Dairy Microbiology Ebook Foodkida 1660321488
Dairy Microbiology Ebook Foodkida 1660321488
Dairy Microbiology Ebook Foodkida 1660321488
Dairy
Dairy
Dairy
Dairy
Dairy
Dairy Microbiology
Dairy Microbiology
The objective of this book is to provide a scientific
Microbiology
background
TheTheobjective
objective toofdairy
The objective ofthis microbiology
thisof
book this isbook
book by re-examining
isprovide
to provide
isto toprovide the
a scientific
a ascientific
scientific
Microbiology
Microbiology
Microbiology
basic concepts
background of general
tomicrobiology
dairy food microbiology
microbiology and the
by re-examining thethe the
Microbiology
background
background to todairy
dairy microbiology byby re-examining
re-examining
microbiology
basic
basic basic
concepts
concepts ofof raw
concepts general
of milk
general while
of general
foodfood offering
food
microbiology
microbiology a and
practical
microbiology and and
the the the
Microbiology
approach
microbiology
microbiology to ofthe
microbiology ofraw following
raw ofmilkraw aspects:
milk
milkwhile while
whileoffering well-known
offering
offering a and
a apracticalpractical
practical
newfound
approach
approach pathogens
approach
to tothethe to
followingthatfollowing
the
following are of major
aspects:
aspects:aspects: concern
well-known toand
well-known
well-known the
andand
A Practical
dairy
newfound
newfound newfound
industry.
pathogens
pathogens pathogensthatthat that
areare of of aremajor
major of concern
major concern
concern to to to the
thethe
AAAPractical
Practical
Practical
dairy dairy industry.
industry.
dairy industry.
Topics addressed include Cronobactersakazakii and its
importance
Topics
Topics
Topics
addressed toaddressed
addressed infant
include
include
includemilk
formula Cronobactersakazakii
or Mycobacterium
Cronobactersakazakii
Cronobactersakazakii and itsand
and its
its
Approach
Approach
Approach
Approach
avium importance to
subspecies infant formula milk
paratuberculosis or that
Mycobacterium
importance
importance to toinfant
infant formula
formulamilk milkor(MAP)
orMycobacterium might
Mycobacterium
beavium avium
connected subspecies
to chronic paratuberculosis
human diseases (MAP)
(Crohn’s), that
themight
avium subspecies
subspecies paratuberculosis
paratuberculosis (MAP)
(MAP) that
thatmight
might
role of be connected
dairy starter to chronic
cultures human diseases
in manufacturing (Crohn’s), the
fermented
be connected
be connected to to
chronic
chronic human
human diseases
diseases (Crohn’s),
(Crohn’s), thethe
dairy role of
products, dairy starter
developing cultures in manufacturing fermented
role of of
role dairy
dairy starter
starter cultures
cultures in novel
in manufacturing functional
manufacturing
dairy products, developing novel functional dairy
fermenteddairy
fermented
products
dairy
dairyproducts,through developing
products, the incorporation
developing novelnoveloffunctional
probiotic
functional strains,
dairy
products through the incorporation of probioticdairystrains,
insights in the field ofincorporation
molecular methods for microbial
Editor:
products
products through
through thethe incorporation of ofprobiotic
probiotic strains,
strains,
insights in the field of molecular methods for microbial
Editor:
insights field of molecular methods microbial
Photis
the compulsory
the systemsapplication
compulsory application
(FSMS) food
of
to the dairy foodsafety
safety
industry. management
management
systems
systems (FSMS)
(FSMS) to theto the dairy industry.
dairy industry.
Photis
The book will provide dairy professionals and students
The book will provide dairy professionals and students
alike
TheThe the
bookbook latest
alike theinformation
willwill
provide
provide
latest dairy on
dairy
information this vast
professionals thistopic.
professionals
on vastand students
and
topic. students
Papademas
alike thethe
alike latest information
latest information ononthis thisvast topic.
vast topic.
Papademas
Editor
Editor
PhotisEditor
Editor
Papademas
Photis Papademas
K24428
Photis
PhotisPapademas
Papademas
6000 Broken Sound Parkway, NW
Suite 300, Boca Raton, FL 33487
711 Third Avenue
New York, NY 10017
an informa business 2 Park Square, Milton Park
w w w. c rc p r e s s . c o m Abingdon, Oxon OX14 4RN, UK 9 7814829 298673
781482 298673 A Science Publishers Book
9 781482 298673
9 781482 298673
DAIRY MICROBIOLOGY
A Practical Approach
DAIRY MICROBIOLOGY
A Practical Approach
Editor
Photis Papademas
Department of Agricultural Sciences
Biotechnology and Food Science
Cyprus University of Technology
Limassol, Cyprus
p,
A SCIENCE PUBLISHERS BOOK
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
This book contains information obtained from authentic and highly regarded sources. Reasonable efforts
have been made to publish reliable data and information, but the author and publisher cannot assume
responsibility for the validity of all materials or the consequences of their use. The authors and publishers
have attempted to trace the copyright holders of all material reproduced in this publication and apologize to
copyright holders if permission to publish in this form has not been obtained. If any copyright material has
not been acknowledged please write and let us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmit-
ted, or utilized in any form by any electronic, mechanical, or other means, now known or hereafter invented,
including photocopying, microfilming, and recording, or in any information storage or retrieval system,
without written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copyright.
com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood
Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and
registration for a variety of users. For organizations that have been granted a photocopy license by the CCC,
a separate system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used
only for identification and explanation without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
Dedication v
Preface vii
1. Basic Concepts of Food Microbiology 1
Peter Raspor, Sonja Smole Smožina and Mateja Ambrožič
2. The Microbiology of Raw Milk 22
Apostolos S. Angelidis
3. Dairy Pathogens: Characteristics and Impact 69
Photis Papademas and Maria Aspri
4. Dairy Starter Cultures 114
Thomas Bintsis and Antonis Athanasoulas
5. Application of Probiotics in the Dairy Industry: 155
The Long Way from Traditional to Novel Functional Foods
Adele Costabile and Simone Maccaferri
6. Application of Molecular Methods for Microbial 177
Identification in Dairy Products
Paul A. Lawson and Dimitris Tsaltas
7. Application of Food Safety Management Systems (FSMS) 217
in the Dairy Industry
Thomas Bintsis
Index 237
Color Plate Section 239
CHAPTER 1
INTRODUCTION
Food has never been safer than it is today, but at the same time it has also
never been at greater risk. Concerns over food safety and quality as well as
its production, processing and preservation have enhanced the importance
of food microbiology as it encompasses the study of microorganisms which
can have beneficial as well as harmful effects on the quality and safety of
food. Food microbiology has begun to implement all necessary precautions
to improve quality and safety of food products. The milk industry was one
of the first to use food microbiology to select relevant starter cultures, to
eliminate spoilage and reduce zoonoses, which were transmitted along
food supply chain.
It is important to stress that milk production and processing was on the
front lines of implementation of technical and regulatory novelties like novel
process design, innovative equipment with most advanced pasteurization
and sterilisation concepts, but also with variety of products that had more
1
Vice Dean for Research and International Cooperation, Faculty of Health Sciences, University
of Primorska, Head of the Institute for Food, Nutrition and Health, Polje 42 SI – 6310, Izola,
Slovenia.
Email: [email protected]
2
Chair of Microbiology, Biotechnology and Food Safety, Department of Food Science and
Technology, Biotechnical Faculty, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
Emails: [email protected]; [email protected]
* Corresponding author
2 Dairy Microbiology: A Practical Approach
Starter Cultures
In the middle of the 19th century, Pasteur and others finally demonstrated
that microorganisms cause food spoilage. Thousands of years before this
sensational experimental proof, man had developed actions for preventing
spoilage based on the use of experiences to identify suitable methods to
prevent this spoilage. Raw milk in its natural state is highly perishable
because it is susceptible to spoilage from naturally occurring enzymes and
Basic Concepts of Food Microbiology 5
On one side finding the right culture for the right application often includes
the simplification of a process or adding an attribute value to the consumer,
who is the last link in food supply chain. Optimal speed of acidification,
reaching target viscosity, developing a new flavour profile or safety of
the final dairy product are just some issues concerning added values in
production lines. Bacteriophages and their impact on starter cultures
are still a challenge for milk starter developers. On the other side novel
starter technology concept is eliminating the huge natural biodiversity of
microorganisms and consequently also products, which were offered by
traditional technologies. The future activities on the production side are
challenged with the issue of how to preserve the natural heritage of many
dairy products in this regard.
Milk-borne Pathogens
Since ancient time milk and products derived from milk have been part
of the human diet. Milk is a complex source of nutrients which includes
protein, carbohydrate, lipid, vitamins and minerals. The components that
make it nutritious for humans also provide ideal growth medium for
producing both beneficial and detrimental microorganisms.
Basic Concepts of Food Microbiology 7
(GMP), good hygiene practice (GHP), hazard analysis and critical control
points (HACCP) and including private standards as well. Quality assurance
is a term used for describing the control, evaluation and audit of a food
processing system.
Food safety defined according to Codex Alimentarius as assurance
that food will not cause harm to the consumer when it is prepared and/
or eaten according to its intended use (Codex Alimentarius Commission/
RCP 1 2003) is an international challenge requiring close cooperation
between producers, processors, retailers and consumers on the one side and
governmental and nongovernmental organisations on the other side. There
are many factors affecting food safety such as global trade, socio-economic
and technological development, urbanization and agricultural land use. In
a food safety program we should be able to identify all hazards, analyse
them, assess them, assess the likelihood of their occurrence and identify
measures to their control. Hazard is a biological, chemical or physical
agent in, or condition of, food with the potential to cause an adverse health
effect (Codex Alimentarius Commission 2003). The aim of HACCP based
systems is to ensure that food is produced safely. HACCP is a tool which
identifies, evaluates, and controls hazards throughout the food chain from
primary production stages to final consumption which are significant for
food safety (Codex Alimentarius Commission 2003). It assesses hazards
and establishes control systems that focus on prevention rather than relying
mainly on end-product testing. A HACCP plan proves that the controls are
in place and that the system is functioning effectively (Food and Agriculture
Organization 1998).
In spite of major advances in science and technology, microbial food-
borne illnesses are considered a significant public health issue (European
Food Safety Authority 2012). To provide safety, stability and quality of food
products detection of microbial contamination is therefore important. The
spectrum of food-borne infections has been changing over time, as well
established pathogens have been controlled or eliminated (Tauxe 2002) and
new ones have emerged because of the changed consumer eating habits and
patterns. In most cases food-borne diseases are associated with diarrhoea,
vomiting, other gastrointestinal and/or extra intestinal manifestations, but
secondary complications can occur. Managing the microbiological food
safety risk with the goal of reducing the burden of microbial food-borne
illnesses is still one of the important challenges today due to the fact that
factors affecting the microbiological food safety are changing dramatically.
Over time we have been witnessed to rapid and huge technological changes
in food processing and production procedures and also due to the changes
in methods of microbiological analysis.
Microbiological criteria are tools that can be used in assessing the safety
and quality of foods. They are necessary to assist in setting critical limits in
Basic Concepts of Food Microbiology 11
Consumers in developed countries ask for food products that are high and
consistent in quality, possible to get in broad assortments throughout the
year and also for competitive prices. All food producers and processors are
responsible for quality products and to guarantee safety of foods. Collecting
products from animals leads to transfer of microorganisms from animals
to those products. Contamination of milk may also arise at any stage of
milking and also during the later stages such as farm storage, transport and
processing. That is why milk and milk products present a unique challenge
for food safety, because numerous microorganisms including bacteria,
12 Dairy Microbiology: A Practical Approach
yeasts and moulds constitute the complex ecosystem present in milk and
fermented dairy products. Beside microbiological hazards chemical hazards
also present hazards to the public health, because toxic chemicals present in
animals’ bodies can be shed into the milk. Chemical contaminants include
industrially derived contaminants like dioxins, furans, PCBs and elemental
heavy metals; biologically derived contaminants like mycotoxins and
phytotoxins; pesticides and residues of plant agrichemicals like pesticides,
and residues of animal remedies (O’Mahoney et al. 2009).
At this point climate changes should also be mentioned because it
may have an impact on the occurrence of food safety hazards at various
stages of any food chain, not just the milk supply chain. Climate change
is widely recognized globally as the major environmental problem to be
faced. There are multiple pathways through which climate related factors
may impact food safety, such as changes in temperature, extreme weather
events and others. Climate change may also affect aspects related to food
safety systems such as agriculture, animal production and trade (Tirado
et al. 2010). The experts selected feed related issues like raw materials,
pasture, silage, storage and manufacturing feed and also animal health as
the most critical factors that affect the occurrence of food safety hazards
due to climate change (van der Spiegel et al. 2012).
An increasing number of people are consuming raw unpasteurized
milk although numerous epidemiological studies have shown clearly that
raw milk can be contaminated by a variety of detrimental microorganisms
(Oliver et al. 2009). Reasons for increased interest in raw milk consumption
is seen in enhanced nutritional qualities and consequently in health benefits.
However, pasteurization reduces spoilage and eliminates pathogens.
Spoilage microorganisms (sporoforms) in raw milk cannot be completely
eliminated and growth can take place readily. For this reason shelf life of
pasteurized milk, even when refrigerated, is limited. In many countries sale
of raw milk for direct consumption is restricted or completely prohibited
because of the potential risk to public health (LeJeune and Rajala-Schultz
2009) but a small amount of milk is still sold as raw or unpasteurized. In
conventional distribution two main heat treatments, pasteurization and
sterilisation are used for milk sold in the retail sector. The main aims of
heat treatment developed for market milks are to eliminate or reduce
the microbial population associated with pathogens, potential spoilage
organisms and degradative enzymes (Lewis and Deeth 2009). A very good
microbial quality of raw milk is also important to prevent production
losses and to achieve an optimal shelf life of dairy products. To ensure a
good microbial quality of milk, quality assurance systems of dairy farms
are being developed and bacteriological schemes are being implemented.
Many aspects of food safety and quality management are involved in the
control of the microbial contamination, especially on farms as main areas
Basic Concepts of Food Microbiology 13
global livestock industry. Global climate change is also challenging the dairy
industry to develop sustainable initiatives to reduce their environmental
impact. Carbon footprint has become a widely used term and concept of
global climate change. Carbon footprint by its definition is the total amount
of greenhouse gas (GHG) emissions associated with a product, along its
supply chain, and sometimes includes emissions from consumption, end of
life recovery and disposal. It is usually expressed in kilograms or tonnes of
carbon dioxide equivalent (Food and Agriculture Organization 2010). FAO
reported (Food and Agriculture Organization 2010) that the dairy sector in
2007 emitted 1,969 million tonnes CO2-eq of which 1,328 million tonnes are
attributed directly to milk. Recent studies have estimated that cradle-to-
farm gate emissions of milk globally contribute 4.0% of global greenhouse
gas emissions such as methane, nitrous dioxide and carbon dioxide. The
overall contribution of the global milk production, the processing and
transportation to global GHG emissions is estimated at 2.7%. Or put in
another way the global average of GHG emissions per kilogram of milk
and related milk products is estimated at 2.4 kg CO2 equivalent. This
estimation includes emissions associated with milk production, processing
and transportation of milk and milk products only. A significant source of
emissions in the dairy supply chain is methane, produced from the natural
digestive process of cows. Nitrous oxide and carbon dioxide are also by-
products of dairy production.
Dairy industry worldwide has engaged to lower the GHG emissions in
next key areas (Global Dairy Agenda for Action 2009):
• Emissions reduction (i.e., optimising animal feeding, optimising use
of resources and optimising manure management).
• Energy efficiency (i.e., on farm energy use in milking and refrigeration,
optimised processing, renewable energy).
• Transport efficiency (i.e., optimised milk collections, optimised
transport and distribution).
• Reduction in loss of milk and milk products (i.e., shelf life improvement,
reduce household waste).
• Resource efficiency (i.e., recycling of packaging, increase recovery of
waste, use of packaging with lowest environmental impact).
• Management (i.e., development of a global standard for measuring
monitoring and reducing GHG emissions).
Transport is highly related to almost all human activities. It stimulates
economy, improves people’s wellbeing and comfort, but at the same
time contributes to negative environmental impacts such as pollution,
depletion of ozone layers, depletion of resources, global warming, waste,
noise, vibration, barriers and congestion in populated areas (Hesse and
Basic Concepts of Food Microbiology 15
Rodrigue 2004, Anderson et al. 2005, Russo and Comi 2010). Food reaches
our plates nowadays via different routes and the present logistic challenge
for food suppliers. Transportation and distribution are one of the most
visible elements of our current food system. We can see trucks labelled
with commercials of food giants on our roads every day. Transport of food
and agricultural produce is a significant component of goods transport as a
whole (Gebresenbet et al. 2011). Considering the importance of maintaining
the food supply system and reducing the environmental impact of the
transport system, increasing the efficiency of food distribution appears
to be a major challenge not only in the dairy sector, but in the food sector
in general. The transportation of foodstuffs is having a considerable
environmental impact through fuel used for transportation and energy
used for refrigeration. According to EUROSTAT statistics road plays a
predominant role in European countries in both passengers and goods
transportation. 46% of total goods required for transportation are done so
by roads. Road transports consumed 26% of total final energy consumption
of the 27 member countries of European Union. Statisatics showed that
road transportations are responsible for emitting 93% of greenhouse gases
of total European transport (Statistical Office of European Communities
2009). Road transportation of agricultural products and foodstuffs is largely
a national operation, where majority of goods were transported less than
150 km, although certain foods are moved and delivered over considerable
distances by road (Statistical Office of European Communities 2011).
Since the dairy industry concentrate their primary production and
processing facilities we cannot ignore the fact that they substantially
contribute to carbon footprint. In addition to this, food transport refrigeration
is a critical link in the food chain, not only in terms of maintaining the
temperature integrity, but also its impact on energy consumption and CO2
emissions (Tassou et al. 2009). Refrigerated storage is one of the most widely
practiced methods of preserving perishable foods like dairy products. It is
important in maintaining the safety and quality of many perishable foods
and it enables that food is supplied to their destination. Refrigeration
stops or reduces the rate at which changes occur in food. These changes
can be microbiological (i.e., growth of microorganisms), physiological (i.e.,
ripening, respiration), biochemical (i.e., browning reactions, lipid oxidation
and pigment degradation) and/or physical (i.e., moisture loss). An efficient
and effective cold chain is designed to prove the best conditions for slowing
and preventing these changes for as long as it is practical (James and James
2010), but the data suggest that currently the cold-chain accounts for
approximately 1% of CO2 production in the world. The cold chain is vital
part of modern global trade as it impacts on all food commodities.
16 Dairy Microbiology: A Practical Approach
Union (EU) law, 105 bacteria ml−1 at this point of the process are the critical
limits enforced (EC 2004).
The availability and application of culture-independent tools that
enable a detailed investigation of the microflora and microbial biodiversity
of food systems has had a major impact. However, it has become apparent
that approaches that include a culturing step can lead to inaccuracies due
to species present in low numbers being outcompeted in laboratory media
by numerically more abundant microbial species (Hugenholtz et al. 1998)
or the fact that others may simply not be amenable to cultivation in the
laboratory (Head et al. 1998). For these reasons approaches to access the
microbial composition of food have had to change dramatically. To address
this, there has been an increased focus in recent year on the use of culture
independent investigations through the direct analysis of DNA or RNA
from food without a culturing step (Quigley et al. 2011). The shift from
culture dependent assessment to culture independent analysis has led to a
revolution in food microbiology.
Advent of biotechnology has greatly altered food testing methods.
Improvements in the field of immunology, molecular biology, and
automation and computer technology continue to have a positive effect
on the development of faster, more sensitive and more convenient and
reliable methods in food microbiology. Further, development of on-line
microbiology, including ATP bioluminescence and cell counting methods,
is important for rapid monitoring of cleanliness in HACCP programs.
However, the important challenge is still sample preparation. More
research is needed on techniques for separating microorganisms from the
food matrices. The possibilities of combining different rapid methods,
including immunological and DNA based methods are occurring more and
more regularly in milk microbiology. Analytical technology is improving
constantly and the current generation of assays potentially has the capability
for near real time and online monitoring of multiple pathogens. Modern
methods are based on molecular biology techniques like PCR, RFLP, DNA
microarray assay, immunological techniques like ELISA, biophysical
and biochemical principles with the application of biosensors like
bioluminescence sensor, bio-analytical sensors utilizing enzymes, electrical
impedimentry and flow cytometry (Mandal et al. 2011).
Concerns over milk safety and quality as well as production, processing
and preservation have increased the importance of food microbiology.
Raw milk is known to comprise a diverse microbial community. The high
nutritional value of raw milk, its high water content and near neutral
pH allows the growth of many, maybe even all microorganisms. These
microorganisms include microflora of technological relevance like starter
cultures as well. On the other hand the presence of spoilage bacteria can
have significant negative effects on the quality of milk and milk products,
18 Dairy Microbiology: A Practical Approach
while one of the most difficult and fundamental issues in food safety is
the detection of pathogens that can have severe effects on human health.
Current transformation of methods to fast and reliable microbiological
analytics is a real challenge since it also needs rapid improvement of the
skills and knowledge of diary/food microbiologist to be able to serve the
system in a fast and reliable way.
Conclusions
The food/milk supply chain is emerging as the most integrated system on
the globe. Milk production, processing and consumption are connected to
human nutrition requirements and the ability of current systems to keep
quality and safety of milk and milk products during assigned shelf life
periods. Novel technologies are offering efficient primary production and
secondary processing with less and less impact on quality reduction, and
at the same time promoting safety of final product to be consumed.
Substantial development was realised by environment engineering and
process design, which was efficiently underpinned with fast and reliable
analytical methods to trace physical, chemical and microbial hazards. The
development in last fifteen years is slowly fusing together novelties on
processing, technical and regulatory level. It is offering us new solutions
in safe and quality management practices which can deliver numerous
traditional and manmade novel products with the intention to serve needs
of contemporary consumer who have less knowledge about food and less
time for learning and handling basics nutritional needs.
References Cited
Accum, F. 1820. A Treatise on the Adulteration of Food and Culinary Poisons. Exhibiting the
Fraudulent Sophistications of Bread, Beer, Wine, Spirituous Liquors, Tea, Coffee, Cream,
Confectionary, Vinegar, Mustard, Pepper, Cheese, Olive Oil, Pickles and Other Articles
Employed in Domestic Economy. And Methods of Detecting Them. 2nd ed. Longman,
Hurst, Rees, Orme and Brown, London. UK.
Anaelom, N.J., O.J. Ikechukwu, E.W. Sunday and U.C. Nnaemeka. 2010. Zoonotic tuberculosis:
A review of epidemiology, clinical presentation, prevention and control. J. Public Health
Epidemiol. 2: 118–124.
Anderson, S., J. Allen and M. Brown. 2005. Urban logistics—how can it meat policy markers’
sustainability objectives? J. Transp. Geogr. 13: 71–81.
Aruoma, I.O. 2006. The impact of food regulation on the food supply chain. Toxicology 221:
119–127.
Arvanitoyannis, I.S., T.H. Varzakas and M. Koukaliaroglou-van Houwelingen. 2009. Dairy
Foods. pp. 91–180. In: I.S. Arvanitoyannis [ed.]. HACCP and ISO 22000: Application to
Foods of Animal Origin. Wiley-Blackwell. Oxford. UK.
Boor, K.J. and S.C. Murphy. 2002. Microbiology of market milks. pp. 91–122. In: R.K. Robinson
[ed.]. Dairy Microbiology Handbook. Wiley-Interscience. New York. USA.
Basic Concepts of Food Microbiology 19
Chambers, J.V. 2002. The microbiology of raw milk. pp. 39–90. In: R.K. Robinson [ed.]. Dairy
Microbiology Handbook. Wiley-Interscience, New York. USA.
Codex Alimentarius Commission. 2003. General principles of food hygiene CAC/RCP 1-1969.
FAO/WHO, Rome, 31 pp.
Codex Alimentarius Commission. 2004. Code of hygienic practice for milk and milk products
CAC/RCP 57-2004. FAO/WHO, Rome, 49 pp.
De Boer, E. and R.R. Beumer. 1999. Methodology for detection and typing of foodborne
microorganisms. Int. J. Food Microbiol. 50: 119–130.
Deak, T. 2009. Testing methods in food microbiology. pp. 99–118. In: R. Laztity [ed.]. Food
Quality and Standards Vol. III. Encyclopedia of Life Support Systems (EOLSS), Developed
under the Auspices of the UNESCO, Eolss Publishers, Oxford. UK.
Derrick, E.H. 1937. Q fever, a new fever entity: chemical features, diagnosis and laboratory
investigation. M.J. Australia 2: 281–299.
EC No 853/2004 of the European Parliament and of the Council of 29 April 2004 laying down
specific hygiene rules for food of animal origin. Off. J. Eur. Communities L 226: 22–82.
EC No 2073/2005 of the European Parliament and of the Council of 15 November 2005 on
microbiological criteria for foodstuffs. Off. J. Eur. Communities L 338: 1–26.
Eckles, C.H., W.B. Combs and H. Macy. 1936. Milk and Milk Products. McGraw-Hill, New
York. USA.
European Food Safety Authority. 2012. The European Union Summary Report on Trends and
Sources of Zoonoses, Zoonotic Agents and Food-borne Outbreaks in 2010. EFSA Journal
10: 2597: 442 pp. Available online: www.efsa.europa.eu/efsajournal.
Food and Agriculture Organization of the United Nations and International Dairy Federation.
2004. Guide to good dairy farming practice. FAO, Rome, 33 pp.
Food and Agriculture Organisation of the United Nations. 1998. Food quality and safety
systems—a training manual on food hygiene and the HACCP system. Food Quality and
Standards Service, Rome, pp. v–viii.
Food and Agriculture Organization of the United Nations. 2010. Greenhouse Gas Emissions
from the Dairy Sector. A Life Cycle Assessment. FAO, Rome, 98 pp.
Fuller, R. 1992. Probiotics. The scientific basis. Chapman & Hall, London. UK.
Gebresenbet, G., I. Nordmark, T. Bosona and D. Ljungberg. 2011. Potential for optimised food
deliveries in and around Uppsala city, Sweden. J. Transp. Geogr. 19: 1456–1464.
Griffiths, M.W. 2000. Milk and unfermented milk products. pp. 507–534. In: B.M. Lund, T.C.
Baird-Parker and G.W. Gould [eds.]. The Microbiological Safety and Quality of Food.
Aspen Publishers, Gaithersburg. USA.
Hansen, T.K. and M. Jakobsen. 2004. Yeast in the dairy industry. pp. 441–460. In: D.K. Arora,
P.D. Bridge and D. Bhatnagar [eds.]. Fungal Biotechnology in Agricultural, Food and
Environmental Applications. Marcel Dekker, New York. USA.
Head, I.M., J.R. Saunders and R.W. Pickup. 1998. Microbial evolution, diversity, and ecology: a
decade of ribosomal RNA analysis of uncultivated microorganisms. Microb. Ecol. 35: 1–21.
Hesse, M. and J.P. Rodrigue. 2004. The transport geography of logistics and freight distribution.
J. Transp. Geogr. 12: 171–184.
Holsinger, V.H., K.T. Rajkowski and J.R. Stabel. 1997. Milk pasteurization and safety: a brief
history and update. Rev. Sci. Tech. Off. Int. Epiz. 16: 441–451.
Hugenholtz, P., B.M. Goebbel and N.R. Pace. 1998. Impact of culture independent studies
on the emerging phylogenetic view of bacterial diversity. J. Bacteriol. 180: 4765–4774.
Hutkins, R.V. 2006. Microbiology and Technology of Fermented Foods. Blackwell Publishing,
Ames. USA.
Institute of Food and Science Technology. Professional food microbiology group. 1997.
Development and use of microbiological criteria for foods. Food Sci. Technol. Today
11: 137–176.
International Dairy Federation. 2010. The world dairy situation 2010. Bulletin of the IDF
446/2010. Brussels, 212 pp.
20 Dairy Microbiology: A Practical Approach
Statistical Office of European Communities. 2011. Farm to fork—a statistical journey along
the EU´s food chain. Statistics in focus 27/2011. Publications Office of the European
Union: Luxembourg, 12 pp.
Straus, G.L. 1913. Diseases in Milk. The Remedy Pasteurization. E.P. Dutton and Co., New
York. USA.
Tamime, A.Y. 2002. Microbiology of starter cultures. pp. 261–366. In: R.K. Robinson [ed.]. Dairy
Microbiology Handbook. Wiley-Interscience, New York. USA.
Tauxe, R.V. 2002. Emerging foodborne pathogens. Int. J. Food Microbiol. 78: 31–41.
Tassou, S.A., G. De-Lille and Y.T. Ge. 2009. Food transport refrigeration—Approaches to
reduce energy consumption and environmental impacts of road transport. Appl. Therm.
Eng. 29: 1467–1477.
The Global Dairy Agenda for Action—Climate Change. 2009. Available at: http://www.dairy-
sustainability-initiative.org/Files/media/Declarations/Declaration_Final-Text_English-
18-June-2009.pdf.
Tirado, M.C., R. Clarke, L.A. Jaykus, A. McQuatters-Gallop and J.M. Frank. 2010. Climate
change and food safety: A review. Food Res. Int. 43: 1745–1765.
United States Public Health Service and Food and Drug Administration. 2011.
Grade “A” p asteurized milk ordinance. Available at: http://www.fda.gov/
downloads/Food/FoodSafety/ProductSpecificInformation/MilkSafety/
NationalConferenceonInterstateMilkShipmentsNCIMSModelDocuments/UCM291757.
pdf.
van der Spiegel, M., H.J. van der Fels-Klerx and H.J.P. Marvin. 2012. Effects of climate change
on food safety hazards in the dairy production chain. Food Res. Int. 46: 201–208.
Von Wright, A. and L. Axelsson. 2011. Lactic acid bacteria: an introduction. pp. 1–16. In:
S. Lahtinen, A.C. Ouwehand, S. Salminen and A. von Wright [eds.]. Lactic Acid Bacteria:
Microbiological and Functional Aspects. CRC Press, Boca Raton. USA.
Westhoff, D.C. 1978. Heating milk for microbial destruction: A historical outline and update.
J. food Prot. 41: 122–130.
CHAPTER 2
INTRODUCTION
Raw milk quality is of utmost importance for the dairy industry because
the principle requirement for the production of high quality dairy products
is the production and use of high quality raw milk. This chapter aims to
cover the main aspects of the microbiology of raw milk and is confined
to the study of the milk of the most common domestic lactating animal
species, i.e., cows, goats and sheep. The chapter discusses aspects pertaining
mostly to bacteria (eubacteria) and to a lesser extent to fungi and viruses.
Other groups of (micro) organisms that might be occasionally present in
raw milk such as protozoa (e.g., Toxoplasma, Cryptosporidium), helminths or
other parasites are not covered.
period (with at least one sample per month) should not exceed 100,000 cfu/mL.
The production of milk from lactating animals other than bovine (e.g., sheep,
goats or buffalos) often presents practical difficulties. For instance, in south-
western Europe, a considerable number of sheep and/or goat farms are
located in mountainous regions, distant from milk processing facilities. The
farms’ herd size is often small and therefore does not produce sufficient
revenues for the producers to invest on mechanization. Therefore frequent
pick-up and collection of milk is not cost-effective or even plausible. In
some cases access to electrical power is not for granted and automated
milking systems and/or milk cooling equipment are not easy to install
and/or operate. These limitations may have led European legislators to
(understandably) adopt a more lenient limit with regards to the microbial
content of milk from “other species”, which is currently set at 1,500,000 cfu/
mL. However, much better milk microbiological quality is required (i.e., a
maximum of 500,000 cfu/mL) for raw milk from species other than cows,
if such milk is intended to be used for the manufacture of dairy products
in a fashion that does not involve a heat treatment (sanitation) step. An
example where the “500,000 cfu/mL” criterion applies is the use of raw
sheep milk for the manufacture of cheeses that undergo a minimum ripening
time of two or more months. No SCC limits are listed in Regulation 853
for the milk of species other than bovine. This is likely the result of the fact
that the SCC in small ruminants’ milk (particularly in goats’ milk) varies
considerably, even among healthy animals and several non-infectious
factors (i.e., factors other than inflammation of the mammary gland) can
influence SCC. Milk secretion in small ruminants, particularly in goats,
is apocrine in nature, and cytoplasmic particles, often similar in size to
milk somatic cells, are normally present in their milk (Souza et al. 2012). A
great body of research worldwide has been allocated on the investigation
of (non-pathological) factors affecting milk SCC levels in sheep and goats,
milk and such factors include, amongst others, parity, stage of lactation,
or even sample handling, sample storage practices and measuring method
(Paape et al. 2001, Contreras et al. 2007, Raynal-Ljutovac et al. 2007, Souza
et al. 2012). Currently, there does not seem to be a consensus on establishing
a unique and scientifically justifiable discriminatory SCC standard (limit)
for sheep and goat milk quality because research studies on the SCC of
small ruminant milk have sometimes resulted in conflicting outcomes and
conclusions. Hence, at present, no legislative upper limits regarding small
ruminant raw milk SCC values are in place in the EU.
As mentioned previously, the microbiological and SCC (where
applicable) upper allowable limits for raw milk are expressed as geometric,
rather than arithmetic means (averages). When evaluating bulk tank SCCs
or microbial counts the use of geometric averages helps to decrease the
variation or impact of high or low counts (often the result of sampling or
The Microbiology of Raw Milk 25
Table 1. Arithmetic and geometric average of total mesophilic counts obtained from the
analysis of ovine bulk tank milk samples (n = 5) collected randomly over a three-month period.
Sampling no. Microbial counts (cfu/mL) Log microbial counts (log cfu/mL)
1 820,000 5.91
2 950,000 5.67
3 3,900,000 6.54
4 1,200,000 5.85
5 720,000 5.71
Arithmetic mean 1,518,000 6.084
Geometric mean 1,212,895 10 6.084
they will give rise to a single colony after incubation. Injured cells may
not form (visible) colonies during the time frame of incubation whereas,
depending on their spatial distribution in the agar milieu (e.g., towards the
surface as opposed to towards the bottom of the petri dish), some of the
strictly anaerobic bacteria present in raw milk will most likely not grow.
Hence, viable but non-culturable microorganisms are not counted. For all
the aforementioned reasons, the SPC method might underestimate the total
number of viable microorganisms present in raw milk. Most importantly,
the method is impractical for the dairy industry because of the prohibitive
length of time necessary to obtain results. Additional colony count methods
that are more suitable for routine testing (i.e., routine methods) have been
developed and used, such as the plate loop method (Thompson 1960) and
the spiral plate method (Gilchrist et al. 1973).
An alternative approach to determine the bacterial counts of raw milk
is the total microscopic count method. This method relies on counting
stained bacteria on a special glass slide on which a known volume of milk is
applied (typically 0.01 mL), following sample treatments to fix the sample.
Several optical fields are counted using bright field illumination (Hill 1991).
The method is relatively fast, yet tedious to the operator, particularly when
a high number of samples must be examined. Another limitation is the
inability to discriminate between living and dead bacteria, i.e., the method
estimates total bacterial counts as opposed to total viable counts.
Additional available direct or indirect methods for assessing the
bacteriological content of raw milk are available and their advantages
and disadvantages are described in comprehensive reviews (O’Toole
1983, International Dairy Federation 1991a, Vasavada 1993, Suhren and
Reichmuth 2000). Today, however, most milk processors rely on direct,
automated, rapid (ca. 10 min), and high-throughput (up to 200 milk samples
per hr) enumeration methods for determining total bacterial counts in
incoming raw milk. These automated bacterial cell counters operate by
degradation and separation (via centrifugation) of milk somatic cells and
milk particles and constituents that are similar in size to bacterial cells,
followed by fluorescent staining and counting of individual bacterial cells
using flow cytometry. The results of automated analyzers are expressed as
IBC/mL (Individual Bacterial Counts per mL), rather than CFU/mL. IBC
estimates can be converted to CFU values via specific algorithms obtained
through correlation analyses between the two parameters (Cassoli et al.
2007). Several parameters of automated flow-cytometry based bacterial
counters have been evaluated (Lachowsky et al. 1997, Bolzoni et al. 2000,
2001). Manufacturers of automated, high-throughput milk analyzers
continuously come up with more advanced and more reliable models. Thus,
the interested user should consult the most updated relevant technical/
32 Dairy Microbiology: A Practical Approach
Bacteria
There are hundreds of different bacterial genera known to date and likely
thousands of different species. Different bacterial genera can differ in their
structural or physiological properties. For instance, most bacterial genera
can be classified into Gram-positive or Gram-negative based on structural
differences in their bacterial cell envelopes. In addition, differences in their
physiological responses to oxygen, osmotic pressure, acidity or temperature
as well as in their biosynthetic and metabolic activities are further used
to classify bacteria into groups of special interest in food microbiology.
Research in bacteriology over the last three decades or so has revealed that
bacteria are quite versatile and adaptive organisms towards various forms
of environmental stress (cold, osmotic, acid or heat stress). These adaptation
mechanisms can be quite elaborate and include both physiological and
genetic responses.
34 Dairy Microbiology: A Practical Approach
Gram-negative bacteria
Spore-forming bacteria
to 240 spores/mL with most of the milk samples having a spore count
between 1 and 10 spores/mL. Spores of clostridia that germinate and grow
fermentatively (converting lactic acid to butyric acid, carbon dioxide and
hydrogen) towards the later stages of cheese maturation are the causative
agents of the “late blowing” defect in cheeses (butyric acid fermentation)
with long ripening times (Cocolin et al. 2004). C. tyrobutyricum is considered
the primary cause of “late blowing” although additional species of clostridia
such as C. sporogenes may play secondary roles in cheese defects (Klijn et
al. 1995, Le Bourhis et al. 2007, Garde et al. 2011).
Clostridium spp. are also important in dairy microbiology as they are
involved in food poisoning episodes. C. perfringens food poisoning is usually
linked with spore germination and proliferation of vegetative cells after
temperature abuse of heat-treated foods. Other pathogenic clostridia include
C. botulinum, the causative agent of food-borne botulism, C. baratii and
C. butyricum. C. butyricum has been implicated in outbreaks of food-borne,
type-E botulism (Meng et al. 1997, Peck 2009). It has been reported that
clostridial proliferation in freshly produced raw milk is improbable due to
its highly positive Eh value (Goudkov and Sharpe 1965). Nonetheless, the
botulinum neurotoxin is the most toxic substance known to date and the
(raw) milk supply has been considered as a likely target for terrorist attacks
in terms of deliberate contamination with botulinum toxin. C. botulinum
neurotoxins type A and B and their corresponding complexes are inactivated
by at least 99.99% and 95%, respectively during raw milk pasteurization at
72°C for 15 s (Weingart et al. 2010).
Psychrotrophic bacteria
communities occurs during cold storage of raw milk, but the taxonomic
diversity decreased with storage time as a consequence of some microbial
populations’ dominance during refrigeration. In their study, raw milk
samples were collected from bulk tanks of dairy farms and silo tanks of an
associated industrial processing plant. According to their analyses Gram-
positive bacteria (Bacilli, Clostridia, and Actinobacteria) prevailed in the
milk from the farm tanks during cold incubation, whereas milk samples
from the dairy plant were dominated by Gram-negative species belonging
to Gammaproteobacteria, especially Pseudomonadales.
Reduced quality of raw and processed milks is frequently a consequence
of the proliferation and metabolic activities of psychrotrophic bacteria.
Although the Gram-negative psychrotrophs are effectively destroyed by
pasteurization, some species of psychrotrophic bacteria can produce lipolytic
and proteolytic enzymes during growth in raw milk. These enzymes are
either not inactivated, or only marginally affected by the time/temperature
schemes used in the dairy industry for the manufacture of processed dairy
products (e.g., pasteurization, UHT processing). These lipases and proteases
can lead to the development of off-odors in raw milk. Additionally, being
heat-stable, they retain activity after the heat treatment of milk and therefore
gradually degrade milk proteins and lipids, leading to noticeable off-odors
and reducing dairy products’ shelf-life (Griffiths et al. 1981, Sørhaug and
Stepaniak 1997, Nörnberg et al. 2010). Therefore numerous published
manuscripts have emphasized the importance of the initial microbiological
quality of raw milk and the importance of the rapid and efficient cooling
of milk upon milking in terms of milk and dairy products’ quality (Banks
et al. 1988, Griffiths et al. 1987, 1988, Zeng et al. 2007).
As stated above, some aerobic spore-forming bacteria belonging to the
genus Bacillus (e.g., B. cereus) are psychrotrophic (Grosskopf and Harper
1974). Psychrotrophic strains of aerobic spore-formers are a major concern
to the dairy industry because they are able to survive milk pasteurization
and can subsequently proliferate in dairy products such as pasteurized
milk stored at low temperatures, often leading to spoilage (Meer et al.
1991). For instance, in the absence of post-pasteurization re-contamination
of milk by Gram-negative psychrotrophic bacteria (Eneroth et al. 2000),
the shelf-life of pasteurized milk largely depends on the presence (levels)
and spoilage potential of psychrotolerant spore-formers (Fromm and Boor
2004). Psychrotrophic aerobic spore-forming bacteria may be also significant
in terms of food safety. Psychrotrophic strains of B. cereus producing
enterotoxin have been identified (Van Netten et al. 1990). Also, the ability of
certain B. cereus strains to produce toxins in milk stored at 8°C under aerated
conditions has been demonstrated (Christiansson et al. 1989). Other well-
known psychrotrophic pathogens (Schofield 1992) that have been isolated
from raw milk are Listeria monocytogenes, Yersinia enterocolitica and Aeromonas
The Microbiology of Raw Milk 41
Thermoduric bacteria
Lactic acid bacteria (LAB) constitute one of the main groups of raw-milk
microorganisms. The most common source of LAB in raw milk is the
udder skin and teat canal, but LAB also colonize animal sites such as the
vagina or the intestine. The term often used to denote LAB present in
raw milk is autochthonous, or non-starter LAB, to distinguish them from
the starter LAB, i.e., the known and characterized strains of LAB that are
deliberately added to milk for the manufacture of fermented dairy products.
Traditionally, the manufacture of fermented dairy products relied upon
The Microbiology of Raw Milk 43
the presence and acidification activity of LAB present in raw milk. This
practice is still used nowadays for the manufacture of some artisanal dairy
products. The biopreservation effect of LAB relies on the fermentation
of the major milk sugar (lactose) and the production of organic acids
with a concomitant reduction of milk pH and therefore the creation of an
acidic environment, unfavorable for the proliferation and/or survival of
pathogenic microorganisms. The rate of acid production as well as the
final pH is of vital importance for the safety of fermented dairy products.
The production of several antimicrobial compounds such as bacteriocins,
which are active against several food-borne pathogens, also contributes to
the bio-preservative effects of LAB. Nowadays a vast majority of fermented
dairy products is produced using well-defined (mixtures of) LAB strains.
Most LAB do not grow at refrigeration temperatures and souring of raw
milk is highly unlikely under adequately refrigerated conditions. Elevated
titratable acidity in milk nowadays is rare and usually an indication of
raw-milk temperature abuse in the farm or during transport. In certain
instances, the presence of adventitious LAB or the use of inappropriately
selected (e.g., overactive) starter LAB, can lead to quality defects in
fermented dairy products (e.g., bitterness, production of gas, or extreme
acidity). On the other hand, underactive LAB, due to the presence of
antibiotic residues or bacteriophages in raw milk, can delay or even halt
the fermentation process.
LAB are usually classified based on: a) their optimum growth
temperature into mesophilic (around 30°C) and thermophilic (around 43°C),
and b) fermentation reactions/metabolic pathways into homofermentative
(lactic acid is the primary and most abundant end-product of lactose
fermentation) and heterofermentative (other compounds in addition to
lactic acid are produced in significant amounts, such as acetic acid, or
carbon dioxide). Raw-milk LAB belong to all major LAB genera (Lactococcus,
Lactobacillus, Streptococcus, Leuconostoc, Pediococcus, Enterococcus). There are
probably hundreds of published articles on the isolation, physiological and
technological characterization of LAB isolated from raw bovine, caprine or
ovine milk and raw-milk cheeses and one such study is cited here (Franciosi
et al. 2009). Details on LAB used in the manufacture of dairy products are
presented in Chapter 4 of this book.
Mastitis-causing organisms
consumption of raw milk are addressed and data on the prevalence of food-
borne pathogens in raw milk and raw milk-borne disease outbreaks in the
US from 2000 until 2008 are summarized. In addition, under the section
“Pathogens in milk”, the second edition of the encyclopedia of dairy sciences
(Fuquay et al. 2011) contains chapters dedicated to 14 pathogenic bacterial
families, genera or species commonly associated with raw milk (Bacillus
cereus, Brucella spp., Campylobacter spp., Clostridium spp., Coxiella burnetii,
Escherichia coli, Enterobacteriaceae, Enterobacter spp., Listeria monocytogenes,
Mycobacterium spp., Salmonella spp., Shigella spp., Staphylococcus aureus and
Yersinia enterocolitica). The chapters have been written by highly-respected
experts in the field and contain updated information on pathogens’
characteristics, physiology, resulting milk-borne illness, toxins, outbreaks,
incidence in dairy products including raw milk, potential sources and
suggested control measures at the farm. Pathogens in dairy products are
also covered in Chapter 3 in this book. The remaining of this section focuses
on specific (emergent?) bacterial agents with known or suspected public
health importance, recently shown to be occasionally present in raw milk.
Mycobacterium avium subsp. paratuberculosis (MAP) is the causative
agent of paratuberculosis (Johne’s disease), a chronic debilitating disease
(infectious enteritis with cachexia) affecting ruminants and other animals
worldwide. Johne’s disease has been reported in many countries worldwide.
Across the EU there are many published studies on the prevalence of MAP
in cattle and a smaller number of studies on sheep and goats (Nielsen and
Toft 2009). Infected animals are shedding the organism periodically in both
feces and milk (Streeter et al. 1995). The presence of MAP in milk for human
consumption poses concerns due to its possible association with Crohn’s
disease in humans (Grant 2005). The occurrence of MAP in bulk tank milk
and individual milk samples at cattle dairy farms worldwide was reviewed
by Okura et al. (2012). The authors reported a considerable variation of MAP
in bulk tank milk and individual cows’ milk. The overall MAP apparent
prevalence and 95% CI based on PCR and culture were summarized to 0.1
(0.04–0.22) or 10% on a per cent basis in bulk tank milk samples, and 0.2
(0.12–0.32) or 20% on a per cent basis in individual milk samples.
