(Mónica V. Cunha, João Inácio) Veterinary Infec PDF
(Mónica V. Cunha, João Inácio) Veterinary Infec PDF
(Mónica V. Cunha, João Inácio) Veterinary Infec PDF
Mnica V. Cunha
Joo Incio Editors
Veterinary Infection
Biology: Molecular
Diagnostics and
High-Throughput
Strategies
METHODS
IN
MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Mnica V. Cunha
Instituto Nacional de Investigao Agrria e Veterinria, Lisbon, Portugal
Centro de Biologia Ambiental, Faculdade de Cincias, Universidade de Lisbon, Lisbon, Portugal
Joo Incio
Instituto Nacional de Investigao Agrria e Veterinria, Lisbon, Portugal
School of Pharmacy and Biomolecular Sciences, University of Brighton, United Kingdom
Editors
Mnica V. Cunha
Instituto Nacional de Investigao
Agrria e Veterinria
Lisbon, Portugal
Joo Incio
Instituto Nacional de Investigao
Agrria e Veterinria
Lisbon, Portugal
ISSN 1064-3745
ISSN 1940-6029 (electronic)
ISBN 978-1-4939-2003-7
ISBN 978-1-4939-2004-4 (eBook)
DOI 10.1007/978-1-4939-2004-4
Springer New York Heidelberg Dordrecht London
Library of Congress Control Number: 2014953901
Springer Science+Business Media New York 2015
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction
on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material supplied specifically for
the purpose of being entered and executed on a computer system, for exclusive use by the purchaser of the work.
Duplication of this publication or parts thereof is permitted only under the provisions of the Copyright Law of the
Publishers location, in its current version, and permission for use must always be obtained from Springer. Permissions
for use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable to prosecution
under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of publication, neither
the authors nor the editors nor the publisher can accept any legal responsibility for any errors or omissions that may be
made. The publisher makes no warranty, express or implied, with respect to the material contained herein.
Printed on acid-free paper
Humana Press is a brand of Springer
Springer is part of Springer Science+Business Media (www.springer.com)
Foreword
Infectious diseases of domestic and wild animals have a tremendous impact on the economy
and public health worldwide. Sixty percent of the pathogens that cause diseases in humans
have their source in animals (whether domestic or wild), as do 75 % of emerging human
diseases and 80 % of the pathogens that potentially could be used in bioterrorism. Animal
diseases that are transmissible to humans (collectively known as zoonoses), such as avian
influenza, rabies, bovine tuberculosis, and brucellosis, represent a very relevant public
health threat worldwide, being imperative to prevent and/or control them. Likewise,
pathogens that are not zoonotic but have a negative impact on livestock production, and
the associated financial income, should not be overlooked. Some estimates suggest that
world production of food animals is reduced by more than 20 % due to disease, which may
be particularly important in developing countries where the unavailability of animal protein
is socially impacting and inevitably exacerbates infectious diseases risk.
The World Organization for Animal Health (OIE) was created in 1924 with the aim of
controlling the international spread of infectious animal diseases, but currently, over and
above this original mission, the new mandate is to improve animal health worldwide. The
OIE builds its expertise on a global network of Reference Laboratories and Collaborating
Centers that play an essential role in prevention, detection, surveillance, and control of
animal diseases worldwide. Reference Laboratories serve as centers of scientific and technical expertise for official listed diseases and have particular responsibility for carrying out
confirmatory diagnostic tests for these diseases. Collaborating Centers are centers of expertise in key competences related to the management of a particular animal health issue, such
as epidemiology, diagnosis, risk analysis, welfare, or veterinary training. This global network
of excellence enables OIE to remain at the forefront of world veterinary scientific expertise
and successfully carry out its activities, providing authoritative scientific opinions and advice
on key topics, such as animal health and welfare and diagnostic techniques. Moreover, the
OIE is involved in the preparation of standards on diagnostic assays and their official validation which are published in the OIE Terrestrial Manual and Aquatic Manual and updated
annually by the World Assembly of the Delegates of the OIE (available from www.oie.int).
The achievements of biotechnology are significantly contributing to the development
of novel, rapid, and powerful diagnostic assays for animal diseases, namely based on molecular approaches, such as standard and real-time polymerase chain reaction and isothermal
nucleic acid amplification methods. The recent advent of high-throughput sequencing
technologies is also boosting the prompt detection of all microbes in a sample, uncovering
novel pathogens and significantly advancing veterinary diagnostic microbiology. Furthermore,
the analysis of single nucleotide and large sequence polymorphisms in the genome of
pathogens provides novel information on their traits, such as virulence and antimicrobial
resistance determinants, and supports epidemiological studies. Outstandingly, biotechnology is enabling the development of diagnostic kits that can ultimately be used in point-ofdecision settings, away from centralized laboratories to assist in local decision-making, for
instance during an outbreak. OIE is aware of the opportunities and challenges that these
novel molecular-based approaches bring to veterinary diagnosis and several OIE Reference
vi
Foreword
Laboratories and Collaborating Centers are also dedicated to Research and Development
activities on these fields. In this context, the present Springers book on the use of molecular biology strategies in veterinary laboratory practice showcases the versatility of the veterinarian profession in meeting the challenges posed by the continuous advance of science,
namely in the biotechnology field. The editors have succeeded in bringing together an
impressive group of renowned collaborating authors, primarily veterinary doctors and
researchers of veterinary science, from institutions with established and prized expertise in
the fields of molecular biology and veterinary diagnostics, including several OIE Reference
Laboratories and Collaborating Centers. This book provides therefore an excellent and
authoritative overview of molecular biology strategies for pathogen detection and characterization, along with the most recent technological innovations and their potential to
reconstruct transmission chains and to disclose pathogen biology, evolution, and interaction with the host. Beyond the techniques, the book also discusses the integration of these
new molecular approaches in the framework of veterinary practice and animal health
management. In recognition of the challenge faced by the public and private components
of Veterinary Services and all veterinarians to improve the animal health status worldwide,
this book may aid veterinary laboratories in setting up molecular diagnosis valences and is
a valuable resource to veterinary doctors and laboratory technicians, researchers, and
students of veterinary medicine and science interested in knowing more about these
challenging, but promising molecular strategies to unravel animal infectious diseases.
Director General
World Organization for Animal Health (OIE)
Bernard Vallat
Preface
Infectious diseases of animals represent a significant economic burden to economies worldwide and may raise relevant Public Health concerns. Molecular diagnostics has emerged as
the fastest growing and trending segment of the in vitro diagnostics industry and seems also
promising in the veterinary sector, providing novel and powerful tools that enable the fast
and accurate diagnosis of animal diseases.
In this book, we provide an overview of molecular biology tools for pathogen detection
and characterization, along with potential applications to disclose pathogen biology and
interaction with the host. In Part I, biobanking, biosafety and good laboratory practices,
biological specimen collection and processing, quality assurance and control, and validation
of molecular diagnostic assays for veterinary use are concisely introduced as a basis for the
molecular strategies enclosed in protocol sections. The significance and integration of
molecular biology methods in the framework of veterinary practice are discussed, along
with a SWOT analysis presenting our view on the advantages, limitations, opportunities
and weaknesses, or challenges, of these approaches. The rationale is on the techniques and
applications rather than the pathogen or disease models. Part II is dedicated to the molecular detection and identification of animal pathogens using a wide range of established techniques and covering, also, emerging diagnosis approaches. A brief overview of the advances
on nanoscience and microfluidics is provided, including a discussion on how the recent
developments in these research fields may be translated into field deployable biosensors.
Part III addresses genotyping tools for assessing the genetic landscape and epidemiology of
pathogens. Part IV introduces integrative omics and high-throughput technologies as powerful research tools, yielding massive amounts of information that may ultimately improve
the control and management of infectious diseases. We strived to span a broad range of
molecular approaches, from simple and affordable to emerging and sophisticated, which we
anticipate will be useful for veterinary doctors and laboratory technicians, researchers, and
students of veterinary medicine and science.
Lisbon, Portugal
Mnica V. Cunha
Joo Incio
vii
Acknowledgements
The editors were supported by Fundaco para a Cincia e Tecnologia (FCT, Portugal)
through projects PTDC/CVT/111634/2009 (JI) and PTDC/CVT/117794/2010 and
in the framework of Projecto 3599Promover a Produo Cientfica e Desenvolvimento
Tecnolgico e a Constituio de Redes Temticas (MVC). Alice Santos (INIAV IP, Lisbon,
Portugal) is acknowledged for the cover image artwork. The authors of Chap. 17 are
acknowledged for providing the fluorescence microscopy image used in the artwork.
ix
Contents
Foreword . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
PART I
v
vii
ix
xv
IN THE
PART II
19
31
43
61
77
xi
109
125
133
xii
Contents
PART III
145
153
163
175
183
193
209
219
235
245
253
287
311
323
Contents
PART IV
xiii
335
349
361
373
391
401
415
437
475
491
513
529
Contributors
AHMED AHMED WHO/FAO/OIE and National Collaborating Centre for Reference
and Research on Leptospirosis, Department of Biomedical Research, Royal Tropical
Institute (KIT), Amsterdam, The Netherlands
SANDRA ANTUNES Instituto de Higiene e Medicina Tropical, Universidade Nova de
Lisboa, Lisbon, Portugal
ALICIA ARANAZ Departamento de Sanidad Animal, Facultad de Veterinaria,
Universidad Complutense de Madrid, Madrid, Spain
MARIA BALLESTER Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA,
Campus de la Universitat Autnoma de Barcelona, Bellaterra (Cerdanyola del
Valls), Spain; Centre de Recerca en Agrigenmica (CRAG), Consorci CSIC-IRTAUAB-UB, Bellaterra, Spain; Departament de Cincia Animal i dels Aliments,
Facultat de Veterinria, Universitat Autnoma de Barcelona, Bellaterra, Spain
KRISTIANE BARINGTON Department of Veterinary Disease Biology, Faculty of Health
and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
CARRIE BATTEN The Pirbright Institute, Surrey, UK
SNDOR BELK OIE Collaborating Centre for the Biotechnology-based Diagnosis of
Infectious Diseases in Veterinary Medicine, Swedish University of Agricultural
Sciences (SLU), Uppsala, Sweden; Department of Biomedical Sciences and Veterinary
Public Health (BVF), Swedish University of Agricultural Sciences (SLU), Uppsala,
Sweden; The National Veterinary Institute (SVA), Uppsala, Sweden
THOMAS BJARNSHOLT Department of International Health, Immunology and
Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences,
University of Copenhagen, Frederiksberg C, Denmark; Department of Clinical
Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
STEVEN VAN BORM Veterinary and Agrochemical Research Center, Brussels, Belgium
ANA BOTELHO Instituto Nacional de Investigao Agrria e Veterinria IP, Lisbon,
Portugal
METTE BOYE National Veterinary Institute, Technical University of Denmark,
Kongens Lyngby, Denmark
BRONWYN E. CAMPBELL Department of Veterinary Science, The University of
Melbourne, Parkville, VIC, Australia
CINZIA CANTACESSI Department of Veterinary Science, The University of Melbourne,
Parkville, VIC, Australia
ANA CANTO Instituto Nacional de Investigao Agrria e Veterinria IP, Lisbon,
Portugal
ANA CLUDIA CARREIRA iMed.ULResearch Institute for Medicines and
Pharmaceutical Sciences, Nanomedicine & Drug Delivery Systems Group, Faculdade
de Farmcia, Universidade de Lisboa, Lisbon, Portugal; Instituto Nacional de
Investigao Agrria e Veterinria, I.P. (INIAV, I.P.), Lisbon, Portugal
xv
xvi
Contributors
Contributors
xvii
xviii
Contributors
CHRIS OURA School of Veterinary Medicine, University of the West Indies, St. Augustine,
Trinidad and Tobago
MARINA POPARA SaBio, Instituto de Investigacin en Recursos Cinegticos IREC,
UCLM-JCCM, Ciudad Real, Spain
SUSANNE ELISABETH PORS Department of Veterinary Disease Biology, Faculty of
Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
FERNANDO RODRIGUEZ Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA,
Campus de la Universitat Autnoma de Barcelona, Bellaterra (Cerdanyola del
Valls), Spain
FLORIAN ROEBER Faculty of Veterinary and Agricultural Sciences, The University
of Melbourne, Parkville, VIC, Australia; AusDiagnostics Pty. Ltd., Beaconsfield,
NSW, Australia
TOON ROSSEEL Veterinary and Agrochemical Research Center, Brussels, Belgium
ANKE RUETTGER Institute of Molecular Pathogenesis at Friedrich-Loeffler-Institut
(Federal Research Institute for Animal Health), Jena, Germany
KONRAD SACHSE Institute of Molecular Pathogenesis at Friedrich-Loeffler-Institut
(Federal Research Institute for Animal Health), Jena, Germany
MONA SALEH Clinical Division of Fish Medicine, University of Veterinary Medicine,
Vienna, Austria
SUSAN SANCHEZ Athens Veterinary Diagnostic Laboratory, College of Veterinary
Medicine, The University of Georgia, Athens, GA, USA
ILDA SANTOS-SANCHES Centro de Recursos Microbiolgicos, Departamento de Cincias
da Vida, Faculdade de Cincias e Tecnologia, Universidade Nova de Lisboa, Lisbon,
Portugal
NICK SAUNDERS Public Health England (PHE), Microbiology Services Colindale,
London, UK
BEVERLY SCHMITT United States Department of Agriculture (USDA), Animal
and Plant Health Inspection Service, National Veterinary Services Laboratories,
Ames, Iowa, USA
CHRISTIANE SCHNEE Institute of Molecular Pathogenesis at Friedrich-Loeffler-Institut
(Federal Research Institute for Animal Health), Jena, Germany
HOLLY SELLERS Poultry Diagnostic and Research Center, College of Veterinary
Medicine, The University of Georgia, Athens, GA, USA
IAN SHARP Public Health England (PHE), Microbiology Services Colindale, London,
UK
HATEM SOLIMAN Clinical Division of Fish Medicine, University of Veterinary
Medicine, Vienna, Austria; Fish Medicine and Managements, Department
of Animal Medicine, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
LINDA D. STEWART Institute for Global Food Security, School of Biological Sciences,
Queens University Belfast, Northern Ireland, UK
FERNANDO TELES Laboratory of Mycology/Microbiology Unit and Centre for Malaria
and Tropical Diseases (CMDT), Institute of Hygiene and Tropical Medicine
(IHMT), Universidade Nova de Lisboa (UNL), Lisbon, Portugal
ROGRIO TENREIRO Centro de Biodiversidade, Genmica Integrativa e Funcional,
Faculdade de Cincias, Universidade de Lisboa, Lisbon, Portugal
Contributors
xix
Part I
Introducing Nucleic Acid Testing in the Veterinary Laboratory
Chapter 1
Overview and Challenges of Molecular Technologies
in the Veterinary Microbiology Laboratory
Mnica V. Cunha and Joo Incio
Abstract
Terrestrial, aquatic, and aerial animals, either domestic or wild, humans, and plants all face similar health
threats caused by infectious agents. Multifaceted anthropic pressure caused by an increasingly growing and
resource-demanding human population has affected biodiversity at all scales, from the DNA molecule to
the pathogen, to the ecosystem level, leading to species declines and extinctions and, also, to host-pathogen
coevolution processes.
