolyte complexes of chi-
tosan: Formation, properties and applications. Russ. Chem. Rev., 77 (9), 799–813.
104. Divya, K. and Jisha, M.S. (2018) Chitosan nanoparticles preparation and applications. Environ.
Chem. Lett., 16 (1), 101–112.
105. Fang, Z., and Bhandari, B. (2010) Encapsulation of polyphenols – a review. Trends Food Sci. Technol.,
21 (10), 510–523.
106. Shao, Y., Wu, C., Wu, T., Li, Y., Chen, S., Yuan, C., and Hu, Y. (2018) Eugenol‐chitosan nanoemul-
sions by ultrasound‐mediated emulsification: Formulation, characterization and antimicrobial activ-
ity. Carbohydr. Polym., 193, 144–152.
107. Li, F., Jin, H., Xiao, J., Yin, X., Liu, X., Li, D., and Huang, Q. (2018) The simultaneous loading of
catechin and quercetin on chitosan‐based nanoparticles as effective antioxidant and antibacterial
agent. Food Res. Int., 111, 351–360.
108. Mascheroni, E., Figoli, A., Musatti, A., Limbo, S., Drioli, E., Suevo, R., Talarico, S., and Rollini, M.
(2014) An alternative encapsulation approach for production of active chitosan‐propolis beads. Int. J.
Food Sci. Technol., 49 (5), 1401–1407.
109. Zhang, H., Fu, Y., Niu, F., Li, Z., Ba, C., Jin, B., Chen, G., and Li, X. (2018) Enhanced antioxidant
activity and in vitro release of propolis by acid‐induced aggregation using heat‐denatured zein and
carboxymethyl chitosan. Food Hydrocoll., 81, 104–112.
110. Liang, J., Yan, H., Wang, X., Zhou, Y., Gao, X., Puligundla, P., and Wan, X. (2017) Encapsulation of
epigallocatechin gallate in zein/chitosan nanoparticles for controlled applications in food systems.
Food Chem., 231, 19–24.
111. Sabliov, C.M. and Astete, C.E. (2008) Encapsulation and controlled release of antioxidants and vita-
mins, in Delivery and Controlled Release of Bioactives in Foods and Nutraceuticals, Elsevier, pp.
297–330.
346 Chitin and Chitosan: Properties and Applications
112. Di Martino, A., Trusova, M.E., Postnikov, P.S., and Sedlarik, V. (2018) Enhancement of the antioxi-
dant activity and stability of β‐carotene using amphiphilic chitosan/nucleic acid polyplexes. Int. J.
Biol. Macromol., 117, 773–780.
113. Varankovich, N., Martinez, M.F., Nickerson, M.T., and Korber, D.R. (2017) Survival of probiotics in
pea protein‐alginate microcapsules with or without chitosan coating during storage and in a simulated
gastrointestinal environment. Food Sci. Biotechnol., 26 (1), 189–194.
114. Tripathi, M.K.K. and Giri, S.K.K. (2014) Probiotic functional foods: Survival of probiotics during
processing and storage. J. Funct. Foods, 9 (1), 225–241.
115. Hill, C., Guarner, F., Reid, G., Gibson, G.R., Merenstein, D.J., Pot, B., Morelli, L., Canani, R.B.,
Flint, H.J., Salminen, S., Calder, P.C., and Sanders, M.E. (2014) Expert consensus document: The
international scientific association for probiotics and prebiotics consensus statement on the scope and
appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol., 11 (8), 506–514.
116. Brinques, G.B., and Ayub, M.A.Z. (2011) Effect of microencapsulation on survival of Lactobacillus
plantarum in simulated gastrointestinal conditions, refrigeration, and yogurt. J. Food Eng., 103 (2),
123–128.
117. Gandomi, H., Abbaszadeh, S., Misaghi, A., Bokaie, S., and Noori, N. (2016) Effect of chitosan‐algi-
nate encapsulation with inulin on survival of Lactobacillus rhamnosus GG during apple juice storage
and under simulated gastrointestinal conditions. LWT – Food Sci. Technol., 69, 365–371.
118. Zanjani, M.A.K., Ehsani, M.R., Ghiassi Tarzi, B., and Sharifan, A. (2018) Promoting Lactobacillus
casei and Bifidobacterium adolescentis survival by microencapsulation with different starches and
chitosan and poly L‐lysine coatings in ice cream. J. Food Process. Preserv., 42 (1), e13318.
119. Bepeyeva, A., de Barros, J.M.S., Albadran, H., Kakimov, A.K., Kakimova, Z.K., Charalampopoulos,
D., and Khutoryanskiy, V. V. (2017) Encapsulation of Lactobacillus casei into calcium pectinate‐chi-
tosan beads for enteric delivery. J. Food Sci., 82 (12), 2954–2959.
120. Singh, P., Medronho, B., Alves, L., da Silva, G.J., Miguel, M.G., and Lindman, B. (2017) Development
of carboxymethyl cellulose‐chitosan hybrid micro‐ and macroparticles for encapsulation of probiotic
bacteria. Carbohydr. Polym., 175, 87–95.
121. Fareez, I.M., Lim, S.M., Lim, F.T., Mishra, R.K., and Ramasamy, K. (2017) Microencapsulation of
Lactobacillus sp. using chitosan‐alginate‐xanthan gum‐β‐cyclodextrin and characterization of its
cholesterol reducing potential and resistance against pH, temperature and storage. J. Food Process
Eng., 40 (3), e12458.
122. Trabelsi, I., Ktari, N., Ben Slima, S., Bouchaala, K., and Ben Salah, R. (2016) Effects of supplemen-
tation with L. plantarum TN8 encapsulated in alginate‐chitosan in broiler chickens. Int. J. Biol.
Macromol., 89, 677–681.
123. Trabelsi, I., Bejar, W., Ayadi, D., Chouayekh, H., Kammoun, R., Bejar, S., and Ben Salah, R. (2013)
Encapsulation in alginate and alginate‐coated chitosan improved the survival of newly probiotic in
oxgall and gastric juice. Int. J. Biol. Macromol., 61, 36–42.
124. Zhao, Z., Liu, N., Yang, L., Wang, J., Song, S., Nie, D., Yang, X., Hou, J., and Wu, A. (2015) Cross‐
linked chitosan polymers as generic adsorbents for simultaneous adsorption of multiple mycotoxins.
Food Control, 57, 362–369.
125. Khan, F.I., Rahman, S., Queen, A., Ahamad, S., Ali, S., Kim, J., and Hassan, M.I. (2017) Implications
of molecular diversity of chitin and its derivatives. Appl. Microbiol. Biotechnol., 101 (9),
3513–3536.
126. Kyzas, G. and Bikiaris, D. (2015) Recent modifications of chitosan for adsorption applications: A
critical and systematic review. Mar. Drugs, 13 (1), 312–337.
127. Becker‐Algeri, T.A., Castagnaro, D., de Bortoli, K., de Souza, C., Drunkler, D.A., and Badiale‐
Furlong, E. (2016) Mycotoxins in bovine milk and dairy products: A review. J. Food Sci., 81 (3),
R544–R552.
128. Quintela, S., Villarán, M.C., De Armentia, I.L., and Elejalde, E. (2012) Ochratoxin A removal from
red wine by several oenological fining agents: Bentonite, egg albumin, allergen‐free adsorbents, chi-
tin and chitosan. Food Addit. Contam. Part A, 29 (7), 1168–1174.
Food Applications of Chitosan and its Derivatives 347
129. Luo, Y., Li, Z., Yuan, Y., and Yue, T. (2016) Bioadsorption of patulin from kiwi fruit juice onto a
superior magnetic chitosan. J. Alloys Compd., 667, 101–108.
130. Abou El Fadl, F.I. (2014) Radiation grafting of ionically crosslinked alginate/chitosan beads with
acrylic acid for lead sorption. J. Radioanal. Nucl. Chem., 301 (2), 529–535.
131. Yen, M., Tseng, Y., Li, R., and Mau, J. (2006) Antioxidant properties of fungal chitosan from shiitake
stripes. Fung. Sci., 21 (1‐2), 1–11
132. Zhu, K.X., Li, J., Li, M., Guo, X.N., Peng, W., and Zhou, H.M. (2013) Functional properties of chi-
tosan‐xylose Maillard reaction products and their application to semi‐dried noodle. Carbohydr.
Polym., 92 (2), 1972–1977.
133. Lamas, A., Anton, X., Miranda, J.M., Roca‐Saavedra, P., Cardelle‐Cobas, A., Ibarra, I.S., Franco,
C.M., and Cepeda, A. (2016) Technological strategies for the development of egg‐derived products
with reduced content of cholesterol. Food Bioprocess Technol., 9 (1), 81–90.
134. do Amaral, D.S., Cardelle‐Cobas, A., do Nascimento, B.M.S., Monteiro, M.J., Madruga, M.S., and
Pintado, M.M.E. (2015) Development of a low fat fresh pork sausage based on chitosan with health
claims: Impact on the quality, functionality and shelf‐life. Food Funct., 6 (8), 2768–2778.
135. Taştan, Ö. and Baysal, T. (2017) Chitosan as a novel clarifying agent on clear apple juice production:
Optimization of process conditions and changes on quality characteristics. Food Chem., 237,
818–824.
14
Potential of Chitosans in the
Development of Edible
Food Packaging
Véronique Coma1 and Artur Bartkowiak2
University of Bordeaux, LCPO, UMR 5629, Centre National de la Recherche Scientifique (CNRS),
1
Pessac, France
2
Center of Bioimmobilisation and Innovative Packaging Materials, Faculty of Food Sciences and
Fisheries, West Pomeranian University of Technology, Szczecin, Poland
OH
NH2
O
O HO O
HO O
NH O
C
H3C O OH
y
n
Figure 14.1 Repeating units of partially acetylated chitosan characterized by an average degree of
acetylation.
edible bioactive packaging have been motivated by both increasing consumer demand for
safe, convenient, and stable foods and also awareness of the negative environmental impacts
of nonbiodegradable packaging waste. Chitosans, therefore, have potential in a large
domain of applications, especially due to their strong film‐ and gel‐forming properties.
Chitosan is a family of polysaccharides with many different structures. It is a high‐
molecular‐weight linear basic polycationic heteropolysaccharide, consisting of two mono
saccharides, N‐acetyl‐D‐glucosamine and D‐glucosamine, linked together by β (1→4)
glycosidic bonds (Figure 14.1).
Due to variable relative amounts of both monosaccharides, chitosans are not a uniquely
defined compound. This diversity in structure leads to diversity in biological activities.
Much of the commercial interest in chitosans and their derivatives arises from the fact that
they combine several favorable biological characteristics, including nontoxicity and poten
tial antimicrobial activity. Regarding the latter, numerous publications are now available
showing not only the wide range of chitosan activities toward human pathogens as well as
foodborne organisms but also the influence of different factors on this activity (i.e., molec
ular weight, deacetylation degree, pH). There is still a lack of knowledge about the mecha
nism of this antimicrobial action, but on bacteria, the site of action is more probably the
microbial cell surface, such as the majority of cationic antibacterial agents.
The use of chitosans in the formulation of edible films or coatings can, thus, lead to new
active food packaging materials. This is a modern strategy seeking to promote an additional
active role to assist in maintaining product quality. The focal point of this chapter is to
provide an overview of bioactive chitosan edible coatings that have the potential to enhance
the preservation of packaged products with a particular emphasis on (i) criteria limiting
their market introduction, (ii) studies based on real food applications and emerging new
technologies that can improve their future commercialization. Only non‐chemically‐
modified chitosans are targeted.
than traditional products. However, we have seen many scientific publications during the
last decades with no real market introduction. In this regard, the main aim of the current
Actinpack COST FP1405 (active and intelligent fiber‐based packaging—innovation and
market introduction) is to develop a knowledge‐based network around sustainable, active,
and intelligent bio‐based packaging in order to overcome current technological, industrial,
and social limitations that hinder the wide deployment of existing and newly developed
solutions in market applications. In the following text, only the limitations connected to
chitosans will be discussed.
14.1.2 Solubility
Chitosans have a great advantage over chitin regarding solubility but this is still a point that
limits its real application. Chitin is insoluble in most organic solvents, while chitosans are
readily soluble in dilute acidic solutions. The presence of the amino groups indicates that
pH substantially alters the charged state and properties of chitosans, and the soluble–
insoluble transition occurs at their pKa value around pH values between 6 and 6.5 ([5],
Figure 14.2). In practice, chitosan film‐forming solutions were prepared at low pH, in
OH OH
pH>PKa
O O O O
OH OH
O pH<PKa O
NH2 NH3+
Water solubility
Potential
antimicrobial
action
Figure 14.2 Impact of pH on the protonation of the amino group of the glucosamine monomer of
chitosans.
352 Chitin and Chitosan: Properties and Applications
general close to 3–5, and even if an adjustment can be made before applying to the food
surface, the pH of edible chitosan solutions is generally close to 5. This insolubility at pH
higher than 6.5 could be a disadvantage for many industrial applications such as edible
coatings for pH‐sensitive food. For food coatings produced from unmodified chitosans,
different dilute acid solutions can be used and the frequent counter ions found in the
literature are acetate, lactate, citrate.
14.1.3 Source—Origin
The question is “Which chitosan is best for a specific application such as edible packag
ing?” Because a large amount of the crustacean exoskeleton is readily available as a by‐
product of the seafood processing industry, chitosans from crab shell or the fishing
industry are the most frequent one on the market. However, the marine origin of chi
tosans is sometimes considered as problematic. Chitosans are also found in nature, for
example, in the cell walls of fungi of Zygomycetes as well as in insect cuticles. Recent
advances in fermentation technology suggest that the cultivation of fungi (Aspergillus
niger) can provide an alternative source of chitosan. In addition, Greenaltech in Spain
has recently discovered the presence of natural chitosans in certain green microalgae
species (Derek Latil). The microalgal chitosans do not undergo any chemical
modifications.
One important point is that various chitosan sources can be responsible for structural
differences [6]. Some authors have shown that whereas the acetyl groups in chitosan
produced from crustacean chitin are uniformly distributed along the polymer chain, a
chitosan of similar degree of deacetylation (DD) isolated from fungal cell walls would
possess acetyl residues that are grouped into clusters [4, 7]. This variability in relation to
the source or origin of chitosans can be a key point for the industrial development of
chitosan‐based edible coatings and films.
14.1.4 Structure Variability
As already mentioned, chitosan structure may differ regarding not only the sources but
also the methods used to transform chitin into chitosans. Today, commercially available
chitosans are often produced from fungal strains or shrimp shell chitin. The main reason
for using the latter is the current context of waste by‐product valorization, particularly
waste from fish‐ and sea‐food factories. Variations in chemical structure could be due to
batch‐to‐batch differences, posing a problem of reproducibility. In addition, the end
product sometimes does not offer the required purity needed for food applications. The
problem is that in order to be able to explore the benefits of chitosans in depth, they
need to have a defined structure. One idea could be to start from materials readily avail
able from natural resources and then transform them through chemical reactions into
monomers designed to self‐condense in a well‐defined manner when reacting in the
presence of an engineered enzyme. In the same way, Dr. Moerschbacher’s team from
the University of Munster in Germany is working on the production of well‐defined
chitosans with known structures and functionalities through biotechnological
approaches. The tools required for this approach come from enzymes such as chitin
synthases, which produce chitin from small sugar molecules, and chitin deacetylases,
Potential of Chitosans in the Development of Edible Food Packaging 353
which convert chitin into chitosan. The recombinant enzymes were then characterized
and used for the biotechnological conversion of chitin into chitosans. However, and
perhaps interestingly, the chitosans obtained differ in their fine structure from all cur
rently available chitosans, which are invariably produced from chitin using chemical
methods. This could be the first step toward “third generation chitosans,” with poten
tially different biological activities.
In any event, chitosans from marine organisms are currently the most studied ones.
Despite the limitations raised previously, many scientific papers related to chitosan‐based
materials for food applications are published every year, showing that these biopolymers
have great potential and could be developed and used in the future because of some specific
properties discussed below.
and the surface to be coated, thus, avoiding the disruption of air between the solution and
the surface. As discussed by Lin and Zhao (2007) [14] and Arnon‐Rips et al. (2018) [15],
the viscosity, density, and surface tension of the coating formulation should be adjusted to
the surface tension and roughness of a specific food product.
Chitosan as
intrinsically active
polymer
Food
Edible
coating
Chitosan as a
carrier of
bioactive agents
Bioactive agent Desorption, and
or active diffusion
functional groups
OH y
Adsorption to negatively
Protonation charged cell membrane
Decrease in osmotic stability
pH < PKa of the cell
Leakage of intracellular
constituants
Indirect antimicrobial
mechanisms Protonation Formation of an impermeable
coat on the microbial surface
pH < PKa
Interaction with exoenzymes
14.2.2.5 Carrier Properties
The incorporation of additives or extracts from natural sources into edible chitosan films
and coatings is one of the methods being explored to increase the shelf life of foods and
then to provide high‐quality products (fresh/safe). Chitosans can be used as a simple carrier
of various molecules such as antimicrobials, antioxidants, vitamins, flavors, and prebiotics
[26–29]. Interestingly, the use of chitosan as a carrier can lead to adequately dispersed
agents. Chitosans can increase the solubility of some hydrophobic molecules in water‐
based formulations.
Regarding studies of the release of active agents, the process responsible for mass
transfer within a system is diffusion, and diffusion coefficients have a much wider range of
values in solids compared to liquids and gases, differing by a factor of more than 1010 as
specified by Flores‐Martinez [30]. When an additive is expected to diffuse through a
material, this corresponds to diffusion in a solid system. A fairly new concept in interactive
packaging is to control this release of additives [31]. Diffusion in polymers is relatively
complex and depends strongly on the concentration and degree of polymer swelling. This
degree of swelling also depends on the free volume in the materials, that is, space not
occupied by polymeric chains or molecule segments. An increase in the amount of free
volume leads to an increased diffusion rate. As a result, external factors such as temperature
greatly affect the amount of free volume (Flores‐Martinez et al. 2015) [30]. As a result, in
terms of practical importance, active agent diffusivity values are essential in predicting the
Potential of Chitosans in the Development of Edible Food Packaging 357
shelf life of coated food products under changing storage conditions. Regarding the
literature, there is clearly a lack of studies on the diffusivity of active agents. A better
understanding of the mechanism of preservative transport in chitosan‐based edible films is,
thus, needed.
Direct incorporation into the film‐forming solution is used in numerous studies. However,
because of some disadvantages with such blends, other studies are considering the
development of multilayer systems, leading to more sophisticated processes. According to
Suppalkul (2015) [32] and Han (1996) [33], a multilayer structure for bioactive‐release
packaging systems can be organized comprising an outer bioactive barrier layer (a barrier
layer to prevent the loss of bioactive agents), an active‐containing matrix layer (provides a
very rapid diffusion rate of the bioactive agent), a release‐control layer (controls the initial
lag period and the subsequent flux of the substance), and food. Chitosans could be good
options to create this multilayer design, which has the advantage of the control layer to alter
the diffusivity of bioactive substances.
were found to significantly affect the formation of the NPs. It was postulated [43] that the
pH of TPP affects the electronegative potential of the molecule in its reaction with free
amine groups in chitosan. At lower pH values, TPP becomes less reactive for chemical
interactions with chitosan because it is buffered by more positive ions in solution (H3O+ and
H+). TPP, therefore, reacts with fewer amino groups of chitosan (−NH3+), leading to the
formation of smaller‐sized NPs that are more monodisperse. The interactive effect of
molecular weight (MW) and pH on the formation of CSNPs showed that optimal particle
size was obtained when low‐MW CS at a pH of 4.6 was used [44].
Due to their polymeric cationic characteristics, CSNPs can interact with negatively
charged molecules and polymers. CSNPs have been successfully used as fillers to improve
mechanical and barrier properties as well as the thermo‐stability of films, decrease solubility
and produce more compact and dense materials [45–47].
The use of NPs in food packaging has been proposed on the basis that it could improve
the protection of foods by reducing the permeation of gases, minimizing odor loss, and
increasing mechanical strength and thermal stability. The typical process of edible
nanocomposite film formation is divided into two stages: the first is the synthesis of CSNPs,
and the second is the production of the films containing NPs.
within 15 min using of both inoculated E. coli and S. typhimurium populations, suggesting
their potential use as an effective disinfectant in washing fresh vegetables.
Incorporation of CSNPs produced TG films less effective against E. coli compared to S.
aureus than films with bulk chitosan, and their antimicrobial activity was reduced at high
concentrations of CSNPs probably due to agglomeration [58]. It has been shown that
antibacterial activity of CSNPs could be significantly enhanced by metal ion loaded,
especially for Cu2+, and Zn2+, compared to those of CSNPs and related metal ions, where
Gram‐negative bacteria were more sensitive than Gram‐positive bacteria. It was found that
antibacterial activity was directly proportional to the zeta potential [63].
New active packaging films based on chitosan printed with chitosan–TPP NPs loaded
with thymol using inkjet printing were evaluated by Caro et al. (2016) [64]. The printed
films had enhanced barrier properties compared to the control. The efficiency of thymol
incorporation by inkjet printing was dependent on the number of printed layers, the contact
angle, the amount of glycerol added to the dispersion, and the film type. The printed films
exhibited higher antimicrobial activity against Listeria innocua, S. aureus, S. typhimurium,
Enterobacter aerogenes, Pseudomonas aeruginosa, and E. coli than both non‐printed and
control films.
Recently, several chitosan/sodium–TPP NPs embedded with Torreya grandis aril
essential oils (TEOs) were synthesized using an emulsion‐ionic gelation technique and in
order to replace acetic acid, an ionic liquid (IL) was employed to dissolve chitosan [65].
The obtained results revealed that TEO‐loaded NPs synthesized in acid and IL–aqueous
systems have stronger antibacterial activities against S. aureus than CSNPs alone.
(w/v) chitosan, 1.25% (w/v) chitosan, and CSNPs were stored at ambient temperature
(25 ± 1°C). The shelf life of banana, starch content, weight loss, pulp to peel ratio, total
soluble solid, surface morphology of banana peel, and sensory evaluation were analyzed.
Molecular analysis on the effect of chitosan was also conducted. Based on the results of
Lustriane et al. (2018) [74], it was confirmed that CSNPs and chitosan can provide good
effects on postharvest quality of banana including shelf life, starch content, weight loss,
pulp to peel ratio, total soluble solids, and sensory quality.
by atmospheric restoration for 15 min [86]. The chitosan–fish oil coating reduced the lipid
oxidation process in both fresh and frozen samples, and also decreased the drip loss of
frozen samples by 14.1–27.6%. Chitosan coatings resulted in 0.37–1.19 and 0.27–
1.55 log CFU/g reductions in total plate and psychrotrophic counts in cold‐stored and
frozen‐stored samples, respectively. The positive effect of chitosan coating on the shelf life
extension of salmon (Salmo salar) fillets during storage at 0°C was demonstrated by Souza
et al. (2010b) [87].
The results of Ojagh et al. (2010) [88] indicated that the chitosan coating enriched with
cinnamon oil enabled the good quality characteristics of rainbow trout (Oncorhynchus
mykiss) to be retained longer and helped extend the shelf life during refrigerated storage.
The possibility of improving the shelf life of fresh sea bass (Dicentrarchus labrax) fillets
by using vacuum packaging and wrapping with chitosan‐based edible films during cold
storage at 4°C was assessed by Günlü and Koyun (2013) [89]. The results showed that the
shelf life of the control and vacuum‐packaged groups ended within 5 days, whereas that of
vacuum‐packaged, chitosan‐film‐wrapped samples ended at 25–30 days. Therefore, the
shelf life of sea bass fillets wrapped in chitosan was prolonged by about 20 days.
The potential of CSNPs produced via ionotropic gelation with sodium TPP as a glazing
material for shrimp was investigated by Solval et al. (2014) [90]. Glazing containing
CSNPs reduced the lipid oxidation in shrimp during frozen storage at −20°C. CSNP glazing
had not affected the weight of frozen shrimp as compared to the controls. In addition,
CSNPs reduced total aerobic counts, yeast, and molds without affecting the color and
texture properties of frozen shrimp during 30 days of storage at −20°C.
Recent results of Wang et al. (2015) [91] demonstrated that CSNP coating exhibited a
better effect on the quality maintenance of shrimp as compared to carboxymethyl–chitosan
coating. On day 10 of storage at 4°C, the highest hardness and springiness and lowest total
viable counts of microorganisms were obtained in case of coating with CSNP and followed
by carboxymethyl–chitosan coating. Lower thiobarbituric acid and higher inosinic acid
content were obtained in NP‐coated sample than those of the control on day 10. Similarly,
Wang and Li (2011) [92] proved that CSNPs also improved fat composition and inosinic
acid content of tilapia (Oreochromis niloticus) as compared to normal chitosan materials.
A chitosan/sodium TPP NP solution was developed and applied to shrimp through
vacuum tumbling, and the quality characteristics during frozen storage were evaluated
[93]. Chitosan and CSNP treatments could preserve the color, texture, and moisture content
of shrimp throughout 120 days of frozen storage at −20°C. In addition, chitosan‐ and
CSNP‐treated shrimp had the highest reduction in lipid oxidation as compared to other
treatments at 120 days of storage at −20°C. This study showed that vacuum tumbling with
a chitosan or CSNP solution can be effective at reducing aerobic plate counts and lipid
oxidation in shrimp during frozen storage while maintaining desired physicochemical
properties.
The responsive intelligent film based on poly(vinyl alcohol) (PVA), CSNPs, and
mulberry extracts (MBEs) were successfully prepared and characterized by Ma et al.
(2018) [94]. CSNPs were prepared by using ionotropic with sodium TPP gelation method
to enhance the mechanical properties of PVA‐based films. The film with 6% (w/v) CSNPs
had the highest tensile strength (∼73.43 MPa). The film containing 20% (w/w) MBE had
the highest tensile strength and showed visible color responses to variations across pH
1–13. This film changed from red to green during the process of fish spoilage due to the
364 Chitin and Chitosan: Properties and Applications
change of pH, indicating that it can detect quality changes in fish. Hence, CSNP coating
might represent a potentially efficient and beneficial natural preservative and freshness
indicator for seafood quality, although the further safety evaluation and elucidation of
biological mechanisms are still required for the transition from laboratory systems to
commercial applications in the seafood industry.
14.5 Conclusions
Although chitosans are bio‐based polymers, their environmental impact must be investigated
further, from the extraction of raw materials to the finished product. One point to keep in
mind is that applied chemical methods to produce chitosans from animal chitin require
large amounts of chemicals and produce a lot of wastewater, which has a harsh impact on
the environment. Life cycle assessments have to be conducted more regularly, especially
with the data from the real chitosan producers.
As a conclusion, taking into account that one of the greatest challenges of the food
industry is microbial contamination, chitosan‐based materials including NPs will certainly
be developed considerably during the next decade.
References
1. Kirwan, M. J. and Strawbridge, J. W. (2003). Plastics in food packaging. In Food Packaging Technology
(eds. R. Coles, D. McDowell, M. J. Kirwan), Blackwell Publishing, CRC Press, 174–240.
2. Kendall, H., Kaptan, G., Stewart, G., Grainger, M., Kuznesof, S., Naughton, P. et al. (2018). Drivers
of existing and emerging food safety risks: Expert opinion regarding multiple impacts. Food Control,
90, 440–458.
3. Singla, A. K. and Chawla, M. (2001). Chitosan: Some pharmaceutical and biological aspects – an
update. Journal of Pharmacy and Pharmacology, 53, 1047–1067.
4. Fouad, D. R. G., Thesis “Chitosan as an antimicrobial compound: Mode of action and resistance
mechanisms” (2008). Diese Dissertation ist auf dem Hochschulschriftenserver der ULB Bonn, Bonn,
Germany, pp. 1–215.
5. Shiekh, R. A., Malik, M. A., Al‐Thabaiti, S. A., and Shiekh, M. A. (2013). Chitosan as a novel edible
coating for fresh fruits. Food Science and Technology Research, 19(2), 139–155.
6. Zimoch‐Korzycka, A., Gardrat, C., Al Kharboutly, M., Castellan, A., Pianet, I., Jarmoluk, A., and
Coma, V. (2016). Chemical characterization, antioxidant and anti‐listerial activity of non‐animal
chitosan‐glucan complexes. Food Hydrocolloids, 61, 338–343.
7. Rabea, E. I., Badawy, M. E. T., Stevens, C. V., Smagghe, G., and Steurbaut, W. (2003). Chitosan as
antimicrobial agent: Applications and mode of action. Biomacromolecules, 4(6), 1457–1465.
8. De Azeredo, H. M. C. (2012). Edible coatings. Advances in Fruit Processing Technologies, 345–361.
9. Šuput, D. Z., Lazic ,́ V. L., Popovic ,́ S. Z., and Hromiš, N. M. (2017). Edible films and coat
ings – sources, properties and application. Food and Feed Research, 42, 11–22.
10. Souza, B. W. S., Cerqueira, M. A., Teixeira, J. A., and Vicente, A. A. (2010a). The use of electric fields
for edible coatings and films development and production: A review. Food Engineering Reviews, 2,
244–255.
11. Fernández‐Pan, I., Ignacio, J., and Caballero, M. (2011). In Biopolymers–New Materials for
Sustainable Films and Coatings (ed. D. Plackett), Padstow, UK, Wiley, 233–254.
12. Kester, J. and Fennema, O. (1986) Edible films and coatings: A review. Food Technology, 40(12),
47–59.
13. Moncayo, D., Buitago, G., and Algecira, N. (2013). The surface properties of biopolymer‐coated fruit:
A review. Ingeniería e Investigación, 33(3), 11–16.
Potential of Chitosans in the Development of Edible Food Packaging 365
14. Lin, D. and Zhao, Y. (2007). Innovations in the development and application of edible coatings for
fresh and minimally processed fruits and vegetables. Comprehensive Reviews in Food Science and
Food Safety, 6(2007), 60–75.
15. Arnon‐Rips, H. and Poverenov, E. (2018). Improving food products’ quality and storability by using
layer by layer edible coatings. Trends in Food Science & Technology, 75, 81–92.
16. Poverenov, E., Danino, S., Horev, B., Granit, R., Vinokur, Y., and Rodov, V. (2014). Layer‐by‐layer
electrostatic deposition of edible coating on fresh cut melon model: Anticipated and unexpected effects
of alginate–chitosan combination. Food Bioprocess Technology, 7, 1424–1432
17. Gonzalez‐Aguilar, G. A., Valenzuela‐Soto, E., Lizardi‐Mendoza, J., Goycoolea, F., Martínez‐Téllez,
M. A., Villegas‐Ochoa, M. A., Monroy‐García, I. N., and Ayala‐Zavala, J. F. (2009). Effect of chitosan
coating in preventing deterioration and preserving the quality of fresh‐cut papaya “Maradol.” Journal
of the Science of Food & Agriculture, 89, 15–23.
18. Cazón, P., Vázquez, M., and Velazquez, G. (2018). Novel composite films based on cellulose rein
forced with chitosan and polyvinyl alcohol: Effect on mechanical properties and water vapour perme
ability. Polymer Testing, 69, 536–544.
19. Fundo, J. F., Galvis‐Sanchez, A. C., Delgadillo, I., Silva, C. L., and Quintas, M. A. (2015). The effect
of polymer/plasticiser ratio in film forming solutions on the properties of chitosan films. Food
Biophysics, 10, 324–333.
20. Suyatma, N. E., Tighzert, L., Copinet, A., and Coma, V. (2005). Effect of hydrophilic plasticizers on
mechanical, thermal and surface properties of chitosan films. Journal of Agricultural and Food
Chemistry, 53, 3950–3957.
21. Yousuf, B., Qadri, O. S., and Srivastava, A. K. (2018). Recent developments in shelf‐life extension of
fresh‐cut fruits and vegetables by application of different edible coatings: A review. LWT – Food
Science and Technology, 89, 198–209.
22. Homez‐Jara, A., Daza, L. D., Aguirre, D. M., Muñoz, J. A., Solanilla, J. F., and Váquiro, H. A. (2018).
Characterization of chitosan edible films obtained with various polymer concentrations and drying
temperatures. International Journal of Biological Macromolecules, 113, 1233–1240.
23. Verlee, A., Mincke, S., and Stevens, C. V. (2017). Recent developments in antibacterial and antifungal
chitosan and its derivatives. Carbohydrate Polymers, 164, 268–283.
24. Coma, V. and Verestiuc, L. (2012). Chitosan‐based antimicrobial materials to biomedical applications.
In Medical Applications of Polymers (eds. M. Popa, R. M. Ottenbritte, C.V. Uglea), American Scientific
Publishers, USA, Vol. 1, 64–108, 2011, ISBN: 1‐58883‐194‐9.
25. Raafat, D., Von Bargen, K., Haas, A., and Sahl, H. G. (2008). Insights into the mode of action of chi
tosan as an antibacterial compound. Applied and Environmental Microbiology, 74(12), 3764–3773.
26. Hajji, S., Younes, I., Affes, S., Boufi, S., and Nasri, M. (2018). Optimization of the formulation of
chitosan edible coatings supplemented with carotenoproteins and their use for extending strawberries
postharvest life. Food Hydrocolloids, 83, 375–392.
27. Ventura‐Aguilar, R. I., Bautista‐Baños, S., Flores‐García, G., and Zavaleta‐Avejar, L. (2018). Impact
of chitosan based edible coatings functionalized with natural compounds on Colletotrichum fragariae
development and the quality of strawberries. Food Chemistry, 262, 142–149.
28. Vu, K. D., Hollingsworth, R. G., Leroux, E., Salmieri, S., and Lacroix, M. (2011). Development of
edible bioactive coating based on modified chitosan for increasing the shelf life of strawberries. Food
Research International, 44, 198–203.
29. Yuan, G., Chen, X., and Li, D. (2016). Chitosan films and coatings containing essential oils: The anti
oxidant and antimicrobial activity, and application in food systems. Food Research International, 89,
117–128.
30. Flores‐Martinez N.L., Garcia‐Rosas M.D. and Perez‐Perez C.I. (2015). Diffusion of bioactive com
pounds from edible packaging material to the contained food/modelling migration, In Bioactive Food
Packagings: Strategies, Quality and Safety (ed. M. Kontominas), DEStech Publications, Inc.,
Lancaster, PA, USA, 189–239.
366 Chitin and Chitosan: Properties and Applications
31. Lacoste, A., Schaich, K.M., Zumbrunnen, D., and Yam, K.L. (2005). Advancing controlled release
packaging through smart blending. Packaging Technology and Science, 18(1), 77.
32. Suppakul, P. (2015). Bioactive agents and polymers. Bioactive Food Packaging: Strategies, Quality,
Safety (ed. M. Kontominas), DEStech Publications, Inc., Lancanster, PA, USA, 48–118.
33. Han, J. H. (1996). Modeling the inhibition kinetics and the mass transfer of controlled releasing potas
sium sorbate to develop an antimicrobial polymer for food packaging. PhD dissertation, Purdue
University, West Lafayette.
34. Sorrentino, A., Gorrasi, G., and Vittoria, V. (2007). Potential perspectives of bio‐nanocomposites for
food packaging applications. Trends in Food Science and Technology, 18(2), 84–95.
35. Fu, Y., Sarkar, P., Bhunia, A. K., and Yao, Y. (2016). Delivery systems of antimicrobial compounds to
food. Trends in Food Science and Technology, 57(September 2016), 165–177.
36. Zambrano‐Zaragoza, M. L., González‐Reza, R., Mendoza‐Muñoz, N., Miranda‐Linares, V., Bernal‐
Couoh, T. F., Mendoza‐Elvira, S., and Quintanar‐Guerrero, D. (2018). Nanosystems in edible coatings:
A novel strategy for food preservation. International Journal of Molecular Sciences, 19(3), 705–729.
37. Cheng, C. J., Tietjen, G. T., Saucier‐Sawyer, J.K., and Saltzman, W. M. (2015). A holistic approach to
targeting disease with polymeric nanoparticles. Nature Reviews Drug Discovery, 14, 239–247.
38. Sozer, N. and Kokini, J. L. (2009). Nanotechnology and its applications in the food sector. Trends in
Biotechnology, 27(2), 82–89.
39. Agnihotri, S. A., Mallikarjuna, N. N., and Aminabhavi, T. M. (2004). Recent advances on chitosan‐
based micro‐ and nanoparticles in drug delivery. Journal of Controlled Release, 100, 5–28.
40. Wang, Y., Li, P., Truong‐Dinh Tran, T., Zhang, J., and Kong, L. (2016). Manufacturing techniques and
surface engineering of polymer based nanoparticles for targeted drug delivery to cancer.
Nanomaterials, 6(2), 26.
41. De Campos, A. M., Sanchez, A., and Alonso, M. J. (2001). Chitosan nanoparticles: A new vehicle for
the improvement of the delivery of drugs to the ocular surface. Application to cyclosporin A.
International Journal of Pharmacology, 224(1–2), 159–168.
42. Janes, K. A., Fresneau, M. P., Marazuela, A., Fabra, A., and Alonso, M. J. (2001). Chitosan nanoparti
cles as delivery systems for doxorubicin. Journal of Controlled Release, 73(2–3), 255–267.
43. Masarudin, J., Cutts, S., Evison, B., Phillips, D., Pigram, P., and Pietersz, G. (2016). Factors determin
ing the stability, size distribution, and cellular accumulation of small, monodisperse chitosan nanopar
ticles as candidate vectors for anticancer drug delivery: Application to the passive encapsulation of
14C]‐doxorubicin. Nanotechnology, Science and Applications, 9, 47–48.
44. Sullivan, D. J., Cruz‐Romero, M., Collins, T., Cummins, E., Kerry, J. P., and Morris, M. A. (2018).
Synthesis of monodisperse chitosan nanoparticles. Food Hydrocolloids, 83, 355–364.
45. Chang, P. R., Jian, R., Yu, J., and Ma, X. (2010). Fabrication and characterisation of chitosan nanopar
ticles/plasticised‐starch composites. Food Chemistry, 120(3), 736–740.
46. De Moura, M. R., Avena‐Bustillos, R., McHugh, T., Krochta, J., and Mattoso, L. (2008). Properties of
novel hydroxypropyl methylcellulose films containing chitosan nanoparticles. Journal of Food
Science, 73(7), N31–N37, doi: 10.1111/j.1750‐3841.2008.00872x.
47. De Moura, M. R., Aouada, F. A., Avena‐Bustillos, R. J., McHugh, T. H., Krochta, J. M., and Mattoso, L.
H. (2009). Improved barrier and mechanical properties of novel hydroxypropyl methylcellulose edible
films with chitosan/tripolyphosphate nanoparticles. Journal of Food Engineering, 92(4), 448–453.
48. El‐Shabouri, M. H. (2002). Positively charged nanoparticles for improving the oral bioavailability of
cyclosporin‐A. International Journal of Pharmaceutics, 249(1–2), 101–108.
49. Ngan, L. T. K., Wang, S. L., Hiep, I. M., Luong, P. M., Vui, N. T., Dink, T. M., and Dzung, N. A.
(2014). Preparation of chitosan nanoparticles by spray drying, and their antibacterial activity. Research
on Chemical Intermediates, 40(6), 2165–2175.
50. Calvo, P., Vila‐Jato, J. L., and Alonso, M. J. (1997). Evaluation of cationic polymer‐coated nanocap
sules as ocular drug carriers. Internationl Journal of Pharmacology, 53, 41–50.
Potential of Chitosans in the Development of Edible Food Packaging 367
51. Hassani, S., Laouini, A., Fessi, H., and Charcosset, C. (2015). Preparation of chitosan‐TPP nanoparti
cles using microengineered membranes – Effect of parameters and encapsulation of tacrine. Colloids
and Surfaces A: Physicochemical and Engineering Aspects, 482, 34–43.
52. Nguyen, T. V., Nguyen, T. T. H., Wang, S. L., Vo, T. P. K., and Nguyen, A. D. (2017). Preparation of
chitosan nanoparticles by TPP ionic gelation combined with spray drying, and the antibacterial activity
of chitosan nanoparticles and a chitosan nanoparticle–amoxicillin complex. Research on Chemical
Intermediates, 43(6), 3527–3537.
53. Mitra, S., Gaur, U., Ghosh, P. C., and Maitra, A. N. (2001). Tumor targeted delivery of encapsulated
dextran–doxorubicin conjugate using chitosan nanoparticles as carrier. Journal of Controlled Release,
74, 317–323.
54. Lorevice, M. V., de Moura, M. R., Aouada, F. A., and Mattoso, L. H. C. (2012). Development of novel
guava puree films containing chitosan nanoparticles. Journal of Nanoscience and Nanotechnology,
12(3), 2711–2717.
55. Lorevice, M. V., de Moura, M. R., and Mattoso, L. H. C. (2014). Nanocompósito de polpa de mamão
e nanopartículas de quitosana para aplicasão em embalagem. Química Nova, 37(6), 931–936.
56. Souza, H. K. S., Campiña, J. M., Sousa, A. M. M., Silva, F., and Gonçalves, M. P. (2013). Ultrasound‐
assisted preparation of size‐controlled chitosan nanoparticles: Characterization and fabrication of
transparent biofilms. Food Hydrocolloids, 31(2), 227–236.
57. Hosseini, S. F., Rezaei, M., Zandi, M., and Farahmandghavi, F. (2015). Fabrication of bio‐nanocomposite
films based on fish gelatin reinforced with chitosan nanoparticles. Food Hydrocolloids, 44,
172–182.
58. Antoniou, J., Liu, F., Majeed, H., and Zhong, F. (2015). Characterization of tara gum edible films
incorporated with bulk chitosan and chitosan nanoparticles: A comparative study. Food Hydrocolloids,
44, 309–319.
59. Martelli, M. R., Barros, T. T., de Moura, M. R., Mattoso, L. C. H., and Assis, O. B. G. (2012). Effect
of chitosan nanoparticles and pectin content on mechanical properties and water permeability of
banana puree films. Journal of Food Science, 78, N98–N104.
60. Lorevice, M. V., Otoni, C. G., de Moura, M. R., and Mattoso, L. H. C. (2016). Chitosan nanoparticles
on the improvement of thermal, barrier, and mechanical properties of high‐ and low‐methyl pectin
films. Food Hydrocolloids, 52, 732–740.
61. Qi, L., Xu, Z., Jiang, X., Hu, C., and Zou, X. (2004). Preparation and antibacterial activity of chitosan
nanoparticles. Carbohydrate Research, 339(16), 2693–2700.