Milk pasteurization is a heat treatment process for a given period of
time that has been designed to inactivate the most heat-resistant, non-
spore-forming pathogenic bacteria that may be present in raw milk, i.e.,
Mycobacterium bovis and Coxiella burnetti, the causative agents of tuberculosis
and Q-fever in humans, respectively. The legal minimum time/temperature
combination for pasteurized milk is the heating of milk at 72°C for 15 s
(HTST) or 63°C for 30 min [low-temperature, long-time pasteurization
(LTLT)] (Commission Regulation (EC) No. 853/2004). There are several
published reports indicating survival of low numbers of MAP after
pasteurization of either artificially contaminated or naturally contaminated
The Microbiology of Raw Milk 47
milk. Therefore it has been suggested that, depending on the initial MAP
load of raw milk, low numbers of MAP may occasionally survive HTST
pasteurization (Grant 2006, Van Brandt et al. 2011b). Review articles have
been published summarizing the available (and often contradictive) data
regarding the prevalence of MAP in raw and pasteurized milks and other
dairy products, as well as the effects of pasteurizing treatments on MAP
survival in milk (Eltholth et al. 2009, Gill et al. 2011). Only a few studies
have been published on the behavior of MAP during the production and
storage of fermented milk products (cheddar cheese, yogurt, acidophilus
milk and kefir). It appears that the type of fermented dairy product, the type
of starter culture, the stage of MAP inoculation (pre- vs. post-fermentation
inoculation), the MAP strain, and the pH of the finished product affect the
(extent of) survival of MAP (Donaghy et al. 2004, Van Brandt et al. 2011a,
Klanicova et al. 2012). More studies are warranted, however in this field.
Helicobacter pylori is the causative agent of gastric ulcers and other
pathogenic conditions in humans. The presence of H. pylori in a small
fraction of raw cows’ milk samples examined was demonstrated via
fluorescence in situ hybridization (Angelidis et al. 2011), although its
presence had been proposed/suspected by the findings of earlier studies
based on PCR (Fujimura et al. 2002, Quaglia et al. 2008). It has been
hypothesized that sheep may constitute a reservoir of H. pylori (Dore et al.
2001). At present, the risk of human H. pylori infection via consumption of
contaminated raw milk or dairy products manufactured with contaminated
raw milk remains unknown.
Arcobacter spp. are Gram-negative, spiral-shaped bacteria belonging
to the family Campylobacteraceae. Arcobacter spp. have been repeatedly
isolated from feces of livestock animals including cows, whereas only
a few investigations have reported their presence in feces of goats and
sheep (Van Driessche et al. 2003, De Smet et al. 2011). Arcobacter spp. are
considered emerging waterborne and food-borne human pathogens with
infection symptoms similar to campylobacteriosis. Consumption and
handling or raw or undercooked meats (mainly poultry) are potential
sources of infections in humans (Lehner et al. 2005). The potential role of
Arcobacter spp. in human disease, however, needs further evaluation. A few
studies from different countries have reported the isolation of Arcobacter
spp. at different frequencies (5.8–46%) from raw bovine milk (Scullion et
al. 2006, Shah et al. 2012).
Generalizations in food microbiology/food safety should be avoided
or at least made with caution. Nonetheless, from a practical point of view
if raw milk is to be used for the manufacture of processed milks or other
dairy products whose technology involves a heat treatment step equal to
or more intense than that of pasteurization, the main food-safety concerns
48 Dairy Microbiology: A Practical Approach
species, not isolated via the traditional cultural approach. Chen et al. (2009)
reported on the biodiversity of yeasts in raw milk from dairies in China.
The authors identified 11 different species of yeasts using an integrated
approach including phenotypic and molecular (RAPD-PCR analysis and
partial sequencing of the 26S rDNA) methods. A recently published article
reported the results of a Canadian survey aiming on characterizing the
fungal microflora of raw cows’ milk (111 samples) and raw-milk cheeses
collected from 19 dairy farms (Lavoie et al. 2012). Molecular identification
analyses of the isolates using the ITS1-5.8S-ITS2 rDNA region showed that
almost two-thirds of the fungal isolates were yeasts that could be assigned
to 37 species/11 genera and the remaining isolates were molds that could
be assigned to 33 species/25 genera. Debaryomyces hansenii was the most
abundant among the yeast species (21% of the 339 yeast isolates) and was
detected in the milk of 14 from the 19 farms. Candida was the most abundant
(44% of the isolates) and most diverse (11 species) genus. Other frequently
identified yeast genera were Cryptococcus spp. (10.9% of the yeast isolates)
and Pichia spp. (8% of the yeast isolates). Eurotium was the most abundant
mold genus (about one-quarter of the mold isolates) and was isolated
from the milk of 13 from the 19 farms. Lichtheimia was the second most
abundant mold genus (13.3% of the mold isolates) and was isolated from
nine farms. The authors argued that only a fraction of the fungal species
may have been actually identified. Nonetheless, the findings of their study
indicate that the fungal profile of milk differs from farm to farm. In France,
Delavenne et al. (2011) evaluated the fungal diversity in a smaller collection
of raw milk samples (nine), which also included goat and ewe samples,
in addition to cow milk samples. Following DNA extraction from milk
samples, a semi-nested PCR method was used to amplify the ITS1 region of
fungal DNA and PCR products were analyzed by ion-pair, reverse-phase,
denaturing, high-performance liquid chromatography (D-HPLC). Fractions
of each peak were retrieved and sequenced. The approach enabled the
identification of 27 fungal species (18 yeast species belonging to 9 genera,
Candida, Cryptococcus, Debaryomyces, Geotrichum, Kluyveromyces, Malassezia,
Pichia, Rhodotorula and Trichosporon and 9 mold species belonging to 7
genera, Aspergillus, Chrysosporium, Cladosporium, Engyodontium, Fusarium,
Penicillium and Torrubiella). The authors reported the highest fungal diversity
in cow milk with a total recovery of 23 different species (five mold and 18
yeast), whereas only six (one mold and five yeast) and seven (one mold
and six yeast) species were recovered in goat and ewe milk, respectively.
The most common fungal species among the nine milk samples were
G. candidum, K. marxianus and Candida spp. such as C. parapsilosis (Delavenne
et al. 2011). In Italy, Corbo et al. (2001) analyzed 26 samples of raw cow,
ewe, goat and water buffalo milk for the presence of yeasts using cultural
and biochemical methods and reported the isolation of 36 yeast species.
50 Dairy Microbiology: A Practical Approach
Viruses
There are many pathogenic viruses for humans known to be transmitted via
consumption of contaminated foods, and according to a literature review
by Newell et al. (2010) food-borne viruses belong to at least 11 known viral
families. Contamination of foods with most of these food-borne viruses often
results from non-hygienic food-handling practices by human carriers. In
addition, the actual involvement of foods in viral food-borne outbreaks is
very difficult to approximate, because in many cases, following the initial
food-borne incident (“seeding event”), viruses easily spread from one
infected individual to another, without involvement of contaminated food
sources. Among food-borne viruses, noroviruses and the hepatitis A virus
have been the causative agents in well-documented causes of food-borne
illness (Koopmans and Duizer 2004). Cliver (1997) stated that “all known
food-borne viruses except the agent of tick-borne encephalitis are human
specific and transmitted by a fecal-oral cycle”.
There are some publications dated back in the seventies about the
incidence/contamination of raw milk with animal viruses (zoonotic or
not) and human viral outbreaks that have occurred as a consequence
of consumption of contaminated milk or raw-milk cheeses (Kefford et
al. 1979). It should be stressed, however, that in most cases the origin of
viral contamination of raw milk in these instances was external, i.e., from
environmental sources such as contaminated water or human carriers, and
52 Dairy Microbiology: A Practical Approach
Lactic bacteriophage
specific antibodies), the non specific defense agents lactoferrin and lysozyme
and the enzyme lactoperoxidase (International Dairy Foundation 1991b,
Korhonen et al. 2000). Several peptides and other organic compounds (e.g.,
free fatty acids) in milk have shown to possess antimicrobial properties. In
addition, a wide variety of bacteriocins with variable antimicrobial spectra
can be produced by LAB. In recent years, the presence of additional host-
defense related minor proteins and peptides has been documented in cow’s
milk (Hettinga et al. 2011, Wheeler et al. 2012).
Bovine milk contains low levels of lysozyme (ca. 0.1 µg/mL) but its
concentration increases during mastitis (1–2 µg/mL). The antibacterial
role of lysozyme relies on the cleavage of the glycosidic bond between
N-acetylmuramic acid and N-acetylglucosamine in bacterial cell-wall
peptidoglycan (International Dairy Federation 1991b, Benkerroum 2008).
Lactoferrin is a whey protein with iron chelating properties. Its principle
antimicrobial action relies on depriving bacteria of iron, which is an
essential element for bacterial growth. Its concentration in bovine milk
(ca. 0.2 mg/mL) can increase several-fold during mastitis (International
Dairy Federation 1991b). Recent studies have attributed additional beneficial
properties to lactoferrin such as anti-cancer, immunomodulatory and
antioxidant properties (García-Montoya et al. 2012). Lactoperoxidase (LP),
i.e., the milk peroxidase, is one of the most abundant enzymes in bovine milk
constituting ca. 0.5% of the whey proteins and one of the three components
of the LP system of raw milk. LP is an oxidoreductase which catalyses the
oxidation of thiocyanate ions (SCN-) that are present in milk as a result of the
cows’ diet into hypothiocyanate ions (SCNO-) at the expense of hydrogen
peroxide (H2O2). Hypothiocyanate ions and other intermediates are the
reactive products of the LP system of milk and are potent antimicrobials.
The LP activity and thiocyanate content in raw milk can be affected by
factors such as the individual animal, the lactating species, the animal feed,
the time after milking and the stage of lactation (Althaus et al. 2001, Fonteh
et al. 2002, Yaqub et al. 2012). The hydrogen peroxide needed to activate
the LP system can be generated by the LAB of milk or by an indigenous
enzymatic system (xanthine oxidase/hypoxanthine). Most typically, H2O2
is provided exogenously in order to activate the LP system and thus help
preserve raw milk in situations where refrigerated storage of raw milk
between milking and processing is not possible (Haddadin et al. 1996). The
concentrations of the components of the LP system, the extent and range
of its antimicrobial action and its applications in milk and dairy products
have been presented in related review articles (Wolfson and Sumner 1993,
Kussendrager and van Hooijdonk 2000, Seifu et al. 2005).
The raw-milk natural antimicrobial compounds exert a bacteriostatic
effect for a time period following milking, the duration of which may
depend on several factors including storage temperature. For instance, the
The Microbiology of Raw Milk 55
Concluding Remarks
The determination of the microbiological flora of raw milks has been the
subject of scientific research for more than one century. Depending on the
nature of the organisms sought, traditionally milk samples (diluted or
undiluted) are plated onto agar media (selective or nonselective) and are
incubated (aerobically, under microaerophilic or even anaerobic conditions)
at temperatures thought or known to be optimum for the growth of the
target organism(s). Enrichment steps in semi-selective or selective broths are
frequently used to enable preferential proliferation of the target organism(s)
against the background microbial flora of raw milk. Other approaches and
steps such as filtration and centrifugation have been used as a means of
bacterial concentration, assisting subsequent detection steps. Following
the isolation of any given microorganism, further identification strategies
include the direct observation under the microscope (wet mounts and
stained-preparations) in which characteristics such as the size, morphology
and motility of the microorganism can be evaluated, as well as the presence
of specific structures (e.g., capsules, flagella, spores). Additional laboratory
tests are then conducted to narrow down the identity of unknown isolates,
such as biochemical and serological tests. Biochemical assays most often
target enzymatic activities that rely on the possession and expression of
specific genes by the isolate. Biochemical tests include the utilization of
sugars, the determination of proteolytic and lipolytic traits, types of energy
metabolism, physiological growth boundaries (e.g., temperature, pH,
osmolarity), tolerance to antimicrobial agents and other enzymatic activities.
The methods for bacterial identification and enumeration based on
culture, microscopy and biochemistry have supported dairy microbiology
for decades and have provided invaluable service to the scientific
community and public health agencies. However, the phenotypic culture-
based identification approaches are often hampered by difficulties.
The morphology and/or motility of certain bacterial species under the
microscope may vary depending on temperature and/or other culture
56 Dairy Microbiology: A Practical Approach
available for the analysis of the microbial composition of raw milk and raw
milk cheeses, covering technical aspects, advantages and disadvantages.
Most experts nowadays seem to agree that it is probably most beneficial
to apply polyphasic approaches, i.e., both culture-dependent and culture
independent methods when assessing the diversity of the raw milk
microbiota.
References Cited
Ajitkumar, P., H.W. Barkema and J. De Buck. 2012. Rapid identification of bovine mastitis
pathogens by high-resolution melt analysis of 16S rDNA sequences. Vet. Microbiol. 155:
332–340.
Althaus, R.L., M.P. Molina, M. Rodríguez and N. Fernández. 2001. Analysis time and lactation
stage influence on lactoperoxidase system components in dairy ewe milk. J. Dairy Sci.
84: 1829–1835.
Amagliani, G., A. Petruzzelli, E. Omiccioli, F. Tonucci, M. Magnani and G. Brandi. 2012.
Microbiological surveillance of a bovine raw milk farm through multiplex real-time
PCR. Foodborne Path. Dis. 9: 1–6.
Ammor, M.S., C. Michaelidis and G.-J.E. Nychas. 2008. Insights into the role of quorum sensing
in food spoilage. J. Food Prot. 71: 1510–1525.
Angelidis, A.S., L.T. Smith and G.M. Smith. 2002. Elevated carnitine accumulation by Listeria
monocytogenes impaired in glycine betaine transport is insufficient to confer wild-type
cryotolerance in milk whey. Int. J. Food Microbiol. 5: 1–9.
Angelidis, A.S., I. Tirodimos, M. Bobos, M.S. Kalamaki, D.K. Papageorgiou and M.
Arvanitidou. 2011. Detection of Helicobacter pylori in raw bovine milk by fluorescence
in situ hybridization. Int. J. Food Microbiol. 151: 252–256.
Anonymous. Pasteurized Milk Ordinance. 2011. U.S. Department of Health and Human Services.
http://www.fda.gov/downloads/Food/Food Safety/Product-Specific Information/
Milk Safety/NationalConferenceonInterstateMilkShipmentsNCIMSModelDocuments/
UCM291757.pdf. Accessed Oct. 29 2012.
Ash, C., F.G. Priest and M.D. Collins. 1993. Molecular identification of rRNA group 3 bacilli
(Ash, Farrow, Wallbanks and Collins) using a PCR probe test. Antonie van Leeuwenhoek
64: 253–260.
Balkhy, H.H. and Z.A. Memish. 2003. Rift Valley Fever: an uninvited zoonoses in the Arabian
peninsula. Int. J. Antimicrob. Agents 21: 153–157.
Banks, J.M., M.W. Griffiths, J.D. Phillips and D.D. Muir. 1988. A comparison of the effects of
storage of raw milk at 2°C and 6°C on the yield and quality of Cheddar cheese. Food
Microbiol. 5: 9–16.
Bartoszewicz, M., B.M. Hansen and I. Swiecicka. 2008. The members of the Bacillus cereus group
are commonly present contaminants of fresh and heat-treated milk. Food Microbiol. 25:
588–596.
Bauer, K. 1997. Foot-and-mouth disease as zoonoses. Arch. Virol. 13(suppl.): 95–97.
Beales, N. 2004. Adaptation of microorganisms to cold temperatures, weak acid preservatives,
low pH, and osmotic stress: A review. Comp. Rev. Food Sci. Food Saf. 3: 1–20.
Benkerroum, N. 2008.Antimicrobial activity of lysozyme with special relevance to milk. Afr.
J. Biotechnol. 7: 4856–4867.
Berry, E.D. and P.M. Foegeding. 1997. Cold adaptation and growth of microorganisms. J. Food
Prot. 12: 1583–1594.
58 Dairy Microbiology: A Practical Approach
Bhatt, V.D., V.B. Ahir, P.G. Koringa, S.J. Jakhesara, D.N. Rank, D.S. Nauriyal, A.P. Kunjadia
and C.G. Joshi. 2012. Milk microbiome signatures of subclinical mastitis-affected cattle
analysed by shotgun sequencing. J. Appl. Microbiol. 112: 639–650.
Bolzoni, G., A. Marcolini and G. Varisco. 2000. Evaluation of the Bactoscan FC. 1 Accuracy,
comparison with the Bactoscan 8000 and somatic cell effect. Milchwissenschaft 55: 67–70.
Bolzoni, G., A. Marcolini and G. Varisco. 2001. Evaluation of the Bacoscan FC. 2. Stability,
repeatability, carry-over and linearity. Milchwissenschaft 56: 318–321.
Bona, C., B. Dewals, L. Wiggers, K. Coudijzer, A. Vanderplasschen and L. Gillet. 2005.
Pasteurization of milk abolishes bovine herpesvirus 4 infectivity. J. Dairy Sci. 88:
3079–3083.
Boor, K.J., D.P. Brown, S.C. Murphy, S.M. Kozlowski and D.K. Bandler. 1998. Microbiological
and chemical quality of raw milk in New York state. J. Dairy Sci. 81: 1743–1748.
Burgess, S.A., D. Lindsay and S.H. Flint. 2010. Thermophilic bacilli and their importance in
dairy processing. Int. J. Food Microbiol. 144: 215–225.
Caini, S., K. Szomor, E. Ferenczi, Á.S. Gáspár, Á. Csohán, K. Krisztalovics, Z. Molnár and J.K.
Horváth. 2012. Tick-borne encephalitis transmitted by unpasteurized cow milk in western
Hungary, September to October 2011. Euro. Surveil. 17: pii=20128.
Callon, C., F. Duthoit, C. Deblés, M. Ferrand, Y. Le Frileux, R. De Crémoux and M.-C. Montel.
2007. Stability of microbial communities in goat milk during a lactation year: Molecular
approaches. Syst. Appl. Microbiol. 30: 547–560.
Cassoli, L.D., P.F. Machado, A.C.D.O. Rodrigues and A. Coldebella. 2007. Correlation study
between standard plate count and flow cytometry for determination of raw milk total
bacterial count. Int. J. Dairy Tech. 60: 44–48.
Cavirani, S. 2008. Cattle industry and zoonotic risk. Vet. Res. Commun. 32(suppl. 1): S19–S24.
CDC Centers for Disease Control and Prevention. 1999. Mass treatment of humans who
drank unpasteurized milk from rabid cows-Massachusetts, 1996–1998. Morb. Mortal.
Wkly Rep. 48: 228–229.
CDC Centers for Disease Control and Prevention. 2014. http://www.cdc.gov/foodsafety/
outbreaks/multistate-outbreaks/outbreaks-list.html.
Celestino, E.L., M. Iyer and H. Roginski. 1996. The effects of refrigerated storage on the quality
of raw milk. Aust. J. Dairy Tech. 51: 59–63.
Champagne, C.P., R.R. Laing, D. Roy and A.A. Mafu. 1994. Psychrotrophs in dairy products:
Their effects and their control. Crit. Rev. Food Sci. Nutr. 34: 1–30.
Chen, L.-S., Y. Ma, J.-L. Maubois, L.-J. Chen, Q.-H. Liu and J.-P. Guo. 2009. Identification of
yeasts from raw milk and selection for some specific antioxidant properties. Int. J. Dairy
Tech. 63: 47–54.
Christiansson, A., A.S. Naidu, I. Nilsson, T. Wadsrtöm and H.-E. Pettersson. 1989. Toxin
production by Bacillus cereus dairy isolates in milk at low temperatures. Appl. Environ.
Microbiol. 55: 2595–2600.
Christiansson, A., J. Bertilsson and B. Svensson. 1999. Bacillus cereus spores in raw milk: Factors
affecting the contamination of milk during the grazing period. J. Dairy Sci. 82: 305–314.
Cisak, E., A. Wójcik-Fatla, V. Zając, J. Sroka, A. Buczek and J. Dutkiewicz. 2010. Prevalence of
tick-borne encephalitis virus (TBEV) in samples of raw milk taken randomly from cows,
goats and sheep in eastern Poland. Ann. Agric. Environ. Med. 17: 283–286.
Cliver, D.O. 1997. Virus transmission via food. Food Technol. 51: 71–78.
Cocolin, L., D. Aggio, M. Manzano, C. Cantoni and G. Comi. 2002. An application of PCR-
DGGE analysis to profile the yeast populations in raw milk. Int. Dairy J. 12: 407–411.
Cocolin, L., N. Innocente, M. Biasutti and G. Comi. 2004. The late blowing in cheese: a new
molecular approach based on PCR and DGGE to study the microbia1 ecology of the
alteration process. Int. J. Food Microbiol. 90: 83–91.
Collins, E.B. 1981. Heat resistant psychrotrophic microorganisms. J. Dairy Sci. 64: 157–160.
Commission Regulation No 853/2004 of the European Parliament and of the Council of 29
November 2004 laying down specific hygiene rules for food of animal origin. Official
J. Eur. Union L 139.
The Microbiology of Raw Milk 59
Commission Regulation No 1881/2006 of 19 December 2006 setting maximum levels for certain
contaminants in foodstuffs. Official J. Eur. Union L 364.
Contreras, A., D. Sierra, A. Sánchez, J.C. Corrales, J.C. Marco, M.J. Paape and C. Gonzalo.
2007. Mastitis in small ruminants. Small Rumin. Res. 68: 145–153.
Contreras, A.G. and J.M. Rodríguez. 2011. Mastitis: Comparative etiology and epidemiology.
J. Mammary Gland Biol. Neoplasia 16: 339–356.
Coorevits, A., V. De Jonghe, J. Vandroemme, R. Reekmans, J. Heyrman, W. Messens, P. De
Vos and M. Heyndrickx. 2008. Comparative analysis of the diversity of aerobic spore-
forming bacteria in raw milk from organic and conventional dairy farms. Syst. Appl.
Microbiol. 31: 126–140.
Corbo, M.R., R. Lancioti, M. Albenzio and M. Sinigaglia. 2001. Occurrence and characterization
of yeasts isolated from milks and dairy products of Apulia region. Int. J. Food Microbiol.
69: 147–152.
Cousin, M.A. 1982. Presence and activity of psychrotrophic microorganisms in milk and dairy
products: a review. J. Food Prot. 45: 172–207.
Council Directive 96/23/EC on measures to monitor certain substances and residues thereof
in live animals and animal products and repealing Directives 85/358/EEC and 86/469/
EEC and Decisions 89/187/EEC and 91/664/EEC. Official J. Eur. Communities L 125/10.
Council Regulation (EEC) No. 2377/90 of 26 June 1990 laying down a Community procedure
for the establishment of maximum residue limits of veterinary medicinal products in
foodstuffs of animal origin. Official J. Eur. Commun. L224/1.
Cremonesi, P., L. Vanoni, T. Silvetti, S. Morandi and M. Brasca. 2012. Identification of Clostridium
beijerinckii, Cl. butyricum, Cl. sporogenes, Cl. tyrobutyricum isolated from silage, raw milk
and hard cheese by a multiplex PCR assay. J. Dairy Res. 79: 318–323.
Cullor, J.S. 1993. The control, treatment and prevention of the various types of bovine mastitis.
Vet. Med. Food Anim. Pract. 88: 571–579.
Cullor, J.S. 1997. HACCP (Hazard Analysis Critical Control Points): is it coming to the dairy?
J. Dairy Sci. 80: 3349–3352.
Davidson, P.M., L.A. Roth and S.A. Gambrel-Lenarz. 2004. Coliform and other indicator
bacteria. pp. 187–226. In: H.M. Wehr and J.F. Frank [eds.]. Standard Methods for
the Examination of Dairy Products. 17th edn. American Public Health Association,
Washington, DC, USA.
De Jonghe, V., A. Coorevits, J. Vandroemme, J. Heyrman, L. Herman, P. De Vos and M.
Heyndrickx. 2008. Intraspecific genotypic diversity of Bacillus species from raw milk.
Int. Dairy J. 18: 496–505.
De Jonghe, V., A. Coorevits, J. De Block, E. Van Coillie, K. Grijspeerdt, L. Herman, P. De Vos
and M. Heyndrickx. 2010. Toxinogenic and spoilage potential of aerobic spore-formers
isolated from raw milk. Int. J. Food Microbiol. 136: 318–325.
De Jonghe, V., A. Coorevits, K. Van Hoorde, W. Messens, A. Van Landschoot, P. De Vos and
M. Heyndrickx. 2011. Influence of storage conditions on the growth of Pseudomonas
species in refrigerated raw milk. Appl. Environ. Microbiol. 77: 460–470.
Delavenne, E., J. Mounier, K. Asmani, J.-L. Jany, G. Barbier and G. Le Blay. 2011. Fungal diversity
in cow, goat and ewe milk. Int. J. Food Microbiol. 151: 247–251.
Delavenne, E., J. Mounier, F. Déniel, G. Barbier and G. Le Blay. 2012. Biodiversity of antifungal
lactic acid bacteria isolated from raw milk samples from cow, ewe and goat over one-year
period. Int. J. Food Microbiol. 155: 185–190.
Deperrois-Lafarge, V. and T. Meheut. 2012. Use of the rpoB gene as an alternative to the V3
gene for the identification of spoilage and pathogenic bacteria species in milk and milk
products. Lett. Appl. Microbiol. 55: 99–108.
Desmasures, N., F. Bazin and M. Guéguen. 1997a. Microbiological composition of raw milk
from selected farms in the Camembert region of Normandy. J. Appl. Microbiol. 83: 53–58.
Desmasures, N., W. Opportune and M. Guéguen. 1997b. Lactococcus spp., yeasts and
Pseudomonas spp. on teats and udders of milking cows as potential sources of milk
contamination. Int. Dairy J. 7: 643–646.
60 Dairy Microbiology: A Practical Approach
De Smet, S., L. De Zutter and K. Houf. 2011. Small ruminants as carriers of the emerging
foodborne pathogen Arcobacter on small and medium farms. Small Rumin. Res. 97:
124–129.
Donaghy, J.A., N.L. Totton and M.T. Rowe. 2004. Persistence of Mycobacterium paratuberculosis
during manufacture and ripening of cheddar cheese. Appl. Environ. Microbiol. 70:
4899–4905.
Donaldson, A.I. 1997. Risks of spreading foot and mouth disease through milk and dairy
products. Rev. Sci. Tech. Off. Int. Epiz. 16: 117–124.
Dore, M.P., A.R. Sepulveda, H. El-Zimaity, Y. Yamaoka, M.S. Osato, K. Mototsugu, A.M. Nieddu,
G. Realdi and D.Y. Graham. 2001. Isolation of Helicobacter pylori from sheep—Implications
for transmission to humans. Am. J. Gastroenterol. 96: 1396–1401.
Elmoslemany, A.M., G.P. Keefe, I.R. Dohoo and B.M. Jayarao. 2009. Risk factors for
bacteriological quality of bulk tank milk in Prince Edward Island dairy herds. Part 2:
Bacterial count-specific risk factors. J. Dairy Sci. 92: 2644–2652.
Elmoslemany, A.M., G.P. Keefe, I.R. Dohoo, J.J. Wichtel, H. Stryhn and R.T. Dingwell. 2010.
The association between bulk tank milk analysis for raw milk quality and on-farm
management practices. Prev. Vet. Med. 95: 32–40.
Eltholth, M.M., V.R. Marsh, S. Van Winden and F.J. Guitian. 2009. Contamination of food
products with Mycobacterium avium paratuberculosis: a systematic review. J. Appl.
Microbiol. 107: 1061–1071.
Eneroth, A., S. Ahrné and G. Molin. 2000. Contamination of milk with Gram-negative spoilage
bacteria during filling of retail containers. Int. J. Food Microbiol. 57: 99–106.
Ercolini, D., F. Russo, I. Ferrocino and F. Villani. 2009. Molecular identification of mesophilic
and psychrotrophic bacteria from raw cow’s milk. Food Microbiol. 26: 228–231.
Eur-Lex website, http://eur-lex.europa.eu/homepage.html.
Fleet, G.H. 1990. Yeasts in dairy products. J. Appl. Bacteriol. 68: 199–211.
Fonteh, F.A., A.S. Grandison and M.J. Lewis. 2002. Variations of lactoperoxidase activity
and thiocyanate content in cows’ and goats’ milk throughout lactation. J. Dairy Res.
69: 401–409.
Fox, L.K., J.H. Kirk and A. Britten. 2005. Mycoplasma mastitis: A review of transmission and
control. J. Vet. Med. B 52: 153–160.
Fox, L.K. 2012. Mycoplasma mastitis causes, transmission and control. Vet. Clin. Food Anim.
28: 225–237.
Franciosi, E., L. Settanni, A. Cavazza and E. Poznanski. 2009. Biodiversity and technological
potential of wild lactic acid bacteria from raw cows’ milk. Int. Dairy J. 19: 3–11.
Frank, J.F. and A.E. Yousef. 2004. Tests for groups of microorganisms. pp. 227–247. In: H.M.
Wehr and J.F. Frank [eds.]. Standard Methods for the Examination of Dairy Products.
17th edn. American Public Health Association, Washington, DC, USA.
Fricker, M., B. Skånseng, K. Rudi, B. Stessl and M. Ehling-Schulz. 2011. Shift from farm to
dairy tank milk microbiota revealed by a polyphasic approach is independent from
geographical origin. Int. J. Food Microbiol. 145: S24–S30.
Fromm, H.I. and K.J. Boor. 2004. Characterization of pasteurized fluid milk shelf-life attributes.
J. Food Sci. 69: 207–214.
Fujimoto, H. 2011. Mycotoxins: Classification, occurrence and determination. In: J.W. Fuquay,
P.F. Fox and P.H.L. McSweeney [eds.]. Encyclopedia of Dairy Sciences, 2nd edition.
Academic Press, New York.
Fujimura, S., T. Kawamura, S. Kato, H. Tateno and A. Watanabe. 2002. Detection of Helicobacter
pylori in cow’s milk. Lett. Appl. Microbiol. 35: 504–507.
Fuquay, J.W., P.F. Fox and P.L.H. McSweeney. 2011. Encyclopedia of dairy sciences, second
edition. Elsevier, Academic Press, New York.
Galvano, F., V. Galofaro and G. Galvano. 1996. Occurrence and stability of aflatoxin M1 in milk
and milk products: A worldwide review. J. Food Prot. 59: 1079–1090.
García-Montoya, I.A., T.S. Cendón, S. Arévalo-Gallegos and Q. Rascón-Cruz. 2012. Lactoferrin
a multiple bioactive protein: An overview. Biochim. Biophys. Acta 1820: 226–236.
The Microbiology of Raw Milk 61
Garde, S., R. Arias, P. Gaya and M. Nuñez. 2011. Occurrence of Clostridium spp. in ovine milk
and Manchego cheese with late blowing defect: Identification and characterization of
isolates. Int. Dairy J. 21: 272–278.
Giacometti, F., A, Serraino, G. Finazzi, P. Daminelli, M.N. Losio, P. Bonilauri, N. Arrigoni,
A. Garigliani, R. Mattioli, S. Alonso, S. Piva, D. Florio, R. Riu and R.G. Zanoni. 2012.
Foodborne pathogens in in-line milk filters and associated on-farm risk factors in
dairy farms authorized to produce and sell raw milk in northern Italy. J. Food Prot. 75:
1263–1269.
Giannino, M.L., M. Aliprandi, M. Feligini, L. Vanoni, M. Brasca and F. Fracchetti. 2009. A
DNA array based assay for the characterization of microbial community in raw milk. J.
Microbiol. Meth. 78: 181–188.
Gilchrist, J.E., J.E. Campbell, C.B. Donnelly, J.T. Peeler and J.M. Delaney. 1973. Spiral plate
method for bacterial determination. Appl. Microbiol. 25: 244–252.
Gill, J.J., P.M. Sabour, J. Gong, H. Yu, K.E. Leslie and M.W. Griffiths. 2006. Characterization of
bacterial populations recovered from the teat canals of lactating dairy and beef cattle by
16S rRNA gene sequence analysis. FEMS Microbiol. Ecol. 56: 471–481.
Gill, C.O., L. Saucier and W.J. Meadus. 2011. Mycobacterium avium subsp. Paratuberculosis in
dairy products, meat, and drinking water. J. Food Prot. 74: 480–499.
Gillet, L., F. Minner, B. Detry, F. Farnir, L. Willems, M. Lambot, E. Thiry, P.-P. Pastoret, F.
Schynts and A. Vanderplasschen. 2004. Investigation of the susceptibility of human cell
lines to bovine herpesvirus 4 infection: Demonstration that human cells can support a
nonpermissive persistent infection which protects them against tumor necrosis factor
alpha-induced apoptosis. J. Virol. 78: 2336–2347.
Goudkov, A.V. and M.E. Sharpe. 1965. Clostridia in dairying. J. Appl. Bacteriol. 28: 63–73.
Grant, I.R. 2005. Zoonotic potential of Mycobacterium avium subsp. paratuberculosis: the current
position. J. Appl. Microbiol. 98: 1282–1293.
Grant, I.R. 2006. Mycobacterium avium subsp. paratuberculosis in foods: current evidence and
potential consequences. Int. J. Dairy Tech. 59: 112–117.
Graumann, P. and M.A. Marahiel. 1996. Some like it cold: response of microorganisms to cold
shock. Arch. Microbiol. 166: 293–300.
Griffiths, M.W., J.D. Phillips and D.D. Muir. 1981. Thermostability of proteases and lipases
from a number of species of psychrotrophic bacteria of dairy origin. J. Appl. Bacteriol.
50: 289–303.
Griffiths, M.W., J.D. Phillips and D.D. Muir. 1987. Effect of low-temperature storage on the
bacteriological quality of raw milk. Food Microbiol. 4: 285–291.
Griffiths, M.W., J.D. Phillips, I.G. West and D.D. Muir. 1988. The effect of extended low-
temperature storage of raw milk on the quality of pasteurized and UHT milk. Food
Microbiol. 5: 75–87.
Grosskopf, J.C. and W.J. Harper. 1974. Isolation and identification of psychrotrophic
sporeformers in milk. Milchwissenschaft 29: 467–470.
Haddadin, M.S., S.A. Ibrahim and R.K. Robinson. 1996. Preservation of raw milk by activation
of the natural lactoperoxidase systems. Food Cont. 7: 149–152.
Hantsis-Zacharov, E. and M. Halpern. 2007. Culturable psychrotrophic bacterial communities in
raw milk and their proteolytic and lipolytic traits. Appl. Environ. Microbiol. 73: 7162–7168.
Hassan, A.N. and J.F. Frank. 2011. Microorganisms associated with milk. pp. 447–457. In:
J.W. Fuquay, P.F. Fox and P.H.L. McSweeney [eds.]. Encyclopedia of Dairy Sciences, 2nd
edition. Academic Press, New York.
Headrick, M.L., S. Korangy, N.H. Bean, F.J. Angulo, S.F. Altekruse, M.E. Potter and K.C.
Klontz. 1998. The epidemiology of raw-milk-associated foodborne disease outbreaks in
the United States, 1973 through 1992. Am. J. Pub. Health. 88: 1219–1221.
Hettinga, K., H. van Valenberg, S. de Vries, S. Boeren, T. van Hooijdonk, J. van Arendonk and
J. Vervoort. 2011. The host defense proteome of human and bovine milk. PLoS One 6:
e19433: 1–8.
Hill, B.M. 1991. Direct microscopic count method. IDF Bullet. 256: 17–20.
62 Dairy Microbiology: A Practical Approach
Hill, B., B. Smythe, D. Lindsay and J. Zealand. 2012. Microbiology of raw milk in New Zealand.
Int. J. Food Microbiol. 157: 305–308.
Hoffmann, S., M.B. Batz and J.G. Morris, Jr. 2012. Annual cost of illness and quality-adjusted life
year losses in the United States due to 14 foodborne pathogens. J. Food Prot. 75: 1292–1302.
Hogan, J.S., K.L. Smith, K.H. Hoblet, D.A. Todhunter, P.S. Schoenberger, W.D. Hueston, D.E.
Pritchard, G.L. Bowman, L.E. Heider, B.L. Brockett and H.R. Conrad. 1989. Bacterial
counts in bedding materials used on nine commercial dairies. J. Dairy Sci. 72: 250–258.
Hogan, J. and K.L. Smith. 2012. Managing environmental mastitis. Vet. Clin. Food Anim. 28:
217–224.
Huck, J.R., N.H. Woodcock, R.D. Ralyea and K.J. Boor. 2007. Molecular subtyping and
characterization of psychrotolerant endospore-forming bacteria in two New York state
fluid milk processing systems. J. Food Prot. 70: 2354–2364.
Huck, J.R., M. Sonnen and K.J. Boor. 2008. Tracking heat-resistant, cold-thriving fluid milk
spoilage bacteria from farm to packaged product. J. Dairy Sci. 91: 1217–1228.
Hutchison, M.L., D.J.I. Thomas, A. Moore, D.R. Jackson and I. Ohnstad. 2005. An evaluation
of raw milk microorganisms as markers of on-farm hygiene practices related to milking.
J. Food Prot. 68: 764–772.
IDF International Dairy Federation. 1988. Code of practice for preservation of raw milk by
lactoperoxidase system. IDF Bulletin No. 234.
IDF International Dairy Federation. 1991a. Methods for assessing the bacteriological quality
of raw milk from the farm. IDF Bulletin No. 256.
IDF International Dairy Federation. 1991b. Significance of the indigenous antimicrobial agents
of milk to the dairy industry. IDF Bulletin No. 256: 2–19.
International Organization for Standardization. 4833: 2003. Microbiology of food and animal
feeding stuffs—Horizontal method for the enumeration of microorganisms—Colony-
count technique at 30 degrees C. International Organization for Standardization. Geneva,
Switzerland.
International Organization for Standardization 707|IDF 050 2008. Milk and milk products
—Guidance on sampling. International Organization for Standardization. Geneva,
Switzerland.
International Organization for Standardization 6887-5 2010. Microbiology of food and animal
feeding stuffs—Preparation of test samples, initial suspension and decimal dilutions for
microbiological examination—Part 5: Specific rules for the preparation of milk and milk
products. International Organization for Standardization. Geneva, Switzerland.
Ivy, R.A., M.L. Ranieri, N.H. Martin, H.C. den Bakker, B.M. Xavier, M. Wiedmann and K.J.
Boor. 2012. Identification and characterization of psychrotolerant sporeformers associated
with fluid milk production and processing. Appl. Environ. Microbiol. 78: 1853–1864.
Jakobsen, M. and J. Narvhus. 1996. Yeasts and their possible beneficial and negative effects
on the quality of dairy products. Int. Dairy J. 6: 755–768.
Jackson, E.E., E.S. Erten, N. Maddi, T.E. Graham, J.W. Larkin, R.J. Blodgett, J.E. Schlesser
and R.M. Reddy. 2012. Detection and enumeration of four foodborne pathogens in raw
commingled silo milk in the United States. J. Food Prot. 75: 1382–1393.
Jayarao, B.M. and L. Wang. 1999. A Study on the prevalence of Gram-negative bacteria in bulk
tank milk. J. Dairy Sci. 82: 2620–2624.
Jones, P.G. and M. Inouye. 1994. The cold-shock response—a hot topic. Mol. Microbiol. 11:
811–818.
Julien, M.-C., P. Dion, C. Lafrenière, H. Antoun and P. Drouin. 2008. Sources of clostridia in
raw milk on farms. Appl. Environ. Microbiol. 74: 6348–6357.
Kefford, B., R. Borland and A.J. Sinclair. 1979. Viral and coxiella contamination of milk. Austr.
J. Dairy Tech. 34: 102–105.
Klanicova, B., I. Slana, P. Roubal, I. Pavlik and P. Kralik. 2012. Mycobacterium avium subsp.
paratuberculosis survival during fermentation of soured milk products detected by culture
and quantitative real time PCR methods. Int. J. Food Microbiol. 157: 150–155.
The Microbiology of Raw Milk 63
Kliem, K.E. and D.I. Givens. 2012. Dairy products in the food chain: their impact on health.
Ann. Rev. Food Sci. Tech. 2: 21–36.
Klijn, N., F.F. Nieuwenhof, J.D. Hoolwerf, C.B. van der Waals and A.H. Weerkamp. 1995.
Identification of Clostridium tyrobutyricum as the causative agent of late blowing in cheese
by species-specific PCR amplification. Appl. Environ. Microbiol. 61: 2919–2924.
Klijn, N., L. Herman, L. Langeveld, M. Vaerewijck, A.A. Wagendorp, I. Huemer and
A.H. Weerkamp. 1997. Genotypical and phenotypical characterization of Bacillus
sporothermodurans strains, surviving UHT sterilization. Int. Dairy J. 7: 421–428.
Koopmans, M. and E. Duizer. 2004. Foodborne viruses: an emerging problem. Int. J. Food
Microbiol. 90: 23–41.
Korhonen, H., P. Marnila and H.S. Gill. 2000. Milk immunoglobulins and complement factors.
Brit. J. Nutr. 84 Suppl. 1: S75–S80.
Kussendrager, K.D. and A.C.M. van Hooijdonk. 2000. Lactoperoxidase: physic-chemical
properties, occurrence, mechanism of action and applications. Br. J. Nutr. 84: S19–S25.
Lachowsky, W.M., W.B. McNab, M. Griffiths and J. Odumeru. 1997. A comparison of the
Bactoscan 8000S to three cultural methods for enumeration of bacteria in raw milk. Food
Res. Int. 30: 273–280.
Lafarge, V., J.-C. Ogier, V. Girard, V. Maladen, J.-Y. Leveau, A. Gruss and A. Delacroix-Buchet.
2004. Raw cow milk bacterial population shifts attributable to refrigeration. Appl. Environ.
Microbiol. 70: 5644–5650.
Langer, A.J., T. Ayers, J. Grass, M. Lynch, F.J. Angulo and B.E. Mahon. 2012. Nonpasteurized
dairy products, disease outbreaks, and State laws-United States, 1993–2006. Emerg. Inf.
Dis. 18: 385–391.
Lavoie, K., M. Touchette, D. St-Gelais and S. Labrie. 2012. Characterization of the fungal
microflora in raw milk and specialty cheeses of the province of Quebec. Dairy Sci. Technol.
DOI 10.1007/s13594–011-0051-4.
Le Bourhis, A.G., J. Doré, J.-P. Carlier, J.-F. Chamba, M.-R. Popoff and J.-L. Tholozan. 2007.
Contribution of C. beijerinckii and C. sporogenes in association with C. tyrobutyricum to
the butyric fermentation in Emmental type cheese. Int. J. Food Microbiol. 113: 154–163.
Leedom, J.M. 2006. Milk of nonhuman origin and infectious diseases in humans. Clin. Inf.
Dis. 43: 610–615.
Leggett, M.J., G. McDonnell, S.P. Denyer, P. Setlow and J.-Y. Maillard. 2012. Bacterial spore
structures and their protective role in biocide resistance. J. Appl. Microbiol. 113: 485–498.
Lehner, A., T. Tasara and R. Stephan. 2005. Relevant aspects of Arcobacter spp. as potential
foodborne pathogen. Int. J. Food Microbiol. 102: 127–135.
LeJeune, J.T. and P.J. Rajala-Schultz. 2009. Unpasteurized milk: A continued public health
threat. Clin. Inf. Dis. 49: 93–100.
Leclercq-Perlat, M.-N. 2011. Camembert, Brie, and Related Varieties. pp. 773–782. In: J.W.
Fuquay, P.F. Fox and P.H.L. McSweeney [eds.]. Encyclopedia of Dairy Sciences, 2nd
edition. Academic Press, New York.
Lievaart, J.J., J.P.T.M. Noordhuizen, E. van Beek, C. van der Beek, A. van Risp, J. Schenkel and
J. van Veersen. 2005. The Hazard Analysis Critical Control Point’s (HACCP) concept as
applied to some chemical, physical and microbiological contaminants of milk on dairy
farms. A prototype. Vet. Quarterly 27: 21–29.
Lindström, M., J. Myllykoski, S. Sivelä and H. Korkeala. 2010. Clostridium botulinum in cattle
and dairy products. Crit. Rev. Food Sci. Nutr. 50: 281–304.
Lukasova, J., J. Vyhnalkova and Z. Pacova. 2001. Bacillus species in raw milk and in the farm
environment. Milchwissenschaft 56: 609–611.
Lundén, J., R. Tolvanen and H. Korkeala. 2004. Human listeriosis outbreaks linked to dairy
products in Europe. J. Dairy Sci. (E. supp.): E6–E11.
Lyne, J. 2011. Technological importance in the dairy industry. In: J.W. Fuquay, P.F. Fox and
P.H.L. McSweeney [eds.]. Encyclopedia of Dairy Sciences, 2nd edition. Academic Press,
New York.
64 Dairy Microbiology: A Practical Approach
Magnusson, M., A. Christiansson and B. Svensson. 2007. Bacillus cereus spores during housing
of dairy cows: Factors affecting contamination of raw milk. J. Dairy Sci. 90: 2745–2754.
Mallet, A., M. Guéguen, F. Kauffmann, C. Chesneau, A. Sesboué and N. Desmasures. 2012.
Quantitative and qualitative microbial analysis of raw milk reveals substantial diversity
influenced by herd management practices. Int. Dairy J. 27: 13–21.
Mansfield, K.L., N. Johnson, L.P. Phipps, J.R. Stephenson, A.R. Fooks and T. Solomon. 2009.
Tick-borne encephalitis virus—a review of an emerging zoonoses. J. Gen. Virol. 90:
1781–1794.
Marchand, S., J. De Block, V. De Jonghe, A. Coorevits, M. Heyndrickx and L. Herman. 2012.
Biofilm formation in milk production and processing environments; influence on milk
quality and safety. Comp. Rev. Food Sci. Food Safety 11: 133–147.
Martin, N.H., M.L. Ranieri, S.C. Murphy, R.D. Ralyea, M. Wiedmann and K.J. Boor. 2011.
Results from raw milk microbiological tests do not predict the shelf-life performance of
commercially pasteurized fluid milk. J. Dairy Sci. 94: 1211–1222.
Marques, S., E. Silva, J. Carvalheira and G. Thompson. 2010. Phenotypic characterization of
Mastitic Prototheca spp. isolates. Res. Vet. Sci. 89: 5–9.