Technological developments over the last century have also led to quantic jumps in laboratorial testing that have highly impacted animal health and welfare, ameliorated animal management and animal
trade, safeguarded public health, and ultimately helped to secure biodiversity. In particular, the field of
molecular diagnostics experienced tremendous technical progresses over the last two decades that significantly have contributed to our ability to study microbial pathogens in the clinical and research laboratories.
This chapter highlights the strengths, weaknesses, opportunities, and threats (or challenges) of molecular
technologies in the framework of a veterinary microbiology laboratory, in view of the latest advances.
Key words Molecular diagnostics, Veterinary microbiology, Nucleic acid testing, Animal health,
Animal welfare, Public health, Biodiversity, Validation, In-house, Molecular assays, Quality assurance,
Quality control
Introduction
Active and passive surveillance of infectious diseases in farmed and
wild animals enable the detection of (re)emergent pathogens and the
monitoring of endemic and/or zoonotic diseases and, ultimately,
back up stakeholders, supporting national and international policies
aimed at the protection of animal health and welfare, food security
and public health, and the trade of animals and animal products.
During recent years, the world has witnessed the emergence or
reemergence of a number of major animal diseases such as footand-mouth disease (FMD), bluetongue, classical swine fever (CSF),
and avian influenza, which are emblematic examples of the overwhelming economic, social, and public health impacts that animal diseases may cause. Effective surveillance and control of animal
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_1, Springer Science+Business Media New York 2015
Table 1
SWOT analysis of molecular diagnostics technologies in the veterinary microbiology laboratory
Strengths
Weaknesses
(continued)
10
Table 1
(continued)
Opportunities
Threats
11
12
13
14
directed to all the people in the diagnostic chain, from the owner,
farmer, or professional that collects the sample to the person that
reports the result and the veterinarian that has to make an enlighten
evidence-based decision.
During easily transmitted and economically impacting disease
outbreaks, such as FMD, the early detection of infection is particularly important but is often impractical to test all the samples in
centralized reference laboratories. Moreover, in many regions of
the globe, even the veterinary reference laboratories may not be
adequately equipped to face these threats. In this context, the
availability of diagnostic methods for use in the point of decision
(e.g., directly at farms, slaughterhouses, or local veterinary clinics
or laboratories) is particularly relevant and is forecasted to represent a major opportunity for the IVD market. Molecular diagnostics methods generally are not applicable in these circumstances,
requiring the use of sophisticated and expensive equipments operated by skilled technicians. The increasing availability of portable
thermal cyclers may be potentially useful for a wider use of molecular technologies, allowing their implementation closest to the
regions where outbreaks occur (e.g., in regional laboratories or in
the field) [15]. The use of isothermal technologies for the amplification of nucleic acids, such as LAMP, may also prove useful in this
context because of its lesser requirement for sophisticated equipments. The detection of amplified products by electrophoresis is
not practical outside the laboratory, but disposable strips for lateral
flow detection of nucleic acids are also already commercially available [19]. Other fields experiencing great developments are microfluidics and nanotechnology. The procedures normally required
for the molecular detection of pathogens in biological samples,
including cell lysis, extraction of nucleic acids, amplification of
specific genomic targets, and their detection, can all potentially be
integrated into autonomous miniaturized microfluidic systems,
enabling the development of molecular diagnostics devices (commonly referred to as lab-on-a-chip), which can be used in the point
of decision [3540]. Nanotechnologies have also sparked interest
in the development of new diagnostic tests, including molecular
tests, taking advantage of the unique properties that many materials exhibit at the nanoscale [41, 42]. Biosensors based on these
technologies are likely to eliminate the need for any steps of extraction and amplification of nucleic acids from samples [42], and several prototypes were recently described in the literature for the
detection of viruses and bacteria.
Conclusion
The unprecedented growth of human population, increasingly
demanding for more food supplies and animal protein, changes in
land use, globalization, industrialization, deforestation, and climate
15
Acknowledgments
This work was supported by Fundaco para a Cincia e Tecnologia
(FCT) through projects PTDC/CVT/111634/2009 and PTDC/
CVT/117794/2010 and in the framework of Projecto 3599
Promover a Produo Cientfica e Desenvolvimento Tecnolgico e
a Constituio de Redes Temticas.
16
References
1. Hughes MD (2013) Molecular diagnostics,
market trends and outlook. Enterprise Analysis
Corporation (www.eacorp.com), Stamford,
United States
2. Anonymous (2014) Veterinary diagnostics
market by products (clinical chemistry, hematology analyzers, molecular diagnostics, immunodiagnostics, diagnostics imaging), by animals
(companion, food-producing, dog, livestock,
poultry, swine): global forecast to 2018.
Markets and Markets, Dallas, United States
(Executive Summary)
3. Dorman MA, Blair CD, Collins JK et al (1985)
Detection of bovine herpesvirus 1 DNA immobilized on nitrocellulose by hybridization with
biotinylated DNA probes. J Clin Microbiol
22:990995
4. Viseshakul N, Panyim S (1990) Specific DNA
probe for the sensitive detection of Trypanosoma
evansi. Southeast Asian J Trop Med Public
Health 21:2127
5. Almeida A, Albuquerque P, Arajo R, Ribeiro
N, Tavares F (2013) Detection and discrimination of common bovine mastitis-causing streptococci. Vet Microbiol 164:370377
6. Saiki RK, Scharf S, Faloona F et al (1985)
Enzymatic amplification of beta-globin genomic
sequences and restriction site analysis for diagnosis of sickle cell anemia. Science 230:13501354
7. Kawasaki ES, Clark SS, Coyne MY et al (1988)
Diagnosis of chronic myeloid and acute lymphocytic leukemias by detection of leukemiaspecific mRNA sequences amplified in vitro.
Proc Natl Acad Sci U S A 85:56985702
8. Belk S, Ballagi-Pordny A, Flensburg J et al
(1989) Detection of pseudorabies virus DNA
sequences by the polymerase chain reaction.
Arch Virol 108:279286
9. Biswas B, Mukherjee D, Mattingly-Napier BL
et al (1991) Diagnostic application of polymerase
chain reaction for detection of Ehrlichia risticii in
equine monocytic ehrlichiosis (Potomac horse
fever). J Clin Microbiol 29:22282233
10. Ballagi-Pordny A, Klingeborn B, Flensburg J
et al (1990) Equine herpesvirus type 1: detection of viral DNA sequences in aborted fetuses
with the polymerase chain reaction. Vet
Microbiol 22:373381
11. Wheeler R, Wilmore H, Savva D et al (1990)
Diagnosis of ovine toxoplasmosis using
PCR. Vet Rec 126:249
12. Figueroa JV, Chieves LP, Johnson GS et al
(1992) Detection of Babesia bigemina-infected
carriers by polymerase chain reaction amplification. J Clin Microbiol 30:25762582
34.
35.
36.
37.
38.
39.
40.
41.
42.
17
Chapter 2
Significance and Integration of Molecular Diagnostics
in the Framework of Veterinary Practice
Alicia Aranaz
Abstract
The field of molecular diagnostics in veterinary practice is rapidly evolving. An array of molecular techniques
of different complexity is available to facilitate the fast and specific diagnosis of animal diseases. The choice
for the adequate technique is dependent on the mission and attributions of the laboratory and requires
both a knowledge of the molecular biology basis and of its limitations.
The ability to quickly detect pathogens and their characteristics would allow for precise decisionmaking and target measures such as prophylaxis, appropriate therapy, and biosafety plans to control disease
outbreaks. In practice, taking benefit of the huge amount of data that can be obtained using molecular
techniques highlights the need of collaboration between veterinarians in the laboratory and practitioners.
Key words Molecular diagnostics, PCR, RT-PCR, Veterinary clinical microbiology, Pathogens
Introduction
Molecular techniques, fueled by polymerase chain reaction
(PCR), have revolutionized the diagnostics in veterinary practice.
Molecular approaches are now widely accepted for identification
of etiological agents and in many cases have replaced traditional
systems based on the more cumbersome phenotypic identification. Several assays are currently described for the detection and
identification of many pathogens of the OIE-listed diseases and
other diseases of importance to international trade. However,
compared to serological techniques, molecular assays are still officially recommended as prescribed tests only for a limited number
of diseases, when concerning the international trade and movement of animals and animal products (http://www.oie.int/en/
international-standard-setting/terrestrial-manual).
The development of molecular methods has provided new
tools for enhanced surveillance and outbreak detection, and this
may result in better implementation of infection control programs.
Benefits of using molecular diagnostics are clear in terms of time to
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_2, Springer Science+Business Media New York 2015
19
20
Alicia Aranaz
21
Level of Information
Most molecular diagnosis approaches currently used in the routine
veterinary laboratory rely in the PCR and its variants, such as
nested-PCR and real-time PCR. The performance of these assays
mainly depends upon the primers used, which are usually a pair
of short oligonucleotide fragments complementary to flanking
regions of the target sequence and that will specifically anneal to
the microbial genetic material present in the sample. The laboratory
can design in-house PCRs based on previously well-characterized
and validated genomic targets and primers sequences available in
scientific publications and online databases. Otherwise, the laboratory staff would need to select novel targets and to design appropriate primers (free software can be downloaded and used for this
purpose). Ideally, the primers should be 100 % specific and, at the
same time, should detect all variants that are being hunted; this
may require a balance between the use of conserved and of highly
variable genetic regions as complementary targets for those primers. Depending on the primer design strategy, molecular tests can
allow the identification of the etiological agents further than the
species level, such as distinct groups (lineages) with a characteristic
geographic spread, for instance, the Mycobacterium bovis clonal
complexes [3] or the Brucella melitensis groups [4]. The possibility
22
Alicia Aranaz
23
24
Alicia Aranaz
25
26
Alicia Aranaz
27
28
Alicia Aranaz
Conclusion
The field of molecular diagnostics is rapidly evolving, with an
expanding number of techniques and assays. The advisability of
each technique depends on the purpose of use and the existing
limitations. The ability to quickly detect pathogens and their characteristics (e.g., subtypes, virulence factors, and drug-resistance
traits) would allow for precise decision-making and target measures such as prophylaxis, appropriate therapy, and biosafety plans
to promptly control disease outbreaks. New tools are needed for
diagnosis and surveillance of emergent or reemergent pathogens
that may arise from globalization of travels and trade, changes in
vector distribution, anthropological pressure on pristine areas, and
many other factors. Keeping abreast with this exponentially growing knowledge requires constant dedication. In this sense, to take
advantage of these possibilities, future veterinarians must have a
global understanding regarding the type of tests that can be applied
in the laboratory and the conclusions that can, or cannot, be drawn
from the results. In this sense, fundamentals of sequence analysis
should become an important item in the teaching program of veterinary medicine, including genes, methods, and databases that
should be accessed for sequence comparison [18]. This requirement for long-life learning is extensive to all veterinarians.