62. Paomephan, P., Assavanig, A., Chaturongakul, S., Cady, N. C., Bergkvist, M., and Niamsiri, N. (2018).
Insight into the antibacterial property of chitosan nanoparticles against Escherichia coli and Salmonella
typhimurium and their application as vegetable wash disinfectant. Food Control, 86, 294–301.
63. Du, W. L., Niu, S. S., Xu, Y. L., Xu, Z. R., and Fan, C. L. (2009). Antibacterial activity of chitosan
tripolyphosphate nanoparticles loaded with various metal ions. Carbohydrate Polymers, 75(3),
385–389.
64. Caro, N., Medina, E., Díaz‐Dosque, M., López, L., Abugoch, L., and Tapia, C. (2016). Novel active
packaging based on films of chitosan and chitosan/quinoa protein printed with chitosan‐TPP‐thymol
nanoparticles via thermal ink‐jet printing. Food Hydrocolloids, 52, 520–532.
65. Wu, J., Shu, Q., Niu, Y., Jiao, Y., and Chen, Q. (2018). Preparation, characterization, and antibacterial
effects of chitosan nanoparticles embedded with essential oils synthesized in an ionic liquid containing
system. Journal of Agricultural and Food Chemistry, acs.jafc.8b01428.
66. De Lima, R., Feitosa, L., Pereira, A. do E. S., De Moura, M. R., Aouada, F. A., Mattoso, L. H. C., and
Fraceto, L. F. (2010). Evaluation of the genotoxicity of chitosan nanoparticles for use in food packag
ing films. Journal of Food Science, 75(6), 89–96.
67. Duran, M., Aday, M. S., Zorba, N. N. D., Temizkan, R., Büyükcan, M. B., and Caner, C. (2016).
Potential of antimicrobial active packaging “containing natamycin, nisin, pomegranate and grape seed
368 Chitin and Chitosan: Properties and Applications
extract in chitosan coating” to extend shelf life of fresh strawberry. Food and Bioproducts Processing,
98, 354–363.
68. Perdones, Á., Escriche, I., Chiralt, A., and Vargas, M. (2016). Effect of chitosan–lemon essential oil
coatings on volatile profile of strawberries during storage. Food Chemistry, 197, 979–986.
69. Valenzuela, C., Tapia, C., López, L., Bunger, A., Escalona, V., and Abugoch, L. (2015). Effect of edible
quinoa protein‐chitosan based films on refrigerated strawberry (Fragaria× ananassa) quality. Electronic
Journal of Biotechnology, 18(6), 406–411.
70. Xu, D., Qin, H., and Ren, D. (2018). Prolonged preservation of tangerine fruits using chitosan/
montmorillonite composite coating. Postharvest Biology and Technology, 143, 50–57.
71. Gao, Y., Kan, C., Wan, C., Chen, C., Chen, M., and Chen, J. (2018). Quality and biochemical changes
of navel orange fruits during storage as affected by cinnamaldehyde‐chitosan coating. Scientia
Horticulturae, Amsterdam, Netherlands, 239, 80–86.
72. Mannozzi, C., Tylewicz, U., Chinnici, F., Siroli, L., Rocculi, P., Dalla Rosa, M., and Romani, S. (2018).
Effects of chitosan based coatings enriched with procyanidin by‐product on quality of fresh blueber
ries during storage. Food Chemistry, 251, 18–24.
73. Pilon, L., Spricigo, P. C., Miranda, M., de Moura, M. R., Assis, O. B. G., Mattoso, L. H. C., and
Ferreira, M. D. (2015). Chitosan nanoparticle coatings reduce microbial growth on fresh‐cut apples
while not affecting quality attributes. International Journal of Food Science and Technology, 50(2),
440–448.
74. Lustriane, C., Dwivany, F. M., Suendo, V., and Reza, M. (2018). Effect of chitosan and chitosan‐nano
particles on post harvest quality of banana fruits. Journal of Plant Biotechnology, 45(1), 36–44.
75. McMillin, K. W. (2017). Advancements in meat packaging. Meat Science, 132, 153–162.
76. Schumann, B. and Schmid, M. (2018). Packaging concepts for fresh and processed meat–Recent pro
gresses. Innovative Food Science & Emerging Technologies, 47, 88–100.
77. Quesada, J., Sendra, E., Navarro, C., and Sayas‐Barberá, E. (2016). Antimicrobial active packag
ing including chitosan films with Thymus vulgaris L. essential oil for ready‐to‐eat meat. Foods,
5(3), 57.
78. Fang, Z., Lin, D., Warner, R. D., and Ha, M. (2018). Effect of gallic acid/chitosan coating on fresh pork
quality in modified atmosphere packaging. Food Chemistry, 260, 90–96.
79. Vardaka, V. D., Yehia, H. M., and Savvaidis, I. N. (2016). Effects of Citrox and chitosan on the survival
of Escherichia coli O157: H7 and Salmonella enterica in vacuum‐packaged turkey meat. Food
Microbiology, 58, 128–134.
80. Noshirvani, N., Ghanbarzadeh, B., Gardrat, C., Rezaei, M. R., Hashemi, M., Le Coz, C., and Coma, V.
(2017). Cinnamon and ginger essential oils to improve antifungal, physical and mechanical properties
of chitosan‐carboxymethyl cellulose films. Food Hydrocolloids, 70, 36–45.
81. Rollini, M., Mascheroni, E., Capretti, G., Coma, V., Musatti, A., and Piergiovanni, L. (2017). Propolis
and chitosan as antimicrobial and polyphenols retainer for the development of paper based active
packaging materials. Food Packaging and Shelf Life, 14, 75–82.
82. Vargas, M., Albors, A., and Chiralt, A. (2011). Application of chitosan‐sunflower oil edible films to
pork meat hamburgers. Procedia Food Science, 1, 39–43.
83. Alnoman, M., Udompijitkul, P., and Sarker, M. R. (2017). Chitosan inhibits enterotoxigenic
Clostridium perfringens type A in growth medium and chicken meat. Food Microbiology, 64, 15–22.
84. Ozvural, E. B. and Huang, Q. (2018). Quality differences of hamburger patties incorporated with
encapsulated β carotene both as an additive and edible coating. Journal of Food Processing and
Preservation, 42(1), 1–9.
85. Fan, W. J., Sun, J. X., Chen, Y. C., Qiu, J., Zhang, Y., and Chi, Y. L. (2009). Effects of chitosan coating
on quality and shelf life of silver carp during refrigerated storage. Food Chemistry, 115, 66–70.
86. Duan, J. Y., Cherian, G., and Zhao, Y. Y. (2010). Quality enhancement in fresh and refrigerated lingcod
(Ophiodon elongates) fillets. Food Chemistry, 119, 524–532.
Potential of Chitosans in the Development of Edible Food Packaging 369
87. Souza, B.W. S., Cerqueira, M. A., Ruiz, J. T. M., Casariego, A., Teixeira, J. A., and Vicente, A. A.
(2010b). Effect of chitosan‐based coatings on the shelf life of salmon (Salmo salar). Journal of
Agricultural and Food Chemistry, 58, 11456–11462.
88. Ojagh, S. M., Rezaei, M., Razavi, S. H., and Hosseini, S. M. H. (2010). Effect of chitosan coatings
enriched with cinnamon oil on the quality of refrigerated rainbow trout. Food Chemistry, 120,
193–198.
89. Günlü, A. and Koyun, E. (2013). Effects of vacuum packaging and wrapping with chitosan‐based
edible film on the extension of the shelf life of sea bass (Dicentrarchus labrax) fillets in cold storage
(4°C). Food and Bioprocess Technology, 6(7), 1713–1719.
90. Solval, K. M., Espinoza Rodezno, L. A., Moncada, M., Bankston, J. D., and Sathivel, S. (2014).
Evaluation of chitosan nanoparticles as a glazing material for cryogenically frozen shrimp. LWT – Food
Science and Technology, 57(1), 172–180.
91. Wang, Y., Liu, L., Zhou, J., Ruan, X., Lin, J., and Fu, L. (2015). Effect of chitosan nanoparticle coat
ings on the quality changes of postharvest whiteleg shrimp, litopenaeus vannamei, during storage at
4 °C. Food Bioprocess Technology, 8, 907–915.
92. Wang, Y. and Li, J. (2011). Effects of chitosan nanoparticles on survival, growth and meat quality of
tilapia, Oreochromis nilotica. Nanotoxicology, 5(3), 425–431.
93. Chouljenko, A., Chotiko. A., Bonilla F., Moncad, M., Reyes, V., and Sathivel, S. (2017). Effects of
vacuum tumbling with chitosan nanoparticles on the quality characteristics of cryogenically frozen
shrimp. LWT ‐ Food Science and Technology, 75, 114–123.
94. Ma, Q., Lianga T., Cao, L., and Wang, L. (2018). Intelligent poly (vinyl alcohol)‐chitosan nanoparti
cles‐mulberryextracts films capable of monitoring pH variations. International Journal of Biological
Macromolecules, 108, 576–584.
15
The Use of Chitosan‐Based
Nanoformulations for Controlling
Fungi During Storage of
Horticultural Commodities
Silvia Bautista‐Baños1, Zormy Nacary Correa‐Pacheco2,
and Rosa Isela Ventura‐Aguilar2
1
Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional (IPN),
Yautepec, Morelos, Mexico
2
CONACYT-CEPROBI, Instituto Politécnico Nacional, Yautepec, Morelos, Mexico
15.1 Introduction
Interest in the use of chitosan has increased significantly in comparison to synthetic fun-
gicides. Chitosan can be considered a bioactive natural product, since it possesses impor-
tant attributes such as antimicrobial, low‐toxicity, biocompatibility, and biodegradability
characteristics. Chitosan (poly‐D‐glucosamine) is provided by an extensive deacetylation
of the polymer chitin (poly‐N‐acetyl‐glucosamine) and can be found in fungal cell walls
of Zygomycetes, some insects and algae, and from various marine origin exoskeletons,
including crabs, shrimps, and shells, among others [1]. This biopolymer provides a wide
range of biological functions due to its cationic characteristics (free amino groups present
in its structure). Its interactions with negatively charged structures like those of patho-
genic fungal cell walls are of great interest. Such interactions provide a fungicidal activity
of paramount importance for controlling many species of microorganisms [2, 3]. In addi-
tion, there is plenty of evidence that chitosan is a reliable elicitor, inducing resistance
mechanisms through a systemic induced resistance (SAR) in the affected horticultural
commodity ([4]).
Chitosan can be dissolved in diluted aqueous acidic solvents allowing the formation of
structures, such as edible films and coatings, that combined with some natural antimicrobials
(essential oils and plant extracts, among others) and one or two bioactive products result in
active films/coatings that protect the horticultural commodity against fast moisture and
oxygen losses and from external environmental factors, including temperature, relative
humidity, heat, and microorganisms [5, 6]. At the present time, the application of
nanotechnology has shown great benefits in different fields, such as medicine, energy, the
environment, and food preservation. In the latter, the study of food conservation and control
of microorganisms that cause decay and disease during the postharvest stage through the
designing of new materials (nanostructured coatings) has been noteworthy [6, 7]. The
application of nanotechnology using nanostructured coatings on agricultural products
based on botanical and animal derivatives, such as chitosan, has a remarkable potential to
inhibit microorganisms as compared to the uncoated products. Its application has no
adverse effects on the ripening process of the product [8].
This chapter discusses the current studies and future research on chitosan coating,
formulated alone or with new materials, including nanoemulsions, for preserving fresh
fruits and vegetables during storage, emphasising their effects on the fruit quality and the
control of different microorganisms.
Table 15.1 Response of various fungal pathogens causing plant and fruit diseases to chitosan
application.
Stage of fungal
Fungal disease/microorganism development affected Range of tested doses References
Black rot, leaf, and fruit spot/Alternaria Mycelium, sporulation, 0.1–2.5% (w/v) [27–29]
alternata conidial germination
Aspergillus ear, kernel rot/Aspergillus Mycelium 0.1–5.0 g L−1 [30]
flavus
Black mould/Aspergillus niger Mycelium, conidia [31]
Brown spot/Bipolaris oryzae Germ tube, conidial 300–1000 mg L−1 [32]
germination
Gray mould/Botrytis cinerea Mycelium 0.75–6.0 mg mL−1; [17]
0.01–1.0% (w/v) [33]
Root rot/Cylindrocarpon destructans/; Mycelium 0.5–20 mg mL−1 [34]
damping‐off, root rot, foliage
blight/Cylindrocladium floridanum
Scab of cucurbits/Cladosporium Mycelium 50–1000 μg mL−1 [35]
cucumerinum
Anthracnose/Colletotrichum Mycelium, conidial 0.10–2.5% (w/v) [23, 24, 36]
gloeosporioides sporulation,
germination
Anthracnose/Colletotrichum musae Mycelium 10–2.0 % (w/v) [26]
Damping off/Fusarium acuminatum Mycelium 0.5–2.0 mg mL−1 [34]
Vascular wilt, Fusarium wilt/Fusarium Mycelium 0.5–2.0 mg mL−1; [34]
oxysporum 50–1000 μg mL−1; [35] [37]
0.30–4.5 g L−1
Fusarium wilt/Fusarium oxysporum f.sp. Mycelium, spore 0.6 mg mL−1; 0.5–2.0 [38, 39]
radicis‐lycopersici germination mg mL−1
Root and fruit rot/Fusarium solani Mycelium, sporulation, 1–5 g L−1; 0.05–0.20% [40, 41]
spore germination (w/v)
Ear rot, kernel rot/Fusarium verticillioides Mycelium 1.0–4.0 g L−1 [42]
Take all/Gaeumannomyces graminis var. trici Mycelium 0.5–2.0 mg mL−1 [39]
Basal canker/Leptographium procerum Mycelium 0.05 and 0.075% [43]
Damping off, seedling blight, and root Mycelium 1–5 g L−1 [40]
rot/Macrophomina phaseolina
Brown rot/Monilinia fructicola Mycelium 50–1000 μg mL−1 [35]
Green mould/Penicillium digitatum Mycelium 0.5–3.0% (w/v) [44]
Damping off, basal rot/Pythium sp. Mycelium 0.5–2.0% (w/v) [45]
Damping off, root rot/Pythium ultimum Mycelium 0.5–2.0% (w/v); [39, 45]
0.5–2.0 mg mL−1
Root rot, late blight/Phythophthora Mycelium 0.1–2.0 mg mL−1 [46]
botryosa
Rice blast/Pyricularia grisea Mycelium 5 g L−1 [47]
Collar rot, root rot, wire stem/Rhizoctonia Mycelium 1–5 g L−1 [40]
solani
Soft rot/Rhizopus stolonifer Mycelium 0.75–6.0 mg mL−1; [17] [28]
100–1000 μg L−1; [48]
2–6 g L−1
White mould/Sclerotinia sclerotiorum Mycelium 1–5 g L−1 [40]
Maize head smut, sorghum head Mycelium, sporidia, 10–10 000 μg L−1 [49]
smut/Sphacelotheca reiliana teliospore
Anthracnose, scab /Sphaceloma pelinum Mycelium 1000–100 000 ppm [50]
Diplodia tip blight/Sphaeropsis sapinea Mycelium 0.05 and 0.075% (w/v) [43]
Corn smut/Ustilago maydis Mycelium 10–10 000 μg L−1 [49]
Verticillium wilt/Verticillium dahliae Mycelium 0.5–2.0 mg mL−1 [39]
Control FEC
Volatile compounds 0 17 0 17
Ethyl acetate + nd + +
Butanoic acid methyl ester + nd + +
Butanoic acid, ethyl ester + nd + +
Hexanoic acid, methyl ester + nd + +
Hexanoic acid, ethyl ester + nd + +
3‐Hexen‐1‐ol, (Z) + nd + +
Acetic acid, phenyl methyl ester + nd + +
2,4‐Di‐tert‐butylphenol + nd + +
p‐Mentha‐1(7),8(10)‐dien‐9‐ol + nd + nd
2‐Octyn‐1‐ol nd nd nd +
Decanoic acid, 10‐bromo‐methyl ester nd nd nd +
N‐Methyl‐D‐glucamine nd nd nd +
E‐9‐Tetradecenoic acid nd nd nd +
1‐Butanol, 3‐methyl, acetate + nd + +
4‐Penten‐1‐ol, 3‐methyl, acetate nd nd nd +
2‐Hexen‐1‐ol, acetate, (E) nd nd nd +
3(2H)‐Furanone, 4‐methoxy‐2,5‐dimethyl nd nd nd +
(+)‐3‐Carene nd nd nd +
Alpha‐terpineol nd nd nd +
Camphene nd nd nd +
Beta‐pinene nd nd nd +
Acetic acid pentyl ester nd nd nd +
4‐Hexen‐1‐ol, acetate nd nd nd +
Note: nd: data not available; + flavour component present; FEC: non‐inoculated fruit with chitosan coating.
s upply 16% of magnesium, 19% of iron, and 9% of total calories. Legume vegetables,
such as potatoes, provide about 5% of proteins, which contain high quality essential
amino acids. Other important nutrients supplied by these agricultural commodities include
folacin, riboflavin, zinc, calcium, potassium, and phosphorus. Thus, some human dis-
eases can be closely related to the lack of appropriate intake of nutrients. The most signifi-
cant nutrition‐related disease is chronic undernutrition; other diseases such as beriberi,
scurvy, and anaemia are associated with the lack of vitamin B3, vitamin C, and iron,
respectively, all principally present in fruits and vegetables (https://www.britannica.com/
science/nutritional‐disease).
additional risk due to pathogenic infection is the mycotoxin contamination, which can
occur when the agricultural commodity has undergone further processing. Fungal genera
such as Penicillium, Alternaria, and Fusarium are known to produce mycotoxins under
certain conditions. The main fungi causing postharvest diseases belong to the following
genera: Alternaria, Colletotrichum, Monilinia, Penicillium, Aspergillus, Rhizopus, Botrytis,
Fusarium, and Thielaviopsis [13].
the inhibition was 100% as compared to the untreated fruit, in which little or no disease
control was observed. With regard to this fruit, Kyu Kyu Win et al. [25] mention that in the
cv. Kluai Hom Thong treated with 1% (w/v) chitosan, there was a considerable reduction
of up to 75% in the development of crown rot after five weeks of storage, while Jinasena
et al. [26] reported very similar results with the cv. Embul, since in this case the anthrac-
nose decreased by up to 90% during 14 days of storage.
their intrinsic attributes related to the physical aspects of a product, such as colour, fla-
vour, shape, and appearance, and also from extrinsic attributes which are related to the
nature of the product, such as brand name, stamp of quality, price, country of origin, stor-
age, packaging, and production information, in response to rising concerns of safety,
health, convenience, ethical factors, and nutritional composition [56]. Thus, the quality of
fruits and vegetables will depend on their sensory and functional features, before and after
storage. Hence, postharvest technologies for preserving the product’s quality are limited
to the use of nontoxic compounds to ensure its acceptability by consumers. In this regard,
chitosan is widely used in different areas. For example, the United States Environmental
Protection Agency (EPA) has approved it as a plant growth enhancer, and as a substance
that boosts the ability of plants to trigger certain defence mechanisms against fungal
infections [57]. In addition, Shields et al. [58] reported the use of this polymer as a human
dietary supplement for weight loss and cholesterol reduction. Particularly in the food sec-
tor, the use of chitosan and chitin is regulated by international standards organisations,
such as the Codex Alimentarius Commission and the Japanese Ministry of Health and
Welfare [59, 60].
At present, more than one hundred research papers are published every year on the physi-
ological, biochemical, and physical features of fruits and vegetables treated with chitosan
coatings alone, or enriched with bioactive compounds as a technology for improving their
quality and safety. As outlined previously, horticultural products are highly perishable due
to their composition. Many agricultural commodities have particular compositions that
make them susceptible to deterioration. This includes a volume of water up to 90% of the
total mass, and other compounds that promote microbial growth, including (a) carbohy-
drates, representing 50–80% of the total dry weight, (b) proteins, which represent less than
1% of the fresh mass with the exception of leguminous seeds that are rich in protein, con-
taining up to 15–30%, (c) fats that are usually below fresh 1%, with exceptions in products,
such as avocado (35–70%), olive (30–70%), grapes (0.2%), banana (0.1%), and apple
(0.06%), and (d) vitamins and antioxidants, such as vitamin C (30–150 mg per 100 g fresh
weight) [61]. Consequently, a wide variety of experiments have been conducted on chitosan
coatings, since they preserve different quality parameters of numerous agricultural com-
modities, such as weight. Fruits and vegetables are basically formed by water, and it repre-
sents an economic value during their marketing. The water interchange that occurs between
the film and fruit during storage has been very well explained by Lin et al. [62]. They indi-
cated that, during the interaction between the agricultural commodity and the chitosan coat-
ing, one side, exposed to air, presents a smooth surface, being the chitosan molecular chains
in an orderly arrangement, and the other side, in contact with the pericarp, presents an
irregular face, being the chitosan molecular chains randomly arranged, due to the high con-
tent of moisture. It was observed that the greater roughness resulted in the higher water
vapour permeability (WVP), which meant that the moist surface facilitates the transport of
water. On the contrary, the air‐dried surface prevents the transfer of water, due to its higher
uniform and compact molecular chains (–NH2 and –OH groups of water inside).
Consequently, the firmness and volatile compounds of aroma are preserved, too.
Another important feature is that chitosan coatings neither modify the appearance (firm-
ness, colour, and absence of visual defects) nor the taste of the treated commodities after
application, even if some additives are also incorporated. According to Arnon‐Rips and
Poverenov [63] the advantage of using this coating is that the firmness and texture of
378 Chitin and Chitosan: Properties and Applications
horticultural products can be improved through the addition of Ca2+ ions and other addi-
tives, thus reducing the water vapour permeability of the product by utilising water‐insoluble
or poorly soluble components in the coating formulation, and, therefore, avoiding the
incidence of microorganisms that cause softening as a consequence of the damage in the
cell wall of the products. In addition, the colour of the horticultural products can be
maintained because chitosan is a polymer without colour and it is able to adhere to their
surface. It is possible that the NH3 group with its positive charge is able to form bonds
with the compounds present on the surface. On the other hand, the coatings may limit the
diffusion rate of oxygen; consequently, the oxidation reactions and enzymatic browning,
which cause undesired colour changes, can occur slowly. Additionally, the shape, size,
and brightness of the products are improved by using chitosan coatings, because they
prevent the loss of water and dehydration. Consequently, the deformation of products
is reduced and the gloss on the external surface is perceivable. Also, skin blemishes
are caused by handling, and microbial and mechanical damages are reduced, since the
coatings act as packaging [63, 64].
Sweetness, sourness, and bitterness of the fruit treated with chitosan coatings can be
slightly modified in the first few days after application, but this abnormality disappears
during storage, as reported by Devlieghere et al. [65]. Chitosan solution is normally
adjusted to a pH value below 6, but this value can be modified by interacting with the
constituents of the product until reaching its pH of the product. Regarding aroma, our
working group trials have shown an increase in volatile compounds after 17 days in 0.4%
(w/v) chitosan‐treated strawberries to which an aqueous extract of Roselle calyces (1%
w/v) and cinnamon essential oil (0.025% w/v) were added. Results indicated that at the end
of the storage, terpenes and esters were synthesised. These are compounds that are
recognised for providing pleasant and fruity aromas. Esters are synthesised by esterases
that show considerable activity during the maturation stage of the fruit. Similar compounds
of aroma were reported by Ayala‐Zavala et al. [66]. Thus, it is confirmed that the chitosan
coatings do not cause changes in the aroma of the product after prolonged storage
(Table 15.2). This confirms the fact that chitosan coatings can be a suitable technology for
quality retention including turgidity, colour, aroma, and total soluble solids (TSS) content,
among others.
and firmness. No significant changes were observed for the other evaluated variables, such
as solid soluble content, titratable acidity, pH (pulp), and sugar content. However, starch
degradation decreased. Therefore, the chitosan coating was considered to be effective in
preserving the quality attributes and starch degradation rate for mango.
Additionally, Pagno et al. [71] reported that chitosan coating at 1.5% (w/v) influenced
the nutraceutical quality of tomatoes, delaying the peak of accumulation of β‐carotene and
lycopene, slowing down the loss of caffeic and ferulic acids during storage, and although
transiently, increasing the levels of efficient antioxidants, such as chlorogenic acid (135%),
caffeic acid (121%), and quercetin (115%), until the final day of storage.
Also, an effect of chitosan coatings on the oxidative stress on fruit has been reported
through changes of enzymatic and nonenzymatic antioxidants, and malondialdehyde
(MDA). In this regard, Li et al. [72] studied the oxidative stress of yali pears (Pyrus bretsch-
neideri) and observed that chitosan treatment of pears, before and after being bruised, not
only decreased the MDA content as an indicator of the breakage of cell membranes, but also
enhanced the activities of antioxidant enzymes in this fruit. Based on these results, it can be
hypothesised that chitosan might delay the senescence of damaged pears by means of regu-
lating the antioxidant enzymes system.
Batista et al. [74] evaluated the physicochemical and antioxidant activities of chitosan‐
coated guava using the same coatings. Cosme et al. [69] reported that chitosan coating sup-
pressed the respiratory rate, weight loss, firmness, and skin colour, and the antioxidant
process was reduced for concentrations of 2% and 3%, thus maintaining fruit quality and
delaying ripening during storage for 96 h. On the other hand, Kaya et al. [70] used chitosan
coating to preserve the shelf life of red kiwifruit (Actinidia deliciosa) berries kept at room
temperature (20 ± 2°C) for a period of 26 days. The coating was effective for the first 12
days of storage. The coated berries preserved higher amounts of biologically active com-
pounds (phenolic and ascorbic acid) at the end of the storage period. The decomposition
rate was lower for the coated samples, as evidenced by the changes in soluble solids.
Therefore, chitosan coating was effective in preserving the short shelf life of the berry fruit.
Another study related to mango was undertaken by Jongsri et al. [77] in which the fruit
was coated with chitosan combined with spermidine and evaluated after storage for 9 days
at 25 ± 2°C. The effect of the coating on anthracnose disease of the mango fruit after
inoculation with Colletotrichum gloeosporioides was studied. Combination of both
components in the formulation induced the fruit’s defence mechanism against anthracnose.
The mango softening was reduced due to the decrease of soluble pectin during the ripening
stage. The coating was effective for improving firmness and delaying the deterioration of
postharvest mangoes.
Shen and Yang [78] elaborated the three coating formulations based on chitosan, chitosan
and salicylic acid, and chitosan‐g‐salicylic acid, studying the effects on decay incidence,
quality, and shelf life during the cold storage of grape fruit. The chitosan‐g‐salicylic acid
enhanced the activity of diverse enzymes (lyase, chitinase, and glucanase), also promoting
the accumulation of phenolic compounds with antifungal activity against B. cinerea. Other
benefits were the decrease of respiration rate, decay incidence, and weight loss. Also, TSS,
titratable acidity, and sensory properties were improved due to the coating during storage at
0°C for 42 days. Pomegranate arils were coated with a formulation containing chitosan and
ascorbic acid [79], and stored at 5 ± 1°C for 28 days. During storage, bacterial and fungal
growth was inhibited. The shelf life of the coated arils was extended up to the storage of 21
days, in contrast with noncoated products with a shelf life of less than 10 days.
Awad et al. [80] evaluated the behaviour of bananas coated with a mixture of 2% (w/v)
chitosan and 1% (w/v) gallic acid. The inclusion of the gallic acid compound into the
chitosan matrix improved the antioxidant properties of the fruit. Moreover, the chitosan–
gallic acid coating delayed the fruit ripening as reflected by a higher firmness, lower TSS
and TSS/acid ratio, and slightly higher vitamin C (0.064 g kg−1) during storage at 20 ± 2°C
as compared to the control group.
Regarding the incorporation of essential oils into chitosan‐based formulations, Shahbazi
[81] applied a carboxymethyl cellulose‐chitosan coating containing mentha (Mentha
spicata) essential oil to fresh strawberries to study the antimicrobial effect and the
physicochemical and sensory properties after 12 days of shelf life extension. It was tested
against Listeria monocytogenes, being the coating effective as an antimicrobial agent. Also,
positive effects for weight loss, titratable acidity, pH, water vapour resistance, and
respiration rate were observed for the coating used as an active packaging.
Yuan et al. [82] prepared chitosan coatings, and the in vitro and in vivo activities of
essential oils as antioxidants, antibacterials, and antifungals were evaluated. Their use in
food composites was also considered. The efficacy of essential oils as antioxidants, anti-
bacterials, and antifungals was increased for the in vitro test. Also, the coating extended the
shelf life of fish and meat products.
Perdones et al. [83] studied the volatile profile of strawberries coated with chitosan‐
lemon essential oil during storage at 20°C for 7 days. They found that the coating was
effective for controlling the fruit’s fungal decay, affecting the metabolic pathways and
volatile profile of the strawberries, and also enhancing the perception of aroma. Coatings
based on chitosan and M. piperita or M. villosa essential oils were tested on cherry tomatoes
by Dantas et al. [84] to prevent postharvest mould infections caused by Aspergillus niger,
B. cinerea, Penicillium expansum, and R. stolonifer, and to maintain the quality of the fruit.
The physicochemical and sensory properties were tested for two storage conditions: at
room temperature (25°C for 12 days) and low temperature (12°C for 24 days), measuring
The Use of Chitosan-Based Nanoformulations for Controlling Fungi During Storage 381
the minimum inhibitory concentration (MIC) for the different fungi. The results showed
mycelial growth and spore germination inhibition for concentrations of 4 mg mL−1
(chitosan) and 2.5 or 1.25 μL mL−1 for the essential oils for both tested temperatures. Also,
physicochemical and sensory properties such as weight loss, firmness, titratable acidity,
and soluble solids were not affected.
Antimicrobial activity of chitosan coatings and films against L. monocytogenes on
black radish was assessed by Jovanović et al. [85]. Chitosan‐gelatin coatings were evalu-
ated on shredded black radish samples stored for 7 days at 4°C. The highest antibacterial
activity was observed for chitosan prepared with acetic acid, and for films with higher
chitosan concentration and essential oil incorporation. Especially films with 1% (w/v)
chitosan and 0.2% (w/v) thyme essential oil were most effective. However, these films
had a negative impact on the consumers due to the flavour and aroma of coated radish.
Therefore, mint essential oil, also tested, but having lower antibacterial activity, was
selected, because it had the least negative impact on the sensory properties mentioned
earlier.
With regard to the interaction between chitosan and essential oils, Devlieghere et al. [65]
showed that, during this interaction, the chitosan is positioned at the outside of the emulsion
drops, due to the interaction between the positively charged chitosan and the negatively
charged free fatty acids. In this way, the chitosan formed larger positively charged drops
that still maintained their antimicrobial activity. Additionally, when the essential oil is used
in combination with chitosan, the antimicrobial effect can be enhanced. This is possible
because essential oils are complex mixtures of volatile metabolites that can be isolated by
pressing and distillation from a whole plant or plant part. The main compounds contained
in the essential oils are derived from three biosynthetic pathways: 1) the mevalonate
pathway leading to sesquiterpenes, 2) the methyl‐erythritol pathway leading to mono and
diterpenes, and 3) the shikimic acid pathway en route to phenylpropenes. Each one has a
specific mode of action on the microorganism. Dorman and Deans [86] explained that the
antimicrobial activity can be enhanced by the effect of the functional groups present in the
essential oil. There are three examples of this: 1) in phenols, an alkyl substitution occurs in
the nucleus and this chemical operates to affect gram‐negative bacterial growth, 2) the
bacteriostatic and fungistatic actions of terpenoids are increased when they are carbonylated,
i.e., when a carbon atom is bonded to an oxygen atom by a double bond, and 3) an aldehyde
group conjugated to a carbon‐to‐carbon double bond is a highly electronegative arrangement
that can interfere in biological processes involving electron transfer and react with vital
nitrogen components, e.g. proteins and nucleic acids. Therefore, it is able to inhibit the
growth of the microorganisms, among others.
Other bioactive compounds with antimicrobial and antioxidant properties, such as plant
extracts, have been incorporated into chitosan‐based formulations. Plant extracts used for
edible coatings are obtained from fruits and herbs, thus adding medicinal benefits to the
coating. Edible coatings incorporated with plant extracts can replace non‐biodegradable
polyethylene plastic films used for the packaging of fruits and vegetables allowing product
cooling, among other benefits [63].
On this subject, Sneha et al. [87] studied the effect of chitosan‐alginate‐based coatings
enriched with pomegranate peel extract to extend the shelf life of guava. The formulation
composition was 1% (w/v) chitosan, 2% (w/v) alginate, and 1% (w/v) of the plant extract.
After the storage of 20 days at 10°C, changes in respiration, ripening, and colour were
382 Chitin and Chitosan: Properties and Applications
observed. The guavas coated with the pomegranate plant extract maintained their quality
compared to the uncoated ones. The visual attributes and nutritional properties of the guava
were improved, retarding respiration rate and delaying senescence. Likewise, Duran et al.
[88] observed that the strawberries treated with chitosan 1.5% (w/v) and pomegranate seed
extract 1% (w/v) showed a reduction of mesophilic bacteria count, but the surface colour
and texture profile analysis were not improved.
Chitosans incorporated with different concentrations of phenolic compound from
blueberry leaf‐and‐fruit extracts were tested by Yang et al. [87] against Staphylococcus
aureus, L. monocytogenes, Salmonella typhimurium, and Escherichia coli to preserve the
postharvest quality of fresh blueberries. Fruits were stored at 2 ± 1°C for 35 days and then
for 3 days at room temperature. Five concentrations were used containing 2% (w/v)
chitosan and 4–12% (w/v) plant extracts. Also, for the 12% (w/v) plant extract, a modified
atmosphere packaging (3 kPa O2 + 12 kPa CO2) was used. Proper antimicrobial activity
against tested microorganisms was observed, for a minimum inhibition concentration from
25 to 50 g L−1. More effective control on fruit decay was obtained for the modified
packaging atmosphere with 12% (w/v) leaf plant extract. All treated samples maintained
higher nutritional properties, total phenolic content, and radical scavenging activity as
compared to the control.
Breda et al. [90] did not find a clear significant difference of colour, firmness, or
weight loss in tomatoes coated with chitosan alone or in combination with the extract
of pequi (4:1) (Caryocar brasiliense), and stored at 22 ± 2°C. However, chitosan coat-
ings and extracts of pequi showed benefits by means of maintaining 70% lower levels
of mould and yeast counts during all storage periods of tomatoes than the control group.
Sun et al. [91] reported that carvacrol at ≥0.1% (w/v) or trans‐cinnamaldehyde at ≥0.2%
(w/v) incorporated with chitosan coatings improved the antimicrobial activity against
E. coli and Penicillium digitatum, and retarded the loss of firmness of blueberries dur-
ing storage. The results suggest that the antimicrobial properties of the compounds
added to chitosan, and their effect on quality will depend on their concentration and
structure.
Studies on propolis or bee glue, a resinous mixture produced by honey bees, have dem-
onstrated effective antimicrobial characteristics; therefore, its incorporation into chitosan
coatings has also been the subject of experimentation. For example, Migliori et al. [92]
tested a propolis coating on “Vesuviano” tomatoes at room temperature for 120 days. The
coating formulation was compared with the effect of thyme essential oil and chitosan on
the postharvest fruit quality and organoleptic properties, and carotenoids and phenol con-
tents were evaluated. Thyme essential oil decreased the number of rotten fruits after stor-
age of 40 days. However, propolis and chitosan had the same effect after 80 days.
Moreover, chitosan retarded the senescence for the whole storage period. The postharvest
quality of the fruit was not affected by the different formulations. However, total soluble
sugar (for chitosan and thyme), total carotenoids and flavonols (for chitosan), and total
organic acids and terpenes (for propolis) were better retained during the postharvest
period. As a conclusion, the best result as a fungicide was obtained for chitosan, which
most effectively reduced the fruit’s senescence maintaining tomato quality for a long
period of time.
In Tables 15.3 and 15.4, the different coatings, bioactive components, and their effects
on the horticultural commodity are summarised.
The Use of Chitosan-Based Nanoformulations for Controlling Fungi During Storage 383
Table 15.3 Chitosan edible coatings on preserving the quality and nutraceutical compounds of fruits
and vegetables.
Topics References
• Potential applications and modes of action of chitosan and its derivatives as [93]
antimicrobials: a review
• Chitosan as a novel edible coating for fresh fruits: a review [94]
• Influence of chitosan coating on loquat’s nutritional quality during cold storage [95]
• Chitosan coating and storage effect on food storage chamber in banana stored at 25°C [96]
• Chitosan coating at concentrations from 0.7 to 2 g L−1 on physicochemical, nutritional, [97]
and microbiological parameters of fresh‐cut “Rocha” pear
• Chitosan coating on the enzymatic antioxidant system and the membrane integrity [98]
maintenance of sweet cherry during cold storage
• Effect of chitosan/nanosilica on enhancing chilling tolerance in loquat fruit during cold [99]
storage
• Edible coatings combined with repetitively pulsed light treatment on microbiological [100]
stability, quality, and physicochemical changes of fresh‐cut cantaloupes during storage
at 4°C
• Application of Cu‐nanoparticles, introduced in chitosan–polyvinyl alcohol hydrogels, [101]
on the growth of jalapeño pepper plants, the antioxidant content in fruits, and their
postharvest characteristics
• Improvement in the fruit storability of Punica granatum using prestorage chitosan [102]
coating technique
• Chitosan coating added with rosemary extracts, chitosan with ascorbic acid on the [103]
browning characteristics, and the physicochemical and sensory properties of fresh‐cut pears
Coating/bioactive
component Effects Fruit References
Chitosan/procyanidin Decreased yeast and mould growth, intact Blueberry [76]
firmness, and increased antioxidant activity
Chitosan/aloe vera Microbial growth and water loss were reduced Blueberry [75]
Chitosan Climacteric peak, water loss, and firmness were Mango [69]
delayed
Chitosan/spermidine Defence mechanism against anthracnose was Mango [77]
induced. Firmness was improved and
deterioration of fruit was delayed.
Chitosan Respiratory rate, weight loss, firmness, and Guava [74]
antioxidant process were reduced. Ripening
was delayed.
Chitosan Biologically active compounds (phenolics and Red kiwifruit [70]
ascorbic acid) were preserved. Lower berries
decomposition rate.
Chitosan, chitosan/ Activities of lyase, chitinase, and glucanase Grape [78]
salicylic acid, and enzymes were enhanced. Decrease on
chitosan‐g‐salicylic respiration rate, decay incidence, and weight
acid loss.
Chitosan/ascorbic acid Bacterial and fungal growth was inhibited. Pomegranate [79]
arils
Chitosan‐ carboxymethyl The coating effective as antimicrobial agent. Strawberries [81]
cellulose/Mentha Positive effects for weight loss, titratable acidity,
spicata essential oil pH, water vapour resistance, and respiration rate.
(Continued)
384 Chitin and Chitosan: Properties and Applications
Coating/bioactive
component Effects Fruit References
Chitosan/lemon essential Fruit fungal decay was controlled. Aroma Strawberries [81]
oil perception was enhanced.
Chitosan/Mentha piperita Inhibition of mycelial growth and spore Cherry [84]
L. or Mentha × villosa germination. Physicochemical and sensory tomato
Huds. essential oils properties did not change during storage.
Chitosan‐gelatin/thyme Higher antibacterial activity, higher flavour and Black radish [85]
or mint essential oil aroma (thyme), and better sensory properties
with low antibacterial activity (mint).
Chitosan‐alginate/ Visual attributes and nutritional properties were Guava [87]
pomegranate peel improved. Respiration rate and senescence
extract were delayed.
Chitosan/phenolic Good antimicrobial activity. Effective control of Blueberry [89]
compound from fruit decay.
blueberry
Chitosan vs. propolis vs. Total soluble sugar (for chitosan and thyme), total Tomato [92]
thyme essential oil carotenoids and flavonols (for chitosan), and
total organic acids and terpenes (for propolis)
were retained. The best result as fungicide was
obtained for chitosan.
soluble solids, and sugar contents were reduced. Moisture retention was increased and
titratable acidity was intact. In conclusion, the postharvest quality of the grapes was pre-
served with the use of the edible coating.
Polyphenols of strawberry extract were encapsulated in chitosan nanoparticles and the
release kinetics was studied by Pulicharla et al. [108]. Their final results showed that the
encapsulation increased the bioavailability and release effectivity of the phytochemical.
The encapsulation efficiency was 58% of polyphenols. With regard to the incorporation of
essential oils into chitosan nanoparticles, Sotelo‐Boyás et al. [109] also studied the release
kinetics and inhibitory effect of thyme essential oil‐loaded chitosan nanoparticles and
nanocapsules against six foodborne bacteria (S. aureus, L. monocytogenes, Bacillus cereus,
S. typhi, Shigella dysenteriae, and E. coli). The results showed 68% of encapsulation
efficiency. The release from thyme was faster for nanoparticles than for nanocapsules. The
highest inhibitory effect was observed against S. aureus for the nanoparticles.
The antimicrobial activity and stability of cinnamon (Cinnamomum zeylanicum)
essential oil‐loaded chitosan nanoparticles against Phytophthora drechsleri as a coating on
cucumber were studied by Mohammadi et al. [110] during 7 days of storage at 4°C and 2–3
additional days at 20°C. The physicochemical properties were evaluated. Encapsulation
efficiency was about 2–17%. Encapsulated oil decreased the disease severity and incidence
more efficiently in Phytophthora‐inoculated cucumbers. The shelf life of cucumbers coated
with cinnamon essential oil‐loaded chitosan nanoparticles was extended up to 21 days at 10
± 1°C, while it was 15 days for the uncoated vegetable. As a conclusion, the coating
improved the physicochemical quality of the cucumbers, preserving firmness, and main-
taining colour and water content. Lower microbial counts were found for the coated fruit.
Mohammadi et al. [111] studied the in vitro and in vivo antifungal activities of satar (Zataria
multiflora) essential oil encapsulated in chitosan nanoparticles on strawberries. The encap-
sulation efficiency was 45%. The coating was effective in controlling the incidence and
severity of B. cinerea in inoculated strawberries during the storage for 7 days at 4°C fol-
lowed by 2–3 more days at 20°C. The same essential oil was tested on cucumber by
Mohammadi et al. [112], who studied the antioxidant activity on the shelf life extension of
nano‐chitosan‐based coating with the addition of this essential oil for a 21‐day storage
period. The coating improved the quality of cucumber and protected the bioactive
compounds during storage. Firmness, respiration rate, and 2,2‐diphenyl‐1‐picrylhydrazyl
(DPPH) radical scavenging capacity were best kept when the coating was used. The coating
was effective in inhibiting the growth of total aerobic bacteria, yeasts, and mould.