Martins, M.L., C.L.O. Pinto, R.B. Rocha, E.F. de Araújo and M.C.D. Vanetti. 2006. Genetic
diversity of Gram-negative, proteolytic, psychrotrophic bacteria isolated from refrigerated
raw milk. Int. J. Food Microbiol. 111: 144–148.
McKillip, J.L. 2000. Prevalence and expression of enterotoxins in Bacillus cereus and other
Bacillus spp., a literature review. Antonie van Leeuwenhoek 77: 393–399.
Meer, R.R., J. Baker, F.W. Bodyfelt and M.W. Griffiths. 1991. Psychrotrophic Bacillus spp. in
fluid milk products: A review. J. Food Prot. 54: 969–979.
Melville, P.A., N.R. Benites, M. Ruz-Peres and E. Yokoya. 2011. Proteinase and phospholipase
activities and development at different temperatures of yeasts isolated from bovine milk.
J. Dairy Res. 78: 385–390.
Meng, X., T. Karasawa, K. Zou, X. Kuang, X. Wang, C. Lu, C. Wang, K. Yamakawa and S.
Nakamura. 1997. Characterization of a neurotoxigenic Clostridium butyricum strain
isolated from the food implicated in an outbreak of food-borne type E botulism. J. Clin.
Microbiol. 35: 2160–2162.
Morse, P.M., H. Jackson, C.H. McNaughton, A.G. Leggatt, G.B. Landerkin and C.K. Johns.
1968. Investigation of factors contributing to the bacterial count of bulk tank milk. II.
Bacteria in milk from individual cows. J. Dairy Sci. 51: 1188–1191.
Murphy, S.C. and K.J. Boor. 2000. Trouble-shooting sources and causes of high bacteria counts
in raw milk. Dairy Food Environ. Sanit. 20: 606–611.
Munsch-Alatossava, O. and T. Alatossava. 2006. Phenotypic characterization of raw milk-
associated psychrotrophic bacteria. Microbiol. Res. 161: 334–346.
Nada, S., D. Ilija, T. Igor, M. Jelena and G. Ruzica. 2012. Implication of food safety measures
on microbiological quality of raw and pasteurized milk. Food Control 25: 728–731.
Newell, D.G., M. Koopmans, L. Verhoef, E. Duizer, A. Aidara-Kane, H. Sprong, M. Opsteegh,
M. Langelaar, J. Threfall, F. Scheutz, J. van der Giessen and H. Kruse. 2010. Food-borne
diseases—The challenges of 20 years ago still persist while new ones continue to emerge.
Int. J. Food Microbiol. 139: S3–S15.
Nielsen, S.S. and N. Toft. 2009. A review of prevalences of paratuberculosis in farmed animals
in Europe. Prev. Vet. Med. 88: 1–14.
Nickerson, S.C. and R.M. Akers. 2001. Anatomy. pp. 1680–1689. In: J.W. Fuquay, P.F. Fox and
H. Roginski [eds.]. Encyclopedia of Dairy Sciences. Academic Press, New York.
Nörnberg, M.F.B.L., R.S.C. Friedrich, R.D.N. Weiss, E.C. Tondo and A. Brandelli. 2010.
Proteolytic activity among psychrotrophic bacteria isolated from refrigerated raw milk.
Int. J. Dairy Tech. 63: 41–46.
Okura, H., N. Toft and S.S. Nielsen. 2012. Occurrence of Mycobacterium avium subsp.
paratuberculosis in milk at dairy cattle farms: A systematic review and meta-analysis.
Vet. Microbiol. 157: 253–263.
The Microbiology of Raw Milk 65
Oliver, S.P., B.M. Jayarao and R.A. Almeida. 2005. Foodborne pathogens in milk and the
dairy farm environment: Food safety and public health implications. Foodborne Path.
Dis. 2: 115–129.
Oliver, S.P., K.J. Boor, S.C. Murphy and S.E. Murinda. 2009. Food safety hazards associated
with consumption of raw milk. Foodborne Path. Dis. 6: 793–806.
Oliver, S.P., D.A. Patel, T.R. Callaway and M.E. Torrence. 2009b. Developments and future
outlook for preharvest food safety. J. Anim. Sci. 87: 419–437.
Omiccioli, E., G. Amagliani, G. Brandi and M. Magnani. 2009. A new platform for real-time
PCR detection of Salmonella spp., Listeria monocytogenes, and Escherichia coli O157 in milk.
Food Microbiol. 26: 615–622.
O’Toole, D.K. 1983. Methods for the direct and indirect assessment of the bacterial content of
milk. J. Appl. Bacteriol. 55: 187–201.
Paape, M.J., B. Poutrel, A. Contreras, J.C. Marco and A.V. Capuco. 2001. Milk somatic cells and
lactation in small ruminants. J. Dairy Sci. 84(E. Suppl.): E237–E244.
Paduch, J.-H., E. Mohr and V. Krömker. 2012. The association between teat end hyperkeratosis
and teat canal microbial load in lactating dairy cattle. Vet. Microbiol. 158: 353–359.
Pantoja, J.C.F., D.J. Reinemann and P.L. Ruegg. 2009. Associations among milk quality
indicators in raw bulk milk. J. Dairy Sci. 92: 4978–4987.
Peck, M.W. 2009. Biology and genomic analysis of Clostridium botulinum. Adv. Microb. Phys.
55: 183–265.
Pettersson, B., F. Lembke, P. Hammer, E. Stackebrandt and F.G. Priest. 1996. Bacillus
sporothermodurans, a new species producing highly heat-resistant endospores. Int. J. Syst.
Bacteriol. 46: 759–764.
Pinto, U.M., E. de S. Viana, M.L. Martins and M.C.D. Vanetti. 2007. Detection of acylated
homoserine lactones in gram-negative proteolytic psychrotrophic bacteria isolated from
cooled raw milk. Food Control 18: 1322–1327.
Pitt, J.I. 2000. Toxigenic fungi and mycotoxins. Brit. Med. Bull. 56: 184–192.
Polonelli, L., L. Giovati, W. Magliani, S. Conti, S. Sforza, A. Calabretta, C. Casoli, P. Ronzi, E.
Grilli, A. Gallo, F. Masoero and G. Piva. 2011. Vaccination of lactating dairy cows for the
prevention of aflatoxin B1 carry over in milk. PLoS One 6: e26777. doi:10.1371/journal.
pone.0026777.
Postollec, F., A.-G. Mathot, M. Bernard, M.-L. Divanac’h, S. Pavan and D. Sohier. 2012. Tracking
spore-forming bacteria in food: From natural biodiversity to selection by processes. Int.
J. Food Microbiol. 158: 1–8.
Prandini, A., G. Tansini, S. Sigolo, L. Filippi, M. Laporta and G. Piva. 2009. On the occurrence
of aflatoxin M1 in milk and dairy products. Food Chem. Toxicol. 47: 984–991.
Prempeh, H., R. Smith and B. Müller. 2001. Foot and mouth disease: the human consequences.
Brit. Med. J. 322: 565–566.
Quaglia, N.C., A. Dambrosio, G. Normanno, A. Parisi, R. Patrono, G. Ranieri, A. Rella and
G.V. Celano. 2008. High occurrence of Helicobacter pylori in raw goat, sheep and cow milk
inferred by glmM gene: A risk of food-borne infection? Int. J. Food Microbiol. 124: 43–47.
Quigley, L., O. O’Sullivan, T.P. Beresford, R.P. Ross, G.F. Fitzgerald and P.D. Cotter. 2011.
Molecular approaches to analyzing the microbial composition of raw milk and raw milk
cheese. Int. J. Food Microbiol. 150: 81–94.
Raats, D., M. Offek, D. Minz and M. Halpern. 2011. Molecular analysis of bacterial communities
in raw cow milk and the impact of refrigeration on its structure and dynamics. Food
Microbiol. 28: 465–471.
Ramsahoi, L., A. Gao, M. Fabri and J.A. Odumeru. 2011. Assessment of the application of an
automated electronic milk analyzer for the enumeration of total bacteria in raw goat
milk. J. Dairy Sci. 94: 3279–3287.
Ranieri, M.L. and K. Boor. 2009. Bacterial ecology of high-temperature, short-time pasteurized
milk processed in the United States. J. Dairy Sci. 92: 4833–4840.
66 Dairy Microbiology: A Practical Approach
Rasolofo, E.A., D. St-Gelais, G. LaPointe and D. Roy. 2010. Molecular analysis of bacterial
population structure and dynamics during cold storage of untreated and treated milk.
Int. J. Food Microbiol. 138: 108–118.
Raynal-Ljutovac, K., A. Pirisi, R. de Crémoux and C. Gonzalo. 2007. Somatic cells of goat and
sheep milk: Analytical, sanitary, productive and technological aspects. Small Rumin.
Res. 68: 126–144.
Rendos, J.J., R.J. Eberhart and E.M. Kesler. 1975. Microbial populations of teat ends of dairy
cows, and bedding materials. J. Dairy Sci. 58: 1492–1500.
Roberts, R.F. and G.S. Torrey. 1988. Inhibition of psychrotrophic bacterial growth in refrigerated
milk by addition of carbon dioxide. J. Dairy Sci. 71: 52–60.
Rohm, H., F. Eliskases-Lechner and M. Bräuer. 1992. Diversity of yeasts in selected dairy
products. J. Appl. Bacteriol. 72: 370–376.
Romen, F., P. Backes, M. Materniak, R. Sting, T.W. Vahlenkamp, R. Riebe, M. Pawlita, J. Kuzmak
and M. Löchelt. 2007. Serological detection systems for identification of cows shedding
bovine foamy virus via milk. Virol. 364: 123–131.
Ruegg, P.L. 2003. Practical food safety interventions for dairy production. J. Dairy Sci.
86(E. suppl.): E1–E9.
Sanjuan, S., J. Rúa and M. Garcia-Armesto. 2003. Microbial flora of technological interest in
raw ovine milk during 6°C storage. Int. J. Dairy Tech. 56: 143–148.
Scheldeman, P., K. Goossens, M. Rodriguez-Diaz, A. Pil, J. Goris, L. Herman, P. De Vos, N.A.
Logan and M. Heyndrickx. 2004. Paenibacillus lactis sp. nov. isolated from raw and heat-
treated milk. Int. J. Syst. Evol. Microbiol. 54: 885–891.
Scheldeman, P., A. Pil, L. Herman, P. De Vos and M. Heyndrickx. 2005. Incidence and diversity
of potentially highly heat-resistant spores isolated at dairy farms. Appl. Environ.
Microbiol. 71: 1480–1494.
Scheldeman, P., L. Herman, S. Foster and M. Heyndrickx. 2006. Bacillus sporothermodurans
and other highly heat-resistant spore formers in milk. J. Appl. Microbiol. 101: 542–555.
Schofield, G.M. 1992. Emerging food-borne pathogens and their significance in chilled foods.
J. Appl. Bacteriol. 72: 267–273.
Sculion, R., C.S. Harrington and R.H. Madden. 2006. Prevalence of Arcobacter spp. in raw milk
and retail raw meats in northern Ireland. J. Food Prot. 69: 1986–1990.
Schukken, Y., M. Chuff, P. Moroni, A. Gurjar, C. Santisteban, F. Welcome and R. Zadoks. 2012.
The “other” gram-negative bacteria in mastitis. Vet. Clin. Food Anim. 28: 239–256.
Seifu, E., E.M. Buys and E.F. Donkin. 2005. Significance of the lactoperoxidase system in the
dairy industry and its potential applications: a review. Trends Food Sci. Tech. 16: 137–154.
Shah, A.H., A.A. Saleha, M. Murugaiyah, Z. Zunita and A.A. Memon. 2012. Prevalence and
distribution of Arcobacter spp. in raw milk and retail raw beef. J. Food Prot. 75: 1474–1478.
Shah, N.P. 1994. Psychrotrophs in milk: a review. Milchwissenschaft 49: 432–437.
Shaheen, R., B. Svensson, M.A. Andersson, A. Christiansson and M. Salkinoja-Salonen.
2010. Persistence strategies of Bacillus cereus spores isolated from dairy silo tanks. Food
Microbiol. 27: 347–355.
Sierra, D., A. Sánchez, A. Contreras, C. Luengo, J.C. Corrales, C. de la Fe, I. Guirao, C.T.
Morales and C. Gonzalo. 2009. Effect of storage and preservation on total bacterial
counts determined by automated flow cytometry in bulk tank goat milk. J. Dairy Sci.
92: 4841–4845.
Singh, P., A.A. Wani, A.A. Karim and H.-C. Langowski. 2012. The use of carbon dioxide in
the processing and packaging of milk and dairy products: A review. Int. J. Dairy Tech.
65: 161–177.
Sørhaug, T. and L. Stephaniak. 1997. Psychrotrophs and their enzymes in milk and dairy
products: Quality aspects. Trends Food Sci. Tech. 8: 35–41.
Souza, F.N., M.G. Blagitz, C.F.A.M. Penna, A.M.M.P. Della Libera, M.B. Heinemann and
M.M.O.P. Cerqueira. 2012. Somatic cell count in small ruminants: Friend or foe? Small
Rumin. Res. 107: 65–75.
Sperber, W.H. 2005. HACCP does not work from farm to table. Food Control 16: 511–514.
The Microbiology of Raw Milk 67
Streeter, R.N., G.F. Hoffsis, S. Bech-Nielsen, W.P. Shulaw and D.M. Rings. 1995. Isolation of
Mycobacterium paratuberculosis from colostrum and milk of subclinically infected cows.
Am. J. Vet. Res. 56: 1322–1324.
Sturino, J.M. and T.R. Klaenhammer. 2004. Bacteriophage defense systems and strategies for
lactic acid bacteria. Adv. Appl. Microbiol. 56: 331–378.
Suhren, G. and J. Reichmuth. 2000. Interpretation of quantitative microbiological results.
Milchwissenschaft 55: 18–22.
Tabata, S. 2011. Mycotoxins: Aflatoxins and related compounds. In: J.W. Fuquay, P.F. Fox and
P.H.L. McSweeney [eds.]. Encyclopedia of Dairy Sciences, 2nd edition. Academic Press,
New York.
Tacket, C.O., J.P. Narain, R. Sattin, J.P. Lofgren, C. Konigsberg Jr., R.C. Rendtorff, A. Rausa,
B.R. Davis and M.L. Cohen. 1984. A multistate outbreak of infections caused by Yersinia
enterocolitica transmitted by pasteurized milk. J. Am. Med. Assoc. 251: 483–486.
Tasara, T. and R. Stephan. 2006. Cold stress tolerance of Listeria monocytogenes: A review of
molecular adaptive mechanisms and food safety implications. J. Food Prot. 69: 1473–1484.
te Giffel, M.C., A. Wagendorp, A. Herrewegh and F. Driehuis. 2002. Bacterial spores in silage
and raw milk. Antonie van Leeuwenhoek 81: 625–630.
Thomas, S.B. 1966. Sources, incidence and significance of psychrotrophic bacteria in milk.
Milchwissenschaft 21: 270–275.
Thompson, D.I. 1960. A plate loop method for determining viable counts of raw milk. J. Milk
Food Tech. 23: 167–171.
Tomáška, M., G. Suhren, O. Hanuš, H.-G. Walte, A. Slottová and M. Hofericová. 2006. The
application of flow cytometry in determining the bacteriological quality of raw sheep’s
milk in Slovakia. Lait. 86: 127–140.
Tomasula, P.M. and R.P. Konstance. 2004. The survival of foot-and-mouth disease virus in raw
and pasteurized milk and milk products. J. Dairy Sci. 87: 1115–1121.
Tormo, H., C. Agabriel, C. Lopex, D. Ali Haimoud Lekhal and C. Roques. 2011. Relationship
between the production conditions of goat’s milk and the microbial profiles of milk. Int.
J. Dairy Sci. 6: 13–28.
Uraz, G. and S. Çitak. 1998. The isolation of Pseudomonas and other Gram(–) psychrotrophic
bacteria in raw milks. J. Basic Microbiol. 2: 129–134.
Vacheyrou, M., A.-C. Normabd, P. Guyot, C. Cassagne, R. Piarroux and Y. Bouton. 2011.
Cultivable microbial communities in raw cow milk and potential transfers from stables
of sixteen French farms. Int. J. Food Microbiol. 146: 253–262.
Vadillo Machota, S., M.J. Paya Vicens, M.T. Cutuli de Simon and S. Suarez Fernandez. 1987.
Raw milk mycoflora. Milchwissenschaft 42: 20–22.
Van Brandt, L., K. Coudijzer, L. Herman, C. Michiels, M. Hendrickx and G. Vlaemynck. 2011a.
Survival of Mycobacterium avium ssp. paratuberculosis in yoghurt and in commercial
fermented milk products containing probiotic cultures. J. Appl. Microbiol. 110: 1252–1261.
Van Brandt, L., I. Van der Plancken, J. De Block, G. Vlaemynck, E. Van Coillie, L. Herman
and M. Hendrickx. 2011b. Adequacy of current pasteurization standards to inactivate
Mycobacterium paratuberculosis in milk and phosphate buffer. Int. Dairy J. 21: 295–304.
Van Driessche, E., K. Houf, J. Van Hoof, L. De Zutter and P. Vandamme. 2003. Isolation of
Arcobacter species from animal feces. FEMS Microbiol. Lett. 229: 243–248.
Van Netten, P., A. Van de Moosdijk, P. Van Hoensel, D.A.A. Mossel and I. Perales. 1990.
Psychrotrophic strains of Bacillus cereus producing enterotoxin. J. Appl. Bacteriol. 69:
73–79.
Van Schothorst, M. and T. Kleiss. 1994. HACCP in the dairy industry. Food Control 5: 162–166.
Vasavada, P.C. 1993. Rapid methods and automation in dairy microbiology. J. Dairy Sci. 76:
3101–3113.
Verdier-Metz, I., V. Michel, C. Deblès and M.-C. Montel. 2009. Do milking practices influence
the bacterial diversity of raw milk? Food Microbiol. 26: 305–310.
68 Dairy Microbiology: A Practical Approach
Dairy Pathogens:
Characteristics and Impact
Photis Papademas* and Maria Aspri
INTRODUCTION
Milk and dairy products can be an ideal medium for the growth of a variety
of microorganisms, both pathogenic and spoilage. Even though milk
and milk products are amongst the safest food worldwide and account
only a small percentage of all food-borne diseases, they have an inherent
potential for causing illness as they are potentially the source of a very
broad range of microbial, chemical, and physical hazards. The presence of
food-borne pathogens in milk and milk products is due to direct contact
with contaminated sources in the dairy farm environment, to excretion
from the udder of an infected animal and during processing and handling.
Some important pathogens for milk and milk products include
Salmonella spp., Listeria monocytogenes, Staphylococcus aureus, Campylobacter
spp., pathogenic Escherichia coli, Cronobacter sakazakii and will be discussed
in detail in this chapter.
Figure 1. Sources of Contamination of Milk and Dairy Products (After: Oliver et al. 2005)
Listeria monocytogenes
Introduction
Characteristics
Milkborne Pathogens
Organism Disease Characteristics Associated Foods
Salmonella spp. Typhoid like fever, headache, Raw milk, dairy products
abdominal pain, diarrhea, Others: Eggs, meat fish,
nausea, vomiting, constipation, poultry, shrimp, salad dressings
malaise, chronic symptoms (especially home-made that
(arthritis) contain raw eggs)
Listeria Nausea, vomiting, diarrhea, Fresh soft cheeses, raw milk,
monocytogenes fever, influenza like symptoms, inadequately pasteurised milk,
meningitis, encephalitis. In Others: Ready to eat deli-meals,
pregnant women can lead to hot dogs, raw and smoked fish
spotaneous abortion, premature
delivery, or still birth
Staphylococcus Nause, vomiting, abdominal Milk and dairy products Others:
aureus cramps, retching, prostration Meat and meat products, salads
(chicken, potato, egg and
makaroni), cream pastries
Campylobacter jejuni Diarrhea, abdominal pain Raw milk Others: Raw
and cramps, fever, vomiting, and undercooked poultry,
headache contaminated water, shellfish
Bacillus cereus Diarrheal toxin: Watery diarrhea, Milk, dairy products Others:
abdominal cramps, nausea Meats, vegetables, rice, pasta,
Emetic toxing: nausea and soups, fish
vomiting, diarrhea may be
present
Escherichia coli Watery diarrhea, abdominal Raw milk Others: Contaminated
cramps, low-grade fever, nausea, water, undercooked ground beef,
malaise raw apple juice and cider, alfalfa
sprouts, cut melons
Cronobacter sakazakii Meningitis, Enteritis Powdered Infant formula
(BHI), trypticase-soy broth with 0.6% yeast extract (TSBYE), Luria broth
(LB), etc. L. monocytogenes is catalase positive, oxidase negative and ferments
rhamnose, dextrose, esculin, and maltose, but does not ferment xylose and
manitol. These biochemical properties are used for the differentiation of
L. monocytogenes from other members of the genus Listeria.
The organism’s ability to grow and reproduce in harsh conditions makes
it a food-borne pathogen of great concern. Optimum growth temperature is
30–37°C, although L. monocytogenes grows reasonably well at temperatures
over a wide range of temperature from 0–42°C. The growth rate reduces
as the growth temperature decreases. The pH range for growth is 5–9 with
an optimum pH of 6–8. However, these values depend on the acidulant,
strain, and temperature. L. monocytogenes is also quite salt tolerant being
able to grow in 10% sodium chloride. The organism is ubiquitous in the
environment. It has been isolated from fresh and salt water, soil, sewage
sludge, decaying vegetation and silage.
Food-borne Illness
Detecting L. monocytogenes
Sample
Plating
Purification
Confirmation
Purified colonies are confirmed with standard tests:
• Gram staining
• Motility test
• Carbohydrate fermentation test
• D-hemolysis and CAMP test (lysis tests)
Figure 2. ISO 11290 for detection of L. monocytogenes in food samples (After: Scharlau 2007).
Color image of this figure appears in the color plate section at the end of the book.
Dairy Pathogens: Characteristics and Impact 75
Dairy products such as raw and pasteurized milk and soft cheeses have
been associated with a number of major outbreaks of listeriosis (Kasalica
2011), since Listeria population may be extremely acid and salt resistant.
L. monocytogenes’ association with soft cheeses is due to the cheese
ripening process. It survives poorly in unriped soft cheeses such as cottage
cheese but well in products such as Camembert and Brie (especially if
made from raw milk). During this ripening process microbial utilization
of lactate and release of amines increase the surface pH allowing Listeria
spp. to multiply. Hard cheese such as Parmesan (Parmigiano Reggiano)
does not favour growth (Yousef and Marth 1990) while in white-brined
cheeses, i.e., Feta, the acidic environment of the cheese (pH 4.6) will inhibit
the growth of Listeria spp. but the pathogen will survive even for 90 days
(Papageorgiou and Marth 1989). It was also reported by Belessi et al. (2008)
that the growth of L. innocua in Feta cheese was assisted by the presence
of fungi. The findings indicated that the growth of fungi on the surface of
Feta cheese and yogurt may compromise the safety of these products by
enhancing survival of the bacterium. Particularly, when fungi increase the
pH of Feta cheese, L. innocua demonstrates better survival and prolonged
storage may raise concerns for the development of acid-resistant Listeria
populations.
In high-scalding cheeses, i.e., Mozzarella, Halloumi the bacteria will not
survive the stretching/cooking temperatures. For fresh Halloumi cheese
(pH 6.2) post-scalding contamination may result in a favorable environment
for Listeria spp. growth therefore, careful handling of the cheese is crucial.
Gougouli et al. (2008) showed that in ice cream the freezing conditions
did not cause a severe stress in L. monocytogenes cells capable of leading
to a significant “additional” lag phase during the subsequent growth
of the pathogen at chilling conditions. Additionally they observed that
under freezing conditions, no significant changes in the population of the
pathogen were observed throughout a 90-d storage period for either of the
inoculum levels tested. The above statements highlight the importance of
post-pasteurisation hygiene conditions to be employed in all dairy factories.
Salmonella spp.
Introduction
Salmonella are one of the most important food-borne pathogens and have
been recognized for over 100 years as the cause of illnesses ranging from
mild to severe food poisoning (gastroenteritis), and even more severe
typhoid (enteric fever), paratyphoid, bacteraemia, septicaemia and a variety
of associated longer-term conditions (sequelae).
76 Dairy Microbiology: A Practical Approach
Characteristics
Food-borne Illness
Color image of this figure appears in the color plate section at the end of the book.
have been related to the same plant, suggesting that the strain had persisted
in the plant and repeatedly contaminated milk after pasteurization.
Staphylococcus aureus
Introduction
Characteristics
Food-borne Illness
Detecting S. aureus
The most successful and widely used medium for S. aureus is called
Baird Parker. This contains tellurite, glycine and lithium chloride as
selective agents, pyruvate and egg yolk to assist the recovery of damaged
cells. Reduction of the tellurite by S. aureus gives characteristic shiny,
jet-black colonies which are surrounded by a zone of clearing resulting
82 Dairy Microbiology: A Practical Approach
from hydrolysis of the egg yolk lipovitellenin and often with an inner
cloudy zone due to the precipitation of fatty acid salts. The appearance
of the colonies on Baird-Parker agar gives a presumptive identification of
S. aureus which is confirmed through the production of coagulase (Asperger
and Zangerl 2003).
Molecular techniques based on the polymerase chain reaction (PCR)
have been developed using specific primers directed against a variety of
sequences in the S. aureus genome including genes for thermostable nuclease
and enterotoxins. There are also kits available for detection based on nucleic
acid hybridization. Enterotoxins can be detected based on immunological
methods following extraction from the food matrix, and there are numerous
kits available using maenzyme linked immunoassay or latex agglutination.
powder and ice cream production critical stages are the pre-drying and pre-
freezing periods in which S. aureus can multiply if temperature is favorable.
Cronobacter sakazakii
Introduction
Characteristics
Food-borne Illness
Detecting C. sakazakii
Bacillus cereus
Introduction
There are several Bacillus species that have been responsible for food-borne
illness, although the only one that is frequently involved is Bacillus cereus.
Hauge (1955) was the first to establish B. cereus as a food poisoning organism.
Characteristics
The Bacillus cereus group is a very homogeneous cluster within the genus
Bacillus and consists of six species: B. cereus, B. thuringiensis, N. anthracis,
B. mycoides, N. pseudomycoides and B. weihenstephanensis (Ehling-Schulz et
al. 2004). B. cereus is a Gram-positive, catalase positive motile, facultative,
aerobic sporeformer. Its cells are rod-shaped, hence the name “Bacillus”
(“rod”). The name “cereus” (“wax”) was given because its colonies have
a waxy appearance on agar plates. Dimensions of vegetative cells are
typically 1.0–1.2 µm in diameter by 3.0–5.0 µm in length, with rounded or
square ends and are often arranged in pairs or chains (Arnesen et al. 2008).
The temperature range for the growth of B. cereus has been reported to
be between 8–55°C with the optimum being in the range of 28–35°C. The
range of pH for growth of B. cereus has been reported to be 4.9–9.3 with the
optimum being 6.0–7.0, and a water activity (aw) greater than 0.94.
Dairy Pathogens: Characteristics and Impact 87
Food-borne Illness
al. 2013). The wide range of infective dose is partly due to the consumption
of spores which can survive the acidic environment in the stomach and
partly due to the ability of different strains to produce different amounts
of enterotoxins. Therefore, any food containing more than 103 B. cereus per
gram cannot be considered completely safe for consumption.
Detecting B. cereus
Campylobacter spp.
Introduction
Campylobacter spp. has been known as a veterinary problem from the early
twentieth century. Campylobacter jejuni subsp. jejuni (referred to as C. jejuni
throughout this chapter) is a major cause of bacterial human gastroenteritis
in both developed and developing nations. It is one of the most important
milk-borne pathogens that has come to rival or even surpass Salmonella
as an etiological agent of human gastroenteritis worldwide.
Dairy Pathogens: Characteristics and Impact 89
Characteristics
Food-borne Illness
Figure 4. Procedure for Isolation and Identification of Campylobacter spp. from Milk (After:
United States Food and Drug Administration).
Color image of this figure appears in the color plate section at the end of the book.
Yersinia spp.
Introduction
Characteristics
Y. enterocolitica are included in the genus Yersinia, which are classified into
the family Enterobacteriaceae, a group of gram-negative, oxidase-negative,
catalase positively, non spore-forming and facultatively anaerobic bacteria.
Y. enterocolitica are rods or coccobacilli of 0.5–0.8 x 1–3 µm in size. Of the 11
species, only 3 species of Yersinia are recognised to be pathogenic to humans,
the plague bacillus, Yersinia pestis, and two that cause gastroenteritis;
Y. enterocolitica and Y. pseudotuberculosis (Bottone 1997). Strains belonging
to Y. enterocolitica are urease positive and can be differentiated from
Y. pseudotuberculosis with a positive result for fermentation of sucrose, and
negative reactions for rhamnose and melibiose fermentation. Most of the
strains are motile at 25°C but non-motile at 37°C. It can be isolated from
many sources, such as food, drinking water, sewage, environmental water
and human clinical samples with a world-wide distribution.
Y. enterocolitica can grow over a pH range of 4–10, generally with an
optimum pH of 7–8 depending on the acidulant used, environmental
temperature, composition of the medium, and growth phase of the
bacteria. It is a psychotropic bacterium therefore it has the ability to grow at
temperatures below 4°C, but the optimum growth temperature is 28–30°C.
Food-borne Illness
Human clinical infections with this species ensue after ingestion of the
microorganisms in contaminated food or water or by direct inoculation
through blood transfusion (Sabina et al. 2011). Infection with Y. enterocolitica
can cause a variety of symptoms depending on the age of the person
Dairy Pathogens: Characteristics and Impact 93
Detection
Y. entercolitica has been isolated from raw cow’s milk in Mexico City
(Bernardino-Varo et al. 2013) while other authors (Ackers et al. 2000)
have reported that the same pathogen was associated with pasteurized
94 Dairy Microbiology: A Practical Approach
bottled milk sold from a local dairy. This incident was connected to
postpasteurization contamination, indicating the need for controls and
training/awareness of dairy farmers. Tacket et al. (1984) stated that
outbreaks of enteric disease caused by pasteurized milk are rare; although
the ability of Y. enterocolitica to grow in milk at refrigeration temperatures
makes pasteurized milk a possible vehicle for virulent Y. enterocolitica.
Shigella spp.
Introduction
The genus Shigella was first reported by the Japanese microbiologist Kiyoshi
in 1898. It was classified into four species based on the O antigen; Shigella
dysenteriae, Shigella flexneri, Shigella boydii and Shigella sonnei which were
also known as Shigella subgroups A, B, C and D respectively.
Characteristics
Food-borne Illness
Figure 5. Detection of Shigella spp. in Food Samples (After: AES Chemunex 2008).
Color image of this figure appears in the color plate section at the end of the book.
4.3%, respectively. The results may show a low prevalence but since the
bacterium may cause dysentery it should not be underestimated.
Escherichia coli
Introduction
Escherichia coli, originally called “Bacterium coli commune”, was first isolated
from children feces in 1885 by the German bacteriologist Theodor Escherich.
Nowadays it is the one of the most understood bacteria and is a common
inhabitant of the gastrointestinal tract of humans and animals.
Characteristics
Food-borne Illness
Even though E. coli is a group of harmless bacteria that are most often
used as indicator organisms for fecal contamination and breaches in
hygiene, there are some strains that have acquired virulence factors
that has allowed them to cause serious disease especially diarrhea.
Pathogenic Escherichia coli is categorized into the following six categories
according to their virulence properties, clinical manifestations, pathogenic
mechanisms, clinical syndromes and O:H serogroups: enteropathogenic
E. coli (EPEC), enterotoxigenic E. coli (ETEC), enterohaemorrhagic E. coli
(EHEC), enteroinvasive E. coli (EIEC), diffusely adherent E. coli (DAEC),
and enteroaggregative E. coli (EAEC) (Jafari et al. 2012).
98 Dairy Microbiology: A Practical Approach
DAEC are a major cause of urinary tract infections worldwide, but its role
as a causative agent of diarrhea is not completely understood.
Detecting E. coli
Shiga toxin producing E. coli (STEC) are an important cause of human food-
borne outbreaks. The consumption of raw milk dairy products may be an
important route of STEC infection. There are 2 STEC strains of serotypes
O157:H7 and O26:H11, and Miszczycha et al. 2014, reported that the survival
of E. coli O26:H11 was 13 times greater than that of E. coli O157:H7 at the
end of digestion in contaminated raw milk cheeses. E. coli O157:H7 has also
been isolated from dairy farms, even though a survey in Italy reported from
Conedera et al. 2004 showed that none of the dairy products collected (2948
including mozzarella cheese, pasteurized and raw bovine and ovine milk)
had tested positive for VTEC E. coli 0157:H7. A very comprehensive review
on Shiga-toxin-producing Escherichia coli (STEC) and their significance in
dairy production has been published by Farrokh et al. (2013), where the
authors review the survival of STEC during cheese making and in particular
the factors affecting the growth of STEC during cheesemaking (i.e., salt,
pH, temperature, water activity), behaviour of STEC during cheesemaking.
As a conclusion, the International Dairy Federation in its IDF Factsheet
(2012) states that in the processing industry, milk pasteurization is
recommended, since pasteurization will eliminate pathogens including
STEC. Good hygienic practices at the processing facility are also to be
applied in order to prevent post-pasteurization contamination. At farm
level, strict hygiene practices should be followed during the production and
transport of raw milk. Additional hygiene practices are to be implemented
in the production of raw milk cheese and other dairy products.
Mycobacterium spp.
Introduction
Characteristics
Food-borne Illness
MAP and its association (or not) to human Crohn’s Disease is a debate
which started in 1984, when a Mycobacterium similar to the agent of
Johne’s disease was isolated and cultivated from the intestinal tissue of three
patients with Crohn’s disease. Crohn’s disease is a chronic inflammatory
bowel disease which currently is of unknown cause. The most widely
accepted hypothesis is that Crohn’s disease occurs as a result of an abnormal
immune response within the gastrointestinal mucosa. Grant (2006) reports
that transmission to humans via consumption of animal-derived foods is a
distinct possibility as milk, other dairy products, beef and water have been
identified as possible food vehicles of transmission. To date, viable MAP
has been cultured from raw cows’, sheep and goats’ milk, retail pasteurized
cows’ milk, and some retail cheeses in several countries during recent
studies. MAP has not been isolated from retail beef to date, although limited
testing has been carried out.
The public health consequences, if any, of low numbers of viable MAP
being periodically consumed by susceptible individuals are uncertain.
Moreover, an association between MAP and Crohn’s disease is not proven,
but neither can it be discounted on the basis of current evidence.
102 Dairy Microbiology: A Practical Approach
Dairy Outbreaks
Dairy products have frequently been implicated in food poisoning
outbreaks. There are two main reasons for that; one is the fact that the gross
chemical composition for most of the dairy products favors the growth and
survival of microorganisms (i.e., high protein content, presence of lactose,
available moisture—water activity, aw), and second, the sheer volumes of
dairy products consumed by almost all population groups making dairy
products a very good vehicle for microorganisms. Fortunately, processing of
milk and milk products (i.e., pasteurization, ultra high temperature, drying,
evaporating, filtrating of bacteria, fermenting/acidification) minimizes risks
either by posing hurdles for bacteria to grow (low pH), decreasing available
water (lowering water activity), or by destroying bacteria (temperature).
Nonetheless, milk and other dairy products (i.e., cheese) could also be
consumed raw (according to the provisions of legislation EC 853/2004);
therefore strict hygienic conditions must apply; at farm, during milk
transportation, during manufacture at the factory and at retail level in order
to avoid food poisoning outbreaks. Table 3 below summarizes recent alert
Dairy Pathogens: Characteristics and Impact 103
cases as reported by the Rapid Alert System for Food and Feed (RASFF),
European Union concerning milk and milk products and certain common
pathogens.
Langer et al. (2012) published a study (results are shown in Table 4)
where they describe the outbreaks related to dairy products both raw
and pasteurized in the USA between the years 1993–2006. They reported
that the causative agent was identified for all 73 outbreaks involving raw
dairy products; all were caused by bacteria. One outbreak was caused by
Campylobacter spp. and shigatoxin producing Escherichia coli. Among the
remaining 72 outbreaks, 39 (54%) were caused by Campylobacter spp., 16
(22%) by Salmonella spp., 9 (13%) by shiga-toxin producing E. coli, 3 (4%)
by Brucella spp., 3 (4%) by Listeria spp., and 2 (3%) by Shigella spp. Among
the 30 outbreaks involving pasteurized dairy products for which the
Table 3. Milk and milk products involved in recent alert cases (RASFF, EU).
Raw
Fluid milk 46 930 71 0
Cheese 27 641 131 2
Total 73 1,571 202 2
Pasteurized
Fluid milk 10 2,098 20 0
Cheese 38 744 17 1
Total 48 2,842 37 1
All dairy 121 4,413 239 3
#
The criterion does not apply to products intended for further processing in the food industry.
* E. coli is used here as an indicator for the level of hygiene.
106 Dairy Microbiology: A Practical Approach
** For cheeses which are not able to support the growth of E. coli, the E. coli count is usually
the highest at the beginning of the ripening period, and for cheeses which are able to
support the growth of E. coli, it is normally at the end of the ripening period.
+
Excluding cheeses where the manufacturer can demonstrate, to the satisfaction of the
competent authorities, that the product does not pose a risk of staphylococcal enterotoxins.
#
The criterion does not apply to products intended for further processing in the food industry.
##
m = M.
Dairy Pathogens: Characteristics and Impact 107
++
Only ice creams containing milk ingredients.
+
m=M.
$
Parallel testing for Enterobacteriaceae and E. sakazakii shall be conducted, unless a
correlation between these micro-organisms has been established at an individual plant
level. If Enterobacteriaceae are detected in any of the product samples tested in such a plant,
the batch has to be tested for E. sakazakii. It shall be the responsibility of the manufacturer
to demonstrate to the satisfaction of the competent authority whether such a correlation
exists between Enterobacteriaceae and E. sakazakii.
108 Dairy Microbiology: A Practical Approach
Conclusions
Dairy pathogens are of paramount importance as they can cause major food
scares to the consumers. The fact that dairy products are widely consumed,
daily, in many forms by the majority of the world’s population including
special groups (i.e., infants, the elderly, and the immune-compromised)
makes their strict control inherent. We should also be aware of emerging
dairy-related pathogens (i.e., Mycobacterium avium subsp. paratuberculosis)
and assess their impact to human health, apart from the obvious food
poisoning. Surveys from underdeveloped countries as well as from the
developed part of the world should continue to yield results in order to
enhance our understanding on dairy pathogens that up to now may not have
been given the attention they required. Finally, the acquired knowledge on
prevalence and impact of dairy pathogens in the food chain would enable
the scientific community to make informed decisions regarding issues such
as the direct selling (or not) of raw milk, from animal species.
Dairy Pathogens: Characteristics and Impact 109
References Cited
Ackers, M.L., S. Schoenfeld, J. Markman, M.G. Smith, M.A. Nicholson, W. DeWitt, D.N.
Cameron, P.M. Griffin and L. Slutsker. 2000. An outbreak of Yersinia enterocolitica O:8
infections associated with pasteurized milk. J. Infect Dis. 181(5): 1834–1837.
AES Chemunex. Culture Media for the Food Industry, Rapid Microbiology Company, France.
Ahmed, A.M. and T. Shimamoto. 2014. Isolation and molecular characterization of Salmonella
enterica, Escherichia coli O157: H7 and Shigella spp. from meat and dairy products in
Egypt. International Journal of Food Microbiology 168-169: 57–62.
Andersson, M.A., E.L. Jaaskelainen, R. Shaheen, T. Pirhonen, L.M. Wijnands and M.S. Salkinoja-
Salonen. 2004. Sperm bioassay for rapid detection of cereulide-producing Bacillus cereus
in food and related environments. Int. J. Food Microbiol. 94: 175–183.
Argudín, M.A., M.C. Mendoza and M.R. Rodicio. 2010. Food Poisoning and Staphylococcus
aureus enterotoxins. Toxins 2(7): 1751–1773.
Arnesen, L.P., A. Fagerlund and P.E. Granum. 2008. From soil to gut: Bacillus cereus and its
food poisoning toxins, FEMS Microbiol. Rev. 32: 579–606.
Arsalam, A., Z. Anwar, I. Ahmad, Z. Shad and S. Ahmed. 2013. Cronobacter sakazakii: an
emerging contaminant in pediatric infant milk formula. Int. Res. J. Pharm. 4(4): 17–22.
Asperger, H. and P. Zangerl. 2003. Staphylococcus aureus. pp. 2563–2569. In: H. Roginski, J.
Fuquay and P. Fox [eds.]. Encyclopedia of Dairy Science. San Diego: Academic Press.
Belessi, C.I.A., S. Papanikolaou, E.H. Drosinos and P.N. Skandamis. 2008. Survival and Acid
Resistance of Listeria innocua in Feta Cheese and Yogurt, in the Presence or Absence of
Fungi. J. Food Prot. 4: 742–749.
Bernardino-Varo, L., E.I. Quiñones-Ramírez, F.J. Fernandez and C. Vazquez-Salinas. 2013.
Prevalence of Yersinia enterocolitica in raw cow’s milk collected from stables of Mexico
City. J. Food Prot. 75(4): 694–698.
Botsaris, G., I. Slana, M. Liapi, C. Dodd, C. Economides, C. Rees and I. Pavlik. 2010. Rapid
detection methods for viable Mycobacterium avium subspecies paratuberculosis in milk
and cheese. Int. J. Food Microbiol. 141: S87–S90.
Bottone, E.J. 1997. Yersinia enterocolitica: the charisma continues. Clin. Microbiol. Rev. 10(2):
257–276.
Bottone, E.J. 2010. Bacillus cereus, a Volatile Human Pathogen. Clin. Microbiol. Rev. 23(2): 382.
Ceuppens, S., N. Boon and M. Uyttendaele. 2013. Diversity of Bacillus cereus group strains is
reflected in their broad range of pathogenicity and diverse ecological lifestyles. FEMS
Microbiol. Ecol. 84: 433–450.
Commission Regulation (EC) No. 853/2004 of the European Parliament and of the Council
laying down specific hygiene rules for the hygiene of foodstuffs. OJ L139.
Commission Regulation (EC) No 2073/2005 of 15 November 2005 on microbiological criteria
for foodstuffs. OJ L338.
Commission Regulation (EC) No 1441/2007 of 5 December 2007 amending Regulation (EC)
No 2073/2005 on microbiological criteria for foodstuffs. OJ L322/12.
Commission Regulation (EU) No. 365/2010 amending Regulation (EC) No. 2073/2005 on
microbiological criteria for foodstuffs as regards Enterobacteriaceae in pasteurised milk
and other pasteurised liquid dairy products and Listeria monocytogenes in food grade
salt. OJ L107. 9–11.
Conedera, G., P. Dalvit, M. Martini, G. Galierod, M. Gramaglia, E. Goffredo, G. Loffredo, S.
Morabito, D. Ottaviani, F. Paterlini, G. Pezzotti, M. Pisanu, P. Semprini and A. Caprioli.
2004. Verocytotoxin-producing Escherichia coli O157 in minced beef and dairy products
in Italy. Int. J. Food Microbiol. 96(1): 67–73.
De Buyser, M.L., B. Dufour, M. Mair and V. Lafarge. 2001. Implication of milk and milk
products in food-borne diseases in France and in different industrialised countries. Int.
J. Food Microbiol. 67(1-2): 1–17.
110 Dairy Microbiology: A Practical Approach
Dumen, E. 2010. Cronobacter sakazakii (Enterobacter sakazakii): Only An Infant Problem? Kafkas
Univ. Vet. Fak. Derg. 16: S171–S178.
Ehling-Schulz, M., M. Fricker and S. Scherer. 2004. Bacillus cereus, the causative agent of an
emetic type of food-borne illness. Mol. Nutr. Food Res. 48: 479–487.
El-Gazzar, F.E. and E.H. Marth. 1992. Salmonellae, salmonellosis, and dairy foods: a review.
J. Dairy Sci. 75: 2327–2343.
El Sharoud, W.M. 2009. Prevalence and survival of Campylobacter in Egyptian dairy products.
Food Res. Int. 42(5-6): 622–626.
European Food Safety Authority. 2003. Opinion of the Scientific Committee on Veterinary
Measures relating to public health on Staphulococcal enterotoxins in milk products,
particularly cheeses.
Farmer, J.J., M.A. Asbury, F.W. Hickman and D.J. Brenner. 1980. Enterobacter sakazakii, new
species of Enterobacteriaceae isolated from clinical specimens. Int. J. Syst. Bacteriol. 30:
569–584.
Farrokh, C., K. Jordan, F. Auvray, K. Glass, H. Oppegaard, S. Raynaud, D. Thevenot, R.
Condron, K. De Reu, A. Govaris, K. Heggum, M. Heyndrickx, J. Hummerjohann, D.
Lindsay, S. Miszczycha, S. Moussiegt, K. Verstraete and O. Cerf. 2013. Review of Shiga-
toxin-producing Escherichia coli (STEC) and their significance in dairy production. Int. J.
Food Microbiol. 162(2): 190–212.
Food and Drug Administration. 2002. Isolation and enumeration of Enterobacter sakazakii from
dehydrated powdered infant formula.
Food Safety news. Dairy-Related Outbreaks, Illnesses, Recalls: 2010 to Present. http://www.
foodsafetynews.com/2012/04/dairy-related-outbreaks-illnesses-recalls-2010-to-present.
Date accessed 17/3/2014.
Fredriksson-Ahomaa, M. and H. Korkeala. 2003. Low occurrence of pathogenic Yersinia
enterocolitica in clinical, food, and environmental samples: a methodological problem.
Clin. Microbiol. Rev. 16: 220–229.
Gasanov, U., D. Hughes and P.M. Hansbro. 2005. Methods for the isolation and identification of
Listeria spp. and Listeria monocytogenes: a review. FEMS Microbiology Reviews 29: 851–875.
Glickman, M.S. and W.R. Jacobs. 2001. Microbial Pathogenesis Review of Mycobacterium
tuberculosis: dawn of discipline. Cell. 104: 477–485.
Gougouli, M., A.S. Angelidis and K. Koutsoumanis. 2008. A study on the kinetic behavior of
Listeria monocytogenes in ice cream stored under static and dynamic chilling and freezing
conditions. J. Dairy Sci. 91(2): 523–530.