In practice, taking benefit of the huge amount of data that can
be obtained using molecular techniques highlights the need of collaboration between veterinarians in the laboratory and practitioners. The possibility to detect an extensive number of pathogens in
the laboratory does not preclude the need for adequate samples
collection and shipment, an adequate anamnesis, and a definition
of the objective of the diagnosis from the practitioner. The laboratory must provide an accurate and fast diagnosis and assistance in
the interpretation of results. The final diagnosis of the disease and
the decision to apply a treatment or a control program must be
made by the practitioner according to laboratory results but also to
other epidemiological factors.
29
9.
10.
11.
12.
13.
14.
15.
30
16.
17.
18.
19.
20.
21.
22.
Alicia Aranaz
Mycobacterium tuberculosis complex. Proc Natl
Acad Sci USA 99:36843689
Smith NH, Kremer K, Inwald J, Dale J, Driscoll
JR, Gordon SV, van Soolingen D, Hewinson
RG, Smith JM (2006) Ecotypes of the
Mycobacterium tuberculosis complex. J Theor
Biol 239:220225
Cottam EM, Wadsworth J, Shaw AE, Rowlands
RJ, Goatley L, Maan S, Maan NS, Mertens
PPC, Ebert K, Li Y, Ryan ED, Juleff N, Ferris
NP, Wilesmith JW, Haydon DT, King DP,
Paton DJ, Knowles NJ (2008) Transmission
pathways of foot-and-mouth disease virus in
the United Kingdom in 2007. PLoS Pathog
4:e1000050
de Lamballerie X (2009) Essential veterinary
education in modern molecular tools for the
detection of disease: what veterinarians will
need to know about genomics and molecular
biology and diagnostics (including bioterrorist
weapons) in 2025. Rev Sci Tech 28:657662
Hoffmann B, Scheuch M, Hper D, Jungblut
R, Holsteg M, Schirrmeier H, Eschbaumer M,
Goller KV, Wernike K, Fischer M, Breithaupt
A, Mettenleiter TC, Beer M (2012) Novel
orthobunyavirus in cattle, Europe, 2011.
Emerg Infect Dis 18:469472
Hoffmann B, Beer M, Reid SM, Mertens P,
Oura CA, van Rijn PA, Slomka MJ, Banks J,
Brown IH, Alexander DJ, King DP (2009) A
review of RT-PCR technologies used in veterinary virology and disease control: sensitive and
specific diagnosis of five livestock diseases notifiable to the World Organisation for Animal
Health. Vet Microbiol 139:123
Ojha S, Kostrzynska M (2008) Examination of
animal and zoonotic pathogens using microarrays. Vet Res 39:04
Yang S, Rothman RE (2004) PCR-based diagnostics for infectious diseases: uses, limitations,
23.
24.
25.
26.
27.
28.
29.
30.
Chapter 3
Biosafety Principles and Practices for the Veterinary
Diagnostic Laboratory
Joseph Kozlovac and Beverly Schmitt
Abstract
Good biosafety and biocontainment programs and practices are critical components of the successful
operation of any veterinary diagnostic laboratory. In this chapter we provide information and guidance on
critical biosafety management program elements, facility requirements, protective equipment, and procedures necessary to ensure that the laboratory worker and the environment are adequately protected in the
challenging work environment of the veterinary diagnostic laboratory in general and provide specific guidance for those laboratories employing molecular diagnostic techniques.
Key words Biosafety, Biocontainment, Laboratory biosecurity
Introduction
Veterinary diagnostic laboratories play a critical and fundamental
role in a countrys individual veterinary health and public health
systems but also act as a key component to regional and worldwide
veterinary health and public health systems. The field of biosafety
promotes safe laboratory practices, procedures, and proper use of
containment equipment and facilities; promotes responsible activities among laboratory workers; and provides advice on laboratory
design. Individuals working in veterinary diagnostic laboratories
have occupational exposure to a variety of hazardous materials in
the workplace. Diagnostic laboratories are demanding work environments where the pace is hectic, the work load is often heavy,
and the pressure to provide rapid, accurate, and reliable results is
ever present. Therefore it is of primary importance that institutions have a robust biosafety program in place which ensures that
laboratory staffs are well aware of the risks present in the laboratory but more importantly that they understand how to use the
protective equipment provided and practice those critical safety
behaviors which are based on internationally recognized, scientifically valid best practices.
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_3, Springer Science+Business Media New York 2015
31
32
Mission
Statement
Program
Assessments
Biosafety
Policy
Organizational
Vision
Program
Goals
Training and
Assistance
Biosafety
Manual
(SOPs)
Risk
Assessment
(IBC)
33
The biosafety program vision and mission for maximum effectiveness should be able to directly link to the institutions vision
and mission statements or goals. An institutions biosafety policy is
especially critical in that it defines the roles and responsibilities for
the program at all levels, as well as the methodology for defining
program goals and how individual accountability at all levels is
measured. It is important that the biosafety policy clearly states the
intent and direction of institutional leadership, and supporting
documentation must identify a chain of responsibility for information on biosafety-related issues. It is important to note that the
written biosafety/biocontainment policy is just another mechanism of communicating the policy to the employees. Laboratory
supervisors, principal investigators, biosafety and safety professionals, and other institutional leaders attitude toward biosafety
and what they do both as individuals and collectively, or fail to do,
is a more powerful expression of the actual biosafety policy and
culture than the actual written words of the policy. In other words,
a strong biosafety culture will only be created and maintained when
all levels of management demonstrate their commitment personally by embodying and rewarding culture-supportive behaviors and
conclusively addressing behaviors in others that undermine the
culture of safety.
Following clockwise on the diagram of Fig. 1, the fourth
through seventh balloon collectively represent universal employee
participation through hazard identification and assessment, hazard
prevention and control, information and training, and evaluation
of program effectiveness. Employee participation is critical to the
success of any biosafety program. Employees from all levels within
the organization need to be engaged and provided opportunities
to actively participate within the biosafety program, such as participating on various safety committees engaged in risk assessment
and policy development, participating in active discussions of biosafety topics at regularly scheduled laboratory meetings, inclusion
of lab staff on biosafety inspection and audit teams, and, finally,
ensuring that employees are involved in determining biosafetyrelated goals and metrics for their specific work areas. Organizations
need to ensure that employees are actively encouraged to report
unsafe conditions, accidents, and near misses, as well as making
34
35
Are there effective prophylaxes, treatments, or vaccines available (for animals and/or humans)?
Will the agent be introduced into species for which there are
no data?
What are the environmental stability, quantity, and concentration of the agent?
What is the host range of the agent and are there ongoing
surveillance testing programs?
Although risk assessment methodology can be either quantitative or qualitative, it is recognized that most life science institutions
36
37
Biocontainment Levels
Many institutions and organizations throughout the world must
consider the risks presented by proposed research with agricultural
pathogens and make decisions regarding the placement of these
pathogens into proper biocontainment and/or biosafety categories
[6]. Many countries and international organizations such as the
World Organization for Animal Health (OIE) have published
guidance to define biocontainment and/or biosafety levels, which
provide specific combinations of facility design features, engineering controls, work practices, and personal protective equipment
(or alternative barriers) for a range of assigned risks from low to
high. The majority of guidance documents recommend that the
selection of facility design features, protective equipment, and
administrative controls must be informed by a robust, site-specific
risk assessment (as earlier described) which takes into consideration
agent characteristics, type of work to be conducted, and training
and competency of staff conducting the work. The new OIE
Manual chapter Standard for managing biorisk in the veterinary
laboratory and animal facilities was adopted at the May, 2014
OIE General Session. It has not yet been combined with the old
Manual chapter on biosafety; that still is a stand-alone chapter
although efforts are being made to combine the two in the near
future. The new chapter draws heavily from concepts articulated in
the CEN Workshop Agreement (CWA) 15793 on Laboratory
Biorisk Management which was scheduled to expire in September
2014 but has been transferred to the International Standard
Organization (ISO) to be used as the primary source for an ISO
deliverable. The new chapter moves away from the traditional
approach of describing minimal recommended biocontainment
levels to a laboratory-specific risk control measures approach. The
authors are supportive of the concepts contained within CWA
15793 and firmly believe that biorisk management programs can
be a valuable tool in facilitating good biosafety and laboratory
38
39
40
6.2 Movement
of Extracted DNA/RNA
from High to Low
Containment
Laboratory Areas
6.3 Operational
Challenges
41
Conclusion
Implementing and maintaining appropriate biosafety measures is
one of the most important parts of veterinary diagnostic laboratory
operations. Robust and comprehensive risk assessments must be
conducted to determine the appropriate protection measures to be
utilized in the laboratory. The use of molecular techniques does
not preclude the application of adequate biosafety measures to
handle the biological risks of pre-extraction diagnostic samples, as
well as the general laboratory safety measures needed to address
chemical and physical hazards inherent in these procedures.
References
1. Roughton J, Mercurio J (2002) Developing an
effective safety culture: a leadership approach.
Butterworth-Heinemann, Oxford, UK
2. Gershon R, Karkashian C, Grosch J et al (2000)
Hospital safety climate and its relationship with
safe work practices and workplace exposure
incidents. Am J Infect Control 28:211221
3. American Veterinary Medical Association
(2008) One health: a new professional imperative. http://www.avma.org/onehealth. Accessed
23 Dec 2013
4. National Association of State Public Health
Veterinarians (2006) Compendium of veterinary standard precautions: zoonotic disease
prevention in veterinary personnel. http://
scav.org/Edit/uploads/compendium%20
of%20veterinar y%20standard%20precautions%202006.pdf. Accessed 23 Dec 2013
5. Siegel JD, Rhinehart E, Jackson M, Chiarello L
et al. (2007) Guideline for isolation precautions: preventing transmission of infectious
agents in healthcare settings. http://www.cdc.
gov/hicpac/pdf/isolation/isolation2007.pdf.
Accessed 23 Dec 2013
6. Heckert R, Kozlovac JP (2007) Biosafety levels
for animal agriculture pathogens. Appl Biosaf
12:168174
7. Kozlovac J, Thacker E, Ellis R (2012)
Introduction to biocontainment and biosafety
concepts as they relate to research with large
livestock and wildlife species. In: Richmond JY,
Jackman J (eds) Anthology of biosafety
XIII. Animal production and protectionchallenges, risks and best practices. American
Biological Safety Association, Mundelein, IL
8. Pauli U, Griot C, Thr B et al (2012) From the
field to the laboratory in an animal disease outbreak situation. In: Richmond JY, Jackman J
9.
10.
11.
12.
13.
14.
15.
16.
(eds) Anthology of biosafety XIII. Animal production and protection-challenges, risks and
best practices. American Biological Safety
Association, Mundelein, IL
Centers for Disease Control and Prevention
(2012) Guidelines for safe work practices in
human and animal medical diagnostic laboratories. MMWR 61(Suppl):1101
Centers for Disease Control and Prevention
(2011) Guidelines for biosafety laboratory
competency. MMWR 60(02):16
Centers for Disease Control (2009) Laboratory
facilities (secondary barriers). In: U.S. Health
and Human Services (ed) Biosafety in microbiological and biomedical laboratories, 5th
edn. p 3335
Office des Epizooties (2012) Biosafety and
biosecurity in the veterinary microbiology laboratory and animal facilities. In: Manual of
diagnostic tests and vaccines for terrestrial animals. 6th edn, Vol 1, Chapter 1.1.2, p 111
Blow JA, Dohm DJ, Negley DL et al (2004)
Virus inactivation by nucleic acid extraction
reagents. J Virol Methods 119:195198
Theriault S (2009). Advances in applied
research. Public Health Agency of Canada.
www.stcu.int/bb2009/download/download.
php?id=115. Accessed 23 Dec 2013
Crossley B, Hietala S, Shih L et al (2005)
High-throughput real-time RT-PCR assay to
detect the exotic Newcastle Disease Virus during the California 20022003 outbreak. J Vet
Diagn Invest 17:124132
Office des Epizooties (2012) Principles and
methods of validation of diagnostic assays for
infectious diseases. In: Manual of diagnostic
tests and vaccines for terrestrial animals. 6th
edn, Vol 1, Chapter 1.1.5, p 116
Chapter 4
Veterinary Biobank Facility: Development and Management
for Diagnostic and Research Purposes
Tina Lombardo, Silvia Dotti, Riccardo Villa, Stefano Cinotti,
and Maura Ferrari
Abstract
Biobanking is an essential tool for ensuring easy availability of high-quality biomaterial collections that
combine essential samples and epidemiological, clinical, and research data for the scientific community.