On the other hand, the antibacterial activity of chitosan nanoparticles and nanocapsules
incorporated with lime essential oil was assessed for S. aureus, L. monocytogenes, S. dys-
enteriae, and E. coli by Sotelo‐Boyás et al. [113]. It was found that the antibacterial activity
of the nanoparticles was higher than that for nanocapsules, and the highest inhibition halo
was found for S. dysenteriae for 40 mL of minimum inhibitory volume.
The in vitro and in vivo activities of the coating of chitosan nanoparticles and chitosan‐
thyme (Thymus sp.) essential oil nanoparticles against C. gloeosporioides on avocado cv.
“Hass” and its fruit quality were evaluated by Correa‐Pacheco et al. [8] after 6 days of stor-
age. The fungal activity and fruit quality were evaluated. In vitro results showed that the
growth of C. gloeosporioides was completely controlled with the incorporation of thyme
essential oil into the nanoparticles at thyme essential oil concentrations of 3% (w/v) and 5%
(w/v). A coating composed of 55% of chitosan‐thyme essential oil nanoparticles resulted in
386 Chitin and Chitosan: Properties and Applications
a reduction of up to 60% of anthracnose disease without affecting the quality of the fruit. In
addition, firmness was better maintained for the coated fruit in comparison with the uncoated
one. On the other hand, the use of plant extracts incorporated into chitosan nanoparticles
was assessed by Barrera‐Necha et al. [114]. In this study, the antifungal activity of chitosan
nanoparticles loaded with ethanolic extracts of nanche (Byrsonima crassifolia) and blue-
berry (Vaccinium corymbosum) was evaluated on A. alternata isolated from fig (Ficus car-
ica) and rosemary (Rosmarinus officinalis) and from C. gloeosporioides isolated from
papaya and soursop. The ethanolic extract of nanche was effective on C. gloeosporioides
showing the mycelial growth inhibition of 79% and 82% for the fungi isolated from papaya
and soursop, respectively. Germination inhibition was 100%. Furthermore, the ethanolic
extract of blueberry inhibited the mycelial growth of A. alternata by 83% and 76% for fig
and rosemary, respectively, and germination inhibition was 75% for rosemary.
Tables 15.5 and 15.6 summarise the chitosan nanoparticle–based coatings and the effects
of pathogenic microorganism development on fruits and vegetables.
Coating/bioactive Fruits/
component Effects of the coating vegetables References
Chitosan nanoparticles Good antifungal and antioxidant activities. Tomato, [106]
(NPs) Reduction of weight loss. chilly, and
eggplant
Chitosan Good antibacterial activity. Fruit ripening was Grape [107]
delayed. Reduction on weight loss, soluble
solids, and sugar contents. Moisture retention
was increased. Titratable acidity was preserved.
Chitosan/Cinnamomum Disease severity and incidence were decreased. Cucumber [110]
zeylanicum essential oil Firmness, colour, and water content were intact.
Chitosan/Zataria multiflora Firmness, respiration rate, and DPPH radical Strawberry [111]
essential oil scavenging capacity were preserved. Bacteria,
yeast, and mould growth were inhibited.
Chitosan/thyme essential Good fungal inhibition. Incidence of fungi was Avocado [8]
oil reduced and firmness was maintained.
The Use of Chitosan-Based Nanoformulations for Controlling Fungi During Storage 387
15.11 Outlook
Horticultural products have proven to be an excellent source of minerals, vitamins, and
functional compounds for human consumption, resulting in food production evolving from
a local to a globalised environment. Contamination of fruits and vegetables by fungal and
bacterial pathogens that causes serious economic losses to producers, traders, and consum-
ers can occur any time from early growth to harvest and during handling and final market-
ing operations.
Controlling practices include chemical and nonchemical methods. Within the last few
years, coatings have become increasingly more important as a means to deliver fresh hor-
ticultural commodities to the consumer in safe conditions. Currently, there is a growing
trend to design and evaluate formulations based on nontoxic, naturally occurring com-
pounds. In this respect, chitosan‐based coatings can be a good alternative as an active food
packaging allowing the shelf life extension and quality preservation of fruits and vegeta-
bles by means of controlling the growth of microorganisms. Development of coatings
based on versatile biopolymers like chitosan will have an important role in promising hor-
ticultural applications, including those for food preservation. The incorporation and release
of bioactive compounds, such as antimicrobials, antioxidants, and nutrients, among others,
will produce deliverable solutions for protection against pathogens, enhancing at the same
time with sensory and functional properties. Certainly, this will depend on the interactions
among the coating, the bioactive ingredients, and the fruit or vegetable at issue.
We firmly believe that the use of nanotechnology will enable some drawbacks of the
existing edible coatings to be solved and better functional properties obtained. However,
ethical, security, and legislative problems regarding the use of nanotechnology, and scaling
from the laboratory to industry are issues that must be resolved. At present, there is a need
for further research related to the use of chitosan or chitosan nanobased formulations for
the control of microorganisms during the storage of fruits and vegetables, in particular
emphasising the incorporation of nontoxic compounds.
Acknowledgments
The authors wish to thank Mexico’s National Science and Technology Council for support-
ing professorships of Rosa Isela Ventura‐Aguilar (No. 772) and Zormy Correa‐Pacheco
(No. 1549).
References
1. Rodríguez Pedroso, T., Ramírez Arrebato M.A., Rivero‐González, D., Bosquez Molina, E., Barrera
Necha, L.L., and Bautista Baños, S. (2009). Propiedades químico‐estructurales y actividad
biológica de la quitosana en microorganismos fitopatógenos. Revista Chapingo, serie Horticultura,
15, 307–317.
2. Bautista‐Baños, S., Hernández‐Lauzardo, A.N., Velázquez del Valle, M.G., Hernández‐López, M.,
Ait Barka, E., Bosquez‐Molina, E., and Wilson, C.L. (2006). Chitosan as a potential natural com-
pound to control pre‐ and postharvest diseases of horticultural commodities. Crop Protection, 25,
108–118.
3. Bautista‐Baños, S., Barrera‐Necha, L.L. Hernández‐López, M., and Rodríguez‐González, F. (2016).
Morphological and ultrastructural modifications of chitosan‐treated fungal phytopathogens. In: Chitosan
in the Preservation of Agricultural Commodities (Bautista‐Baños, S., Romanazzi, G., Jiménez‐Aparicio,
A. eds.). Academic Press/Elsevier, USA. pp. 251–275.
388 Chitin and Chitosan: Properties and Applications
4. Romanazzi, G. and Feliziani, E. (2016). Use of chitosan to control postharvest decay of temperate
fruit: Effectiveness and mechanisms of action. In: Chitosan in the Preservation of Agricultural
Commodities (Bautista‐Baños, S., Romanazzi, G., and Jiménez‐Aparicio, A. eds.). Academic Press/
Elsevier, USA. pp. 155–177.
5. Sivakumar, D., Bill, M., Korsten, L., and Thompson, K. (2016). Integrated application of chitosan
coating with different postharvest treatments in the control of postharvest decay and maintenance of
overall fruit quality. In: Chitosan in the Preservation of Agricultural Commodities (Bautista‐Baños, S.,
Romanazzi, G., and Jiménez‐Aparicio, A. eds.). Academic Press/Elsevier, USA. pp. 127–153.
6. Bautista Baños, S., Correa Pacheco, Z., Barrea Necha, L.L., and Ventura Aguilar, R.I. (2017). Inserción
del CEPROBI en el campo de la nanotecnología con temas sobre la conservación de productos agrícolas.
In: Agronanotecnología: nuevo paradigma científico en la producción de alimentos (Lira, S.R.H.,
Méndez, A.B., and Vera, R.I. eds.). CIQA, Saltillo Coahuila, Mexico. pp. 88–103.
7. Sotelo‐Boyás, M.E., Bautista‐Baños, S., Correa‐Pacheco, Z.N., Jiménez‐Aparicio, A. y Sivakumar, D.
(2016). Biological activity of chitosan nanoparticles against pathogenic fungi and bacteria. In:
Chitosan in the Preservation of Agricultural Commodities (Bautista‐Baños, S., Romanazzi, G., and
Jiménez‐Aparicio, A. eds.). Academic Press/Elsevier, USA. pp. 339–349.
8. Correa‐Pacheco, Z., Bautista‐Baños, S., Marquina‐Valle, M., and Hernández‐López, M. (2017). The
effect of nanostructured chitosan and chitosan‐thyme essential oil coatings on Colletotrichum gloe-
osporioides growth in vitro and on cv. Hass avocado and fruit quality. Journal of Phytopathology, 165,
297–305.
9. FAO (2017). Fruit and vegetables for healthy initiative. Food and Agriculture Organization of the
United Nations. p. 8.
10. Bautista‐Baños, S., Velázquez‐del Valle, M.G., Hernández‐Lauzardo A.N., and Ait Barka, E.
(2008).The Rhizopus stolonifer‐tomato interaction. In: Plant Microbe Interaction (Ait Barka, E. and
Clement, C. eds.). Research Signpost, Kerala, India. pp. 269–289.
11. Kader, A. (2001). Importance of fruits, nuts, and vegetables in human nutrition. Perishable Handling
Quarterly, 106, 1–6.
12. Wills, R., McGlasson, B., Graham, D., and Joyce, D. (2004). Postharvest. An introduction to the physiol-
ogy & handling of fruit, vegetables & ornamental. CAB International, Adelaide, South Australia. p. 262.
13. Hernández‐Albíter, C., Bravo‐Luna, L., Bautista‐Baños, S., and Barrera‐Necha, L.L. (2005). Actividad
antifúngica de extractos vegetales en hongos postcosecha. Estado actual del conocimiento. Biótica, 2,
115–155.
14. Coates, L.M. and Johnson, G.I. (1997). Postharvest diseases of fruit and vegetables. In: Plant Pathogens
and Plant Diseases (Brown, J. and Ogle, H. eds.). Rockvale Publications. pp. 533–547.
15. Ramos‐García, M., Bautista‐Baños, S.,Barrera‐Necha, L.L., Bosquez‐Molina, E., Alia‐Tejacal, I., and
Estrada Carrillo, M. (2010). Compuestos antimicrobianos adicionados en recubrimientos comestibles
para uso en productos hortofrutícolas. Revista Mexicana de Fitopatología, 28, 44–57.
16. Hernández‐Téllez, C.N., Plascencia‐Jatomea, M., and Cortez‐Rocha, M.O. (2016). Chitosan‐based
bionanocomposites: development and perspectives in food and agricultural applications. In: Chitosan
in the Preservation of Agricultural Commodities (Bautista‐Baños, S., Romanazzi, G., and Jiménez‐
Aparicio, A. eds.). Academic Press/Elsevier, USA. pp. 315–338.
17. El Ghaouth, A., Arul, J., Grenier, J., and Asselin, A. (1992). Antifungal activity of chitosan on two
postharvest pathogens of strawberry fruits. Phytopathology, 82, 398–402.
18. Romanazzi, G., Nigro, F., Ippolito, A., Venere, Di., and Salerno, M., (2002). Effects of pre‐ and post-
harvest chitosan treatments to control storage gray mold of table grapes. Journal of Food Science, 67,
1862–1867.
19. Jiang, Y.M., Zhang, Z.Q., Joyce, D.C., and Ketsa, S. (2002). Postharvest biology and handling of lon-
gan fruit (Dimocarpus longan Lour.) Postharvest Biology and Technology, 26, 241–252.
20. Zhang, D. and Quantick, P.C. (1997). Effects of chitosan coating on enzymatic browning and decay
during postharvest storage of litchi (Litchi sinensis Sonn.) fruit. Postharvest Biology and Technology,
12, 195–202.
The Use of Chitosan-Based Nanoformulations for Controlling Fungi During Storage 389
21. Sivakumar, D., Sultanbawa, Y., Ranasingh, N., Kumara, P., and Wijesundera, R.L.C. (2005). Effect of
the combined application of chitosan and carbonate salts on the incidence of anthracnose and on the
quality of papaya during storage. Journal of Horticultural Science and Biotechnology, 80, 447–452.
22. Al Eryani‐Raqeeb, A., Mahmud, T.M.M., Syed Oar, S.R., Mohamed Zaki, A.R., and Al Eryani, A.R.
(2009). Effects of calcium and chitosan treatments on controlling anthracnose and postharvest quality
of papaya (Carica papaya L.). International Journal of Agricultural Research, 4, 53–68.
23. Muñoz, Z., Moret, A., and Garcés, S. (2009). Assessment of chitosan for inhibition of Colletotrichum
sp. on tomatoes and grapes. Crop Protection, 28, 36–40.
24. Gutiérrez‐Martínez, P., Bautista‐Baños, S., Berúmen‐Varela, G., Ramos‐Guerrero, A., and Hernández‐
Ibañez, A.M. (2017). In vitro response of Colletotrichum to chitosan. Effect on incidence and quality
on tropical fruit. Enzymatic expression in mango. Acta Agronómica, 66(2), 282–289.
25. Win N.K.K., Jitareetat, P., Kanlayanarat, S., and Sangchote, S. (2007). Effects of cinnamon extracts,
chitosan coating, hot water treatments and their combination on crown rot disease and quality of
banana fruit. Postharvest Biology and Technology, 45, 333–340.
26. Jinasena, D., Pathirathana, P., and Wickramarachi Marasinghe, E. (2011). Effect of chitosan (unirradi-
ated and irradiated) treatments on anthracnose disease and its potential to increase the shelf life of
“Embul” banana. IJEST, 2, 248–252.
27. Sánchez‐Dómínguez, D., Bautista‐Baños, S., and Castillo‐Ocampo, P. (2007). Efecto del quitosano en
el desarrollo y morfología de Alternaria alternata (Fr.:Fr.) Keissl. Annals of Biology, 29, 23–32.
28. de Oliveira, Jr. E.N., El Gueddari, N.E., Moershbacher, B.M., and Franco, T.T. (2012). Growth rate
inhibition of phytopathogenic fungi by characterized chitosans. Brazilian Journal of Microbiology, 80,
800–809.
29. López, M.L.I., Gutiérrez, M.P., Bautista, B.S., Jiménez, G.L.F., and Zavaleta, M.H.A. (2013).
Evaluación de la actividad antifúngica del quitosano en Alternaria alternata y en la calidad del
mango cv. ‘Tommy Atkins’ durante el almacenamiento. Revista Chapingo serie Horticultura, 19,
315–331.
30. Plascencia‐Jatomea, M., Viniegra, G., Olayo, R., Castillo‐Ortega, M.M., and Shirai, K. (2003). Effect
of chitosan and temperature on spore germination on Aspergillus niger. Macromolecular BioScience,
3, 582–586.
31. Li, X.‐F., Feng, X.‐Q., Yang, S., Wang, T.‐P., and Su, Z.‐X. (2008). Effects of molecular weight and
concentration of chitosan on antifungal activity against Aspergillus niger. Iranian Polymer Journal
2008, 17, 843–852.
32. Rivero, D., Cruz, A., Martínez, B., Rodríguez, A.T., and Ramírez, M.A. (2008). Actividad antifúngica
in vitro de las quitosanas Ki y Sigma frente a Bipolaris oryzae (B. de Haan) Shoem. Revista de
Protección Vegetal, 23, 43–47.
33. Pak, H.A., Rasim, S, Manning, M.A., and Fullerton, R.A. (1998). Soft fungicides for the control of
Botrytis cinerea in vitro and in vivo on kiwifruit. Proceedings of the 51st International Plant Protection
Conference. New Zealand Plant Protection Society. p. 123–129.
34. Laflamme, P., Benhamou, N., Bussières, G., and Dessureault, M. (1999). Differential effect of chitosan
on root rot fungal pathogens in forest nurseries. Canadian Journal of Botany 1999, 77, 1460–1468.
35. Ren, J., Liu, J., Li, R., Dong, F., and Guo, Z. (2012). Antifungal properties of chitosan salts in labora-
tory media. Journal of Applied Polymer Science 124, 2501–2507.
36. Bautista‐Baños, S., Hernández‐López, M., Trejo‐Tapia, J.L., Hernández‐Lauzardo, A.N., Bautista‐
Cerón, M.K., and Melo‐Giorgana, M.G. (2005). Effect of chitosan on in vitro development and mor-
phology of two isolates of Colletotrichum gloeosporioides Penz. Mexican Journal of Phytopathology,
23, 62–67.
37. Ragab, M.M.M., Ashour, A.M.A., Abdel‐Kader, M.M., and Abderl‐Aziz, A. (2012). In vitro evaluation
of some fungicides alternatives against Fusarium oxysporum the casual of wilt disease of pepper
(Capsicum annum L.) IJAF, 2, 70–77.
38. Benhamou, N. (1992). Ultrastructural and cytochemical aspects of chitosan of Fusarium oxysporum f.
sp. radicis‐lycopersici, agent of tomato crown and root rot. Phytopathology, 82, 1185–1193.
390 Chitin and Chitosan: Properties and Applications
39. Palma‐Guerrero, J., Jansson, H.‐B., Salinas, J., and Lopez‐Llorca, L.V. (2008). Effect of chitosan on
hyphal growth and spore germination of plant pathogenic and biocontrol fungi. Journal of Applied
Microbiology, 104, 541–553.
40. El‐Mohamedy, R.S.R., Abdel‐Kader, M.M., El‐Karem, F.A., and El‐Mougy, N.S. (2013). Inhibitory
effect on antagonistic bio‐agents and chitosan on the growth of tomato root rot pathogens in vitro.
IJAT, 9, 1521–1533.
41. Bhattacharya, A. (2013). Fungicidal potential of chitosan against phytopathogenic Fusarium solani.
JEBAS 2013, 1, 259–263.
42. Quintana‐Obregón, E.A., Plascencia‐Jatomea, M., Sánchez‐Mariñez, R.I., Rosas‐Burgos, E.C., and
Cortez‐Rocha, M.O. (2010). Inhibición del crecimiento radial “in vitro” de la Fusarium verticillioides
en presencia de quitosano. Revista Iberoamericana de Polímeros, 11, 386–391.
43. Chittenden, C. and Singh, T. (2009). In vitro evaluation of combination of Thrichoderma harzianum
and chitosan for the control of sapstain fungi. Biological Control, 50, 262–266.
44. Bautista‐Baños, S., Hernández‐López, M., and Bosquez‐Molina, E. (2004). Growth inhibition of
selected fungi by chitosan and plant extracts. Mexican Journal of Phytopathology, 22, 178–186.
45. Arias, R.B., McGee, D.C., and Burris, J.S. (1998). Evaluación del potencial de polímeros como agen-
tes envolventes de fungicidas en el tratamiento de semillas de maíz para el control de Pythium spp.
Agronomía Tropical, 48(4), 471–448.
46. Sunpapao, A. and Pornsuriya, C. (2013). Chitosan inhibits the growth of Phytophthora botryose: The
casual agent of para rubber leaf fall disease. Plant Pathology Journal, 12(2), 92–97.
47. Badawy, M.E.I., Rabea, E.I., Steurbaut, W., Rogge, T.M., Stevens, C.V., Smagghe, G., and Höfte, M.
(2005). Fungicidal activity of some O‐acyl chitosan derivatives against grey mould Botrytis cinerea and
rice leaf blast Pyricularia grisea. Communications in Agriculture and Applied Biology, 70, 215–218.
48. Yarahmadi, M., Safaei, Z., and Azizi, M. (2014). Study the effect of chitosan, vanillin, and acetic acid
on fungal diseases control of Rhizopus stolonifer in strawberry fruits in vitro and in vivo. EJEBAU,
2014, 219–2125.
49. Kosiada, T. (2011), In vitro influence of selected fungicides on Sphacelothea reiliana and Ustilago
maydis. Journal of Plant Protection Research, 51(4). Doi: 10.2478/v10045‐011‐0056‐2.
50. Singburaudom, N., Piasi, O., and Dethaub, T. (2011). Antimicrobial activity of different molecular
weight chitosans to inhibit some important plant pathogenic fungi. Natural Science, 4, 644–655.
51. Bautista‐Baños, S., Hernández‐Lauzardo, A.N., Velázquez del Valle, M.G., Bosquez‐Molina, E.,
Sánchez‐Domínguez, D. (2005). Quitosano: una alternativa natural para reducir microorganismos
postcosecha y mantener la vida de anaquel de productos hortofrutícolas. Revista Iberoamericana de
Tecnología Postcosecha, 67, 1–6.
52. Fortunati, E. (2016). Multifunctional films, blends, and nanocomposites based on chitosan: Use in
antimicrobial packaging in antimicrobial food packaging. In: Antimicrobial Food Packaging (J.
Barros‐Velazquez ed.). Elsevier, CA, USA. pp. 467–477.
53. Li, Y., Wang, X., and Sun, J. (2012). Layer‐by‐layer assembly for rapid fabrication of thick polymeric
films. Chemical Society Reviews, 41, 5998–6009.
54. Muxica, A., Etxabide, A., Uranga, J., Guerrero, P., and de la Caba, K (2017). Chitosan as a bioactive
polymer: Processing, properties and applications. International Journal of Biological Macromolecules,
105, 1358–1368.
55. Espejel, J., Fandos, C., and Flavián, C. (2007). The role of intrinsic and extrinsic quality attributes on
consumer behaviour for traditional food products. Managing Service Quality, 17(6), 681–701.
56. Bernués, A., Olaizolab, A., and Corcoranc, K. (2003). Extrinsic attributes of red meat as indicators of
quality in Europe: An application for market segmentation. Food Quality and Preference, 14,
265–276.
57. EPA (2003). Chitosan; Poly‐D‐Glucosamine (128930) Fact Sheet. Biopesticide Active Ingredient.
USA. Retrieved from http://www.talunecoproducts.com/wp‐content/uploads/2016/08/EPA‐
ChitosanFactSheet.pdf
The Use of Chitosan-Based Nanoformulations for Controlling Fungi During Storage 391
58. Shields, KM., Smock, N., McQueen, C.E., and Bryant, P. (2003). Chitosan for weight loss and choles-
terol management. American Journal of Health‐System Pharmacy, 60(13), 1310–1312.
59. Codex Alimentarius, C. (1991). Report of the Twenty Second Session of the Codex committee on food
additives and contaminants. Rome.
60. Thirunavukkarasu, N. and Shanmugam, A. (2009). Extraction of chitin and chitosan from mud crab
Scylla tranquebarica (Fabricius, 1798). International Journal on Applied Bio‐Engineering, 3(2),
31–33.
61. Vicente, A.R., Manganaris, G.A., Sozzi, G.O., and Crisoto, C.H. (2009). Nutritional quality of fruits
and vegetable. In: Postharvest Handling: A Systems Approach (Second Edition) (Florkowski, W.J.,
Shewfelt, R.L., Bruekner, B., and Prussia, S. eds.). Academic Press/Elsevier, USA. pp. 58–93.
62. Lin, B., Du, Y., Liang, X., Wang, X., Wang, X., and Yang, J. (2011). Effect of chitosan coating on res-
piratory behavior and quality of stored litchi under ambient temperature. Journal of Food Engineering,
102(1), 94–99.
63. Arnon‐Rips, H. and Poverenov, E. (2018). Improving food products’ quality and storability by using
layer by layer edible coatings. Trends in Food Science & Technology, 75, 81–92.
64. Ncama, K., Magwaza, L.S., Mditshwa, A., and Tesfay, S.Z. (2018). Plant‐based edible coatings for
managing postharvest quality of fresh horticultural produce: A review. Food Packaging and Shelf Life,
16, 157–167.
65. Devlieghere, F., Vermeulen, A., and Debevere, J. (2004). Chitosan: Antimicrobial activity, interactions
with food components and applicability as a coating on fruit and vegetables. Food Microbiology, 21(6),
703–714.
66. Ayala‐Zavala, J.F., Wang, S.Y., Wang, C.Y., and González‐Aguilar, G.A. (2004). Effect of storage
temperatures on antioxidant capacity and aroma compounds in strawberry fruit. LWT – Food Science
and Technology, 37(7), 687–695.
67. Khaliq, G., Mohamed, M.T.M., Ding, P., Ghazali, H.M., and Ali, A. (2016). Storage behaviour and
quality responses of mango (Mangifera indica L.) fruit treated with chitosan and gum arabic coatings
during cold storage conditions. International Food Research Journal, 2, S141–S148.
68. Kumar, P., Sethi, S., Sharma, R.R., Srivastav, M., and Varghese, E. (2017). Effect of chitosan coating
on postharvest life and quality of plum during storage at low temperature. Scientia Horticulturae, 226,
104–109.
69. Cosme, G., Batista, W., Medeiros, D., Rodrigues, A., Menezes, M., Martins, N., Bortolini, D., and
Polete, G. (2017). The chitosan affects severely the carbon metabolism in mango (Mangifera indica L.
cv. Palmer) fruit during storage. Food Chemistry, 237, 372–378.
70. Kaya, M., Česonienė, L., Daubaras, R., Leskauskaitcė, D., and Zabulionė, D. (2016). Chitosan coating
of red kiwifruit (Actinidia melanandra) for extending of the shelf life. International Journal of
Biological Macromolecules, 85, 355–360.
71. Pagno, C.H., Castagna, A., Trivellini, A., Mensuali‐Sodi, A., Ranieri, A., Ferreira, E.A. et al. (2018).
The nutraceutical quality of tomato fruit during domestic storage is affected by chitosan coating.
Journal of Food Processing and Preservation, 42(1), 1–9.
72. Li, J., Yan, J., Wang, J., Zhao, Y., Cao, J., and Jiang, W. (2010). Effects of chitosan coating on oxidative
stress in bruised Yali pears (Pyrus bretschneideri Rehd.). International Journal of Food Science and
Technology, 45(10), 2149–2154.
73. Elsabee, M. and Abdou, E. (2013). Chitosan based edible films and coatings: A review. Materials
Science Engineering C, 33, 1819–1841.
74. Batista, W., Cosme, G., Bortolini, D., Rodrigues, A., Barbosa, D., Belghith, I., Martins, N., Menezes,
M., and Polete, G. (2018). Chitosan delays ripening and ROS production in guava (Psidium guajava
L.) fruit. Food Chemistry, 242, 232–238.
75. Vieira, J., Flores‐López, M., Jasso, D., Sousa, M., Vicente, A., and Martins, J. (2016). Effect of chi-
tosan–Aloe vera coating on postharvest quality of blueberry (Vaccinium corymbosum) fruit. Postharvest
Biology and Technology, 116, 88–97.
392 Chitin and Chitosan: Properties and Applications
76. Mannozzi, C., Tylewicz, U., Chinnici, F., Siroli, L., Rocculi, P., Dalla Rosa, M., and Romany, S.
(2018). Effects of chitosan based coatings enriched with procyanidin by‐product on quality of fresh
blueberries during storage. Food Chemistry, 251, 18–24.
77. Jongsri, P., Rojsitthisak, P., Wangsomboondee, T., and Seraypheap, K. (2017). Influence of chitosan
coating combined with spermidine on anthracnose disease and qualities of “Nam Dok Mai” mango
after harvest. Scientia Horticulturae, 224, 180–187.
78. Shen, Y. and Yang, H. (2017). Effect of preharvest chitosan‐g‐salicylic acid treatment on postharvest
table grape quality, shelf life, and resistance to Botrytis cinerea‐induced spoilage. Scientia
Horticulturae, 224, 367–373.
79. Özdemir, K. and Gökmen, V. (2017). Extending the shelf‐life of pomegranate arils with chitosan‐
ascorbic acid coating. LWT – Food Science and Technology, 76, 172–180.
80. Awad, M.A., Al‐Qurashi, A.D., Mohamed, S.A., El‐Shishtawy, R.M., and Ali, M.A. (2017). Postharvest
chitosan, gallic acid and chitosan gallate treatments effects on shelf life quality, antioxidant com-
pounds, free radical scavenging capacity and enzymes activities of “Sukkari” bananas. Journal of Food
Science and Technology, 54(2), 447–457.
81. Shahbazi, Y. (2018). Application of carboxymethyl cellulose and chitosan coatings containing Mentha
spicata essential oil in fresh strawberries. International Journal of Biological Macromolecules, 112,
262–274.
82. Yuan, G., Chen, X., and Li, D. (2016). Chitosan films and coatings containing essential oils: The anti-
oxidant and antimicrobial activity, and application in food systems. Food Research International, 89,
117–128.
83. Perdones, A., Escriche, I., Chiralt, A., and Vargas, M. (2016). Effect of chitosan‐lemon essential oil
coatings on volatile profile of strawberries during storage. Food Chemistry, 197, 979–986.
84. Dantas, I., Lima, P., Lima, A., Carneiro, A., Fechine, J., Barbosa‐Filho, J., Madruga, M., and de Souza,
E. (2015). Coatings comprising chitosan and Mentha piperita L. or Mentha × villosa Huds essential
oils to prevent common postharvest mould infections and maintain the quality of cherry tomato fruit.
International Journal of Food Microbiology, 214, 168–178.
85. Jovanović, G., Klaus, A., and Nikšić, M. (2016). Antimicrobial activity of chitosan coatings and films
against Listeria monocytogenes on black radish. Revista Argentina de Microbiología, 48, 128–136.
86. Dorman, H.J. and Deans, S.G. (2000). Antimicrobial agents from plants: antibacterial activity of plant
volatile oils. Journal of Applied Microbiology, 88(2), 308–316.
87. Sneha, M., Saxena, A., and Kaur, C. (2018). Effect of chitosan and alginate based coatings enriched
with pomegranate peel extract to extend the postharvest quality of guava (Psidium guajava L.). Food
Chemistry, 240, 245–252.
88. Duran, M., Aday, M.S., Zorba, N.N.D., Temizkan, R., Büyükcan, M.B., and Caner, C. (2016). Potential of
antimicrobial active packaging “containing natamycin, nisin, pomegranate and grape seed extract in chi-
tosan coating” to extend shelf life of fresh strawberry. Food and Bioproducts Processing, 98, 354–363.
89. Yang, G., Yue, J., Gong, X., Qian, B., Wang, H., Deng, Y., and Zhao, Y. (2014). Blueberry leaf extracts
incorporated chitosan coatings for preserving postharvest quality of fresh blueberries. Postharvest
Biology and Technology, 92, 46–53.
90. Breda, C.A., Morgado, D.L., de Assis, O.B.G., and Duarte, M.C.T. (2017). Effect of chitosan coating
enriched with pequi (Caryocar brasiliense Camb.) peel extract on quality and safety of tomatoes
(Lycopersicon esculentum Mill.) during storage. Journal of Food Processing and Preservation, 41(6),
1–8.
91. Sun, X., Narciso, J., Wang, Z., Ference, C., Bai, J., and Zhou, K. (2014). Effects of chitosan‐essential
oil coatings on safety and quality of fresh blueberries. Journal of Food Science, 79(5), M955–M960.
92. Migliori, C., Salvati, L., Di Cesare, L., Lo Scalzo, R., and Parisi, M. (2017). Effects of preharvest
applications of natural antimicrobial products on tomato fruit decay and quality during long‐term stor-
age. Scientia Horticulturae, 222, 193–202.
93. Rabea, E.I., Badawy, M.E.T., Stevens, C.V., Smagghe, G., and Steurbaut, W. (2003). Chitosan as anti-
microbial agent: Applications and mode of action. Biomacromolecules, 4(6), 1457–1465.
The Use of Chitosan-Based Nanoformulations for Controlling Fungi During Storage 393
94. Shiekh, R.A., Mailk, M.A., Al‐Thabaiti, S.A., and Shiekh, M.A. (2013). Chitosan as a novel edible
coating for fresh fruits. Food Science and Technology Research, 19(2), 139–155.
95. Petriccione, M., De Sanctis, F., Pasquariello, M.S., Mastrobuoni, F., Rega, P., Scortichini, M., and
Mencarelli, F. (2015). The effect of chitosan coating on the quality and nutraceutical traits of sweet
cherry during postharvest life. Food and Bioprocess Technology, 8(2), 394–408.
96. Sa Pratiwi, A., Dwivany, F.M., Larasati, D., Islamia, H.C., and Martien, R. (2015). Effect of chitosan
coating and bamboo FSC (fruit storage chamber) to expand banana shelf life. AIP Conference
Proceedings, 1677.
97. Sánchez, C., Lidon, F.C., Vivas, M., Ramos, P., Santos, M., and Barreiro, M.G. (2015). Effect of
chitosan coating on quality and nutritional value of fresh‐cut “Rocha” pear. Emirates Journal of Food
and Agriculture, 27(2), 206–214.
98. Pasquariello, M.S., Di Patre, D., Mastrobuoni, F., Zampella, L., Scortichini, M., and Petriccione, M.
(2015). Influence of postharvest chitosan treatment on enzymatic browning and antioxidant enzyme
activity in sweet cherry fruit. Postharvest Biology and Technology, 109, 45–56.
99. Song, H., Yuan, W., Jin, P., Wang, W., Wang, X., Yang, L., and Zhang, Y. (2016). Effects of chitosan/
nano‐silica on postharvest quality and antioxidant capacity of loquat fruit during cold storage.
Postharvest Biology and Technology, 119, 41–48.
100. Koh, P.C., Noranizan, M.A., Nur Hanani, Z.A., Karim, R., and Rosli, S.Z. (2017). Application of
edible coatings and repetitive pulsed light for shelf life extension of fresh‐cut cantaloupe (Cucumis
melo L. reticulatus cv. Glamour). Postharvest Biology and Technology, 129, 64–78.
101. Pinedo‐Guerrero, Z.H.D., Hernández‐Fuentes, A., Ortega‐Ortiz, H., Benavides‐Mendoza, A.,
Cadenas‐Pliego, G., and Juárez‐Maldonado, A. (2017). Cu nanoparticles in hydrogels of chitosan‐
PVA affect the characteristics of post‐harvest and bioactive compounds of jalapeño pepper. Molecules,
22(6), e926.
102. Varasteh, F., Arzani, K., Barzegar, M., and Zamani, Z. (2017). Pomegranate (Punica granatum L.)
Fruit storability improvement using pre‐storage chitosan coating technique. Journal of Agricultural
Sciences and Technology, 19, 389–400.
103. ChenLong, X., Wen, L., MingYang, L., LiWei, Z., Min, L., HongShun, Y., and Yun, D. (2010). Quality
of fresh‐cut pears (Pyrus bretschneideri Rehd cv. Huangguan) coated with chitosan combined with
ascorbic acid and rosemary extracts. Philippine Agricultural Scientist, 93(1), 66–75.
104. Rasdem, J. (2016). What is Nanotechnology? In: Nanotechnology. An Introduction (Second Edition).
Elsevier, Oxford, UK. p.18.
105. Kumar, A., Singla, R., Guliani, A., and Kumar, S. (2016). Biodegradable nanoparticles and their in
vivo fate. In: Nanoscale Materials in Targeted Drug Delivery, Theragnosis, and Tissue Regeneration
(S. Kumar ed.). Springer, Singapore. pp. 21–40.
106. Divya, K., Smitha, V., and Jisha, M. (2018). Antifungal, antioxidant and cytotoxic activities of chi-
tosan nanoparticles and its use as an edible coating on vegetables. International Journal of Biological
Macromolecules, 114, 572–577.
107. Ferrão, N., de Mendoça, B., Marques, K., Ferreira, C., Canto, D., Flores, M., da Costa, J., Galembeck,
A., Montenegro, T., Montenegro, T., and Montenegro, T. (2018). Effects of fungal chitosan nanopar-
ticles as eco‐friendly edible coatings on the quality of postharvest table grapes. Postharvest Biology
and Technology, 139, 56–66.
108. Pulicharla, R., Marques, C., Kumar, R., Rouissi, T., and Kaur, S. (2016). Encapsulation and release
studies of strawberry polyphenols in biodegradable chitosan nanoformulation. International Journal
of Biological Macromolecules, 88, 171–178.
109. Sotelo‐Boyás, M., Correa‐Pacheco, Z., Bautista‐Baños, S., and Gómez y Gómez, Y. (2017). Release
study and inhibitory activity of thyme essential oil‐loaded chitosan nanoparticles and nanocapsules
against foodborne bacteria. International Journal of Biological Macromolecules, 103, 409–414.
110. Mohammadi, A., Hashemi, M., and Masoud, S. (2015). Chitosan nanoparticles loaded with
Cinnamomum zeylanicum essential oil enhance the shelf life of cucumber during cold storage.
Postharvest Biology and Technology, 110, 203–213.
394 Chitin and Chitosan: Properties and Applications
111. Mohammadi, A., Hashemi, M., and Hosseini, S. (2015). Nanoencapsulation of Zataria multiflora
essential oil preparation and characterization with enhanced antifungal activity for controlling
Botrytis cinerea, the causal agent of gray mould disease. Innovative Food Science and Emerging
Technologies, 28, 73–80.
112. Mohammadi, A., Hashemi, M., and Hosseini, S.M. (2016). Postharvest treatment of nanochitosan‐
based coating loaded with Zataria multiflora essential oil improves antioxidant activity and extends
shelf‐life of cucumber. Innovative Food Science and Emerging Technologies, 33, 580–588.
113. Sotelo‐Boyás, M., Correa‐Pacheco, Z., Bautista‐Baños, S., and Corona‐Rangel, M. (2017).
Physicochemical characterization of chitosan nanoparticles and nanocapsules incorporated with lime
essential oil and their antibacterial activity against food‐borne pathogens. LWT‐Food Science and
Technology, 77, 15–20.
114. Barrera‐Necha, L., Correa‐Pacheco, Z., Bautista‐Baños, S., Hernández‐López, M., Martínez, J., and
Morán, A. (2018). Synthesis and characterization of chitosan nanoparticles loaded botanical extracts
with antifungal activity on Colletotrichum gloeosporioides and Alternaria species. Advances in
Microbiology, 8, 286–296.
115. Kaur, P., Thakur, R., and Choudhary, A. (2012). An in vitro study of the antifungal activity of silver/
chitosan nanoformulations against important seed borne pathogens. IJST, 1, 83–86.
116. Chookhongkha, N., Sopondilok, T., and Photchanachai, S. (2013). Effect of chitosan and chitosan
nanoparticles on fungal growth and chilli seed quality. Acta Horticultare, 973, 231–237.
117. Saharan, V., Mehrotra, A., Khatik, R., Rawal, P., Sharma, S., and Pal, A. (2013). Synthesis of chitosan
based nanoparticles and their in vitro evaluation against phytopathogenic fungi. International Journal
of Biology and Macromolecules, 62, 677–683.
118. Beyki, M., Zhaveh, S., Tahere, S., Rahmani‐Cherati, T., Abollahi, A., Mansour, B., Bayat Tabatabaei,
M., and Mohsenifarc, A. (2014). Encapsulation of Mentha piperita essential oils in chitosan–cin-
namic acid nanogel with enhanced antimicrobial activity against Aspergillus flavus. Industrial Crop
Production, 54, 310–319.
119. Zahid, N., Alderson, P., Ali, A., Maqbool, M., and Manickam, S. (2013). In vitro control of
Colletotrichum gloeosporioides by using chitosan loaded nanoemulsions. ActaHorticulturae, 1012,
769–774.
120. Sotelo, B.M.E., Bautista, B.S., Aldana, L.L., Solorza, F.J., Jiménez, A.A., Barrera, N.L.L., Valverde,
A.G., and Plascencia, J.M. (2015). La nanotecnología en el control de microorganismos patógenos e
insectos de importancia económica. In: Nanotecnología: fundamentos y aplicaciones (Laréz‐Velázquez,
C., Koteich Khatib, S., and López‐González, F. eds.). Universidad de los Andes, Venezuela. pp.
307–321.
16
Chitosan Application in Textile
Processing and Fabric Coating
Thomas Hahn1, Leonie Bossog2, Tom Hager3, Werner Wunderlich3,
Rudi Breier2, Thomas Stegmaier3, and Susanne Zibek1
1
Fraunhofer Institute of Interfacial Engineering and Biotechnology, Stuttgart, Germany
2
Textilchemie Dr. Petry GmbH, Reutlingen, Germany
3
German Institutes of Textile and Fiber Research, Denkendorf, Germany
Textile manufacturers and related industries are undergoing a change due to legal regula-
tions becoming effective soon, as well as their commitment for social responsibility and the
increased ecological awareness of customers. Turning from established fossil‐based value
chains towards more sustainable bio‐based products is one of the key factors to achieve
these goals. This means to substitute petrol‐based agents with reactants and products
derived from renewable resources, like cellulose and chitosan. Cellulose is, as the main
content of cotton, an omnipresent and abundant material already known in the textile
industry for centuries. By comparison, the utilisation of chitosan in textile processing is
relatively new but not less promising. The benefits of chitosan include biodegradability,
antibacterial effects and further functionalisations that enable a broad range of applications
within the textile industry. Together with a previous view on the textile processing value
chain in general, the most promising application fields of chitosan are elucidated within
this review. A summary of the commonly applied physical–mechanical or application‐
specific test methods for textile fabrics is also part of the chapter in order to assess chi-
tosan performance. A brief overview about chitin fibre production introduces the different
methods used to manufacture chitosan fibres, revealing that wet spinning and electrospin-
ning are techniques of choice. The physical–mechanical properties are discussed in more
detail whereby chitosan fibres show low to medium tensile properties. A further application
field being considered is the utilisation of chitosan as a sizing agent that efficiently protects
yarns during weaving. Chitosan films exhibit a suitable value range of significant proper-
ties, such as viscosity, surface tension, adhesion power and mechanical properties. Removal
of chitosan from the yarn or fabric as well as recycling after weaving is explained in differ-
ent sections. A further part of the article deals with the application of native or modified
chitosan as a permanent coating or finishing agent on a textile fabric meant to be non‐
removable. Due to the amine functionality of chitosan, which offers a reactive coupling
site, the polysaccharide could be modified with versatile reagents to gain various desired
properties. Furthermore, a cross‐linking is desired in order to increase the washing fastness
required for a marketable product. Besides antibacterial activity, which has already resulted
in the development of commercial chitosan‐containing formulations, further beneficial
properties that have commercial viability are anticipated. The removal of dyes and heavy
metals with chitosan‐grafted fabrics, the increased dyeing ability of fabrics, and advanta-
geous crease recovery properties are presented in the last section of this chapter.