Grant, I.R. 2006. Mycobacterium avium subsp. paratuberculosis in foods: Current evidence and
potential consequences. Int. J. Dairy Technol. 59: 112–117.
Granum, P.E. and T. Lund. 1997. Bacillus cereus and its food poisoning toxins. FEMS Microbiol.
Lett. 157: 223–228.
Gurtler, J.B., J.L. Kornacki and L.R. Beuchat. 2005. Enterobacter sakazakii: A coliform of increased
concern to infant health. Int. J. Food Microbiol. 104: 1–34.
Hammer, P., C. Kiesner and H.G. Walte. 2014. Short communication: Effect of homogenization
on heat inactivation of Mycobacterium avium subspecies paratuberculosis in milk. J.
Dairy Sci. 97(4): 2045–2048.
Hariharan, H., G.A. Murphey and I. Kempf. 2004. Campylobacter jejuni: Public health hazards
and potential control methods in poultry: a review Vet. Med. – Czech 49(11): 441–446.
Hauge, S. 1955. Food Poisoning caused by aerobic spore-forming bacilli. J. Applied Bacter.
18(3): 591–595.
Heuvelink, A.E., C. Van Heerwaarden, A. Zwartkruis-Nahuis, J.J Tilburg, M.H. Bos,
F.G. Heilmann, A. Hofhuis, T. Hoekstra and E. de Boer. 2009. Two outbreaks of
campylobacteriosis associated with the consumption of raw cows’ milk. Int. J. Food
Microbiol. 134(1-2): 70–74.
Hitchins, A.D. 2003. Detection and Enumeration of Listeria monocytogenes in Foods.
Bacteriological Analytical Manual Online. U.S. department of Health and Human
Dairy Pathogens: Characteristics and Impact 111
Services, U.S. Food and Drug Administration Center for Food Safety & Applied Nutrition.
Chapter 10.
Holt, J.G. 1994. Bergey’s manual of determination bacteriology, 9th Edition.
Huang, D.B., A. Mohanty, H.L. DuPont, P.C. Okhuysen and T. Chiang. 2006. A review of An
emerging enteric pathogen: enteroaggregative Escherichia coli. J. Med. Microbiol. 55:
1303–1311.
International Dairy Federation [IDF] Technical Bulletin. 2012. Shiga-toxin-producing Escherichia
coli (STEC), IDF.
International Organization for Standardization. 1996. Microbiology of food and animal feeding
stuffs—Horizontal method for the detection and enumeration of Listeria monocytogenes
—Part 1: Detection method. International Standard ISO 11290–1, Geneva, Switzerland.
International Organization for Standardization. 1998. Microbiology of food and animal feeding
stuffs—Horizontal method for the detection and enumeration of Listeria monocytogenes—
Part 2: Enumeration method. International Standard ISO 11290–2, Geneva, Switzerland.
International Organization for Standardization. 2002. ISO 6579: (Annex D) Detection of
Salmonella spp. In animal faeces and in samples of the primary production stage—
Horizontal method for the detection of Salmonella spp., International Organization for
Standardization, 1, rue de Varembé, Case postale 56 CH-1211 Geneva 20, Switzerland.
International Organization for Standardization. 1995. Microbiology of Food and Animal
Feeding Stuffs—Horizontal Method for Detection of Thermotolerant Campylobacter,
ISO 10270:1995(E). Geneva, Switzerland.
International Organization for Standardization. 2006. Microbiology of Food and Animal
Feeding Stuffs—Horizontal Method for Detection and Enumeration of Campylobacter
spp., ISO 10272–1:2006(E). Geneva, Switzerland.
International Organization for Standardization. 2006. Milk and milk products—Detection of
Enterobacter sakazakii ISO/TS 22964:2006. Geneva, Switzerland.
International Organization for Standardization. 2001. Microbiology of food and animal feeding
stuffs—Horizontal method for the detection of Escherichia coli O157. ISO 16654:2001.
Geneva, Switzerland.
International Organization for Standardization. 2004. Microbiology of food and animal feeding
stuffs —Horizontal methods for the detection and enumeration of Enterobacteriaceae—
Part 2: Colony-count method. ISO 21528-2:2004. Geneva, Switzerland.
International Organization for Standardization. 2001. Microbiology of food and animal
feeding stuffs —Horizontal method for the enumeration of beta-glucuronidase-positive
Escherichia coli—Part 1: Colony-count technique at 44 degrees C using membranes and
5-bromo-4-chloro-3-indolyl beta-D-glucuronide. ISO 16649-1:2001. Geneva, Switzerland.
International Organization for Standardization. 1999. Microbiology of food and animal feeding
stuffs —Horizontal method for the enumeration of coagulase-positive staphylococci
(Staphylococcus aureus and other species)—Part 2: Technique using rabbit plasma
fibrinogen agar medium. ISO 6888-2:1999. Geneva, Switzerland.
International Organization for Standardization. 2004. Microbiology of food and animal feeding
stuffs —Horizontal method for the enumeration of presumptive Bacillus cereus—Colony-
count technique at 30 degrees C. ISO 7932:2004. Geneva, Switzerland.
International Organization for Standardization. 2009. Prerequisite programmes on food
manufacturing safety—Part 1: Food ISO/TS 22002-1:2009. Geneva, Switzerland.
International Organization for Standardization. 2005. Food safety management systems—
Requirements for any organization in the food chain. ISO 22000:2005. Geneva, Switzerland.
Iversen, C. and S. Forsythe. 2003. Risk profile of Enterobacter sakazakii, an emergent pathogen
associated with infant milk formula. Trends Food Sci. Technol. 14: 443–454.
Iversen, C., N. Mullane, B. McCardell, B.D. Tall, A. Lehner, S. Fanning, R. and H. Joosten.
2008. Cronobacter gen. nov., a new genus to accommodate the biogroups of Enterobacter
sakazakii, and proposal of Cronobacter sakazakii gen. nov., comb. nov., Cronobacter malonaticus
sp. nov., Cronobacter turicensis sp. nov., Cronobacter muytjensii sp. nov., Cronobacter
dublinensis sp. nov., Cronobacter genomospecies 1, and of three subspecies, Cronobacter
112 Dairy Microbiology: A Practical Approach
Papageorgiou, D.K. and E.H. Marth. 1989. Fate of Listeria monocytogenes during the
manufacture and ripening of blue cheese. J. Food Prot. 52: 459–465.
Phalipon, A. and P.J. Sansonetti. 2007. Shigella’s ways of manipulating the host intestinal innate
and adaptive immune system: a tool box for survival? Immun. Cell Biol. 85(2): 119–129.
Pui, C.F., W.C. Wong. L.C. Chai, R. Tunung, P. Jeyaletchumi, M.S. Noor Hidayah, A. Ubong,
M.G. Farinazleen, Y.K. Cheah and R. Son. 2011. Salmonella: A Foodborne Pathogen. Int.
Food Res. J. 18(2): 465–473.
Ryan, C.A., M.K. Nickels, N.T. Hargrett-Bean, M.E. Potter, T. Endo, L. Mayer, C.W. Langkop,
C. Gibson, R.C. McDonald, R.T. Kenney, N.D. Puhr, P.J. McDonnell, R.J. Martin, M.L.
Cohen and P.A. Blake. 1987. Massive Outbreak of Antimicrobial-Resistant Salmonellosis
Traced to Pasteurized Milk JAMA. 258(22): 3269–3274.
Ryser, E.T. and E.H. Marth. 2004. Listeria, listeriosis and food safety. Third Edition. Marcel
Dekker Inc., New York, USA.
Sabina, Y., A. Rahman, R.C. Ray and D. Montet. 2011. Yersinia enterocolitica: Mode of
Transmission, Molecular Insights of Virulence, and Pathogenesis of Infection. J. Pathog.
429069.
[Scharlau] Standardized procedures for microbiological examinations of food products. Culture
Medium Handbook, Edition 9.
Shaker, R., T. Osaili, W. Al-Omary, Z. Jaradat and M. Al-Zuby. 2007. Isolation of Enterobacter
sakazakii and other Enterobacter sp. from food and food production environments. Food
Control 18: 1241–1245.
Silva, J., D. Leite, M. Fernandes, C. Mena, P.A. Gibbs and P. Teixeira. 2011. Campylobacter spp.
as a foodborne pathogen: a review. Front Microbiol. 2: 200.
Singh, P. and A. Prakash. 2010. Prevalence of coagulase positive pathogenic Staphylococcus
aureus in milk and milk products collected from unorganized sector of Agra. Acta
Argiculturae Slovenica 96(1): 37–41.
Tacket, C.O., J.P. Narain, R. Sattin, J.P. Lofgren, C. Konigsberg, R.C. Rendtorff, A. Rausa, B.R.
Davis and M.L. Cohen. 1984. A Multistate Outbreak of Infections caused by Yersinia
enterocolitica Transmitted by pasteurized milk. JAMA 251(4): 483–486.
Teunis, P.F.M., K. Takumi and K. Shinagawa. 2004. Dose response for infection by Escherichia
coli O157:H7 from outbreak data. Risk Analysis. 24: 401–408.
Tindall, B.J., P.A. Grimont, G.M. Garrity and J.P. Euzeby. 2005. Nomenclature and taxonomy
of the genus Salmonella. Int. J. Syst. Evol. Microbiol. 55: 521–524.
United States Department of Agriculture (USDA). 2013. Isolation and Identification of Listeria
monocytogenes from Red Meat, Poultry and Egg Products, and Environmental Samples,
9th Rev. 1–20.
Yan, Q.Q., O. Condell, K. Power, F. Butler, B.D. Tall and S. Fanning. 2012. Cronobacter species
(formerly known as Enterobacter sakazakii) in powdered infant formula: a review of our
current understanding of the biology of this bacterium. J. of Appl. Microbiol. 113: 1–15.
Young, K.T., L.M. Davis and V.J. DiRita. 2007. Campylobacter jejuni: molecular biology and
pathogenesis. Nat. Rev. Microbiol. 5: 665–679.
Yousef, A.E. and E.H. Marth. 1990. Fate of Listeria monocytogenes During the Manufacture and
Ripening of Parmesan Cheese. J. Dairy Sc. 73(12): 3351–3356.
Zaika, L.L. and J.G. Phillips. 2005. Model for the combined effects of temperature, pH and
sodium chloride concentration on survival of Shigella flexneri strain 5348 under aerobic
conditions. Int. J. Food Microbiol. 101: 179–187.
CHAPTER 4
INTRODUCTION
Fermented milk products, such as yogurt and cheese, appeared in human
diet about 8000–10000 years ago. Up to the 20th century, milk fermentation
remained an unregulated process, and, the discovery and characterization
of lactic acid bacteria (LAB) have changed the views on milk fermentation.
In the early 1960s, commercial starter culture companies developed
the production technology to freeze-dry liquid cultures and produce
concentrated frozen starter cultures for the direct inoculation of bulk starter
tanks at the dairy. In the beginning of 1980s Chr. Hansen released the first
Direct-Vat-Set (DVS) culture comprising defined single strains. Today,
several commercial starter companies offer an extensive range of frozen
and freeze-dried concentrated cultures for direct inoculation, eliminating
the need for use of bulk starters, and thus, propagation problems. In fact,
these fermentation businesses, together with probiotic products, represent
a total global market value of over 100 billion Euros (de Vos 2011).
While the traditional method for the manufacture of fermented dairy
products was the “inoculation” of the milk with a sample of a previous
day product, i.e., back-slopping. This method had certain drawbacks and
is not used anymore, except for some home-made products. There was a
significant microbial variability both in the milk and the natural starter,
hence a great fluctuation in the quality of the product. Thus, the substitution
1
PhD, Dairy Science, 25 Kappadokias St. 55134 Thessaloniki, Greece.
2
Starter Cultures Technical Manager, TYRAS SA 5th klm Trikala-Pili, 42100, Trikala, Greece.
* Corresponding author: [email protected]
Dairy Starter Cultures 115
Table 1. Types of starter cultures and species of lactic acid bacteria used in some dairy products.
and produce no gas. The L cultures and D cultures also exist, but are only
used to a minor degree in the cheese industry. Traditional, mesophilic O
cultures are used in many cheeses, where the main focus is on a rapid
and consistent acidification of the milk. The LD cultures are used in most
continental semi-hard and soft cheeses (Table 1).
Starters are also subdivided into defined- and mixed-strain cultures.
Defined-strain cultures are pure cultures with known physiological
characteristics and technological properties. These consist of 2–6 strains,
used in rotation as paired single strains or as multiple strains and enable
industrial-scale production of high quality products. Mixed-strain cultures
contain unknown numbers of strains of the same species and may also
contain bacteria from different species or genera of LAB (Sheehan 2007). For
a detailed classification of starter cultures see Tamime (2002) and Parente
and Cogan (2004).
Dairy Starter Cultures 117
Adjunct cultures are defined as any cultures that are deliberately added at
some point of the manufacture of cheese, but whose primary role is not acid
production (Chamba and Irlinger 2004). The application of adjuncts could
be seen as an attempt by the cheese-maker to add back to the product, in
a controlled manner, some of the biodiversity removed by pasteurization,
improved hygiene and the defined strain starter system. Adjuncts can be
added in the milk or at a later stage of the cheese-making, e.g., in the salting
brines (Bintsis et al. 2002).
During cheese ripening, the starter bacteria and most other organisms
in the curd die; they autolyse and release intracellular enzymes as ripening
progresses (Khalid and Marth 1990a). On the other hand, the nonstarter
lactic acid bacteria (NSLAB) persist and can grow from levels of 102 to 104
cfu/g to ~108 cfu/g after manufacture, where they can have a significant
impact on flavour, either positively or negatively (Khalid and Marth 1990,
Cogan et al. 2007). The role of the secondary micro-flora is evident in certain
cheese varieties, e.g., “eye” formation due to fermentation of lactate by
propionibacteria in Swiss-type cheeses (i.e., Emmental, Gruyère de Comte)
and neutralization of the pH of the surface of a cheese due to metabolism
of lactate by yeasts and moulds in mould- and smear-ripened cheeses
(i.e., Camembert and Limburger) (Dezmazeaud and Cogan 1996). Penicillium
camemberti cultures are commercially available for the manufacture of
mould-ripened soft cheeses (i.e., Brie and Camembert), and Penicillium
roqueforti for blue-veined cheeses (i.e., Danish Blue, Stilton, Gorgonzola
and Roquefort). In addition, Geotrichum candidum are used in mould-
ripened cheeses since its deacidification activity stimulates the growth of
moulds on the cheese surface, and yeasts such as Debaryomyces hansenii,
Kluyveromyces lactis, Candida utilis, Saccharomyces cerevisiae and Rhodosporium
infirmominiatum are used in bacterial smear cheeses to stimulate the growth
of smear bacteria, due to their deacidification potential, and for their
debittering effect, due to their aminopeptidase activity (Chamba and Irlinger
2004). For the manufacture of bacterial smear-ripened cheeses, cultures of
Brevibacterium linens and Brevibacterium casei are available, since they belong
118 Dairy Microbiology: A Practical Approach
to the typical surface bacterial flora of such cheeses and contribute to the
formation of aromatic sulphur compounds (Bockelmann 1999).
In addition, it has been suggested that addition of the adjunct cultures
may be an indirect solution for controlling the growth of non-desirable
bacteria in cheese (Crow et al. 2001, Di Cagno et al. 2003). The term
“protective cultures” has been applied to microbial food cultures exhibiting
a metabolic activity contributing to inhibiting or controlling the growth
of undesired microorganisms in food (European Food and Feed Cultures
Association 2011). These undesired microorganisms could be pathogenic
or toxigenic bacteria and fungi but spoilage causing species may also be
included.
The technological properties of a number of Lactobacillus strains isolated
from cheese have been studied, including milk acidification kinetics and
proteolytic properties, as well as tolerance to salts and phage resistance
(Briggiler-Marco et al. 2007). For example, Lactobacillus casei I90 and
Lactobacillus rhamnosus I91 with low acidifying activity were found to be
appropriate for use as adjunct cultures as they did not alter the composition
of the cheese products and improved their sensory attributes. Interestingly,
the contribution of Lactobacillus paracasei subsp. paracasei and Lactobacillus
plantarum to proteolysis of Cheddar cheese appears to be mainly at the
level of free amino acids (i.e., due to exopeptidase activity) (Lynch et al.
1999). Thus, depending on the biochemical activities of the selected strains,
together with the rate of autolysis and/or release of enzymes, the water-
soluble peptides can be further degraded to small peptides and free amino
acids. Khalid and Marth (1990) emphasized the role of the proteolytic
and lipolytic activities in the development of cheese flavour and Peterson
and Marshall (1990) concluded that the high proteolytic and peptidolytic
activities of lactobacilli have an influence on the extent of proteolysis, but the
growth of heterofermentative strains have been associated with undesirable
flavours in Cheddar cheese.
In order to become an appropriate adjunct culture, it has been suggested
that a microorganism needs to be able to:
1. reach and maintain high levels of cell density during ripening,
2. cause no defect in the product, and
3. impact positively on the overall quality of the cheese.
Probiotic cultures have been extensively used during the last 10 years in
the dairy industry. Probiotic bacteria are “live microorganisms which when
administered in adequate amounts confer a health benefit on the host” (Food
and Agriculture Organization/World Health Organization 2001). Recently,
the addition of probiotic bacteria to cheese has been increasing applied (Ross
et al. 2002, Karimi et al. 2011, Kasimoglu et al. 2004, Souza et al. 2008, Rodgers
2008, Ong et al. 2006, Gardiner et al. 2002, Cruz et al. 2009, Araujo et al. 2010).
Dairy Starter Cultures 119
Genetics
LAB used for starter cultures in dairy products belong to a number
of bacterial genera including Lactococcus, Streptococcus, Lactobacillus,
Pediococcus, and Leuconostoc, all members of the Firmicutes. Moreover, some
probiotic cultures are mostly members of the genus Bifidobacterium, which
also produce lactic acid as one of their major fermentation end-products,
however, from the taxonomical point of view, they are members of the
Actinobacteria. LAB have either homofermentative metabolism, which
mainly produce lactic acid, or heterofermentative metabolism, which, apart
from lactic acid, yield a variety of fermentation products such as acetic acid,
ethanol, carbon dioxide and formic acid (Hutkins 2001).
The genetics of the LAB used as starter cultures in the dairy industry
have been reviewed (Broadbent 2001, Callanan and Ross 2004, Klaenhammer
et al. 2002, Morelli et al. 2004, Mills et al. 2010) and an overview is presented
below. In addition, complete genome sequences of a number of LAB have
been published, and an updated review of the available nucleic acid
databases is published every year in the Nucleic Acid Research (Galperin
and Fernandez-Suarez 2012).
Lactococcus spp.
Lactococci are mesophilic LAB that were first isolated from green plants
(Klaenhammer et al. 2002). These bacteria, previously designated as the
lactic streptococci (Streptococcus lactis subsp. lactis or S. lactis subsp. cremoris)
were placed in this new taxon in 1987 by Schleifer (Klaenhammer et al.
2002). Lactococci are selected for use as starters based on their metabolic
stability, their resistance to bacteriophage, and their ability to produce
unique compounds—often from amino acid catabolism. Lc. lactis subsp.
lactis form one of the main constituents in starter cultures (Table 1) where
120 Dairy Microbiology: A Practical Approach
their most important role lies in their ability to produce acid in milk and
to convert milk fat and protein into flavour compounds.
To date, the complete genome sequences of three lactococcal strains have
been published (Mayo et al. 2008); Lc. lactis subsp. lactis IL1403 (Bolotin et al.
2001), Lc. lactis subsp. cremoris MG1363 and NZ9000 (Wegmann et al. 2007,
Linares et al. 2010) and Lc. lactis subsp. cremoris SK11 (Makarova et al. 2006).
There are noticeable differences between strains, e.g., the chromosome
of Lc. lactis subsp. lactis MG1363 is 160 kb larger than that of Lc. lactis subsp.
lactis IL1403 and has an average Guanine + Cytosine (G+C) content of 35.8%,
and thus, encodes more proteins (Table 2).
Lc. lactis subsp. cremoris strains are preferred over Lc. lactis subsp. lactis
strains because of their superior contribution to product flavor via unique
metabolic mechanisms (Salama et al. 1991). With the knowledge of the
complete genome sequences, Lc. lactis subsp. cremoris was found to contain
greater genome sizes than Lc. lactis subsp. lactis IL1403 (approximately 2.37
Mb), with Lc. lactis subsp. cremoris MG1363 containing the largest genome
size of approximately 2.53 Mb, followed by Lc. lactis subsp. cremoris SK11
with a genome size of 2.44 Mb (Mills et al. 2010). Approximately 85% DNA
sequence identity was observed between the coding domains of Lc. lactis
subsp. lactis IL1403 and Lc. lactis subsp. cremoris MG1363, whereas 97.7%
identity was observed between Lc. lactis subsp. cremoris MG1363 and
Lc. lactis subsp. cremoris SK11 (Wegmann et al. 2007). Interestingly, a
complete set of competence genes was observed on the Lc. lactis subsp. lactis
IL1403 genome, indicating that the strain may have the ability to undergo
natural DNA transformation (Mills et al. 2010).
Many of the traits in lactococci which render these microorganisms
suitable for dairy fermentations are in fact encoded on plasmids (Mills et
al. 2006, McKay 1985). Traits such as lactose utilization, casein breakdown,
bacteriophage resistance, bacteriocin production, antibiotic resistance,
Table 2. Comparison of characteristics of sequences for Lc. lactis subsp. lactis MG1363 and
Lc. lactis subsp. lactis IL1403.
Streptococcus thermophilus
Table 3. Genomes of lactic acid bacteria used as starter cultures in dairy industry.
strains examined (Madera et al. 2003, Turgeon et al. 2004, Hols et al. 2005).
Streptococcal plasmids are generally small, ranging in size from 2.1 to 10
kb and encode few industrially useful phenotypic traits, which include
low molecular weight, heat-stress proteins and specificity subunits of
bacteriophage-resistant restriction modification (R⁄M) systems (O’ Sullivan
et al. 1999) (Solow and Somkuti 2000, El Demerdash et al. 2006).
Lactobacillus spp.
among the largest of lactic acid bacteria. In addition, the strain harbours
three plasmids of ~36 kb (G+C 40.8%), 2.3 kb (G+C 34.3%), and ~1.9 kb
(G+C 39.5%), respectively (Klaenhammer et al. 2002).
Lactobacillus rhamnosus is one of the few species of Lactobacillus that
have been used as probiotic organisms in functional foods. A strain of
L. rhamnosus, designated HN001, has been identified that has both flavour
enhancing and probiotic attributes, therefore, it can be used as an adjunct
during cheese manufacture to reduce adventitious microflora, accelerate
cheese ripening, and improve cheese flavour (Klaenhammer et al. 2002).
Lactobacillus johnsonii strains have been mainly isolated from the
feces of humans and animals (Johnson et al. 1980, Fujisawa et al. 1992),
suggesting that these bacteria constitute part of the natural intestinal flora.
L. johnsonii La1 (formerly L. acidophilus La1) has been extensively studied
for its probiotic properties and is commercialized in the LC1 fermented
milk products (Klaenhammer et al. 2002). La1 shows immunomodulatory
properties (Link-Amster et al. 1994, Schiffrin et al. 1995) and antimicrobial
properties (Bernet-Camard et al. 1997, Felley et al. 2001, Pérez et al. 2001).
L. helveticus is quite closely related (< 10% sequence divergence) to
L. amylovorus, L. acidophilus, L. delbrueckii, L. acetotolerans, L. gasseri, and
L. amylophilus (Schleifer and Ludwig 1995). The genome size of L. helveticus
has been determined to be 2.4 Mb by PFGE (Klaenhammer et al. 2002).
Approximately 40 chromosomal genes and four plasmids have been
sequenced from L. helveticus. L. helveticus is a component of “thermophilic”
starter cultures used in the manufacture of a number of fermented dairy
products (Hassan and Frank 2001) and grows on a relatively restricted
number of carbohydrates that includes lactose and galactose and typically
requires riboflavin, pantothenic acid and pyridoxal for growth (Hammes
and Vogel 1995).
The L. delbrueckii species contains three subspecies, L. delbrueckii subsp.
delbrueckii, L. delbrueckii subsp. lactis, and L. delbrueckii subsp. bulgaricus.
L. delbrueckii subsp. bulgaricus grows on a relatively restricted number of
carbohydrates and typically requires pantothenic acid and niacin (Hammes
and Vogel 1995).
Phylogenetically, L. delbrueckii subsp. bulgaricus is closely related to
L. amylovorus, L. acidophilus, L. helveticus, L. acetotolerans, L. gasseri, and
L. amylophilus (Schleifer and Ludwig 1995). The G+C ratio of L. delbrueckii
subsp. bulgaricus (49–51%) is somewhat higher than that found among other
species (34–46%) within this phylogenetic tree (Hammes and Vogel 1995).
The genome size of L. delbrueckii subsp. bulgaricus has been determined to
be 1.8 Mb by PFGA (Klaenhammer et al. 2002).
Very few chromosomal genes (< 15) have been sequenced from
L. delbrueckii subsp. bulgaricus, however the complete sequence of a small
cryptic plasmid and the partial sequence of a bacteriophage are known.
124 Dairy Microbiology: A Practical Approach
Pediococcus spp.
Leuconostoc spp.
Brevibacterium spp.
Propionibacterium spp.
Bifidobacterium spp.
al. 1985, Axelson 1998, Hammes and Vogel 1995, Tannock 1999). In general,
phenotypic methods suffer from a lack of reproducibility generated by
conditions of culture related to different laboratories, and are often unable
to distinguish between closely related strains (Albuquerque et al. 2009,
Huys et al. 2006, Schleifer et al. 1995).
Lactose Metabolism
LAB need a sugar for energy production and subsequent growth. In dairy
products, the sugar is lactose—a disaccharide composed of glucose and
galactose. Transport of lactose into the cell is carried out using a translocation
system involving the phospoenol pyruvate phosphotransferase (PTS) in
lactococci, while S. thermophilus use a proton motive force (PMF) (Cogan
and Hill 1993). Lactose fermentation by LAB has been reviewed by Broome
et al. (2002), Cogan and Hill (1993), Cocaign-Bousquet et al. (1996), Poolman
(2002), and Tamime and Robinson 1999.
After transporting into the cell, lactose is fermented with one of the
four pathways as shown in Fig. 1. For example, in lactococci the tagarose
pathway is followed and lactose transport and the enzymes for the pathway
are plasmid encoded (Crow et al. 1983). Galactose is only metabolized by
L. helveticus and some strains of L. delbrueckii subsp. lactis (Gal+) and probably
leuconstoc via the Leloir pathway. Glucose-6-P is metabolized by the
glucolytic pathway in the lactobacilli and by the phosphoketolase pathway
in leuconstoc. L-lactate is generally the sole product of fermentation, but
when LAB are grown on galactose, maltose or low levels of glucose other
products are formed, form pyruvate metabolism (Fig. 2).
Citrate is present at a low concentration in milk and is metabolizes by
Leuconostoc spp. and some strains of Lc. lactis subsp. lactis (citrate-utilizing,
Cit+) to CO2 which is responsible for “eye” formation in some cheeses
(Parente and Cogan 2004). In addition, other important aroma compounds
are produced in fermented milks, cheese and butter (Fig. 3). Cit+ strains of
Lc. lactis subsp. lactis contain a plasmid which encodes the transport of
citrate. Citrate metabolism has been reviewed by Hugenholtz (1993).
The presence of a citrate permease is essential for the metabolism of
citrate. The citrate permeases of both Lc. lactis subsp. lactis and Leuconostoc
spp. were found to be pH dependant and their highest acidity was between
pH 5.0 and 6.0. The citrate inside the cell is converted to oxaloacetate, by the
enzyme citrate lyase, and then oxaloacetate is decarboxylated to pyruvate.
In lactococci, pyruvate is then converted to acetate, diacetyl, acetoin,
2, 3-butanediol and CO2. The enzyme pyruvate formate lyase is able to
convert pyruvate to formate, acetate, acetaldehyde and ethanol under
anaerobic conditions and at high pH (> 7.0). Under aerobic conditions
and at pH 5.5 to 6.5, pyruvate can be converted to acetate, acetaldehyde,
ethanol and the minor products acetoin, diacetyl and 2, 3-butanediol via the
multienzyme pyruvate dehydrogenase complex (Fig. 3). In Leuconostoc spp.,
the pyruvate produced from citrate is converted to lactate, although at low
pH and in the absence of glucose (or lactose) Leuconostoc spp. will produce
diacetyl and acetoin. Acetate is also formed via the heterofermentative
metabolism of lactose during co-metabolism with citrate (Broome et al.
2002).
130 Dairy Microbiology: A Practical Approach
Lactose
ADP NADH
Fructose 6-P 6-Phosphogluconate
Tagarose 1,6-biP ATP NAD+
ADP NADH
Fructose 1,6-biP Ribulose 5-P CO2
Xylulose 5-P
Pi
Glyceraldehyde 3-P
Pi NAD+ Acetyl-P
CoAsh
NADH
1,3-diPhosphoglycerate Pi
ADP Acetyl-CoA
NADH
ATP
3-Phosphoglycerate CoAsh NAD+
Acetaldehyde
NADH
2-Phosphoglycerate NAD+
Ethanol
Phosphoenolpyruvate
ADP
ATP
Pyruvate
NADH
NAD+
Lactate
Figure 1. Lactose metabolism pathways in lactic acid bacteria (After: Cogan and Hill 1993,
Hutkins 2001, Tamime and Robinson 1999).
Over the last 15–20 years numerous attempts have been made to change
metabolite production in LAB, via metabolic engineering, from lactic acid
to production of other flavour compounds—usually by removing lactate
Dairy Starter Cultures 131
NAD+ NADH
Pyruvate Acetyl-CoA
NADH NADH ADP
C
á -Acetolactate
ATP
NAD+ NAD+
Diacetyl Acetoin Lactate Ethanol Acetate
Figure 2. Pyruvate metabolism in lactic acid bacteria (After: Cogan and Hill 1993, Hutkins
2001, Tamime and Robinson 1999).
Citrate
Acetate
Oxaloate
CO2
Pyruvate
TPP
α-Acetolactate
CO2 O2
Acetaldehyde-TPP
CO2
CO2
2,3-Butanediol Acetoin Diacetyl
Figure 3. Citrate metabolism in lactic acid bacteria (After: Cogan and Hill 1993, Hutkins 2001,
Tamime and Robinson 1999).
Protein Metabolism
Table 4. Some peptidases derived from lactic acid bacteria used as dairy starter cultures .
Peptidase
Peptidase Substrate
Substrate hydrolyzed
Aminopeptidases
PepC X (X)n
PepL Leu X
PepN X (X)n
Dipeptidases
PepV X X
PepD X X
Tripeptidase T (PepT) X X X
Endopeptidases
subsp. cremoris SK11 (Liu et al. 2010). Members of both the PepE/PepG
(endopeptidases) and PepI/PepR/PepL (proline peptidases) superfamilies
are absent in lactococci and streptococci. On the other hand, many of the
peptidases, such as aminopeptidases PepC, PepN, and PepM, and proline
134 Dairy Microbiology: A Practical Approach
peptidases PepX and PepQ are present in all LAB genomes, usually with
one gene per genome, and seem to be essential for bacterial growth or
survival (Liu et al. 2010).
The formation of amino acids from the proteolysis of cheese caseins
is very important for the development of flavour during the maturation
process. Free amino acids contribute directly to the cheese flavour (Seth
and Robinson 1988) and, additionally, they serve as substrates for other
flavour-generating compounds (Fox and Wallace 1997). In addition,
undesirable bitter peptides may lead to the formation of off-flavours, and
the ability of the micro-flora to hydrolyse bitter peptides is equally important
(Arora and Lee 1990). The conversion of free amino acids in more specific
flavour components is the next, and much more complicated, process in
overall flavour generation. The principal compounds formed are amines,
other amino acids, α-keto acids and sulphur compounds. In lactococci,
transamination in which the α-amino group of the amino acid is transferred
to a keto acid acceptor appears to be the first step in the degradation of
aromatic and branched chain amino acids. One of the key amino acids
in the formation of flavour compounds is methionine. This amino acid
is transaminated using α-ketoglutarate as the amino group acceptor to
form 4-methylthio-2-oxobutyrate which appears to be degraded either
nonenzymically or enzymically to form methanethiol. Methanethiol in
turn is the precursor to a number of potential flavour compounds such as
dimethyl sulphide, dimethyl disulphide and S-methylthioesters (Broome
et al. 2002).
Overproduction of specific amino acid-converting enzymes in starter
bacteria, such as cystathione-β-lyase involved in methionine- and cysteine-
metabolism has had little effect on the ripening in cheese (Fernandez et al.
2002), while increased production of α-ketoglutarate and overproduction
of aminotransferases gave a clear stimulation of the flavour formation by
LAB (Rijnen et al. 1999, 2000). The limited impact of metabolic engineering
in this area is mainly due to the lack of descriptive, and predictive, models
for overall amino acid metabolism in the LAB, which could guide the
different metabolic engineering strategies. Genomics techniques, however,
have been very productive in this area with the development of different
high throughput analyses for amino acid-converting enzymes and DNA
microarrays for analysis of the presence and expression of relevant genes
in different Lc. lactis strains (Hugenholtz 2008).
Lipid Metabolism
certain free fatty acids are essential flavor compounds in certain cheeses
(e.g., caprine milk cheeses). In addition, they react with alcohols or free
sulphydryl groups to form esters and thioesters, respectively, or act as
precursors of a number of other flavour compounds, such as lactones (Fox
and Wallace 1997). Esterase activity has been detected in various lactobacilli
(Khalid et al. 1990), and esters contribute to the characteristic flavor of Swiss-
type cheeses (Fox and Wallace 1997) and Feta cheese (Bintsis et al. 2003).
Exopolysaccharide Production
Extracellular polysaccharides (EPSs) are produced by a variety of
bacteria and are present as capsular polysaccharides (CPS and LPS)
bound to the cell surface, or are released into the growth medium
(Hassan 2008). These polymers can consist of a single type of sugar
(homopolysaccharides) or are regular, repeating units consisting of different
sugars (heteropolysaccharides). EPSs play a major role in the production of
yogurt, cheese, fermented cream and milk-based desserts (Jolly et al. 2002)
where they contribute to texture, mouth-feel, taste perception and stability
of the final products (Hassan 2008). Capsular EPSs form capsules around
the cell wall and are not secreted into the medium. Unattached EPSs are
secreted outside the cell wall of the producer and are responsible for the
“ropy” phenotype observed in fermented milks. The genetic machinery
responsible for EPSs production in lactococci is generally encoded on
large operons containing more than 10 genes. As the producing strains
are food-grade, the EPSs are also considered food-grade. In addition, it
has been suggested that these EPSs or fermented milks containing these
EPSs are active as prebiotics (Gibson and Robertfroid 1995), cholesterol-
lowering (Nakajima et al. 1992) and immunomodulants (Hosono et al. 1997).
EPS-producing strains of S. thermophilus and L. delbreuckii subsp. bulgaricus
have been shown to enhance the texture and viscosity of yogurt and to
reduce syneresis (Hassan et al. 2003, Doleyres et al. 2005). Broadbent et al.
(2003) extensively reviewed the topic of EPS production in S. thermophilus,
providing a detailed overview of its biochemistry, genetics and applications.
With regard to the genetic machinery encoding EPS production in
S. thermophilus, it has been suggested that a dozen or more unique EPSs
gene clusters may occur (Rallu et al. 2002). These clusters range in size from
15 to 20 kb and are chromosomally encoded.
Bacteriophage Resistance
Bacteriophages (phages) are viruses infecting bacteria and multiply in
the bacterial host cells resulting in cell lysis (i.e., virulent phages). The
136 Dairy Microbiology: A Practical Approach
word originates from the Greek word “φάγω”, which means “eat”, so
bacteriophages are the viruses that attack bacteria and they are the main
reason for slow or faulty fermentations during dairy-products making.
They cause no harm to human beings, animals or plants and cannot
cause any human diseases. With a size in the range of 0.1–0.2 µm, phage
are considerably smaller than bacteria. Therefore they can be transferred
through air and can thus be present in all production areas (Chr. Hansen
2002). The general morphology of a bacteriophage consists of a head and
protruding tail. The hexagonal head is a protein envelope. Inside this
envelope is the genetic information (DNA or RNA) of the phage. Under
the head, the collar and the tail are located through which the DNA is
injected into the bacterium. At the end of the tail, a baseplate and tail fibres
are located. They will read and recognise the host specific receptors on the
bacterial cell wall (Chr. Hansen 2002).
Over the years different methods have been proposed to classify
bacteriophages, but they were not accepted universally. However, a recent
approach to bacteriophage taxonomy, which is accepted universally, has
identified three groups known as bacteriophage families, namely the
Myoviridae, Podoviridae and Siphoviridae (Tamime et al. 1999). Based on
morphological distinctions, there are three main types of bacteriophages:
morphotypes A, B, and C, with the B or Siphoviridae group being the most
common type that infect lactic acid bacteria. The group B phages have long
tails with heads that have either a small isometric (B1 sub-group), prolate
(B2), or elongated (B3) shape. The lactococcal phages generally belong to
the B1 or B2 group, whereas the phages that infect S. thermophilus are all in
the B1 group (Hutkins 2001).
While total loss of the final product as a result of phage infection
is infrequent nowadays, phage infection continues to result in quality
defects that affect the flavour, texture and even safety of dairy products
and so continues to receive attention in both research and manufacturing
communities. Phage resistance mechanisms are probably best characterized
in lactococci. Indeed, within lactococci there are four main cellular defences
which interfere with different stages of the phage lytic cycle, namely,
adsorption inhibition (Ads), injection blocking, R⁄M and abortive infection
(Abi). The Ads phenotype prevents the attachment of the phage particle
to the cell surface and can often be induced through the generation of
bacteriophage insensitive mutants (BIMs) (Mills et al. 2007). In Lactococcus
spp., this has been attributed to nonspecific point mutations in chromosomal
genes encoding cell receptor sites by masking of receptors through,
for example, polysaccharide production (Forde and Fitzgerald 1999).
Potentially, one of the most exciting breakthroughs in phage resistance
research recently is the study of S. thermophilus BIMs. Interestingly, within
streptococci the CRISPR loci have recently been shown to play a role in BIM
Dairy Starter Cultures 137
Bacteriocin Production
Bacteriocins are polypeptides synthesized ribosomally by bacteria and can
have a bacteriocidal or bacteriostatic effect on other bacteria (McAuliffe
et al. 2001, Ross et al. 2002). In general, bacteriocins lead to cell death by
inhibiting cell wall biosynthesis or by disrupting the membrane through
pore formation (Twomey et al. 2002). Bacteriocins are therefore important in
food fermentations where they can prevent food spoilage or the inhibition
of food pathogens. Although different classification schemes have been
proposed (Klaenhammer 1993, Nes et al. 1996, Kemperman et al. 2003), the
bacteriocins of LAB have been classified into two major classes. Bacteriocins
are classified into either lanthionine-containing bacteriocins (class I)
(lantibiotics) or the non-lanthionine-containing bacteriocins (class II). The
class II family is relatively diverse in terms of sequence variations as well as
in modes of action, and has therefore been divided further into subgroups
(Diep et al. 2009). Lantibiotics contain post-translationally modified amino
acids such as lanthionine, β-methyllanthionine and the dehydrated residues
dehydroalanine and dehydrobutyrine. The best known lantibiotic is nisin,
which has gained widespread application in the food industry and is used
as a food additive in at least 50 countries, particularly in processed cheese,
dairy products and canned foods (Delves-Broughton et al. 1996). Another
useful bacteriocin in terms of starter culture improvement is lacticin 3147,
encoded on the 60.2 kb plasmid, pMRC01, and this plasmid has been
transferred to over 30 different lactococcal hosts (Ryan et al. 1996, Coakley et
al. 1997). Lacticin 3147 has been shown to be effective in many food systems
for the control of food spoilage or pathogenic bacteria (McAuliffe et al.
1999, Ross et al. 1999, Morgan et al. 2001, O’Sullivan et al. 2006). Recently,
Streptococcus macedonicus ACA-DC 198, was found to produce a lantibiotic
named macedovicin, which inhibits a broad spectrum of lactic acid bacteria,
several food spoilage species (e.g., Clostridium spp.) and oral streptococci
(Georgalaki et al. 2013); Streptococcus macedonicus ACA-DC 198 has been
Dairy Starter Cultures 139
isolated from Greek Kasseri cheese and produce the lacticin 481 lantibiotic
macedocin (Georgalaki et al. 2002).
Bacteriocin-producing LAB have also been applied to improve the
flavour and quality of fermented foods through two strategies; by using
bacteriocin-producing LAB to control adventitious microbial populations
(Ryan et al. 1996) and secondly by using bacteriocin-producing LAB as cell
lysis-inducing agents to increase the rate of proteolysis in cheese (Garde
et al. 2002, Oumer et al. 2001). Many strategies have been developed for
the detection of antibacterial production by microorganisms including
phenotypic methods using indicator strains (Morgan et al. 2000) and liquid
chromatography/mass spectrometry techniques (Zendo et al. 2008). The
detection and identification of bacteriocins in producing strains has also
been greatly aided by PCR amplification of putative bacteriocin genes using
specific primers designed to known bacteriocins (Trmcic et al. 2008) and by
in silico searching amongst bacterial DNA sequences (Draper et al. 2009).
Since LAB are widely used as starter cultures for fermentation in the
dairy, meat and other food industries and have a long history of use by man
for food production and preservation, they are considered as food-grade
microorganisms.
The European Food Safety Authority (EFSA) has recently introduced the
“Qualified Presumption of Safety” (QPS) for a premarket safety assessment
of microorganisms used in food and feed production (European Food
Safety Authority 2006). QPS is applicable to food and feed additives, food
enzymes and plant protection products. The QPS system was proposed to
harmonize approaches to the safety assessment of microorganisms across
the various EFSA scientific panels. The QPS approach is meant to be a fast
track for species for which there is a sufficient body of knowledge that all
strains within a species are assumed to be safe.
In the QPS approach, safety assessment will depend upon the body of
knowledge available for a given microorganism. For some bacterial groups
(e.g., most lactobacilli, including those dominating natural cultures such
as L. helveticus and L. delbrueckii, or S. thermophilus) the identification at
genus/species level and the lack of acquired antibiotic resistance will be
sufficient to assure a safety status (European Food Safety Authority 2006),
while identification and characterization at strain level should be required
for some microbial groups, such as for example enterococci (European Food
Safety Authority 2010).
The QPS list covers only selected groups of microorganisms which
have been referred to EFSA for a formal assessment of safety (European
Food Safety Authority 2006, Leuschner et al. 2010). Seventy-nine species of
microorganisms have so far been submitted to EFSA for a safety assessment
(Pedersen et al. 2005); the list is updated annually (European Food Safety
Authority 2010). It should be noted that the absence of a particular organism
from the QPS list does not necessarily imply a risk associated with its use.
Individual strains may be safe, but this cannot be ascertained from the
existing knowledge of the taxonomic unit to which it belongs. Another
reason for a species not being on the list could be that EFSA has not been
asked to assess the safety of any strains of the species.
Specifications
Commercial starter cultures are supplied either in a frozen or freeze-dried
form and a food safety management system based on the principles of
HACCP is applied in the manufacturing procedure in order to assure the
stability and safety of the product (Tamime 2002). The required specifications
are presented in Table 5.
142 Dairy Microbiology: A Practical Approach
Conclusions
Starter cultures are a part of a rapidly developing field, and the results from
research in the last 15 years, since the discovery of the complete genome
sequence of Lc. lactis subsp. lactis IL1403 by Bolotin et al. (2001), have
led to the development of commercial starters with desirable properties.
Advances in the genetics, molecular biology, physiology, and biochemistry
of LAB have provided new insights and applications for these bacteria in
the dairy industry. On the other hand, the constant risk of bacteriophage
attack justifies the continuous need to search for new strains for improved
processes.
Dairy industry is now capable of producing safe and nutritious
products with different flavours, sometimes with special health-promoting
properties, which satisfy the demands of all consumer and market niches,
and resemble the characteristics of the traditional products. In addition, the
use of selected strains of given species with known metabolic properties
and high technological performances has improved the total quality
control of the manufacturing process. Interestingly, the potential intended
and unintended effects and related risks of using genetic modified starter
cultures can be predicted in an accurate way and verification is feasible.
This combined with profound safety assessments, such as QPS, ensures
safety for consumers and safety for the environment.
These developments could lead to novel functional foods, produced
from processes with well-characterized metabolic pathways, illustrating
new possibilities for the application of these food-grade microorganisms.
References Cited
Albuquerque, P., M.V. Mendes, C.L. Santos, P. Moradas-Ferreira and F. Tavares. 2009. DNA
signature-based approaches for bacterial detection and identification. Sci. of the Total
Environm. 407: 3641–3651.
Araujo, E.A., A.F. Carvalho, E.S. Leandro, M.M. Furtado and C.A. Moraes. 2010. Development
of a symbiotic cottage cheese added with Lactobacillus delbrueckii UFV H2b20 and inulin.
J. of Functional Foods 2: 85–89.
Arora, G. and B.H. Lee. 1990. Comparative studies of peptidases of Lactobacillus casei subspecies.
Journal of Dairy Science 73: 274–279.
Atlan, D., P. Laloi and R. Portalier. 1990. X-prolyldipeptidyl-aminopeptidase of Lactobacillus
delbrueckii subsp. bulgaricus: characterization of the enzyme and isolation of deficient
mutants. Appl. Environ. Microbiol. 56: 2174–2179.
Axelsson, L.T. 1998. Lactic acid bacteria: Classification and physiology. pp. 1–72. In: S. Salminen
and A. von Wright [eds.]. Lactic Acid Bacteria, Marcel Dekker, New York.
Axelsson, L.T. 2004. Lactic acid bacteria: Classification and physiology. pp. 1–65. In: S. Salminen,
A. von Wright and A. Ouwehand [eds.]. Lactic Acid Bacteria, Marcel Dekker, New York.
Baker, G.C., J.J. Smith and D.A. Cowan. 2003. Review and re-analysis of domain-specific 16S
primers. J. Microbiol. Meth. 55: 541–555.
144 Dairy Microbiology: A Practical Approach
Ballongue, J. 2004. Bifidobacteria and Probiotic Action. pp. 66–124. In: S. Salminen, A. von
Wright and A. Ouwehand [eds.]. Lactic Acid Bacteria, Marcel Dekker, New York.