Specimen collection is an integral part of clinical research. Indeed, every year throughout the world, millions of biological samples are stored for diagnostics and research, but in many fields the lack of biological
material and models is a major hindrance for ongoing research. A biobank facility provides suitable samples
for large-scale screening studies and database repositories. Software dedicated to biological banks simplify
sample registration and identification, the cataloging of sample properties (type of sample/specimen, associated diseases and/or therapeutic protocols, environmental information, etc.), sample tracking, quality
assurance, and specimen availability characterized by well-defined features. Biobank facilities must adopt
good laboratory practices (GLPs) and a stringent quality control system and also comply with ethical
issues, when required.
The creation of a veterinary network can be useful under different aspects: the first one is related to
the importance of animal sciences itself to improve research and strategies in the different branches of the
veterinary area, and the second aspect is related to the possibility of data management harmonization to
improve scientific cooperation.
Key words Biobanking, Biological resources, Quality controls, Cryopreservation, Veterinary medicine
Introduction
The word biobank is a little over a decade old. It has been used
since 1996 [1] to describe collections of various types of biological
samples. Biobanks have been defined in a variety of different ways
and this has been a major challenge [2]. As reported by Hallmans
and Vaught, a biobank may be defined as the long-term storage of
biological samples for research or clinical purposes [3]. In addition
to storage facilities, a biobank can comprise a complete organization,
including biological samples, data, personnel, policies, and procedures, for handling specimens and performing other services, such
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_4, Springer Science+Business Media New York 2015
43
44
45
2.1
Sample Census
46
2.3 Quality
Management System
and Quality Controls
Currently, biobanks must follow internationally accepted guidelines and best practices issued by ISBER (International Society for
Biological and Environmental Repositories) [11] and BBMRI
(Biobanking and Biomedical Resources Research Infrastructure).
In particular, BBMRI aims to improve biobanking accessibility
and interoperability by harmonizing similar biobanks in different
2.2
47
48
This is one of the main points that must be monitored and controlled. In fact, it represents a potential risk in the whole storage
process. Starting from this point of view, monitoring temperature
within mechanical freezers and refrigerators is an important quality
assurance measure in addition to general building safety and security to protect against fire, unauthorized entrance, and other usual
hazards. Furthermore, all freezers must be protected by a real-time
temperature monitoring and alarm system. Alarm systems should
be in place to monitor the temperature conditions of mechanical
freezers or, for nitrogen freezers, the liquid nitrogen level (Fig. 1).
Furthermore, a well-designed biobank software system has been
developed to provide the possibility of monitoring the temperature
values with an alarm service. Such temperature logging information must be automatically transferred via electronic interface into
the biorepository management system and be linked with the
2.4
49
Fig. 1 Illustration of the real-time monitoring and alarm system of the IZSLER Veterinary Biobank facility
50
Blood Samples
The routine use and collection of blood samples for diagnosis has
provided general information on optimal methodologies and
potential pitfalls.
Although procedures for collecting, processing, storing, and
shipping blood components are generally standardized and well
documented, several important factors need to be considered.
An important early decision in blood collection is whether to
collect anticoagulated blood (consisting of plasma, buffy coat, and
red blood cells) or coagulated blood (consisting of serum and red
blood cell clot). Hemolysis of the specimen affects the accuracy of
laboratory tests, particularly chemical and serological tests; therefore, it should be prevented by employing careful handling techniques according to optimum needle size, proper handling of the
tubes, and proper pipetting techniques; however, if hemolysis is
observed (pink to red tinge in sample), this information should be
recorded [21]. In fact, hemolyzed samples would not be used for
proteomics analysis, but destroying them may be unnecessary as
any sample is worth saving, unless storage space is constrained.
Annotation of all pertinent information about the samples allows
the identification of potential factors that could influence outcomes. Serum and plasma specimens are of better quality for analysis if smaller-volume aliquots are initially prepared rather than
larger ones that have to be thawed, handled, and refrozen, perhaps
several times. Indeed, the ability to provide ready-to-use (RTU)
aliquots without additional handling steps facilitates the sharing of
samples and provides multiple replicates handled in an identical
manner. It is also important to consider new or alternative methods and reagents that may offer longer-term stability or increased
efficiency in the collection and preservation of blood [22, 23].
3.2
Cell Cultures
51
Chlamydiaceae
3.4 Field
and Immune Sera
52
3.5
Hybridomas
3.6 Microbiological
and Parasitological
Samples
3.7
Prototheca Algae
The identification of Prototheca algae (species and eventually subspecies) is currently carried out through microscopic examination,
growth in specific culture media, and molecular assays (HMRPCR, end point PCR, or sequencing).
3.8
Tissue Samples
53
Cell-associated viruses that can be grown in adherent or suspension cell cultures or chorioallantoic membranes of embryonated
hens eggs can be isolated from several types of samples. The main
principle of isolating viruses is to choose the most suitable cell line
and mechanically lyse infected cells and subsequently carry out several amplification passages to increase the titer in order to produce
the master sample and then the working samples. Virus batches are
tested for potential microbiological and viral adventitious contaminations; the tests are performed using microbiology, virology,
serology, and molecular biology methods. The primary sources of
potential viral contamination come from infected animal tissues
used to prepare biological reagents and media and during laboratory manipulation. Virus detection and identification can be made
by employing several methods, mainly based on serology tests
using monoclonal antibodies and standard and real-time PCR.
These assays amplify specific viral genome sequences known to be
characteristics of a virus with a nucleotide sequence available in
database collections. Extraneous viral contaminations can be verified through tests based on molecular biology techniques that
allow the detection of viral DNA and RNA of other viruses.
Mycoplasma contamination can be detected using real-time PCR
methods. Bacterial contamination is determined through inoculation of nonselective culture media. In addition, electron microscopy
is available for viral detection and identification using negative
staining methods. Such catchall methods (able to detect even
non-suspected/unknown viruses) benefit from Airfuge ultracentrifugation, increasing the sensitivity of the detection level.
Indeed, immune electron microscopy methods (IEM and IEM
gold) based on the use of hyperimmune sera and/or monoclonal
antibodies may help viral identification and classification.
54
55
56
57
58
59
Acknowledgments
The authors thank Dr. G. Bontempi, L.R. Scorrano, colleagues of
IZSLER, and the personnel of Biobank Infrastructure for practical
support and collaboration.
References
1. Loft S, Poulsen HE (1996) Cancer risk in oxidative DNA damage in man. J Mol Med 74:
297312
2. Kaye J (2006) Do we need a uniform regulatory system for biobanks across Europe? Eur J
Hum Genet 14:245248
3. Hallmans G, Vaught JB (2011) Best practices
for establishing a biobank. Methods Mol Biol
675:241260
4. OECD (2006) Creation and governance of
human genetic research databases. OECD
Publishing, 25 October 2006. ISBN-92-6402852-8
5. Yuille M, Van Ommen GJ, Brchot C et al
(2008) Biobanking for Europe. Brief Bioinform
9:1424
6. Prokosch HU, Beck A, Ganslandt T et al
(2010) IT infrastructure components for biobanking. Appl Clin Inf 1:419429
7. Carter A, Betsou F (2011) Quality assurance in
cancer biobanking. Biopreserv Biobank 9:
157163
8. Vgvari A, Welinder C, Lindberg H et al
(2011) Biobank resources for future patient
care: developments, principles and concepts.
J Clin Bioinforma 1:24
60
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
Chapter 5
Biological Specimen Collection and Processing
for Molecular Analysis
Susan Sanchez and Holly Sellers
Abstract
This chapter provides a standardized best practice approach to sample collection and handling for the
purpose of nucleic acid (NA) extraction and PCR. These methods are described through text and clearly
illustrated figures. Furthermore, for those cases in which transportation of samples cannot be accomplished due to the inability to maintain the cold chain or limitations on the import of live disease agents in
diagnostic samples, the chapter describes the use of FTA cards for collection transport and NA preparation
of samples for PCR. This chapter also makes emphasis in the best safety practices for handling of samples/
tissues to avoid unnecessary disease exposures to laboratory personnel. Finally, NA extraction best practices
and methods will be briefly outlined.
Key words Tissue, Swab, Culture, Virus, Bacteria, Fungi, Protozoa, Nucleic acid, Animals, Disease,
Organs, FTA card, Polymerase chain reaction, PCR, Typing, DNA extraction, RNA extraction
Introduction
Sample collection is the first and most important step for accurate
diagnostic testing. The sample must be appropriate for the test that
is going to be performed. In this chapter, the primary focus will be
on sampling and sample preparation for molecular analyses. The
molecular analysis will most commonly be a polymerase chain reaction (PCR)-based test. Because the pathogens affecting both farm
and companion animals are varied and include viral, bacterial,
protozoal, and fungal pathogens, the processing of the samples
outlined in this chapter will allow for both DNA and/or RNA
extraction. Therefore, nucleic acid extraction (NA) will be used as
an encompassing term that will be applicable to any pathogen.
PCR requires nucleic acid extraction from the diagnostic sample.
Understanding the location of the pathogen in the host during an
infection (this may vary during the course of the disease), as well as
the duration of the infection, will improve the success of detecting
the causative agent. It is also important to note that surveillance
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_5, Springer Science+Business Media New York 2015
61
62
Table 1
Examples of clinical signs and tissue samples that should be collected from chickens
Clinical signs
Hepatitis
Liver
Lameness
Respiratory
Dyspnea
Kidneys
Tumors
Immunosuppression
Skin lesions
Scab
Fowlpox
Neurological signs
Brains
Enteritis, malabsorption
Proventriculitis
Proventriculus
These samples will allow for accurate molecular testing of virus and bacteria
63
64
Materials
2.1.1 Manual
and Mechanical Grinder
Common Materials Needed
1. Small manual tissue grinder. Many manufacturers provide disposable grinders in sterile packages. Nevertheless, pestle and
mortar or other similar manual instruments can be used. The
advantage of the disposable is the fact that they have a plastic
sleeve that will protect the user from splashes enhancing safety
as well as minimize sample to sample contamination. See
Fig. 1b for an example of a disposable manual grinder.
65
Fig. 1 Solid tissue preparation for nucleic acid extraction (see Subheading 3 for further information)
66
Fig. 2 FTA tissues and tissues scraping sampling (see Subheading 3 for further information)
67
Fig. 3 FTA fluid and swab sampling (see Subheading 3 for further information)
68
Fig. 4 FTA punch processing for nucleic acid amplification (see Subheading 3 for further information)
69
Methods
1. Label a sterile bead tube with the sample identification number.
3.1 Tissues
Preparation (Fig. 1;
See Notes 1, 2, and 5)
2. Using a sterile scalpel cut a small piece (~1 cm3) of affected tissue and place in a weigh boat (Fig. 1b).
70
71
Fluids
1. Sanitize the Harris Uni-Core punch and mat with 70 % ethanol and allow to dry prior to use (Fig. 4b1, b2).
2. Place the mat under the FTA card circle to be punched
(Fig. 4c).
72
Notes
1. Safety considerations
(a) Always wear a clean laboratory outer garment when working with diagnostic samples.
(b) Wear protective glasses to avoid splashes to the eyes.
(c) Wear gloves when handling animal tissues.
(d) Fresh tissue manipulations should be carried out inside a
biosafety cabinet.
(e) Do not use the mechanical grinder if working with nonhuman primate tissues, non-captive marine mammals, and
tissues suspect of any Risk Group 3 agents. There is a small
risk that tubes may break.
2. Quality assurance
The standardization of procedures is critical for the credibility
of all diagnostic laboratories. Results should be repeatable; this
is hampered occasionally by the type and volume of the samples received. Therefore, standardizing the sample of tissue by
weighing, resuspending in standard volumes, and applying
standard amounts to FTA cards is critical for your molecular
diagnostic laboratory. Too much tissue or tissues that are not
properly homogenized can lead to false positives or negatives.
The protocols described here have been quality tested; this
means that multiple tissue samples have been serially tested to
determine the best procedure to yield positives and negatives
consistently. It is always good practice to check the quality of
73
74
75
Acknowledgments
Photography credits to Christopher B. Herron, Digital Media
Coordinator, College of Veterinary Medicine, The University of
Georgia, Athens, USA; art credits to Kip Carter, Chief of Medical
Illustration Services, College of Veterinary Medicine, The
University of Georgia, Athens, USA.
References
1. Vosloo W, Morris J, Davis A et al (2013)
Collection of oral fluids using cotton ropes as a
sampling method to detect foot-and-mouth
disease virus infection in pigs. Transbound
Emerg Dis. doi: 10.1111/tbed.12196
2. Prickett J, Simer R, Christopher-Hennings J
et al (2008) Detection of porcine reproductive
and respiratory syndrome virus infection in
porcine oral fluid samples: a longitudinal study
under experimental conditions. J Vet Diagn
Invest 20:156163
3. Committee on Foreign and Emerging Diseases
of the United States Animal Health Association
(2008) Foreign animal diseases, 7th edn.
USHAA, St. Joseph, MO
4. Dufour-Zavala L (ed) (2008) A laboratory
manual for the isolation, identification and
characterization of avian pathogens, 5th edn.