4. Chitosan has antibacterial activity: Medical fabrics, for example, wound dressings, are
commonly applied in order to support wound healing after surgery or to treat harmless
injuries. The predominant requirement of medical fabrics is sterility in order to avoid
bacterial and viral infections. Thus, the most common commercial biocidal textile fab-
rics include metals and metal salts, triclosan, polyhexamethylene biguanide (PHMB)
and quaternary ammonium compounds [18]. Chitosan provides the opportunity to
incorporate a bio‐based bacteriostatic and hence an anti‐odour property to fabrics. This
activity is based on the interaction of the polycationic molecule with the negative
charges of the microorganism’s cell membranes [19].
Because of these features, there are several possible chitosan application fields in the
textile industry. Consequently, the polysaccharide can be potentially applied – in textile
manufacturing or in processes directly linked to fabric production – in the native or modi-
fied form as [20]:
• A sizing agent to protect the yarn during weaving [21, 22]
• An adsorbent for the removal of dyes to purify textile effluents [23, 24]
• An auxiliary in the textile dyeing process in order to increase the uptake of anionic dyes
[25, 26]
• A carrier and linker of flame retardation agents [27]
• A thickener in the cotton print process [28]
• A fibre material or mixed/blended fibres with natural and/or synthetic polymers [29]
• A finishing agent for anti‐creasing properties or enhanced crease recovery of textile
fabrics [30, 31]
• An antibacterial finishing or anti‐odour agent [32, 33]
• A superhydrophobic coating for cotton fabrics [34, 35]
• A shell material for microencapsulation processes [36]
The list emphasises the versatility of chitosan in the textile industry, although it has to be
stated that chitosan is frequently used as an auxiliary rather than as an agent with the
desired functionality. This is due to the amine groups providing a suitable site for anchor-
ing and grafting. However, several applications seem to be more promising than others,
giving rise to the first technical chitosan products on the market, described in the course of
this review. The most important applications indicated by increased research activities of
the scientific community will be discussed in the following pages. Since most readers are
probably not familiar with the processes and relevant measurements taking place during
textile manufacture, the section will start with a brief overview for a comprehensive under-
standing. This is required to evaluate chitosan performance in textile processes described
for fibres, sizing and finishing agents.
• Dissolving cellulose
• Transfer into the
spinning mass
Petroleum Plant, animal
Gas, hydrocarbons Cellulose, cotton, wool, silk
• Finishing • Finishing
• Oiling
Yarn
Spun fibres
• Sizing • Sizing
• Weaving preparation • Pretreatment
• Dyeing • Dyeing
Textile fabric
Weaving, knit fabric, fleece
• Pretreatment • Pretreatment
• Dyeing • Dyeing
• Finishing • Finishing
Everyday textiles
Clothing, home textiles, technical textiles
Figure 16.1 Textile value chains from the basic commodity to the textile. The figure was reprinted and
modified according to [37] and permission was obtained by the Fund of the chemical industry and
TEGEWA e. V.
properties, blend yarns are produced by mixing natural and chemical fibres. To increase the
efficiency of weaving processes due to the high mechanical load, as well as to conserve the
quality of the fabric, warp yarns get a protective coating before weaving [21]. After the
weaving process, the surface structure of the fabric is pretreated for dyeing, then dyed and
printed. Following this procedure, if a desired property is not an inherent characteristic of
the fibre, the fabric obtains a finishing, such as making shirts or blouses easier for ironing
or making seat covers and carpets in aircraft or train compartments flame retardant [37].
The last processing step converts the fabric into the designed textile ready for further use.
Considering the value chain of natural fibres, it should be noted that the number of chemi-
cal processing steps and the amount of chemicals consumed (textile auxiliaries) is as high as
for the production of man‐made fibres. This is due to the requirements that textiles must
meet. Many properties required by customers (e.g. water‐ and dirt‐repellence, moisture
absorption, water resistance, elasticity, the feel of the fabric on the skin, and resistance to
heat, cold, bacteria, etc.) are adjusted by applying thin coatings within the textile finishing
process. These chemicals end up in wastewater and could pollute the environment. This
confirms the need to integrate a higher content of environment‐friendly raw materials, such
as renewable resources. For this reason, there is a trend towards using bio‐based materials
and products. At the same time, customers expect equal or even better performance and
properties substituting fossil‐based compounds. Chitosan has, as an abundant natural poly-
mer, the potential to substitute several petrol‐based agents while providing new properties to
the fabrics. To evaluate chitosan properties and to guarantee quality criteria for commercial
textiles, specific measurement methods are necessary to confirm the required performance.
Table 16.1 A selection of test methods applied in the textile industry dependent on the material.
Abbreviations: ISO: International Organization for Standardization; DIN: German Industry Norm ; AATCC: American Norm.
They do not only perform textile‐ and clothing‐relevant measurements of fibres, yarns and
threads of woven, knitted, braid, and laid textile fabrics, but also carry out health‐relevant
investigations of textile medicinal products. This is especially important since the predomi-
nant property of chitosan – that is, being relevant for commercial use – is the result of
antimicrobial activity. On the one hand, antibacterial activity can be implemented by
finishing/coating the textile fabric at the end of the production chain. On the other hand, the
application of chitosan fibres and yarns offers the opportunity to implement these proper-
ties at the initial step of the value chain.
(a) (b)
6 μm 6 μm
(c) (d)
6 μm 3 μm
Figure 16.2 Scanning electron microscope (SEM) figures of chitosan fibres obtained by solubilisation of
the polysaccharide in different solvent mixtures; (a) 90:10 v/v TFA/dichloromethane (DCM), (b) 80:20
v/v TFA/DCM, (c) 70:30 v/v TFA/DCM (5000x magnified), (d) 70:30 v/v TFA/DCM (10,000x magnified).
Reprinted from Ohkawa et al.[55] with permission from Wiley.
1. Modification of chitosan prior to or during spinning: It can be assumed that the graft-
ing, especially at the free primary amine groups, affects the properties, since they are
particularly responsible for chitosan characteristics. There are only a few studies per-
formed regarding the spinning of derivatised chitosan, but the majority of the investiga-
tions concentrate on electron microscopy pictures or medicinal or biological tests rather
than investigation of the tensile properties [69, 70]. Electrospinning of PEGylated or
carboxymethyl chitosan is reported to be challenging and could only be performed in
the presence of auxiliaries [71, 72].
2. Adaption of spinning conditions: There are various tuning parameters to adjust either
wet or dry spinning in order to obtain chitosan fibres with optimised properties.
However, comprehensive studies focusing on the systematic optimisation of spinning
parameters have not been carried out yet. Using ionic liquids rather than acetic acid as
the chitosan solvent increases the tenacity by about four times, emphasising the poten-
tial of adapting the spinning conditions [73]. Post‐spinning drying is also considered as
a relevant process step to influence the mechanical properties of chitosan fibres.
Researchers confirmed that drying with methanol, based on the increase in the degree
of crystallinity, improves tensile strength and biodegradation resistance [74].
3. Post‐modification of chitosan fibres: Cross‐linking is an effective way to modulate the
properties. Glyoxal or glutardialdehyde application led to an improvement of tenacity
from 1 to 2 cN/dtex [75, 76]. Treatment with acyl anhydrides is an alternative method
to hydrophobise chitosan fibres. This revealed decreased strength but increased elonga-
tion value with increasing chain length of the acyl chain [77]. Post‐modification does
406 Chitin and Chitosan: Properties and Applications
not necessarily result in altered properties. Constant or even worse properties were
reported with regard to breaking elongation and tenacity after derivatising chitosan
fibres with acrylic acid [78].
4. Admixture of other polymers in the spinning dope (co‐spinning): Different polymers
could be solubilised together in a suitable solvent, enabling the production of fibres
with properties completely different from the fibres derived from pure compounds.
Zheng et al. solubilised PVA and chitosan in an acetic acid solution containing urea.
The dry and wet tensile strengths of PVA/chitosan fibres were 10–40% higher than that
of pure chitosan [79]. There are other known studies that utilise the wet‐spinning tech-
nique to process chitosan with molecules such as heparin or cellulose. This has led to
the formation of bio‐filaments, providing a holistic renewable approach. Yan et al.
observed that a spike in chitin nanocrystals in the chitosan spinning dope raised tenac-
ity and young modulus by 54% and 85%, respectively [80].
5. Applying chitosan fibres with other polymer fibres to gain blended yarns: The merits
pointed out to blend two polymers as stated earlier apply here in the same way. Blending
chitosan with polyacrylonitrile fibres, while providing a non‐sustainable fibre, showed
superior yarn performance and minimised wastage [81]. Lam et al. investigated the pro-
duction of chitosan/cotton blend yarns and evaluated their mechanical properties. They
stated that the mechanical properties of the resulting yarns can be optimised by applying
chitosan fibres of shorter length and blending them with the sliver technique [82].
As can be obtained from the quoted examples, mechanical properties of chitosan fibres
can be affected and improved by several techniques. Nevertheless, the tensile strengths of
chitosan fibres or fibre blends with a major content of chitosan are at a disadvantage as
compared to other man‐made fibres, although the potential for improvement has not yet
been fully exploited and should be part of prospective research projects. Based on this
reason, few chitosan fibres or blends thereof are currently commercialised, for example
Crabyon (Swicofil), Sunray (Hismer Bio‐technology Co. Ltd) and Ultex (Shanghai
Nantec), and predominantly offered for special applications. To strengthen the turnover, the
following challenge needs to be overcome: developing an economic process and material
with higher mechanical properties.
The majority of sizing agents contain fossil‐based polymers like PVA, PAC and PES or
biopolymer‐like starch‐based compounds. Starch and its derivatives are the predominant
sizing agents of earlier times, but their manufacturing competes with food production.
Furthermore, they are not very effective in the current applied high‐speed weaving looms.
Hence, chitosan as a polysaccharide from renewable resources exhibiting film‐forming and
biodegradation properties could be an alternative. An advantage is that chitosan has limited
valuable use in the food or feed industry. The competitive pressure within the textile indus-
try forces, besides the textile material, the use of cheap auxiliaries and reagents. However,
at the same time, customers demand more sustainable products. Sustainability of chitosan
is proven but the costs are nowadays higher as compared to conventional sizing agents.
Thus, it should show superior characteristics while, as with all other sizing agents, meeting
the following criteria [83, 84]:
• The liquid should have high wetting ability and appropriate surface tension with regard
to the applied yarn.
• The ability to form a homogeneous and closed film on the warp thread unaffected by
different fibre finishes is mandatory. Additionally, the film‐forming property should not
be influenced by atmospheric conditions.
• The resulting film should have a certain elasticity so that no breakage of the film occurs
during the various stretching and bending stresses of the weaving process.
• A high adhesive power is required to stick the protruding fibre ends to the yarn surface,
resulting in the prevention of friction during the weaving process.
• A significant reduction of the warp thread elongation by the sizing agent is not
required.
Chitosan‐specific requirements can be summarised in five statements as follows:
1. For economic reasons, chitosan should be miscible with conventional, inexpensive siz-
ing agents and additives. It should be possible to produce homogeneous solutions.
2. To ensure good workability in standard sizing boxes, the sizing liquid should have, at
maximum, a viscosity of 200 mPa·s (at a shear rate of 500 s−1).
3. In order to achieve good solubility and film formation as well as suitable viscosity in
the solution, the DDA should be at least 80%.
4. Easy removal of chitosan after weaving is highly demanded. This is negligible if chi-
tosan does not affect further processing of the fabric.
5. Ash content should be lower than 1% (w/w) of the solid in order to achieve high adhe-
sive power.
The general and specific requirements for chitosan will be discussed in detail in the fol-
lowing text.
of precipitates, the phase separation during long‐time storage. Since chitosan is a polymer
whose application is demonstrated as a coagulation/flocculation auxiliary in wastewater
treatment, it readily forms precipitates, especially with anionic compounds. Renault et al.
gives an excellent overview about the chitosan coagulation/flocculation application with a
variety of compounds stating the effective interaction between them [85]. However, inves-
tigations in blending pure starch and chitosan revealed miscibility over the entire range
measured [86]. Sudhakar et al. indicated positive interactions only up to 40% starch content
in the blend but stated a significant temperature effect [87]. A German research project
investigated the miscibility of chitosan with different sizing agents [88]. Studies performed
by the authors indicated that there are incompatibilities with carboxymethylated starch,
carboxymethyl cellulose, PVA and polyacrylate. In combination with other selected modi-
fied starch solutions, chitosan application is possible. Nevertheless, preliminary miscibility
investigations in a long‐term test are a prerequisite to evaluate the performance and exclude
undesired effects while sizing. Another fact to be considered while applying different com-
pounds in an approach is that the molecular interaction could result in high‐viscosity solu-
tions aggravating the application. Besides, viscosity is also affected by several other factors,
as described in the following chapter.
350 350
LMW Chitosan
300 HMW Chitosan 300
Viscosity [mPa·s]
Viscosity [mPa·s]
250 250
Threshold value
200 200
3%
150 150
2.5%
100 100 2%
50 50 1.5%
1%
0 0
0.5 1.0 1.5 2.0 2.5 3.0 500 1000 1500 2000 2500 3000
Concentration [%] Shear rate [s–1]
(c) (d)
90 80
80
70
70
Viscosity [mPa·s]
Viscosity [mPa·s]
60
60
50
40 50
30
40
20
10
30 40 50 60 70 80 90 0 1 2 3 4 5
Figure 16.3 (a) Viscosity of low‐molecular‐weight (LMW) and high‐molecular‐weight (HMW) crab shell chitosan in 2% acetic acid at different concentrations
(% w/v) and at a constant shear rate of 500 s−1; (b) viscosity of LMW chitosan in 2% acetic acid at different chitosan concentrations as a function of the shear
rate; (c) viscosity of 2% LMW chitosan in 2% acetic acid as a function of the temperature (at 500 s−1); and (d) viscosity of 2% LMW chitosan solution in 2%
acetic acid at 500 s−1 as a function of the storing time. LMW chitosan: deacetylation degree (DDA) = 90%; HMW chitosan: DDA = 85%, measured with cone‐
plate rheometer.
410 Chitin and Chitosan: Properties and Applications
between the surface free energy of the solid and surface tension of the liquid is, in general,
beneficial. Concerning the surface free energy of chitosan films, literature offers opposing
data. On the one hand, there are investigations reporting surface free energy of the films
from 36 to 41 mN·m−1. Based on this value, chitosan has a suitable adhesion to surfaces
with low surface free energy [95, 96]. Other studies revealed high surface tension of chi-
tosan solutions (65–75 mN·m−1) [97, 98]. The differences are likely related to various
measurement methods, water content of the films, concentration or the physical–chemical
properties of the polysaccharide applied. With regard to these inconsistencies, Cunha et al.
agreed that nonpolar impurities greatly affect measurement and result in high errors, con-
cluding that a high purification degree is essential for performance. Examining a film com-
posed of pure chitosan resulted in a surface free energy of 60 mN·m−1 [99]. As is the case
with other polymer solutions, the surface tension of chitosan solutions also increase with
increasing concentration and molecular weight of the chitosan used [100]. Chitosan is a
known, experimentally proven, well‐performing adhesive due to its positively charged
amino groups that can interact with negative surface charges via electrostatic forces exhib-
iting a high amount of polar forces. The contribution of hydrogen bonds and van der Waals
forces to the adhesive strength was also described, together with strong cohesive interac-
tions in acidic conditions [101].
The aforementioned properties resulted in the commercialisation of chitosan adhe-
sives for biomedical applications, as reported by Mati‐Baouche et al. [102]. In contrast,
only a few commercial sizing agents contain chitosan, although there are major advan-
tages as compared to conventional starch sizing, for example, of cotton. It can be assumed
that the adhesion of chitosan to cotton is probably due to the structural similarity with
cellulose and/or the opposite charge of the cotton derived by residual pectins or waxes.
However, high adhesion power in comparison to conventional PVA is also confirmed by
own investigations. Furthermore, the tensile strength of the yarn is also improved by the
increased adhesion of individual fibres to each other. The extent of improvement and
protection efficiency depends on the mechanical properties of the films formed in the
sizing process.
properties can also be achieved by the application of plasticizers, such as, for example,
glycols, glycerol, sorbitol, sucrose and citrate [108]. At higher moisture content, modulus
and tensile strength decrease, whereas elongation until breakage greatly increases [109].
The same obviously applies for the organic acid concentration in the films previously
applied for acidification of the solution in order to solubilise chitosan. The properties are
altered in an order of the power of ten [110]. Hence, the protectability of the sizing agent
cannot necessarily be determined by the mechanical properties of chitosan films, but is
rather based on application‐specific trials, whereas the sizing agent demonstrated to be
eligible for this purpose. This also holds true for the water uptake behaviour with swelling
ratios measured in a range from 190 to 500% [104, 107], which is a crucial property for the
removal of the film after weaving (desizing) [111].
Polyester
Cotton
Figure 16.4 Fabrics treated with ninhydrin, a chitosan‐specific dye. In the original state, ninhydrin gives
only a slight colour. After sizing with 1% (w/v) chitosan, the dark green colour is recognisable, indicating
the presence of adsorbed chitosan. Desizing the polyester fabric with acidic water results in the complete
removal of chitosan, whereas cotton still exhibits an excessive colouration. In comparison, enzymatic
desizing of chitosan‐sized cotton revealed a slightly lower colouration, indicating the presence of residual
chitosan on the fabric.
on the cotton surface or not. However, it must be stated that optical brighteners were not
used for the trials, since their anionic character could result in a strong electrostatic
interaction with the positively charged chitosan.
• Impact on dyeing: After desizing, it was possible to dye the polyester content of the
mixed fabric (polyester–cotton) without any problems. The single yarns of the mixed
fabric exhibited a comparable colouration. Chitosan‐sized cotton showed a slightly
deeper colouration after dyeing with direct dyes. Concerning reactive dyes, there were
no differences between chitosan‐sized cotton and cotton sized with usually applied
agents, for example, starch. The chitosan hardly affected the fastness of the dyeing.
• Impact on the fabric handle: No significant differences were determined concerning the
handle between fabrics sized with chitosan or with conventional sizing liquid after
desizing.
• Impact on the finishing process: Chitosan‐sized fabrics subjected to high‐grade finish-
ing (easy‐care finishing) exhibited slightly lower tensile strength and wrinkle recovery
angle. The impact of residual chitosan on fluorocarbon finishing was marginal.
As can be seen from the preceding text, the influence of chitosan on the subsequent pro-
cessing is low in general, even if chitosan washout was not completely performed. On the
other hand, if dyeing with acidic agents, chitosan should be completely separated to avoid
interaction. Further investigations must be carried out for this purpose. To the best of the
authors’ knowledge, comprehensive studies on performing a total removal of chitosan from
a cotton surface via enzymes are lacking. If not treated with enzymes, the removal solution
could theoretically be recycled to obtain a closed sizing loop.
Chitosan in its native form is a biodegradable, non‐toxic polysaccharide. However,
uncontrolled release into the wastewater system or the environment needs to be avoided,
since it is still expensive as compared to other polymers, and adverse effects on the
414 Chitin and Chitosan: Properties and Applications
is linked, whether covalently, ionic or via non‐ionic interactions, to the textile, conferring
the following properties [121, 122]:
• Wrinkle resistance or resiliency for cellulosic materials (durable‐press finish)
• Soil, oil and water repellence
• Chemical resistance
• Durable cleanability (soil release)
• Self‐cleaning ability
• Shrink‐proofing of wool and cellulose
• Pilling, abrasion and wear resistance
• Antistatic properties
• Resistance to UV light, heat and pollutants
• Minimising ironing or pressing (easy‐care finish)
• Resistance towards microorganisms and insects (especially wool fibres)
• Flame retardancy
• Thermal conductivity
The increasing trend towards the functionalisation of textile surfaces also sometimes
requires that textiles possess several such functionalities. The functional coating of techni-
cal textiles is usually based on the use of synthetic chemical auxiliaries based on polyure-
thanes, polyacrylates, polyvinyl chloride, silicone elastomers and fluoropolymers, which
are dispersed in water. However, Holme already stated, in the 1990s, that prospective com-
pounds should meet the demand for green finishing agents, specifically for ease of applica-
tion on automated machinery [123]. Since some of the properties listed are high‐tech
applications, the use of natural heterogeneous materials is not suitable for all of these func-
tionalities. Nevertheless, chitosan was already investigated for use in finishing formula-
tions, and revealed the most promising results concerning antibacterial or wrinkle‐free
finishes, but is not restricted to these [124]. As follows, chitosan is contained in several
coating or finishing formulations in order to modify the surface of textiles. Two options for
using chitosan in surface modification formulations are conceivable: on the one hand, the
application as an active agent conferring the desired property to the textile; and, on the
other hand, as a compound supporting the efficiency, respectively, of the release of active
agents or of performing as a carrier material. A sole use as one or the other is normally not
possible independently of each other, but the definition of a focus is. Both application pur-
poses will be explained in detail in the text that follows.
Roy et al. highlighted the tuning of the active ingredient release by the layer‐by‐layer for-
mation of chitosan/carboxymethyl cellulose microcapsules and the succeeding application
of glutardialdehyde [125]. The impregnation of the textile with the capsules commonly
results in weak interactions. Thus, researchers grafted chitosan/gelatine microcapsules
onto a cotton fabric with the help of dimethylol dihydroxy ethylene urea. Herein, it was
demonstrated that patchouli oil in the core of the microcapsules also develops a significant
antibacterial effect, even after several wash cycles [126]. Ionic gelation with sodium trip-
olyphosphate (TPP) is a method to produce chitosan‐containing nanocapsules. The authors
described the incorporation of rose fragrance in the core of these particles as enhancing the
permanence on cotton fabrics [127]. Another study has focused on the use of microcapsules
with shell material based on TPP and chitosan formed by ionic gelation and grafting.
Incorporated ingredients such as rosehip oil and ceramide greatly increased the antibacte-
rial activity of the cotton fabric [128]. The same applies for Ag/ZnO particles embedded in
a chitosan matrix, resulting in a highly active antibacterial finishing agent for a cotton/
polyester blend [129].
In conclusion, mainly oils were investigated for incorporation into the capsules. The
release of active ingredients by chitosan capsules can be steered by the encapsulation pro-
cess itself derived by adaption to the conditions. A slow release is often desired, whereas
some applications require permanent immobilisation of the functional molecule. Chitosan
itself has a high affinity to, for example, cotton, resulting in tight adsorption (see
Section 16.5.5), but covalent bonds are usually necessary to implement a permanent effect
on the textile.
16.7 Outlook
In the course of human history, textile production has developed from manual time‐intensive
production of individual garments to highly automated production with high unit num-
bers. As a result, products with a higher degree of modification and quality were also
produced, which were either based on petrochemically produced raw materials or at least
treated with synthetic agents in the course of production. Today, due to customer demand
and ecological necessity, the trend is towards more sustainable and environmentally
friendly textile products. According to the results shown earlier, chitosan could contribute
to this. Chitosan can be implemented as a fibre material at the very first process step of the
textile value chain. Unfortunately, the spinning of chitosan is challenging and results in
fibres with poor mechanical properties. Adaption of spinning conditions, derivatisation or
blending can be applied to unfold a strengthening effect in order to resist the mechanical
loads in further processing. Weaving is one of these processes which cause high stress to
the fibres requiring sizing that protects the fibre. It turned out that chitosan is a sizing
agent providing a suitable protection film around natural and man‐made fibres, enabling
them to withstand stress. Prospective works should focus on the complete removal of
chitosan from the cotton fibre after sizing. However, residual chitosan adsorbed at the
fibre has no or marginal influence on the following process steps. A permanent fixation of
chitosan on the fabric is even desired in the case of coating or finishing. On the one hand,
chitosan can act as a carrier material or linker, transferring the beneficial properties of
another encapsulated or grafted active agent to the fabric. For a durable chitosan coating
or finishing, several synthetic or bio‐based cross‐linkers are suitable. On the other hand,
chitosan could act as the active agent itself, conferring antibacterial activity to the textiles,
which already led to commercial biomedical applications. A few more research studies
were promising and could lead to the development of marketable products, such as the
removal of dyes or metal ions from textile wastewater, the increased dyeability of textiles
or the crease resistance.
In contrast to the beneficial properties described, the production process requires much
more research and improvement to satisfy the demands of a holistic sustainable approach.
The positive is the substantial progress based on intensive research in the past, resulting in
lower consumption of chemicals and the reduction in energy input during the conversion of
chitin into chitosan [158]. Nevertheless, the development of biotechnological or at least
milder methods to produce chitosan is mandatory. As a consequence, the relatively high
cost of chitosan (US$10–100/kg) [159, 160] would considerably decrease and make chi-
tosan more competitive to other synthetic agents used. A price cut can also be expected
from a rise in further commercialised chitin sources, such as fungi or insects. As part of the
arthropods, insects do not only contain proteins in their exoskeleton but also high amounts
of chitin. Cultivation of insects and the products thereof are currently a ‘hot topic’, impli-
cating that a larger quantity of insect‐based chitosan will be available in the future. Due to
the more controlled conditions in the breeding of insects and the cultivation of fungi, alter-
ing external conditions do not influence the homogeneity and thus the quality of chitin and
chitosan, respectively, as is the case for crab shells. The application of defined and homo-
geneous chitosan is highly desirable for the substitution of synthetic polymers in the textile
value chain. However, chitosan is already showing promising results in its current form.
Now the existing potential must be fully exploited through intensive research work and the
420 Chitin and Chitosan: Properties and Applications
Nomenclature
Carding The process of preparing the fibres of cotton, wool, etc., for
spinning.
Dyes:
Direct Water‐soluble azo compounds for the direct adsorptive dyeing
of cellulose or cotton in alkaline or neutral solution.
Reactive Water‐soluble anionic compounds for the dyeing of fibres via
covalent bonds at alkaline conditions exhibiting high washing
and light fastness.
Fabric Artificial network of yarns and threads of fibres that has been
woven or knitted.
Colour fastness Describes the property of a material to maintain the colouration
by external influences.
Finishing The last process step to improve the textile’s aesthetic or func-
tional properties.
Easy‐care Cross‐linking of cellulose or cotton fibres with aldehyde‐
containing compounds to reduce swelling and improve wrinkle
recovery.
Knitting Knitted fabric consists of consecutive loops called stitches. As
each row progresses, a new loop is pulled through an existing
loop. The active stitches are held on a needle until another loop
can be passed through them.
Sizing process The process of applying an adhesive protective coating on the
warp yarns’ surface is called sizing. Sizing is done during weav-
ing preparation for getting a good weaving behaviour.
Sizing agent A compound applied to protect the warp yarn during the weav-
ing process.
Tenacity/specific strength The tenacity of a material is the mass stress at break. It is defined
as the specific stress corresponding to the maximum force on a
force/extension curve.
Spinning:
Primary Cellulosic or synthetic polymers are converted into fibres by
extrusion of a polymer melt or solution.
Secondary Processing of fibres into staple yarn.
Tensile strength The tensile strength of a material is the maximum amount of
tensile stress it can take before failure, such as breaking or per-
manent deformation. Tensile strength specifies the point when a
material goes from elastic to plastic deformation.
Wash fastness The resistance of a material to change in any of its colour char-
acteristics; when subjected to washing, is called colour fastness
to washing.
Chitosan Application in Textile Processing and Fabric Coating 421
References
1. Bellis, M. The History of Clothing: Thought Co [updated 05.02.2018]. Available from: https://www.
thoughtco.com/history‐of‐clothing‐1991476.
2. Wragg‐Skyes, R. Humans evolve. In: Fewster, H., editor. Big History: Our Incredible Journey, from
Big Bang to Now. London: DK; 2016. p. 0241225906.
3. Barlex D. Developing a Technology Curriculum. In: Williams, J.P., Jones, A., Buntting, C., editors.
The Future of Technology Education: Springer; 2015.
4. Kvavadze, E., Bar‐Yosef, O., Belfer‐Cohen, A., Boaretto, E., Jakeli, N., Matskevich, Z. et al. 30,000‐
year‐old wild flax fibers. Science (New York, NY). 2009;325(5946):1359.
5. Wild, J.P. Part I. Textile industries of the ancient world to AD 1000. In: Jenkins D, editor. The
Cambridge History of Western Textiles. Cambridge: Cambridge University Press; 2003.
6. Cothren, J.T. Preface. In: Smith CW, editor. Cotton: Origin, History, Technology, and Production.
Wiley Series in crop sciences. New York: Wiley; 1999.
7. Pearson, G.P. The stocking’s escape from the cocoon. Journal of the Society of Dyers and Colourists.
1993;109(10):314.
8. Sanches, R.A., Marcicano, J.P.P., de Held, M.S.B., Guimaraes, B.M.G., Alonso, R.S., Takamune,
K.M. et al. Organic cotton, lyocell and SPF: a comparative study. International Journal of Clothing
Science and Technology. 2015;27(5):692–704.
9. Younes, I. and Rinaudo, M. Chitin and chitosan preparation from marine sources. Structure, properties
and applications. Marine Drugs. 2015;13(3):1133–1374.
10. Islam, S., Bhuiyan, M.A.R., and Islam, M.N. Chitin and chitosan: Structure, properties and appli-
cations in biomedical engineering. Journal of Polymers and the Environment. 2017;25(3):
854–866.
11. Akovali, G. Thermoplastic polymers used in textile coatings. In: Akovali G, editor. Advances in
Polymer Coated Textiles. Shwabury: Smithers Rapra Technology Ltd.; 2012.
12. Shahid‐ul, I. and Sun, G. Thermodynamics, kinetics, and multifunctional finishing of textile materials
with colorants extracted from natural renewable sources. ACS Sustainable Chemistry & Engineering.
2017;5(9):7451–7466.
13. Zheng, C., Zhang, Z., Xu, D., National Engineering Research Center for Dyeing and Finishing of
Textiles; Minist Educ KLS, Technol Eco‐Textile; Donghua Univ CCCE, et al. Dyeing and finishing
technical strategies for wool to prepare for REACH Law 2010. 781‐+ p.
14. Jain, A., Gulbake, A., Shilpi, S., Jain, A., Hurkat, P., and Jain, S.K. A new horizon in modifications of
chitosan: Syntheses and applications. Critical Reviews In Therapeutic Drug Carrier Systems.
2013;30(2):91–181.
15. Bordenave, N., Grelier, S., and Coma, V. Hydrophobization and antimicrobial activity of chitosan and
paper‐based packaging material. Biomacromolecules. 2010;11(1):88–96.
16. Kyzas, G.Z. and Bikiaris, D.N. Recent modifications of chitosan for adsorption applications: A critical
and systematic review. Marine Drugs. 2015;13(1):312–337.
17. Kildeeva, N.R., Perminov, P.A., Vladimirov, L.V., Novikov, V.V., and Mikhailov, S.N. About mecha-
nism of chitosan cross‐linking with glutaraldehyde. Russian Journal of Bioorganic Chemistry.
2009;35(3):360–369.
422 Chitin and Chitosan: Properties and Applications
18. Morais, D.S., Guedes, R.M., and Lopes, M.A. Antimicrobial approaches for textiles: From research to
market. Materials. 2016;9(6):498.
19. Lim, S.‐H. and Hudson, S.M. Review of chitosan and its derivatives as antimicrobial agents and their
uses as textile chemicals. Journal of Macromolecular Science, Part C. 2003;43(2):223–269.
20. Roy, J., Salaün, F., Giraud, S., Ferri, A., and Guan, J. Chitosan‐based sustainable textile technology:
Process, mechanism, innovation, and safety. In: Shalaby E, editor. Biological Activities and Application
of Marine Polysaccharides: InTech; 2017.
21. Stegmaier, T., Wunderlich, W., Hager, T., Siddique, A.B., Sarsour, J., and Planck, H. Chitosan – A siz-
ing agent in fabric production – Development and ecological evaluation. CLEAN – Soil, Air, Water.
2008;36(3):279–286.
22. Hebeish, A., Higazy, A., and El‐Shafei, A. New sizing agents and flocculants derived from chitosan.
Starch – Stärke. 2006;58(8):401–410.
23. Herrera‐González, A.M., Peláez‐Cid, A.A., and Caldera‐Villalobos, M. Adsorption of textile dyes pre-
sent in aqueous solution and wastewater using polyelectrolytes derived from chitosan. Journal of
Chemical Technology & Biotechnology. 2017;92(7):1488–1495.
24. Trung, T.S., Ng, C.‐H., and Stevens, W.F. Characterization of decrystallized chitosan and its applica-
tion in biosorption of textile dyes. Biotechnology Letters. 2003;25(14):1185–1190.
25. Dev, V.R.G., Venugopal, J., Sudha, S., Deepika, G., and Ramakrishna, S. Dyeing and antimicrobial
characteristics of chitosan treated wool fabrics with henna dye. Carbohydrate Polymers. 2009;
75(4):646–650.
26. Mahbubul Bashar, M. and Khan, M.A. An overview on surface modification of cotton fiber for apparel
use. Journal of Polymers and the Environment. 2013;21(1):181–190.
27. El‐Shafei, A., ElShemy, M., and Abou‐Okeil, A. Eco‐friendly finishing agent for cotton fabrics to
improve flame retardant and antibacterial properties. Carbohydrate Polymers. 2015;118:83–90.
28. Yuen, C.W.M., Ku, S.K.A., and Kan, C.W. Use of a biomaterial as a thickener for textile ink‐jet print-
ing. Journal of Applied Polymer Science. 2008;107(2):1057–1065.
29. Ravi Kumar, M.N.V. Chitin and chitosan fibres: A review. Bulletin of Materials Science. 1999;22(5):905.
30. Huang, K.‐S., Wu, W.‐J., Chen, J.‐B., and Lian, H.‐S. Application of low‐molecular‐weight chitosan
in durable press finishing. Carbohydrate Polymers. 2008;73(2):254–260.
31. Hebeish, A., Abdel‐Mohdy, F.A., Fouda, M.M.G., Elsaid, Z., Essam, S., Tammam, G.H. et al.
Green synthesis of easy care and antimicrobial cotton fabrics. Carbohydrate Polymers. 2011;86(4):
1684–1691.
32. Tawiah, B., Badoe, W., and Fu, S.H. Advances in the development of antimicrobial agents for textiles:
The quest for natural products. Review. Fibres & Textiles in Eastern Europe. 2016;24(3):136–149.
33. Hasebe, Y., Kuwahara, K., and Tokunaga, S., editors. Chitosan hybrid deodorant agent for finishing
textiles. The New Millennium of Textiles, AATCC International Conference &Exhibition; 2000;
Winston‐Salem.
34. Ivanova, N.A. and Philipchenko, A.B. Superhydrophobic chitosan‐based coatings for textile process-
ing. Applied Surface Science. 2012;263:783–787.
35. El‐Bisi, M.K., Ibrahim, H., Rabie, A.M., Elnagar, K., Taha, G.M., and El‐Alfy, E.A. Super hydropho-
bic cotton fabrics via green techniques. Der Pharma Chemica. 2016;8(19):57–69.
36. Souza, J.M., Caldas, A.L., Tohidi, S.D., Molina, J., Souto, A.P., Fangueiro, R. et al. Properties and
controlled release of chitosan microencapsulated limonene oil. Revista Brasileira de Farmacognosia.
2014;24(6):691–698.
37. Bode, A., Hardt, P., Pöhlig, M., Rauch, W., Schröder, V., Tausch, M.W. et al. Informationsserie
Textilchemie. In: V. VdcIe, editor. Funds of the Chemical Industry and TEGEWA e.V.; 2007. p. 63.
38. Agboh, O.C. and Qin, Y. Chitin and chitosan fibers. Polymers for Advanced Technologies.
1997;8(6):355–365.
39. Roberts, G.A.F. Solubility and solution behaviour of chitin and chitosan. Chitin Chemistry. London:
Macmillan Education UK; 1992. p. 274–329.
Chitosan Application in Textile Processing and Fabric Coating 423
40. Ansari, R. Sizing Agents: Chemistry behind sizing of warp yarns and methods of desizing: Textile
Digest; 2016 [Available from: http://www.textiledigest.com/sizing‐agents‐chemistry‐behind‐sizing‐
of‐warp‐yarns‐and‐methods‐of‐desizing/].
41. Rana, S., Pichandi, S., Parveen, S. and Fangueiro, R. Biosynthetic fibers: Production, processing,
properties and their sustainability parameters. In: Muthu, S.S., editor. Roadmap to Sustainable Textiles
and Clothing: Eco‐friendly Raw Materials, Technologies, and Processing Methods. Singapore:
Springer Singapore; 2014. p. 109–138
42. Mundsinger, K., Müller, A., Beyer, R., Hermanutz, F., and Buchmeiser, M.R. Multifilament cellulose/
chitin blend yarn spun from ionic liquids. Carbohydrate Polymers. 2015;131:34–40.
43. Muzzarelli, C., Francescangeli, O., Tosi, G., and Muzzarelli, R.A.A. Susceptibility of dibutyryl chitin
and regenerated chitin fibres to deacylation and depolymerization by lipases. Carbohydrate Polymers.
2004;56(2):137–146.
44. Szosland, L. and East, G.C. The dry spinning of dibutyrylchitin fibers. Journal of Applied Polymer
Science. 1995;58(13):2459–2466.
45. Salas, C., Thompson, Z., and Bhattarai, N. 15 – Electrospun chitosan fibers A2 – Afshari, Mehdi.
Electrospun Nanofibers: Woodhead Publishing; 2017. p. 371–98.
46. El‐Hefian, E. A., Nasef, M.M., and Yahaya, A.H. Preparation and characterization of chitosan/
poly(vinyl alcohol) blended films: Mechanical, Thermal and Surface Investigations. E‐Journal of
Chemistry. 2011;8(1):91–96.
47. Notin, L., Viton, C., Lucas, J.‐M., and Domard, A. Pseudo‐dry‐spinning of chitosan. Acta Biomaterialia.
2006;2(3):297–311.
48. Knaul, J., Hooper, M., Chanyi, C., and Creber, K.A.M. Improvements in the drying process for wet‐
spun chitosan fibers. Journal of Applied Polymer Science. 1998;69(7):1435–1444.
49. East, G.C. and Qin, Y.M. Wet Spinning of chitosan and the acetylation of chitosan fibers. Journal of
Applied Polymer Science. 1993;50(10):1773–1779.
50. Hirano, S., Nagamura, K., Zhang, M., Kim, S.K., Chung, B.G., Yoshikawa, M. et al. Chitosan staple
fibers and their chemical modification with some aldehydes. Carbohydrate Polymers.
1999;38(4):293–298.
51. Cruz, R., Diniz, L.G.M., Lisboa, H.M., and Fook, M.V.L. Effect of different carboxylic acids as
solvent on chitosan fibers production by wet spinning. Materia‐Rio De Janeiro. 2016;21(2):
525–531.
52. Huang, Z.‐M., Zhang, Y.Z., Kotaki, M., and Ramakrishna, S. A review on polymer nanofibers by elec-
trospinning and their applications in nanocomposites. Composites Science and Technology.
2003;63(15):2223–2253.
53. Bhardwaj, N. and Kundu, S.C. Electrospinning: A fascinating fiber fabrication technique. Biotechnology
Advances. 2010;28(3):325–347.
54. Min, B.‐M., Lee, S.W., Lim, J.N., You, Y., Lee, T.S., Kang, P.H. et al. Chitin and chitosan nanofibers:
electrospinning of chitin and deacetylation of chitin nanofibers. Polymer. 2004;45(21):7137–7142.
55. Ohkawa, K., Cha, D., Kim, H., Nishida, A., and Yamamoto, H. Electrospinning of chitosan.
Macromolecular Rapid Communications. 2004;25(18):1600–1605.
56. Wan, Y., Cao, X., Zhang, S., Wang, S., and Wu, Q. Fibrous poly(chitosan‐g‐dl‐lactic acid) scaffolds
prepared via electro‐wet‐spinning. Acta Biomaterialia. 2008;4(4):876–886.
57. De Vrieze, S., Westbroek, P., Van Camp, T., and Van Langenhove, L. Electrospinning of chitosan
nanofibrous structures: Feasibility study. Journal of Materials Science. 2007;42(19):8029–8034.
58. Elsabee, M.Z., Naguib, H.F., and Morsi, R.E. Chitosan based nanofibers, review. Materials Science
and Engineering: C. 2012;32(7):1711–1726.
59. El‐Tahlawy, K. and Hudson, S.M. Chitosan: Aspects of fiber spinnability. Journal of Applied Polymer
Science. 2006;100(2):1162–1168.
60. Sun, K. and Li, Z.H. Preparations, properties and applications of chitosan based nanofibers fabricated
by electrospinning. eXPRESS Polymer Letters. 2011;5(4):342–361.
424 Chitin and Chitosan: Properties and Applications
61. Guo, H.F., Lam, N.Y.K., Yang, C., and Li, L. Simulating three‐dimensional dynamics of flexible fibers in
a ring spinning triangle: Chitosan and cotton fibers. Textile Research Journal. 2017;87(11):1403–1410.
62. Toskas, G., Brunler, R., Hund, H., Hund, R.D., Hild, M., Aibibu, D. et al. Pure Chitosan Microfibres
for biomedical applications. Autex Research Journal. 2013;13(4):134–140.
63. Whang, H.S., Whang, N., Frey, M., Hudson, S.M., and Cuculo, J.A. 12 – Conversion of cellulose,
chitin and chitosan to filaments with simple salt solutions A2 – Blackburn, R.S. Biodegradable and
Sustainable Fibres: Woodhead Publishing; 2005. p. 367–397.
64. Lee, S.‐H. Ripening time and fiber formation of chitosan spinning dope. Journal of Applied Polymer
Science. 2003;90(10):2870–2877.
65. Ma, B., Qin, A., Li, X., and He, C. High tenacity regenerated chitosan fibers prepared by using the
binary ionic liquid solvent (Gly·HCl)‐[Bmim]Cl. Carbohydrate Polymers. 2013;97(2):300–305.
66. Notin, L., Viton, C., David, L., Alcouffe, P., Rochas, C., and Domard, A. Morphology and mechanical
properties of chitosan fibers obtained by gel‐spinning: Influence of the dry‐jet‐stretching step and age-
ing. Acta Biomaterialia. 2006;2(4):387–402.
67. Batra, K.S.and Pourdeyhimi, B. Raw Materials: Properties and Use in Fiberweb Nonwovens.
Introduction to Nonwovens Technology. Lancaster: DEStech Publications, Inc.; 2012.
68. Reddy, N. and Yang, Y. Chitin fibers. Innovative Biofibers from Renewable Resources. Berlin,
Heidelberg: Springer Berlin Heidelberg; 2015. p. 95–98.
69. Mincheva, R., Manolova, N., and Rashkov, I. Bicomponent aligned nanofibers of N‐carboxyethylchi-
tosan and poly(vinyl alcohol). European Polymer Journal. 2007;43(7):2809–2818.