Ben Amor, K., E.E. Vaughan and W.M. de Vos. 2007. Advanced Molecular Tools for the
Identification of Lactic Acid Bacteria. The Journal of Nutrition, Supplement: Effect of
Probiotics and Prebiotics 741S–747S.
Beresford, T.P., N.A. Fitzsimons, N.L. Brennan and T.M. Cogan. 2001. Recent advances in
cheese microbiology. Int. Dairy J. 11: 259–274.
Bernet-Camard, M.-F., V. Liιvin, D. Brassart, J.-R. Neeser, A.L. Servin and S. Hudault. 1997.
The human Lactobacillus acidophilus strain La1 secretes a non bacteriocin antibacterial
substance active in vitro and in vivo. Appl. Environ. Microbiol. 63: 2747–2753.
Bintsis, T., A. Vafopoulou-Mastrojiannaki, E. Litopoulou-Tzanetaki and R.K. Robinson. 2003.
Protease, peptidase and esterase activities by lactobacilli and yeast isolates from Feta
cheese brine. Journal of Applied Microbiology 95: 68–77.
Bintsis, T., A.S. Angelidis and L. Psoni. 2008. Some modern laboratory practices—analysis
of dairy products. pp. 183–261. In: T. Britz and R.K. Robinson [eds.]. Advanced Dairy
Technology, Blackwell Publishing Ltd., Oxford.
Bintsis, T., E. Litopoulou-Tzanetaki and R.K. Robinson. 2000. Review: Existing and potential
applications of ultraviolet light in the food industry—a critical review. J. Sci. Food Agric.
80: 637–645.
Bintsis, T., E.-Z. Chissimelli, A. Papadimitriou and R.K. Robinson. 2002. Brine microbiology
and the chemical and sensory properties of mature feta-type cheese. Australian Dairy
Foods 8: 30–32.
Bockelmann, W., M. Fobker and M. Teuber. 1991. Purification and characterisation of the
X-prolyldipeptidyl-aminopeptidase from Lactobacillus delbrueckii ssp. bulgaricus and
Lactobacillus acidophilus. Int. Dairy J. 1: 51–66.
Bockelmann, W. 1999. Secondary cheese cultures. pp. 132–162. In: B.A. Law [ed.]. Technology
of cheesemaking. Sheffild Academic Press, Sheffild, UK.
Bolotin, A., B. Quinquis, P. Renault, A. Sorokin, S.D. Ehrlich, S. Kulakauskas, A. Lapidus, E.
Goltsman, M. Mazur, G.D. Pusch, M. Fonstein, R. Overbeek, N. Kyprides, B. Purnelle, D.
Prozzi, K. Nqui, D. Masuy, F. Hancy, S. Burteau, M. Boutry, J. Delcour and P. Hols. 2004.
Complete sequence and comparative genome analysis of the dairy bacterium Streptococcus
thermophilus. Nature Biotechnology 22: 1554–1558.
Bolotin, A., P. Wincker, S. Mauger, O. Jaillon, K. Malarme, J. Weissenbach, S.D. Ehrlich and A.
Sorokin. 2001. The complete genome sequence of the lactic acid bacterium Lactococcus
lactis ssp. lactis IL1403. Genome Research 11: 731–753.
Booth, M., D. Fhaolain, P.V. Jennings and G. O’Cuinn. 1990. Purification and characterization
of a postproline dipeptidyl-aminopeptidase from Streptococcus cremoris AM2. J. Dairy
Res. 57: 89–99.
Bourdichon, F., P. Boyaval, Casaregola, J. Dupont, C. Forrokh, J.C. Frisvad, W.P. Hammes, J.
Harnett, G. Huys, J.L. Jany, S. Laululd, A. Ouwehand, Y. Seto, A. Zgoda and E.B. Hansen.
2012a. The 2012 Inventory of Microbial Species with technological beneficial role in
fermented food products. Bulletin of the IDF 455: 22–61.
Bourdichon, F., B. Berger, S. Casaregola, C. Forrokh, J.C. Frisvad, M.L. Gerds, W.P. Hammes,
J. Harnett, G. Huys, S. Laululd, A. Ouwehand, I.B. Powell, J.B. Prajapati, Y. Seto, E. Ter
Schure, A. Van Boven, V. Vankerckhoven, A. Zgoda and E.B. Hansen. 2012b. A Safety
Assessment of Microbial Food Cultures with History of Use in Fermented Dairy Products.
Bulletin of the IDF 455: 2–12.
Bourdichon, F., S. Casaregola, C. Forrokh, J.V. Frisvad, M.L. Gerds, W.P. Hammes, J. Harnett,
G. Huys, S. Laululd, A. Ouwehand, I.B. Powell, J.B. Prajapati, Y. Seto, E.T. Schure, A.
van Boven, V. Vankerckhoven, A. Zgoda, S. Tuijelaars and E.B. Hansen. 2012c. Food
fermentations: Microorganisms with technological beneficial use. International Journal
of Food Microbiology 154: 87–97.
Briggiler-Marco, M., M.L. Capra, A. Quiberoni, G. Vinderola, J.A. Reinheimer and E. Hynes.
2007. Nonstarter Lactobacillus strains as adjunct cultures for cheese making: in vitro
Dairy Starter Cultures 145
characterization and performance in two model cheeses. Journal of Dairy Science 90:
4532–4542.
Broadbent, J.R., D.J. McMahon, D.L. Welker, C.J. Oberg and S. Moineau. 2003. Biochemistry,
genetics, and applications of exopolysaccharide production in Streptococcus thermophilus:
a review. Journal of Dairy Science 86: 407–423.
Broadbent, J.R. 2001. Genetics of lactic acid bacteria. In: J.L. Steele and E.H. Marth [eds.].
Applied Dairy Microbiology, 2nd ed. Marcel Dekker, New York.
Broome, M.C., I.B. Powell and G.K.Y. Limsowtin. 2002. Starter cultures: Specific Properties. pp.
269–275. In: H. Roginski, J.W. Fuguay and P.F. Fox [eds.]. Encyclopedia of Dairy Science,
Vol. I, Elsevier Science Ltd., London.
Caldwell, S., D.J. McMahon, C.J. Oberg and J.R. Broadbent. 1996. Development and
characterization of lactose-positive Pediococcus species for milk fermentation. Appl.
Environ. Microbiol. 62: 936–941.
Caldwell, S., R.W. Hutkins, D.J. McMahon, C.J. Oberg and J.R. Broadbent. 1998. Lactose
and galactose uptake by genetically engineered Pediococcus species. Appl. Microbiol.
Biotechnol. 49: 315–320.
Callanan, M., P. Kaleta and J. O’Callaghan. 2008. Genome sequence of Lactobacillus helveticus, an
organism distinguished by selective gene loss and insertion sequence element expansion.
Journal of Bacteriology 190: 727–735.
Callanan, M.J. and R.P. Ross. 2004. Starter cultures: genetics. pp. 149–161. In: P.F. Fox, P.L.H.
McSweeney, T.M. Cogan and T.P. Guinee [eds.]. Vol 1, 4th Ed. Elsevier Academic Press,
London, UK.
Chamba, J.-F. and F. Irlinger. 2004. Secondary and adjunct cultures. pp. 191–206. In: P.F. Fox,
P.L.H. McSweeney, T.M. Cogan and T.P. Guinee [eds.]. Vol 1, 4th Ed. Elsevier Academic
Press, London, UK.
Chevallier, B., J.C. Hubert and B. Kammerer. 1994. Determination of chromosome size and
number of rrn loci in Lactobacillus plantarum by pulsed-field gel electrophoresis. FEMS
Microbiol. Lett. 120: 51–56.
Chopin, A. 1993. Organization and regulation of genes for amino acid biosynthesis in lactic
acid bacteria. FEMS Microbiol. Rev. 12: 21–38.
Chopin, M.C., A. Chopin and E. Bidnenko. 2005. Phage abortive infection in lactococci:
variations on a theme. Current Opinion in Microbiology 8: 473–479.
Chr. Hansen. 2002. Bacteriophage: A latent problem in the cheese industry. 3rd Edition,
Horsholm, Denmark, pp. 2–7.
Christensen, J.E., E.G. Dudley, J.A. Pederson and J.L. Steele. 1999. Peptidases and amino acid
catabolism in lactic acid bacteria. Antonie van Leeuwenhoek 76: 217–246.
Coakley, M., G. Fitzgerald and R.P. Ross. 1997. Application and evaluation of the phage
resistance- and bacteriocin encoding plasmid pMRC01 for the improvement of dairy
starter cultures. Applied and Environmental Microbiology 63: 1434–1440.
Cocaign-Bousquet, M., C. Garrigues, L. Novak, P. Loubiere and N.D. Lindley. 1996. Physiology
of pyruvate metabolism in Lactococccus lactis. Antonie van Leewenhoek 70: 253–267.
Coenye, T. and P. Vandamme. 2003. Intragenomic heterogeneity between multiple 16S
ribosomal RNA operons in sequenced bacterial genomes. FEMS Microbiol. Lett. 228:
45–49.
Cogan, T.M. and C. Hill. 1993. Cheese starter cultures. pp. 193–255. In: P.F. Fox [ed.]. Cheese:
Chemistry, Physics and Microbiology, Vol. 1, 2nd Ed. Chapman & Hall, London, UK.
Cogan, T.M., M. O’Dowd and D. Mellerick. 1981. Effects of sugar on acetoin production from
citrate by Leuconostoc lactis. Appl. Environ. Microbiol. 41: 1–8.
Cogan, T.M., T.P. Beresford, J. Steele, J. Broadbent, N.P. Shah and Z. Ustunol. 2007. Invited
review: advances in starter cultures and cultured foods. Journal of Dairy Science 90:
4005–4021.
Crittenden, R. 2004. An update on probiotic bifidobacteria. pp. 125–158. In: S. Salminen, A.
von Wright and A. Ouwehand [eds.]. Lactic Acid Bacteria, Marcel Dekker, New York.
146 Dairy Microbiology: A Practical Approach
Crow, V.L., B. Curry and M. Hayes. 2001. The ecology of non-starter lactic acid bacteria
(NSLAB) and their use as adjuncts in New Zealand Cheddar. International Dairy Journal
11: 275–283.
Crow, V.L., G.P. Davey, L.E. Pearce and T.D. Thomas. 1983. Plasmid linkage of the D-tagatose
6-phosphate pathway in Streptococcus lactis: Effect on lactose and galactose metabolism. J.
Bacteriol. 153: 76–83.
Cruz, A.G., F.C.A. Buriti, C.H.B. Souza, J.A.F. Faria and S.M.I. Saad. 2009. Probiotic cheese:
health benefits, technological and stability aspects. Trends in Food Science & Technology
20: 344–354.
Curtin, A.C. and P.L.H. McSweeney. 2004. Catabolism of amino acids in cheese during ripening.
pp. 435–454. In: P.F. Fox, P.L.H. McSweeney, T.M. Cogan and T.P. Guinee [eds.]. Vol 1, 4th
Ed. Elsevier Academic Press, London, UK.
Daeschel, M.A. and T.R. Klaenhammer. 1985. Association of a 13.6-megadalton plasmid in
Pediococcus pentosaceus with bacteriocin activity. Appl. Environ. Microbiol. 50: 1528–1541.
Daniel, P. 1995. Sizing the Lactobacillus plantarum genome and other lactic bacteria species by
transverse alternating field electrophoresis. Curr. Microbiol. 30: 243–246.
Davidson, B., N. Kordis, M. Dobos and A. Hillier. 1996. Genomic organization of lactic acid
bacteria. Antonie van Leeuwenhoek 70: 161–183.
de Visser, J.A., A.D. Akkermans, R.F. Hoekstra and W.M. de Vos. 2004. Insertion-sequence
mediated mutations isolated during adaptation to growth and starvation in Lactococcus
lactis. Genetics 168: 1145–1157.
de Vos, W.M. 2001. Advances in genomics for microbial food fermentations and safety. Curr.
Opin. Biotechnol. 12: 493–498.
de Vos, W.M. 2011. Systems solutions by lactic acid bacteria: from paradigms to practice.
Microbial. Cell Factories 10(Suppl. 1): S2.
de Vos, W.M. and J. Hugenholtz. 2004. Engineering metabolic highways in Lactococci and
other lactic acid bacteria. Trends in Biotechnology 22: 72–79.
Delves-Broughton, J., P. Blackburn, R.J. Evans and J. Hugenholtz. 1996. Applications of the
bacteriocin, nisin. Antonie Van Leeuwenhoek 69: 193–202.
Dezmazeaud, M. and T.M. Cogan. 1996. Role of cultures in cheese ripening. pp. 207–232.
In: T.M. Cogan and J.-P. Accolas [eds.]. Dairy Starter Cultures. VCH Publishers Ltd.,
Cambridge, UK.
Di Cagno, R., M. De Angelis, V. Upadhyay, P.L.H. McSweeney, F. Minervini, G. Gallo and M.
Gobbetti. 2003. Effect of proteinases of starter bacteria on the growth and proteolytic
activity of Lactobacillus plantarum DPC2741. International Dairy Journal 13: 145–157.
Diep, D.B., D. Straume, M. Kjos, C. Torres and I.F. Nes. 2009. An overview of the mosaic
bacteriocin pln loci from Lactobacillus plantarum. Peptides 30: 1562–1574.
Doleyres, Y., L. Schaub and C. Lacroix. 2005. Comparison of the functionality of
exopolysaccharides produced in situ or added as bioingredients on yogurt properties.
Journal of Dairy Science 88: 4146–4156.
Draper, L.A., K. Grainger, L.H. Deegan, P.D. Cotter, C. Hill and R.P. Ross. 2009. Cross-
immunity and immune mimicry as mechanisms of resistance to the lantibiotic lacticin
3147. Molecular Microbiology 71: 1043–1054.
Dunny, G. and L.L. McKay. 1999. Group II introns and expression of conjugative transfer
functions in lactic acid bacteria. Antonie van Leeuwenhoek 76: 77–88.
EC European Council. 2003. Regulation (EC) No 1829/2003 of the European Parliament and of
the Council of 22 September 2003 on genetically modified food and feed. Official Journal
of the European Union. L268: 1–23.
[EC] European Council 2006. Regulation (EC)No 1924/2006 of the European Parliament and of
the Council of 20 December 2006 on nutrition and health claims made on foods. Official
Journal of the European Union. L12: 3–18.
El Abboudi, M., M. El Soda, S. Pandian, R.E. Simard and N.F. Olson. 1992. Purification of
X-prolyl dipeptidyl aminopeptidase from Lactobacillus casei subspecies, Int. J. Food
Microbiol. 15: 87–98.
Dairy Starter Cultures 147
El Demerdash, H.A.M., J. Oxmann, K.J. Heller and A. Geis. 2006. Yoghurt fermentation at
elevated temperatures by strains of Streptococcus thermophilus expressing a small heat-
shock protein: application of a two-plasmid system for constructing food-grade strains
of Streptococcus thermophilus. Biotechnol. Journal 1: 398–404.
Eom, H.S., B.H. Hwang, D.H. Kim, I.B. Lee, Y.H. Kim and H.J. Cha. 2007. Multiple detection of
food-borne pathogenic bacteria using a novel 16S rDNA-based oligonucleotide signature
chip. Biosens. Bioelectron 22: 845–853.
European Food & Feed Cultures Association. 2011. Protective Cultures. http://www.effca.
org/sites/effca.drupalgardens.com/files/Protective-cultures-effca-statement_0.pdf.
European Food Safety Authority. 2010. Scientific Opinion on the maintenance of the list of
QPS biological agents intentionally added to food and feed (2010 update). EFSA Journal
8, 1944: 1–56.
European Food Safety Authority. 2007. Introduction of a Qualified Presumption of Safety
(QPS) approach for assessment of selected microorganisms referred to EFSA. The EFSA
Journal 587: 1–16.
European Food Safety Authority. 2006. Qualified Presumption of Safety of Micro-organisms
in Food and Feed. EFSA Scientific Colloquium Summary Report. Brussels, Belgium.
Felley, C.P., I. Corthιsy-Theulaz, J.-L. Blanco Rivero, P. Sipponen, M. Kaufmann, P. Bauerfeind,
P.H. Wiesel, D. Brassart, A. Pfeifer, A.L. Blum and P. Michetti. 2001. Favourable effect
of an acidified milk (LC-1) on Helicobacter pylori gastritis in man. Eur. J. Gastroenterol.
Hepatol. 13: 25–29.
Ferchichi, M., D. Hemme, M. Nardi and N. Pamboukdjian. 1985. Production of methanethiol
from methionine by Brevibacterium linens CNRZ 918. J. Gen. Microbiol. 131: 715.
Fernandez, M., M. Kleerebezem, O.P. Kuipers, R.J. Siezen and R. Kranenburg. 2002. Regulation
of the metC-cysK operon, involved in sulphur metabolism in Lactococcus lactis. Journal
of Bacteriology 184: 82–90.
Food and Agriculture Organization and World Health Organization. 2001. Health and
Nutritional Properties of Probiotics in Food including Powder Milk with Live Lactic
Acid Bacteria. http://www.who.int/foodsafety/publications/fs_management/en/
probiotics.pdf.
Forde, A. and G.F. Fitzgerald. 1999. Bacteriophage defence systems in lactic acid bacteria.
Antonie van Leeuwenhoek 76: 89–113.
Fox, P.F. and J.M. Wallace. 1997. Formation of flavour compounds in cheese. Advances in
Applied Microbiology 45: 17–85.
Fujisawa, T., Y. Benno, T. Yaeshima and T. Mitsuoka. 1992. Taxonomic study of the Lactobacillus
acidophilus group, with recognition of Lactobacillus gallinarum sp. nov. and Lactobacillus
johnsonii sp. nov. and synonymy of Lactobacillus acidophilus group A3 with the type strain
of Lactobacillus amylovorus. Int. J. System. Bacteriol. 42: 487–491.
Galperin, M.Y. and X.M. Fernandez. 2012. The Nucleic Acids Research Database Issue and
the online Molecular Biology Database Collection. Nucleic Acids Research 40, Database
Issue: D1–D8.
Garde, S., J. Tomillo, P. Gaya, M. Medina and M. Nunez. 2002. Proteolysis in Hispanico cheese
manufactured using a mesophilic starter, a thermophilic adjunct culture and bacteriocin-
producing Lactococcus lactis subsp. lactis INIA 415 adjunct culture. J. Agric. Food Chem.
50: 3479–3485.
Gardiner, G.E., P. Bouchier, E. O’sullivan, J. Kelly, K. Collins, G. Fitzgerald, R.P. Ross and
C. Stanton. 2002. A spray-dried culture for porbiotic Cheddar chesse manufacture.
International Dairy Journal 12: 749–756.
Garvie, E.I. 1986. Genus Leuconostoc. pp. 1071–1075. In: P.H.A. Sneath, N.S. Mair, M.E. Sharpe
and J.G. Holt [eds.]. Bergey’s Manual of Systematic Bacteriology, vol 2, 9th ed. Williams
and Wilkins, Baltimore, US.
Gasson, M.J. 1990. In vivo genetic systems in lactic acid bacteria. FEMS Microbiol. Rev. 7: 43–60.
148 Dairy Microbiology: A Practical Approach
Gasson, M.J., J. Godon, C.J. Pillidge, T.J. Eaton, K. Jury and C.J. Shearman. 1995. Characterization
and exploitation of conjugation in Lactococcus lactis. International Dairy Journal 5: 757–762.
Georgalaki, M.D., K. Papadimitriou, R. Anastasiou, B. Pot, G. Van Driessche, B. Devreese
and E. Tsakalidou. 2013. Macedovicin, the second food-grade lantibiotic produced by
Streptococcus macedonicus ACA-DC 198. Food Microb. 33: 124–130.
Georgalaki, M.D., E. Van Den Berghe, D. Kritikos, B. Devreese, J. Van Beeumen, G.
Kalantzopoulos, L. De Vuyst and E. Tsakalidou. 2002. Macedocin, a food-grade lantibiotic
produced by Streptococcus macedonicus ACA-DC 198. Appl. Environ. Microbiol. 68:
5891–5903.
Gibson, G.R. and M.B. Robertfroid. 1995. Dietary modulation of the human colonic microbiota:
introducing the concept of prebiotics. Journal of Nutrition 124: 1401–1412.
Gilbert, C., D. Atlan, B. Blanc, R. Portailer, J.E. Germond, L. Lapierre and B. Mollet. 1996. A
new cell surface proteinase: sequencing and analysis of the prtB gene from Lactobacillus
delbruekii subsp. bulgaricus. J. Bacteriol. 178: 3059–3065.
Giraffa, G., M. Gatti, L. Rossetti, L. Senini and E. Neviani. 2000. Molecular diversity within
Lactobacillus helveticus as revealed by genotypic characterization. Appl. Environ. Microbiol.
66: 1259–1265.
Gonzalez, C.F. and B.S. Kunka. 1986. Evidence for plasmid linkage of raffinose utilization and
associated α-galactosidase and sucrose hydrolase activity in Pediococcus pentosaceus. Appl.
Environ. Microbiol. 51: 105–109.
Graham, D.C. and L.L. McKay. 1985. Plasmid DNA in strains of Pediococcus cerevisiae and
Pediococcus pentosaceus. Appl. Environ. Microbiol. 50: 532–534.
Guinee, T.P. and D.J. O’Callaghan. 2010. Control and prediction of quality characteristics in
the manufacture and ripening of Cheese. pp. 260–329. In: B.A. Law and A.Y. Tamime
[eds.]. Technology of Cheesemaking 2nd Edition, John Wiley & Sons Ltd., Chichester, UK.
Hammes, W. P. and R. F. Vogel. 1995. The genus Lactobacillus. pp. 19–54. In: B.J.B. Wood and
W.H. Holzapfel [eds.]. The lactic acid bacteria. Vol 2. The genera of lactic acid bacteria.
Blackie Academic and Professional, London.
Hansen, E.B. 2002. Commercial bacterial starter cultures for fermented foods of the future.
International Journal of Food Microbiology 78: 119–131.
Hassan, A.N. 2008. ADSA Foundation Scholar Award: possibilities and challenges of
exopolysaccharide-producing lactic cultures in dairy foods. Journal of Dairy Science
91: 1282–1298.
Hassan, A.N. and J.F. Frank. 2001. Starter cultures and their use. pp 151–206. In: E.H. Marth and
J.L. Steele [eds.]. Applied Dairy Microbiology, 2nd edition. Marcel Dekker, Inc, New York.
Hassan, A.N., R. Ipsen, T. Janzen and K.B. Qvist. 2003. Microstructure and rheology of yogurt
made with cultures differing only in their ability to produce exopolysaccharides. Journal
of Dairy Science 86: 1632–1638.
Høier, E., T. Janzen, F. Rattray, K. Sørensen, M.W. Børsting, E. Brockmann and E. Johansen.
2010. The production, application and action of Lactic cheese starter cultures. pp. 166–192.
In: B.A. Law and A.Y. Tamime [eds.]. Technology of Cheesemaking, 2nd ed. John Wiley
& Sons Ltd., Oxford.
Hols, P., F. Hancy, L. Fontaine, B. Grossiord, D. Prozzi, N. Leblond-Bourget, B. Decaris, A.
Bolotin, C. Delorme, S.D. Ehrlich, E. Guedon, V. Monnet, P. Renault and M. Kleerebezem.
2005. New insights in the molecular biology and physiology of Streptococcus thermophilus
revealed by comparative genomics. FEMS Microbiology Reviews 29: 435–463.
Hols, P., M. Kleerebezem, A.N. Schanck, T. Ferain, J. Hugenholtz, J. Delcour and W.M. de
Vos. 1999. Conversion of Lactococcus lactis from homolactic to homoalanine fermentation
through metabolic engineering. Nature Biotechnology 17: 588–592.
Horvath, P., A.C. Coute-Monvoisin, D.A. Romero, P. Boyaval, C. Fremaux and R. Barrangou.
2009. Comparative analysis of CRISPR loci in lactic acid bacteria genomes. International
Journal of Food Microbiolology 131: 62–70.
Dairy Starter Cultures 149
Klaenhammer, T.R. 1993. Genetics of bacteriocins produced by lactic acid bacteria. FEMS
Microbiology Reviews 12: 39–85.
Kleerebezem, M., P. Hols and J. Hugenholtz. 2000. Lactic acid bacteria as a cell factory:
rerouting of carbon metabolism in Lactococcus lactis by metabolic engineering. Enzyme
Microb. Technol. 26: 840–848.
Kleerebezem, M. and J. Hugenholtz. 2003. Metabolic pathway engineering in lactic acid
bacteria. Current Opinion in Biotechnology 14: 232–237.
Kok, J., G. Buist, A. Zomer, S.A.F.T. vn Hijum and O.P. Kuipers. 2005. Comparative and
functional genomics of lactococci. FEMS Microb. Rev. 29: 411–433.
Kunji, E.R.S., I. Mierau, A. Hagting, B. Poolman and W.N. Konings. 1996. The proteolytic
system of lactic acid bacteria. Antonie van Leewenhoek 70: 187–221.
Lane, D.J., B. Pace, G.J. Olsen, D.A. Stahl, M.L. Sogin and N.R. Pace. 1985. Rapid determination
of 16S ribosomal RNA sequences for phylogenetic analyses. Proceedings of the National
Academy of Sciences of the United States of America 82: 6955–6959.
Law, B.A. 1999. Cheese ripening and cheese flavour technology. pp. 163–192. In: B.A. Law [ed.]
Technology of Cheesemaking. Sheffield Academic Press Ltd., Sheffield, UK.
Leenhouts, L., G. Buist, A. Bolhuis, A.A. ten Berge, J. Kiel, I. Mierau, M. Dabrowska, G. Venema
and J. Kok. 1996. A general system for generating unlabelled gene replacements in bacterial
chromosomes. Molecular and General Genetics 253: 217–224.
Leuschner, R.-G.K., T. Robinson, M. Hugas, P.S. Cocconcelli, F. Richard-Forget, G. Klein, T.R.
Licht, C. Nguyen-The, A. Querol, M. Richardson, J.E. Suarez, J.M. Vlak and A. von Wright.
2010. Qualified presumption of safety (QPS): a generic risk assessment approach for
biological agents notified to the European Food Safety Authority (EFSA). Trends Food
Sci. Technol. 21: 425–435.
Lima, P.T. and A.M. Correia. 2000. Genetic fingerprinting of Brevibacterium linens by pulsed-
field gel electrophoresis and ribotyping. Curr. Microbiol. 41: 50–55.
Linares, D.M., J. Kok and B. Poolman. 2010. Genome Sequences of Lactococcus lactis MG1363
(Revised) and NZ9000 and Comparative Physiological Studies. Journal of Bacteriology
192: 5806–5812.
Link-Amster, H., F. Rochat, K.Y. Saudan, O. Mignot and J.-M. Aeschlimann. 1994. Modulation
of a specific humoral immune response and changes in intestinal flora mediated through
fermented milk intake. FEMS Immunol. Med. Microbiol. 10: 55–64.
Liu, M., J.R. Bayjanov, B. Renckens, A. Nauta and R.J. Siezen. 2010. The proteolytic system of
lactic acid bacteria revisited: a genomic comparison. BMC Genomics 11: 36–51.
Lynch, C.M., D.D. Muir, J.M. Banks, P.L.H. McSweeney and P.F. Fox. 1999. Influence of adjunct
cultures of Lactobacillus paracasei ssp. paracasei or Lactobacillus plantarum on Cheddar cheese
ripening. Journal of Dairy Science 82: 1618–1628.
Madera, C., P. Garcia, T. Janzen, A. Rodriguez and J.E. Suarez. 2003. Characterization of
technologically proficient wild Lactococcus lactis strains resistant to phage infection. Int.
Journal of Food Microb. 86: 213–222.
Makarova, K., A. Slesarev, Y. Wolf, A. Sorokin, B. Mirkin, E. Koonin, A. Pavlov, N. Pavlova,
V. Karamychev, N. Polouchine, V. Shakhova, I. Grigoriev, Y. Lou, D. Rohksar, S. Lucas,
K. Huang, D.M. Goodstein, T. Hawkins, V. Plengvidhya, D. Welker, J. Hughes, Y. Goh,
A. Benson, K. Baldwin, J.H. Lee, I. Díaz-Muñiz, B. Dosti, V. Smeianov, W. Wechter, R.
Barabote, G. Lorca, E. Altermann, R. Barrangou, B. Ganesan, Y. Xie, H. Rawsthorne,
D. Tamir, C. Parker, F. Breidt, J. Broadbent, R. Hutkins, D. O’Sullivan, J. Steele, G.
Unlu, M. Saier, T. Klaenhammer, P. Richardson, S. Kozyavkin, B. Weimer and D. Mills.
2006. Comparative genomics of the lactic acid bacteria. Proc. Natl. Acad. Sci. USA 103:
15611–15616.
Mayo, B., D. van Sinderen and M. Ventura. 2008. Genome analysis of food grade lactic acid-
producing bacteria: from basics to applications. Current Genomics 9: 169–183.
McAuliffe, O., C. Hill and R.P. Ross. 1999. Inhibition of Listeria monocytogenes in cottage
cheese manufactured with a lacticin 3147-producing starter culture. Journal of Applied
Microbiology 86: 251–256.
Dairy Starter Cultures 151
McAuliffe, O., R.P. Ross and C. Hill. 2001. Lantibiotics: structure, biosynthesis and mode of
action. FEMS Microbiol. Rev. 25: 285–308.
McCartney, A. 2002. Application of molecular biological methods for studying probiotics and
the gut flora. Br. J. Nutr. 88: 29–37.
McKay. L.L. 1985. Roles of plasmids in starter cultures. pp. 159–174. In: S.E. Gilliland [ed.].
Bacterial Starter Cultures for Food. CRC Press, Boca Raton, US.
Mills, S., A. Coffey, O.E. McAuliffe, W.C. Meijer, B. Hafkamp and R.P. Ross. 2007. Efficient
method for generation of bacteriophage insensitive mutants of Streptococcus thermophilus
yoghurt and mozzarella strains. Journal of Microbiological Methods 70: 159–164.
Mills, S., C. Griffin, A. Coffey, W.C. Meijer, B. Hafkamp and R.P. Ross. 2009. CRISPR analysis
of bacteriophage insensitive mutants (BIMs) of industrial Streptococcus thermophilus—
implications for starter design. Journal of Applied Microbiology 108: 945–955.
Mills, S., O. O’Sullivan, C. Hill, G. Fitzgerald and R.P. Ross. 2010. The changing face of dairy
starter culture research: From genomics to economics. International Journal of the Dairy
Technology 63: 149–170.
Mills, S., O.E. McAuliffe, A. Coffey, G.F. Fitzgerald and R.P. Ross. 2006. Plasmids of lactococci
—genetic accessories or genetic necessities? FEMS Microbiology Reviews 30: 243–273.
Mollet, B. and M. Delley. 1990. Spontaneous deletion formation within the beta-galactosidase
of Lactobacillus bulgaricus. J. of Bacteriol. 172: 5670–5676.
Morandi, A., O. Zhaxybayeva, J.P. Gogarten and J. Graf. 2005. Evolutionary and diagnostic
implications of intragenomic heterogeneity in the 16S rRNA gene in Aeromonas strains.
J. Bacteriol. 187: 6561–6564.
Morelli, L., F.K. Vogensen and A. von Wright. 2004. Genetics of Lactic Acid Bacteria. pp.
249–293. In: S. Salminen, A. von Wright and A. Ouwehand [eds.]. Lactic Acid Bacteria
—Microbiological and Functional Aspects, 3rd Edition. Marcel Dekker Inc., New York.
Morgan, S.M., M. Galvin, R.P. Ross and C. Hill. 2001. Evaluation of a spray-dried lacticin 3147
powder for the control of Listeria monocytogenes and Bacillus cereus in a range of food
systems. Letters in Applied Microbiology 33: 387–391.
Morgan, S.M., R. Hickey, R.P. Ross and C. Hill. 2000. Efficient method for the detection of
microbially-produced antibacterial substances from food systems. Journal of Applied
Microbiology 89: 56–62.
Myers, K.M., J. Gaba and S.F. Al-Khaldi. 2006. Molecular identification of Yersnia enterocolitica
isolated from pasteurized whole milk using DNA microarray chip hybridization. Mol.
Cell Probes 20: 71–80.
Nakajima, H., Y. Suzuki, H. Kaizu and T. Hirota. 1992. Cholesterol-lowering activity of ropy
fermented milk. Journal of Food Science 57: 1327–1329.
Nes, I.F., D.B. Diep, L.S. Havarstein, M.B. Brurberg, V. Eijsink and H. Holo. 1996. Biosynthesis
of bacteriocins in lactic acid bacteria. Antonie van Leeuwenhoek 70: 113–128.
O’Sullivan, D.J. 1999. Methods for analysis of the intestinal microflora. In: G.W. Tannock [ed.].
Probiotics: A Critical Review. Horizon Scientific Press Norfolk, UK.
O’Sullivan, L., E.B. O’Connor, R.P. Ross and C. Hill. 2006. Evaluation of live-culture-producing
lacticin 3147 as a treatment for the control of Listeria monocytogenes on the surface of
smear-ripened cheese. Journal of Applied Microbiology 100: 125–143.
O’Sullivan, O., J. O’Callaghan, A. Sangrador-Vegas, O. McAuliffe, L. Slattery, P. Kaleta, M.
Callanan, G.F. Fitzgerald, R.P. Ross and T. Beresford. 2009. Comparative genomics of lactic
acid bacteria reveals a niche-specific gene set. BMC Microbiology 9: 50–58.
Ong, L., A. Henriksson and N.P. Shah. 2006. Development of probiotic Cheddar cheese
containing Lactobacillus acidophilus, Lb. casei, Lb. paracasei and Bifidobacterium spp. and
the influence of these bacteria on proteolytic patterns and production of organic acid.
International Dairy Journal 16: 446–456.
Oumer, B.A., E. Fernandez-Garcia, R. Marciaca, S. Garde, M. Medina and M. Nunez. 2001.
Proteolysis and formation of volatile compounds in cheese manufactured with a
bacteriocin-producing adjunct. J. Dairy Res. 68: 119–127.
152 Dairy Microbiology: A Practical Approach
Parente, E. and T.M. Cogan. 2004. Starter Cultures: General Aspects. pp. 123–147. In: P.F. Fox,
P.L.H. McSweeney, T.M. Cogan and T.P. Guinee, Vol. 1, 4th Ed. Elsevier Academic Press,
London, UK.
Park, Y.H., H. Hori, K. Suzuki, S. Osawa and K. Komagata. 1987. Phylogenetic analysis of the
coryneform bacteria by 5S rRNA sequences. J. Bacteriol. 169: 1801–1806.
Parvez, S., K.A. Malik, S.A. Kang and H.Y. Kim. 2006. Probiotics and their fermented food
products are beneficial for health. Journal of Applied Microbiology 100: 1171–1185.
Pastink, M.I., B. Teusink, P. Hols, S. Visser, W.M. de Vos and J. Hugenholtz 2009. Genome-Scale
Model of Streptococcus thermophilus LMG18311 for Metabolic Comparison of Lactic Acid
Bacteria. Appl. and Environm. Microb. 75: 3627–3633.
Pedersen, M.B., S.L. Iversen, K.K. Sorensen and E. Johansen. 2005. The long and winding
road from the research laboratory to industrial applications of lactic acid bacteria. FEMS
Microbiology Reviews 29: 611–624.
Pérez, P.F., J. Minnaard, M. Rouvet, C. Knabenhans, D. Brassart, G.L. De Antoni and E.J.
Schiffrin. 2001. Inhibition of Giardia intestinalis by extracellular factors from lactobacilli: an in
vitro study. Appl. Environ. Microbiol. 67: 5037–5042.
Peterson, S.D. and R.T. Marshall 1990. Nonstarter Lactobacilli in Cheddar Cheese: A Review.
Journal of Dairy Science 73: 1395–1410.
Poolman, B. 2002. Transporters and their role in LAB cell physiology. Antonie van Leewenhoek
82: 147–164.
Rallu, F., P. Taillez, D. Ehrlich and P. Renault. 2002. Common scheme of evolution between eps
clusters of the food bacteria Streptococcus thermophilus and cps clusters of the pathogenic
streptococci. 6th American Society of Microbiology Conference on Streptococcal Genetics,
112. Asheville, NC, USA.
Rattray, F.P., P.F. Fox and A. Healy. 1997. Specificity of an extracellular proteinase from
Brevibacterium linens ATCC 9174 on bovine beta-casein. Appl. Environ. Microbiol. 63:
2468–2471.
Rattray, F.P. and P.F. Fox. 1999. Aspects of enzymology and biochemical properties of
Brevibacterium linens relevant to cheese ripening: a review. J. Dairy Sci. 82: 891–909.
Rijnen, L., P. Courtin, J.C. Gripon and M. Yvon. 2000. Expression of heterologous glutamate
dehydrogenase gene in Lactococcus lactis highly improves the conversion of amino acids
to aroma compounds. Applied and Environmental Microbiology 66: 1354–1359.
Rijnen, L., A. Delacroix Buchet, D. Demaiziere, J.L. Le Quere, J.C. Gripon and M. Yvon. 1999.
Inactivation of lactococcal aromatic aminotransferase prevents the formation of floral
aroma compounds from aromatic amino acids in semi-hard cheese. International Dairy
Journal 9: 877–885.
Rodgers, S. 2008. Novel applications of live bacteria in food services: probiotics and protective
cultures. Trends Food Sci. Tech. 19: 188–197.
Ross, R.P., M. Galvin, O. McAuliffe, S.M. Morgan, M.P. Ryan, D.P. Twomey, W.J. Meaney
and C. Hill. 1999. Developing applications for lactococcal bacteriocins. Antonie van
Leeuwenhoek 76: 337–346.
Ross, R.P., S. Morgan and C. Hill. 2002. Preservation and fermentation: past, present and future.
International Journal of Food Microbiology 79: 3–16.
Roy, D., P. Ward, D. Vincent and F. Mondou. 2000. Molecular identification of potentially
probiotic lactobacilli. Curr. Microbiol. 40: 40–46.
Rudi, K., M. Zimonja, B. Kvenshagen, J. Rugtveit, T. Midtvedt and M. Eggesbo. 2007.
Alignment-independent comparisons of human gastrointestinal tract microbial
communities in a multidimensional 16S rRNA gene evolutionary space. Appl. Environ.
Microbiol. 73: 2727–2734.
Ryan, M.P., M.C. Rea, C. Hill and R.P. Ross. 1996. An application in cheddar cheese manufacture
for a strain of Lactococcus lactis producing a novel broad-spectrum bacteriocin, lacticin
3147. Applied and Environmental Microbiology 62: 612–619.
Dairy Starter Cultures 153
Salama, M., W.E. Sandine and S. Giovannoni. 1991. Development and application of
oligonucleotide probes for identification of Lactococcus lactis subsp. cremoris. Appl.
Environ. Microbiol. 57: 1313–1318.
Schiffrin, E.J., F. Rochat, H. Link-Amster, J.-M. Aeschlimann and A. Donnet-Hughes. 1995.
Immunomodulation of human blood cells following the ingestion of lactic acid bacteria.
J. Dairy Sci. 78: 491–497.
Schleifer, K.H., J. Kraus, C. Dvorak, R. Kilpper-Balz, M.D. Collins and W. Fisher. 1985. Transfer
of Streptococcus lactis and related streptococci to the genus Lactococcus. Systematic and
Applied Microbiology 6: 183–195.
Schleifer, K.-H., M. Ehrmann, C. Beimfohr, E. Brockmann, W. Ludwig and R. Amann. 1995.
Application of molecular methods for the classification and identification of Lactic Acid
Bacteria. Int. Dairy Journal 5: 1081–1094.
Schleifer, K.H. and W. Ludwig. 1995. Phylogenetic relationships of lactic acid bacteria. pp.
7–18. In: B.J.B. Wood and W.H. Holzapfel [eds.]. The Genera of Lactic Acid Bacteria.
Chapman & Hall, London.
Serror, P., T. Sasaki, S.D. Ehrlich and E. Maguin. 2002. Electrotransformation of Lactobacillus
delbrueckii subsp. bulgaricus and L. delbrueckii subsp. lactis with various plasmids. Appl.
Environ. Microbiol. 68: 46–52.
Seth, R.J. and R.K. Robinson. 1988. Factors contributing to the flavour characteristics of mould-
ripened cheese. pp 23–47. In: R.K. Robinson [ed.]. Developments in Food Microbiology,
Vol. 4. Elsevier Applied Science, London, UK.
Sheehan, J.J. 2007. What are starters and what starter types are used for cheesemaking? pp.
36–37. In: P.L.H. McSweeney [ed.]. Cheese problems solved. Woodhead Publishing Ltd.,
Boca Raton, US.
Simpson, W.J. and H. Taguchi. 1995. The genus Pediococcus, with notes on the genera
Tetratogenococcus and Aerococcus. pp 125–172. In: B.J.B. Wood and W.H. Holzapfel [eds].
The Genera of Lactic Acid Bacteria. Chapman & Hall, London.
Smit, G., B.A. Smit and W.J. Engels. 2005. Flavour formation by lactic acid bacteria and
biochemical flavour profiling of cheese products. FEMS Microbiology Reviews 29:
591–610.
Solow, B.T. and G.A. Somkuti. 2000. Molecular properties of Streptococcus thermophilus plasmid
pER35 encoding a restriction modification system. Curr. Microbiol. 42: 122–128.
Sorek, R., V. Kunin and P. Hugenholtz. 2008. CRISPR—a widespread system that provides
acquired resistance against phages in bacteria and archaea. Nature Reviews Microbiology
6: 181–186.
Souza, C.H.B., F.C.A. Buriti, J.H. Behrens and S.M.I. Saad. 2008. Sensory evaluation of probiotic
Minas fresh cheese with Lactobacillus acidophilus added solely or in co-culture with a
thermophilic starter culture. International Journal of Food Science and Technology 43:
871–877.
Souza, M.J., Y. Ardo and P.L.H. McSweeney. 2001. Advances in the study of proteolysis in
cheese. Int. Dairy J. 11: 327–345.
Sybesma, W., J. Hugenholtz, W.M. de Vos and E.J. Smid. 2006. Safe Use of Genetically Modified
Lactic Acid Bacteria in Food. Electronic Journal of Biotechnology 9: 1–25.
Tamime, A.Y. 2002. Microbiology of Starter Cultures, pp. 261–366. In: R.K. Robinson [ed.].
Dairy Microbiology Handbook. 3rd Edition John Wiley & Sons Inc., New York.
Tamime, A.Y. and R.K. Robinson. 1999. Yoghurt Science and Technology. 2nd Edition.
Woodhead Publishing Ltd. Cambridge.
Tannock, G.W. 1999. Probiotics: A Critical Review. Horizon Scientific Press, Wymondham, U.K.
Tetrapak. 1995. Dairy Processing Handbook, Lund, Sweden 64: 233–240.
Thomas, T.D. and O.E. Mills 1981. Proteolytic enzymes of starter bacteria. Netherlands Milk
and Dairy Journal 35: 255–273.
Thompson, J.K., K.J. McConville, C. McReynolds and M.A. Collins. 1999. Potential of conjugal
transfer as a strategy for the introduction of recombinant genetic material into strains of
Lactobacillus helveticus. Appl. Environ. Microbiol. 65: 1910–1914.
154 Dairy Microbiology: A Practical Approach
Thunell, R.K. and W.E. Sandine. 1985. Types of starter cultures. pp. 127–144. In: S.E. Gilliland
[ed.]. Bacterial Starter Cultures for Foods. CRC Press Inc. Boca Raton, US.
Torriani, S., G. Zapparoli and F. Dellaglio. 1999. Use of PCR-based methods for rapid
differentiation of Lactobacillus delbrueckii subsp. bulgaricus and L. delbrueckii subsp. lactis.
Appl. Environ. Microbiol. 65: 4351–4356.
Trmcic, A., T. Obermajer, I. Rogelj and B. Bogovic Matijasic. 2008. Short communication:
culture-independent detection of lactic acid bacteria bacteriocin genes in two traditional
slovenian raw milk cheeses and their microbial consortia. J. Dairy Sci. 91: 4535–4541.
Turgeon, N., M. Frenette and S. Moineau. 2004. Characterization of a theta-replicating plasmid
from Streptococcus thermophilus. Plasmid 51: 24–36.
Twomey, D., R.P. Ross, M. Ryan, B. Meaney and C. Hill. 2002. Lantibiotics produced by lactic
acid bacteria: structure, function and applications. Antonie Van Leeuwenhoek 82: 165–185.
Tynkkynen, S., R. Satokari, M. Saarela, T. Mattila-Sandholm and M. Saxelin. 1999. Comparison
of ribotyping, randomly amplified polymorphic DNA Analysis, and pulsed-field gel
electrophoresis in typing of Lactobacillus rhamnosus and L. casei strains. Appl. Environ.
Microbiol. 65: 3908–3914.
Ummadi, M. and B.C. Weimer. 2001. Tryptophan metabolism in Brevibacterium linens BL2. J.
Dairy Sci. 84: 1773–1782.
Upadhyay, V.K., PL.H. McSweeney, A.A.A. Magboul and P.F. Fox. 2004. Proteolysis in Cheese
during Ripening. pp. 391–433. In: P.F. Fox, P.L.H. McSweeney, T.M. Cogan and T.P. Guinee
[eds.]. Vol 1, 4th Ed. Elsevier Academic Press, London, UK.
[U.S. Food and Drug Administration. 2012. Generally Recognized as Safe (GRAS). http://
www.fda.gov/Food/FoodIngredientsPackaging/GenerallyRecognizedasSafeGRAS/
default.htm.
Valdes-Stauber, N. and S. Scherer. 1996. Nucleotide sequence and taxonomical distribution of
the bacteriocin gene lin cloned from Brevibacterium linens M18. Appl. Environ. Microbiol.
62: 1283–1286.
Van de Guchte, M., S. Penaud, C. Grimaldi, V. Barbe, K. Bryson, P. Nicolas, C. Robert, S.
Oztas, S. Mangenot, A. Couloux, V. Loux, R. Dervyn, R. Bossy, A. Bolotin, J.-M. Batto,
T. Walunas, J.-F. Gibrat, P. Bessieres, J. Weissenbach, S.D. Ehrlich and E. Maguin. 2006.
The complete genome sequence of Lactobacillus bulgaricus reveals extensive and ongoing
reductive evolution. Microbiology 103: 9274–9279.