American Association of Avian Pathologists,
Madison, WI
5. Saif YM, Fadly AM, Glisson JR, McDougald
LR, Nolan L, Swayne DE (eds) (2008) Diseases
of poultry, 12th edn. Blackwell Publishing
Professional, Ames, IA
6. Greene CE (2012) Infectious diseases of the
dog and cat, 4th edn. Elsevier/Saunders, St.
Louis, Mo
7. Lo YMD, Chiu RWK, Chan KCA (2006)
Clinical applications of PCR, 2nd edn.
Humana, Totowa, NJ
8. Radostits OM, Done SH (2007) Veterinary
medicine: a textbook of the diseases of cattle,
sheep, pigs, goats, and horses, 10th edn.
Elsevier Saunders, New York, NY
9. Smith BP (2009) Large animal internal medicine, 4th edn. Mosby Elsevier, St. Louis, MO
10. Zimmerman JJ (2012) Diseases of swine, 10th
edn. Wiley-Blackwell, Chichester
11. Granato A, Giantin M, Ariani P et al (2014)
DNA and RNA isolation from canine oncologic
12.
13.
14.
15.
16.
17.
18.
19.
20.
Chapter 6
Validation of Molecular Diagnostic Assays and Quality
Assurance and Control in the Veterinary Laboratory
Nick Saunders and Ian Sharp
Abstract
This chapter describes the process of validating in-house molecular assays although the principles described
are equally relevant to all diagnostic assays. The best practice principles described below are based on the
In Vitro Diagnostic Medical Devices Directive (IVDD) and associated documentation. Although compliance with these regulations is not required for diagnostic reagents used on animals, the principles are
equally relevant to validation of all diagnostic assays, whatever their purpose.
Key words Validation, In-house, Molecular assays, Quality assurance, Quality control
Introduction
The introduction of new or modified diagnostic tests for veterinary
application is associated with significant risks, and a consistent
approach to assay validation and verification is therefore a requirement (Fig. 1).
In vitro diagnostics (IVDs) for human use come under statutory regulations in most jurisdictions (e.g., the US Food and Drug
Administration (FDA), the European Commission (EC)), and
although veterinary IVDs are generally free of these controls, there
is a responsibility, and usually a commercial necessity, to ensure
that tests are fit for purpose. Both in-house and commercial assays
(commercial kits used in modified form or for off-label applications
fall within the in-house status) need to be backed by the same level
of evidence supporting their performance against a specification.
Once an assay has been introduced, systematic quality assurance (QA) activities are essential to ensure that the assay continues
to perform according to the requirements of the specification and
fulfills its purpose.
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_6, Springer Science+Business Media New York 2015
77
78
Project planning
(panel, assay
definition,
materials
sourcing) and
commercial
analysis
Post-implementation
surveillance and
exploitation
Validation
core tasks
Review
meeting
Technical transfer
including training,
final SOP, reagent
manufacture and
quality control, batch
record collection,
verification
Followup/final
tasks
Data
analysis
&
technical
report
Panel
review
OR
Sign-off
79
80
Quality Assurance
Quality assurance is the process whereby the quality of laboratory
reports can be guaranteed and comprises all the different measures
taken to ensure the reliability of investigations. It is not limited to the
technical procedures performed in the laboratory. Therefore,
although procedures within the laboratory that ensure that the testing procedures (the analytical phase) are reliable are important, consideration must also be given to the pre- and post-analytical phases,
where the majority of the errors in the entire testing pathway occur.
The laboratory does not have control of many of the preanalytical steps, such as what and how specimens were taken, labeling
(of samples and request forms), and transportation. However, it
can influence these activities by providing guidance in the form of
a user manual. This provides details of those key factors related to
the specimen which are known to affect the performance of the
test or the interpretation of the results and instructions for transportation of samples, including any special handling needs. The
laboratory should also have a procedure which defines the criteria
for specimen rejection. It is good practice to notify the user concerning rejected specimens.
Many veterinary laboratories are accredited by the national
accreditation body to the international standard ISO 17025 [3].
Accreditation is a means of assessing the technical competence of
the laboratory and provides assurance to the laboratory and its customers that its service is fit for purpose.
The key elements of accreditation are competency of staff; use
of documented, validated procedures; appropriate management
and use of equipment and reagents; and a system of evaluation and
quality improvement, involving internal audit, recording, and
management of user complaints and nonconformities.
There is a much used saying among quality professionals, If
it isnt written down it didnt happen. Good record keeping is
essential, for audit purposes (internal and external), to aid identification of root causes when problems are identified (e.g., determining which reagents or instruments might be responsible for a poor
performing test) and evidence of due diligence if the quality of
testing is questioned by a customer.
81
Quality Control
Quality assurance of test methods will be provided by a combination of internal quality control (IQC), internal quality assessment
(IQA), and external quality assessment (EQA).
IQC is the analysis of material of known content in order to determine in real time if the procedures are performing within predetermined specifications. It is primarily the day-to-day monitoring of
reproducibility or precision designed to detect errors in any single
days analytical procedure. Performance of control material within
predefined limits is essential for technical validation of a diagnostic
test. The type of control used will depend on the type of assay. For
qualitative assays, controls may just consist of a positive or negative
sample. For quantitative assays, quality requirements of controls
need to be determined for high and/or low clinical decision limits,
depending on the analyte. A good understanding of the assay is
essential in order to ensure that the appropriate controls are used.
There is further discussion on the use of controls for polymerase
chain reaction (PCR) assays later in this chapter. A number of
papers have been published on the use and interpretation of controls in veterinary laboratories [4, 5].
Commercial assays will include assay controls but the laboratory should not rely on kit controls alone. Manufacturers adjust
controls from batch to batch to give consistent results although
assay sensitivity may vary between batches. This consistency is an
obvious aid to the user, but it does mean that if you only use the
controls that come with the kit, you will not detect any batch to
batch variation. Therefore, it is recommended that the laboratory also use independent internal quality control materials,
either purchased from a commercial source or prepared in-house.
82
83
External quality assessment is usually an externally organized function that monitors the efficacy of quality assurance procedures. It
compares the performance of different testing sites by allowing the
analysis of an identical specimen at many laboratories, followed by
comparison of individual results with those of other sites and with
the correct answer. EQA acts as a check on the efficacy of internal
quality control procedures.
EQA is performed on a limited number of samples, and the
process is inevitably retrospective, providing an assessment of performance rather than a true control for each test performed. It
gives participants an insight into their routine performance so that
they can take action to achieve improvements. EQA is an educational tool. To achieve the greatest benefit from EQA, samples
must be treated the same way as routine samples.
The diagram in Fig. 2 is a good illustration of how to interpret
QC results in terms of assay performance. The correct EQA result
84
could be the dart closest to the bulls eye in the first dartboard on
the left. Therefore, the correct EQA result does not show that the
laboratory is capable of getting consistent results or of getting the
correct result consistently. If IQA results are consistently the same
as the original test results, they show good precision as shown in
dart boards 2 and 3 but do not distinguish between these two
options, i.e., show good process control but not whether the assay
is good. If IQC results are consistently accurate (mean close to
expected value of the control and small SD values), then this shows
good precision and a good assay, as indicated by dartboard 3.
However, only the examination phase is assessed by IQC.
Clearly, EQA, IQA, and IQC all have their role to play. EQA
allows comparison with results obtained by all other laboratories
participating in the scheme. IQA shows whether or not the laboratory has the pre-examination, examination, and post-examination
activities in control, and IQC shows whether or not the actual
results are correct. By performing EQA, IQA, and IQC and
reviewing the data obtained, you get a powerful indication whether
or not you have a well-controlled process (and competent operator) and also a good assay.
4.4
Personnel
4.5
Equipment
All equipment used in the assay validation exercise must be maintained, serviced, calibrated, and monitored as appropriate to ensure
that it is suitable for use. This is essential to ensure that all conditions can be reproduced accurately during subsequent routine production of reagents and performance of the assay.
5.1 Establishment
of the Project
The drivers for introduction of new diagnostics are most commonly gaps in capability and capacity or opportunities for improved
service presented by new knowledge and technology. During the
planning phase, the aim is to produce a clear, agreed project plan.
A vital part of the process is to ensure that the project is properly
resourced.
5.2 Establishment
of a Review Team
85
86
87
Validation Design
The project leader should prepare the validation plan including the
following points that are based on the STARD initiative [8] and
external literature including MIQE [9]:
1. Define the purpose and objectives of the validation study. For
example, the study may be intended to validate the performance of a new assay or may aim to demonstrate that a significantly modified assay or protocol variant gives results within
the tolerance of the original.
2. Identify any training requirements to ensure everyone involved
in the validation has suitable levels of competency. Ensure training records are up to date for procedures being carried out.
3. Identify any risk assessments which need to be reviewed or
written.
4. Identify the available standards or reference materials. These
act as controls to allow the assay to be standardized, facilitate
assay comparison, and permit stability of the assay to be determined over time.
5. Identify the assay to be used for comparison with the assay
undergoing validation. This should be the currently accepted
gold standard where one is available.
6. Design an analytical validation study to test the sensitivity and
specificity of the assay using control materials, extracts from a
wide range of strains/variants of the target organism, specimens spiked with the target organisms, and a range of unrelated strains or species that could be present in a sample, but
that should not give a positive result.
7. Design a clinical validation study appropriate to the clinical
context (e.g., surveillance, screening, clinical diagnosis).
Choose the study group including species, case definitions,
inclusion/exclusion criteria, and study settings.
8. Identify the types (i.e., specimen, method of sampling, transport and processing) and numbers of samples to be tested.
Consider the need to include known positives, known negatives, low and high positives, and samples which are known or
likely to be problematic (e.g., containing inhibitors or possibly
cross-reactive markers).
9. Select appropriate statistical tools for determination of an appropriate sample size and to avoid bias. It is essential to consider
statistical requirements to ensure that results are statistically
significant. The sample size needed to ensure statistically significant results depends on a number of variables. Some guidance is
given in Tables 1 and 2. The validation design should avoid
discrepant analysis bias. Some samples may give discordant
88
Table 1
Relationship between sample size and 95 % confidence interval
Estimated test sensitivity (or specificity)b
Number of infected (noninfected)
subjects requireda
50 %
60 %
70 %
80 %
90 %
95 %
50
13.9 %
13.6 %
12.7 %
11.1 %
8.3 %
100
9.8 %
9.6 %
9.0 %
7.8 %
5.9 %
4.3 %
150
8.0 %
7.8 %
7.3 %
6.4 %
4.8 %
3.5 %
200
6.9 %
6.8 %
6.4 %
5.5 %
4.2 %
3.0 %
500
4.4 %
4.3 %
4.0 %
3.5 %
2.6 %
1.9 %
1,000
3.1 %
3.0 %
2.8 %
2.5 %
1.9 %
1.4 %
89
Table 2
Design parameters for validation experiments
Parameter
Design
Analytical sensitivity
Four different assay runs with at least three replicates per dilution of the sample
Diagnostic sensitivity Testing of samples (that have been tested using the gold-standard or
appropriate alternative assay) from cases with the defined clinical profile(s).
The minimum numbers of samples to be tested (to give a required level of
reliability of the sensitivity measurement) will depend on the prevalence of
disease and can be calculated from the minimum sensitivity levels and the
95 % CI shown in Table 1
Analytical specificity
Diagnostic specificity Testing of >50 samples that were positive and >50 samples that were negative
using the gold-standard assay
Efficiency
(quantitative
assays)
Linearity
(quantitative
assays)
Measurement range
Precision
(quantitative
assays)
Three samples (high, medium, and low positive) assayed at least four times or
more in one run and over at least four different runs on different days
Reproducibility
(quantitative
assays)
Three samples (high, medium, and low positive) assayed at least four times or
more in one run and in at least four different runs on different days. These
to be run in different laboratories or using different reagent batches or
different instruments
Analytical accuracy
(quantitative
assays)
Three analytical standards (high, medium, and low positive) assayed at least
four times or more in one run and over at least four different runs on
different days
Clinical accuracy
(quantitative
assays)
Three clinical standards (high, medium, and low positive) assayed at least four
times or more in one run and over at least four different runs on different
days
Reference intervals
Testing of >100 samples (that have been tested using the gold-standard assay)
from cases with the defined clinical profile(s)
Clinical validation
Analysis of samples from cases with the defined clinical profile(s) with
follow-up. This is on-going audit of assay performance
Shelf life
90
8.1 General
Principles
8.2
Controls
Table 3
Definitions
Reference test results (gold standard)
Positive
Negative
Positive
TP
FP
Negative
FN
TN
91
extraction
PCR set-up
sample
The analytical sensitivity of an assay is its ability to detect a low concentration of a given substance in a biological sample. This type of
sensitivity is expressed as a concentration (e.g., as copies, colonyforming units (cfu), plaque-forming units (pfu), or genome equivalents per ml or per g of sample material) in acceptable units. A lower
detectable concentration shows a greater analytical sensitivity.