70. Cooper, A., Bhattarai, N., Kievit, F.M., Rossol, M., and Zhang, M. Electrospinning of chitosan deriva-
tive nanofibers with structural stability in an aqueous environment. Physical Chemistry Chemical
Physics. 2011;13(21):9969–9972.
71. Du, J. and Hsieh, Y.‐L. Nanofibrous membranes from aqueous electrospinning of carboxymethyl chi-
tosan. Nanotechnology. 2008;19(12):125707.
72. Du, J. and Hsieh, Y.‐L. PEGylation of chitosan for improved solubility and fiber formation via electro-
spinning. Cellulose. 2007;14(6):543–552.
73. Li, L., Yuan, B., Liu, S., Yu, S., Xie, C., Liu, F. et al. Preparation of high strength chitosan fibers by
using ionic liquid as spinning solution. Journal of Materials Chemistry. 2012;22(17):8585–8593.
74. Hermawan, J., Hadyiswanto, I.O.C., Sitohang, R., and Winiati, W. Improvements of tensile properties and
durability of chitosan fiber using methanol drying treatment. Macromol Symp. 2015;353(1):147–153.
75. Yang, Q., Dou, F., Liang, B., and Shen, Q. Studies of cross‐linking reaction on chitosan fiber with
glyoxal. Carbohydrate Polymers. 2005;59(2):205–210.
76. Knaul, J.Z., Hudson, S.M., and Creber, K.A.M. Crosslinking of chitosan fibers with dialdehydes:
Proposal of a new reaction mechanism. Journal of Polymer Science Part B: Polymer Physics.
1999;37(11):1079–1094.
77. Choi, C.Y., Kim, S.B., Pak, P.K., Yoo, D.I., and Chung, Y.S. Effect of N‐acylation on structure and
properties of chitosan fibers. Carbohydrate Polymers. 2007;68(1):122–127.
78. Yang, S., Dong, Q., Yang, H., Liu, X., Gu, S., Zhou, Y. et al. N‐carboxyethyl chitosan fibers prepared
as potential use in tissue engineering. International Journal of Biological Macromolecules.
2016;82:1018–1022.
79. Zheng, H., Du, Y.M., Yu, J.H., Huang, R.H., and Zhang, L. Preparation and characterization of chi-
tosan/poly(vinyl alcohol) blend fibers. Journal of Applied Polymer Science. 2001;80(13):2558–2565.
80. Yan, W., Shen, L., Ji, Y., Yang, Q., and Shen, X. Chitin nanocrystal reinforced wet‐spun chitosan fibers.
Journal of Applied Polymer Science. 2014;131(19):40852.
81. Liu, S., Hua, T., Luo, X., Lam, N.Y., Tao, X.‐M., and Li, L. A novel approach to improving the quality of
chitosan blended yarns using static theory. Textile Research Journal. 2015;85(10):1022–1034.
82. Lam, N.Y.K., Zhang, M., Guo, H.‐F., Ho, C.P., and Li, L. Effect of fiber length and blending method
on the tensile properties of ring spun chitosan–cotton blend yarns. Textile Research Journal.
2017;87(2):244–257.
Chitosan Application in Textile Processing and Fabric Coating 425
83. Schlüter, K. Textile Auxiliaries, 3. Sizing Agents. Ullmann’s Encyclopedia of Industrial Chemistry.
Weinheim: Wiley‐VCH Verlag; 2011.
84. Siddique, A.B. Entwicklung von neuen Schlichtemitteln für die Gewebeherstellung auf Basis von
Chitosan unter technologischen und ökologischen Gesichtspunkten [Dissertation]. Denkendorf:
University of Stuttgart; 2005.
85. Renault, F., Sancey, B., Badot, P.M., and Crini, G. Chitosan for coagulation/flocculation pro-
cesses – An eco‐friendly approach. European Polymer Journal. 2009;45(5):1337–1348.
86. Singh, V. and Kumari, K. Some physicochemical measurements of chitosan/starch polymers in acetic
acid‐water mixtures. Macromol Symp. 2012;320(1):81–86.
87. Sudhakar, Y.N., Sowmya, Selvakumar M., and Bhat, D.K. Miscibility studies of chitosan and starch
blends in buffer solution. Journal of Macromolecular Science, Part A. 2012;49(12):1099–1105.
88. Stegmaier, T. Entwicklung von hocheffizienten, biologischabbaubaren Schlichtemitteln auf der Basis
von Chitosan. Denkendorf: Institut für Textil‐ und Verfahrenstechnik; 2005.
89. Hall, D.M. Slashing. In: Adanur S, editor. Wellington Sears Handbook of Industrial Textiles: CRC
Press 1995. p. 832.
90. Morey, F.C. Thickness of a liquid film adhering to a surface slowly withdrawn from the liquid.
Journal of Research of the National Bureau of Standards. 1940;25:385–393.
91. Wang, W. and Xu, D.S. Viscosity and flow properties of concentrated solutions of chitosan with dif-
ferent degrees of deacetylation International Journal of Biological Macromolecules.
1994;16(3):149–152.
92. Nguyen, T.T.B., Hein, S., Ng, C.H., and Stevens, W.F. Molecular stability of chitosan in acid solu-
tions stored at various conditions. Journal of Applied Polymer Science. 2008;107(4):2588–2593.
93. Yuan, Y., Chesnutt, B.M., Haggard, W.O., and Bumgardner, J.D. Deacetylation of chitosan: Material
characterization and in vitro evaluation via albumin adsorption and pre‐osteoblastic cell cultures.
Materials. 2011;4(8):1399.
94. Choi, W.Y., Park, H.J., Ahn, D.J., Lee, J., and Lee, C.Y. Wettability of chitosan coating solution on
‘Fuji’ apple skin. Journal of Food Science. 2002;67(7):2668–2672.
95. Kurek, M., Brachais, C.‐H., Ščetar, M., Voilley, A., Galić, K., Couvercelle, J.‐P. et al. Carvacrol
affects interfacial, structural and transfer properties of chitosan coatings applied onto polyethylene.
Carbohydrate Polymers. 2013;97(1):217–225.
96. Katalinich, M. Characterization of chitosan films for cell culture applications [Master Thesis]. Orono:
The University of Maine; 2001.
97. Siddique, A.B., Stegmaier, T., Wunderlich, W., Hager, T., and Planck, H., editors. Analysis of thermal
behavior of chitosan to use in textile sizing. 2nd International Conference of Textile Research
Division. 2005; Cairo.
98. Obaidat, R., Al‐Jbour, N., Al‐Sou’d, K., Sweidan, K., Al‐Remawi, M., and Badwan, A. Some phys-
ico‐chemical properties of low molecular weight chitosans and their relationship to conformation in
aqueous solution. Journal of Solution Chemistry. 2010;39(4):575–588.
99. Cunha, A.G., Fernandes, S.C.M., Freire, C.S.R., Silvestre, A.J.D., Neto, C.P., and Gandini, A. What
is the real value of chitosan’s surface energy? Biomacromolecules. 2008;9(2):610–614.
100. Qun, G. and Ajun, W. Effects of molecular weight, degree of acetylation and ionic strength on surface
tension of chitosan in dilute solution. Carbohydrate Polymers. 2006;64(1):29–36.
101. Lee, D.W., Lim, C., Israelachvili, J.N., and Hwang, D.S. Strong adhesion and cohesion of chitosan in
aqueous solutions. Langmuir. 2013;29(46):14222–14229.
102. Mati‐Baouche, N., Elchinger, P.‐H., de Baynast, H., Pierre, G., Delattre, C., and Michaud, P. Chitosan
as an adhesive. European Polymer Journal. 2014;60:198–212.
103. Baumann, M.G.D. and Conner, A.H. Carbohydrate polymers as adhesives. In: Pizzi A. and Kashmiri,
L.M., editors. Handbook of Adhesive Technology. 2 ed. Boca Raton: CRC Press; 2003.
104. Hu, Z., Hong, P.Z., Liao, M.N., Kong, S.Z., Huang, N., Ou, C.Y. et al. Preparation and characteriza-
tion of chitosan‐agarose composite films. Materials. 2016;9(10):816.
426 Chitin and Chitosan: Properties and Applications
105. Singh, D.K. and Ray, A.R. Characterization of grafted chitosan films. Carbohydrate Polymers.
1998;36(2):251–255.
106. Krkić, N., Lazić, V., and Šuput, D. Improvement of mechanical properties of chitosan film. Journal
on Processing and Energy in Agriculture. 2012;16:103–105.
107. Souza, V.G.L., Fernando, A.L., Pires, J.R.A., Rodrigues, P.F., Lopes, A.A.S., and Fernandes, F.M.B.
Physical properties of chitosan films incorporated with natural antioxidants. Industrial Crops and
Products. 2017;107:565–572.
108. Patel, A.K. Chitosan: Emergence as potent candidate for green adhesive market. Biochemical
Engineering Journal. 2015;102:74–81.
109. Lazaridou, A. and Biliaderis, C.G. Thermophysical properties of chitosan, chitosan–starch and chi-
tosan–pullulan films near the glass transition. Carbohydrate Polymers. 2002;48(2):179–190.
110. Bujang, A., Adila, S., and Suyatma, N., editors. Physical properties of chitosan films as affected by
concentration of lactic acid and glycerol. 4th International Conference on Biology, Environment and
Chemistry; 2013; Singapore: IACSIT Press.
111. Gebert, K. Textile Auxiliaries, 4. Pretreatment Auxiliaries. Ullmann’s Encyclopedia of Industrial
Chemistry. Weinheim: Wiley‐VCH Verlag GmbH & Co. KGaA; 2012.
112. Aly, A.S., Sayed, S.M., and Zahran, M.K. One‐step process for enzymatic desizing and bioscouring
of cotton fabrics. Journal of Natural Fibers. 2010;7(2):71–92.
113. Madhu, A. and Chakraborty, J.N. Developments in application of enzymes for textile processing.
Journal of Cleaner Production. 2017;145:114–133.
114. Cakara, D., Fras, L., Bracic, M., and Kleinschek, K.S. Protonation behavior of cotton fabric with
irreversibly adsorbed chitosan: A potentiometric titration study. Carbohydrate Polymers.
2009;78(1):36–40.
115. Jabli, M., Baouab, M.H.V., Roudesli, M.S., and Bartegi, A. Adsorption of acid dyes from aqueous
solution on a chitosan‐cotton composite material prepared by a new pad‐dry process. Journal of
Engineered Fibers and Fabrics. 2011;6(3):1–12.
116. Mitchell, R., Carr, C.M., Parfitt, M., Vickerman, J.C., and Jones, C. Surface chemical analysis of raw
cotton fibres and associated materials. Cellulose. 2005;12(6):629–639.
117. Feitkenhauer, H., Fischer, D., and Fah, D. Microbial desizing using starch as model compound:
Enzyme properties and desizing efficiency. Biotechnology Progress. 2003;19(3):874–879.
118. Schindler, W.D. and Hauser, P.J. 1 – Introduction to chemical finishing. Chemical Finishing of
Textiles: Woodhead Publishing; 2004. p. 1–6.
119. Araujo, R., Casal, M., and Cavaco‐Paulo, A. Application of enzymes for textile fibres processing.
Biocatalysis and Biotransformation. 2008;26(5):332–349.
120. Mojsov, K. Trends in bio‐processing of textiles. Advanced Technologies. 2014;3(2):135–138.
121. Roy Choudhury, A.K. 1 – Introduction to finishing. Principles of Textile Finishing: Woodhead
Publishing; 2017. p. 1–19.
122. Ian, H. Innovative technologies for high performance textiles. Coloration Technology.
2007;123(2):59–73.
123. Holme, I. New Developments in the chemical finishing of textiles. The Journal of The Textile Institute.
1993;84(4):520–533.
124. Bhala, R., Dhandhania, V., and Periyasamy, A.P. Bio‐finishing of fabrics. Wet Processing.
2012;9(4):45–49.
125. Roy, J.C., Ferri, A., Salaun, F., Giraud, S., Chen, G., Jinping, G. et al. Chitosan‐carboxymethylcel-
lulose based microcapsules formulation for controlled release of active ingredients from cosmeto
textile. 17th World Textile Conference Autex 2017 – Shaping the Future of Textiles. IOP Conference
Series‐Materials Science and Engineering. 254. Bristol: Iop Publishing Ltd; 2017.
126. Liu, J., Liu, C., Liu, Y., Chen, M., Hu, Y., and Yang, Z. Study on the grafting of chitosan–gelatin
microcapsules onto cotton fabrics and its antibacterial effect. Colloids and Surfaces B: Biointerfaces.
2013;109:103–108.
Chitosan Application in Textile Processing and Fabric Coating 427
127. Hu, J., Xiao, Z.‐B., Zhou, R.‐J., Ma, S.‐S., Li, Z., and Wang, M.‐X. Comparison of compounded
fragrance and chitosan nanoparticles loaded with fragrance applied in cotton fabrics. Textile Research
Journal. 2011;81(19):2056–2064.
128. Wang, H., Yu, L., and Chen, C. Antibacterial and moisture retention evaluation of cotton fabric treated
with chitosan incorporated ceramide microcapsules. Sen’i Gakkaishi. 2010;66(8):204–208.
129. Busila, M., Musat, V., Textor, T., and Mahltig, B. Synthesis and characterization of antimicrobial
textile finishing based on Ag:ZnO nanoparticles/chitosan biocomposites. RSC Advances. 2015;
5(28):21562–21571.
130. Shahid ul I., Shahid, M., and Mohammad, F. Green chemistry approaches to develop antimicrobial
textiles based on sustainable biopolymers—A review. Industrial & Engineering Chemistry Research.
2013;52(15):5245–5260.
131. Kitkulnumchai, Y., Ajavakom, A., and Sukwattanasinitt, M. Treatment of oxidized cellulose
fabric with chitosan and its surface activity towards anionic reactive dyes. Cellulose. 2008;
15(4):599–608.
132. Cheng, X., Ma, K., Li, R., and Ren, X., and Huang, T.S. Antimicrobial coating of modified chitosan
onto cotton fabrics. Applied Surface Science. 2014;309:138–143.
133. Aly, A.S., Hashem, A., and Hussein, S.S. Utilization of chitosan citrate as crease‐resistant and
antimicrobial finishing agent for cotton fabric. Indian Journal of Fibre & Textile Research.
2004;29(2):218–222.
134. Chung, Y.‐S., Lee, K.‐K., and Kim, J.‐W. Durable press and antimicrobial finishing of cotton fabrics
with a citric acid and chitosan treatment. Textile Research Journal. 1998;68(10):772–775.
135. Pascual, E. and Julià, M.R. The role of chitosan in wool finishing. Journal of Biotechnology.
2001;89(2):289–296.
136. Ranjbar‐Mohammadi, M., Arami, M., Bahrami, H., Mazaheri, F., and Mahmoodi, N.M. Grafting of
chitosan as a biopolymer onto wool fabric using anhydride bridge and its antibacterial property.
Colloids and Surfaces B: Biointerfaces. 2010;76(2):397–403.
137. Ranjbar‐Mohammadi, M., Bahrami, S.H., and Arami, M. Eco‐friendly grafting of natural biopolymer
chitosan onto acylated wool fabrics using ultrasonic and study its properties. Journal of Applied
Polymer Science. 2013;129(2):707–713.
138. Goy, R.C., Britto, D.D., and Assis, O.B.G. A review of the antimicrobial activity of chitosan.
Polímeros. 2009;19:241–247.
139. Kong, M., Chen, X.G., Xing, K., and Park, H.J. Antimicrobial properties of chitosan and mode of
action: A state of the art review. International Journal of Food Microbiology. 2010;144(1):51–63.
140. Raafat, D. and Sahl, H.G. Chitosan and its antimicrobial potential – a critical literature survey.
Microbial Biotechnology. 2009;2(2):186–201.
141. Gao, Y., Cranston R. Recent advances in antimicrobial treatments of textiles. Textile Research
Journal. 2008;78(1):60–72.
142. Samanta, K.K., Basak, S., and Chattopadhyay, S.K. Specialty chemical finishes for sustainable luxu-
rious textiles. In: Gardetti, M.A. and Muthu, S.S., editors. Handbook of Sustainable Luxury Textiles
and Fashion: Volume 1. Singapore: Springer Singapore; 2015. p. 145–184.
143. Simoncic, B. and Tomsic, B. Structures of novel antimicrobial agents for textiles – A review. Textile
Research Journal. 2010;80(16):1721–1737.
144. Kardas, I., Struszczyk, M.H., Kucharska, M., van den Broek, L.A.M., van Dam, J.E.G., and
Ciechańska, D. Chitin and chitosan as functional biopolymers for industrial applications. In: Navard
P, editor. The European Polysaccharide Network of Excellence (EPNOE): Research Initiatives and
Results. Vienna: Springer Vienna; 2013. p. 329–373.
145. Periolatto, M. and Ferrero, F. Cotton filter fabrics functionalization by chitosan UV‐grafting for
removal of dyes. In: Pierucci, S. and Klemes, J.J., editors. Icheap‐11:11th International Conference
on Chemical and Process Engineering, Pts 1–4. Chemical Engineering Transactions. 32. Milano:
Aidic Servizi Srl; 2013. p. 85–90.
428 Chitin and Chitosan: Properties and Applications
146. Ali N, Awais, Kamal, T., Ul‐Islam, M., Khan, A., Shah, S.J. et al. Chitosan‐coated cotton cloth sup-
ported copper nanoparticles for toxic dye reduction. International Journal of Biological
Macromolecules. 2018;111:832–838.
147. Lalmi, S., Kameche, M., Innocent, C., Haddou, B., Derriche, Z., and Pourcelly, G. Adsorption of
biodegradable polyelectrolyte onto cotton for fixation of copper and lead: Comparison to a Cation
Exchange Textile. Separation Science and Technology. 2013;48(1):156–165.
148. Qu, R., Sun, C., Ma, F., Zhang, Y., Ji, C., Xu, Q. et al. Removal and recovery of Hg(II) from aqueous
solution using chitosan‐coated cotton fibers. Journal of Hazardous Materials. 2009;167(1):
717–727.
149. Tonetti, C., Ferrero, F., Periolatto, M., Vineis, C., Varesano, A., and Mazzuchetti, G., editors. Chitosan
coated cotton textiles for copper and chromium ions adsorption. 9th International Izmir Textile and
Apparel Symposium; 2014; 2.4–5.4.2014; Izmir.
150. Campos, J., Diaz‐Garcia, P., Montava, I., Bonet‐Aracil, M., and Bou‐Belda, E. Chitosan pretreatment
for cotton dyeing with black tea. 17th World Textile Conference Autex 2017 – Shaping the Future of
Textiles. IOP Conference Series‐Materials Science and Engineering. 2542017.
151. Hao, L.Y., Wang, R., Fang, K.J., and Cai, Y.Q. The modification of cotton substrate using chitosan for
improving its dyeability towards anionic microencapsulated nano‐pigment particles. Industrial Crops
and Products. 2017; 95:348–356.
152. Periolatto, M., Ferrero, F., Vineis, C., and Rombaldoni, F. Multifunctional finishing of wool fabrics
by chitosan UV‐grafting: An approach. Carbohydrate Polymers. 2013;98(1):624–629.
153. Kotb, R.M. Innovative Multi‐Protection treatments and free‐salt dyeing of cotton and silk fabrics.
Journal of Engineered Fibers and Fabrics. 2017;12(3):60–71.
154. Roberts, G.A.F. and Wood, F.A. A study of the influence of structure on the effectiveness of chitosan
as an anti‐felting treatment for wool. Journal of Biotechnology. 2001;89(2–3):297–304.
155. Muzaffar, S., Bhatti, I.A., Zuber, M., Bhatti, H.N., and Shahid, M. Synthesis, characterization and
efficiency evaluation of chitosan‐polyurethane based textile finishes. International Journal of
Biological Macromolecules. 2016;93:145–155.
156. Shi, J., Han, X., and Kelu, Y. A novel bio‐functional finishing agent for wool based on waterborne
polyurethane mixed with chitosan. Textile Research Journal. 2014;84(11):1174–1182.
157. Gupta, D. and Haile, A. Multifunctional properties of cotton fabric treated with chitosan and carboxy-
methyl chitosan. Carbohydrate Polymers. 2007;69(1):164–171.
158. Roberts GAF. Thirty years of progress in chitin and chitosan. Progress on Chemistry and Application
of Chitin and its Derivatives. 2008;13:7–15.
159. Einbu, A. Characterisation of chitin and a study of its scid‐catalysed hydrolysis [Doctoral Thesis].
Trondheim: Norwegian University of Science and Technology; 2007.
160. Ríos, D.A.G. Technical and economic feasibility analysis of a plant process for production of chi-
tosan from shrimp shells in Colombia. Medellín: University of Antioquia; 2015.
17
Chitin and Chitosan for Water
Purification
Petrisor Samoila, Andra Cristina Humelnicu, Maria Ignat,
Corneliu Cojocaru, and Valeria Harabagiu
‘Petru Poni’ Institute of Macromolecular Chemistry, Romanian Academy, Iași, Romania
The interest in developing new and more efficient materials and technologies for the
removal of different pollutants from wastewater is constantly growing. To this end, chitin
and chitosan have attracted the attention of many researchers because of their interesting
removal capacities for certain hazardous pollutants such as water‐soluble organics (phe-
nols, dyes, pesticides, herbicides, etc.) and heavy metal ions. Moreover, these biocompat-
ible and biodegradable carbohydrate polymers fall in the category of low‐cost sorbents that
are abundant and renewable resources. Nevertheless, the limited solubility of chitin and the
high sensitivity to pH of chitosan generate some feasibility issues. In this respect, many
studies suggest the chemical modification of chitin and chitosan, in order to develop deriv-
atives and composites with improved properties for water purification purposes. In this
chapter, chitin and chitosan, together with their derivatives and composites, are reviewed
from the perspective of water purification applications, underlining their advantages for the
removal of representative organic pollutants and heavy metals from aqueous solutions.
Likewise, the principles and mechanisms governing the purification processes such as
adsorption, coagulation/flocculation and ultrafiltration are detailed.
17.1 Introduction
Water contamination by a wide range of toxic compounds, particularly organic pollutants
and heavy metals, has become a serious environmental concern because of the negative
impact on the quality of drinking water for generations to come. Under these circum-
stances, the interest on developing new and more efficient materials and technologies for
removal of polluting substances from wastewaters is in constant growth. Depending on the
processes involved, wastewater treatments can be classified as: physical, chemical and bio-
logical. The selection of the appropriate treatment is made by considering the pollutant
nature and concentration, as well as the investment and operational costs. Because of their
versatility and wide range of properties, polymers (either synthetic or natural) and polymer‐
based composites are playing a key role in wastewater treatments such as: adsorption [1],
coagulation/flocculation [2] or membrane separation by (micro‐/ultra‐/nano‐/hyper‐) filtra-
tion processes [3].
In comparison with synthetic polymers, natural polymers gained the attention of
researchers in order to develop cheaper and more effective adsorbents. Biopolymers repre-
sent a real alternative to synthetic polymers owing to their distinguished structure, physico-
chemical characteristics, high reactivity and chemical stability. The presence of different
functional groups in the chemical structure (hydroxyl, carboxyl, amino or acetamido) per-
mits the synthesis of new bioadsorbents with high selectivity to aromatic compounds and
metals [4]. Among these, polysaccharides such as chitin [5], chitosan [6], starch [7], cel-
lulose [8] and their derivatives are known for their eco‐friendly properties such as biodeg-
radability, non‐toxicity and biocompatibility [9]. Likewise, their ability to bind with a large
variety of molecules by physical and chemical interactions is of great interest. Therefore,
their efficient adsorption property for toxic compounds made them ideal candidates for
water treatment (removal of pollutants) [10] and for separation processes (recovery of
important metals) [11].
Chitin and chitosan were extensively studied and proved to have attractive removal
capacities for certain pollutants such as water‐soluble organics and metal ions. Moreover,
these carbohydrate polymers (chitin and chitosan) fall in the category of low‐cost sorbents,
if compared to commercial activated carbons or synthetic ion exchange materials.
Industrially, chitin – known as the second most abundant natural polymer on a global
scale – is extracted mainly from crustaceans (crab, krill, crayfish) by a three‐step procedure
as follows: (i) calcium carbonate dissolution by means of an acid treatment; (ii) protein
solubilisation by alkaline extraction; and (iii) pigment removal by decolourisation.
Consequently, chitosan – the most important chitin derivative – is usually obtained by par-
tial deacetylation in alkaline media [12]. In spite of the beneficial properties of these natu-
ral polymers, such as being environment‐friendly, abundant, biodegradable and renewable
resources, the limited solubility of chitin and the high sensitivity to pH of chitosan impose
some feasibility issues [13, 14]. In this respect, many studies have been published on the
chemical modification of chitosan. According to these studies, the chemical modification
of chitosan yielded derivatives and composites with improved properties for wastewater
treatment purposes [14].
To obtain chitosan‐based sorbents with adequate physical–chemical properties, cross‐
linking agents are often employed. Such cross‐linking agents imply: glutaraldehyde [12, 15],
genipine [16], glyoxal [17], epichlorohydrin [18] or ethylene glycol diglycidyl ether [19].
Some of the commonly used cross‐linking agents are detailed in Figure 17.1.
Chitin and Chitosan for Water Purification 431
CHEMICAL CROSS-LINKING
CS OH OH
CS H
CS OH CS N
H-C=O N=C-H H OH
NH2 NH2 O
+ + H
NH2 NH2
N=C-H O NH
H-C=O H
CS CS O OCH3 CS
CS
Glutaraldehyde Genipine
NH2 OH
OH NH2 NH2
OH OH
CS CS
CS CS CS
CS O CS
O O
N O O O
NH2 NH2
+ + OH sau
NH2 sau NH2 OH
O CS CI O O
CS O N
O
OH CS CS CS
OH CS
CS
OH NH2 NH2
OH
NH2
Glyoxal Epichlorohydrin
OH OH
CS CS
OH
NH OH
NH
O=C=N O
CS O NH CS HO
NH2 NH2 O O
+
NH2 +
NH2
O O
CS O=C=N CS
O NH OH
O
OH NH NH
OH
CS CS
OH OH
4,4ʹ-Methylenebis (phenyl isocyanate) Ethylene glycol diglycidyl ether
PHYSICAL CROSS-LINKING
OH
CS
OH
O– Na+ O– NH3+
CS Na O P = O
+ –
Na O– P = O
+
O O
NH2
+ Na+ O– P = O Na+ O– P = O
NH2
O O
CS Na+ O– P = O Na+ O–
P= O
+
O– Na+ O– NH3
OH
CS
OH
Pentasodium triphosphate
Regarding solute affinity, it can be distinguished into three types of adsorption: physical,
ion exchange and chemical adsorption. Physical adsorption (or physisorption) is explained
mainly by Van der Waalls attraction complemented by electrostatic forces between the
adsorbate and the surface of the adsorbent [28]. The second type of adsorption relies mostly
on electrostatic forces occurring between ions retained on the surface, and is often called
ion exchange adsorption [29]. Chemical adsorption (or chemisorption) is the result of the
chemical interaction between the solid and the adsorbed substance. Chemisorption takes
place when the substance is linked to the solid surface by covalent bonds. Usually, physical
adsorption may occur on all surfaces, provided the pressure and temperature conditions are
favourable. In contrast, chemisorption is highly more selective. Also, it is possible only
between specific adsorptive and adsorbent species, especially if the active sites are not
blocked by previously adsorbed molecules [30].
The adsorption process is dependent on several factors, from the adsorbent nature and
structure to the physical and chemical properties of the adsorbate and the reaction medium.
Therefore, the most important factors that affect absorption capacity are: properties of the
sorbate (functional groups, polarity, hydrophobic/hydrophilic character, size and molecular
weight), medium condition (pH and temperature of solution, sorbent concentration and ionic
strength) and the type of sorbent (surface area, porosity, functional groups distribution) [31].
The adsorption process is based on the solute’s removal from a solution and changing the
surface concentration until an equilibrium between the solution and adsorbent is attained.
The adsorption amount at equilibrium is usually determined in accordance with the follow-
ing equation [32]:
C0 C V
q Eq. 17.1
m 1000
where q denotes the adsorption capacity (mg·g−1); C0 and C are, respectively, the initial and
final concentrations of pollutant in solution (mg·L−1); V is the volume of the solution (mL);
and m represents the mass of the adsorbent (g).
Frequently, adsorption isotherms are used to describe the equilibrium of adsorption of a
material on a surface (more generally, on a boundary surface) at a constant temperature.
Literature reports often provide isotherm models, such as Freundlich, Langmuir, Redlich–
Peterson, Sips and Dubinin–Radushkevich [33]. The conventional adsorption isotherm
equations are Langmuir and Freundlich isotherms, since both can be transformed into lin-
ear forms, and so the main parameters of the adsorption process are easily calculated either
by linear regression or by graphical means [34]. In addition, the Dubinin–Radushkevich
model is often used to determine the adsorption process type (physical, ion exchange or
chemisorption). The mean free energy of sorption ES (kJ·mol−1) is the main parameter of
this model that indicates the nature of adsorption. Generally, the process is considered to be
based on physisorption if ES <8 (kJ·mol−1). In turn, the sorption process relies on ion
exchange if ES ranges from 8 to 16 kJ·mol−1 [35]. For higher values of mean free energy,
the adsorption mechanism might be attributed to chemical interactions (chemisorptions).
In addition, thermodynamic parameters such as Gibbs free energy of adsorption (ΔGad) can
also indicate the nature of adsorption. For example, ΔGad ranges of 0 to −20 kJ·mol−1 and
434 Chitin and Chitosan: Properties and Applications
from synthetic waters by chemically modifying chitosan. In this respect, chitosan was sub-
jected to a cross‐linking process using 2‐hydroxy‐1‐naphthaldehyde. Further, the function-
alised biopolymer was modified by grafting with copper(II) chloride. The maximum
adsorption capacities were very sensitive to the modification of chitosan. The cross‐linked
chitosan exhibited the highest pesticide adsorption (39.1 mg·g−1) followed by the grafted
material (35.4 mg·g−1) and the unmodified polysaccharide (24.4 mg·g−1). These observations
shed light on the beneficial effect of chemical modification over the capacity of chitosan to
adsorb organic pollutants.
In their seminal paper, El Harmoudi et al. [45] recovered chitin by extraction technique
from shellfish waste and produced chitosan by the deacetylation of chitin. Further, they
used the obtained biopolymers for the removal of one of the most common organic herbi-
cides: 2,4‐dichlorophenoxyacetate (2,4‐D). During the adsorption tests, the researchers
studied the influence of some factors such as pH, the contact time and initial pollutant
concentration over the maximum adsorption capacity. The adsorption of the herbicide onto
chitosan and chitin surface was optimal at pH 3.7. According to their findings, at this pH
value, the 2,4‐D was in anionic form, and the functional groups (hydroxyl, amine and
amide groups) of the polysaccharides were protonated, favouring electrostatic attraction of
the system components. The adsorption capacities estimated from Langmuir equation were
equal to 11 mg·g−1 for chitosan and 6 mg·g−1 for chitin.
Other authors [39] dealt with montmorillonite–chitosan bionanocomposites applied as
adsorbents for 3,6‐dichloropyridine‐2‐carboxylic acid (clopyralid) as a representative mol-
ecule for anionic herbicides. They conducted their study by simulating waters and soil/
water suspensions polluted with clopyralid in order to determine the potential of their mate-
rials to prevent and remediate both water and soil contaminated with herbicides. The results
reported in this work underlined the main role of pH over adsorption performances. The
montmorillonite–chitosan composites showed good adsorption capacities at pH values that
enabled the protonated form of chitosan and anionic form of clopyralid, suggesting an ion
exchange adsorption mechanism. Moreover, the addition of bionanocomposites to acidic
soil significantly increased the pollutant adsorption, whereas negligible adsorption perfor-
mances were observed for alkaline soils. These observations recommended the immobilisa-
tion of montmorillonite–chitosan materials in certain soils to prevent the pollution of ground
waters. In another study, Ding et al. [46] designed the same kind of bionanocomposites for
removing the herbicide 3,7‐dichloroquinoline‐8‐carboxylic acid (quinclorac) from aqueous
solution. This study highlighted once again the influence of pH over adsorption perfor-
mances of montmorillonite–chitosan materials. These authors observed that the increase in
pH from 2.9 to 5.0 positively affected the adsorption capacity. In contrast, the pH increase
from 5.0 to 10.3 was detrimental for the maximum adsorption capacity. Furthermore, the
authors conducted desorption studies in order to elucidate the adsorption mechanism. The
results indicated a very high desorption rate in alkaline media as compared to distilled
water, suggesting that adsorption of quinclorac onto montmorillonite–chitosan biocompos-
ites occurred mainly by electrostatic attraction.
One of the most concerning environmental problem regarding wastewaters is caused by
the textile industry. The coloured effluents that result from textile manufacturing are gener-
ally considered to be highly toxic, since dyes are reported to cause allergy, skin irritation,
dermatitis and even cancer. Thus, the removal of dyes from effluents prior to their mixing
with clean natural water bodies is of crucial importance. Adsorption via natural biopolymers
436 Chitin and Chitosan: Properties and Applications
such as chitin and chitosan was proved to be an effective treatment for discolouration of
textile dyes from effluents. Chemical modifications of these polysaccharides can induce
changes on their properties by promoting new materials with more appropriate characteris-
tics for the removal of various dyes.
The literature survey has indicated that chitosan is very effective for the adsorption of
anionic dyes (reactive and acid dyes), especially in an acidic environment, following the
ion exchange mechanism illustrated in Figure 17.2.
According to the adsorption mechanism proposed by Tan et al. [47], the amino groups of
chitosan are protonated in the presence of high concentrations of protons. Also, in acidic
conditions, anionic dye molecules are dissociated, favouring the electrostatic attraction
between anionic dye ions and protonated amino group of chitosan.
Recently, Humelnicu et al. [33] have reported on the adsorptive removal of anionic dyes
by rare‐earth‐doped cobalt spinel ferrite functionalised with chitosan cross‐linked with
epichlorohydrine. The selection of the cross‐linking agent was made in order to preserve
the amino groups’ number from the pristine polysaccharide (chitosan). The developed
composite material (CoFe1.98Sm0.02O4@Chitosan‐cL‐Epichlorohydrine) was used as sorb-
ent for Orange II removal in batch conditions. The maximal adsorption capacity was found
to be 182 mg·g−1. The model‐based optimisation was performed for the adsorption process
to evaluate the effects of pH and sorbent dosage over adsorption performances. The model‐
based optimisation performed by stochastic simulation using the Monte‐Carlo method sug-
gested that the optimal values of factors were pH 5.2 and sorbent dosage 1.60 g·L−1.
Furthermore, the energy values (ES) determined from Dubinin–Radushkevich isotherm
ranged from 9.85–11.78 kJ mol−1 depending on the temperature. These values were
included into the interval 8–16 kJ mol−1, suggesting that the mechanism predominant for
this adsorption process was ion exchange mechanism.
Nevertheless, chitosan‐based materials were also used for the removal of some cationic
dyes. Kamal et al. [48] investigated a new type of titanium oxide/chitosan nanocomposite
as an adsorbent for thymol violet dye removal. Both composite material and pristine chi-
tosan were adhered on the cellulose microfibre mat by using the doctor‐blade method. The
adsorption properties were evaluated as a function of pH, adsorbent dosage and contact
time. As expected, the optimum pH value for the adsorption process was found to be 7.96.
These authors argued that the superior values of adsorption capacities could be due to the
increasing number of negatively charged sites on the adsorbent. However, no details were
H+
CS NH2 CS NH3+
Protonation
H2O
Dye SO3Na Dye SO3– + Na+
Dissociation
Figure 17.2 Schematic representation of the adsorption mechanism of anionic dyes onto the chitosan
surface; adapted from [47].
Chitin and Chitosan for Water Purification 437
given about the nature of these negatively charged sites onto the adsorbent surface.
Likewise, adsorption of the studied dye and adsorbent dosage were into a directly propor-
tional relation. Kinetic studies revealed that the composite adsorbed the dye rapidly and
equilibrium was achieved in 90 min, comparing with pure chitosan coated with cellulose.
In addition, the adsorption process involved particle diffusion mechanism and followed
pseudo‐second order kinetics. The calculated maximum adsorption capacities for the
adsorbents were equal to 84.32 mg·g−1 (chitosan–cellulose microfibre mat) and 97.51
mg·g−1 (TiO2/chitosan–cellulose microfibre mat), respectively.
Composite hydrogel beads based on chitosan and halloysite nanotubes were used as
absorbents for the removal of both methylene blue and malachite green from aqueous solu-
tions by Peng et al. [49]. The adsorbent materials were prepared by the dropping and pH‐
precipitation method. Adsorption data were fitted using Langmuir and Freundlich isotherm
models. It was observed that the addition of clay mineral into chitosan beads led to a sig-
nificant increase in the adsorption rate for both pollutants. Therefore, the maximum absorp-
tion capacity achieved for methylene blue was 270.27 mg·g−1 and for malachite green was
303.03 mg·g−1, attributed mainly to the numerous active sites of halloysite nanotubes.
Moreover, a regeneration experiment was done and showed that the adsorption capacity of
all hydrogel beads could be restored, especially for the case of methylene blue uptake.
Ultimately, it was inferred that chitosan/halloysite composite showed great potential as
reusable adsorbents for dye removal.
Table 17.1 summarises the performances of some adsorption processes dealing with the
uptake of organic dyes by chitosan‐based materials along with the adopted conditions of
experimentation. As one may notice from Table 17.1, most studies were focused on anionic
dyes (but not exclusively) and conducted in acidic environments.
No. Chitosan‐based sorbent Form Dye Qmax (mg·g−1, mmol·g−1) pH T (°C) Reference
1 Chitosan Microparticles Acid Green 25 525 3 [51]
2 Chitosan Powder Acid Blue 9 210 3 25 [52]
Food Yellow 3 295
3 Chitosan Flakes Acid Green 25 645.1 4 25 [53]
Acid Orange 10 922.9
Acid Orange 12 973.3
Acid Red 18 693.2
Acid Red 73 728.2
4 Chitosan/Ethylenediamine Nanoparticles Acid Orange 7 3.47 4 25 [54]
Acid Orange 10 2.25 3
5 Chitosan/graphene Nano‐composite Acid Orange 7 42.7 3 25 [55]
6 Chitosan/TPP Nanoparticles Acid Green 27 1051.8 5 25 [56]
7 Chitosan/4‐formyl‐benzene‐1,3‐disulphonic acid Powder Basic Blue 3 166.5 3 25 [57]
8 Chitosan‐graft‐l‐monoguluronic acid Powder Congo Red 459.75 5 30 [58]
9 Chitosan‐g‐poly (acrylic acid) Microparticles Methylene Blue 1873 5 30 [59]
Chitosan‐g‐attapulgite 1848
10 Chitosan/Al2O3/Fe2O3 Nanoparticles Methyl Orange 417 25 [60]
11 Chitosan/Epichlorohydrine Beads Metanil Yellow 1334 3 30 [61]
Reactive Blue 15 722
12 Chitosan/montmorillonite Hydrogel Nitrazine Yellow 177 5 25 [62]
13 Chitosan/Epichlorohydrine Microparticles Remazol Black 2941 3 30 [63]
14 Chitosan‐graft‐poly(methylmethacrylate) Powder Procion Yellow MX 250 7 20–30 [64]
Remazol Brilliant Violet 357
Reactive Blue H5G 178
15 Chitosan‐graft‐poly(acrylamide) Powder Remazol Violet 1428 4 30 [65]
Procion Yellow 1000
16 Chitosan‐g‐poly(acrylamide) Powder Remazol Yellow Gelb 3RS 1211 2 25 [66]
Basic Yellow 37 528
17 Chitosan‐N‐lauryl/Fe2O3 Particles Remazol Red 198 267 25 [67]
18 Chitosan/Epichlorohydrine Beads Reactive Red 189 1642–1936 3 30 [68]
19 Chitosan/Sodium tripolyphosphate/Epichlorohydrine Beads Reactive Red 189 1802–1840 3 30 [69]
20 Chitosan‐g‐poly(methyl methacrylate) Powder Orange‐G 435 7 [70]
(a) (b)
CT CS
NHAc NH2
Mn+ Mn+
NHAc NH2
CT CS
Figure 17.3 Schematic representation of the adsorption mechanism of heavy metals onto (a) chitin and
(b) chitosan materials.
formed involving the pair of electrons shared by the nitrogen atoms of biopolymers (donor)
with the metal cations (acceptor).
Chitin/chitosan are reported as efficient adsorbents for a very wide range of heavy met-
als, starting with elements usually present in the living organisms in low/moderate quantity,
but toxic at higher concentrations (Fe, Zn, Co, Cu, Mn, etc.), and finishing with Cd, As, Pb,
Hg or Cr, which are extremely hazardous even at lower concentrations.
For example, Forutan et al. [71] studied the adsorption of Pb(II) from synthetic wastewa-
ters by the chitin extracted from shells of pink shrimps. In order to evaluate the influence of
pH, sorbent dosage and initial concentration of lead, they conducted experiments varying
exclusively from the studied parameter. They reported a maximum adsorption capacity of
7.003 mg·g−1 in the following experimental conditions: initial concentration of lead equal to
7.99 ppm, absorbent dosage of 5 g·L−1, temperature 30°C and pH 9. In addition, Karthik and
Meenaksh [72] utilised chitin grafted with polypyrol for Pb(II) and Cd(II) ions removal
from aqueous solutions. They reported maximum adsorption capacities of 9.14 mg·g−1 for
lead and 6.49 mg·g−1 for cadmium, at 50°C. The adsorption mechanism suggested by these
authors involved electrostatic attraction followed by a final complexation step.
Singh and Nagendran [73] compared the adsorption efficiencies of chitin and chitosan
for chromium (III) from synthetic wastewaters. The maximum adsorption values estimated
from the Langmuir isotherm were very sensitive to the biopolymer nature. According to
their results, the deacetylated polysaccharide is far more suitable for Cr3+ removal. For
instance, the maximum adsorption capacity values were equal to 51.12 mg·g−1 for Cr3+/
chitosan, and only 7.738 mg·g−1 for the Cr3+/chitin adsorption system.
Ariff, Hanafiah and Ngah [74] prepared cross‐linked chitosan‐coated bentonite beads to
investigate their adsorption efficiency regarding Cu (II) removal form aqueous solution.