Varmanen, P., K. Savijoki, S. Εval, A. Palva and S. Tynkkynen. 2000. X-Prolyl dipeptidyl
aminopeptidase gene (pepX) is part of the glnRA operon in Lactobacillus rhamnosus, J.
Bacteriol. 182: 146–154.
Ventura, M. and R. Zink. 2002. Specific identification and molecular typing analysis of
Lactobacillus johnsonii by using PCR-based methods and pulsed-field gel electrophoresis.
FEMS Microbiol. Lett. 217: 141–154.
Vesanto, E., K. Savijoki, T. Rantanen, J.L. Steele and A. Palva. 1995. An X-prolyl-dipeptidyl
aminopeptidase (pepX) gene from Lactobacillus helveticus. Microbiology 141: 3067–3075.
Wegmann, U., M. O’Connell-Motherway, A. Zomer, G. Buist, C. Shearman, C. Canchaya, M.
Ventura, A. Goesmann, M.J. Gasson, O.P. Kuipers, D. van Sinderen and J. Kok. 2007.
Complete genome sequence of the prototype lactic acid bacterium Lactococcus lactis subsp.
cremoris MG1363. J. Bacteriol. 189: 3256–3270.
Zendo, T., J. Nakayama, K. Fujita and K. Sonomoto. 2008. Bacteriocin detection by liquid
chromatography ⁄ mass spectrometry for rapid identification. Journal of Applied
Microbiology 104: 499–507.
Zhong, W., K. Millsap, H. Bialkowska-Hobrzanska and G. Reid. 1998. Differentiation of
Lactobacillus species by molecular typing. Appl. Environ. Microbiol. 64: 2418–2423.
CHAPTER 5
Application of Probiotics in
the Dairy Industry: The Long
Way from Traditional to Novel
Functional Foods
Adele Costabile1,* and Simone Maccaferri2
INTRODUCTION
Dairy products have always been very important functional foods, and
they currently represent an important sector in the “health and wellbeing”
industry.
Dairy foods are generally divided in three categories (Saxelin 2008):
• Basic products (milk, fermented milks, cheeses, ice creams, etc.).
• Value-added products, in which the milk composition has been
modified for enhancing some of the basic properties ascribable to the
food. Examples of value-added products are low-lactose or lactose-free
dairy products, sodium-reduced or calcium-enriched milk products,
among others.
• Functional dairy products with proven health benefit. These kinds
of products are based on the enrichment of the dairy food with a
functional component. Most commonly, functional dairy foods are
1
Research Fellow, Department of Food and Nutritional Sciences, The University of Reading,
PO Box 226, Whiteknights, Reading RG6 6AP.
2
Department of Pharmacy and Biotechnology, University of Bologna Via Belmeloro 6, 40126
Bologna, Italy.
* Corresponding author: [email protected]
156 Dairy Microbiology: A Practical Approach
Dairy Products
Yogurt and Fermented Milks
Yogurt and fermented milks are considered to be the main carrier for the
delivery of probiotics in the dairy industry. In recent years, since many
consumers’ associate yogurt—especially probiotics yogurt, with good
health, per capita consumption of these dairy products has increased
drastically (Hekmat and Reid 2006).
Traditionally, yogurts are prepared through a fermentation procedure
of milk by specific pure cultures of lactic acid bacteria, such as Streptococcus
thermophilus and Lactobacillus bulgaricus. However, even if the simple and
standardized procedure at the basis of the yogurt production is widely
established and commonly utilized, many factors influence the probiotics
viability and its efficacy in promoting human health. Indeed, probiotic
cell survival during the product’s shelf life is a striking factor influenced
by culture conditions, strain selection, level of inoculation, medium
composition, the interactions among starter and probiotics species, final
acidity, availability of nutrients and sugars, and storage and logistics
conditions along the distribution chain (Talwalkar and Kailaspathy 2004,
Dave and Shah 1997, Donkor et al. 2006, Vinderola et al. 2002, Ranadheera
et al. 2010, Plessas et al. 2012).
Dairy Probiotics and Novel Functional Foods 157
Probiotic Cheeses
Probiotic Ice-creams
STRAIN IDENTIFICATION
Molecular taxonomic identification
using 16S rRNA
SAFETY ASSESMENT
Pre-requisite in order to in depth study
the activity and the efficacy of the
probiotics
FUNCTIONAL EVALUATION
In vitro and animal models for
assessing main probiotic traits
EFFICACY EVALUATION
In vivo human study for substantiating
health effect
Figure 1. Scheme of the guidelines for the Evaluation of Health and Nutritional Properties of
Probiotics in Food (Food and Agriculture Organisation of the United Nations/World Health
Organisation 2001) (After: Collado et al. 2009).
Dairy Probiotics and Novel Functional Foods 161
Strain Identification
Safety Assessment
Functional Activity
Adhesion
Antimicrobial substances
Immune modulation
SCFAs production
Enhancement of
barrier function
Improved lactose
tolerance and lowering Suppression of
of cholesterol levels pathogenic bacteria
infection
Stabilization of
Improvement of gut microbiota Production of
mineral absorption and composition antimicrobials/
production vitamins/ bacteriocins
micronutrients
Color image of this figure appears in the color plate section at the end of the book.
The use of LAB as unique probiotic strains did not show relevant results
in the management of UC. Zocco et al. (2006) did not show any efficacy of
probiotic L. rhamnosus GG in maintaining UC remission, whereas Fujimori
et al. (2009) demonstrated that the utilization of B. longum was effective
in improving general IBS symptoms only when administered within a
synbiotic formulation.
In addition to a single strain probiotics formulation, multispecies
probiotics were investigated and tested in clinical trials. Sood et al. (2009)
demonstrated that VSL#3 probiotic mixture was effective in inducing UC
remission in adults suffering from mild to moderate UC, whereas Tursi
et al. (2010) demonstrated a significant clinical response in the group of
UC patients subjected to probiotics therapy. Recently, Miele et al. (2009)
further demonstrated that VSL#3 was useful in the maintenance of UC
remission. Notably, the clinically active probiotic combination VSL#3 has
been demonstrated to exert a potent induction of IL-10 by intestinal and
blood dendritic cells and inhibited generation of pro-inflammatory Th1
cells (Hart et al. 2004).
In the last decades, a rapid rise in the prevalence of allergic and autoimmune
disorders has been observed (Bach 2002). In particular, atopic dermatitis
(AD) is the most common chronic inflammatory skin disease in infancy.
Allergic diseases are generally associated with an imbalance in the TH1/TH2
cytokine response and with stimulation of IgE and IgA synthesis (Winkler
et al. 2007). Since different probiotic strains have been demonstrated to
induce a reduction in TH2 cytokines (i.e., IL-4, IL-5, IL-6, IL-10, IL-13) or a
shift towards TH1-mediated immunity (i.e., IL-2, TNF-α, IFN-γ production)
(Vanderpool et al. 2008, Thomas and Versalovic 2010), a potential role for
probiotics in the management of allergies and AD could be suggested.
Most studies assessing probiotic effects in the treatment of allergic
diseases have focused on AD with or without associated food allergies in
infants and children. Studies aimed at investigating the role of probiotics
in adult-type AD are less frequent. Trials evaluating probiotic efficacy have
investigated on either the treatment or prevention of AD and, to date, only
a few rigorous randomized controlled studies have been performed.
The majority of studies have evaluated probiotic formulations
containing Lactobacillus species, alone or in combination with Bifidobacterium
species. The primary outcome of these trials was the change in the score
for the evaluation of AD severity (SCORAD).
First pioneering studies from Majamaa and Isolauri (1997) and Isolauri
et al. (2000), performed in small numbers of children who were administered
with L. rhamnosus GG, demonstrated that probiotic treatment resulted in
170 Dairy Microbiology: A Practical Approach
References Cited
Alamprese, C., R. Foschino, M. Rossi, C. Pompei and L. Savani. 2002. Survival of Lactobacillus
johnsonii La1 and influence of its addition in retail-manufactured ice cream produced
with different sugar and fat concentrations. Int. Dairy J. 12: 201–208.
Alamprese, C., R. Foschino, M. Rossi, C. Pompei and S. Corti. 2005. Effects of Lactobacillus
rhamnosus GG addition in ice cream. Int. J. Dairy Technol. 58: 200–206.
Andriulli, A., M. Neri, C. Loguercio, N. Terreni, A. Merla, M.P. Cardarella, A. Federico, F.
Chilovi, G.L. Milandri, M. De Bona, S. Cavenati, S. Gullini, R. Abbiati, N. Garbagna, R.
Cerutti and E. Grossi. 2008. Clinical trial on the efficacy of a new symbiotic formulation,
Flortec, in patients with irritable bowel syndrome: a multicenter, randomized study. J.
Clin. Gastroenterol. 42 suppl. 3: S218–223.
Aragon, G., D.B. Graham, M. Borum and D.B. Doman. 2010. Probiotic therapy for Irritable
Bowel Syndrome. Gastroenterol. Hepatol. 6: 39–44.
Bach, J.F. 2002. The effect of infections on susceptibility to autoimmune and allergic diseases.
New Engl. J. Med. 347: 911–920.
Boirivant, M. and W. Strober. 2007. The mechanism of action of probiotics. Curr. Opin.
Gastroenterol. 23(6): 679–692.
Boyle, R. J. and M.L.K. Tang. 2006. The role of probiotics in the management of allergic disease.
Clin. Exp. Allergy. 36: 568–576.
Boyle, R.J., F.J. Bath-Hextall, J. Leonardi-Bee, D.F. Murrell and M.L.K. Tang. 2008. Probiotics
for treating eczema. Cochrane Database of Systematic Reviews (4): CD006135.
Brenner, D.M., M.J. Moeller, W.D. Chey and P.S. Schoenfeld. 2009. The utility of probiotics
in the treatment of irritable bowel syndrome: a systematic review. Am. J. Gastroenterol.
104: 1033–1049.
Burgain, J., C. Gaiani, M. Linder and J. Scher. 2011. Encapsulation of probiotic living cells: from
laboratory scale to industrial applications. J. Food Eng. 104: 467–483.
Cammarota, M., M. De Rosa, A. Stellavato, M. Lamberti, I. Marzaioli and M. Giuliano. 2009.
In vitro evaluation of Lactobacillus plantarum DSMZ 12028 as a probiotic: emphasis on
innate immunity. Int. J. Food Microbiol. 135: 90–98.
Camilleri, M. 2008. Probiotics and irritable bowel syndrome: rationale, mechanisms, and
efficacy. J. Clin. Gastroenterol. 42 Suppl. 3: S123–125.
Candela, M., F. Perna, P. Carnevali, B. Vitali, R. Ciati, R. Gionchetti, F. Rizzello, M. Campieri
and P. Brigidi. 2008. Interaction of probiotic Lactobacillus and Bifidobacterium strains
with human intestinal epithelial cells: adhesion properties, competition against
enteropathogens and modulation of IL-8 production. Int. J. Food Microbiol. 125: 286–292.
Chapman, C.M.C., G.R. Gibson and I. Rowland. 2011. Health benefits of probiotics: are mixtures
more effective than single strains? Eur. J. Nutr. 50: 1–17.
Chen, H. and D.G. Hoover. 2003. Bacteriocins and their food applications. Comp. Rev. Food
Sci. Food Safety 2: 82–100.
Choi, C.H., S.Y. Jo, H.J. Park, S.K. Chang, J.-S. Byeon and S.-J. Myung. 2011. A randomized,
double-blind, placebo-controlled multicenter trial of Saccharomyces boulardii in irritable
bowel syndrome: effect on quality of life. J. Clin. Gastroenterol. 45: 679–683.
Clarke, G., J.F. Cryan, T.G. Dinan and E.M. Quigley. 2012. Review article: probiotics for the
treatment of irritable bowel syndrome—focus on lactic acid bacteria. Aliment. Pharmacol.
Ther. 35(4): 403–413.
172 Dairy Microbiology: A Practical Approach
eczema: randomized controlled trial with follow-up until age 3 years. Clin. Exp. Immunol.
42: 112–122.
Granato, D., G.F. Branco, A.G. Cruz, J. de A.F. Faria and N.P. Shah. 2010. Probiotic Dairy
Products as Functional Foods. Comp. Rev. Food Sci. Food Safety 9: 455–470.
Guandalini, S., G. Magazzù, A. Chiaro, V. La Balestra, G. Di Nardo, S. Gopalan, A. Sibal,
C. Romano, R.B. Canani, P. Lionetti and M. Setty. 2010. VSL#3 improves symptoms in
children with irritable bowel syndrome: a multicenter, randomized, placebo-controlled,
double-blind, crossover study. J. Pediatr. Gastroenterol. Nutr. 51: 24–30.
Gueimonde, M. and S. Salminen. 2006. New methods for selecting and evaluating probiotics.
Dig. Liver Dis. 38 Suppl 2: S242–247.
Guglielmetti, S., D. Mora, M. Gschwender and K. Popp. 2011. Randomised clinical trial:
Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and
improves quality of life—a double-blind, placebo-controlled study. Aliment. Pharmacol.
Ther. 33: 1123–1132.
Guslandi, M., G. Mezzi, M. Sorghi and P.A. Testoni. 2000. Saccharomyces boulardii in
maintenance treatment of Crohn’s disease. Dig. Dis. Sci. 45: 1462–1464.
Haller, D. 2010. Nutrigenomics and IBD: the intestinal microbiota at the cross-road between
inflammation and metabolism. J. Clin. Gastroenterol. 44 Suppl. 1: S6–9.
Hart, A.L., K. Lammers, P. Brigidi, B. Vitali, F. Rizzello, P. Gionchetti, M. Campieri, M.A.
Kamm, S.C. Knight and A.J. Stagg. 2004. Modulation of human dendritic cell phenotype
and function by probiotic bacteria. Gut. 53: 1602–1609.
Heenan, C.N., M.C. Adams, R.W. Hosken and G.H. Fleet. 2004. Survival and sensory
acceptability of probiotic microorganisms in a nonfermented frozen vegetarian dessert.
Food Sci. Technol. 37: 461–466.
Hekmat, S. and G. Reid. 2006. Sensory properties of probiotic yogurt is comparable to standard
yogurt. Nutr. Res. 26: 163–166.
Helwig, U., K.M. Lammers, F. Rizzello, P. Brigidi, V. Rohleder, E. Caramelli, P. Gionchetti,
J. Schrezenmeir, U.R. Foelsch, S. Schreiber and M. Campieri. 2006. Lactobacilli,
bifidobacteria and E. coli nissle induce pro- and anti-inflammatory cytokines in peripheral
blood mononuclear cells. World J. Gastroenterol. 12: 5978–5986.
Hong Y.-S., K.S. Hong, M.-H. Park, Y.-T., Ahn, J.-H. Lee, C.-S. Huh, J. Lee, I.-K. Kim, G.-S.
Hwang and J.S. Kim. 2011. Metabonomic understanding of probiotic effects in humans
with irritable bowel syndrome. J. Clin. Gastroenterol. 45: 415–25.
Isolauri, E. 2000. The use of probiotics in paediatrics. Hosp. Med. 61: 6–7.
Kailasapathy, K., I. Harmstorf and M. Phillips. 2008. Survival of Lactobacillus acidophilus
and Bifidobacterium animalis ssp. lactis in stirred fruit yogurts. Food Sci. Technol. 41:
1317–1322.
Kajander, K., K. Hatakka, T. Poussa, M. Färkkilä and R. Korpela. 2005. A probiotic mixture
alleviates symptoms in irritable bowel syndrome patients: a controlled 6-month
intervention. Aliment. Pharmacol. Ther. 22: 387–394.
Kamada, N., N. Inoue, T. Hisamatsu, S. Okamoto, K. Matsuoka, T. Sato, H. Chinen, K.S. Hong,
T. Yamada, Y. Suzuki, T. Suzuki, N. Watanabe, K. Tsuchimoto and T. Hibi. 2005. Non-
pathogenic Escherichia coli strain Nissle1917 prevents murine acute and chronic colitis.
Inflamm. Bow. Dis. 11: 455–463.
Kim, H.J., M. Camilleri, S. McKinzie, M.B. Lempke, D.D. Burton, G.M. Thomforde and A.R.
Zinsmeister. 2003. A randomized controlled trial of a probiotic, VSL#3, on gut transit and
symptoms in diarrhoea-predominant irritable bowel syndrome. Aliment. Pharmacol.
Ther. 17: 895–904.
Kim, H.J., M.I. Vazquez Roque, M. Camilleri, D. Stephens, D.D. Burton, K. Baxter and G.
homforde. 2005. A randomized controlled trial of a probiotic combination VSL#3 and
placebo in irritable bowel syndrome with bloating. Neurogastroenterol. Motil. 17(5):
687–696.
174 Dairy Microbiology: A Practical Approach
Koll, O., S. von Wallpach and M. Kreuzer. 2010. Multi-Method research on consumer—brand
associations: Comparing free associations, storytelling, and collages. Psychol. Market.
27(6): 584–602.
Konieczna, P., D. Groeger, M. Ziegler, R. Frei, R. Ferstl, F. Shanahan and E.M.M. Quigley.
2012. Bifidobacterium infantis 35624 administration induces Foxp3 T regulatory cells in
human peripheral blood: potential role for myeloid and plasmacytoid dendritic cells.
Gut. 61(3): 354–366.
Kruis, W., P. Fric, J. Pokrotnieks, M. Lukás, B. Fixa, M. Kascák and M.A. Kamm. 2004.
Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917
is as effective as with standard mesalazine. Gut. 53(11): 1617–1623.
Kumar, R., S. Grover and V.K. Batish. 2011. Hypocholesterolaemic effect of dietary inclusion
of two putative probiotic bile salt hydrolase-producing Lactobacillus plantarum strains
in Sprague-Dawley rats. Br. J. Nutr. 105(4): 561–573.
Lee, J., D. Seto and L. Bielory. 2008. Meta-analysis of clinical trials of probiotics for prevention
and treatment of pediatric atopic dermatitis. J. Allergy Clin. Immunol. 121(1).
Lisotti, A., G.L. Cornia, A.M. Morselli-Labate, A. Sartini, L. Turco and V. Grasso. 2011. Effects of
a fermented milk containing Kluyveromyces marxianus B0399 and Bifidobacterium lactis
Bb12 in patients with irritable bowel syndrome. Minerva Gastroenterol. Dietol. 57: 1–12.
Maccaferri, S., A. Klinder, P. Brigidi, P. Cavina and A. Costabile. 2012. Potential Probiotic
Kluyveromyces marxianus B0399 Modulates the immune response in caco-2 cells
and peripheral blood mononuclear cells and impacts the human gut microbiota in an
in vitro colonic model system. Appl. Environment. Microbiol. 78: 956–964.
Majamaa, H. and E. Isolauri. 1997. Probiotics: a novel approach in the management of food
allergy. J. Allergy Clin. Immunol. 99(2): 179–185.
Marteau, P., M. Lémann, P. Seksik, D. Laharie, J.F. Colombel, Y. Bouhnik and G. Cadiot. 2006.
Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence
in Crohn’s disease: a randomised, double blind, placebo controlled GETAID trial. Gut.
5(6): 842–847.
McCarthy, J., L. O’Mahony, C. Dunne, P. Kelly, M. Feeney, B. Kiely, G. O’Sullivan, J.K. Collins
and F. Shanahan. 2001. An open trial of a novel probiotic as an alternative to steroids in
mild/moderately active Crohn’s disease. Gut. 49: A2447.
Michail, S.K., A. Stolfi, T. Johnson and G.M. Onady. 2008. Efficacy of probiotics in the treatment
of pediatric atopic dermatitis: a meta-analysis of randomized controlled trials. Ann.
Allergy Asthma Immunol. 101(5): 508–516.
Michail, S. and H. Kenche. 2011. Gut microbiota is not modified by Randomized, Double-blind,
Placebo-controlled Trial of VSL#3 in Diarrhea-predominant Irritable Bowel Syndrome.
Probiotics Antimicrobial Proteins. (1): 1–7.
Miele, E., F. Pascarella, E. Giannetti, L. Quaglietta, R.N. Baldassano and A. Staiano. 2009.
Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in
children with ulcerative colitis. Am. J. Gastroenterol. 104(2): 437–443.
Moroi, M., S. Uchi, K. Nakamura, S. Sato, N. Shimizu, M. Fujii and T. Kumagai. 2011. Beneficial
effect of a diet containing heat-killed Lactobacillus paracasei K71 on adult type atopic
dermatitis. J. Dermatol. 38(2): 131–139.
Nazzaro, F., F. Fratianni, P. Orlando and R. Coppola. 2012. Biochemical traits, survival and
biological properties of the probiotic Lactobacillus plantarum grown in the presence of
prebiotic inulin and pectin as energy source. Pharmaceuticals 5(5): 481–492.
Niedzielin, K., H. Kordecki and B. Birkenfeld. 2001. A controlled, double-blind, randomized
study on the efficacy of Lactobacillus plantarum 299V in patients with irritable bowel
syndrome. Eur. J. Gastroenterol. Hepatol. 13(10): 1143–1147.
O’Mahony, L., J. McCarthy, P. Kelly, G. Hurley, F. Luo, K. Chen and G.C. O’Sullivan. 2005.
Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and
relationship to cytokine profiles. Gastroenterology 128(3): 541–551.
Dairy Probiotics and Novel Functional Foods 175
Packey, C.D. and R.B. Sartor. 2009. Commensal bacteria, traditional and opportunistic
pathogens, dysbiosis and bacterial killing in inflammatory bowel diseases. Curr. Opin.
Infect. Dis. 22(3): 292–301.
Parassol, N., M. Freitas, K. Thoreux, G. Dalmasso, R. Bourdet-Sicard and P. Rampal. 2005.
Lactobacillus casei DN-114 001 inhibits the increase in paracellular permeability of
enteropathogenic Escherichia coli-infected T84 cells. Res. Microbiol. 156(2): 256–262.
Plessas, S., L. Bosnea, A. Alexopoulos and E. Bezirtzoglou. 2012. Potential effects of probiotics
in cheese and yogurt production: A review. Eng. Lif. Sci. 12(4): 433–440.
Prantera, C., M.L. Scribano, G. Falasco, A. Andreoli and C. Luzi. 2002. Ineffectiveness of
probiotics in preventing recurrence after curative resection for Crohn’s disease: a
randomised controlled trial with Lactobacillus GG. Gut. 51(3): 405–409.
Preidis, G.A. and J. Versalovic. 2009. Targeting the human microbiome with antibiotics,
probiotics, and prebiotics: Gastroenterology Enters the Metagenomics Era.
Gastroenterology 136: 2015–2031.
Ranadheera, R.D.C.S., S.K. Baines and M.C. Adams. 2010. Importance of food in probiotic
efficacy. Food Res. Internat. 43(1): 1–7.
Rembacken, B.J., A.M. Snelling, P.M. Hawkey, D.M. Chalmers and A.T. Axon. 1999. Non-
pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a
randomised trial. Lancet. 354(9179): 635–639.
Roessler, A., S.D. Forssten, M. Glei, A.C. Ouwehand and G. Jahreis. 2012. The effect of probiotics
on faecal microbiota and genotoxic activity of faecal water in patients with atopic
dermatitis: a randomized, placebo-controlled study. Clin. Nutr. 31(1): 22–29.
Rousseaux, C., X. Thuru, A. Gelot, N. Barnich, C. Neut, L. Dubuquoy, C. Dubuquoy et al. 2007.
Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid
receptors. Nat. Med. 13(1): 35–37.
Saxelin, M. 2008. Probiotic formulations and applications, the current probiotics market, and
changes in the marketplace: a European perspective. Clin. Infect. Dis. 46(Suppl. 2): S76–79.
Schillinger, U. and W.H. Holzapfel. 1996. Guidelines for manuscripts on bacteriocins of lactic
acid bacteria. Int. J. Food Microbiol. 33: 3–4.
Schlee, M., J. Wehkamp, A. Altenhoefer, T.A. Oelschlaeger, E.F. Stange and K. Fellermann.
2007. Induction of human beta-defensin 2 by the probiotic Escherichia coli Nissle 1917
is mediated through flagellin. Infection and Immunity 75(5): 2399–2407.
Senok, A.C., A.Y. Ismaeel and G.A. Botta. 2005. Probiotics: facts and myths. Clin. Microbiol.
Infect. 11(12): 958–966.
Shah, S. 2007. Dietary factors in the modulation of inflammatory bowel disease activity. Med.
Gen. Med. 9(1): 60.
Sinn, D.H., J.H. Song, H.J. Kim, J.H. Lee, H.J. Son, D.K. Chang and Y.-H. Kim. 2008. Therapeutic
effect of Lactobacillus acidophilus-SDC 2012, 2013 in patients with irritable bowel
syndrome. Dig. Dis. Sci. 53(10): 2714–2718.
Sood, A., V. Midha, G.K. Makharia, V. Ahuja, D. Singal, P. Goswami and R.K. Tandon. 2009.
The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately
active ulcerative colitis. Clin. Gastroenterol. Hepatol. 7(11): 1202–1209.
Spiller, R. 2008. Review article: probiotics and prebiotics in irritable bowel syndrome. Aliment.
Pharmacol. Ther. 28(4): 385–396.
Stanton C., G. Gardiner, P.B. Lynch, J.K. Collins, G. Fitzgerald and R.P. Ross. 1998. Probiotic
Cheese. Int. Dairy J. 8(5): 6.
Talwalkar, A. and K. Kailasapathy. 2004. The role of oxygen in the viability of probiotic bacteria
with reference to L. acidophilus and Bifidobacterium spp. Curr. Iss. Intest. Microbiol.
5(1): 1–8.
Thomas, C.M. and J. Versalovic. 2010. Probiotics-host communication: Modulation of signaling
pathways in the intestine. Gut Microbes 1(3): 148–163.
Tursi, A., G. Brandimarte, A. Papa, A. Giglio, W. Elisei, G.M. Giorgetti and G. Forti. 2010.
Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as
176 Dairy Microbiology: A Practical Approach
Application of Molecular
Methods for Microbial
Identification in Dairy Products
Paul A. Lawson1,* and Dimitris Tsaltas2
INTRODUCTION
Although traditional approaches such as cultivation, physiological and chemo
taxonomic methods are the cornerstone of the isolation and characterization
of individual organisms and complex communities; molecular methods
have made, and continue to make, incredible contributions to the study of
microbial diversity. A major advantage of molecular methods is the ability
to process large numbers of samples simultaneously and have been termed
high-throughput methods. Principle investigators and students alike,
therefore, favor such methods due to the huge amount of data that can be
generated in a relatively short period of time in a cost-effective manner.
Indeed many would argue that molecular methods have surpassed the
more traditional methods, but this viewpoint is unwise. No one method
can answer all questions and even the most powerful approaches such as
genome analysis must be complemented by physiological investigations to
provide a comprehensive polyphasic approach (Rainey 2011). For example,
a gene may well be present in the genome but is it expressed at all, and if
1
Department of Microbiology and Plant Biology, University of Oklahoma, OK 73019, USA.
Email: [email protected]
2
Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University
of Technology, P.O. Box 50329, 3603 Limassol, CYPRUS.
Email: [email protected]
* Corresponding author
178 Dairy Microbiology: A Practical Approach
Method Principle
16S rDNA gene sequencing Sequence determination of the 16S rDNA gene that contains
both conserved regions to allow sequence alignment and
variable regions that allow diagnostic identification and
provide evolutionary relationships.
Random Amplified Employs short arbitrary primers and low stringency
Polymorphic DNA (RAPD) hybridisation to randomly amplify DNA fragments which
are separated on agarose to give a fingerprint pattern.
Denaturing or Temporal Small PCR amplicons (distinguished by differences in their
Temperature Gradient Gel DNA sequences) are separated from a low to high gradient.
Electrophoresis (DGGE or DGGE uses a chemical gradient (urea or formamide)
TTGE) while TTGE has a temperature gradient and a constant
concentration of denaturing agents.
Real-Time PCR (qPCR) Uses species-specific primers to target a gene/organism. A
fluorescent probe or dye is used to monitor the amplification
of the target DNA in real time enabling quantification of a
target organism.
Terminal Restriction Fluorescent, end labeled, PCR products are digested with
Fragment Length restriction endonucleases and separated by electrophoresis.
Polymorphisms (tRFLP) The end-labelled terminal restriction fragments are
compared with DNA size standards. Different groups have
a difference in the number and location of restriction sites
giving rise to different fragment lengths.
Intergenic Transcribed Analyses the bacterial ITS region located between the 16S
Spacer Analysis (ITS) and 23S ribosomal genes. Allows the differentiation between
strains of the same species or closely related species.
Application of Molecular Methods for Microbial Identification in Dairy Products 181
Genomic Methods
Isolation of DNA
DNA extraction—Protocol 1
2. Add 8 µl each of Proteinase K and RNase (10 mg/ml), mix. Incubate for
1h at 65°C. (The increased temperature inhibits non-specific nucleases,
which would otherwise degrade the DNA).
3. Add 120 µl 10% SDS and return immediately to the 65°C water-bath
for a further 10 min. When the cells lyse the solution may become clear.
4. Remove from water-bath and leave to cool. Add an equal volume of
phenol/chloroform mix until emulsion forms. Centrifuge on low-speed
setting 1500–6500 rpm for 10 min.
5. Three layers should form, a lower solvent layer, an upper aqueous
layer containing the DNA and a layer of protein/cell debris separating
the two other layers. Carefully take off the top layer into a clean
microcentrifuge tube using a wide bore blue tip (i.e., cut with scissors
and smooth with bunsen flame). If the solution is too concentrated
the solution may be very cloudy. Add TES until the solution clears
and repeat the phenol/chloroform step until no more protein material
remains.
6. To the DNA solution add 2.5 volumes of 100% ethanol (–20°C). Mix
gently, DNA strands should start to precipitate. Centrifuge on high-
speed for 5 min. Air-dry for 20 min (or speed vacuum for 5 min)
and resuspend in 50–500 µl (depending on the yield) sterile water
or TE buffer (10 mM Tris, 1 mM EDTA, pH 8.0). Keep the solution
concentrated and fairly viscous (Even if no DNA is seen, continue with
the protocol as very small amounts of DNA can be amplified by PCR).
7. Run 5 µl of the DNA solution on a 0.8% agarose gel with 1 µg lambda
standard to get an approximate concentration. Usually very high
quality DNA is seen as a tight band almost as good as the Lambda
standard. Make a dilution 10 ng/µl for PCR.
Although the first step for many molecular methods is the extraction of high
quality DNA that requires the lysis of bacterial cells prior to downstream
manipulations, for amplification of genes by PCR, a simple boiling method
is often satisfactory.
1. Colonies are suspended on 100 µl of sterile water and incubated at
95–100ºC for 10 min.
2. Centrifuge at 12000 rpm for 5 min to remove cell debris, transfer
supernatant to a clean tube making sure none of the cell pellet is
disturbed.
(If cells prove to be more recalcitrant to lysis, a simple lysis buffer (0.5
M NaOH, 0.05 M sodium citrate) may be used. After 5 min incubation
at room temperature, samples are pelleted and the supernatant
transferred to a clean tube and treated as above).
184 Dairy Microbiology: A Practical Approach
Many research groups also apply the so called “Colony PCR” method
where very small amount of cells are picked up via a pipette tip and
immediately transferred to the PCR reaction mix. Although it is a very
successful technique for the most common bacteria, special care and practice
is required in order to acquire very small amount of cells.
Rapid, sensitive and accurate methods for microbial identification have been
expanded with the introduction of real time PCR or also called quantitative
real time PCR. As a procedure, real time PCR follows the same principle
of PCR with the difference of detecting the end product of the reaction
in real time using fluorescent dyes (Mackay 2004). These dyes are either
186 Dairy Microbiology: A Practical Approach
Fluorescent DNA binding dyes bind to all double stranded DNA thus DNA
increased product during PCR leads to increased fluorescence intensity
measured at each cycle. The most commonly used dye is SYBR Green and
is sold either alone or incorporated in master mixes by a wide range of
companies.
Reactions are prepared as usual with the addition of the dye and runs
are performed on special PCR instruments carrying UV or LED lamps for
excitation of the dye and sensitive detectors/cameras for the measurement
of emitted light.
Some major pitfalls the reader should have in mind and seriously consider
are the following:
• Poor Primer Design. The use of primer design software is strongly
recommended. Most primer design software includes adjustable
parameters for optimal primer design. These parameters consider
primer melting temperature (Tm), complementarity, and secondary
structure as well as amplicon size. The primer melting temperature
(Tm) of each PCR primer should be between 58–60°C and the Tm
of both primers should be within 1°C. Regions of low-complexity
sequence can be problematic in designing a unique primer. The
best option would be to select an alternative region and if that is
not possible, choosing longer primer sequences with higher Tm or
optimization of the thermal cycling protocol may be necessary to help
reduce nonspecific binding. Designing primers that generate very long
amplicons may lead to poor amplification efficiency. Ideally, amplicon
length should be 50 to 150 bases for optimal PCR efficiency. In cases
in which longer amplicons are necessary, optimization of the thermal
cycling protocol and reaction components may be necessary.
• Poor Quality DNA. Degraded or impure DNA can limit the efficiency
of the sensitive PCR reaction and reduce yield. It not uncommon to
have a positive PCR reaction and negative real time PCR reaction using
the same set of primers. Residues of cell (proteins, polysaccharides,
etc.) and phenol and/or salts from DNA isolation method used are
common PCR inhibitory substances.
Application of Molecular Methods for Microbial Identification in Dairy Products 187
Fluorescent reporter probes will detect only DNA containing the probe
sequence. This significantly increases specificity because detection and
quantification does not take place in non-specific DNA amplification. In
addition fluorescent probes labeled with different color flurophores can
be used in multiplex assays detecting several genes in one reaction. The
reactions are as those commonly use in normal PCR. Typical probes used
in many laboratories are TaqMan probes which are hydrolysis probes.
TaqMan probes consist of a fluorophore covalently attached to the 5’–end
of the oligonucleotide probe and a quencher at the 3’–end. Several different
fluorophores (FAM, TET, HEX, Cy3, Cy5, JOE, VIC, ROX, and Texas Red)
and quenchers (TAMRA, MGB) are available. The quencher molecule
quenches the fluorescence emitted by the fluorophore when excited by the
cycler’s light source. As long as the fluorophore and the quencher are in
proximity, quenching inhibits any fluorescence signals. During the reaction,
primers and probe anneal to DNA target and once polymerase reaches the
probe its exonuclease degrades the probe releasing the fluorescent reporter
from the quenching molecule also attached to the probe and as a result
fluorescence is detected.
188 Dairy Microbiology: A Practical Approach
Similarly to Real Time PCR with dsDNA binding dyes, there are
additional major pitfalls that the reader should consider for Real Time PCR
with fluorescent reporter molecules:
• Poor probe design. TaqMan® probe Tm should be ~10°C higher than
the primer Tm.
• Concentration of probes is incorrect. A common range of working stock
concentrations for probes is 2–10 µM.
• Ordering a probe labeled with a dye not calibrated or supported on
the real-time PCR instrument being used.
• Combination of flurophores and quenchers. A well-chosen combination
of flurophores and quenchers is very important for maximum
fluorescence, minimum background, maximum signal to noise ratio
and maximum sensitivity (Marras 2006). Good advice for compatibility
of instruments and the combination of fluorophores/quenchers is
usually provided on instrument and molecular probes manufactures
websites.
et al. 2005, 2006, Sanchez et al. 2006a). Analysis is based on the restriction
endonuclease digestion of fluorescently end labeled PCR products using
a genetic analyzer and appropriate software (Applied Biosystems 2005).
various genes. By placing two samples side-by-side on the gel and allowing
them to denature together, we can easily see even the smallest differences
in two samples or fragments of DNA. Main disadvantages to this technique
are the small reproducibility due to difficulties preparing the acrylamide
gels and the difficulties handling and preparing polyacrylamide. These
problems are partially addressed by TGGE/TTGE (Temperature Gradient
Gel Electrophoresis or Temporal Temperature Gel Electrophoresis), which
uses temperature, rather than chemical, gradient to denature the sample.
In this case the denaturing agent is added at a constant concentration.
DGGE has been used by an extensive number of dairy microbiologists
worldwide Randazzo et al. 2002, 2009, Chen et al. 2008, Dolci et al. 2008a,
2010, Gala et al. 2008, Nikolic et al. 2008, Rantsiou et al. 2008b, Van Hoorde
et al. 2008, Alegria et al. 2009), and the same happened for TGGE (Henri-
Dubernet et al. 2004, 2008, Abriouel et al. 2008).
Resolution problems shown by all electrophoretic methods could be
resolved by the addition of a GC-clamp to one of the primers increasing
resolution when using DGGE (Sheffield et al. 1989, Cocolin et al. 2001, Chen
et al. 2008). Also, fragments with identical migration are strongly suggested
to be analyzed by sequencing since closely related species might co-migrate
(Ogier et al. 2004, Parayre et al. 2007, Giannino et al. 2009, Masoud et al.
2011). Confirmatory results may be acquired via DNA sequencing of the
same or other appropriate PCR products (Chen et al. 2008, Masoud et al.
2011).
In order to reveal metabolically active microbiota of dairy products
scientists have used RNA processed through reverse transcription and
analyzed also by PCR-DGGE (Randazzo et al. 2002, Florez and Mayo 2006,
Rantsiou et al. 2008a, Masoud et al. 2011). In a recent attempt by Porcellato
et al. (2012a, 2012b) combining DGGE with high resolution melt analysis of
DGGE bands, the authors compared reference strain bands with unknown
bands, concluding in high accuracy results of identification.
Multiple Locus VNTR Analysis is a method for genetic analysis that takes
advantage of the polymorphism of tandemly repeated DNA sequences.
VNTR stands for Variable Number of Tandem Repeats. This method is
well known in forensic science since it is the basis of DNA fingerprinting in
humans. When applied to bacteria, it contributes to forensic microbiology
through which the source of a particular strain might eventually be traced
back. For this reason it has been extensively used for pathogenic bacteria
(Stefano et al. 2008, Chen et al. 2011, Radtke et al. 2012, Seale et al. 2012,
Tilburg et al. 2012). In MLVA a number of well-selected and characterized
(in terms of mutation rate and diversity) loci are amplified by polymerase
chain reaction (PCR) so that the size of each locus can be measured. From
this size, the number of repeat units at each locus can be deduced. Repeat
unit sizes and repeat sequences can vary when multiple loci are examined
in a number of different isolates of an individual microbial species. It has
been documented on many occasions that the number of repeat units
per locus is a strain-defining parameter. Consequently, there is isolate
specificity in the number of repeats per locus, when different strains of a
given bacterial species are compared. The resulting information is a code
which can be easily compared to reference databases (http://www.mlva.
eu/). MLVA had limited use in dairy microbiology up to now (Diancourt et
al. 2007, Matamoros et al. 2011) but has found extensive use in pathogenic
and food spoilage microorganisms (Duffy 2009). Well-designed multiplex
PCR primers producing MLVA banding patterns in lactic acid bacteria or
other dairy microflora microorganisms will provide ample new data. A
cornerstone for the adoption of this method will be the development of open
access reference online databases with MLVA patterns of such organisms.
Variable Tandem Repeats can be found using online tools; Tandem
Repeats Finder [http://tandem.bu.edu/trf/trf.html (Benson 1999)] or the
Microorganism Tandem Repeat Database [http://minisatellites.u-psud.fr/
GPMS/(Denoeud and Vergnaud 2004)].
Without doubt the single most important advance both in the identification
of individual species/strains and in the characterization of communities
194 Dairy Microbiology: A Practical Approach
Table 3. Shows commonly used 16S rRNA primers for amplification and complete 16S gene
sequencing. The numbering refers to the positions of the 16S rRNA of Escherichia coli (Brosius
et al. 1978).
Sequencing Reactions
For each reaction add the following:
• Template DNA (see below) 2 to 5 µl
• Primer (1.6 µM) 2.0 µl
• Sequencing dilution buffer or TM (2.5X) 2.0 µl
• BigDye Mix 0.65 µl
• Water to total 10.0 µl
Mix well and spin briefly.
PCR Program
Use hot lid
Initial denaturation 96°C for 30 sec
45 cycles of:
96oC for 10 sec
55°C for 15 sec
60°C for 4 min
Reactions are light sensitive and will degrade. Wrap in foil or put in
box to limit effects of light on samples.
Application of Molecular Methods for Microbial Identification in Dairy Products 197
MicroSeq®
The MicroSeq® Full Gene 16S rDNA and 500 16S rDNA Bacterial
Identification Kit provide all the reagents necessary for determining the
sequence of the 16S rDNA or a part of it. The resulting DNA sequence is
analyzed and compared to a library of 16S rDNA bacterial gene sequences
using MicroSeq® ID Analysis Software and the MicroSeq® ID 16S rDNA
Full Gene Library. MicroSeq® ID Analysis Software enables you to analyze
sequences obtained with any of the MicroSeq® 500 16S rDNA Bacterial
Identification Sequencing Kit, MicroSeq® Full Gene 16S rDNA Bacterial
Identification Sequencing Kit and the MicroSeq® D2 rDNA Fungal
Sequencing Kit. The MicroSeq® ID 16S rDNA Full Gene Library (v1.0)
includes over 1200 validated 16S rDNA sequences. Fungal identification,
Applied Biosystems offers MicroSeq® D2 rDNA Fungal Identification
Sequencing Kit which contains reagents for amplifying and sequencing
the D2 expansion segment region of the nuclear large-subunit (LSU)
ribosomal RNA gene. Variation within this region is sufficient to identify
most organisms at the species level. More than 1070 validated nuclear large-
subunit (LSU) ribosomal RNA gene sequences are included in the library.
Data Analysis
Phylogenetic analysis
Single locus sequence typing has been developed by the use of 16S rRNA
gene sequence analysis and later via the use of more discriminatory
genes such as elongation factor Tu gene (tuf) (Jian et al. 2001), DNA repair
recombinase (recA) (Ventura and Zink 2003), chaperonin Hsp60 (Cpn60)
(Blaiotta et al. 2008), RNA polymerase β subunit (rpoB) (Rantsiou et al. 2004),
and β subunit of DNA gyrase (gyrB) (Itoh et al. 2006).
202 Dairy Microbiology: A Practical Approach
Although single locus sequence typing (De Vos 2011) hasn’t been extensively
used in dairy microbiology, multilocus sequence typing (MLST) has become
a popular approach in dairy fermented products for the characterization
of the microflora. The robustness of MLST derives from the advantage that
sequence data are far less ambiguous and easier to record and interpret than
band patterns produced from all other electrophoresis based techniques
(Spratt 1999).
PCR amplification and sequencing of internal regions of multiple
housekeeping genes (usually seven) assigns numeric allelic designations
and the individual strains are characterized by a seven digit MLST sequence
type. Using algorithm we may identify a parent as the one with the greatest
number of single locus variants. Various tools exist in the internet for
graphical representation of clonal complexes which group a minimum of
5–6 of the 7 allelic designations.
Databases of MLST information are these days easily accessible offering
portability of the data and facilitating global usage of them (http://www.
mlst.net/databases/default.asp, http://pubmlst.org/databases/,http://
www.pasteur.fr/recherche/genopole/PF8/mlst/, http://cge.cbs.dtu.dk/
services/MLST/). Increased cost and time associated with sequencing is
of limited importance since the introduction of chip based sequencing
methods. An MLST analysis for Lactococcus lactis subsp. lactis and cremoris
genotypes gave deeply branched trees (Fernandez et al. 2011). The genes
used were atpA, pheS, rpoA, bcaT, pepN and pepX. For PCR conditions the
reader should refer to Rademaker et al. (2007) and data analysis can be
performed using MEGA software (Tamura et al. 2007).
References Cited
Abriouel, H., A. Martin-Platero, M. Maqueda, E. Valdivia and M. Martinez-Bueno. 2008.
Biodiversity of the microbial community in a Spanish farmhouse cheese as revealed
by culture-dependent and culture-independent methods. Int. J. Food Microbiol. 127(3):
200–208.
Achenbach, L. and C. Woese. 1995. 16S and 23S rRNA-like primers. Archaea: A Laboratory
Manual. K.R.a.S. Sower, H.J. New York, Cold Spring Harbor Laboratory Press 521–523.
Albenzio, M., M.R. Corbo, S.U. Rehman, P.F. Fox, M. De Angelis, A. Corsetti, A. Sevi and M.
Gobbetti. 2001. Microbiological and biochemical characteristics of Canestrato Pugliese
cheese made from raw milk, pasteurized milk or by heating the curd in hot whey. Int. J.
Food Microbiol. 67(1-2): 35–48.
Alcorn, T.M. and S.M. Anderson. 2004. Automated DNA Sequencing. Molecular Microbiology:
Diagnostic Principles and Practice. T.F.C. Persing D.H., Versalovic J., Tang Y.-W., Unger
E.R., Relman D.A., White T.J., ASM Press 153–159.
Alegria, A., P. Alvarez-Martin, N. Sacristan, E. Fernandez, S. Delgado and B. Mayo. 2009.
Diversity and evolution of the microbial populations during manufacture and ripening
of Casin, a traditional Spanish, starter-free cheese made from cow’s milk. Int. J. Food
Microbiol. 136(1): 44–51.
Alegria, A., P. Szczesny, B. Mayo, J. Bardowski and M. Kowalczyk. 2012. Biodiversity in
Oscypek, a traditional Polish cheese, determined by culture-dependent and -independent
approaches. Appl. Environ. Microbiol. 78(6): 1890–1898.
Altschul, S.F., W. Gish, W. Miller, E.W. Myers and D.J. Lipman. 1990. Basic local alignment
search tool. Journal of Molecular Biology 215(3): 403–410.
Amor, K.B., E.E. Vaughan and W.M. De Vos. 2007. Advanced molecular tools for the
identification of lactic acid bacteria. Journal of Nutrition 137(3): 741S–747S.
Andrighetto, C., G. Marcazzan and A. Lombardi. 2004. Use of RAPD-PCR and TTGE for the
evaluation of biodiversity of whey cultures for Grana Padano cheese. Letters in Applied
Microbiology 38(5): 400–405.
Applied Biosystems. 2005. Terminal Fragment Length Polymorphism (T-RFLP) Analysis
on Applied Biosystems Capillary Electrophoresis Systems. Retrieved 12/2005, from
http://www3.appliedbiosystems.com/cms/groups/mcb_marketing/documents/
generaldocuments/cms_042272.pdf.