92
ss RNA transcripts
plasmid clone
amoured RNA
cloned or natural
target in RNA phage
cloned or natural
target in DNA phage
93
8.5 Determination
of Diagnostic
Sensitivity (DSe)
and Diagnostic
Specificity (DSp)
94
95
Linearity
8.8 Measurement
Range
8.9
8.7
Precision
96
8.11 Clinical
Accuracy
8.12
The robustness of the assay may be evaluated by deliberately deviating aspects of the protocol that are perceived as being sensitive to
the outcome, for example, by simulating pipetting errors affecting
critical components, such as enzyme and Mg2+ or by extending the
storage of PCR reaction mix at room temperature or at 4 C before
amplification. Such data are likely to be useful for troubleshooting
if this is required.
Robustness
8.13 Reference
Intervals
The reference intervals, also referred to as the reference range, normal range, or reference limits, are the upper and lower levels of
analyte that you would expect to see in a normal population. Any
values above or below that range are outside normal limits.
Reference intervals are important when the purpose of the assay is
to determine whether the test subject or the sample is in or outside
the normal range. Generally, reference intervals are not relevant for
97
The presence of interferences may be identified during assay development and/or validation. When it is known that samples might
contain materials that potentially interfere with the test result, the
SOP should be modified, wherever possible, to introduce a suitable sample pretreatment. However, the complete removal of
interfering substances cannot be guaranteed, and it is therefore
essential to include appropriate positive controls to ensure that this
problem is detected.
8.15 Limitations
of the Method
8.16 Clinical
Validation
and Implementation
in Diagnostic
Pathways
While analytical validation is designed to show that the test chemistry reliably detects the target, diagnostic validation ensures that
the test detects the target in clinical specimens. The clinical sensitivity and specificity of a test refer to its ability to detect the disease
accurately. Clinical validation can be carried out retrospectively or
prospectively. Often it is useful to establish an additional postimplementation prospective evaluation to add data to the file.
The next step is to integrate the test into routine as part of a
clinical diagnostic pathway. Workflows are established to ensure
that the test is performed at an appropriate point in the diagnostic
cycle, that performance is triggered by specific clinical questions,
and that the results are interpreted in a way that feeds back into the
management of disease. The validation file should include information on the situations that would trigger use of the test and the
appropriate clinical interpretation of the results in relation to identification of the pathogen. Details of appropriate reporting comments in relation to the results in particular scenarios should be
provided. High and low positives in various samples and in different clinical contexts may have different implications. Thorough
clinical validation should provide insight into the relevance of the
test results.
98
The signal to noise ratio of the assay may decline gradually, and
this may be difficult to detect without rigorous application of
multi-rule run QC (i.e., Westgard [6]) since positive controls may
give acceptable results while the assay is performing suboptimally.
The principle of shelf-life studies is straightforward. Batches of
vials of the reagent are stored at the required temperature and samples are tested at different times using a known QC sample. The
European Standard Stability testing of in vitro diagnostic reagents
BS EN 13640 [11] recommends that shelf-life testing is performed
on samples from three batches.
While shelf-life studies determine the stability of the reagents
under specified conditions, reagents may be treated differently during their life. It is best practice to consider conditions that the
reagents might be subjected to and recreate them in the laboratory
to determine whether their performance is affected. For example, if
the reagents might be freeze-thawed several times during their life
aliquots should be subjected to freeze-thaw cycles and retested.
Samples may be tested after each cycle or collected and tested in a
single run. Where reagents are to be transferred between laboratories, they may, during transit, be stored overnight in a warm warehouse. It is recommended that easily envisaged scenarios such as this
are simulated in the laboratory to test the effect on the assay. It is
acceptable to perform stress studies on only one batch of a reagent.
If stress studies reveal that reagents are not stable under defined
conditions, then instructions for use must clearly state this. For
example, if reagents are found to be impaired following several
freeze-thaw cycles, the SOP should require that freeze-thawing is
limited to a number of cycles at which the level of impairment is
acceptable, e.g., instructions for reagents that need to be stored
frozen may need to dictate that the vial is discarded after first use.
Risk Assessment
To minimize the hazards to both users of the assay and to test subjects, a risk assessment should be performed prior to using any
reagent in a diagnostic test [12]. The risk assessment should consider the infection risk of test materials that contain biological substances and take into account all combinations for use, for example,
different platforms or ancillary reagents. The risks of an incorrect
diagnosis, either false positive or negative, should also be assessed.
Depending on what risks are identified, further action may be
required to ensure that appropriate control measures are implemented. These might include treatments to inactivate infectious
agents, a review of the scope of stress studies, a review of the
reagent storage conditions, the provision of additional information
in the instructions for use or revision of the guidance, or the interpretation of results or limitations of the assay.
10
99
11
12
13
100
14
Production of Reagents
Staff involved in the preparation of reagents must be suitably
trained and have documented evidence of competency.
Following the development of a satisfactory assay, it is essential that the procedures for production of reagents for routine use
are clearly defined to ensure that the assay continues to be fit for
purpose when further batches are made. SOPs that describe the
101
15
Rollout of Assays
The collection of diagnostic validation data will necessarily involve
limited technical rollout within the developing laboratory. Full
adoption of the new assay into clinical use will include consideration of the reagent supply chain and arrangements for quality
assessment, including participation in external quality assessment
(EQA) schemes wherever appropriate. If the test is required to
meet the needs of a range of end users, rollout should be considered. A rollout plan should include the timeline, equipment, personnel, and risk management proposals.
Following rollout, an assay verification study and continuing
quality assessment should be performed. This is usually best organized by the developing laboratory. Verification should involve the
testing of validation panels or be limited in scope to a few wellcharacterized samples and standards. In some circumstances, it
may be desirable that validation materials are sourced from laboratories other than that of the developer. Round-robins may be useful in ensuring continuing attention to quality.
16
102
Table 4
Assay validation checklist
Name of assay
Specific targets (pathogens and genes) of the assay
Project manager
Project leader
Staff performing the hands-on development work
Section: planning and inception
Subsection
Contributor
Deadline
Completion
Contributor
Deadline
Completion
103
Table 4
(continued)
Choice of study population
Choice of sample type
Sample size calculation
Performance of clinical validation
Data analysis and interpretation
Assessment of usability
Reagent stability studies
Further optimization if required
Production of documentation
Sign-off process
Section: rollout and verification
Subsection
Contributor
Deadline
Completion
Contributor
Deadline
Completion
17
Conclusions
With the advent of molecular biology, opportunities for development of new diagnostic assays have increased significantly.
Molecular assays are faster, more sensitive, and more specific than
104
Glossary of Terms
Fitness for purpose Fit for purpose means that the kit has to be
validated to such a level to show that the kits results can be interpreted
to have a defined meaning in terms of diagnosis or another biological property being examined (see www.oie.int/our-scientific-expertise/
certification-of-diagnostic-tests/the-register-of-diagnostic-tests).
In-house Any assay developed within the laboratory or commercial
assay which is modified or used off label, i.e., used in a different way,
for a different purpose or using a different sample type from that recommended by the manufacturer.
Validation Confirmation, through the provision of objective evidence,
that the requirements for a specific intended use or application have
been fulfilled [ISO 9000:2000]. Validation is normally performed by
the manufacturer (whether commercial or in-house).
Verification Confirmation, through the provision of objective evidence, that specified requirements have been fulfilled [ISO 9000:2000].
Verification is normally performed by the user prior to introduction of
a validated assay to determine if the assay achieves the required specification in their hands.
105
References
1. OIE (2013) Register of diagnostic kits certified
by the OIE as validated as fit for purpose.
http://www.oie.int/our-scientific-expertise/
certification-of-diagnostic-tests/the-registerof-diagnostic-tests. Accessed 30 December
2013
2. Working Party on Control of Medicines and
Inspections (2001) Annex 15 to the EU guide
to good manufacturing practice. http://ec.
europa.eu/health/files/eudralex/vol-4/pdfsen/v4an15_en.pdf. Accessed 30 December
2013
3. General requirements for the competence of
testing and calibration laboratories (BS EN
ISO/IEC 17025:2005)
4. Freeman KP, Gruenwaldt J (1999) Quality
control validation in veterinary laboratories.
Vet Clin Pathol 28:150155
5. Farr AJ, Freeman KP (2008) Quality control
validation, application of Sigma metrics, and
performance comparison between two biochemistry analyzers in a commercial veterinary
laboratory. J Vet Diagn Invest 20:536544
6. Westgard JO, Barry PL, Hunt MR, Groth T
(1981) A multi-rule Shewhart chart for quality
control in clinical chemistry. Clin Chem
27:493501
7. http://www.westgard.com/westgard-rules
8. Bossuyt PM, Reitsma JB, Bruns DE et al
(2003) Standards for Reporting of Diagnostic
Accuracy. Toward complete and accurate
reporting of studies of diagnostic accuracy: the
STARD initiative. Clin Chem 49:16
9. Bustin SA, Benes V, Garson JA et al (2009)
The MIQE guidelines: minimum information
for publication of quantitative real-time PCR
experiments. Clin Chem 55:611622
10. Burd EM (2010) Validation of laboratorydeveloped molecular assays for infectious diseases. Clin Microbiol Rev 23:550576
11. Stability testing of in vitro diagnostic reagents
(BS EN 13640. 2002)
12. Medical devices. Application of risk management to medical devices (BS EN ISO 14971.
2009)
13. Directive 98/79/EC of the European
Parliament and of the Council of 27 October
1998 on in vitro diagnostic medical devices
(1998-12-07 OJ No L 331/1). In vitro diagnostic medical devices. European Standards
1998
14. Medical devices. Quality management systems.
Requirements for regulatory purposes (BS EN
ISO 13485:2012)
Part II
Molecular Detection and Identification of Animal Pathogens
in Laboratorial Settings
Chapter 7
Molecular Approaches to Recognize Relevant
and Emerging Infectious Diseases in Animals
Fredrik Granberg, Oskar E. Karlsson, Mikael Leijon, Lihong Liu,
and Sndor Belk
Abstract
Since the introduction of the first molecular tests, there has been a continuous effort to develop new and
improved assays for rapid and efficient detection of infectious agents. This has been motivated by a need
for improved sensitivity as well as results that can be easily communicated. The experiences and knowledge
gained at the World Organisation for Animal Health (OIE) Collaborating Centre for Biotechnology-based
Diagnosis of Infectious Diseases in Veterinary Medicine, Uppsala, Sweden, will here be used to provide an
overview of the different molecular approaches that can be used to diagnose and identify relevant and
emerging infectious diseases in animals.
Key words Infectious diseases, Pathogen detection, Molecular diagnosis, Transboundary animal
diseases, Endemic diseases, Zoonoses, PCR, Isothermal amplification, Hybridization, Proximity
ligation assay (PLA), Microarrays, Nanotechnology
Introduction
The increased occurrence and emergence of devastating infectious diseases, in both domestic and wildlife animal populations,
are causing very serious socioeconomic losses at both global and
regional levels. This increase has been attributed to several contributing factors, the most prominent being the accelerated
movements of humans and animals due to increased globalization and international trade, the climatic changes, and the larger
and larger populations kept together in animal husbandry and
breeding. Some of these diseases, termed transboundary animal
diseases (TADs), such as foot-and-mouth disease and classical
swine fever, have a high capacity to spread very rapidly over countries and borders, having a devastating impact on animal productivity and trade, as well as causing other losses in the animal
husbandries and in wildlife. Other diseases, such as anthrax,
bovine tuberculosis, and rabies, have more endemic character,
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_7, Springer Science+Business Media New York 2015
109
110
111
PCR-Based Approaches
Molecular approaches become increasingly important in infectious
disease diagnostics and, with the exception of isolation by culturing, may supersede all other direct detection methods. The main
reasons are that a unique signature of every microorganism is
encoded in its genome, which in principle enables perfect specificity, and that various enzymatic mechanisms can be utilized to
manipulate and amplify the genetic material, yielding an exquisite
sensitivity of the molecular DNA-based assays. While bacteria have
their genome encoded in the form of DNA, some viral genomes
are composed of RNA, and an initial reverse transcription step is
therefore required before further manipulations and amplification
can be carried out. Enzymes typically utilized are polymerases,
reverse transcriptases, ligases, glycosylases, and nucleases. Of these,
the polymerases require a pair of sequence-specific primers, which
enables selective target amplification.
112
2.1
PCR Assays
2.2 Real-Time
PCR Assays
113
114
Isothermal Amplification
Isothermal amplification of nucleic acids is an alternative method to
PCR. The reaction is performed at a constant temperature in simple
devices, such as water baths or heating blocks, which eliminates the
need for high-end equipment and system maintenance. It can be
used to test for infections in regions where resources are limited and
logistic chains are impossible, but a rapid answer is needed.
Isothermal amplification normally takes about an hour or less to
complete, providing a fast specimen-to-result diagnosis at the point
of care (POC). To make the best use of isothermal amplification, a
system should ideally integrate the upstream sample preparation
and the downstream detection steps and be operated by personnel
without extensive training. Several platforms utilizing isothermal
technology are commercially available or close to market [21].