The beads were sieved in order to retain the microparticles greater than 200 μm. Several
factors impaired the absorption capacity. Therefore, the effects of temperature, stirring time
and Cu(II) concentrations were assessed. Adsorption kinetics was evaluated to determine
the optimum stirring time necessary for Cu(II) adsorption. The experimental data were
440 Chitin and Chitosan: Properties and Applications
No. Chitosan‐based sorbent Form Heavy Metal Qmax (mg·g−1, mmol·g−1) pH T (°C) Reference
1 Chitosan Powder/solid Pb2+
141.10 4.5 20 [85]
Ni2+ 52.81 5.5
2 Chitosan Nanofibres Cu2+ 485.44 7 25 [86]
Pb2+ 263.15
3 Chitosan Particles Hg2+ 1127.1 6 25 [87]
4 Chitosan/Epichlorohydrine Powder Cr2+ 78 5 [88]
Cu2+ 80 3–5
5 Chitosan/glutaraldehyde Aerogel Cu2+ 35.08 25 [89]
6 Chitosan/graphene oxide Nanofibres Cr6+ 310.4 3 45 [90]
Cu2+ 423.8 6
Pb2+ 461.3 6
7 Chitosan/Fe3O4 Nanoparticles Cu2+ 21.5 2–5 [91]
8 Chitosan/phenylthiourea Beads Hg2+ 135 ± 3 5 30 [92]
Cd2+ 120 ± 1
Zn2+ 52 ± 1
9 Chitosan/clinoptilolite Beads Cu2+ 11.32 5 25 [93]
Co2+ 7.94
Ni2+ 4.209
10 Chitosan/(polyvinyl alcohol)/zeolite Nanofibres Cr6+ 0.17 [94]
Fe3+ 0.11
Ni2+ 0.03
11 Chitosan/thioglyceraldehyde Beads Hg2+ 85.33 5 [95]
Cu2+ 54.76
Zn2+ 35.12
12 Magnetic chitosan/glutaraldehyde Microsphere Zn2+ 32.16 5 [96]
13 Chitosan/poly(allylamine hydrochloride)–modified E. coli Fibres Pt4+ 290.98 2 40 [97]
14 Chitosan/glutaraldehyde Beads Co2+ 73.98 5 25 [98]
Cd2+ 68.52
Pb2+ 78.67
(Continued)
No. Chitosan‐based sorbent Form Heavy Metal Qmax (mg·g−1, mmol·g−1) pH T (°C) Reference
15 Chitosan/montmorillonite Particles Pb2+
49.33 5 25–50 [99]
Cu2+ 25.28
Cd2+ 20.61
16 Chitosan/Fe3O4/thiourea Microspheres Hg2+ 625.2 28 [100]
Cu2+ 66.7
Ni2+ 15.3
17 Chitosan/glutaraldehyde Membrane Hg2+ 891.4 6 25 [101]
Spheres 647.9 ± 6
18 Chitosan/methyl iodide/EDTA dianhydride Powder Cu2+
0.698 4.5 25 [102]
Co2+ 1.125 4.5
Ni2+ 0.725 7.5
Cr6+ 1.910 2
19 Chitosan‐g‐Polyaniline Powder Pb2+ 16.07 6 50 [103]
Cd2+ 14.33
20 Chitosan/fly ash Particles Cd2+ 87.72 8 35 [104]
time, chemical precipitation was done using inorganic coagulants such as lime alone or in
mixture with metal salts, magnesium chloride or calcium chloride. Later, other chemicals
were studied to be used as coagulants, namely ferric chloride, aluminium sulphate and cati-
onic polymers (e.g. polydiallyldimethyl ammonium chloride) [82–84]. Because of its very
large surface‐to‐volume ratio, a colloidal solution does not agglomerate naturally. Thus, the
small particles in such a stable solution, due to various factors (e.g. adsorption of ions onto
the particle surface, imperfection in crystal structure, ionisation of surface sites), can gain
net charges in aqueous solutions. This effect depends on the pH of the solution (at
pH>pHiso, the particles in solution are negatively charged; at pH<pHiso, the particles in
water are positively charged). The charge neutralisation, adsorption and precipitation are
often exploited to destabilise the colloidal particles.
Thus, over time, researchers found that coagulation using such organic/inorganic coagu-
lants may follow different mechanisms (Table 17.3). At low coagulant dosage and low pH
value, the coagulant neutralises the electrical charge of the suspended particles in wastes
3 Sweep coagulation
causing the clumping effect, which is a predominant phenomenon. On the other hand, at high
coagulant dosage and pH value, the particles are just adsorbed on the coagulant surface.
In general, the most used coagulants are of inorganic nature (Table 17.4), acting as
charge neutralisers. Being added to water, this type of metal‐based coagulant dissociates
and releases metal ions in the solution. The released metal ions react with hydroxyl ions
(OH−), resulting in a hydrolysed species [106]. The formed hydroxides produce polymer‐
like chains allowing microfloc formation and growth to larger flocs.
One may observe that the most commonly used inorganic coagulants are based on alu-
minium and iron(III) salts. Nevertheless, these chemicals show some disadvantages in their
usage because they are effective only in a certain pH range. As a consequence, a good floc-
culation may not be achieved in some wastewaters, yielding an effluent with poor quality.
Despite these disadvantages, these kinds of coagulants can be found as residuals in treated
waters that are returned to nature. As they are not biodegradable, these residuals can cause
illness when the water is intended for human consumption. For example, Alzheimer’s dis-
ease is linked to aluminium accumulated from water sludge that causes problems of dis-
posal and treatment [107].
Consequently, researchers had to look for new solutions. In this context, polymers,
especially derived from certain kinds of plants and animal life, were found to be work-
able alternatives to inorganic coagulants. The natural coagulants were proven to be non‐
toxic flocculants in treatment of organic polluted wastewaters [110] and as a chelator of
toxic (heavy and radioactive) metals [111]. Due to their properties (availability, biocom-
patibility, biodegradability, polyelectrolisity, non‐toxicity and flocculating ability), pol-
ysaccharides can be used in multiple and various fields. For a few decades, these natural
additives attracted the attention of many researchers dealing with environmental protec-
tion. Hence, natural additives were employed as a flocculant/coagulant for wastewater
treatment [112].
The most challenging natural coagulants, over other polysaccharides, are chitin/chitosan
and their derivatives [113]. They have a chemical structure that contain very reactive amino
(–NH2) and hydroxyl (–OH) groups, which recommend them to be used as adsorbent mate-
rial. Also, their chemical structures allow designing chitin/chitosan macromolecules for a
particular application. Owing to the presence of reactive groups, chitin/chitosan is able to
form composites with various compounds (zeolites, magnetite, polyurethane, etc.) [114].
Recently, Dhananasekaran, Palanivel and Pappu [115] have found that α‐chitin
Chitin and Chitosan for Water Purification 445
(a) (b)
Feed
Driving
force
Retentate Feed Retentate
(ΔP)
Membrane Membrane
Flux Flux
Permeate Permeate
Figure 17.4 Principle sketch of the pressure‐driven membrane processes: (a) dead‐end (in‐line) filtration
mode; (b) cross‐flow filtration mode.
Chitin and Chitosan for Water Purification 447
undergoes ultrafiltration. Components of the bulk solution that are retained by the mem-
brane represent the retentate (concentrated solution). As the driving force acts, a higher‐
quality flux crosses through the membrane from the feed side to the permeate side. The
driving force of the filtration process is the transmembrane pressure (ΔP) [128, 129],
which can be calculated by Eq. 17.2 and Eq. 17.3 for dead‐end and cross‐flow filtration,
respectively.
Dead‐end filtration:
Cross‐flow filtration:
Note that the pressure on permeate side (Ppermeate) is always much smaller than the pres-
sures on feed (Pfeed) and retentate (Pretentate) sides, that is, Pfeed>>Ppermeate.
The liquid going through the membrane with a certain velocity is designated as the per-
meate flux, and it is often reported in the units of L/(m2⋅h). Hence, the permeate flux is
calculated with respect to the membrane surface area, as given by [125, 127]:
V P
J Eq. 17.4
A t RM RCP RF
Here, A denotes the membrane area, and ΔV is the filtrate volume produced within time
interval Δt. On the other hand, the permeate flux is directly proportional to the transmem-
brane pressure (ΔP), and inversely proportional with the filtrate viscosity (μ) and total flow
resistance. The latter term involves three components of resistances against the solvent
transport, such as the resistance to flow provided by the membrane (RM); the resistance
caused by concentration polarization effect (RCP); and the resistance caused by membrane
fouling (RF) due to cake formation and pore blocking. Both phenomena, concentration
polarization (reversible) and fouling (irreversible), help achieve flux decline, thereby
reducing the filtration performance. Likewise, the quality of the permeate solution can be
evaluated by determining the rejection efficiency (Y, %), which can be expressed as:
Cp
Y 1 100 Eq. 17.5
Cb
where Cb is the solute concentration in the bulk solution from the feed side, and Cp is the
solute concentration in the permeate solution.
The greater the values of both responses (i.e., rejection efficiency, Y, and permeate flux,
J), the better is the performance of the UF process. Therefore, it is important to design
membranes with good selectivity and improved hydrophilicity. The latter characteristic
(hydrophilicity) helps in diminishing the concentration polarisation and fouling effects.
448 Chitin and Chitosan: Properties and Applications
O
OH O O OH
DMF / H2O
O O O O
+
HO 130 °C/N2 HO
N
NH2 O O
n n
Figure 17.5 Schematic representation of chitosan modification with phthalic anhydride; adapted from
[132–134].
Ghiggi and collaborators demonstrated that the blend membranes (made of PES and N‐
phthaloyl‐chitosan) could significantly diminish the concentration polarisation effect and
fouling resistance, indicating their antifouling properties [132].
Other authors [135–137] reported the chemical modification of chitosan with succinic
anhydride, in mild thermal conditions (35–40°C), yielding N‐succinyl chitosan (NSCS)
derivative. For instance, Kumar, Isloor and Ismail [136] produced blend membranes made
of polysulphone/N‐succinyl chitosan (PSf/NSCS) that were applied for the UF of proteins.
Owing to the low solubility of NSCS in organic solvents, these authors proposed a new
method for blending of NSCS with PSf. According to that method, the maximum blending
of NSCS with PSf was achieved for the ratio of 80:20 (PSf:NSCS). The produced blend
membrane PSf/NSCS (80:20) showed a low contact angle equal to 60.9°, confirming the
improvement of membrane hydrophilicity. All PSf/NSCS membranes disclosed higher per-
meate fluxes during protein UF than the pristine PSf membrane. Hence, by blending NSCS
with PSf, both permeation flux and antifouling properties of membranes were enhanced
significantly. Finally, the authors concluded that N‐succinyl chitosan derivative (NSCS)
acted as a promising substitute for the hydrophilic polymeric additives like PVP, PEG or
polyvinyl alcohol (PVA).
The same authors [137] reported in another paper the synthesis of a different chitosan
derivative as an additive for polysulphone membrane. The water‐soluble chitosan deriva-
tive was designated as N‐propylphosphonic chitosan (NPPCS) having a terminal phospho-
nic acid group, and this derivative was synthesised by reacting chitosan with
hydroxybenzotriazole (HOBt) and propylphosphonic anhydride via a one‐pot reaction. The
produced blend membranes (polysulphone/N‐propylphosphonic chitosan) disclosed
improved antifouling properties when compared to pristine polysulphone membranes.
In their seminal paper, Zhang et al. [138] prepared a chitosan nanofiltration membrane
on polyacrylonitrile (PAN) UF membrane which was used as support. To improve the acid
resistance of the composite membrane, chitosan was cross‐linked by glutaraldehyde
through hydroxyl groups by protecting amine groups using copper ion chelation. The
cross‐linked chitosan/PAN membrane disclosed good rejection efficiency (95%) of γ‐
aminobutyric acid from aqueous solutions at pH 4.69.
A previous study reported by Xue et al. [139] dealt with chitosan‐functionalised gra-
phene oxide applied to improve the permeability and antifouling properties of UF mem-
branes. In their approach, the chitosan‐functionalised graphene oxide (CS‐GO) was
450 Chitin and Chitosan: Properties and Applications
synthesised and incorporated into a polyvinylidene fluoride (PVDF) matrix. The produced
CS‐GO/PVDF blend membranes showed higher permeate flux and rejection efficiency,
when compared to PVDF, CS/PVDF, and GO/PVDF membranes. The optimal content of
CS‐GO into the PVDF matrix was found to be 0.6% wt%. This composite membrane
showed a reduced WCA (64.2°) and an improved antifouling property. Finally, these
authors concluded that the CS‐GO/PVDF composite membrane could be of great potential
for water treatment applications.
In another study, Hamzah et al. [140] designed chitosan/polysulphone (CS/PSf) self‐
assembly membranes to mitigate fouling and enhance the performance of the UF process.
To this end, the native PSf membranes were immersed into the chitosan solution (0.1%
wt%, in acetic acid pH 5) for 30–120 min to deposit chitosan macromolecules onto the PSf
membrane surface. This study highlighted that the integration of hydrophobic polysul-
phone and hydrophilic chitosan successfully upgraded the membrane UF performance by
minimising the fouling phenomena.
Preliminary work on the removal of chromium from wastewater by a cellulose acetate/
chitosan UF membrane was undertaken by Vinodhini and Sudha [141]. In their study, nan-
oparticles of chitosan were first prepared by sol‐gel method using sodium tripolyphosphate
(TPP) as a cross‐linker. Next, the UF membrane was prepared by blending cellulose acetate
(CA) with nanoparticles of chitosan (NCS) and PEG at 1:2:2 ratios. DMF was used as an
aprotic solvent to prepare casting solutions. These authors inferred that the produced blend
membrane CA/NCS/PEG could remove chromium from tannery effluents in an effective
way by the UF process.
(a) (b)
Feed Feed
Solvent (water)
Solute (pollutant)
Binding polymer
Driving
force
(∆P)
Membrane
Permeate Permeate
Figure 17.6 Schematic representation of ultrafiltration process: (a) ultrafiltration in the absence of a binding
polymer; (b) polymer‐enhanced ultrafiltration (PEUF).
17.5 Outlook
Chitin and chitosan are known for their abundance, biodegradability, non‐toxicity, biocom-
patibility, renewable nature, low cost, versatility and ease of chemical modification. Based
on their properties, these polysaccharides are strong candidates for applications in the field
of wastewater treatment. This review was focused on the most relevant methods for water
purification involving chitin/chitosan materials, such as: adsorption, coagulation/floccula-
tion and UF purification processes. In spite of some rather satisfactory performances, the
solubility problems of chitin do limit the utilisation of this biopolymer in the development
of new materials dedicated to water purification treatments. In contrast, chitosan is easily
modified by cross‐linking, grafting, chemical functionalisation and/or forming composites,
thereby extending the range of applications or clearly improving its performance.
The use of chitin/chitosan (pristine and modified forms) for organic pollutant adsorption
is widely reported in literature. The majority of research works suggested that the adsorp-
tion occurred mainly in acidic medium and was realised via an ion exchange mechanism
based on electrostatic attraction between the protonated form of the biopolymer and organic
pollutant molecules frequently in their negatively charged form. Future work should be
dedicated to the application of chitin/chitosan‐based materials as adsorbents in real waste-
waters in order to determine their performances under real environmental conditions.
The performance of chitin/chitosan, both pristine and modified forms, for heavy metal
ion removal from wastewaters is reported by many authors. Nevertheless, the mechanism
governing metal cation adsorption onto adsorbent surface still needs to be detailed for each
particular application.
Many types of wastewaters can also be treated with chitin/chitosan‐based materials in order
to reduce the total solids, suspended solids and turbidity. In these lines, chitin/chitosan‐based
materials may act as coagulants/flocculants in the separation process. Such a process occurs
by a charge neutralisation mechanism or by the mechanism of electrostatic charges, depend-
ing on the coagulant concentration. Biomaterials (chitin/chitosan) showed improved coagu-
lant properties when compared to inorganic coagulants, even if they were used individually or
along with other organic/inorganic materials to treat wastewaters of different origins.
The literature survey has indicated that chitosan‐based materials are also of interest in
the field of membrane separations by UF. In this regard, chitosan derivatives were success-
fully employed to produce blend membranes with improved hydrophilic properties that can
diminish concentration polarisation and fouling phenomena.
The binding capacity of chitosan for metal cations, oxyanions and water‐soluble dyes
proves that this biopolymer is a good candidate substitute for conventional binding polymers
used in complexation–UF processes. Other advantages that make chitosan attractive for the
membrane separation processes are related to its non‐toxicity, recyclability and low cost.
Hence, it turns out that chitosan and its derivatives are prospective materials for environ-
mental applications related to water purification.
Acknowledgement
The authors Petrisor Samoila, Corneliu Cojocaru and Maria Ignat are grateful for the finan-
cial support of the Romanian Ministry of Research and Innovation, CNCS – UEFISCDI,
project number PN‐III‐P1‐1.1‐TE‐2016‐0805, within PNCDI III. Also, the authors Andra
Cristina Humelnicu and Valeria Harabagiu are grateful for the financial support of the
Chitin and Chitosan for Water Purification 453
References
1. Crini, G., Lichtfouse, E., Wilson, L.D., and Morin‐Crini, N. (2019). Conventional and non‐conven-
tional adsorbents for wastewater treatment. Environmental Chemistry Letters, 17(1):195–213.
2. Teh, C.Y., Budiman, P.M., Shak, K.P.Y., and Wu, T.Y. (2016). Recent advancement of coagulation–
flocculation and its application in wastewater treatment. Industrial & Engineering Chemistry Research,
55(16):4363–4389.
3. Geise, G.M., Lee, H.S., Miller, D.J., Freeman, B.D., McGrath, J.E., and Paul, D.R. (2010). Water
purification by membranes: The role of polymer science. Journal of Polymer Science: Part B: Polymer
Physics, 48:1685–1718.
4. Crini, G. (2005). Recent developments in polysaccharide‐based materials used as adsorbents in waste-
water treatment. Progress in Polymer Science, 30(1):38–70.
5. Figueiredo, S.A., Loureiro, J.M., and Boaventura, R.A. (2005). Natural waste materials containing
chitin as adsorbents for textile dyestuffs: Batch and continuous studies. Water Research,
39(17):4142–4152.
6. Zhu, H.Y., Jiang, R., Xiao, L., and Li, W. (2010). A novel magnetically separable γ‐Fe2O3/crosslinked
chitosan adsorbent: Preparation, characterization and adsorption application for removal of hazardous
azo dye. Journal of Hazardous Materials, 179(1–3):251–257.
7. Renault, F., Morin‐Crini, N., Gimbert, F., Badot, P.M., and Crini, G. (2008). Cationized starch‐based
material as a new ion‐exchanger adsorbent for the removal of C.I. Acid Blue 25 from aqueous solu-
tions. Bioresource Technology, 99(16):7573–7586.
8. Liu, M., Deng, Y., Zhan, H., and Zhang, X. (2002). Adsorption and desorption of copper (II) from solu-
tions on new spherical cellulose adsorbent. Journal of Applied Polymer Science, 84:478–485.
9. Thakur, V.K., Thakur, M.K. and Gupta, R.K. (2014). Graft copolymers of natural fibers for green
composites. Carbohydrate Polymers, 104:87–93.
10. Tran, V.S., Ngo, H.H., Guo, W., Zhang, J., Liang, S., Ton‐That, C., and Zhang, X. (2015). Typical low
cost biosorbents for adsorptive removal of specific organic pollutants from water. Bioresource
Technology, 182:353–363.
11. Abdel‐Halim, E.S. and Al‐Deyab, S.S. (2011). Removal of heavy metals from their aqueous solutions
through adsorption onto natural polymers. Carbohydrate Polymers, 84(1):454–458.
12. Rinaudo, M. (2006). Chitin and chitosan: Properties and applications. Progress in Polymer Science,
31(7):603–632.
13. Ifuku, S. (2014). Chitin and chitosan nanofibers: Preparation and chemical modifications. Molecules,
19:18367–18380.
14. Wan Ngah, W.S., Teong, L.C., and Hanafiah, M.A.K.M. (2011). Adsorption of dyes and heavy metal
ions by chitosan composites: A review. Carbohydrate Polymers, 83(4):1446–1456.
15. Wan Ngah, W.S., Endud, C.S., and Mayanar, R. (2002). Removal of copper(II) ions from aqueous solu-
tion onto chitosan and cross‐linked chitosan beads. Reactive and Functional Polymers,
50(2):181–190.
16. Liu, Y., Chen, W., and Kim, H.I. (2012). Removal of lead and nickel ions from wastewater by genipin
crosslinked chitosan/poly(ethylene glycol) films. Journal of Macromolecular Science, Part A: Pure
and Applied Chemistry, 49(3):242–250.
17. Monier, M., Ayad, D.M., and Abdel‐Latif, D.A. (2012). Adsorption of Cu(II), Cd(II) and Ni(II) ions by
cross‐linked magnetic chitosan‐2‐aminopyridine glyoxal Schiff’s base. Colloids and Surfaces B:
Biointerfaces, 94(250–258).
18. Chen, A.H., Liu, S.C., Chen, C.Y., and Chen, C.Y. (2008). Comparative adsorption of Cu(II), Zn(II),
and Pb(II) ions in aqueous solution on the crosslinked chitosan with epichlorohydrin. Journal of
Hazardous Materials, 54(1–3):184–191.
454 Chitin and Chitosan: Properties and Applications
19. Wan Ngah, W.S., Ab Ghani, S., and Kamari, A. (2005). Adsorption behaviour of Fe(II) and Fe(III) ions
in aqueous solution on chitosan and cross‐linked chitosan beads. Bioresource Technology,
96(4):443–450.
20. Ignat, M., Samoila, P., Cojocaru, C., Sacarescu, L., and Harabagiu, V. (2016). Novel synthesis route for
chitosan‐coated zinc ferrite nanoparticles as potential sorbents for wastewater treatment. Chemical
Engineering Communications, 203(12):1591–1599.
21. Nechita P. (2017). Applications of chitosan in wastewater treatment, in Biological Activities and
Application of Marine Polysaccharides (ed. E.A. Shalaby) IntechOpen. DOI: 10.5772/65289.
22. Pérez‐Calderón, J., Santos, M.V., and Zaritzky, N. (2018). Optimal clarification of emulsified oily
wastewater using a surfactant/chitosan biopolymer. Journal of Environmental Chemical Engineering,
6(4):3808–3818.
23. Anastopoulos, I., Bhatnagar, A., Bikiaris, D.N., and Kyzas, G.Z. (2017). Chitin adsorbents for toxic
metals: A review. International Journal of Molecular Sciences, 18(1):114.
24. Ali, I. and Gupta, V.K. (2006). Advances in water treatment by adsorption technology. Nature
Protocols, 1:2661–2667.
25. Crini, G. (2006). Non‐conventional low‐cost adsorbents for dye removal: A review. Bioresource
Technology, 97(9):1061–1085.
26. Faust, S.D. and Aly, O.M. (1987). Kinetics of adsorption, in Adsorption Processes for Water Treatment,
Butterworth‐Heinemann.
27. Bonilla‐Petriciolet, A., Mendoza‐Castillo, D.I., and Reynel‐Avila, H.E. (2017). Adsorption Processes
for Water Treatment and Purification, Springer, Netherlands.
28. Dubinin, M.M., Erashko, I.T., Kadlec, O., Ulin, V.I., Voloshchuk, A.M., and Zolotarev, P.P. (1975).
Kinetics of physical adsorption by carbonaceous adsorbents of biporous structure. Carbon,
13(3):193–200.
29. Chiarle, S., Ratto, M., and Rovatti, M. (2000). Mercury removal from water by ion exchange resins
adsorption. Water Research, 34(11):2971–2978.
30. Webb, P.A. (2003). Introduction to Chemical Adsorption Analytical Techniques and Their Applications
to Catalysis. MIC Technical Publications.
31. Faust, S.D. and Aly, O.M. (1987). Adsorption models, in Adsorption Processes for Water Treatment,
Butterworth‐Heinemann.
32. Samoila, P., Cojocaru, C., Cretescu, I., Stan, C.D., Nica, V., Sacarescu, L., and Harabagiu, V. (2015).
Nanosized spinel ferrites synthesized by sol‐gel autocombustion for optimized removal of azo dye
from aqueous solution. Journal of Nanomaterials. 713802, http://dx.doi.org/10.1155/2015/713802.
33. Humelnicu, A.C., Cojocaru, C., Pascariu Dorneanu, P., Samoila, P., and Harabagiu, V. (2017). Novel
chitosan‐functionalized samarium‐doped cobalt ferrite for adsorptive removal of anionic dye from
aqueous solutions. Comptes Rendus Chimie, 20:1026–1036.
34. Kinniburgh, D.G. (1986). General purpose adsorption isotherms. Environmental Science & Technology,
20(9):895–904.
35. Cojocaru, C., Humelnicu, A.C., Samoila, P., Pascariu, P., and Harabagiu, V. (2018). Optimized formu-
lation of NiFe2O4@Ca‐alginate composite as a selective and magnetic adsorbent for cationic dyes:
Experimental and modeling study. Reactive and Functional Polymers, 125:57–69.
36. Luo, Z., Gao, M., Yang, S., and Yang, Q. (2015). Adsorption of phenols on reduced‐charge montmoril-
lonites modified by bispyridinium dibromides: Mechanism, kinetics and thermodynamics studies.
Colloids and Surfaces A: Physicochemical and Engineering Aspects, 482:222–230.
37. Pigatto, G., Lodi, A., Finocchio, E., Palm, M.S.A., and Converti, A., (2013). Chitin as biosorbent for
phenol removal from aqueous solution: Equilibrium, kinetic and thermodynamic studies. Chemical
Engineering and Processing, 70:131–139.
38. Yoshizuka, K., Lou, Z., and Inoue, K. (2000). Silver‐complexed chitosan microparticles for pesticide
removal. Reactive and Functional Polymers, 44(1):47–54.
Chitin and Chitosan for Water Purification 455
39. Celis, R., Adelino, M.A., Hermosín, M.C., and Cornejo, J. (2012) Montmorillonite–chitosan bionano-
composites as adsorbents of the herbicide clopyralid in aqueous solution and soil/water suspensions.
Journal of Hazardous Materials, 209–210:67–76.
40. Rêgo, T.V., Cadaval, T.R. Jr, Dotto, G.L., and Pinto, L.A. (2013). Statistical optimization, interaction
analysis and desorption studies for the azo dyes adsorption onto chitosan films. Journal of Colloid and
Interface Science, 411:27–33.
41. Dursun, A.Y. and Seda Kalayci, C.¸ (2005). Equilibrium, kinetic and thermodynamic studies on the
adsorption of phenol onto chitin. Journal of Hazardous Materials, B123:151–157.
42. Soni, U., Bajpai, J., Singh, S.K., and Bajpai, A.K. (2017). Evaluation of chitosan‐carbon based bio-
composite for efficient removal of phenols from aqueous solutions. Journal of Water Process
Engineering, 16:56–63.
43. Zhou, L.C., Meng, X.G., Fu, J.W., Yang, Y.C., Yang, P., and Mi, C. (2014). Highly efficient adsorption
of chlorophenols onto chemically modified chitosan. Applied Surface Science, 292:735–741.
44. Shankar, A., Kongot, M., Kumar Saini, V., and Kumar A. (2018). Removal of pentachlorophenol pes-
ticide from aqueous solutions using modified chitosan. Arabian Journal of Chemistry, https://doi.
org/10.1016/j.arabjc.2018.01.016.
45. El Harmoudi, H., El Gaini, L., Daoudi, E., Rhazi, M., Boughaleb, Y., El Mhammedi, M.A., Migalska‐
Zalas, A., and Bakasse, M. (2014). Removal of 2,4‐D from aqueous solutions by adsorption processes
using two biopolymers: Chitin and chitosan and their optical properties. Optical Materials,
36:1471–1477.
46. Ding, C., Gong, D., Yu, P., Shao, J., and Zhong, M.E. (2016). Removal of quinclorac herbicide from
aqueous solution by chitosan/montmorillonite bionanocomposite. Desalination and Water Treatment,
57(52):24970–24981.
47. Tan, K.B., Vakili, M., Horri, B.A., Poh, P.E., Abdullah, A.Z., and Salamatinia, B. (2015). Adsorption
of dyes by nanomaterials: Recent developments and adsorption mechanisms. Separation and
Purification Technology, 150:229–242.
48. Kamal, T., Anwar, Y., Khan, S.B., Chani, M.T.S., and Asiri, A.M. (2016). Dye adsorption and bacteri-
cidal properties of TiO2/chitosan coating layer. Carbohydrate Polymers, 148:153–160.
49. Peng, Q., Liu, M., Zheng, J., and Zhou, C. (2015). Adsorption of dyes in aqueous solutions by chi-
tosan–halloysite nanotubes composite hydrogel beads. Microporous and Mesoporous Materials,
201:190–201.
50. Bhavani, K., Roshan Ara Begum, E., Selvakumar, S., and Shenbagarathai, R. (2016). Chitosan a low
cost adsorbent for electroplating waste water treatment. Journal of Bioremediation & Biodegradation,
7:346.
51. Gibbs, G., Tobin, J.M., and Guibal, E. (2003). Sorption of Acid Green 25 on chitosan: Influence of
experimental parameters on uptake kinetics and sorption isotherms. Journal of Applied Polymer
Science, 90(4):1073–1080.
52. Dotto, G.L. and Pinto, L.A.A. (2011). Adsorption of food dyes onto chitosan: Optimization process
and kinetic. Carbohydrate Polymers, 84(1):231–238.
53. Wong, Y.C., Szeto, Y.S., Cheung, W.H., and McKay, G. (2003). Equilibrium studies for acid dye
adsorption onto chitosan. Langmuir, 19(19), 7888–7894.
54. Zhou, L., Jin, J., Liu, Z., Liang, X., and Shang, C. (2011). Adsorption of acid dyes from aqueous solu-
tions by the ethylenediamine‐modified magnetic chitosan nanoparticles. Journal of Hazardous
Materials, 185(2–3):1045–1052.
55. Sheshmani, S., Ashori, A., and Hasanzadeh, S. (2014). Removal of Acid Orange 7 from aqueous solu-
tion using magnetic graphene/chitosan: A promising nano‐adsorbent. International Journal of
Biological Macromolecules, 68:218–224.
56. Hu, Z.G., Zhang, J., Chan, W.L., and Szeto, Y.S. (2006). The sorption of acid dye onto chitosan nano-
particles. Polymer, 47(16):5838–5842.
456 Chitin and Chitosan: Properties and Applications
57. Crini, G., Gimbert, F., Robert, C., Martel, B., Adama, O., Morin‐Crini, N., De Giorgi, F., and Badot,
P.M. (2008). The removal of Basic Blue 3 from aqueous solutions by chitosan‐based adsorbent: Batch
studies. Journal of Hazardous Materials, 153:96–106.
58. Yuan, B., Qiu, L.G., Su, H.Z., Cao, C.L., and Jiang, J.H. (2016). Schiff base – Chitosan grafted l‐
monoguluronic acid as a novel solid‐phase adsorbent for removal of congo red. International Journal
of Biological Macromolecules, 82:355–360.
59. Wang, L., Zhang, J., and Wang, A. (2011). Fast removal of methylene blue from aqueous solution by
adsorption onto chitosan‐g‐poly (acrylic acid)/attapulgite composite. Desalination, 266(1–3):33–39.
60. Tanhaei, B., Ayati, A., Lahtinen, M., and Sillanpää, M. (2015). Preparation and characterization of a
novel chitosan/Al2O3/magnetite nanoparticles composite adsorbent for kinetic, thermodynamic and
isotherm studies of Methyl Orange adsorption. Chemical Engineering Journal, 259:1–10.
61. Chiou, M.S. and Chuang, G.S. (2006). Competitive adsorption of dye metanil yellow and RB15 in acid
solutions on chemically cross‐linked chitosan beads. Chemosphere, 62(5):731–740.
62. Ngwabebhoh, F. A., Erdem, A., and Yildiz, U., (2016) Synergistic removal of Cu(II) and nitrazine yel-
low dye using an eco‐friendly chitosan‐montmorillonite hydrogel: Optimization by response surface
methodology. Journal of Applied Polymer Science, 133(29):43664.
63. Chen, A.H. and Huang, Y.Y. (2010). Adsorption of Remazol Black 5 from aqueous solution by the
templated crosslinked‐chitosans. Journal of Hazardous Materials, 177(1–3):668–675.
64. Singh, V., Sharma, A.K., Tripathi, D.N., and Sanghi, R. (2009). Poly(methylmethacrylate) grafted chi-
tosan: An efficient adsorbent for anionic azo dyes. Journal of Hazardous Materials, 1612–3, 2009,
955–966.
65. Singh, V., Sharma, A.K., and Sanghi, R. (2009). Poly(acrylamide) functionalized chitosan: An efficient
adsorbent for azo dyes from aqueous solutions. Journal of Hazardous Materials, 166(1):327–335.
66. Kyzas, G.Z. and Lazaridis, N.K. (2009). Reactive and basic dyes removal by sorption onto chitosan
derivatives. Journal of Colloid and Interface Science, 331(1):32–39.
67. Debrassi, A., Baccarin, T., Demarchi, C.A., Nedelko, N., Ślawska‐Waniewska, A., Dłużewski, P.,
Bilska, M., and Rodrigues, C.A. (2012). Adsorption of Remazol Red 198 onto magnetic N‐lauryl
chitosan particles: Equilibrium, kinetics, reuse and factorial design. Environmental Science and
Pollution Research, 19:1594.
68. Chiou, M.‐S. and Li, H.‐Y. (2002). Equilibrium and kinetic modeling of adsorption of reactive dye on
cross‐linked chitosan beads. Journal of Hazardous Materials, 93(2):233–248.
69. Chiou, M.S. and Li, H.Y. (2003). Adsorption behavior of reactive dye in aqueous solution on chemical
cross‐linked chitosan beads. Chemosphere, 50(8):1095–1105.
70. Konaganti, V.K., Kota, R., Patil, S., and Madras, G. (2010). Adsorption of anionic dyes on chitosan
grafted poly(alkyl methacrylate)s. Chemical Engineering Journal, 158(3):393–401.
71. Forutan, R., Ehsandoost, E., Hadipour, S., Mobaraki, Z., Saleki, M., and Mohebbi, G. (2016). Kinetic
and equilibrium studies on the adsorption of lead by the chitin of pink shrimp (Solenocera melantho).
Entomology and Applied Science Letters, 3:20–26.
72. Karthik, R. and Meenakshi, S. (2015) Chemical modification of chitin with polypyrrole for the uptake
of Pb(II) and Cd(II) ions. International Journal of Biological Macromolecules, 78:157–164.
73. Singh, P. and Nagendran, R.A. (2016). Comparative study of sorption of chromium(III) onto chitin and
chitosan. Applied Water Science, 6:199–204.
74. Ariff, M.N.F., Hanafiah, M.A.K.M., and Ngah, W.S.W. (2017). Adsorption of Cu(II) onto cross‐linked
chitosan coated bentonite beads: Kinetic and isotherm studies. Key Engineering Materials,
753:243–248.
75. Šćiban, M., Klašnja, M., Antov, M., and Škrbic ́, B. (2009). Removal of water turbidity by natural
coagulants obtained from chestnut and acorn. Bioresource Technology, 100:6639–6643.
76. Alwi, H., Idris, J., Musa, M., and Hamid, K.H.K. (2013). A preliminary study of banana stem juice
as a plant‐based coagulant for treatment of spent coolant wastewater. Journal of Chemistry,
165057:7.
Chitin and Chitosan for Water Purification 457
77. Choy, S.Y., Prasad, K.M.N., Wu, T.Y., Raghunandan, M.E., and Ramanan, R.N. (2014). Utilization of
plant‐based natural coagulants as future alternatives towards sustainable water clarification. Journal of
Environmental Science, 26:2178–2189.
78. Sancha, A.M. (2006). Review of coagulation technology for removal of arsenic: Case of Chile. Journal
of Health, Population and Nutrition, 24(3):267–272.
79. Camacho, F.P., Bongiovani, M.C., Arakawa, F.S., Shimabuku, Q.L., Shimabuku, Q.L., Vieira, A.M.S.,
and Bergamasco, R. (2013). Advanced processes of cyanobacteria and cyanotoxins removal in supply
water treatment. Chemical Engineering Transactions, 32:421–426.
80. Loloei, M., Alidadi, H., Nekonam, G., and Kor, Y. (2014). Study of the coagulation process in waste-
water treatment of dairy industries. International Journal of Environmental Health Engineering,
3(1):17–21.
81. Reynolds, L.B. (1933). History of chemical precipitation. Sewage Works Journal, 5(4):595–599.
82. Cheng, R., Krasner, S.W., Green, J.F., and Wattier, K.L. (1995). Enhanced coagulation: A preliminary
evaluation. Journal of American Water Works Association, 87(2):91–97.
83. Gregory, J. and Guibai, L. (1991). Effects of dosing and mixing conditions on polymer flocculation of
concentrated suspensions. Chemical Engineering Communication, 108:3–21.
84. Gebbie, P. (2006). An operator’s guide to water treatment coagulants. 31st Annual Qld Water Industry
Workshop – Operations Skills, University Central Queensland – Rockhampton, 4:14–20.
85. Paulino, A.T., Guilherme, M.R., Elias, A.V.R., Tambourgi, B., Nozakia, J., and Muniza, E.C. (2007).
Capacity of adsorption of Pb2+ and Ni2+ from aqueous solutions by chitosan produced from silkworm
chrysalides in different degrees of deacetylation. Journal of Hazardous Materials,
147(1–2):139–147.
86. Haider, S. and Park, S.Y. (2009). Preparation of the electrospun chitosan nanofibers and their applica-
tions to the adsorption of Cu(II) and Pb(II) ions from an aqueous solution. Journal of Membrane
Science, 328(1–2):90–96.
87. Shafaei, A., Ashtiani, F.Z., and Kaghazchi, T. (2007). Equilibrium studies of the sorption of Hg(II) ions
onto chitosan. Chemical Engineering Journal, 133(1–3):311–316.
88. Schmuhl, R., Krieg, H.M., and Keizer, K. (2001). Adsorption of Cu(II) and Cr(VI) ions by chitosan:
Kinetics and equilibrium studies. Water SA, 27(1):1–8.
89. Li, A., Lin, R., Lin, C., He, B., Zheng, T., Lu, L., and Cao, Y. (2016). An environment‐friendly and
multi‐functional absorbent from chitosan for organic pollutants and heavy metal ion. Carbohydrate
Polymers, 148:272–280.
90. Najafabadi, H.H., Irani, M., Rad, L.R., Haratameh, A.H., and Haririan, I. (2015). Removal of Cu2+,
Pb2+ and Cr6+ from aqueous solutions using a chitosan/graphene oxide composite nanofibrous adsor-
bent. RSC Advances, 21:16532–16539.
91. Chang, Y.C. and Chen, D.H. (2005). Preparation and adsorption properties of monodisperse chitosan‐
bound Fe3O4 magnetic nanoparticles for removal of Cu(II) ions. Journal of Colloid and Interface
Science, 283(2):446–451.
92. Monier, M. and Abdel‐Latif, D.A. (2012). Preparation of cross‐linked magnetic chitosan‐phenylthiou-
rea resin for adsorption of Hg(II), Cd(II) and Zn(II) ions from aqueous solutions. Journal of Hazardous
Materials, 209–210:240–249.
93. Dinu, M.V. and Dragan, E.S. (2010). Evaluation of Cu2+, Co2+ and Ni2+ ions removal from aqueous solu-
tion using a novel chitosan/clinoptilolite composite: Kinetics and isotherms. Chemical Engineering
Journal, 160:157–163.
94. Habiba, U., Afifi, A.M., Salleh, A., and Ang, B.C. (2017). Chitosan/(polyvinyl alcohol)/zeolite electro-
spun composite nanofibrous membrane for adsorption of Cr6+, Fe3+ and Ni2+. Journal of Hazardous
Materials, 322(A):182–194.
95. Monier, M. (2012). Adsorption of Hg2+, Cu2+ and Zn2+ ions from aqueous solution using formaldehyde
cross‐linked modified chitosan–thioglyceraldehyde Schiff’s base. International Journal of Biological
Macromolecules, 50(3):773–781.
458 Chitin and Chitosan: Properties and Applications
96. Fan, L., Luo, C., Lv, Z., Lu, F., and Qiu, H. (2011) Preparation of magnetic modified chitosan and
adsorption of Zn2+ from aqueous solutions. Colloids and surfaces B: Biointerfaces 88(2):574–581.
97. Mao, J., Kim, S., Wu, X.H., Kwak, I.S., Zhou, T., and Yun, Y.S. (2015) A sustainable cationic
chitosan/E. coli fiber biosorbent for Pt(IV) removal and recovery in batch and column systems.
Separation and Purification Technology, 143:32–39.
98. Lemrabet, E., Jaafari, K., Benkhouja, K., Touaiher, M., Bakasse, M., Sahraoui, B., Touhtouh, S., and
Soufiáne, A. (2013) Removal of heavy‐metal ion by adsorption on chitosan gel beads. Journal of
Optoelectronics and Advanced Materials, 15(11–12):1298–1302.
99. Hu, C., Zhu, P., Cai, M., Hu, H., and Fu, Q. (2017). Comparative adsorption of Pb(II), Cu(II) and
Cd(II) on chitosan saturated montmorillonite: Kinetic, thermodynamic and equilibrium studies.
Applied Clay Science, 143:320–326.
100. Zhou, L., Wang, Y., Liu, Z., and Huang, Q. (2009). Characteristics of equilibrium, kinetics studies for
adsorption of Hg(II), Cu(II), and Ni(II) ions by thiourea‐modified magnetic chitosan microspheres.
Journal of Hazardous Materials, 161(2–3):995–1002.
101. Vieira, R.S. and Beppu, M.M. (2006). Dynamic and static adsorption and desorption of Hg(II) ions
on chitosan membranes and spheres. Water Research, 40(8):1726–1734.
102. de Almeida, F.T.R., Ferreira, B.C.S., Moreira, A.L.S.L., de Freitas, R.P., Gil, L.F., and Gurgel, L.V.A.
(2016). Application of a new bifunctionalized chitosan derivative with zwitterionic characteristics for
the adsorption of Cu2+, Co2+, Ni2+, and oxyanions of Cr6+ from aqueous solutions: Kinetic and equi-
librium aspects. Journal of Colloid and Interface Science, 466:297–309.
103. Karthik, R. and Meenakshi, S. (2015). Removal of Pb(II) and Cd(II) ions from aqueous solution using
polyaniline grafted chitosan. Chemical Engineering Journal, 263:168–177.