Aquilanti, L., L. Dell’Aquila, E. Zannini, A. Zocchetti and F. Clementi. 2006. Resident lactic
acid bacteria in raw milk Canestrato Pugliese cheese. Letters in Applied Microbiology
43(2): 161–167.
Aquilanti, L., G. Silvestri, E. Zannini, A. Osimani, S. Santarelli and F. Clementi. 2007.
Phenotypic, genotypic and technological characterization of predominant lactic acid
bacteria in Pecorino cheese from central Italy. J. Appl. Microbiol. 103(4): 948–960.
Argyri, A.A., A.A. Nisiotou, A. Mallouchos, E.Z. Panagou and C.C. Tassou. 2014. Performance
of two potential probiotic Lactobacillus strains from the olive microbiota as starters in the
fermentation of heat shocked green olives. International Journal of Food Microbiology
171: 68–76.
Baker, G.C., J.J. Smith and D.A. Cowan. 2003. Review and re-analysis of domain-specific 16S
primers. J. Microbiol. Methods 55(3): 541–555.
Barns, S.M., R.E. Fundyga, M.W. Jeffries and N.R. Pace. 1994. Remarkable archaeal diversity
detected in a Yellowstone National Park hot spring environment. Proc. Natl. Acad. of
Sci. 91(5): 1609–1613.
206 Dairy Microbiology: A Practical Approach
Baruzzi, F., M. Morea, A. Matarante and P.S. Cocconcelli. 2000. Changes in the Lactobacillus
community during Ricotta forte cheese natural fermentation. J. Appl. Microbiol. 89(5):
807–814.
Benson, G. 1999. Tandem repeats finder: a program to analyze DNA sequences. Nucleic acids
Research 27(2): 573–580.
Berbee, M., M. Pirseyedi and S. Hubbard. 1999. Cochliobolus phylogenetics and the origin of
known, highly virulent pathogens, inferred from ITS and glyceraldehyde-3-phosphate
dehydrogenase gene sequences. Mycologia 964–977.
Bianchi, M.A., D. Del Rio, N. Pellegrini, G. Sansebastiano, E. Neviani and F. Brighenti. 2004. A
fluorescence-based method for the detection of adhesive properties of lactic acid bacteria
to Caco-2 cells. Letters in Applied Microbiology 39(3): 301–305.
Blaiotta, G., V. Fusco, D. Ercolini, M. Aponte, O. Pepe and F. Villani. 2008. Lactobacillus strain
diversity based on partial hsp60 gene sequences and design of PCR-restriction fragment
length polymorphism assays for species identification and differentiation. Applied and
Environmental Microbiology 74(1): 208–215.
Blaiotta, G., O. Pepe, G. Mauriello, F. Villani, R. Andolfi and G. Moschetti. 2002. 16S-23S rDNA
intergenic spacer region polymorphism of Lactococcus garvieae, Lactococcus raffinolactis
and Lactococcus lactis as revealed by PCR and nucleotide sequence analysis. Syst. Appl.
Microbiol. 25(4): 520–527.
Blana, V.A., A. Grounta, C.C. Tassou, G.-J.E. Nychas and E.Z. Panagou. 2014. Inoculated
fermentation of green olives with potential probiotic Lactobacillus pentosus and
Lactobacillus plantarum starter cultures isolated from industrially fermented olives.
Food Microbiology 38: 208–218.
Böhme, K., I.C. Fernández-No, J. Barros-Velázquez, J.M. Gallardo, B. Cañas and P. Calo-Mata.
2010. Comparative analysis of protein extraction methods for the identification of seafood-
borne pathogenic and spoilage bacteria by MALDI-TOF mass spectrometry. Analytical
Methods 2(12): 1941–1947.
Böhme, K., I.C. Fernández-No, J. Barros-Velázquez, J.M. Gallardo, B. Cañas and P. Calo-Mata.
2011. Rapid species identification of seafood spoilage and pathogenic Gram-positive
bacteria by MALDI-TOF mass fingerprinting. Electrophoresis 32(21): 2951–2965.
Böhme, K., I.C. Fernández-No, J. Barros-Velázquez, J.M. Gallardo, B. Cañas and P. Calo-Mata.
2012a. SpectraBank: An open access tool for rapid microbial identification by MALDI-
TOF MS fingerprinting. Electrophoresis 33(14): 2138–2142.
Böhme, K., S. Morandi, P. Cremonesi, I.C. Fernández No, J. Barros-Velázquez, B. Castiglioni,
M. Brasca, B. Cañas and P. Calo-Mata. 2012b. Characterization of Staphylococcus aureus
strains isolated from Italian dairy products by MALDI—TOF mass fingerprinting.
Electrophoresis 33(15): 2355–2364.
Bokulich, N.A. and D.A. Mills 2013. House microbiome drives microbial landscapes of artisan
cheesemaking plants. Appl. Environ. Microbiol.
Bonetta, S., S. Bonetta, E. Carraro, K. Rantsiou and L. Cocolin. 2008. Microbiological
characterisation of Robiola di Roccaverano cheese using PCR-DGGE. Food Microbiol.
25(6): 786–792.
Bottari, B., C. Agrimonti, M. Gatti, E. Neviani and N. Marmiroli. 2013. Development of a
multiplex real time PCR to detect thermophilic lactic acid bacteria in natural whey
starters. Int. J. Food Microbiol. 160(3): 290–297.
Bouton, Y., P. Guyot, E. Beuvier, P. Tailliez and R. Grappin. 2002. Use of PCR-based methods
and PFGE for typing and monitoring homofermentative lactobacilli during Comte cheese
ripening. Int. J. Food Microbiol. 76(1-2): 27–38.
Boyer, M.l. and J.r.m. Combrisson. 2013. Analytical opportunities of quantitative polymerase
chain reaction in dairy microbiology. International Dairy Journal 30(1): 45–52.
Brauns, L.A., M.C. Hudson and J.D. Oliver. 1991. Use of the polymerase chain reaction in
detection of culturable and nonculturable Vibrio vulnificus cells. Appl. Environ. Microbiol.
57(9): 2651–2655.
Application of Molecular Methods for Microbial Identification in Dairy Products 207
Brosius, J., M.L. Palmer, P.J. Kennedy and H.F. Noller. 1978. Complete nucleotide sequence
of a 16S ribosomal RNA gene from Escherichia coli. Proc. Natl. Acad. Sci. USA 75(10):
4801–4805.
Bunthof, C.J. and T. Abee. 2002. Development of a flow cytometric method to analyze
subpopulations of bacteria in probiotic products and dairy starters. Appl. Environ.
Microbiol. 68(6): 2934–2942.
Bunthof, C.J., K. Bloemen, P. Breeuwer, F.M. Rombouts and T. Abee. 2001. Flow cytometric
assessment of viability of lactic acid bacteria. Appl. Environ. Microbiol. 67(5): 2326–2335.
Cain, T.C., D.M. Lubman, W.J. Weber and A. Vertes. 1994. Differentiation of bacteria using
protein profiles from matrix-assisted laser desorption/ionization time-of-flight mass
spectrometry. Rapid Communications in Mass Spectrometry 8(12): 1026–1030.
Callon, C., F. Duthoit, C. Delbes, M. Ferrand, Y. Le Frileux, R. De Cremoux and M.C. Montel.
2007. Stability of microbial communities in goat milk during a lactation year: molecular
approaches. Syst. Appl. Microbiol. 30(7): 547–560.
Cardenas, E. and J.M. Tiedje. 2008. New tools for discovering and characterizing microbial
diversity. Current Opinion in Biotechnology 19(6): 544–549.
Castro-Carrera, T., P. Toral, P. Frutos, N. McEwan, G. Hervás, L. Abecia, E. Pinloche, S.
Girdwood and A. Belenguer. 2014. Rumen bacterial community evaluated by 454
pyrosequencing and terminal restriction fragment length polymorphism analyses in
dairy sheep fed marine algae. Journal of Dairy Science.
Chan, R.K., C.R. Wortman, B.K. Smiley and C.A. Hendrick. 2003. Construction and use of a
computerized DNA fingerprint database for lactic acid bacteria from silage. J. Microbiol.
Methods 55(3): 565–574.
Chen, H.C., S.Y. Wang and M.J. Chen. 2008. Microbiological study of lactic acid bacteria in
kefir grains by culture-dependent and culture-independent methods. Food Microbiol.
25(3): 492–501.
Chen, S., J. Li, S. Saleh-Lakha, V. Allen and J. Odumeru. 2011. Multiple-locus variable number
of tandem repeat analysis (MLVA) of Listeria monocytogenes directly in food samples.
International Journal of Food Microbiology 148(1): 8–14.
Chun, J. and F.A. Rainey. 2014. Integrating genomics into the taxonomy and systematics of the
Bacteria and Archaea. International Journal of Systematic and Evolutionary Microbiology
64(Pt 2): 316–324.
Cocolin, L., M. Manzano, C. Cantoni and G. Comi. 2001. Denaturing gradient gel electrophoresis
analysis of the 16S rRNA gene V1 region to monitor dynamic changes in the bacterial
population during fermentation of Italian sausages. Appl. Environ. Microbiol. 67(11):
5113–5121.
Cole, J.R., Q. Wang, E. Cardenas, J. Fish, B. Chai, R.J. Farris, A. Kulam-Syed-Mohideen, D.
McGarrell, T. Marsh and G.M. Garrity. 2009. The Ribosomal Database Project: improved
alignments and new tools for rRNA analysis. Nucleic Acids Research 37(suppl. 1):
D141–D145.
Collins, M., U. Rodrigues, C. Ash, M. Aguirre, J. Farrow, A. Martinez-Murcia, B. Phillips, A.
Williams and S. Wallbanks. 1991. Phylogenetic analysis of the genus Lactobacillus and
related lactic acid bacteria as determined by reverse transcriptase sequencing of 16S
rRNA. FEMS Microbiology Letters 77(1): 5–12.
Coppola, S., G. Blaiotta and D. Ercolini. 2008. Dairy Products. Molecular Techniques in the
Microbial Ecology of Feremnted Foods. L. Cocolin and d. Ercolini, Springer 31–90.
Corroler, D., I. Mangin, N. Desmasures and M. Gueguen. 1998. An ecological study of lactococci
isolated from raw milk in the camembert cheese registered designation of origin area.
Appl. Environ. Microbiol. 64(12): 4729–4735.
Cremonesi, P., L. Vanoni, S. Morandi, T. Silvetti, B. Castiglioni and M. Brasca. 2011.
Development of a pentaplex PCR assay for the simultaneous detection of Streptococcus
thermophilus, Lactobacillus delbrueckii subsp. bulgaricus, L. delbrueckii subsp. lactis,
L. helveticus, L. fermentum in whey starter for Grana Padano cheese. Int. J. Food
Microbiol. 146(2): 207–211.
208 Dairy Microbiology: A Practical Approach
Ercolini, D. 2013. High-throughput sequencing and metagenomics: steps ahead in the culture-
independent analysis of food microbial ecology. Appl. Environ. Microbiol.
Ercolini, D., F. De Filippis, A. La Storia and M. Iacono. 2012. “Remake” by high-throughput
sequencing of the microbiota involved in the production of water buffalo mozzarella
cheese. Appl. Environ. Microbiol. 78(22): 8142–8145.
Ercolini, D., P.J. Hill and C.E. Dodd. 2003a. Bacterial community structure and location in
Stilton cheese. Appl. Environ. Microbiol. 69(6): 3540–3548.
Ercolini, D., P.J. Hill and C.E. Dodd. 2003b. Development of a fluorescence in situ hybridization
method for cheese using a 16S rRNA probe. J. Microbiol Methods 52(2): 267–271.
Ercolini, D., F. Russo, I. Ferrocino and F. Villani. 2009. Molecular identification of mesophilic
and psychrotrophic bacteria from raw cow’s milk. Food Microbiol. 26(2): 228–231.
Fadda, M.E., S. Viale, M. Deplano, M.B. Pisano and S. Cosentino. 2010. Characterization of
yeast population and molecular fingerprinting of Candida zeylanoides isolated from
goat’s milk collected in Sardinia. Int. J. Food Microbiol. 136(3): 376–380.
Fernandez, E., A. Alegria, S. Delgado, M.C. Martin and B. Mayo. 2011. Comparative phenotypic
and molecular genetic profiling of wild Lactococcus lactis subsp. lactis strains of the L. lactis
subsp. lactis and L. lactis subsp. cremoris genotypes, isolated from starter-free cheeses
made of raw milk. Appl. Environ. Microbiol. 77(15): 5324–5335.
Florez, A.B. and B. Mayo. 2006. Microbial diversity and succession during the manufacture
and ripening of traditional, Spanish, blue-veined Cabrales cheese, as determined by
PCR-DGGE. Int. J. Food. Microbiol. 110(2): 165–171.
Fortina, M.G., G. Ricci, A. Acquati, G. Zeppa, A. Gandini and P.L. Manachini. 2003. Genetic
characterization of some lactic acid bacteria occurring in an artisanal protected
denomination origin (PDO) Italian cheese, the Toma piemontese. Food Microbiology
20(4): 397–404.
Frankel, G., L. Riley, J.A. Giron, J. Valmassoi, A. Friedmann, N. Strockbine, S. Falkow and G.K.
Schoolnik. 1990. Detection of Shigella in feces using DNA amplification. The Journal of
Infectious Diseases 161(6): 1252–1256.
Gala, E., S. Landi, L. Solieri, M. Nocetti, A. Pulvirenti and P. Giudici. 2008. Diversity of lactic
acid bacteria population in ripened Parmigiano Reggiano cheese. Int. J. Food Microbiol.
125(3): 347–351.
Gancheva, A., B. Pot, K. Vanhonacker, B. Hoste and K. Kersters. 1999. A polyphasic approach
towards the identification of strains belonging to Lactobacillus acidophilus and related
species. Syst. Appl. Microbiol. 22(4): 573–585.
Giannino, M.L., M. Marzotto, F. Dellaglio and M. Feligini. 2009. Study of microbial diversity
in raw milk and fresh curd used for Fontina cheese production by culture-independent
methods. Int. J. Food Microbiol. 130(3): 188–195.
Giraffa, G. and E. Neviani. 2001. DNA-based, culture-independent strategies for evaluating
microbial communities in food-associated ecosystems. Int. J. Food Microbiol. 67(1-2):
19–34.
Goering, R.V. 2013. Molecular Typing Techniques: State of the Art. Advanced Techniques in
Diagnostic Microbiology. Y.-W. Tang and C.W. Stratton, Springer US 239–261.
Hall, B.G. 2004. Phylogenetic trees made easy: a how-to manual, Sinauer Associates Sunderland.
Handelsman, J., M.R. Rondon, S.F. Brady, J. Clardy and R.M. Goodman. 1998. Molecular
biological access to the chemistry of unknown soil microbes: a new frontier for natural
products. Chemistry & Biology 5(10): R245–R249.
Harmsen, D., J. Heesemann, T. Brabletz, T. Kirchner and H. Müller-Hermelink. 1994.
Heterogeneity among Whipple’s-disease-associated bacteria. The Lancet 343(8908): 1288.
Harmsen, D., J. Rothgänger, M. Frosch and J. Albert. 2002. RIDOM: ribosomal differentiation
of medical micro-organisms database. Nucleic Acids Research 30(1): 416–417.
Hayden, R.T. 2004. In Vitro Nucleic Acid Amplification Techniques. Molecular Microbiology
Diagnostic Principles and Practice. D.H. Persing, F.C. Tenover, J. Versalovic, Y.-W. Tang,
E.R. Unger, D.A. Relman and T.J. White, ASM Press 43–69.
210 Dairy Microbiology: A Practical Approach
Lahtinen, S.J., A.C. Ouwehand, J.P. Reinikainen, J.M. Korpela, J. Sandholm and S.J. Salminen.
2006b. Intrinsic properties of so-called dormant probiotic bacteria, determined by flow
cytometric viability assays. Appl. Environ. Microbiol. 72(7): 5132–5134.
Lederberg, J. and A. Mccray. 2001. The Scientist:\’Ome Sweet\’Omics—A Genealogical
Treasury of Words. The Scientist 17(7).
Lepp, P.W. and D.A. Relman. 2011. Molecular Phylogenetic Analysis. Molecular Microbiology:
Diagnostic Principles and Practice. D.H. Persing, F.C. Tenover, J. Versalovic, Y.W. Tang,
E.R. Unger, D.A. Relman and T.J. White, ASM Press 161–180.
Loman, N.J., C. Constantinidou, J.Z. Chan, M. Halachev, M. Sergeant, C.W. Penn, E.R. Robinson
and M.J. Pallen 2012. High-throughput bacterial genome sequencing: an embarrassment
of choice, a world of opportunity. Nature Reviews Microbiology 10(9): 599–606.
Lopez-Garcia, P., F. Rodriguez-Valera, C. Pedros-Alio and D. Moreira. 2001. Unexpected
diversity of small eukaryotes in deep-sea Antarctic plankton. Nature 409(6820): 603–607.
Ludwig, W., F.O. Glockner and P. Yilmaz. 2011. The use of rRNA gene sequence data in the
classification and identification of prokaryotes. Methods in Microbiology 38: 349–384.
Lusk, T.S., A. R. Ottesen, J.R. White, M.W. Allard, E.W. Brown and J.A. Kase. 2012.
Characterization of microflora in Latin-style cheeses by next-generation sequencing
technology. BMC Microbiol. 12: 254.
Mackay, I.M. 2004. Real-time PCR in the microbiology laboratory. Clinical Microbiology and
Infection 10(3): 190–212.
Maidak, B.L., J.R. Cole, C.T. Parker, G.M. Garrity, N. Larsen, B. Li, T.G. Lilburn, M.J. McCaughey,
G.J. Olsen and R. Overbeek. 1999. A new version of the RDP (Ribosomal Database Project).
Nucleic Acids Research 27(1): 171–173.
Mannu, L. and A. Paba. 2002. Genetic diversity of lactococci and enterococci isolated from
home-made Pecorino Sardo ewes’ milk cheese. J. Appl. Microbiol. 92(1): 55–62.
Marras, S.E. 2006. Selection of fluorophore and quencher pairs for fluorescent nucleic acid
hybridization probes. Fluorescent Energy Transfer Nucleic Acid Probes. V. Didenko,
Humana Press. 335: 3–16.
Martin-Platero, A.M., M. Maqueda, E. Valdivia, J. Purswani and M. Martinez-Bueno. 2009.
Polyphasic study of microbial communities of two Spanish farmhouse goats’ milk cheeses
from Sierra de Aracena. Food Microbiol. 26(3): 294–304.
Masoud, W., M. Takamiya, F.K. Vogensen, S.r. Lillevang, W.A. Al-Soud, S.r.J. S√∏rensen and
M. Jakobsen. 2011. Characterization of bacterial populations in Danish raw milk cheeses
made with different starter cultures by denaturating gradient gel electrophoresis and
pyrosequencing. International Dairy Journal 21(3): 142–148.
Masoud, W., F.K. Vogensen, S. Lillevang, W. Abu Al-Soud, S.J. Sorensen and M. Jakobsen. 2012.
The fate of indigenous microbiota, starter cultures, Escherichia coli, Listeria innocua and
Staphylococcus aureus in Danish raw milk and cheeses determined by pyrosequencing
and quantitative real time (qRT)-PCR. Int. J. Food Microbiol. 153(1-2): 192–202.
Massire, C., D.R. Buelow, S.X. Zhang, R. Lovari, H.E. Matthews, D.M. Toleno, R.R. Ranken,
T.A. Hall, D. Metzgar and R. Sampath. 2013. PCR followed by electrospray ionization
mass spectrometry for broad-range identification of fungal pathogens. Journal of Clinical
Microbiology 51(3): 959–966.
Matamoros, S., P. Savard and D. Roy. 2011. Genotyping of Bifidobacterium longum subsp.
longum strains by multilocus variable number of tandem repeat analysis. J. Microbiol.
Methods 87(3): 378–380.
Maukonen, J., H.-L. Alakomi, L. Nohynek, K. Hallamaa, S. Leppämäki, J. Mättö and M.
Saarela. 2006. Suitability of the fluorescent techniques for the enumeration of probiotic
bacteria in commercial non-dairy drinks and in pharmaceutical products. Food Research
International 39(1): 22–32.
McInerney, J.O., M. Wilkinson, J.W. Patching, T.M. Embley and R. Powell. 1995. Recovery and
phylogenetic analysis of novel archaeal rRNA sequences from a deep-sea deposit feeder.
Appl. Environ. Microbiol. 61(4): 1646–1648.
212 Dairy Microbiology: A Practical Approach
Mellmann, A., D. Harmsen, C.A. Cummings, E.B. Zentz, S.R. Leopold, A. Rico, K. Prior, R.
Szczepanowski, Y. Ji and W. Zhang. 2011. Prospective genomic characterization of the
German enterohemorrhagic Escherichia coli O104: H4 outbreak by rapid next generation
sequencing technology. PloS One 6(7): e22751.
Mora, D., G. Ricci, S. Guglielmetti, D. Daffonchio and M.G. Fortina. 2003. 16S-23S rRNA
intergenic spacer region sequence variation in Streptococcus thermophilus and
related dairy streptococci and development of a multiplex ITS-SSCP analysis for their
identification. Microbiology (Reading, England) 149(Pt 3): 807–813.
Moreira, J.L.S., R.M. Mota, M.F. Horta, S.M.R. Teixeira, E. Neumann, J.R. Nicoli and Ã.C. Nunes.
2005. Identification to the species level of Lactobacillus isolated in probiotic prospecting
studies of human, animal or food origin by 16S-23S rRNA restriction profiling. BMC
Microbiology 5.
Moreno, Y., M.C. Collado, M.A. Ferrús, J.M. Cobo, E. Hernández and M. Hernández. 2006.
Viability assessment of lactic acid bacteria in commercial dairy products stored at 4°C
using LIVE/DEAD® BacLightTM staining and conventional plate counts. International
Journal of Food Science & Technology 41(3): 275–280.
Moschetti, G., G. Blaiotta, M. Aponte, P. Catzeddu, F. Villani, P. Deiana and S. Coppola.
1998. Random amplified polymorphic DNA and amplified ribosomal DNA spacer
polymorphism: powerful methods to differentiate Streptococcus thermophilus strains.
J. Appl. Microbiol. 85(1): 25–36.
Mullis, K.B. and F.A. Faloona. 1987. Specific synthesis of DNA in vitro via a polymerase-
catalyzed chain reaction. Methods in Enzymology 155: 335–350.
Muyzer, G., A. Teske, C.O. Wirsen and H.W. Jannasch. 1995. Phylogenetic relationships of
Thiomicrospira species and their identification in deep-sea hydrothermal vent samples by
denaturing gradient gel electrophoresis of 16S rDNA fragments. Archives of Microbiology
164(3): 165–172.
Naravaneni, R. and K. Jamil. 2005. Rapid detection of food-borne pathogens by using molecular
techniques. Journal of Medical Microbiology 54(Pt 1): 51–54.
NCBI. 2014. Random Amplified Polymorphic DNA (RAPD). Retrieved 1/2/2014, 2014, from
http://www.ncbi.nlm.nih.gov/projects/genome/probe/doc/techRAPD.shtml.
Neviani, E., B. Bottari, C. Lazzi and M. Gatti. 2013. New developments in the study of the
microbiota of raw-milk, long-ripened cheeses by molecular methods: the case of Grana
Padano and Parmigiano Reggiano. Front Microbiol. 4: 36.
Nikolic, M., A. Terzic-Vidojevic, B. Jovcic, J. Begovic, N. Golic and L. Topisirovic. 2008.
Characterization of lactic acid bacteria isolated from Bukuljac, a homemade goat’s milk
cheese. Int. J. Food Microbiol. 122(1-2): 162–170.
Nocker, A. and A.K. Camper. 2006. Selective removal of DNA from dead cells of mixed bacterial
communities by use of ethidium monoazide. Appl. Environ. Microbiol. 72(3): 1997–2004.
Nocker, A. and A.K. Camper. 2009. Novel approaches toward preferential detection of viable
cells using nucleic acid amplification techniques. FEMS Microbiol. Lett. 291(2): 137–142.
Nocker, A., C.Y. Cheung and A.K. Camper 2006. Comparison of propidium monoazide with
ethidium monoazide for differentiation of live vs. dead bacteria by selective removal of
DNA from dead cells. J. Microbiol. Methods 67(2): 310–320.
Nocker, A., A. Mazza, L. Masson, A.K. Camper and R. Brousseau. 2009. Selective detection
of live bacteria combining propidium monoazide sample treatment with microarray
technology. J. Microbiol. Methods 76(3): 253–261.
Nocker, A., T. Richter-Heitmann, R. Montijn, F. Schuren and R. Kort. 2010. Discrimination
between live and dead cellsin bacterial communities from environmental water samples
analyzed by 454 pyrosequencing. Int. Microbiol. 13(2): 59–65.
Nocker, A., P. Sossa-Fernandez, M.D. Burr and A.K. Camper. 2007a. Use of propidium
monoazide for live/dead distinction in microbial ecology. Appl. Environ. Microbiol.
73(16): 5111–5117.
Application of Molecular Methods for Microbial Identification in Dairy Products 213
Nocker, A., K.E. Sossa and A.K. Camper. 2007b. Molecular monitoring of disinfection efficacy
using propidium monoazide in combination with quantitative PCR. J. Microbiol. Methods
70(2): 252–260.
O’Sullivan, D.J., L. Giblin, P.L. McSweeney, J.J. Sheehan and P.D. Cotter. 2013. Nucleic acid-
based approaches to investigate microbial-related cheese quality defects. Front Microbiol.
4: 1.
Ogier, J.C., V. Lafarge, V. Girard, A. Rault, V. Maladen, A. Gruss, J.Y. Leveau and A. Delacroix-
Buchet. 2004. Molecular fingerprinting of dairy microbial ecosystems by use of temporal
temperature and denaturing gradient gel electrophoresis. Appl. Environ. Microbiol.
70(9): 5628–5643.
Pal, K., O. Szen, A. Kiss and Z. Naar. 2012. Comparison and evaluation of molecular methods
used for identification and discrimination of lactic acid bacteria. J. Sci. Food Agric. 92(9):
1931–1936.
Papadimitriou, K., S. Ferreira, N.C. Papandreou, E. Mavrogonatou, P. Supply, B. Pot and
E. Tsakalidou. 2012. Complete genome sequence of the dairy isolate Streptococcus
macedonicus ACA-DC 198. J. Bacteriol. 194(7): 1838–1839.
Parayre, S., H. Falentin, M.N. Madec, K. Sivieri, A.S. Le Dizes, D. Sohier and S. Lortal. 2007.
Easy DNA extraction method and optimisation of PCR-Temporal Temperature Gel
Electrophoresis to identify the predominant high and low GC-content bacteria from
dairy products. J. Microbiol. Methods 69(3): 431–441.
Pearson, W.R. and D.J. Lipman. 1988. Improved tools for biological sequence comparison.
Proc. Natl. Acad. Sci. USA 85(8): 2444–2448.
Pirondini, A., U. Bonas, E. Maestri, G. Visioli, M. Marmiroli and N. Marmiroli. 2010. Yield and
amplificability of different DNA extraction procedures for traceability in the dairy food
chain. Food Control 21(5): 663–668.
Porcellato, D., H. Grønnevik, K. Rudi, J. Narvhus and S.B. Skeie. 2012a. Rapid lactic acid
bacteria identification in dairy products by high-resolution melt analysis of DGGE bands.
Letters in Applied Microbiology 54(4): 344–351.
Porcellato, D., H.M. Ostlie, K.H. Liland, K. Rudi, T. Isaksson and S.B. Skeie. 2012b. Strain-level
characterization of nonstarter lactic acid bacteria in Norvegia cheese by high-resolution
melt analysis. J. Dairy Sci. 95(9): 4804–4812.
Prajapati, J., C. Khedkar, J. Chitra, S. Suja, V. Mishra, V. Sreeja, R. Patel, V. Ahir, V. Bhatt and
M. Sajnani. 2012. Whole-genome shotgun sequencing of Lactobacillus rhamnosus MTCC
5462, a strain with probiotic potential. J. Bacteriol. 194(5): 1264–1265.
Prajapati, J.B., C.D. Khedkar, J. Chitra, S. Suja, V. Mishra, V. Sreeja, R.K. Patel, V.B. Ahir, V.D.
Bhatt, M.R. Sajnani, S.J. Jakhesara, P.G. Koringa and C.G. Joshi. 2011. Whole-genome
shotgun sequencing of an Indian-origin Lactobacillus helveticus strain, MTCC 5463,
with probiotic potential. J. Bacteriol. 193(16): 4282–4283.
Pruesse, E., C. Quast, K. Knittel, B.M. Fuchs, W. Ludwig, J. Peplies and F.O. Glockner. 2007.
SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA
sequence data compatible with ARB. Nucleic Acids Research 35(21): 7188–7196.
Psoni, L., C. Kotzamanides, C. Andrighetto, A. Lombardi, N. Tzanetakis and E. Litopoulou-
Tzanetaki. 2006. Genotypic and phenotypic heterogeneity in Enterococcus isolates from
Batzos, a raw goat milk cheese. Int. J. Food Microbiol. 109(1-2): 109–120.
Quigley, L., O. O’Sullivan, T.P. Beresford, R. Paul Ross, G.F. Fitzgerald and P.D. Cotter. 2012a.
A comparison of methods used to extract bacterial DNA from raw milk and raw milk
cheese. J. Appl. Microbiol. 113(1): 96–105.
Quigley, L., O. O’Sullivan, T.P. Beresford, R.P. Ross, G.F. Fitzgerald and P.D. Cotter. 2012b.
High-throughput sequencing for detection of subpopulations of bacteria not previously
associated with artisanal cheeses. Appl. Environ. Microbiol. 78(16): 5717–5723.
Rademaker, J.L., H. Herbet, M.J. Starrenburg, S.M. Naser, D. Gevers, W.J. Kelly, J. Hugenholtz,
J. Swings and J.E. van Hylckama Vlieg. 2007. Diversity analysis of dairy and nondairy
Lactococcus lactis isolates, using a novel multilocus sequence analysis scheme and (GTG)
5-PCR fingerprinting. Appl. Environ. Microbiol. 73(22): 7128–7137.
214 Dairy Microbiology: A Practical Approach
Rademaker, J.L. W., J.D. Hoolwerf, A.A. Wagendorp and M.C. Te Giffel. 2006. Assessment
of microbial population dynamics during yoghurt and hard cheese fermentation and
ripening by DNA population fingerprinting. International Dairy Journal 16(5): 457–466.
Rademaker, J.L.W., M. Peinhopf, L. Rijnen, W. Bockelmann and W.H. Noordman. 2005. The
surface microflora dynamics of bacterial smear-ripened Tilsit cheese determined by
T-RFLP DNA population fingerprint analysis. International Dairy Journal 15(6-9): 785–794.
Radtke, A., T. Bruheim, J.E. Afset and K. Bergh. 2012. Multiple-locus variant-repeat assay
(MLVA) is a useful tool for molecular epidemiologic analysis of Streptococcus agalactiae
strains causing bovine mastitis. Veterinary Microbiology 157(3-4): 398–404.
Rahn, K., S.A. De Grandis, R.C. Clarke, S.A. McEwen, J.E. Galan, C. Ginocchio, R. Curtiss, 3rd
and C.L. Gyles. 1992. Amplification of an invA gene sequence of Salmonella typhimurium
by polymerase chain reaction as a specific method of detection of Salmonella. Molecular
and Cellular Probes 6(4): 271–279.
Rainey, F.A. 2011. How to describe new species of prokaryotes. Methods in Microbiology
38: 7–14.
Randazzo, C.L., C. Caggia and E. Neviani. 2009. Application of molecular approaches to study
lactic acid bacteria in artisanal cheeses. J. Microbiol. Methods 78(1): 1–9.
Randazzo, C.L., S. Torriani, A.D. Akkermans, W.M. de Vos and E.E. Vaughan. 2002. Diversity,
dynamics, and activity of bacterial communities during production of an artisanal Sicilian
cheese as evaluated by 16S rRNA analysis. Appl. Environ. Microbiol. 68(4): 1882–1892.
Rantsiou, K., V. Alessandria, R. Urso, P. Dolci and L. Cocolin. 2008a. Detection, quantification
and vitality of Listeria monocytogenes in food as determined by quantitative PCR. Int.
J. Food Microbiol. 121(1): 99–105.
Rantsiou, K., G. Comi and L. Cocolin. 2004. TherpoBgene as a target for PCR-DGGE analysis
to follow lactic acid bacterial population dynamics during food fermentations. Food
Microbiology 21(4): 481–487.
Rantsiou, K., R. Urso, P. Dolci, G. Comi and L. Cocolin. 2008b. Microflora of Feta cheese from
four Greek manufacturers. Int. J. Food. Microbiol. 126(1-2): 36–42.
Rasko, D.A., D.R. Webster, J.W. Sahl, A. Bashir, N. Boisen, F. Scheutz, E.E. Paxinos, R. Sebra,
C.-S. Chin and D. Iliopoulos. 2011. Origins of the E. coli strain causing an outbreak of
hemolytic–uremic syndrome in Germany. New England Journal of Medicine 365(8):
709–717.
Rasolofo, E.A., D. St-Gelais, G. LaPointe and D. Roy. 2010. Molecular analysis of bacterial
population structure and dynamics during cold storage of untreated and treated milk.
Int. J. Food Microbiol. 138(1-2): 108–118.
Reysenbach, A.L. and N.R. Pace. 1995. Reliable amplification of hyperthermophilic archaeal
16S rRNA genes by PCR. Thermophiles. F.T. Robb and A. Place. New York, Cold Spring
Habor Press 101–106.
Rodrigues, D., T.A.P. Rocha-Santos, A.C. Freitas, A.C. Duarte and A.M.P. Gomes 2012.
Analytical strategies for characterization and validation of functional dairy foods. TrAC
Trends in Analytical Chemistry 41(0): 27–45.
Sanchez, I., S. Sesena, J.M. Poveda, L. Cabezas and L. Palop. 2006a. Genetic diversity, dynamics,
and activity of Lactobacillus community involved in traditional processing of artisanal
Manchego cheese. Int. J. Food Microbiol. 107(3): 265–273.
Sanchez, J.I., L. Rossetti, B. Martinez, A. Rodriguez and G. Giraffa. 2006b. Application of reverse
transcriptase PCR-based T-RFLP to perform semi-quantitative analysis of metabolically
active bacteria in dairy fermentations. J. Microbiol. Methods 65(2): 268–277.
Sandrin, T.R., J.E. Goldstein and S. Schumaker. 2012. MALDI TOF MS profiling of bacteria at
the strain level: a review. Mass Spectrometry Reviews.
Saubusse, M., L. Millet, C. Delbes, C. Callon and M.C. Montel. 2007. Application of Single
Strand Conformation Polymorphism—PCR method for distinguishing cheese bacterial
communities that inhibit Listeria monocytogenes. Int. J. Food Microbiol. 116(1): 126–135.
Application of Molecular Methods for Microbial Identification in Dairy Products 215
Schoch, C.L., K.A. Seifert, S. Huhndorf, V. Robert, J.L. Spouge, C.A. Levesque, W. Chen and
F.B. Consortium. 2012. Nuclear ribosomal internal transcribed spacer (ITS) region as a
universal DNA barcode marker for Fungi. Proc. Natl. Acad. Sci. USA 109(16): 6241–6246.
Seale, R.B., R. Dhakal, K. Chauhan, H.M. Craven, H.C. Deeth, C.J. Pillidge, I.B. Powell and
M.S. Turnera. 2012. Genotyping of present-day and historical geobacillus species isolates
from milk powders by high-resolution melt analysis of multiple variable-number tandem-
repeat loci. Appl. Environ. Microbiol. 78(19): 7090–7097.
Seifert, K.A. 2009. Progress towards DNA barcoding of fungi. Molecular Ecology Resources
9(s1): 83–89.
Seifert, K.A., R.A. Samson, J. Houbraken, C.A. Lévesque, J.-M. Moncalvo, G. Louis-Seize and
P.D. Hebert. 2007. Prospects for fungus identification using CO1 DNA barcodes, with
Penicillium as a test case. Proc. Natl. Acad. Sci. USA 104(10): 3901–3906.
Senan, S., S. Grover and V.K. Batish. 2008. Comparison of specificity of different primer pairs
for the development of multiplex PCR assays for rapid identification of dairy Lacrobacilli.
International Journal of Science & Technology 3(2): 123–137.
Sergeev, N., D. Volokhov, V. Chizhikov and A. Rasooly. 2004. Simultaneous analysis of multiple
staphylococcal enterotoxin genes by an oligonucleotide microarray assay. Journal of
Clinical Microbiology 42(5): 2134–2143.
Shangkuan, Y.H., Y.S. Show and T.M. Wang. 1995. Multiplex polymerase chain reaction to
detect toxigenic Vibrio cholerae and to biotype Vibrio cholerae O1. The Journal of Applied
Bacteriology 79(3): 264–273.
Sheffield, V.C., D.R. Cox, L.S. Lerman and R.M. Myers. 1989. Attachment of a 40-base-pair
G + C-rich sequence (GC-clamp) to genomic DNA fragments by the polymerase chain
reaction results in improved detection of single-base changes. Proc. Natl. Acad. Sci. USA
86(1): 232–236.
Simon, M.C., D.I. Gray and N. Cook. 1996. DNA extraction and PCR methods for the detection
of Listeria monocytogenes in cold-smoked salmon. Appl. Environ. Microbiol. 62(3):
822–824.
Soggiu, A., E. Bendixen, M. Brasca, S. Morandi, C. Piras, L. Bonizzi and P. Roncada. 2013. Milk
and cheese microbiome for safety and quality of dairy products. Farm animal proteomics
2013. A. Almeida, D. Eckersall, E. Bencurova, S. Dolinska, P. Mlynarcik, M. Vincova and
M. Bhide, Wageningen Academic Publishers 262–265.
Solieri, L., T. Dakal and P. Giudici. 2013. Next-generation sequencing and its potential impact
on food microbial genomics. Ann. Microbiol. 63(1): 21–37.
Spratt, B.G. 1999. Multilocus sequence typing: molecular typing of bacterial pathogens in an era
of rapid DNA sequencing and the internet. Current Opinion in Microbiology 2(3): 312–316.
Stefano, M., B. Milena, L. Roberta and B. Lorenzo. 2008. Molecular typing of Staphylococcus
aureus isolated from Italian dairy products on the basis of coagulase gene polymorphism,
multiple-locus variable-number tandem-repeat and toxin genes. The Journal of Dairy
Research 75(4): 444–449.
Suzzi, G., M. Caruso, F. Gardini, A. Lombardi, L. Vannini, M.E. Guerzoni, C. Andrighetto and
M.T. Lanorte. 2000. A survey of the enterococci isolated from an artisanal Italian goat’s
cheese (semicotto caprino). J. Appl. Microbiol. 89(2): 267–274.
Swofford, D.L., G.J. Olsen, P.J. Waddell and D.M. Hillis. 1996. Chapter 11: Phylogenetic
inference. Molecular Systematics 2.
Tada, J., T. Ohashi, N. Nishimura, Y. Shirasaki, H. Ozaki, S. Fukushima, J. Takano, M. Nishibuchi
and Y. Takeda. 1992. Detection of the thermostable direct hemolysin gene (tdh) and the
thermostable direct hemolysin-related hemolysin gene (trh) of Vibrio parahaemolyticus
by polymerase chain reaction. Molecular and Cellular Probes 6(6): 477–487.
Tamura, K., J. Dudley, M. Nei and S. Kumar. 2007. MEGA4: Molecular Evolutionary Genetics
Analysis (MEGA) software version 4.0. Molecular Biology and Evolution 24(8): 1596–1599.
Tanigawa, K., H. Kawabata and K. Watanabe. 2010. Identification and typing of Lactococcus
lactis by matrix-assisted laser desorption ionization-time of flight mass spectrometry.
Appl. Environ. Microbiol. 76(12): 4055–4062.
216 Dairy Microbiology: A Practical Approach
Thompson, J.D., D.G. Higgins and T.J. Gibson 1994. CLUSTAL W: improving the sensitivity of
progressive multiple sequence alignment through sequence weighting, position-specific
gap penalties and weight matrix choice. Nucleic Acids Research 22(22): 4673–4680.
Tilburg, J.J., H.J. Roest, M.H. Nabuurs-Franssen, A.M. Horrevorts and C.H. Klaassen. 2012.
Genotyping reveals the presence of a predominant genotype of Coxiella burnetii in
consumer milk products. Journal of Clinical Microbiology 50(6): 2156–2158.
Torriani, S., F. Clementi, M. Vancanneyt, B. Hoste, F. Dellaglio and K. Kersters. 2001.
Differentiation of Lactobacillus plantarum, L. pentosus and L. paraplantarum species
by RAPD-PCR and AFLP. Syst. Appl. Microbiol. 24(4): 554–560.
Tsai, Y.L., C.J. Palmer and L.R. Sangermano. 1993. Detection of Escherichia coli in sewage and
sludge by polymerase chain reaction. Appl. Environ. Microbiol. 59(2): 353–357.
Van Hoorde, K., T. Verstraete, P. Vandamme and G. Huys. 2008. Diversity of lactic acid bacteria
in two Flemish artisan raw milk Gouda-type cheeses. Food Microbiol. 25(7): 929–935.
Vancanneyt, M., G. Huys, K. Lefebvre, V. Vankerckhoven, H. Goossens and J. Swings. 2006.
Intraspecific genotypic characterization of Lactobacillus rhamnosus strains intended for
probiotic use and isolates of human origin. Appl. Environ. Microbiol. 72(8): 5376–5383.
Ventura, M. and R. Zink. 2002. Specific identification and molecular typing analysis of
Lactobacillus johnsonii by using PCR-based methods and pulsed-field gel electrophoresis.
FEMS Microbiology Letters 217(2): 141–154.
Ventura, M. and R. Zink. 2003. Comparative sequence analysis of the tuf and recA genes and
restriction fragment length polymorphism of the internal transcribed spacer region
sequences supply additional tools for discriminating Bifidobacterium lactis from
Bifidobacterium animalis. Applied and Environmental Microbiology 69(12): 7517–7522.
Verdier-Metz, I., V. Michel, C. Delbes and M.C. Montel. 2009. Do milking practices influence
the bacterial diversity of raw milk? Food Microbiol. 26(3): 305–310.
Weisburg, W.G. , S.M. Barns, D.A. Pelletier and D.J. Lane. 1991. 16S ribosomal DNA
amplification for phylogenetic study. J. Bacteriol. 173(2): 697–703.
Whitehead, T.R. and M.A. Cotta. 1999. Phylogenetic diversity of methanogenic archaea in
swine waste storage pits. FEMS Microbiol. Lett. 179(2): 223–226.
Williams, A., U. Rodrigues and M. Collins. 1991. Intrageneric relationships of< i> Enterococci</
i> as determined by reverse transcriptase sequencing of small-subunit rRNA. Research
in Microbiology 142(1): 67–74.
Wilson, I. G. 1997. Inhibition and facilitation of nucleic acid amplification. Appl Environ
Microbiol 63(10): 3741–3751.
Wilson, I.G., J.E. Cooper and A. Gilmour. 1991. Detection of enterotoxigenic Staphylococcus
aureus in dried skimmed milk: use of the polymerase chain reaction for amplification and
detection of staphylococcal enterotoxin genes entB and entC1 and the thermonuclease
gene nuc. Appl. Environ. Microbiol. 57(6): 1793–1798.
Wittwer, C.T. and N. Kusukawa. 2004. Real-Time PCR. Molecular Microbiology Diagnostic
Principles and Practice. T.F.C. Persing D.H., Versalovic J., Tang Y.-W., Unger E.R., Relman
D.A., White T.J., ASM Press 71–84.
Woese, C.R. and G.E. Fox. 1977. Phylogenetic structure of the prokaryotic domain: the primary
kingdoms. Proc. Natl. Acad. Sci. USA 74(11): 5088–5090.
CHAPTER 7
INTRODUCTION
It is a generally accepted fact that milk from a healthy udder is sterile.
Therefore, pathogens that enter the milk originate either from the milking
parlor (i.e., equipment, environment, personnel), or from means of
transportation (i.e., equipment) to the factory for processing. This chapter
deals with the application of Hazard Analysis and Critical Control Points
(HACCP), in dairy products (including Risk Assessment) and it highlights
the importance of on-farm HACCP.
developed national standards for the supply of safe food and individual
companies and groupings in the food sector have developed their own
standards or programmes for auditing their suppliers such as BRC, IFS,
Dutch HACCP, etc. (National Advisory Committee on Microbiological
Criteria for Foods 1997, Bernard 1998, Forsythe and Hayes 1998, BRC
2005, Arvanitoyannis and Traikou 2005, Frost 2006). The plethora of more
than 20 different such schemes worldwide generates risks of uneven
levels of food safety, confusion over requirements, and increased cost
and complication for suppliers that find themselves obliged to conform
to multiple programmes (Papademas and Bintsis 2010). To fill this gap,
the International Organization for Standardization (ISO) published a
new food safety management system, the ISO 22000:2005—Food safety
management systems—Requirements for any organization in the food chain
(International Organization for Standardization 2005). These standards
provide a framework of internationally harmonized requirements for the
global approach to food safety issues. However, ISO 22000:2005 doesn’t
contain the non-exhaustive list of Good Manufacturing Practices present
in the Global Food Safety Initiative (GFSI) guidance document (GFSI
2007). Thus, the Publicly Available Specification—PAS 220:2008 developed
by British Standards Institution (BSI 2008) and published in Oct 2008 in
association with major branded dairies (Unilevel, Kraft, Nestle, Danone)
and is a new complementary standard to ISO 22000:2005. It specifies the
prerequisite programmes requirements in detail to assist in controlling
food safety hazards. In fact, it provides harmonization of prerequisite
programmes and industry best practice for food manufacturing. GFSI
agreed that the combination of ISO 22000:2005 and PAS 220:2008 contained
adequate content for approval, but that an industry-owned scheme
governing the combination of these two standards must exist. Consequently,
the Foundation for Food Safety Certification developed the FSSC 22000, an
auditable standard which incorporates food safety elements already known
from previous standards such as HACCP, ISO 22000:2005, BRC (2005) and
IFS as well as from specifications such as PAS 220:2008 (Sansawat and
Muliyil 2009) which has approved by the GFSI as a global benchmark in
food safety management. In addition, ISO, through the Technical Committee
ISO/TC 34 and based on PAS: 2008, has published ISO 22002-1:2009—
Prerequisite programmes on food safety—Part 1: Food manufacturing
(International Organization for Standardization 2009). Thus, ISO 22002-
1:2009, which has completed with the publication of ISO 22002-2:2013—
Prerequisite programmes on food safety—Part 2: Catering (International
Organization for Standardization 2013) and ISO 22002-3:2011—Prerequisite
programmes on food safety—Part 3: Farming (International Organization
for Standardization 2011), is now used in conjunction with ISO 22000:2005
throughout the whole food chain to manage the food safety aspects.