Recently, the field of isothermal amplification technologies has
advanced dramatically, resulting in several different amplification
systems. These have been summarized by Niemz et al. [21] and
include transcription-mediated amplification (TMA) [22], helicasedependent amplification system [23], loop-mediated isothermal
amplification (LAMP) [24], and rolling-circle amplification [25].
Of those methods, LAMP has gained the greatest interest because
of its high specificity, efficiency, and rapidity. By addition of a
reverse transcriptase in the reaction, RNA targets can also be amplified and detected by LAMP, which is referred to as RT-LAMP. The
LAMP utilizes four primers that bind to six distinct regions of the
target DNA to specifically amplify a short region and is catalyzed
by Bst DNA polymerase with strand-displacement activity [24].
Addition of loop primers may accelerate the reaction [26]. As of 8
February 2014, PubMed listed 990 publications with the search
term loop-mediated isothermal amplification. LAMP technology has been applied for the detection of viral pathogens such as
classical swine fever virus [27] and foot-and-mouth disease virus
(FMDV) [28], bacteria such as Clostridium difficile [29], and parasites such as malaria [30]. Commercial developments have progressed: a total of eight LAMP kits are approved in Japan for the
detection of SARS coronavirus, Mycobacterium tuberculosis (TB),
Mycoplasma pneumoniae, Legionella species, influenza A virus, H1
115
With the development of DNA macro- and microarray technologies, it became possible to detect and characterize a wide variety
of bacteria and viruses through simultaneous hybridization against
large numbers of DNA probes immobilized on a solid support
[35, 36].
The probes represent known sequences that may serve as
markers for identification and/or genotyping of bacterial strains,
resistance genes, viruses, etc. These are commonly arranged in an
ordered array of spots (or features), and hybridization with a
labeled target, i.e., the sample to be investigated, will therefore
result in a hybridization profile in which individual probe results
also can be assessed. As the names imply, the main difference
between macro- and microarrays is the number and size of spots
on the support. Macroarrays typically have larger and fewer spots
and have proven particularly effective for detecting smaller subsets
of genes, such as genes involved in antibiotic resistance [37].
Microarrays can contain thousands, and even up to many hundred
thousands, of spots with different oligonucleotide probes and have
116
4.3 Proximity
Ligation Assay
117
5.1 Microfluidic
Analysis Systems
118
119
120
121
Acknowledgments
This work was supported by Epi-SEQ, a research project under
the 2nd joint call for transnational research projects by EMIDA
ERA-NET (FP7 project nr. 219235), the European Union FP7
project RAPIDIA-FIELD (FP7-289364), the Formas Strong
Research Environments BioBridges project (No. 2011-1692),
the Award of Excellence (Excellensbidrag) provided to SB by the
Swedish University of Agricultural Sciences (SLU), and executed
in the framework of the EU-project AniBioThreat (Grant
Agreement: Home/2009/ISEC/AG/191) with the financial
support from the Prevention of and Fight against Crime
Programme of the European Union, European Commission,
Directorate General Home Affairs. This publication reflects the
views only of the authors, and the European Commission cannot
be held responsible for any use, which may be made of the information contained therein. The funders had no role in study
design, data collection and analysis, decision to publish, or preparation of the manuscript.
References
1. Cleaveland S, Laurenson MK, Taylor LH
(2001) Diseases of humans and their domestic
mammals: pathogen characteristics, host range
and the risk of emergence. Philos Trans R Soc
Lond B Biol Sci 356:991999
2. Taylor LH, Latham SM, Woolhouse ME
(2001) Risk factors for human disease emergence. Philos Trans R Soc Lond B Biol Sci 356:
983989
3. Saiki RK, Gelfand DH, Stoffel S et al (1988)
Primer-directed enzymatic amplification of
DNA with a thermostable DNA polymerase.
Science 239:487491
4. Gouvea V, Glass RI, Woods P et al (1990)
Polymerase chain reaction amplification and
typing of rotavirus nucleic acid from stool specimens. J Clin Microbiol 28:276282
5. Kemp DJ, Smith DB, Foote SJ et al (1989)
Colorimetric detection of specific DNA segments amplified by polymerase chain reactions.
Proc Natl Acad Sci U S A 86:24232427
6. Higuchi R, Fockler C, Dollinger G, Watson R
(1993) Kinetic PCR analysis: real-time monitoring of DNA amplification reactions. Bio/
Technology (N Y) 11:10261030
7. Leijon M, Mousavi-Jazi M, Kubista M (2006)
LightUp probes in clinical diagnostics. Mol
Aspects Med 27:160175
8. Zipper H, Brunner H, Bernhagen J, Vitzthum
F (2004) Investigations on DNA intercalation
9.
10.
11.
12.
13.
14.
122
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
123
124
Chapter 8
Real-Time Reverse Transcriptase PCR for the Detection
of Bluetongue Virus
Carrie Batten, Lorraine Frost, and Chris Oura
Abstract
In recent years, real-time reverse transcription polymerase chain reaction (rRT-PCR) has become one of
the most widely used methods for the diagnosis of infectious pathogens. The combined properties of high
sensitivity, specificity, and speed, along with a low contamination risk, have made real-time PCR technology a highly attractive alternative to more conventional diagnostic methods. Numerous robust rRT-PCR
systems have been developed and validated for important epizootic diseases of livestock, and in this chapter
we describe an rRT-PCR protocol for the detection of bluetongue virus. The assay uses oligonucleotide
primers to specifically amplify target regions of the viral genome and a dual-labeled fluorogenic (TaqMan)
probe which allows for the assay to be performed in a closed-tube format, thus minimizing the potential
for cross-contamination.
Key words Bluetongue virus, Molecular biology, Real-time reverse transcriptase PCR, TaqMan,
Diagnosis
Introduction
Techniques based on the amplification of specific nucleic acid
sequences by polymerase chain reaction (PCR) are highly sensitive
and specific. Real-time polymerase chain reaction (rPCR) is currently one of the most widely used methods in the field of molecular diagnostics and research. The rPCR technique avoids the use
of agarose gel electrophoresis, thus reducing the risks of contamination, and enables the amplification of nucleic acids and the
detection of the amplified products in real time. The technique
is fast and simple to perform and can be carried out in a 96-well
format, making it suitable for large-scale testing and epidemiological screening. Portable real-time PCR machines are now available, enabling the application of molecular technologies in the
field with prospects for radically changing diagnostic approaches
in the future.
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_8, Springer Science+Business Media New York 2015
125
126
127
Table 1
Primers and probe sequences
Oligo name
Sequence (53)
Primer Hofm_BTV_IVI_F2
20
Primer Hofm_BTV_IVI_R2
20
Probe Hofm_BTV_IVI_P
a
Materials
2.1 Reagents
and Consumables
2.2
Equipment
1. PCR cabinet.
2. Calibrated pipettes.
3. Freezer (5 to 30 C).
4. Refrigerator (18 C).
5. Ice bucket with ice.
6. Large capacity centrifuge.
7. Real-time PCR machine.
128
Table 2
Composition of the master mix for one-step real-time RT-PCR
Master mix composition
12.5
0.5
0.5
Probe Hofm_BTV_IVI_P (5 M)
1.0
RNase-free water
2.5
2.5 M ROX l
0.5
50 mM MgSO4
1.0
0.5
Total volume
19
The final concentration of the primers and probe per reaction is 0.4 M and 0.2 M,
respectively
a
Methods
3.1 Sample
Preparation
1. dsRNA from BTV test samples should be prepared as per standard methods (e.g., Qiagen/Roche viral nucleic acid extraction kits) and stored at 50 to 90 C. BTV negative and
positive control samples of known origin should also be prepared (see Note 2).
3.2
Assay Setup
2. Working in a clean laminar flow cabinet (see Note 4), add all
reagents to a suitable container (e.g., microcentrifuge tube)
allowing for the total volume of reagents (see Note 5). Mix the
reagents gently with a pipette. Once outside the PCR clean
room, maintain the master mix on ice and shielded from light.
3. In a class II safety cabinet, carefully aliquot 6 l of extracted
RNA for both samples and controls into an optical reaction
PCR plate following a planned layout (see Note 6). Alternatively,
use PCR strips/tubes that are suitable for a real-time PCR
machine.
4. Seal the PCR plate containing the aliquoted RNA with a PCR
adhesive film (or carefully close the caps on the PCR plate/
strips/tubes, hereinafter referred to as PCR plate), and heat
denature the RNA (see Note 7) for 5 min at 95 C using a
129
Table 3
Thermal cycling program
Step
Temperature
Duration
Number of cycles
Reverse transcription
48 C
30 min
95 C
2 min
PCR
95 C
15 s
45
56 C
30 s
72 C
30 s
Notes
1. The primer and probe set described target BTV genome segment 10 and are specific for BTV. This primer and probe set
will detect BTV serotypes 126 [5, 6]. Primers and probe
130
131
8. Placing the plate in a 96-well rack stabilizes the plate and makes
it easier to remove the seal. The bottom of the plate should be
free from moisture, as this can evaporate leaving calcium
deposits on the bottom of the plate during thermocycling.
Calcium fluoresces, and this may interfere with the fluorescence captured during amplification. The same applies when
using PCR strips/tubes.
9. A roller can be used to aid in sealing the PCR plate. Touching
the seal with gloves or fingers may leave marks which affect the
fluorescence detection.
10. A late Ct in a negative control with a sigmoidal character may
indicate cross-contamination.
11. Positive controls and positive samples should generate clean,
sigmoidal curves.
12. Raw data should be inspected if there are any samples/controls
generating a non-sigmoidal curve. This may indicate a problem with the computer selecting a baseline, inhibition from
inefficiencies during the extraction phase, or a fault with the
real-time PCR machine.
References
1. Gibson UEM, Heid CA, Williams PM (1996)
A novel method for real time quantitative RT
PCR. Genome Res 6:9951001
2. Heid CA, Stevens J, Livak KJ, Williams PM
(1996) Real time quantitative PCR. Genome
Res 6:986994
3. Livak KJ, Flood SJA, Marmaro J, Giusti W,
Deetz K (1995) Oligonucleotides with fluorescent dyes at opposite ends provide a quenched
probe system useful for detecting PCR product
and nucleic-acid hybridization. PCR Methods
Appl 4:357362
4. Hoffmann B, Beer M, Reid SM et al (2009) A
review of RT-PCR technologies used in veterinary
Chapter 9
Nested and Multiplex Real-Time PCR Using Dual-Labeled
Probes: Detecting and Discriminating Mycobacterium
tuberculosis Complex Members in Cultures and Animal
Tissues
Pedro Costa, Isabel Couto, Miguel Viveiros, and Joo Incio
Abstract
Members of the Mycobacterium tuberculosis complex (MTC) are causative agents of tuberculosis (TB) in
both humans and animals. In the last two decades, the accumulating knowledge of the nucleotide sequences
of several genes, and of the whole genomes, of MTC members has allowed the development of novel
molecular assays able to detect and discriminate between these species. However, despite the significant
advances in the development of molecular assays for detecting MTC members in human samples, only a
few assays have been described for detecting these agents in animal tissues. In this chapter we describe the
use of two TaqMan-based real-time PCR approaches, highly sensitive and specific and easy to perform, to
detect and identify veterinary-relevant MTC species in both animal tissue samples and cultures.
Key words Mycobacterium tuberculosis complex, Mycobacterium bovis, Bovine tuberculosis,
Real-time PCR
Introduction
Members of the Mycobacterium tuberculosis complex (MTC) are
causative agents of tuberculosis (TB) in both humans and animals.
This complex encompasses several closely related pathogenic species,
notably M. tuberculosis, the main agent of human TB, M. bovis, primarily linked to bovine TB but also often isolated from a wide range
of other domestic and wild animals, and M. caprae, mostly associated to caprine TB [13]. Other less frequently found MTC species
associated to TB disease in animals are M. pinnipedii, M. microti, M.
mungi, and M. orygis [410]. Finally, M. canetti and M. africanum
complement the known list of MTC species, being these last two
closely related with M. tuberculosis and almost always restricted to
human tuberculosis, in spite of some sporadic reports of infections
caused by M. tuberculosis also in domestic and wild animals [1, 8].
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_9, Springer Science+Business Media New York 2015
133
134
135
Materials
1. Animal tissue samples (e.g., lymph nodes, liver, spleen, or lung
tissue samples from TB-suspect animals such as bovine, wild
boar, or deer).
2. Reference strains (e.g., M. tuberculosis ATCC 25177; M. bovis
AN5; M. bovis BCG ATCC 27291) and clinical isolates of MTC.
3. Materials for homogenization and DNA extraction from tissues: pestle and mortar; zirconium beads; DNA extraction kit
(e.g., the QIAamp DNA Mini Kit, Qiagen).