104. Pandey, S. and Tiwari, S. (2015). Facile approach to synthesize chitosan based composite—
Characterization and cadmium(II) ion adsorption studies. Carbohydrate Polymers, 134:646–656.
105. Martínez‐Navarro, F.(2007). Tratamiento de aguas residuales industriales mediante electrocoagu-
lación y coagulación convencional. s.l.: Tesis (Ph.D) Ingeniería Química. Universidad de Castilla La
Mancha.
106. Kushwaha, J.P., Srivastava, V.C., and Mall, I.D. (2010). Treatment of dairy wastewater by inorganic
coagulants: Parametric and disposal studies. Water Research, 44:5867–5874.
107. Sahu, O.P. and Chaudhari, P.K. (2013). Review on chemical treatment of industrial waste water.
Journal of Applied Sciences and Environmental Management, 17:241–257.
108. Flynn, C.M. (1984). Hydrolysis of inorganic iron (III) salts. Chemical Reviews, 84:31–41.
109. Base, C.F. and Mesmer R.E. (1976). The Hydrolysis of Cations. Wiley, New York.
110. Deans, J.R., Heinsohn G.E., and Link B.A. (1995). Removal of organic contaminants from aqueous
media. U.S. Patent No. 5,393,435.
111. Knorr, D. (1984). Use of chitinous polymers in food – a challenge for food research and development.
Food Technology, 38:85–97.
112. Peniston, Q.P. and Johnson, E.L. (1970). Method for treating an aqueous medium with chitosan and
derivatives of chitin to remove an impurity. U.S. patent 3,533, 940.
113. Sandford, P.A. and Hutchings G.P. (1987). Chitosan‐A natural, cationic biopolymer: commercial
applications, in Industrial Polysaccharides: Genetic Engineering, Structure/Property Relations and
Applications (ed. M. Yalpani). Elsevier, Amsterdam.
114. Ganjali, M.R., Rezapour, M., Faridbod, F., and Norouzi, P. (2017). Chitosan composites: Preparation
and applications in removing water pollutants, in Handbook of Composites from Renewable Materials
(eds. V.K. Thakur, M.K. Thakur, and M.R. Kessler). doi:10.1002/9781119441632.ch124
115. Dhananasekaran, S., Palanivel, R., and Pappu, S. (2015). Adsorption of Methylene Blue, Bromophenol
Blue and Coomassie Brilliant Blue by α‐chitin nanoparticles. Journal of Advanced Resources,
7:113–124.
116. Saritha, V., Srinivas, N., and Srikanth Vuppala, N.V. (2017). Analysis and optimization of coagulation
and flocculation process. Applied Water Science, 7(1):451–460.
Chitin and Chitosan for Water Purification 459
117. Dotto, G.L., Rosa, G.S., Moraes, M.A., Weska, R.F., and Pinto, L.A.A., (2013). Treatment of chitin
effluents by coagulation–flocculation with chitin and aluminum sulfate. Journal of Environmental
Chemical Engineering, 1(1–2):50–55.
118. Ahmad, A.L., Sumathi, S., and Hameed, B.H. (2006). Coagulation of residue oil and suspended solid
in palm oil mill effluent by chitosan, alum and PAC. Chemical Engineering Journal, 118:99–105.
119. Frederick, W.P. (2016). Chitosan as a drinking water treatment coagulant. American Journal of Civil
Engineering, 4(5):205–215.
120. Szyguła, A., Guibal, E., Palacín, M.A., Ruiz, M., and Sastre, A.M. (2009). Removal of an anionic
dye (AcidBlue 92) by coagulation–flocculation using chitosan. Journal of Environment Management,
90:2979–2986.
121. Bolto, B.A. (2006). Coagulation and flocculation with organic polyelectrolytes, in Interface Science
in Drinking Water Treatment (eds. G. Newcombe and D. Dixon) Academic Press.
122. Xi, L., Yanhua, X., Wenquan, S., Yongjun, S., and Huaili, Z. (2017). UV‐initiated synthesis of a novel
chitosan‐based flocculant with high flocculation efficiency for algal removal. Science of the Total
Environment, 609:410–418.
123. Strand, S.P., Nordengen, T., and Ostgaard, K. (2002). Efficiency of chitosans applied for flocculation
of different bacteria. Water Research, 36:4745–4752.
124. Bratskaya, S., Avramenko, V., Schwarz, S., and Philippova, I. (2006). Enhanced flocculation of
oil‐in‐water emulsions by hydrophobically modified chitosan derivatives. Colloid Surface A,
275:168–176.
125. Foley, G. (2013) Membrane Filtration: A Problem Solving Approach with Matlab.Cambridge
University Press, Cambridge.
126. Mulder, M. (1996). Basic Principles of Membrane Technology, 2nd ed. Kluwer Academic Publishers,
Springer, Netherlands.
127. Baker, R.W. (2004). Membrane Technology and Applications, 2nd ed. Wiley, Chichester, UK.
128. Yang, J.‐S., Baek, K., and Yang, J.‐W. (2005). Crossflow ultrafiltration of surfactant solutions.
Desalination, 184:385–394.
129. Cojocaru, C., Zakrzewska‐Trznadel, G., and Miskiewicz, A. (2009). Removal of cobalt ions from
aqueous solutions by polymer assisted ultrafiltration using experimental design approach: Part 2:
Optimization of hydrodynamic conditions for a crossflow ultrafiltration module with rotating part,
Journal of Hazardous Materials, 169:610–620.
130. Lalia, B.S., Kochkodan, V., Hashaikeh, R., and Hilal, N. (2013). A review on membrane fabrication:
Structure, properties and performance relationship. Desalination, 326:77–95.
131. Van den Berg, G.B. and Smolders, C.A. (1990). Flux decline in ultrafiltration processes. Desalination,
77:101–133.
132. Ghiggi, F.F., Pollo, L.D., Cardozo, N.S.M., and Tessaro, I.C. (2017). Preparation and characterization
of polyethersulfone/N‐phthaloyl‐chitosan ultrafiltration membrane with antifouling property.
European Polymer Journal, 92:61–70.
133. Padaki, M., Isloor, A.M., and Wanichapichart, P. (2011). Polysulfone/N‐phthaloylchitosan novel
composite membranes for salt rejection application. Desalination, 279:409–414.
134. Kurita, K., Ikeda, H., Yoshida, Y., Shimojoh, M., and Harata, M. (2002). Chemoselective protection
of the amino groups of chitosan by controlled phthaloylation: Facile preparation of a precursor useful
for chemical modifications. Biomacromolecules, 3:1–4.
135. Aiping, Z., Tian, C., Lanhua, Y., Hao, W., and Ping, L., (2006) Synthesis and characterization of N‐
succinyl‐chitosan and its self‐assembly of nanospheres. Carbohydrate Polymers, 66:274–279.
136. Kumar, R., Isloor, A.M., Ismail, A.F., and Matsuura, T. (2013). Performance improvement of polysul-
fone ultrafiltration membrane using N‐succinyl chitosan as additive. Desalination 318:1–8.
137. Kumar, R., Isloor, A.M., Ismail, A.F., and Matsuura, T. (2013). Synthesis and characterization of
novel water soluble derivative of chitosan as an additive for polysulfone ultrafiltration membrane.
Journal of Membrane Science, 440:140–147.
460 Chitin and Chitosan: Properties and Applications
138. Zhang, X., Jin, X., Xu, C., and Shen, X. (2013). Preparation and characterization of glutaraldehyde
crosslinked chitosan nanofiltration membrane. Journal of Applied Polymer Science, 128:3665–3671.
139. Xue, J., Wang, S., Han, X., Wang, Y., Hua, X., and Li, J. (2018). Chitosan‐functionalized graphene
oxide for enhanced permeability and antifouling of ultrafiltration membranes. Chemical Engineering
& Technology, 41:270–277.
140. Hamzah, S., Ali, N., Mohammad, A.W., Ariffin, M.M., and Ali, A., (2012). Design of chitosan/PSf
self‐assembly membrane to mitigate fouling and enhance performance in trypsin separation. Journal
of Chemical Technology & Biotechnology, 87:1157–1166.
141. Vinodhini, P.A. and Sudha, P.N. (2016). Removal of heavy metal chromium from tannery effluent
using ultrafiltration membrane. Textiles and Clothing Sustainability, 2(5):1–15.
142. Zakrzewska‐Trznadel, G. and Harasimowicz, M. (2002). Removal of radionuclides by membrane
permeation combined with complexation. Desalination, 144:207–212.
143. Cojocaru, C. and Zakrzewska‐Trznadel, G. (2007). Response surface modeling and optimization of
copper removal from aqua solutions using polymer assisted ultrafiltration. Journal of Membrane
Science, 298:56–70.
144. Cojocaru, C., Zakrzewska‐Trznadel, G., and Jaworska, A. (2009). Removal of cobalt ions from aque-
ous solutions by polymer assisted ultrafiltration using experimental design approach. part 1:
Optimization of complexation conditions. Journal of Hazardous Materials, 169:599–609.
145. Dasgupta, J., Sikder, J., Mandal, T., and Adhikari, U. (2015). Reactive red 120 retention through
ultrafiltration enhanced by synthetic and natural polyelectrolytes. Journal of Hazardous Materials,
299:192–205.
146. Dasgupta, J., Sikder, J., and Mandal, D. (2017). Modeling and optimization of polymer enhanced
ultrafiltration using hybrid neural‐genetic algorithm based evolutionary approach. Applied Soft
Computing, 55:108–126.
147. Baharuddin, N.H., Sulaiman, N.M.N., and Aroua, M.K. (2015). Removal of heavy metal ions from
mixed solutions via polymer‐enhanced ultrafiltration using starch as a water‐soluble biopolymer.
Environmental Progress & Sustainable Energy, 34:359–367.
148. Baharuddin, N.H., Sulaiman, N.M.N., and Aroua, M.K. (2015). Removal of zinc and lead ions by
polymer‐enhanced ultrafiltration using unmodified starch as novel binding polymer. International
Journal of Environmental Science and Technology, 12:1825–1834.
149. Baharuddin, N.H., Sulaiman, N.M.N., and Aroua, M.K. (2014). Unmodified starch as water‐soluble
binding polymer for chromium ions removal via polymer enhanced ultrafiltration system. Journal of
Environmental Health Science and Engineering, 12(61):1–10.
150. Lam, B., Déon, S., Morin‐Crini, N., Crini, G., and Fievet, P. (2018). Polymer‐enhanced ultrafiltration
for heavy metal removal: Influence of chitosan and carboxymethyl cellulose on filtration perfor-
mances. Journal of Cleaner Production, 171:927–933.
151. Crini, G., Morin‐Crini, N., Fatin‐Rouge, N., Déon, S., and Fievet, P. (2017). Metal removal from
aqueous media by polymer‐assisted ultrafiltration with chitosan. Arabian Journal of Chemistry,
10(S2):3826–3839.
152. Déon, S., Deher, J., Lam, B., Crini, N., Crini, G., and Fievet, P. (2017). Remediation of solutions
containing oxyanions of selenium by ultrafiltration: Study of rejection performances with and with-
out chitosan addition. Industrial & Engineering Chemistry Research, 56:10461–10471.
153. Dasgupta, J., Singh, M., Sikder, J., Padarthi, V., Chakraborty, S., and Curcio, S. (2015). Response
surface‐optimized removal of Reactive Red 120 dye from its aqueous solutions using polyethyle-
neimine enhanced ultrafiltration. Ecotoxicology and Environmental Safety, 121:271–278.
18
Chitosan for Sensors and
Electrochemical Applications
Suse Botelho da Silva1, Guilherme Lopes Batista2, and
Cristiane Krause Santin1,2
1
Food and Chemical Engineering, Polytechnic School, Unisinos University, São Leopoldo, RS, Brazil
2
itt CHIP ‐ Unisinos Semiconductor Institute, São Leopoldo, RS, Brazil
This chapter discusses the use of chitosan for sensors and electrochemical applications.
The development of specific sensors and electrochemical devices is among the most inno-
vative applications of chitosan and its composites and derivatives. These materials present
characteristic chemical and electrical features, along with their interesting mechanical and
biological properties (which have been extensively exploited before), making them unique
materials for the referred applications. Although chitosan may present useful characteris-
tics alone, many applications explore its use through chemical modifications or in compos-
ites, leading to materials that may present mixed characteristics or, in some cases, better
performance due to synergic effects. This chapter will present how the structure of chi-
tosan, whether alone or modified, as well as the composite materials containing this poly-
mer, can present very interesting properties for its use in sensing platforms, solid‐state
batteries, and other electronic devices.
18.1 Introduction
In the last decades, there has been a huge demand for biocompatible, biodegradable, and/
or compostable materials in several areas, including in chemical, cosmetic, pharmaceutical,
and medical industries [1]. More recently, the use of “green materials” has also been shown
as a new trend in microelectronics, for applications as electronic devices, solid‐state
batteries, and sensors [2, 3]. In this sense, chitosan appears as a promising substitute for
synthetic polymers for use in electrochemical devices due to many peculiar properties and
characteristics. Chitosan, in pure form or combined with other compounds, can offer robust
and homogeneous materials in many different forms, such as layered films, hydrogels, or
other three‐dimensional materials. Its structure, containing both hydroxyl and amino
groups, is very versatile for chemical modifications for production of sensors and other
electrochemical devices, and also presents good interaction with different ions as
ionophores. Additionally, many chitosan‐based materials exhibit electrical conductivity
due to their proton binding sites that allow proton mobility. All these properties allow the
development of a wide range of applications. This chapter does not intend to cover
comprehensively all the applications of chitosan that are presented in literature, but rather
present some of the many different approaches in which chitosan can be used, making use
of its unique properties for different sensor technologies and other electrochemical
applications. For better understanding, some key aspects regarding these applications, such
as chitosan structure and relevant properties, the proton conductivity mechanism, and
methods for preparation of chitosan‐based materials, will be also presented in this chapter.
O
H3C
OH
6
NH
3
2
1
O
4
5 O
HO
O 5
HO 2
4 O 3 1
6
NH2
OH
Due to all these properties, chitosan has been intensively studied, and several industrial and
technological applications have been developed in the last years, including uses in water
treatment, agriculture, food, pharmaceutical, biomedical, cosmetics, textile, and electronic
areas [5, 10–13]. Among the many properties exhibited by this polysaccharide, chelating
activity and electrical conductivity are of particular relevance in the development of
applications for sensors and other electrochemical devices. The hydrogen‐donating ability
of chitosan is the key factor for many properties exhibited by this polymer, including
electrical conductivity, which is supported by the hydrogen‐bond network along the
chitosan structure.
In solid state, chitosan is a semi‐crystalline polymer that is insoluble in water and most
organic solvents; however, it is soluble in dilute acid solutions [14]. Acetic acid is often
used for solubilization of chitosan, but other diluted organic acids (formic acid, propionic
acid, lactic acid) and inorganic acids (hydrochloric acid and nitric acid) have also been used
[15]. Chitosan is considered as a base which has primary amino groups of glucosamine
with a pKa value of 6.3 [16]. When chitosan is dissolved in acidic medium, it becomes a
cationic polyelectrolyte due to protonation of amino groups [17], according to the equation:
The degree of protonation is dependent on the concentration (pH) and the type (pKa) of
acid used [17]. Likewise, as the pH increases above the pKa, the amino groups deprotonate,
and the polymer loses its charge and becomes insoluble. The soluble–insoluble transition
occurs at its pKa value between pH 6 and 6.5 [16].
As the solubility of chitosan in the acidic medium depends on the ionic concentration, a
salting‐out effect can be observed in excess of hydrochloric acid (>1 M), making it possible
to isolate the hydrochloride form of chitosan [17]. On the other hand, as reported by
Rinaudo et al. [17], an excess of acetic acid or other weak acid increases the chitosan
solubility and no salting‐out was observed, even in high concentrations of acetic acid
(lower than 16.5 M), due to its small dissociation degree. When the hydrochloride and
acetate forms of chitosan are isolated, they are directly soluble in water giving an acidic
solution with pKa = 6.0 [14]. Chitosan salts can also be obtained by water evaporating from
an acid diluted solution [18].
In blending, chitosan is combined with another polymer leading to a polymer blend with
better properties as compared to those obtained from the polymers alone [20]. Blends can
be homogenous or heterogeneous mixtures showing different morphologies, which depend
mainly on the type of process used to combine polymers (solvent blending, extrusion
blending, or reactive extrusion blending) [21]. In grafting, functional groups (and even
polymers) are inserted onto the backbone of chitosan via a chemical reaction or modification
of amino and hydroxyl groups. Functional groups such as carboxylic acid and hydroxyl
groups may be grafting on chitosan to increase the number of hydrogen bonds or to provide
more reactive groups for subsequent modifications [13].
The presence of amino and hydroxyls groups in the chitosan backbone also represents a
great potential for cross‐linking reactions. Dialdehydes can be used to interact with amino
groups of chitosan via covalent imine bonds (Schiff’s base), producing a cross‐linked
structure [9, 22]. Glutaraldehyde and glyoxal are the most commonly used cross‐linker
agents, but also genipin, diethyl squarate, and oxalic acid can be used [22]. Nevertheless,
cross‐linked chitosan can also be produced without an external cross‐linker, being formed
by direct interaction between polymeric chitosan chains through complexation with another
polymer or with a modified chitosan [7, 22]. A self‐cross‐linked amphiphilic structure was
obtained by (2,2,6,6‐tetramethylpiperidin‐1‐yl)oxyl (TEMPO)‐laccase oxidation of chi-
tosan, which added aldehyde groups to the structure from the oxidation of primary hydrox-
yls, allowing the formation of covalent imine bonds directly with the amino groups [19].
In addition to the covalent and ionic bonds, secondary interactions such as hydrogen
bonds and hydrophobic interactions are present in the three‐dimensional network formed
by the cross‐linked chitosan structures [23]. Three‐dimensional hydrophilic polymeric
networks, which absorb and retain water in their structure, are typically called hydrogels
[24]. Hydrogels can take up considerable amounts of water (up to thousands of times its
dry weight) without dissolving [24, 25]. They can occur as a colloid gel as well, in which
water is the dispersion medium [13].
Hydrogels, as well as films, are among the most used forms of chitosan in the development
of applications involving electrochemical devices and sensors. In some applications,
chitosan films or hydrogels can be used as a carrier or as a substrate for other compounds
or nanoparticles, resulting in composite materials with enhanced properties. It is especially
true for sensing applications. In this area, considerable advances have occurred in recent
years, mainly driven by the development of nanotechnology. Chitosan‐based nanomaterials
tend to exhibit superior physical and chemical properties as compared to conventional
chitosan [13]. Tensile strength, conductivity, and photoluminescence are among the
properties that are improved as a result of the typical high surface area of the nanomaterials
[13]. All these properties are particularly important for the manufacturing of films to be
used in sensor devices.
Chitosan films are usually prepared by the casting method [5]. Following this method,
first chitosan is dispersed in a diluted acid solution, typically in acetic acid. This dispersion
is kept under continuous stirring, generally at room temperature, until complete
solubilization. Then, the film‐forming solution is casted over a surface for drying under
controlled temperature and relative humidity [15, 26, 27]. The complete solubilization of
chitosan in a diluted acid solution may take some hours. After solubilization, plasticizers or
other agents can be added to improve the properties of films that will be formed [2, 20].
Likewise, chitosan solution can be filtered, degassed under vacuum, or centrifuged to
Chitosan for Sensors and Electrochemical Applications 465
remove particulate or air bubbles [15, 28] As a polyelectrolyte, chitosan can also be
employed for the preparation of multilayered films, using electrostatic layer‐by‐layer
techniques based on the alternated adsorption of materials bearing opposite charges [29].
Recently, other methods of obtaining chitosan films, other than casting, have been
reported. Thin films have been prepared by spin‐coating [28] and electrochemical
deposition methods [30]. Even more in these cases, obtaining a filmogenic solution with
adequate viscosity and homogeneity is a necessary requirement. By spin‐coating, a dilute
acid chitosan solution is spread over a planar surface under high rotation speed [28]. While
the solution spins over the surface, the solvent is evaporated, being film thickness dependent
on polymer concentration, solvent volatility, and angular speed of spinning [31]. Controlling
these parameters is possible to prepare uniform ultrathin films [28, 31].
By an electrochemical deposition process, two electrodes (cathode and anode) are
dipped into the chitosan solution subjected to constant voltage [30]. The deposition of
chitosan on the cathode (negatively polarized electrode) is caused by the evolution of
hydrogen (H2) from the electrolysis of water, which consumes protons (H+) and generates
excess hydroxide ions (OH)− in the vicinity of this electrode. Thus, the protonated amino
groups of the chitosan are attracted electrostatically to the cathode, leading to the
insolubilization of the polymer next to the electrode. The continuous flow of electrons and
removal of protons by the cathode lead to the formation of a thin chitosan film on the
surface of the cathode [32]. To avoid the evolution of hydrogen gas and produce more
uniform coatings during the modification, a proton‐consuming reagent such as benzoquinone
or chloramphenicol can be added to the plating solution [33]. According to Suginta [32],
the electrodeposition of chitosan is particularly important in the development of
electrochemical biosensor devices, and in the selective placement of polymer deposits on
the active sites of microelectrode and nanoelectrode arrays that would not be easily
produced with conventional film‐forming procedures.
Apart from the method used to obtain chitosan films, the drying process exerts great
importance in the definition of the conductivity of film, especially in the conductivity–
temperature relationship behavior. Films dried in temperatures below Tg (glass temperature)
have higher conductivity than films processed over Tg. This difference is related to the
water content that remains adsorbed on the films, especially which are dehydrated at low
temperatures. The adsorbed water molecules form inter- and intra-form hydrogen bonds
with side groups of chitosan backbone and contribute for the enhancement of the conduc-
tivity of these films [26]. The mechanism behind this phenomenon will be explained in
more detail in section 18.4.
In dry state, chitosan films exhibit a very low electrical conductivity, less than 10−8 S
cm−1 [8]. However, if these films are hydrated or solvated with saline solutions, they
strongly enhance their conductivity [8, 23, 26]. Chitosan acetate films showed conductivity
as high as 10−4 S cm−1 after 1 h of hydration [8]. As shown by the authors [8], the conductiv-
ity increased greatly within 1 h but remained in the same order of magnitude for a longer
hydration time. This shows that the higher conductivity observed in wet films is due to the
adsorption of water, and gives insight that this may be related to conformational changes in
the chitosan chain. As discussed in section 18.2, the dissolution of chitosan in an acidic
solution causes the protonation of amino groups in the backbone structure, which remains
in chitosan films. In the same way, when dry chitosan films are hydrated, the amino groups,
which are weak alkaline groups, are partially protonated and some hydroxide ions are
formed [26]:
Chit NH 2 H 2 O NH 3 OH (2)
Nevertheless, the increase on the conductivity of wet chitosan films is not due to ionic
conductivity (OH− ion conductivity) [26, 34], otherwise it is due to the “Grotthuss
mechanism” (proton‐hopping mechanism), as discussed by Prokhorov et al. [26]. When
water molecules are adsorbed by the chitosan chain, hydrogen bonds are formed
between water and protonated amino groups and other polar groups [34]. Several intra‐
hydrogen and inter‐hydrogen bonds in the chitosan structure create a kind of network
through which the protons (H+) can “jump” moving along the hydrogen bond network,
following a process of reorientation with cleavage/formation bonds with a local molec-
ular rearrangement [26, 34]. This proton mobility explains the difference in the conduc-
tivity of wet and dried chitosan films. It also supports the prevalent nonlinearity between
conductivity and temperature, described by Vogel–Fulcher–Tammann (VFT) model, in
heating experiments of wet films in the range of 25–70°C [26]. The VFT behavior is
related to the α‐relaxation process including the rotation of hydroxyl and amino groups
and breaking of hydrogen bonds due to water desorption for wet films obtained below
their Tg [26, 36]. These were confirmed by dielectric relaxation measurements [26].
Since the deacetylation degree of chitosan affects the water uptake, the proton
conductivity will also be affected, with highly deacetylated chitosan exhibiting higher
conductivity [23].
The conductivity exhibited by dried chitosan films can be explained by Grotthuss
“packed‐acid mechanism” [26, 36]. “Packed‐acid mechanism” is a kind of proton‐hopping
mechanism in which the conduction occurs by acid–acid interactions without the movement
of water [35, 37]. This mechanism was proposed by Ogawa et al. [37], and it occurs under
low humidity conditions and is typically observed in materials with a high acid concentration
[35]. A packed‐acid structure has a large number of proton donors, which increases the
concentration of protons in a packed‐acid structure, sufficiently to construct a weak
hydrogen‐bond network among the proton‐donors whereby protons move. [37]. Self‐
assembled polymer electrolytes can construct a similar structure as packed acids [37], dried
chitosan films produced by previous acid dissolution would be an example. In acetate
chitosan films, protons will be transferred through neighboring acid groups supported by
the formation of new bonds between −NH3+ groups and acetate ions [26]. Use of different
acids must give rise to different results in proton conductivity, since different acids induce
Chitosan for Sensors and Electrochemical Applications 467
to be measured. There are several approaches for electrochemical sensors and chitosan
derivatives, and composites have been used in many ways. As chitosan usually does not
present good electrical conductivity, it is rather common to make composites with more
conductive materials for making electrodes and sensors. Usually, the materials that have
been used more frequently are carbon‐based, especially carbon paste, graphite, graphene,
and carbon nanotubes. Chitosan composites can make very homogeneous dispersions,
retaining the conductive properties of these fillers and the chemical and catalytic properties
of chitosan and other composite materials.
The amino groups that are present on the chitosan molecules are binding sites for
electrostatic interactions. When in the protonated form, these groups interact well with
anions, and this property has been applied for selective potentiometric sensing of uranyl
ions (UO2−) [40] and monovalent anions [41]. In this type of sensor, the electric potential
of the membrane changes depending on the ion concentration, which is measured relative
to a reference electrode (with known electric potential) at zero current.
Although the surface of chitosan itself may be used as a sensing material for potentiometric
sensors, the surface can be further modified for the determination of different species. The
modification of sensors with enzymes has been extensively used for the fabrication of
analytical devices, and chitosan‐based materials are considered very suitable for this
application [42]. Enzymes have excellent catalytic activity and exhibit high selectivity
toward the target substrate, acting as excellent recognition molecules in sensors [43].
The modification of chitosan‐based materials with enzymes may happen in different
forms, that we can classify broadly as physical and/or chemical, and more than one
mechanism can occur simultaneously. For its modification, the methods may be classified
broadly into four groups: solvent evaporation method, in which a solution of chitosan in an
organic acid is casted and dried over a surface, typically an electrode (for this application,
it may be referred as drop‐casting); neutralization method, in which small particles of
chitosan are formed upon the titration of an acidic chitosan solution by a strong alkali;
cross‐linking method, using chemical agents to cross‐link chitosan molecules; and
coacervation, which precipitates chitosan on acidic solutions with anionic polyelectrolytes,
usually premodified with enzymes. A broad range of applications has been presented in a
review [42]. One example of enzymatic modification for a potentiometric approach used
glucose oxidase on a composite of chitosan with polyvinyl alcohol (PVA) by drop‐casting,
and the hydrogen peroxide formed in the reaction with glucose was sensed on platinized
paper. Although the enzyme is selective towards glucose the platinum surface was modified
with Nafion in order to enhance its selectivity on the potentiometric measurements,
avoiding anions to make contact with the platinum electrode [44].
Other biological molecules that have been incorporated to chitosan composites are
antibodies, which allow specific interaction with more complex molecules. Ma et al. [45]
developed a disposable amperometric immunosensor for determination of the Aflatoxin B1
(AFB1) in wheat based on chitosan and anti‐AFB1 antibody. Electrochemical immunosensors
are sensors based on solid‐state devices in which immunochemical reactions are coupled to
a transducer surface to generate the output as electrochemical signal [46]. Ma et al. [45]
reported that pH can interfere in the current measurements in an AFB1 sensor, therefore,
this effect must be understood and considered in the development of sensors using chitosan.
The modification of chitosan with inorganic materials may also render materials with
very interesting properties. For example, the composite of chitosan with graphite powder
Chitosan for Sensors and Electrochemical Applications 469
and montmorillonite can form a structured three‐dimensional material when the amount of
chitosan is below the cationic exchange capacity (CEC) of the clay. However, when
chitosan is at a concentration higher than CEC, it forms a bidimensional nanostructured
material composed of a bilayer of chitosan intercalated with the interlayer region of the
clay, and the amino groups of the chitosan act as ionic exchange sites. This material was
used as a potentiometric sensor specific for monovalent ions, with more sensitivity for the
detection of nitrite ions [41].
Besides potentiometric measurements, many other modifications have been made to use
chitosan composites for sensor materials. One of the most common approaches for
electrochemical sensors is the measurement of an electric current on a working electrode
(which is a conductive surface) due to an electrochemical reaction, which is driven by a
difference of electrical potential applied between the sample and the electrode. These
modifications improve the sensor selectivity and sensitivity toward selected molecules or
groups of compounds [47].
A novel method for analysis of hydroxymethanesulfinate (HMS) was developed using
chitosan modified with carbon nanotubes and cetyltrimethylammonium (CTAB) [48]. In
bare glassy carbon electrodes, the signal of HMS is superimposed on the sulfite signal. The
modification, however, allows performing the analysis of both compounds simultaneously
due to the interaction of the CTAB in the interface with the sample.
More impressively, the incorporation of chiral compounds on chitosan composites may
allow the selective determination of chiral compounds. For example, an electrode composed
of chitosan modified with 3,4,9,10‐perylenetetracarboxylic acid (PTCA) for the
enantioselective determination of tryptophan presented a synergic effect that increased
substantially the selectivity of the determination of D‐tryptophan (selectivity factor of 2.6),
even comparing with glassy carbon electrode with PTCA or with chitosan alone (selectivity
factors of 1.1 and 1.2) [49]. Another interesting example of chiral determination is the
determination of atenolol using chitosan and chitosan succinamide with cyclodextrin
composites [50]. Although each enantiomer did not present a large difference in signals
between R‐atenolol and S‐atenolol, the enantiomeric determination could be accomplished
by the use of an electrode array with the different modifications combined to the use of
chemometric tools (such as principal component analysis) to evaluate the signals.
Another emerging approach for highly selective electrochemical analysis is by the use of
molecularly imprinted polymers (MIPs). These polymers are synthesized with known
cavity sizes for recognition of specific molecules, and the interaction of this polymer with
the target molecule will render an analytical signal. To build an electrically conductive MIP
on a stable surface, a carbon dots–chitosan composite was prepared and 3‐
aminobenzeneboronic acid was electrodeposited at its surface in the presence of glucose
molecules to build the cavities [51]. The system was tested to determine glucose on human
serum with excellent selectivity and fairly good detection limit (0.09 μM).
Other modifications that have been extensively studied are the incorporation or synthesis
of nanoparticles in chitosan composites. In many cases, the chitosan‐based materials
stabilize the nanoparticles of the composite, avoiding agglomeration and immobilizing on
the surface for sensor fabrication. Chitosan has been extensively used in combination with
carbon nanoparticles and derivatives. The simultaneous determination of mercury, copper,
lead, and cadmium was made by a fullerene (C60)–chitosan composite [52]. Besides
forming a stable and uniform film for the sensing surface, the positively charged chitosan
470 Chitin and Chitosan: Properties and Applications
changes the electrical cloud distribution of C60. In another approach, a highly specific and
sensitive sensor for copper (II) was developed using amino‐functionalized graphene
combined with chitosan [53].
Besides being carbon‐based, much work has been done using metal nanoparticles on
chitosan composites. In many cases, they have catalytic properties on electrochemical
reactions, enhancing sensitivity and selectivity. The detection of ochratoxin A, a mycotoxin
that can be present in grape juice, could be achieved by amperometric detection using a
blend of graphene, multiwalled carbon nanotubes (MWCNT), ionic liquid, chitosan,
collagen, and nickel oxide nanoparticles [54]. While the carbon compounds enhance the
electrical conductivity and surface area of the sample, the nanoparticles increase sensitivity
and selectivity toward ochratoxin A, collagen can stabilize these nanoparticles, and chitosan
provides a film‐forming property to the mixture, as well as mechanical strength. In a
different approach, functionalized MWCNT and cobalt were dispersed on chitosan to the
determination of paracetamol. In this case, chitosan can disperse the f‐MWCNT
homogeneously and prevent its agglomeration. This sensor allowed the determination of
paracetamol in commercial tablets and human serum samples [55].
Glucose‐sensing could be accomplished by amperometry using chitosan modified with
copper (II) oxide [56]. This modification was made through a hydrothermal method, in
which the copper is oxidized after being coordinated with chitosan. This interaction
promotes differences in the homogeneity of the mixture and in the growth of copper oxide.
Besides being nonenzymatic, this method has high sensibility and low detection limit
(LD = 11 μM).
The selective determination of Cr3+ and Cr6+ could be accomplished using a dual
electrochemical detector using gold nanoparticle‐decorated carbon fiber chitosan‐modified
glassy carbon electrode [57]. This electrode presents the catalytic properties for chromium
electrochemical reactions on mild acidic pH and made the speciation of these species
possible with no prior chemical separation.
in‐between the cell and electrode. With this, the system was sensitive enough to detect
H2O2 released at a single cell level [59].
The use of modified chitosan made the development of a wound‐healing membrane
possible, sensitive to the redox potential of the surface. For this, anthraquinone molecules
attached to gold nanoshells were loaded on the surface of a chitosan wound‐dressing
membrane, in order to monitor the redox potential of the material by surface‐enhanced
Raman scattering (SERS). This probe allows in situ measurement of redox potentials in a
noninvasive manner, making it possible to follow the wound‐healing process in a
noninvasive way [60].
Fluorescent detectors have also been implemented using chitosan composites. A
fluorophore, synthesized by cross‐linking chitosan using glutaraldehyde, was able to moni-
tor the concentration of Cr6+ selectively in the environment, by the oxidation of C═N bonds
of the composite, promoting a quenching effect on the fluorescence of the material [61].
A phosphorescent composite was obtained by the stabilization of trinuclear gold(I)
pyrazolate (Au3Pz3) on chitosan [62]. The polysaccharide, in this composite, has been used
as a template for Au3Pz3 synthesis, having an important role in the formation of gold(I)
pyrazolate complexes. The probe is especially sensitive to silver (I) ions in solution,
presenting zero interference from 15 other metals tested.
Another promising energy storage solution which has drawn much research efforts are
the supercapacitors [67]. They are based mainly on two phenomena: electrochemical dou-
ble layer capacitance (EDLC) and pseudocapacitance. In the EDLC, the surface of the
electrode presents a charge separation between the capacitor electrolytes, thus a large sur-
face area is desired. The pseudocapacitance is referred to fast and reversible faradaic reac-
tions between the electrode surface and the electrolyte [67]. The ability of chitosan
composites to form self‐assembling and cross‐linking structures with graphene oxide
results in a high‐porosity and low‐density material which has been proposed for superca-
pacitors [68]. Besides these interesting properties, the material presents exceptional
mechanical properties, such as compressive strength and resilience.
Although the chitosan composites can be used to enhance the surface area of electrodes
(making EDLC higher), its capacitance can benefit from its chemistry as well. Chitosan
composites with activated carbon present higher pseudocapacitance as a modified activated
carbon, due to its nitrogen containing groups, whose stability also favors its applicability
on supercapacitors [69].
In rechargeable batteries, the variation of volume on electrodes demands special
materials. In this context, a chitosan–glutaraldehyde composite linked by cyano bridges,
which behaves as a double network hydrogel, has been used with Sn–Fe nanoparticles
resulting on a 3D structure, confining the alloy particles in a hierarchical carbon framework.
As a result, the material exhibits long life cycles and high rate capability [70]. Chitosan was
also proposed in composition with natural rubber to form an elastic composite for Si anode
Li‐ion batteries [71]. High cyclic performance was achieved once natural rubber made the
material elastic in a reversible manner, while chitosan could anchor Si particles through
hydrogen bonding.
The synaptic transistor is a new type of device that presents nonvolatile memory and
history‐dependent analog‐like states, which simulate the behavior of a neuron and are
recently being proposed for new computing architectures [72]. A novel topology for this
kind of transistor is proposed using chitosan dispersed in acetic acid as a material for the
drain of the transistor, and indium tin oxide (ITO) for electrode materials on a polyethylene
terephthalate (PET) substrate [73], yielding a biocompatible and flexible synaptic
transistor.
18.6 Outlook
Chitosan has been widely investigated as a new alternative material for a broad range of
applications, and it presents unique characteristics that are very suitable in electrochem-
ical devices. Its structure allows physical dispersion and chemical modification, com-
bining conductive and functional materials to build very selective sensors and intelligent
materials. These dispersions can be tailored to synthetize and/or maintain nanoparticles
with reduced agglomeration effects, favoring the use of nanosized materials. Moreover,
the chemical structure of chitosan itself appears to be a convenient and selective mate-
rial for sensing applications. Other applications, moreover, use its proton conduction
property to assemble energy storage and other electrochemical devices. However, the
use of chitosan is still very recent in this area, and much novel applications are surely
about to come.
Chitosan for Sensors and Electrochemical Applications 473
References
1. Kardas, I., Struszczyk, M.H., Kucharska, M., van den Broek, L.A.M., van Dam, J.E.G., and
Ciechańska, D. (2013) Chitin and chitosan as functional biopolymers for industrial applications, in
The European Polysaccharide Network of Excellence (EPNOE): Research Initiatives and Results,
Springer, pp. 329–373.
2. Shukur, M.F., and Kadir, M.F.Z. (2015) Hydrogen ion conducting starch‐chitosan blend based electro-
lyte for application in electrochemical devices. Electrochim. Acta, 158, 152–165.
3. Monisha, S., Mathavan, T., Selvasekarapandian, S., Milton, A., Benial, F., Aristatil, G., Mani, N.,
Premalatha, M., and Vinoth Pandi, D. (2017) Investigation of bio polymer electrolyte based on cellu-
lose acetate‐ammonium nitrate for potential use in electrochemical devices. Carbohydr. Polym., 157,
38–47.
4. Liu, N., Gan, L., Liu, Y., Gui, W., Li, W., and Zhang, X. (2017) Improving pH sensitivity by field‐
induced charge regulation in flexible biopolymer electrolyte gated oxide transistors. Appl. Surf. Sci.,
419, 206–212.
5. Luo, Y., and Wang, Q. (2013) Recent advances of chitosan and its derivatives for novel applications in
food science. J. Food Process. Beverages, 1 (1), 1–13.
6. Lee, D.W., Lim, C., Israelachvili, J.N., and Hwang, D.S. (2013) Strong adhesion and cohesion of chi-
tosan in aqueous solutions. Langmuir, 29 (46), 14222–14229.
7. Croisier, F. and Jérôme, C. (2013) Chitosan‐based biomaterials for tissue engineering. Eur. Polym. J.,
49 (4), 780–792.
8. Wan, Y., Creber, K.A., Peppley, B., and Bui, V.T. (2003) Ionic conductivity of chitosan membranes.
Polymer (Guildf)., 44 (4), 1057–1065.
9. Prashanth, K. V., Harish Tharanathan, R.N., Harish Prashanth, K.V., and Tharanathan, R.N. (2007)
Chitin/chitosan: modifications and their unlimited application potential—an overview. Trends Food
Sci. Technol., 18 (3), 117–131.
10. Kong, M., Chen, X.G., Xing, K., and Park, H.J. (2010) Antimicrobial properties of chitosan and mode
of action: a state of the art review. Int. J. Food Microbiol., 144 (1), 51–63.
11. Zargar, V., Asghari, M., and Dashti, A. (2015) A review on chitin and chitosan polymers: Structure,
chemistry, solubility, derivatives, and applications. ChemBioEng Rev., 2 (3), 204–226.
12. Hamed, I., Özogul, F., and Regenstein, J.M. (2016) Industrial applications of crustacean by‐products
(chitin, chitosan, and chitooligosaccharides): A review. Trends Food Sci. Technol., 48, 40–50.
13. Shukla, S.K., Mishra, A.K., Arotiba, O.A., and Mamba, B.B. (2013) Chitosan‐based nanomaterials: A
state‐of‐the‐art review. Int. J. Biol. Macromol., 59, 46–58.
14. Rinaudo, M. (2006) Chitin and chitosan: Properties and applications. Prog. Polym. Sci., 31 (7),
603–632.
15. Kim, K.M., Son, J.H., Kim, S.‐K., Weller, C.L., and Hanna, M.A. (2006) Properties of chitosan films
as a function of pH and solvent type. J. Food Sci., 71 (3), E119–E124.
16. Pillai, C.K.S., Paul, W., and Sharma, C.P. (2009) Chitin and chitosan polymers: Chemistry, solubility
and fiber formation. Prog. Polym. Sci., 34 (7), 641–678.
17. Rinaudo, M., Pavlov, G., and Desbrières, J. (1999) Influence of acetic acid concentration on the solu-
bilization of chitosan. Polymer (Guildf)., 40 (25), 7029–7032.
18. Demarger‐Andre, S., and Domard, A. (1994) Chitosan carboxylic acid salts in solution and in the solid
state. Carbohydr. Polym., 23 (3), 211–219.
19. Botelho da Silva, S., Krolicka, M., van den Broek, L.A.M., Frissen, A.E., and Boeriu, C.G. (2018)
Water‐soluble chitosan derivatives and pH‐responsive hydrogels by selective C‐6 oxidation mediated
by TEMPO‐laccase redox system. Carbohydr. Polym., 186, 299–309.
20. Elsabee, M.Z. and Abdou, E.S. (2013) Chitosan based edible films and coatings: A review. Mater. Sci.
Eng. C, 33 (4), 1819–1841.
474 Chitin and Chitosan: Properties and Applications
21. van den Broek, L.A.M., Knoop, R.J.I., Kappen, F.H.J., and Boeriu, C.G. (2015) Chitosan films and
blends for packaging material. Carbohydr. Polym., 116, 237–242.
22. Berger, J., Reist, M., Mayer, J.M.M., Felt, O., Peppas, N.A.A., and Gurny, R. (2004) Structure and
interactions in covalently and ionically crosslinked chitosan hydrogels for biomedical applications.
Eur. J. Pharm. Biopharm., 57 (1), 19–34.
23. Ma, J. and Sahai, Y. (2013) Chitosan biopolymer for fuel cell applications. Carbohydr. Polym., 92 (2),
955–975.
24. Singh, A., Narvi, S.S., Dutta, P.K., and Pandey, N.D. (2006) External stimuli response on a novel chi-
tosan hydrogel crosslinked with formaldehyde. Bull. Mater. Sci, 29 (3), 233–238.