Food Safety Management Systems 219
On Farm HACCP
Although the responsibility lies with the manufacturer for ensuring that
the dairy foods manufactured are safe and suitable, there is a continuum of
effective effort or controls needed by other parties, including milk producers,
to assure the safety and suitability of milk products. It is important to
recognize that distributors, competent authorities and consumers also have
220 Dairy Microbiology: A Practical Approach
a role in ensuring the safety and suitability of milk and milk products. Due
to the special importance of the primary production to the safety of the
dairy products, major branded dairies have introduced their own on-farm
HACCP programmes. For example, Arla Foods has established a quality
program entitled Arlagεrden (‘the Arla farm’) to be used by their farmers.
The program specifies Arla Foods’ requirements not only for food safety and
milk composition, but also for animal welfare and environmental protection
(Junedahl et al. 2008). The Canadian dairy industry has begun implementing
an on-farm food-safety program called Canadian Quality Milk (Young et al.
2010). These quality assurance programmes starting at dairy farm level deals
with food safety, animal health and animal welfare issues to take account
of the demands of consumers and retailers (Noordhuizen and Metz 2005).
The HACCP concept, focused on risk management and prevention,
appears to be very promising to control on-farm processes. It can be easily
linked to both operational management and food chain quality assurance
and is suitable for certification (Noordhuizen 2003, Heeschen and Bluthgen
2004, Noordhuizen and Jorritsma 2006). In fact, introduction of HACCP on
dairy farms means nothing more than ‘structuring and formalising what the
truly good farmer would be doing anyway’ (Ryan et al. 1997). Until now,
the introduction of HACCP principles in on-farm management has hardly
been tested in practice, due to many objectively immeasurable processes
in an on-farm situation (Noordhuizen 2003).
Livestock species are an important reservoir of C. jejuni, shiga-toxin
producing E. coli, L. monocytogenes, Salmonella spp., and Y. enterocolitica
(Jayarao and Henning 2001, Murinda et al. 2002, Jayarao et al. 2006) and
other pathogenic bacteria that have been implicated in a number of food-
borne outbreaks (Papademas and Bintsis 2010). These pathogens have
been recovered with various frequencies from dairy-cattle faeces, bulk
milk tanks and the dairy-farm environment (Troutt 1995, Jayarao and
Henning 2001, Murinda et al. 2002, Wiedmann 2006, Van Kessel et al.
2004, Srinivasan et al. 2005, Karns et al. 2007, Vissers and Driehuis 2009).
Molecular epidemiological studies of E. coli O157:H7 have demonstrated
that subtypes of the organism can persist on cattle farms for years (Hancock
et al. 2001, Aspán and Eriksson 2010). The presence of food-borne pathogens
in milk is due to direct contact with contaminated sources in the dairy farm
environment and to excretion from the udder of an infected animal (Oliver
et al. 2005, Kousta et al. 2010). Fox et al. (2009) demonstrated the prevalence
of L. monocytogenes in the dairy farm environment and the need for good
hygiene practices to prevent its entry into the food chain and Hussein
and Sakuma (2005) described pre- and postharvest control measures to
ensure safety of dairy cattle products. D’Amico et al. (2008) reported that
the incidence of food-borne pathogens of concern in raw milk utilized for
farmstead cheese production was very low, whereas, Danielsson-Tham et
Food Safety Management Systems 221
al. (2004) stated that the conditions on a summer farm can hardly fulfill the
requirements for hygienic and strictly controlled conditions necessary for
safe processing of fresh cheese.
Outbreaks due to the consumption of unpasteurized milk (Peterson
2003, Centers for Disease Control and Prevention 2007, Lind et al. 2008,
Heuvelink et al. 2009, Lejeune and Rajala-Schultz 2009, Oliver et al. 2009),
inadequately pasteurized milk (Fahey et al. 1995) and cheeses made from
unpasteurized milk (Honish et al. 2005, Center for Science in the Public
Interest 2008, 2009) continue to occur. Campylobacteriosis and Salmonellosis
was the most common zoonotic diseases in humans in the European Union
during 2008, but incidences of both have fallen, whereas, the number of
cases of Verotoxigenic Escherichia coli (VTEC) rose by almost 9% and the
number of listeriosis cases in humans decreased by 11.1% (European Food
Safety Authority 2010). In 2008, there were 5,332 food-borne outbreaks in
the EU, sickening over 45,000 people and causing 32 deaths. Some 35 per
cent of these were triggered by Salmonella spp., with viruses and bacterial
toxins detailed as the next most common causes.
When milk is intended to be used for the manufacture of raw milk
products, hygienic conditions used at the primary production are one of the
most important public health control measures, as a high level of hygiene
of the milk is essential in order to obtain milk with a sufficiently low initial
microbial load in order to enable the manufacturing of raw milk products
that are safe and suitable for human consumption (Codex Alimentarius
Commission 2004). In such situations, additional control measures may
be necessary. In addition, increased emphasis in certain aspects of the
production of milk for raw milk products (animal health, animal feeding,
and milk hygiene monitoring) are specified and are critical to the production
of milk that is safe and suitable for the intended purpose. Interestingly,
the FDA/Health Canada risk assessment found that the risk of listeriosis
from soft-ripened cheeses made with raw milk is estimated to be 50 to 160
times higher than that from soft-ripened cheese made with pasteurized
milk (Food and Drug Administration/Health Canada 2012). This finding
is consistent with the fact that consuming raw milk and raw milk products
generally poses a higher risk from pathogens than do pasteurized milk and
its products.
E. coli, S. aureus, Corynebacterium bovis, Klebsiella spp., or Pseudomonas
spp., Streptococcus agalactiae, Streptococcus dysgalactiae and Streptococcus
uberis may cause, under certain circumstances, clinical mastitis (Hahn 1996,
Barkema et al. 1998). Special care should be taken for the use of antibiotics,
and Sawant et al. (2007) found that enteric bacteria such as E. coli from
healthy lactating cattle can be an important reservoir for tetracycline and
other antimicrobial resistance determinants.
222 Dairy Microbiology: A Practical Approach
Risk Analysis
More than 100 countries have signed the “Sanitary and Phytosanitary (SPS)
Agreement” of the World Trade Organization (WTO). This agreement states
that “whilst a country has the sovereign right to decide on the degree of
protection it wishes for its citizens, it must provide, if required, the scientific
evidence on which this level of protection rests.” It follows that if a country
sets a microbiological criterion—or any other limit—for a particular health
hazard in a particular food product, they must be able to explain, based
on scientific data, consideration of risk and societal considerations, the
rationale and justification for the criterion. The “Technical Barriers to Trade
(TBT) Agreement,” also requires that a country must not ask for a higher
degree of safety for imported goods than it does for goods produced in its
own country (International Commission on Microbiological Specifications
for Food 2006).
Risk assessment for food safety is part of the risk analysis framework,
provided by the Codex Alimentarius Commission (Codex Alimentarius
Commission 1999), which also includes risk management and risk
224 Dairy Microbiology: A Practical Approach
Hazard identification
• What is the problem?
• What is the hazards involved (chemical,
biological or physical)?
• Which foods are associated?
Risk characterization
• What is the risk to consumers and to sub-groups?
• What is the effect of different mitigation actions?
• What is the key assumptions and uncertainties
in the assessments?
Figure 1. The basic steps in risk assessment (After: Codex Alimentarius Commission (1999)).
must be met during the earlier steps in the food chain before consumption.
The FSO gives flexibility to the food chain to use different operations and
processing techniques that best suit their situation, as long as the maximum
hazard level specified at consumption is not exceeded (e.g., the replacement
of heat treatment with another equivalent technique, i.e., microfiltration).
The position of these concepts appearing in the food chain can be seen in
Fig. 2. Microbiological standards (Buchanan 1995) have been included in
European Legislation (EC 2005).
exposure
Figure 2. A presentation of a model food chain indicating the position of a Food Safety Objective
and derived Performance Objectives (After: International Commission on Microbiological
Specifications for Foods (2006)).
The development and use of a simple tool for food safety risk
assessment has been described by Ross and Sumner (2002) in spreadsheet
software format that embodies established principles of food safety risk
assessment. Microbial predictive modeling techniques have been developed
by many workers (George et al. 1996, Murphy et al. 1996, McClure et al.
1997, Xanthiakos et al. 2006, Membre and Lambent 2008). The validation of
such models has been investigated (Murphy et al. 1996, Ross 1996, Baranyi
et al. 1999, te Giffel and Zwietering 1999). Notermans et al. (1995) suggested
the use of quantitative risk assessment for setting critical limits at the CCPs
of a HACCP system for realistic levels of control.
Several computer programs have been launched for estimating bacterial
growth and inactivation in different products such as the Pathogen Modeling
Program, PMP (United States Department of Agriculture-Agricultural
Research Service 2006), Combase Predictor (Combase 2010), the online
resource for food safety risk analysis (Joint Institute for Food Safety and
Applied Nutrition 2010), the @RISK spreadsheet software (Palisade 2012),
the Crystal Ball Oracle Risk assessment software (Oracle 2012), Seafood
Spoilage and Safety Predictor (National Institute of Aquatic Resources 2009),
and Safe Foods (2010) for helping in development of HACCP-systems or
in performing quantitative risk assessment (McMeekin et al. 2006, 2008,
Hignette et al. 2008).
International agencies and all levels of government are increasingly
relying on, or at least recognizing the need to rely on, risk assessments
for decision-making in public health protection, international trade,
and to support cost-effective resource allocation including prioritizing
research directions (Council for Agricultural Science and Technology
1994, International Life Science Institute 1996, 2005, Food and Agriculture
Orginisation/World Health Orginisation 1997) and several authors have
highlighted the need for the application of risk assessment methods to food
safety (Jaykus 1996, Kindred 1996, Lammerding 1997, Buchanan et al. 1998,
Voysey and Brown 2000). For these reasons, a web-based quantitative risk
assessment system was developed (Chen et al. 2013), which enables users
to assess, compare and rank the risks posed by multiple food-hazard pairs
at all stages of the food supply system, from primary production, through
manufacturing and processing, to retail distribution and, ultimately, to the
consumer.
Full quantitative risk assessments are provided by the three Joint
Food and Agriculture Organisation/World Health Organisation Expert
Bodies: the Joint Expert Committee on Food Additives (JECFA); the Joint
Meeting on Pesticide Residues (JMPR); and the Joint Expert Meeting on
Microbiological Risk Assessment (JEMRA). Additional risk assessments
may be provided, on occasion, by ad hoc expert consultations, and
by member governments that have conducted their own assessments
228 Dairy Microbiology: A Practical Approach
Conclusions
The integration of HACCP plans with the development of fully quantitative
risk assessments offer very useful tools for controlling the entire farm-
to-table food chain. Moreover, the importance of dairy factory hygiene
needs to be highlighted, as well as the need for efficient controls of the
feed administered to production animals. For example, Salmonella spp. is a
food pathogen that according to the Rapid Alert System for Food and Feed
(RASFF) of the European Union (EU) is isolated in animal’s feed. Finally,
one must be aware and be able to follow changes regarding microbiological
standards as these change both in EU and globally.
References Cited
2005. ISO 22000:2005, Food safety management systems—Requirements for any organization
in the food chain. Geneva, Switzerland.
2007. Global Food Safety Initiative Guidance Document, 5th edition. URL http://www.
ciesnet.com/pfiles/programmes/foodsafety/GFSI_Guidance_Document_5th%20
Edition%20_September%202007.pdf.
2009. ISO 22002-1:2009—Prerequisite programmes on food safety—Part 1: Food manufacturing.
Geneva, Switzerland.
2010. A combined database for predictive microbiology. URL http://combase.arserrc.gov/
BrowserHome/SearchOptions/Search.aspx.
2010. CIES—The Food Business Forum Global Food Safety Initiative URL http://http://
www.ciesnet.com/4-press/4.2-press-release/index.asp.
2010. The Community Summary Report on Trends and Sources of Zoonoses and Zoonotic
Agents and Foodborne Outbreaks in the European Union in 2008. The EFSA Journal
1496: 1–288.
2011. ISO 22002-3:2011—Prerequisite programmes on food safety—Part 3: Farming. Geneva,
Switzerland.
2013. ISO 22002-2:2013—Prerequisite programmes on food safety—Part 2: Catering. Geneva,
Switzerland.
Albert, I., R. Pouillot and J.B. Denis. 2005. Stochastically modeling Listeria monocytogenes growth
in farm tank milk. Risk Analysis 25(5): 171–185.
Ali, A.A. and R.M. Fischer. 2002. Implementation of HACCP to bulk condensed milk production
line. Food Reviews International 18(2-3): 177–190.
Ali, A.A. and R.M. Fischer. 2005. Implementation of HACCP to bulk cream and butter
production Line. Food Reviews International 21189–210.
Arvanitoyannis, I.S. and A. Traikou. 2005. A comprehensive review of the implementation of
hazard analysis critical control point (HACCP) to the production of flour and flour-based
products. Critical Reviews in Food Science and Nutrition 45: 327–370.
Food Safety Management Systems 229
Arvanitoyannis, I.S. and A.A. Mavropoulos. 2000. Implementation of the hazard analysis
critical control point (HACCP) system to Kasseri/Kefalotiri and Anevato cheese
production lines. Food Control 11(1) 31–40.
Arvanitoyiannis, I.S., T.H. Varzakas and M. Koukaliaroglou-van Houwelingen. 2009.
Implementing HACCP and ISO 22000 for foods of animal origin—Dairy products. pp
91–180. In: I.S. Arvanitoyiannis [ ed.]. HACCP and ISO 22000—Application to Foods of
Animal Origin. Wiley—Blackwell, Oxford, UK.
Aspán, A. and E. Eriksson. 2010. Verotoxigenic Escherichia coli O157:H7 from Swedish
cattle; isolates from prevalence studies versus strains linked to human infections—A
retrospective study. BMC Veterinary Research 6: 7–14.
Baranyi, J., C. Pin and T. Ross. 1999. Validating and comparing predictive models. International
Journal of Food Microbiology 48: 159–166.
Barkema, H.M., Y.H. Schukken, T.G. Lam, M.L. Beiboer, H. Wilmine, G. Benediktus and A.
Brand. 1998. Incidence of clinical mastitis in dairy herds grouped in three categories by
bulk milk somatic cell counts. Journal of Dairy Science 81: 411–419.
Bemrah, N., M. Sanaa, M.H. Cassin, M.W. Griffiths and O. Cerf. 1999. Quantitative risk
assessment of human listeriosis from consumption of soft cheese made from raw milk.
Preventative Veterinary Medicine 37: 129–145.
Bernard, D. 1998. Developing and implementing HACCP in the USA. Food Control 9(2-3):
91–95.
British Retail Consortium Global Standard. 2005. British Retail Consortium Global Standard
—Food Issue 4. URL http://www.brc.org.uk/standards/downloads/food_std_
background.pdf.
British Standard Institute. 2008. British Standard Institute Group. URL http://www.bsigroup.
com/en/Assessment-and-certification-services/management-systems/Standards-and-
Schemes/PAS-220.
Brouillaud-Delattre, A., M. Maire, C. Collette, C. Mattei and C. Lahellec. 1997. Predictive
microbiology of dairy products: influence of biological factors affecting growth of Listeria
monocytogenes. Journal of AOAC International 80(4): 913–919.
Buchanan, R.L. 1995. The role of microbiological criteria and risk assessment in HACCP. Food
Microbiology 12: 421–424.
Buchanan, R.L. and R.C. Whiting. 1998. Risk assessment: a means for linking HACCP plans
and public health. Journal of Food Protection 61(11): 1531–1534.
Buchanan, R.L., A.M. Lammerding, M. van Schothorst and T.A. Roberts. 1998. International
Commission on Microbiological Specifications for Foods working group on microbial
risk assessment. Potential application of risk assessment techniques to microbiological
issues related to international trade in food and food products. Journal of Food Protection
61: 1075–1086.
Burgess, K., C. Heggum, S. Walker and M. van Schothorst. 1994. Recommendations for the
hygienic manufacture of milk and milk based products. Bulletin of the IDF 292: 12–19.
Center for Science in the Public Interest. 2008. Outbreak alert 2008. URL http://www.cspinet.
org/new/pdf/outbreak_alert_2008_report_final.pdf.
Center for Science in the Public Interest. 2009. Outbreak alert Analyzing foodborne outbreaks
1998–2007. URL http://cspinet.org/new/pdf/outbreakalertreport09.pdf.
Center for Science in the Public Interest. 2010. Outbreaks Alert Database – Dairy. URL http://
www.cspinet.org/foodsafety/outbreak/outbreaks.php?column=food&colval=Dairy.
Centers for Disease Control and Prevention. 2007. Centers for Disease Control and Prevention,
MMWR Weekly Report, http://www.cdc.gov/mmwr/preview/mmwrhtml/mm5608a3.
htm.
Chen, Y., S.B. Dennis, E. Hartnett, G. Paoli, R. Pouillot, T. Ruthman and M. Wilson. 2013. FDA-
iRISK—A comparative risk assessment system for evaluating and ranking food-hazard
pairs: case studies on microbiological hazards. Journal of Food Protection 3: 376–385.
230 Dairy Microbiology: A Practical Approach
Clough, H.E., D. Clancy and N.P. French. 2006. Vero-Cytotoxigenic Escherichia coli O157 in
pasteurized milk containers at the point of retail: a qualitative approach to exposure
assessment. Risk Analysis 26(5): 1291–1309.
Codex Alimentarius Commission. 1999. Principles and Guidelines for the Conduct of
Microbiological Risk Assessment. URL www.codexalimentarius.net/download/standards/.../
CXG_030e.pdf.
Codex Alimentarius Commission. 2003. Recommended international code of practice—General
principles of food hygiene. CAC/RCP 1–1969, Rev. 4–2003. Rome: FAO.
Codex Alimentarius Commission. 2004. Code of Hygiene Practice for Milk and Milk Products.
Codex Alimentarius Committee. CAC/RCP 57–2004. http://www.codexalimentarius.
org/input/download/standards/10087/CXP_057e.pdf.
Codex Alimentarius Commission. 2009. Food Hygiene—Basic texts. 4th Edition. Codex
Alimentarius Commission. Joint FAO/WHO Food Standards Programme. Rome.
Coffie, R., E. Cummins and S. Ward. 2009. Exposure assessment of mycotoxins in dairy milk.
Food Control 20: 239–249.
Council for Agricultural Science and Technology. 1994. Foodborne pathogens: risk and
consequences. Task Force Report No 122. Ames, Iowa, USA.
Cullor, J.S. 1997. HACCP: Is it coming to the dairy? Journal of Dairy Science 80: 3449–3452.
D’Amico, D.J., E. Groves and C.W. Donnelly. 2008. Low incidence of foodborne pathogens of
concern in raw milk utilized for farmstead cheese production. Journal of Food Protection
71(8): 1580–1589.
Danielsson-Tham, M.L., E. Eriksson, S. Helmersson, M. Leffler, L. Lüdtke, M. Steen, S. Sørgjerd
and W. Tham. 2004. Causes behind a human cheese-borne outbreak of gastrointestinal
listeriosis. Foodborne Pathogens Diseases 1(3): 153–159.
Dijkers, J.H., T. Huurnink, P.P.L. Pennings and M.G. van den Berg. 1995. An example of HACCP
application in an existing pasteurized milk plant, following the Codex Alimentarius
model. Bulletin of the IDF 302: 11–34.
EC. 2002. Regulation (EC) No. 178/2002 of the European Parliament and of the Council of
28 January 2002 laying down the general principles and requirements of food safety
law, establishing the European Food Standards Agency and laying down procedures in
matters of food safety. Official Journal of the European Union L31: 1–24.
EC. 2004a. Regulation (EC) No. 852/2004 of the European Parliament and of the Council of 29
April 2004 on the hygiene of foodstuffs ( including HACCP principles). Official Journal
of the European Union L139: 1–54.
EC. 2004b. Regulation (EC) No.853/2004/EC of the European Parliament and of the Council
of 29 April 2004 laying down specific hygiene rules for food of animal origin. Official
Journal of the European Union L139: 55–205.
EC. 2005. Commission Regulation (EC) No. 2073/2005 of 15 November 2005 laying down
microbiological criteria for certain products. Official Journal of the European Union
L338: 1–26.
Fahey, T., D. Morgan, C. Gunneburg, G.K. Adak, F. Majid and E. Kaczmarski. 1995. An outbreak
of Campylobacter jejuni enteritis associated with failed milk pasteurisation. Journal of
Infections 31: 137–143.
Food and Agriculture Organization/World Health Organisation. 1995. Application of Risk
Analysis to Food Standards Issues. Report of the Joint FAO/WHO Expert Consultation,
World Health Organisation, Geneva, Switzerland.
Food and Agriculture Organization/World Health Organisation. 1997. Risk Management
and Food Safety. Report of a Joint FAO/WHO Consultation. Food and Agriculture
Organization of the United Nations, Rome, Italy.
Food and Agriculture Organization/World Health Organisation. 2004. Risk assessment of
Listeria monocytogenes in ready-to-eat foods. Microbiological risk assessment series
5. Food and Agriculture Organization of the United Nations, Rome, Italy. URL http://
www.fao.org/es/esn/food/risk_mra_listeria_report_en.stm.
Food Safety Management Systems 231
Food and Drug Adminsitration/Health Canada. 2012. Quantitative Assessment of the Risk
of Listeriosis from Soft-Ripened Cheese Consumption in the United States and Canada:
Draft Report. Food Directorate—Health Canada/Center for Food Safety and Applied
Nutrition—Food and Drug Administration, U.S. Department of Health and Human
Services.
Forsythe, S.J. and P.R. Hayes. 1998. Food Hygiene, Microbiology and HACCP, 3rd ed. Maryland:
Aspen Publishers, pp. 276–326.
Fox, E., T. O’Mahony, M. Clancy, R. Dempsey, M. O’Brien and K. Jordan. 2009. Listeria
monocytogenes in the Irish dairy farm environment. Journal of Food Protection 72(7):
1450–1456.
Frey, H.C. 1992. Quantitative Analysis of Uncertainty and Variability in Environmental Policy
Making. http://www4.ncsu.edu/~frey/reports/frey_92.pdf.
Frost. 2006. Early adopters underline benefits of new ISO standard for safe food supply chains
ISO Management Systems—March–April 2006: 21– 23.
Gardner, I.A. 1997. Testing to Fulfill HACCP (Hazard Analysis Critical Control Points)
Requirements: Principles and Examples. Journal of Dairy Science 80: 3453–3457.
George, S.M., L.C.C. Richardson and M.W. Peck. 1996. Predictive models of the effect of
temperature, pH and acetic and lactic acids on the growth of Listeria monocytogenes.
International Journal of Food Microbiology 32:(1-2) 73–90.
[Globalgap]. 2007. General Regulations Integrated Farm Assurance, Version 3.0, GLOBALG.A.P.,
Cologne, Germany.
Hahn, G. 1996. Pathogenic bacteria in raw milk-situation and significance. Proceedings of
Symposium on Bacteriological Quality of Raw Milk—Wolfpassing Austria, March 13–15
1996, IDF Special Issue 9601: 67–83.
Hancock, D., T. Besser, J. Lejeune, M. Davis and D. Rice. 2001. The control of VTEC in the
animal reservoir. International Journal of Food Microbiology 66: 71–78.
Hathaway, S.C. 1997. Development of food safety risk assessment guidelines for foods of animal
origin in international trade. Journal of Food Protection 60(11): 1432–1438.
Hauser, R., E. Breidenbach and K.D.C. Stärk. 2007. Swiss federal veterinary office risk
assessments: advantages and limitations of the qualitative method. pp. 519–526 In: J-.L.
Auget, N. Balakrishman, M. Mesbah and G. Molenbergh [eds.]. Advances in Statistical
Methods for the Health Sciences. Birkhäuser, Boston.
Heeschen, W.H. and A.H. Bluthgen. 2004. Carry-over of environmental contaminants into milk
and food hygiene assessment ⁄ management. Bulletin of the IDF 386: 28–39.
Heuvelink, A.E., C. van Heerwaarden, A. Zwartkruis-Nahuis, J.H.C. Tilburg, M.H. Bos,
F.G.C. Heilmann, A. Hofhuis, T. Hoekstra and E. de Boer. 2009. Two outbreaks of
campylobacteriosis associated with the consumption of raw cows’ milk. International
Journal of Food Microbiology 134(1-2): 70–74.
Hignette, G., P. Buche, O. Couvert, J. Dibie-Barthélemy, D. Doussot, O. Haemmerlé, E. Mettler
and L. Soler. 2008. Semantic annotation of Web data applied to risk in food. International
Journal of Food Microbiology 128: 174–180.
Honish, L., G. Predy, N. Hislop, L. Chui, K. Kowalewska-Grochowska, L. Trottier, C. Kreplin
and I. Zazulak. 2005. An outbreak of E. coli O157:H7 hemorrhagic colitis associated with
unpasteurized gouda cheese. Canadian Journal of Public Health 96: 182–184.
Hussein, H.S. and T. Sakuma. 2005. Shiga toxin-producing Escherichia coli: pre- and postharvest
control measures to ensure safety of dairy cattle products. Journal of Food Protection
68(1): 199–207.
International Life Science Institute. 1996. A conceptual framework to assess the risks of
human disease following exposure to pathogens. International Life Science Institute,
North America—Risk Science Institute Pathogen Risk Assessment Working Group, Risk
Analysis 16: 841–848.
International Life Science Institute. 2005. Achieving continuous improvement in reductions
in foodborne listeriosis—a risk-based approach. ILSI Research Foundation Risk Science
Institute Journal of Food Protection 68(9): 1932–94.
232 Dairy Microbiology: A Practical Approach
International Life Science Institute. 2012. Tools for Microbiological Risk Assessment. ILSI
Europe Report Series, ILSI Europe, Brussels, Belgium.
Jayarao, B.M. and D.R. Henning. 2001. Prevalence of Foodborne Pathogens in Bulk Tank Milk.
Journal of Dairy Science 84: 2157–2162.
Jayarao, B.M., S.C. Donaldson, B.A. Straley, A.A. Sawant, N.V. Hegde and J.L. Brown. 2006. A
Survey of Foodborne Pathogens in Bulk Tank Milk and Raw Milk Consumption Among
Farm Families in Pennsylvania. Journal of Dairy Science 89: 2451–2458.
Jaykus, L.A. 1996. The application of quantitative risk assessment to microbial food safety
risks. Critical Review in Microbiology 22: 279–293.
Joint Institute for Food Safety and Applied Nutrition. 2010. FoodRisk.org—The online Resource
for Food Safety Risk Analysis. Joint Institute for Food Safety and Applied Nutrition
[Internet document] URL http://www.foodrisk.org/commodity/animal/dairy/index.
cfm. Accessed 29/01/2010.
Julien, M.-C., P. Dion, C. Lafreniere, H. Antoun and P. Drouin. 2008. Sources of clostridia in
raw milk on farms. Applied and Environmental Microbiology 74(20): 6348–6357.
Junedahl, P., E. Øgaard, H. Alnås and U. Nilsson. 2008. Arla Foods sees ISO 22000 becoming
international benchmark for food safety. ISO Management Systems May–June, 23–28.
Karns, J.S., J.S. Van Kessel, B.J. McClusky and M.L. Perdue. 2007. Incidence of Escherichia coli
O157:H7 and E. coli Virulence Factors in US Bulk Tank Milk as Determined by Polymerase
Chain Reaction. Journal of Dairy Science 90: 3212–3219.
Katz, S.E. and M.S. Brady. 2000. Antibiotic residues in food and their significance. Food
Biotechnology 14: 147–171.
Kindred, T.P. 1996. Risk assessment and its role in the safety of foods of animal origin. Journal
of the American Veterinary Medical Association 209: 2055–2056.
Kousta, M., M. Mataragas, P. Skandamis and E.H. Drosinos. 2010. Prevalence and sources of
cheese contamination with pathogens at farm and processing levels. Food Control 21:
805–815.
Lammerding, A.M. 1997. An overview of microbial food safety risk assessment. Journal of
Food Protection 60: 1420–1425.
Lejeune, J.T. and P.J. Rajala-Schultz. 2009. Food safety: unpasteurized milk: a continued public
health threat. Clinical Infectious Disease 48(1): 93–100.
Lind, L., J. Reeser, K. Stayman, M. Deasy, M. Moll, A. Weltman, V. Urdaneta and M.D.
Ostroff. 2008. Salmonella Typhimurium infection associated with raw milk and cheese
Consumption—Pennsylvania, 2007. Journal of American Medical Associaton 299(4):
402–404.
Maunsell, B. and D.J. Bolton. 2004. Guidelines for Food Safety Management on Farms, The
Food Safety Department. Teagasc—The National Food Centre, Dublin.
Mauropoulos, A.A. and I.S. Arvanitoyannis. 1999. Implementation of hazard analysis critical
control point to Feta and Manouri cheese production lines. Food Control 10: 213–219.
McClure, P.J., A.L. Beaumont, J.P. Sutherland and T.A. Roberts. 1997. Predictive modelling of
growth of Listeria monocytogenes The effects on growth of NaCl, pH, storage temperature
and NaNO2. International Journal of Food Microbiology 34(3): 221–232.
McMeekin, T.A. and T. Ross. 2002. Predictive microbiology: providing a knowledge-based
framework for change management. International Journal of Food Microbiology 78:
133–153.
McMeekin, T.A., J. Baranyi, J. Bowman, P. Dalgaard, M. Kirk, T. Ross, S. Schmid and M.H.
Zwietering. 2006. Information systems in food safety management. International Journal
of Food Microbiology 112: 181–194.
McMeekin, T.A., J. Bowman, O. McQuestin, L. Mellefont, T. Ross and M. Tamplin. 2008. The
future of predictive microbiology: Strategic research, innovative applications and great
expectations. International Journal of Food Microbiology 128: 2–9.
Membré, J.-M. and R.J.W. Lambent. 2008. Application of predictive modelling techniques
in industry: From food design up to risk assessment. International Journal of Food
Microbiology 128: 10–15.
Food Safety Management Systems 233
Mortimore, S. 2001. How to make HACCP really work in practice. Food Control 12: 209–215.
Mortimore, S. and C. Wallace. 1998. HACCP—A practical approach. 2nd Edition. Chapman
and Hall, London.
Murinda, S.E., L.T. Nguyen, S.J. Ivey, B.E. Gillespie, R.A. Almeida, F.A. Draughon and S.P.
Oliver. 2002. Prevalence and molecular characterization of Escherichia coli O157:H7 in
bulk tank milk and fecal samples from cull cows: a 12-month survey of dairy farms in
East Tennessee. Journal of Food Protection 65: 752–759.
Murphy, P.M., M.C. Rea and D. Harrington. 1996. Development of a predictive model for
growth of Listeria monocytogenes in a skim milk medium and validation studies in a
range of dairy products. Journal of Applied Bacteriology 80(5): 557–564.
National Advisory Committee on Microbiological Criteria for Foods. 1997. Hazard Analysis
and Critical Control Point Principles and Application Guidelines. National Advisory
Committee on Microbiological Criteria for Foods. 14 August 1997. URL http://
haccpalliance.org/alliance/microhaccp.pdf.
[National Institute of Aquatic Resources]. 2009. Seafood Spoilage and Safety Predictor (SSSP)
software v. 3.1 URL http://sssp.dtuaqua.dk.
Noordhuizen, J.P.T.M. 2003. HACCP, total quality management and dairy herd health. pp.
1281–1289. In: H. Roginski, J.W. Fuquay and P.F. Fox [eds.]. Encyclopedia of Dairy Sciences,
Vol. 3. Academic Press, London.
Noordhuizen, J.P.T.M. and J.H.M. Metz. 2005. Quality control on dairy farms with emphasis
on public health, food safety, animal health and welfare. Livestock Production Science
94: 51–59.
Noordhuizen, J.P.T.M. and R. Jorritsma. 2006. The role of animal hygiene and animal health
in dairy operations URL http://www.isah-soc.org/documents/2005/keynotespeakers/
mon/nordhuisen%20.doc.
Notermans, S., A.W. Barendsz and F. Rombouts. 2002. The evolution of microbiological
risk assessment in food production. pp. 5–43. In: M. Brown and M. Stringer [eds.].
Microbiological risk assessment in food processing. Woodhead Publishing Ltd.,
Cambridge, UK.
Notermans, S., G. Gallhoff, M.H. Zwietering and G.C. Mead. 1995. The HACCP concept:
specification of criteria using quantitative risk assessment. Food Microbiology 12: 81–90.
Oliver, S.P., B.M. Jayarao and R.A. Almeida. 2005. Foodborne pathogens in milk and the dairy
farm environment: food safety and public health implications. Foodborne Pathogen
Disease 2(2): 115–129.
Oliver, S.P., K.J. Boor, S.C. Murphy and S.E. Murinda. 2009. Food safety hazards associated
with consumption of raw milk. Foodborne Pathogen Disease 6(7): 793–806.
[Oracle]. 2012. The Crystal Ball Oracle Risk assessment software. http://www.oracle.com/
us/products/applications/crystalball/overview/index.html.
[Palisade]. 2012. The @RISK software maker. http://www.palisade.com/risk/?gclid=CPmg
gpv877UCFUbMtAodUnIAYg.
Papademas, P. and T. Bintsis. 2002. Microbiology of ice cream and related products. In: Dairy
Microbiology Handbook—The Microbiology of Milk and Milk Products, pp. 213–260.
Robinson R.K. (ed.). John Wiley and Sons, Inc., New York.
Papademas, P. and T. Bintsis. 2010. Food safety management systems (FSMS) in the dairy
industry: A review. International Journal of Dairy Technology 63: 1–15.
Peeler, J.T. and V.K. Bunning. 1994. Hazard assessment of Listeria monocytogenes in the
processing of bovine milk. Journal of Food Protection 57: 689–697.
Peterson, M.C. 2003. Campylobacter jejuni enteritis associated with consumption of raw milk.
Journal of Environmental Health 65: 20–21.
Presidential Congressional Commission on Risk Assessment and Risk Management. 1997.
Framework for environmental health risk management. Washington DC, USA: The
Presidential/Congressional Commission on Risk Assessment and Risk Management.
234 Dairy Microbiology: A Practical Approach
Reybroeck, W. 1996. Modern methods for bacteriological quality control of raw milk.
Proceedings of Symposium on Bacteriological Quality of Raw Milk, Wolfpassing Austria,
March 13–15, 1996, IDF Special Issue 9601: 131–140.
Ross, T. 1996. Indices for performance evaluation of predictive models in food microbiology.
Journal of Applied Bacteriology 81(5) 501–508.
Ross, T. and J. Sumner. 2002. A simple, spreadsheet-based, food safety risk assessment tool.
International Journal of Food Microbiology 77(1-2): 39–53.
Ryan, M.J., P.G. Wall, G.K. Adak, H.S. Evans and J.M. Cowden. 1997. Outbreaks of infectious
intestinal disease in residential institutions in England and Wales 1992–1994. Journal of
Infection 34: 49–54.
[Safe Foods]. 2010. Promoting Food Safety through a New Integrated Risk Analysis Approach
for Foods URL http://www.safefoods.nl/ default.aspx.
Sanaa, M., B. Pourel, J.L. Menard and F. Serieys. 1993. Risk factors associated with
contamination of raw milk by Listeria monocytogenes in dairy farms. Journal of Dairy
Science 76: 2891–2898.
Sanaa, M., L. Coroller and O. Cerf. 2004. Risk assessment of listeriosis linked to the consumption
of two soft cheeses made from raw milk: Camembert of Normandy and Brie of Meaux.
Risk Analysis 24(2): 389–399.
Sandrou, D.K. and I.S. Arvanitoyannis. 2000a. Implementation of Hazard Analysis Critical
Control Point (HACCP) system to the dairy industry: current status and perspectives.
Food Reviews International 16(1): 77–111.
Sandrou, D.K. and I.S. Arvanitoyannis. 2000b. Application of Hazard Analysis Critical
Control Point (HACCP) system to the cheese-making industry: a review. Food Reviews
International 16(3): 327–368.
Sansawat, S. and V. Muliyil. 2009. Understanding the FSSC 22000 Food Standard. URL http://
www.sgs.com/foodsafety.
Sawant, A.A., N.V. Hegde, B.A. Straley, S.C. Donaldson, B.C. Love, S.J. Knabel and B.
Jayarao. 2007. Antimicrobial-Resistant Enteric Bacteria from Dairy Cattle. Applied and
Environmental Microbiology 73(1): 156–163.
Schlundt, J. 2000. Comparison of microbiological risk assessment studies published.
International Journal of Food Microbiology 58: 197–202.
Srinivasan, V., H.M. Nam, L.T. Nguyen, B. Tamilselvam, S.E. Murinda and S.P. Oliver. 2005.
Prevalence of antimicrobial resistance genes in Listeria monocytogenes isolated from dairy
farms. Foodborne Pathogens Disease 2: 201–211.
Straley, B.A., S.C. Donaldson, N.V. Hedge, A.A. Sawant, V. Srinivasan, S.P. Oliver and B.M.
Jayarao. 2006. Public health significance of antimicrobial-resistant Gram-negative bacteria
in raw bulk tank milk. Foodborne Pathogens Disease 3: 222–233.
te Giffel, M.C. and M.H. Zwietering. 1999. Validation of predictive models describing the
growth of Listeria monocytogenes. International Journal of Food Microbiology 46(2):
135–149.
The International Commission on Microbiological Specifications for Foods. 2006. A simplified
guide to understanding and using Food Safety Objectives and Performance Objectives.
The International Dairy Federation/Food and Agriculture Organization of the United Nations.
2004. Guide to good dairy farming practice. A joint publication of the International Dairy
Federation and the Food and Agriculture Organization of the United Nations. FAO, Rome.
Troutt, H.F., J. Gillespie and B.I. Osburn 1995. Implementation of HACCP program on
farms and ranches. pp 36–57. In: A.M. Pearson and T.R. Dutson [eds.]. HACCP in Meat,
Poultry and Fish Processing—Advances in Meat Research Series, Vol. 10, Chapman &
Hall, New York.
United States Department of Agriculture—Agricultural Research Service. 2006. Pathogens
Modeling Program, United States Department of Agriculture—Agricultural Research
Service, Microbial Food Safety Research Unit URL http://ars.usda.gov/services/docs.
htm?Docid =6786.
Food Safety Management Systems 235
Unnevehr, L.J. and H.H. Jensen. 1999. The economic implications of using HACCP as a food
safety regulatory standard. Food Policy 4: 625–635.
Van Kessel, J.S., J.S. Karns, L. Gorski, B.J. McCluskey and M.L. Perdue. 2004. Prevalence of
Salmonellae, Listeria monocytogenes, and Fecal Coliforms in Bulk Tank Milk on US Dairies.
Journal of Dairy Science 87: 2822–2830.
Vissers, M.M.M. and F. Driehuis. 2009. On-farm Hygienic milk production. pp 1–22. In: A.Y.
Tamime [ed.]. Milk Processing and Quality Management. Blackwell Publishing, Ltd.
Vissers, M.M.M., F. Driehuis, M.C. Te Giffel, P. De Jong and J.M.G. Lankveld. 2006. Improving
farm management by modeling the contamination of farm tank milk with butyric acid
bacteria. Journal of Dairy Science 89: 850–858.
Vissers, M.M.M., F. Driehuis, M.C. Te Giffel, P. De Jong and J.M.G. Lankveld. 2007.
Concentrations of Butyric Acid Bacteria Spores in Silage and Relationships with Aerobic
Deterioration. Journal of Dairy Science 90: 928–936.
Vissers, M.M.M., F. Driehuis, M.C. Te Giffel, P. De Jong and J.M.G. Lankveld. 2007a. Short
Communication: quantification of the transmission of microorganisms to milk via dirt
attached to the exterior of teats. Journal of Dairy Science 90: 3579–3582.
Voysey, P.A. and M. Brown. 2000. Microbiological risk assessment: a new approach to food
safety control. International Journal of Food Microbiology 58(3): 173–179.
Wiedmann, M. 2006. ADSA Foundation Scholar Award—An Integrated Science Based
Approach to Dairy Food Safety: Listeria monocytogenes as a Model System. Journal of
Dairy Science 86: 1865–1875.
Xanthiakos, K., D. Simos, A.S. Angelidis, G.J.-E. Nychas and K. Koutsoumanis. 2006. Dynamic
modeling of Listeria monocytogenes growth in pasteurized milk. Journal of Applied
Microbiology 100: 1289–1298.
Young, I., S. Hendrick, S. Parker, A. Rajić, J.T. McClure, J. Sanchez and S.A. Mc Ewen. 2010.
Attitudes towards the Canadian quality milk program and use of good production
practices among Canadian dairy producers. Preventive Veterinary Medicine 94: 43–53.
Color Plate Section
Chapter 3
Sample
Plating
Purification
Confirmation
Purified colonies are confirmed with standard tests:
• Gram staining
• Motility test
• Carbohydrate fermentation test
• D-hemolysis and CAMP test (lysis tests)
Figure 2. ISO 11290 for detection of L. monocytogenes in food samples (After: Scharlau 2007).
240 Dairy Microbiology: A Practical Approach
Figure 4. Procedure for Isolation and Identification of Campylobacter spp. from Milk (After:
United States Food and Drug Administration).
242 Dairy Microbiology: A Practical Approach
Figure 5. Detection of Shigella spp. in Food Samples (After: AES Chemunex 2008).
Color Plate Section 243
Chapter 5
SCFAs production
Enhancement of
barrier function
Improved lactose
tolerance and lowering Suppression of
of cholesterol levels pathogenic bacteria
infection
Stabilization of
Improvement of gut microbiota Production of
mineral absorption and composition antimicrobials/
production vitamins/ bacteriocins
micronutrients
Dairy
Dairy
Dairy Microbiology
Dairy Microbiology
The objective of this book is to provide a scientific
Microbiology
background
TheTheobjective
objective toofdairy
The objective ofthis microbiology
thisof
book this isbook
book by re-examining
isprovide
to provide
isto toprovide the
a scientific
a ascientific
scientific
Microbiology
Microbiology
Microbiology
basic concepts
background of general
tomicrobiology
dairy food microbiology
microbiology and the
by re-examining thethe the
Microbiology
background
background to todairy
dairy microbiology byby re-examining
re-examining
microbiology
basic
basic basic
concepts
concepts ofof raw
concepts general
of milk
general while
of general
foodfood offering
food
microbiology
microbiology a and
practical
microbiology and and
the the the
Microbiology
approach
microbiology
microbiology to ofthe
microbiology ofraw following
raw ofmilkraw aspects:
milk
milkwhile while
whileoffering well-known
offering
offering a and
a apracticalpractical
practical
newfound
approach
approach pathogens
approach
to tothethe to
followingthatfollowing
the
following are of major
aspects:
aspects:aspects: concern
well-known toand
well-known
well-known the
andand
A Practical
dairy
newfound
newfound newfound
industry.
pathogens
pathogens pathogensthatthat that
areare of of aremajor
major of concern
major concern
concern to to to the
thethe
AAAPractical
Practical
Practical
dairy dairy industry.
industry.
dairy industry.
Topics addressed include Cronobactersakazakii and its
importance
Topics
Topics
Topics
addressed toaddressed
addressed infant
include
include
includemilk
formula Cronobactersakazakii
or Mycobacterium
Cronobactersakazakii
Cronobactersakazakii and itsand
and its
its
Approach
Approach
Approach
Approach
avium importance to
subspecies infant formula milk
paratuberculosis or that
Mycobacterium
importance
importance to toinfant
infant formula
formulamilk milkor(MAP)
orMycobacterium might
Mycobacterium
beavium avium
connected subspecies
to chronic paratuberculosis
human diseases (MAP)
(Crohn’s), that
themight
avium subspecies
subspecies paratuberculosis
paratuberculosis (MAP)
(MAP) that
thatmight
might
role of be connected
dairy starter to chronic
cultures human diseases
in manufacturing (Crohn’s), the
fermented
be connected
be connected to to
chronic
chronic human
human diseases
diseases (Crohn’s),
(Crohn’s), thethe
dairy role of
products, dairy starter
developing cultures in manufacturing fermented
role of of
role dairy
dairy starter
starter cultures
cultures in novel
in manufacturing functional
manufacturing
dairy products, developing novel functional dairy
fermenteddairy
fermented
products
dairy
dairyproducts,through developing
products, the incorporation
developing novelnoveloffunctional
probiotic
functional strains,
dairy
products through the incorporation of probioticdairystrains,
insights in the field ofincorporation
molecular methods for microbial
Editor:
products
products through
through thethe incorporation of ofprobiotic
probiotic strains,
strains,
insights in the field of molecular methods for microbial
Editor:
insights field of molecular methods microbial
Photis
the compulsory
the systemsapplication
compulsory application
(FSMS) food
of
to the dairy foodsafety
safety
industry. management
management
systems
systems (FSMS)
(FSMS) to theto the dairy industry.
dairy industry.
Photis
The book will provide dairy professionals and students
The book will provide dairy professionals and students
alike
TheThe the
bookbook latest
alike theinformation
willwill
provide
provide
latest dairy on
dairy
information this vast
professionals thistopic.
professionals
on vastand students
and
topic. students
Papademas
alike thethe
alike latest information
latest information ononthis thisvast topic.
vast topic.
Papademas
Editor
Editor
PhotisEditor
Editor
Papademas
Photis Papademas
K24428
Photis
PhotisPapademas
Papademas
6000 Broken Sound Parkway, NW
Suite 300, Boca Raton, FL 33487
711 Third Avenue
New York, NY 10017
an informa business 2 Park Square, Milton Park
w w w. c rc p r e s s . c o m Abingdon, Oxon OX14 4RN, UK 9 7814829 298673
781482 298673 A Science Publishers Book
9 781482 298673
9 781482 298673