25. Yucel Falco, C., Sotres, J., Rascón, A., Risbo, J., and Cárdenas, M. (2017) Design of a potentially
prebiotic and responsive encapsulation material for probiotic bacteria based on chitosan and sulfated
β‐glucan. J. Colloid Interface Sci., 487, 97–106.
26. Prokhorov, E., Luna‐Barcenas, G., González‐Campos, J.B., Kovalenko, Y., Garcia‐Carvajal, Z.Y., and
Mota‐Morales, J. (2016) Proton conductivity and relaxation properties of chitosan‐acetate films.
Electrochim. Acta, 215, 600–608.
27. Shukur, M.F., Ithnin, R., and Kadir, M.F.Z. (2014) Electrical properties of proton conducting solid
biopolymer electrolytes based on starch–chitosan blend. Ionics (Kiel)., 20 (7), 977–999.
28. Voznesenskiy, S., Sergeev, A., Mironenko, Ay., Bratskaya, Sy., and Kulchin, Y. (2013) Integrated‐
optical sensors based on chitosan waveguide films for relative humidity measurements. Sensors
Actuators B. Chem., 188, 482–487.
29. Pavinatto, F.J., Caseli, L., and Oliveira, O.N. (2010) Chitosan in nanostructured thin films.
Biomacromolecules, 11 (8), 1897–1908.
30. Shantini, D., Nainggolan, I., Nasution, T.I., Derman, M.N., Mustaffa, R., and Abd Wahab, N.Z. (2016)
Hexanal gas detection using chitosan biopolymer as sensing material at room temperature. J. Sensors,
2016, 1–7.
31. Hall, D.B., Underhill, P., and Torkelson, J.M. (1998) Spin coating of thin and ultrathin polymer films.
Polym. Eng. Sci., 38 (12), 2039–2045.
32. Suginta, W., Khunkaewla, P., and Schulte, A. (2013) Electrochemical biosensor applications of poly-
saccharides chitin and chitosan. Chem. Rev., 113 (7), 5458–5479.
33. Donahue, C.E.T., Miller, D.R., Beger, T.W., Johann, T.W., and Keithley, R.B. (2018) Improved forma-
tion of electrically‐deposited enzyme‐embedded chitosan coatings onto carbon fiber microelectrodes.
Anal. Methods, 10 (13), 1565–1576.
34. Wu, G., Zhang, J., Wan, X., Yang, Y., and Jiang, S. (2014) Chitosan‐based biopolysaccharide proton
conductors for synaptic transistors on paper substrates. J. Mater. Chem. C, 2 (31), 6249–6255.
35. Ogawa, T., Kamiguchi, K., Tamaki, T., Imai, H., and Yamaguchi, T. (2014) Differentiating Grotthuss
proton conduction mechanisms by nuclear magnetic resonance spectroscopic analysis of frozen sam-
ples. Anal. Chem., 86 (19), 9362–9366.
36. Kumar‐Krishnan, S., Prokhorov, E., Ramírez, M., Hernandez‐Landaverde, M.A., Zarate‐Triviño,
D.G., Kovalenko, Y., Sanchez, I.C., Méndez‐Nonell, J., and Luna‐Bárcenas, G. (2014) Novel gigahertz
frequency dielectric relaxations in chitosan films. Soft Matter, 10 (43), 8673–8684.
37. Ogawa, T., Aonuma, T., Tamaki, T., Ohashi, H., Ushiyama, H., Yamashita, K., and Yamaguchi, T.
(2014) The proton conduction mechanism in a material consisting of packed acids. Chem. Sci., 5 (12),
4878–4887.
38. Khiar, A.S.A., Puteh, R., and Arof, A.K. (2006) Conductivity studies of a chitosan‐based polymer
electrolyte. Phys. B Condens. Matter, 373 (1), 23–27.
39. Du, J.F., Bai, Y., Chu, W.Y., and Qiao, L.J. (2010) The structure and electric characters of proton‐
conducting chitosan membranes with various ammonium salts as complexant. J. Polym. Sci. Part B
Polym. Phys., 48 (8), 880–885.
40. Ali, T.A., Mohamed, G.G., Aglan, R.F., and Mourad, M.A. (2018) A novel screen‐printed and carbon
paste electrodes for potentiometric determination of uranyl(II) ion in spiked water samples. Russ.
J. Electrochem., 54 (2), 201–215.
Chitosan for Sensors and Electrochemical Applications 475
41. Darder, M., Colilla, M., and Ruiz‐Hitzky, E. (2005) Chitosan–clay nanocomposites: Application as
electrochemical sensors. Appl. Clay Sci., 28 (1–4), 199–208.
42. Krajewska, B. (2004) Application of chitin‐ and chitosan‐based materials for enzyme immobilizations:
a review. Enzyme Microb. Technol., 35 (2–3), 126–139.
43. Putzbach, W., and Ronkainen, N. (2013) Immobilization techniques in the fabrication of nanomate-
rial‐based electrochemical biosensors: A review. Sensors, 13 (4), 4811–4840.
44. Guadarrama‐Fernández, L., Novell, M., Blondeau, P., and Andrade, F.J. (2018) A disposable, simple,
fast and low‐cost paper‐based biosensor and its application to the determination of glucose in com-
mercial orange juices. Food Chem., 265, 64–69.
45. Ma, H., Sun, J., Zhang, Y., and Xia, S. (2016) Disposable amperometric immunosensor for simple and
sensitive determination of aflatoxin B1 in wheat. Biochem. Eng. J., 115, 38–46.
46. Catanante, G., Rhouati, A., Hayat, A., and Marty, J.L. (2016) An Overview of Recent Electrochemical
Immunosensing Strategies for Mycotoxins Detection. Electroanalysis, 28 (8), 1750–1763.
47. Murray, R.W., Ewing, A.G., and Durst, R.A. (1987) Chemically modified electrodes. Molecular design
for electroanalysis. Anal. Chem., 59 (5), 379A–390A.
48. Wu, Z., Guo, F., Huang, L., and Wang, L. (2018) Electrochemical nonenzymatic sensor based on cetyl-
trimethylammonium bromide and chitosan functionalized carbon nanotube modified glassy carbon
electrode for the determination of hydroxymethanesulfinate in the presence of sulfite in foods. Food
Chem., 259, 213–218.
49. Mo, Z., Niu, X., Gao, H., Li, Z., Meng, S., and Guo, R. (2018) Electrochemical recognition for
tryptophan enantiomers based on 3, 4, 9, 10‐perylenetetracarboxylic acid–chitosan composite film.
J. Solid State Electrochem., 22 (80), 2405–2412.
50. Zilberg, R.A., Maistrenko, V.N., Kabirova, L.R., and Dubrovsky, D.I. (2018) Selective voltammetric
sensors based on composites of chitosan polyelectrolyte complexes with cyclodextrins for the recogni-
tion and determination of atenolol enantiomers. Anal. Methods, 10 (16), 1886–1894.
51. Zheng, W., Wu, H., Jiang, Y., Xu, J., Li, X., Zhang, W., and Qiu, F. (2018) A molecularly‐imprinted‐
electrochemical‐sensor modified with nano‐carbon‐dots with high sensitivity and selectivity for rapid
determination of glucose. Anal. Biochem., 555, 42–49.
52. Han, X., Meng, Z., Zhang, H., and Zheng, J. (2018) Fullerene‐based anodic stripping voltammetry
for simultaneous determination of Hg(II), Cu(II), Pb(II) and Cd(II) in foodstuff. Microchim. Acta,
185 (5), 274.
53. Mo, Z., Liu, H., Hu, R., Gou, H., Li, Z., and Guo, R. (2018) Amino‐functionalized graphene/chitosan
composite as an enhanced sensing platform for highly selective detection of Cu2+. Ionics (Kiel).,
24 (5), 1505–1513.
54. Jalalvand, A.R. (2018) Fabrication of a novel and high‐performance amperometric sensor for highly
sensitive determination of ochratoxin A in juice samples. Talanta, 188, 225–231.
55. Akhter, S., Basirun, W.J., Alias, Y., Johan, M.R., Bagheri, S., Shalauddin, M., Ladan, M., and Anuar,
N.S. (2018) Enhanced amperometric detection of paracetamol by immobilized cobalt ion on function-
alized MWCNTs – Chitosan thin film. Anal. Biochem., 551, 29–36.
56. Figiela, M., Wysokowski, M., Galinski, M., Jesionowski, T., and Stepniak, I. (2018) Synthesis and
characterization of novel copper oxide‐chitosan nanocomposites for non‐enzymatic glucose sensing.
Sensors Actuators, B Chem., 272, 296–307.
57. Nellaiappan, S., and Kumar, A.S. (2018) A bipotentiostat based separation‐free method for simultane-
ous flow injection analysis of chromium (III) and (VI) species. Electrochim. Acta, 273, 248–256.
58. Han, Z., Shu, J., Jiang, Q., and Cui, H. (2018) Coreactant‐free and label‐free eletrochemilumines-
cence immunosensor for copeptin based on luminescent immuno‐gold nanoassemblies. Anal. Chem.,
90 (10), 6064–6070.
59. Liu, G., Ma, C., Jin, B.‐K., Chen, Z., and Zhu, J.‐J. (2018) Direct electrochemiluminescence imaging
of a dingle cell on a chitosan film modified electrode. Anal. Chem., 90 (7), 4801–4806.
60. Sun, J., Han, S., Wang, Y., Zhao, G., Qian, W., and Dong, J. (2018) Detection of redox state evolution during
wound healing process based on a redox‐sensitive wound dressing. Anal. Chem., 90 (11), 6660–6665.
476 Chitin and Chitosan: Properties and Applications
61. Song, J., Zhou, H., Gao, R., Zhang, Y., Zhang, H., Zhang, Y., Wang, G., Wong, P.K., and Zhao, H.
(2018) Selective determination of Cr(VI) by glutaraldehyde cross‐linked chitosan polymer fluoro-
phores. ACS Sensors, 3 (4), 792–798.
62. Upadhyay, P.K., Marpu, S.B., Benton, E.N., Williams, C.L., Telang, A., and Omary, M.A. (2018) A
phosphorescent trinuclear gold(I) tyrazolate chemosensor for silver ion detection and remediation in
aqueous media. Anal. Chem., 90 (8), 4999–5006.
63. Jayakumar, R., Prabaharan, M., Sudheesh Kumar, P.T., Nair, S. V., and Tamura, H. (2011) Biomaterials
based on chitin and chitosan in wound dressing applications. Biotechnol. Adv., 29 (3), 322–337.
64. Bu, Y., Xu, H.X., Li, X., Xu, W.J., Yin, Y.X., Dai, H.L., Wang, X. Bin, Huang, Z.J., and Xu, P.H. (2018)
A conductive sodium alginate and carboxymethyl chitosan hydrogel doped with polypyrrole for
peripheral nerve regeneration. RSC Adv., 8 (20), 10806–10817.
65. Kang, M., Liu, S., Oderinde, O., Yao, F., Fu, G., and Zhang, Z. (2018) Template method for dual net-
work self‐healing hydrogel with conductive property. Mater. Des., 148, 96–103.
66. Kalaiselvimary, J., Sundararajan, M., and Prabhu, M.R. (2018) Preparation and characterization of
chitosan‐based nanocomposite hybrid polymer electrolyte membranes for fuel cell application. Ionics
(Kiel)., 24 (11), 3555–3571.
67. Wang, G., Zhang, L., and Zhang, J. (2012) A review of electrode materials for electrochemical super-
capacitors. Chem. Soc. Rev., 41 (2), 797–828.
68. Zhang, Y., Zhang, L., Zhang, G., and Li, H. (2018) Naturally dried graphene‐based nanocomposite
aerogels with exceptional elasticity and high electrical conductivity. ACS Appl. Mater. Interfaces,
10 (25), 21565–21572.
69. Sun, G., Li, B., Ran, J., Shen, X., and Tong, H. (2015) Three‐dimensional hierarchical porous carbon/
graphene composites derived from graphene oxide‐chitosan hydrogels for high performance superca-
pacitors. Electrochim. Acta, 171, 13–22.
70. Shi, H., Fang, Z., Zhang, X., Li, F., Tang, Y., Zhou, Y., Wu, P., and Yu, G. (2018) Double‐network
nanostructured hydrogel‐derived ultrafine Sn–Fe alloy in three‐dimensional carbon framework for
enhanced lithium storage. Nano Lett., 18 (5), 3193–3198.
71. Lee, S.H., Lee, J.H., Nam, D.H., Cho, M., Kim, J., Chanthad, C., and Lee, Y. (2018) Epoxidized natu-
ral rubber/chitosan network binder for silicon anode in lithium‐ion battery. ACS Appl. Mater. Interfaces,
10 (19), 16449–16457.
72. Shi, J., Ha, S.D., Zhou, Y., Schoofs, F., and Ramanathan, S. (2013) A correlated nickelate synaptic
transistor. Nat. Commun., 4 (1), 2676.
73. Yu, F., Zhu, L.Q., Gao, W.T., Fu, Y.M., Xiao, H., Tao, J., and Zhou, J.M. (2018) Chitosan‐based poly-
saccharide‐gated flexible indium tin oxide synaptic transistor with learning abilities. ACS Appl. Mater.
Interfaces, 10 (19), 16881–16886.
19
Marketing and Regulations
of Chitin and Chitosan
from Insects
Nathalie Berezina and Antoine Hubert
Ynsect, Évry, France
Chitin, the second most abundant biopolymer worldwide, and chitosan, its deacetylated
derivative, are considered as very promising products in the market. Until now, chitin was
mainly sourced as a by‐product of marine arthropods, but with the emergence of the insect
industry this state is about to change. The aim of this chapter is to understand whether and
how this may have an incidence on chitin‐ and chitosan‐derived products, and examine the
main regulation constraints for their arrival in the market.
CH3
O CH3
OH O
NH
HO O NH
O HO Chitin
O O O
O HO O
NH
OH
O
CH3 OH
Deacetylation
CH3
OH O
NH2
HO O NH
O HO
O O O
O HO O Chitosan
NH2
OH
OH
differentiation between these two biopolymers, and, when exploring the exoskeleton of silk
worm, Bombyx mori [2], in 1843, Lassaigne noticed that, in contrast to cellulose, chitin
contained nitrogen in its intimate structure. Further studies on the chitin structure were
conducted, among others, by Ledderhose in 1879 and Gilson in 1894; finally, Purchase and
Braun established the structural information of chitin in 1946, declaring it to be a biopoly-
mer with repeating N‐acetyl‐glucosamine units (Figure 19.1) [2].
Chitosan is the deacetylated version of chitin (Figure 19.1). It is almost absent in its
actual form in nature and was first obtained by C. Rouget by boiling the chitin in a concen-
trated alkali solution [3]. Due to its natural protective function, chitin is extremely resistant
and almost insoluble; therefore, it is difficult to find any direct applications for it, whereas
once deacetylated to give chitosan, the latter becomes soluble in slightly acidic conditions,
allowing a number of applications. Therefore, the main market applications and the related
regulations discussed in this chapter concern chitosan rather than chitin.
important to notice that the determination of the chitin content depends on the methods
used. Different techniques such as infrared or diffractive analysis remain only qualitative,
and the main quantitative method used is the extraction in basic medium, which strongly
depends on the matrix subjected to extraction and the operating conditions [8, 9]. Therefore,
contradictory results were found by different research groups concerning chitin content in
cuttlefish or shrimp [8, 9].
and base treatments are rarely enough for obtaining perfectly pure and white chitin; there-
fore, bleaching steps are often applied at the end of the processes [16].
the scope of fertilizers, as it specifies that fertilizers should provide nutrients to the plants,
which the biostimulants clearly do not [44].
supplementation of chitin in broiler diet inhibited the growth of foodborne pathogens such
as Escherichia coli or Salmonella spp [73].
To the best of our knowledge, no specific regulations have yet been issued for controlling
the addition of these substrates to animal diets. It is anticipated that, in addition to the
application of elicitors in agriculture for the replacement of pesticides, these applications
for the enhancement of the innate immune systems of animals will be highly appreciated,
as they have the potential to reduce the preventive antibiotic treatments of livestock.
19.7 Outlook
Chitin and chitosan are very promising molecules for several applications. In case of chi-
tosan, the initial source of the biopolymer is no longer of interest, as the extraction, purifi-
cation, and deacetylation of chitin tend to uniformize the obtained product, and the applied
process has more influence on the final product and its properties than the original
organism.
Among the different market opportunities reviewed, chitin is mainly concerned with a
few applications in seed protection in agriculture and improvement of the innate immune
system in animals, whereas chitosan has been found to present great possibilities in fields
such as water treatment and biomedical applications.
Bringing a new product into the market is always a costly and long process, as safety
concerns need to be addressed. We can regret that, in many fields, such as active packaging
and elicitors, no unified regulation frameworks are available, and sometimes the same
product can fall under several different categories with specific requirements to be
addressed.
However, we also have reasons to be optimistic, as chitin and chitosan exhibit actual,
diverse, and very interesting properties, with many valuable applications in several fields
where they can replace more drastic or harmful compounds. Also, the emergence of the
insect industry, even if they do not modify the intrinsic properties of these biopolymers,
allows their more consistent production; therefore, in the coming years, an increase in the
market share of chitin‐ and chitosan‐derived products can be anticipated.
References
1. Wisniak, J. and Braconnot H. Revista CENIC. Ciencias Químicas, 2007, 38, 345–355 and references
cited therein.
2. Discover polysaccharides: Chitins and chitosans. Last accessed on 2nd July 2015. http://polysac3db.
cermav.cnrs.fr/discover_chitins_chitosans.html, and references cited therein.
3. Teng, D. From chitin to chitosan. In: Chitosan‐Based Hydrogels: Taylor & Francis group, 2012, 1–33
and references cited therein.
4. Berezina, N. Production and application of chitin. In Biomaterials: Biological Production of Fuels and
Chemicals, De Gruyter, 2016, 61–72.
5. Nwe, N., Furuike, T., and Tamura, H. Chitin and chitosan from terrestrial organisms. In: Chitin,
Chitosan, Oligosaccharides and Their Derivatives. CRC Press, 2010, 3–10.
6. Abdou, E.S., Nagy, K.S.A., and Elsabee, M.Z. Extraction and characterization of chitin and chitosan
from local sources. Bioresour. Technol., 2008, 99, 1359–1367.
7. Gervasi, O., Jeuniaux, C., and Dauby, P. Production de chitine par les crustacés du zooplancton de la
baie de Calvie (Corse). IFREMER: Actes de colloques, 1988, 8, 33–8 in French.
486 Chitin and Chitosan: Properties and Applications
8. Hajji, S., Younes, I., Ghorbel‐Bellaaj, O., Hajj, R., Rinaudo, M., Nasri, M., and Jellouli, K. Structural
differences between chitin and chitosan extracted from three different marine sources. Int. J. Biol.
Macromol., 2014, 65, 298–306.
9. Rhazi, M. and Desbrieres, J. Investigation of different natural sources of chitin: Influence of the source
and deacetylation process on the physicochemical characteristics of chitosan. Polymer Int., 2000, 49,
337–344.
10. Thomas, S., Durand, D., Chassenieux, C., and Jyotishkumar, P. Handbook of Biopolymer‐Based
Materials: From Blends and Composites to Gels and Complex Networks. John Wiley & Sons, 2013.
11. Merzendorfer, H. and Zimoch, L. Chitin metabolism in insects: Structure function and regulation of
chitin synthases and chitinases. J. Exp. Biol., 2003, 206, 4393–4412.
12. Charvolin, J. and Sadoc, J.F. About collagen, a tribute to Yves Bouligand. Interface Focus., 2012,
1–8.
13. Raabe, D., Romano, P., Sachs, C., Al‐Sawalmih, A., Brokmeier, H.G., Yi, S.B., Servos, G., and
Hartwig, H.G. Discovery of a honeycomb structure in the twisted plywood patterns of fibrous biologi-
cal nanocomposite tissue. J. Cryst. Growth, 2005, 283, 1–7.
14. Chen, P.Y., Lin, A.Y.M., McKittrick, J., and Meyers, M.A. Structure and mechanical properties of crab
exoskeletons. ActaBiomaerialia, 2008, 4, 587–596.
15. Sharma, V., Crneb, M., Park, J.O., and Srinivasarao, J. Bouligand structures underlie circularly polar-
ized iridescence of Scarab beetles: A closer view. Materials Today: Proc., 2014, 161–171.
16. Vazquez, J.A., Rodriguez‐Amado, I., Montemayor, M.I., Fraguas, J., del Pilar Gonzalez, M., and
Murado, M.A. Chondroitin sulfate, hyaluronic acid and chitin/chitosan production using marine waste
sources: characteristics, applications and eco‐friendly processes: A review. Marine Drugs, 2013, 11,
747–774.
17. Schiffman, J.D. and Schauer, C.L. Solid state characterization of α‐chitin from Vanessa cardui
Linnaeus wings. Mat. Sci. Eng. C, 2009, 29, 1370–1374.
18. Berezina, N., Hubert, A., Berro, F., Levon, J.‐G., Le Roux, K., Socolsky, C., Sanchez, L., and Laurent,
S. Chitin, hydrolysate and method for the production of one or more desired products from insects by
means of enzymatic hydrolysis. WO 2016108033 patent.
19. Berezina, N., Hubert, A., Berro, F., Levon, J.‐G., Le Roux, K., Socolsky, C., Sanchez, L., and Laurent,
S. Chitin, hydrolysate and method for the production of one or more desired products by means of
enzymatic hydrolysis, including pre‐treatment with an oxidising agent. WO 2016108034 patent.
20. Berezina, N., Hubert, A., Berro, F., Levon, J.‐G., le Roux, K., Socolsky, C., Sanchez, L., and Laurent,
S. Chitin, hydrolysate and production of at least one desired product from insects by means of enzy-
matic hydrolysis, comprising a combination of steps performed prior to the enzymatic hydrolysis. WO
2016108035 patent.
21. Shahidi, F. and Synowiecki, J. Isolation and characterization of nutrients and value‐added products
from snow crab (Chionoecetes opilio) and shrimp (Pandalus borealis) processing discards. J. Agric.
Food Chem., 1991, 39, 1527–1532.
22. Rodde, R.H., Einbu, A., and Varum, K.M. A seasonal study of the chemical composition and chitin
quality of shrimp shells obtained from northern shrimp (Pandalus borealis). Carbohydr. Polym., 2008,
71, 388–393.
23. Le Roux, K., Bergé, J.P., Baron, R., Leroy, E., and Arhliass, A. Extraction de chitines en une seule
étape par hydrolyse enzymatique en milieu acide. WO2012168618 patent.
24. Rinaudo, M. Chitin and chitosan: Properties and applications. Prog. Polym. Sci., 2006, 31, 603–632.
25. Plassard, C., Mousan, D., and Salsac, L. Dosage de la chitine sur des ectomycorhizes de pin maritime
(Pinus pinaster) à Pisolithus tinctorius: évaluation de la masse mycélienne et de la mycorhization.
Can. J. Bot., 1983, 61, 692–699 in French.
26. Lavall, R., Assis, O.B.G., and Campana‐Filho, S.P. β‐Chitin from the pens of Loligo sp.: Extraction
and characterization. Bioresour. Technol., 2007, 98, 2465–2472.
Marketing and Regulations of Chitin and Chitosan from Insects 487
27. Xu, Y., Gallert, C., and Winter, J. Chitin purification from shrimp wastes by microbial deproteination
and decalcification. Environ. Biotechnol., 2008, 79, 687–697.
28. Duarte de Holanda, H. and Netto, F.M. Recovery of components from shrimp (Xiphopenaeus kroyeri)
processing waste by enzymatic hydrolysis. J Food Sci., 2006, 71, 298–303.
29. Synowiecki, J. and Al‐Khateeb, N.A.A.Q. The recovery of protein hydrolysate during enzymatic
isolation of chitin from shrimp Crangon crangon processing discards. Food Chem., 2000, 68,
147–152.
30. Jeuniaux, C., Voss‐Foucart, M.F., and Bussers, J.C. La production de chitine par les crustacés dans les
écosystèmes marins. Aquat. Living Resour., 1993, 6, 331–341 in French.
31. Zhu, A. and Shen, J. Bioactivities of chitosan and its derivatives. In: Chitosan‐Based Hydrogels. Taylor
& Francis, 2012, 109–173.
32. Kim, K.W. and Thomas, R.L. Antioxidative activity of chitosans with varying molecular weights. Food
Chem., 2007, 101, 308–313.
33. Park, P.J., Koppula, S., and Kim, S.K. Antioxidative activity of chitosan, chitooligosaccharides and
their derivatives. Chitin, Chitosan, Oligosaccharides and Their Derivatives. CRC Press, 2010,
241–250.
34. Shepherd, R., Reader, S., and Falshaw, A. Chitosan functional properties. Glyconjug. J., 1997, 14,
535–542.
35. Koide, S.S. Chitin–chitosan: Properties, benefits and risks. Nutr. Res., 1998, 18, 1091–1101.
36. Khoushab, F. and Yamabhai, M. Chitin research revisited. Mar. Drugs, 2010, 8, 1988–2012.
37. Kumar, M.N.V.R. A review of chitin and chitosan applications. React. Funct. Polym., 2000, 46, 1–27.
38. Aranaz, I., Mengibar, M., Acosta, N., and Heras, A. Chitosan: A natural polymer with potential indus-
trial applications. Science Vision, 21, 41–50.
39. Aranaz, I., Acosta, N., Civera, C., Elozra, B., Mingo, J., Castro, C., de los Llanos Gandia, M., and
Caballero, A.H. Cosmetic and cosmeceutical applications of chitin, chitosan and their derivatives.
Polymers, 2018, 10, 1–25.
40. Khor, H. and Wan, A.C.A. Chitin: Fulfilling a Biomaterials Promise. Elsevier Insights, 2nd ed., 2014.
41. Tronsmo, A., Skaugrud, O., and Harman, G.E. Use of chitin and chitosan in biological control of plant
diseases. In: Chitin Enzymology. Eur. Chitin Soc., 1993, 265–270.
42. Kato, J., Shimura, Y., Sogitani, M., Torii, H., Nakajima, K., and Oishi, K. Physiological role of chi-
tinase and chitin‐binding lectin in cucumber. In: Chitin Enzymology. Eur. Chitin Soc., 1993,
257–264.
43. Lienart, Y., Gautier, C., Dubois‐Dauphin, R., and Domard, A. Tetramers of chitin (chitosan) as elicitors
in Rubus protoplasts. In: Chitin Enzymology. Eur. Chitin Soc., 1993, 271–276.
44. Du Jardin, P. Plant biostimulants: Definition, concept, main categories and regulation. Scientia
Horticulturae, 2015, 196, 3–14.
45. Crini, G. Recent developments in polysaccharide‐based materials used as adsorbents in wastewater
treatment. Prog. Polym. Sci., 2005, 30, 38–70.
46. Crini, G. Non‐conventional low‐cost adsorbents for dye removal: A review. Bioresour. Technol., 2006,
97, 1061–1085
47. Renault, F., Sancey, B., Charles, J., Morin‐Crini, N., Badot, P.M., Winterton, P., and Crini, G. Chitosan
flocculation of cardboard‐mill secondary biological wastewater. Chem. Eng. J., 2009, 155, 775–783.
48. Nadarajah, K., Prinyawtwatkui, W., No, H.K., Sathivel, S., and Xu, Z. Sorption behavior of crawfish
chitosan films as affected by chitosan extraction processes and solvent types. J Food Sci., 2006, 71,
33–39.
49. El Ghaouth, A., Arul, J., Grenier, J., and Asselin, A. Antifungal activity of chitosan on two postharvest
pathogens of strawberry fruits. Phytopathology, 1992, 82, 398–402.
50. Parisi, S. Antimicrobials in food today and the role of chitosan – current hopes and new perspectives.
Glob. Drugs Therap., 2017, 2.
488 Chitin and Chitosan: Properties and Applications
51. Eleftheriadou, M., Pyrgiotakis, G., and Demokritou, P. Nanotechnology to the rescue: Using nano‐
enabled approaches in microbiological food safety and quality. Curr. Opin. Biotechnol., 2017, 44,
87–93.
52. Alishahi, A., Proulx, J., and Aider, M. Chitosan as bio‐based nanocomposite in seafood industry and
aquaculture. In Seafood Science: Advances in Chemistry, Technology and Applications. CRC Press,
Boca Raton (Fl, USA), 2015, 211–231.
53. Restuccia, D., Spizzirri, U.G., Parisi, O.I., Cirillo, G., Curcio, M., Iemma, F., Puoci, F., Vinci, G., and
Picci, N. new EU regulation aspects and global market of active and intelligent packaging for food
industry applications. Food Control, 2010, 21, 1425–1435.
54. Valdes, A., Melinas, A.C., Ramos, M., Garrigos, M.C., and Jimenez, A. Natural additives and agricul-
tural wastes in biopolymer formulations for food processing. Front. Chem., 2014, 2, 1–10.
55. Leduc, F., Dez, I., Desbrieres, J., Picton, L., and Madec, J.P. Different ways for grafting ester deriva-
tives of poly(ethylene glycol) onto chitosan: Related characteristics and potential properties. Polymer,
2005, 46, 639–651.
56. Leduc, F., Dez, I., and Madec, J.P. NMR study of the phosphonomethylation reaction on chitosan.
Polymer, 2005, 46, 319–325.
57. Leduc, F., Dez, I., Gulea, M., Madec, J.P., and Jaffres, P.A. Synthesis of phosphorus‐containing chi-
tosan derivatives. In: Phosphorus, Sulfur, and Silicon. Taylor & Francis, 2009, 872–889.
58. Jouanin, C., Vincent, C., Dez, I., Gaumont, A.C., Vincent, T., and Guibal, E. Highly porous cata-
lytic materials with Pd and ionic liquid supported on chitosan. J. Appl. Polym. Sci., 2013, 218,
3122–3130.
59. Moucel, R., Perrigaud, K., Goupil, J.M., Madec, P.J., Marinel, S., Guibal, E., Gaumont, A.C., and Dez,
I. Importance of the conditioning of the chitosan support in a catalyst‐containing ionic liquid phase
immobilized on chitosan: The palladium‐catalysed alkylation reaction case. Adv. Synth. Catal., 2010,
352, 433–439.
60. Clousier, N., Moucel, R., Naik, P., Madec, P.J., Gaumont, A.C., and Dez, I. Catalytic materials based
on catalyst containing ionic liquid phase supported on chitosan or alginate: Importance of the support.
Comptes Rendus Chimie, 2011, 14, 680–684.
61. Baudoux, J., Perrigaud, K., Madec, P.J., Gaumont, A.C., and Dez, I. Development of new SILP cata-
lysts using chitosan as support. Green Chem., 2007, 9, 1346–1351.
62. Muzzarelli, R.A.A. Chitosan scaffolds for bone regeneration. Chitin, Chitosan, Oligosaccharides and
Their Derivatives. CRC Press, 2010, 223–239.
63. Khor, E. and Lim, L.Y. Implantable applications of chitin and chitosan. Biomaterials, 2003, 24,
2339–2349.
64. Kim, M.M. and Kim, S.K. Anti‐inflammatory activity of chitin, chitosan, and their derivatives. In:
Chitin, Chitosan, Oligosaccharides and Their Derivatives. CRC Press, 2010, 215–221.
65. Kim, K. and Ji, H.S. Effect of chitin sources on production of chitinase and chitosanase by Streptomyces
griseus HUT 6037. Biotechnol. Bioprocess Eng., 2001, 6, 18–24.
66. Je, J.Y. and Ahn, C.B. Antihypertensive actions of chitosan and its derivatives. In: Chitin, Chitosan,
Oligosaccharides and Their Derivatives. CRC Press, 2010, 263–270.
67. Ta, H.T., Dunstan, D.E., and Dass, C.R. Anticancer activity and therapeutic applications of chitosan
nanoparticles. In: Chitin, Chitosan, Oligosaccharides and Their Derivatives. CRC Press, 2010,
271–284.
68. Canepa, C., Imperiale, J.C., Berini, C.A., Lewicki, M., Sosnik, A., and Biglione, M.M. Development
of a drug delivery system based on chitosan nanoparticles for oral administration of interferon α.
Biomacromolecules, 2017, 18, 3302–3309: 7
69. Pereira Gomes, C., Ferreira Lopes, C.D., Duarte Moreno, P.M., Varela‐Moreira, A., Alonso, M.J., and
Pego, A.P. Translating chitosan to clinical delivery of nucleic acid‐based drugs. Mat. Res. Society Bull.,
2014, 39, 60–71.
Marketing and Regulations of Chitin and Chitosan from Insects 489
70. Martelli, S.M., Lin, C.S.K., Sun, Z., Berezina, N., Fakhouri, F.M., and Innocentini‐Mei, L.H. Natural
rubber blends with biopolymers. In: Natural Rubber Materials: Volume 1: Blends and IPNs, RSC,
2013, 1, 370–393.
71. Berezina, N. and Martelli, S.M. Polyhydroxyalkanoates: Structure, properties and sources. In:
Polyhydroxyalkanoate (PHA) based blends, composites and nanocomposites; RSC Green Chemistry
N°30, 2015, 18–46.
72. Adjou, K., Mammeri M., Grasset‐Chevillot, A., Marden, J.P., Auclair, E., Mage, C., and Vallée, I.
Maitrise de la cryptosporidiose avec le chitosan: essi in vivo. Proceedings: Journéesnationales
DTV – Nantes 2015, 377–80 in French.
73. Gasco, L., Finke, M., and van Huis, A. Can diets containing insects promote animal health? J. Insects
Food Feed, 2018, 4, 1–4.
Index
Exo‐activity, 191, 194, 209, 211 packaging, 175, 321, 324, 350, 358–359, 483
Exo‐enzyme, 195 preservation, 175, 234, 324, 353, 362, 371–372
Exo‐β‐glucosaminidases, 190, 195–197, 237 products, 174–175, 316, 353, 362
Exoskeleton, 1, 5, 9–10, 212, 291, 315, 352, 372, quality, 174, 349
477–478, 480 shelf life, 174, 324, 357
Eye, 269–270 spoilage, 174–175, 338, 355
waste, 176
Fabrics, 176–177, 396, 398, 400, 413–414, Food and Drug Administration (FDA), 261, 292,
417–418 316, 351
cellulose, 176 Formulations
cotton, 176, 398, 416, 418 chitosan, 179, 376, 380–381, 417
polyester, 176 drug delivery, 261
textile, 395, 400–401, 414 feed, 484
Failure Mode and Effect Analysis (FMEA), 307 macroscopic, 261
Fats, 160, 406, 480 microscopic, 261
Fatty acids, 175, 302, 319, 321, 381, 412 nanoscopic, 261
Feed, 12, 230, 338 pharmaceutical, 259–261
formulation, 484 Fossil fuels, 230, 349, 397
side, 446–447 Fourier‐transform infrared (FT‐IR) spectroscopy,
Ferric ions, 119–120 5, 67, 481
Fertilizer, 230, 483 Fractionation, 89, 232
foliar, 180 Fraction of acetylation, 81, 83, 87, 91
release, 133 Fragmentation, 84–86
synthetic, 177 Free radicals, 74, 114, 252, 296, 318, 320
Fibers, 2, 40, 43, 48, 303–304, 395–396, 398–406, Functional material, 176, 357, 472
410, 412, 419–420, 480 Fungal
Filling material, 173, 305 biomass, 10, 16, 24, 26
Film, 61, 305, 322–323, 353–354, 357–359, 362, cell wall, 10, 207–208, 352, 372
407–408, 410–412, 464–465 lung inflammation, 146, 149
antimicrobial properties, 170, 174, 296, 319, mycelium, 25–26
323, 359–360, 381 pathogens, 147, 177–180, 207, 214
biodegradable, 322, 324, 357 Fungi, 5, 10, 25, 28, 39, 169, 177–179, 207–208,
edible, 174–175, 322, 349–353, 357–360, 213, 375, 419
363, 372 chitosan extraction, 24–25
forming property, 406–407, 410, 462, 465, 470 filamentous, 169–170
gas permeability, 173 Fungicidal activity, 155, 171, 372, 375
hydrophilic properties, 173 Fungine, 1, 477
Fingerprinting, 87, 92 Furan‐based monomers, 229, 238
First derivative of UV spectrum, 69–70 2,5‐Furan dicarboxylic acid (FDCA), 238
Fish, 35, 155–156, 174–175, 291, 352, 362–364, 380
Flame retardation, 398, 400, 415 Gallic acid, 250, 318, 362, 380
Flocculant, 214, 444–445 Gastrointestinal
Flocculation, 408, 429–430, 432, 440, 444–445 tissues, 265
Foam, 301–302, 305, 414 tract, 320, 332, 334, 339
Food, 174–175, 230, 315–316, 318, 322, 324, Gel, 99, 108–109, 111–112, 119–122, 125, 134,
334, 338 172, 275, 297, 410
additive, 315–316, 483 colloid, 464
freshness, 174 core, 100
industry, 174–175, 214, 230, 232, 235, 316, cross‐linked, 120
318, 357 formation, 108, 112
500 Index
Laccase, 250, 252, 318 Lymphocytes, 147–148, 153, 156, 158, 360
Lacrimal clearance, 270 Lymphopoietin, 146
Lactic acid, 320 Lyophilization, 302, 304–305
bacteria, 14 Lysis
biosensor, 332 cell, 155, 170, 355
fermentation, 39, 239 sponge cells, 45
Layer‐by‐layer Lysozyme, 74, 116, 127
assembly, 101 activity, 208
deposition, 261 Lytic polysaccharide monoxygenase (LMPOs),
formation, 416 189–190, 199–200, 204–205, 207–209,
technique, 354, 465 211, 213, 216, 236–237
Lead (Pb), 71, 301, 338, 418, 439, 469
Leakage Macromolecular
cell, 319 chain, 125, 263
cell membrane, 170 cross‐linkers, 112, 135
Leucocyte, 155, 158, 163 network, 110
activity, 155 radical, 318
responses, 155 Macrophage, 146–148, 155–157, 263
Levulinic acid, 232, 235, 238, 240 Maillard reaction, 316, 338
Lewis base liquid, 252 Mannose receptor, 147–148
Liesegang rings, 99 Manufacturing
Lignin, 180, 189, 230 film, 464
Lignocellulosic biomass, 215, 230, 238 medicinal products, 292
Linear process, 292–293, 295
aggregates, 246, 462 textile, 396, 398, 435
backbone, 251 Mark–Houwink equation, 66–67
macromolecules, 399 Mark–Houwink–Sakurada equation, 63
polysaccharide, 40, 98, 103, 232 Mass spectrometric tools, 76, 81
potentiometric titration, 69–70 Mass spectrometry, 83–85
Lipase, 75, 332, 481 Mast cell, 157
Lipid, 230, 266, 355, 376 Mathematical models, 261, 276
bilayers, 266 Matrix assisted laser desorption time of flight mass
composition, 160 spectrometry, 76
layer, 270 Meat, 362
nanocarriers, 271 Mechanical
oxidation, 316, 362–363 properties, 107, 110, 120, 125, 136, 322, 354,
peroxidation, 175, 318, 320–321 358, 472
Lipopolysaccharides, 148–149, 162, 170, 319 fibers/yarns, 404–406
Lobster, 23, 38, 479 films, 411–412
Loops, 191, 193, 199, 201–202, 420 strength, 102, 110, 119, 125, 176, 304
Low‐density lipoprotein (LDL), 61, 158, 160, Medical
162, 321 applications, 71, 171, 214, 292, 308
Lower critical solution temperature, 110 devices, 72, 291–297, 301, 306–308, 484
Low molecular fabrics, 398
chitosan, 75, 153, 158, 170, 215, 261, 319 textiles, 176
weight, 175, 265, 269, 320, 408, 446, 450 Membrane, 259, 276, 305, 447
Lowry method, 73 biological, 177, 266
Lymph blend, 448–450, 452
node‐, 156 cell, 170, 179, 271, 319, 398
stream, 265 cellular, 265–266, 320
504 Index
Ultrafiltration, 75, 429, 447, 450–452 solubility, 98, 103, 234–235, 250, 359, 450
membrane, 432, 446, 448–450 soluble polymers, 251, 450
polymer‐enhanced, 450–451 treatment, 232, 234, 237, 430, 450,
process, 432, 446–447, 452 483, 485
Ultrasonic uptake, 132–133, 135–136, 412, 466
treatment, 45 vapour, 307
waves, 45, 417 permeability, 174, 377–378
Ultrasonication, 237 resistance, 179, 380
Ultrasound, 45–46, 54, 126–127, 271, 359 Wear resistance, 102, 415
UV‐irradiation, 275, 376 Weaving, 400, 419, 421
Weight
Vacuum packaging, 175, 363 average, 86
Validation, 149, 292–293, 297, 307–308 electrophoretic mobility, 89
Vapour permeability, 304 molar mass, 63
Verification, 292–294, 297, 307–308 loss, 158, 163–164, 234, 361, 376–382, 384
Verongula gigantea, 36, 43 Well‐being, 276
Vinyl monomers, 132, 136 Wetting, 407, 410
Virulence, 156–157 Wide‐angle X‐ray scattering, 76
factor, 207–208 Wine, 175, 235, 316, 338
Viruses, 76–77, 177–178, 198–199 Wound
Viscometer, 66, 70 dressing, 72, 171, 241, 292–297, 301–308, 398,
Viscose, 399 417, 471
fibres, 404 gel, 297
Viscosimetry, 86 nonwoven, 297
Viscosity, 66–70, 194–195, 408, 410, 447 powder, 297
average, 86 sponge/foam, 297
molecular weight, 86 healing, 145, 171–172, 174, 275, 291,
number, 66–67 297, 301–302, 305–306, 398,
471, 484
Wastewater, 28, 214, 232, 364, 400, 413–414, treatment, 174, 261, 306
418, 429–430, 434–435, 437, 440, 445,
450–452 X‐ray diffraction, 87, 481
synthetic, 439–440 Xylanase, 75
treatment, 136, 408, 430, 432, 444, 452
adsorption, 430, 432–433 Yarn, 399, 401, 404
coagulation/flocculation, 430, 440, blend, 400, 406
443–444 Yeast, 10, 35, 307, 361–362, 376
membrane separation, 430, 446, 452
Water Zero‐order release kinetics, 260
consumption, 179 Zero‐waste circular economy, 230
WILEY END USER LICENSE AGREE-
MENT
Go to www.wiley.com/go/eula to access Wiley’s ebook EULA.