4. Phosphate-buffered saline (PBS) buffer: 137 mM of NaCl,
2.7 mM of KCl, 8 mM of Na2HPO4, 1.5 mM of KH2PO4,
pH 7.2. Weigh all components into a glass beaker and dissolve
in 800 ml water. Adjust the pH to 7.2 with HCl and NaOH
solutions. Add distilled water to a total volume of 1 l, dispense
the solution into aliquots, and sterilize by autoclaving (20 min,
121 C). Store at room temperature.
5. TE buffer: 10 mM of TrisHCl, pH 8.0, 1 mM of EDTA.
6. TaqMan probes and primers (Table 1) (see Note 1).
7. Reagents for standard PCR amplification: Taq DNA polymerase and respective 10 reaction buffer; deoxynucleotide
triphosphates (dNTPs); MgCl2.
8. Reagents for real-time PCR amplification (e.g., the SsoFast
supermix, BioRad).
9. Homogenizer/bead shaker (e.g., the FastPrep FP120 Bio101,
Savant Instruments Inc., Holbrook, NY).
10. Centrifuge.
11. Standard and real-time PCR equipment (e.g., the CFX96 realtime PCR instrument, BioRad).
Methods
To prevent the risk of human infection, the manipulation of MTC
cultures and TB-suspect tissues must be performed in a confined
biosafety level 3 laboratory.
136
Table 1
Sequences of primers and probes
Primer/probe
Sequence (53)
Complementary target
F_Actin
R_Actin
b
P_Actin
F_IS6110
FN_IS6110
R_IS6110
P_IS6110d
F_16SrDNA
R_16SrDNA
P_16SrDNAf
F-esat6
R-esat6
P-esat6
F-Rv1510
R-Rv1510
GCGACGGTGCCAATCATC
P-Rv1510f
TET-CCATCGTACCCATCCGCT GCG-BHQ1
F-Rv2073c
AGTCGGTGTGCACGATGG
R-Rv2073c
P-Rv2073cg
137
138
139
Tissue sample
Bacteriological culture
1st
1st reaction
standard PCR
16S rDNA
IS6110
-actin +
IS6110
-actin +
IS6110 +
Inconclusive
Not infected
with MTC
Infected with
MTC
16S rDNA +
IS6110 +
Not
Mycobacterium
16S rDNA +
IS6110
Non-tuberculous
mycobacteria
2nd reaction
3400
RD1 +
RD4 +
RD9 +
RFU
2700
2000
1300
600
-100
11
16
21
26
Cycles (Ct)
31
36
41
46
RD1 +
RD4 +
RD9
M. tuberculosis M. caprae
M. canettii M. africanum
M. pinnipedii
M. orygis
RD1 +
RD4
RD9
RD1
RD4
RD9
M. bovis
M. bovis BCG
Fig. 1 Schematics of the detection and identification algorithm for MTC species most commonly associated
with TB in livestock and other animals. DNA directly extracted from tissues can be used as template for the
detection of MTC by nested real-time PCR targeting the IS6110 (left dashed box). The amplification of the
mammal -actin gene is used as control for the occurrence of inhibitors of the reactions. The inset in the bottom illustrates the amplification results usually obtained by the nested duplex real-time PCR assay of an MTCinfected (black lines) and non-infected (gray lines) tissues samples for the amplification targeting the IS6110
(solid lines) and the mammal -actin gene (dashed lines) (RFU-relative fluorescence units). DNA extracted
from cultures can be used as template for the identification of MTC by multiplex real-time PCR (right dashed
box). The DNA extracted directly from tissues can be also used as template for these multiplex assays, but the
sensitivity for the detection of MTC is lower than when using the nested real-time PCR assay. In the first amplification step, the isolate will be assigned as an MTC member (by detecting the presence of the IS6110 element) or, alternatively, as a non-MTC Mycobacterium species. The subsequent RDs-targeted triplex PCR allows
the identification of the most veterinary-relevant MTC members to the species level as M. bovis (or M. bovis
BCG), M. caprae, or M. tuberculosis, according to their distinct patterns of presence or absence of RD1, RD4,
and RD9. Other MTC members such as M. canettii, M. africanum, M. pinnipedii, and M. orygis may present
similar RDs profiles, but these species are rarely found in domestic (particularly livestock) and big game animals. Due to specific deletions spanning at least part of the region RD1, other less frequently found MTC species of M. microti and M. mungi present the alternative profile RD1 (), RD4 (+), and RD9 ()
140
Notes
1. Probes and primers are frequently delivered lyophilized and
need to be diluted with sterile water according to the manufacturers instructions. Stock solutions can be prepared at a
standard concentration of 100 pmol/l and stored at 20 C.
141
142
Acknowledgments
This work was partially funded by the project PTDC/CVT/
111634/2009 from Fundao para a Cincia e a Tecnologia.
References
1. Brosch R, Gordon SV, Marmiesse M et al
(2002) A new evolutionary scenario for the
Mycobacterium tuberculosis complex. Proc Natl
Acad Sci U S A 99:36843689
2. Mostowy S, Cousins D, Brinkman J, Aranaz A,
Behr MA (2002) Genomic deletions suggest a
phylogeny for the Mycobacterium tuberculosis
complex. J Infect Dis 186:7480
3. Michel AL, Mller B, van Helden PD (2010)
Mycobacterium bovis at the animal-human
interface: a problem, or not? Vet Microbiol
140:371381
4. van Soolingen D, de Haas PE, Haagsma J et al
(1994) Use of various genetic markers in differentiation of Mycobacterium bovis strains from
animals and humans and for studying epidemiology of bovine tuberculosis. J Clin Microbiol
32:24252433
5. Aranaz A, Cousins D, Mateos A, Dominguez L
(2003) Elevation of Mycobacterium tuberculosis
subsp. caprae Aranaz et al. 1999 to species rank
as Mycobacterium caprae comb. nov., sp. nov.
Int J Syst Evol Microbiol 53:17851789
6. Cousins DV, Bastida R, Cataldi A et al (2003)
Tuberculosis in seals caused by a novel member
of the Mycobacterium tuberculosis complex:
Mycobacterium pinnipedii sp. nov. Int J Syst
Evol Microbiol 53:13051314
7. Mostowy S, Cousins D, Behr MA (2004)
Genomic interrogation of the dassie bacillus
reveals it as a unique RD1 mutant within the
8.
9.
10.
11.
12.
13.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
143
Chapter 10
A Real-Time PCR Assay for the Diagnosis
of Gastrointestinal Nematode Infections
of Small Ruminants
Florian Roeber, Aaron R. Jex, and Robin B. Gasser
Abstract
The diagnosis of gastrointestinal nematode infections in small ruminants is central to studying the biology
and epidemiology of these parasites and underpins their control. Traditional methods of diagnosis are inaccurate, time-consuming and laborious. Here, we describe a step-by-step protocol for the molecular-based
diagnosis of infections by real-time PCR.
Key words Livestock parasites, Strongylid nematodes, Diagnosis, PCR, Real-time PCR
Introduction
Parasitic worms of livestock cause diseases of major socioeconomic
impact worldwide. The current financial losses caused by parasites
to agriculture have a substantial impact on farm profitability around
the world [1, 2]. Parasitic roundworms (nematodes) of livestock
animals are mainly controlled by treatment with antiparasitic drugs
(anthelmintics) [3]. Even with optimally timed, strategic treatments, this type of control is expensive. The excessive and uncontrolled use of anthelmintics has resulted in substantial and
widespread problems with resistance in nematodes against these
compounds [4, 5]. Given the significant problems with parasitic
diseases and drug resistance, there is an urgent need for an increased
focus on integrated control, which includes the use of diagnostic
methods. Unfortunately, traditional parasitological methods of
diagnosis are often unreliable and/or time-consuming to carry
out [6]. These limitations relate to their inability to accurately
identify parasite stages (eggs or larvae) from fecal samples to species and also to the time that it takes to allow eggs to develop to
larvae (via coproculture) for subsequent microscopic identification [7]. Recently, a DNA approach was developed to overcome
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_10, Springer Science+Business Media New York 2015
145
146
Fig. 1 Step-by-step protocol for the molecular diagnosis of strongylid nematode infections from small
ruminant fecal samples. Following coproscopy (1). the fecal suspension is washed and nematode parasite
eggs are recovered (2). Genomic DNA is isolated from eggs (3). and regions within the second internal transcribed spacer of nuclear ribosomal DNA specifically amplified by real-time PCR (4). After completion of PCR
and melting-curve analysis (5). an estimate of the contribution of individual nematode species or genera to the
egg count is estimated
147
Materials
2.3 For
the Amplification
of Nucleic Acids by
Real-Time PCR
Methods
3.1 Preparation
of Fecal Samples
and Enumeration
of Nematode Eggs
1. Collect feces from the rectum of sheep using a disposable plastic glove. Invert the glove to capture the sample, expel air, tie
a knot in the glove and label with a permanent marker (see
Note 5).
148
Table 1
Parasite-specific primers used in the real-time PCR assays
Melting temperaStart position
Amplicon
ture of target
in ITS-2 regionb length (bp)c sequence (C)
Target species
41
265
78.81
H. contortus
TEL
98
218
78.15
Te. circumcincta
TRI
40
267268
77.87
Trichostrongylus
spp.
CHO
162
82.12
C. ovina
OEV
105
79.38
O. venulosum
222
149
1. Transfer 250 l of the suspension to a PowerSoil DNA isolation tube and perform isolation of nucleic acids according to
the manufacturers instructions.
2. Use DNA samples immediately for molecular analysis or store
frozen (70 C).
Notes
1. It is critical to use a salt solution of this specific gravity to
achieve maximum flotation of nematode eggs.
2. We use the Rotor-Gene 3000 thermal cycler (Qiagen, USA)
and the respective software.
150
151
Acknowledgments
This research was supported through funds from the Australian
Research Council (RBG and ARJ). Other support from Melbourne
Water Corporation, the National Health and Medical Research
Council, and the Alexander von Humboldt Foundation is gratefully acknowledged (RBG). FR was the recipient of scholarships
from the University of Melbourne.
152
References
1. Perry BD, Randolph TF (1999) Improving
the assessment of the economic impact of parasitic diseases and of their control in production
animals. Vet Parasitol 84:145168
2. Sackett D, Holmes P (2006) Assessing the
economic cost of endemic disease on the
profitability of Australian beef cattle and
sheep producers. Meat and Livestock
Australia, Report No. ISBN: 1741910021,
Sydney, Australia
3. Hoste H, Torres-Acosta JF (2011) Non chemical control of helminths in ruminants: adapting solutions for changing worms in a changing
world. Vet Parasitol 180:144154
4. Wolstenholme AJ, Fairweather I, Prichard R,
Samson-Himmelstjerna GV, Sangster NC
(2004) Drug resistance in veterinary helminths. Trends Parasitol 20:469476
5. Kaplan RM (2004) Drug resistance in nematodes of veterinary importance: a status report.
Trends Parasitol 20:477481
6. Gasser RB, Bott NJ, Chilton NB, Hunt P,
Beveridge I (2008) Towards practical, DNAbased diagnostic methods for parasitic nematodes of livestock-bionomic and biotechnological implications. Biotechnol Adv
26:325334
7. Roeber F, Jex AR, Gasser RB (2013) Nextgeneration molecular-diagnostic tools for gastrointestinal nematodes of livestock, with
emphasis on small ruminants: a turning point?
Adv Parasitol 83:267333
8. Bott NJ, Campbell BE, Beveridge I, Chilton
NB, Rees D, Hunt PW et al (2009) A combined
9.
10.
11.
12.
13.
14.
Chapter 11
Improved Detection of Mycobacterium bovis in Bovine
Tissues Using Immunomagnetic Separation Approaches
Irene R. Grant and Linda D. Stewart
Abstract
Immunomagnetic separation (IMS) represents a simple but effective method of selectively capturing and
concentrating Mycobacterium bovis, the causative agent of bovine tuberculosis (bTB), from tissue samples.
It is a physical cell separation technique that does not impact cell viability, unlike traditional chemical
decontamination prior to culture. IMS is performed with paramagnetic beads coated with M. bovis-specific
antibody and peptide binders. Once captured by IMS, M. bovis cells can be detected by either PCR or
cultural detection methods. Increased detection rates of M. bovis, particularly from non-visibly lesioned
lymph node tissues from bTB reactor animals, have recently been reported when IMS-based methods were
employed.
Key words Mycobacterium bovis, Lymph node tissue, Immunomagnetic separation (IMS), Isolation,
Detection, IMS-PCR, IMS-culture
Introduction
Bovine tuberculosis (bTB), caused by Mycobacterium bovis, continues to be a significant animal health issue in many countries of the
world [1]. The single intradermal comparative cervical tuberculin
(SICCT) test, more commonly known as the tuberculin skin test,
is used in many countries to identify bTB reactor animals, which
are then compulsorily slaughtered. At the time of slaughter, meat
inspectors look for visible lesions typical of bTB in the lymph nodes
of the head and chest and in the lungs of these animals and excise
tissue samples to be sent to the bTB testing laboratory to allow confirmation of M. bovis infection status. Tissues are also taken from
bTB reactor animals displaying no visible lesions (NVL). Currently,
confirmation of diagnosis of bTB is reliant on successful isolation of
M. bovis by culture, but this approach can be problematic for a variety of reasons. There may be small numbers of mycobacteria present
in the selected tissues, at or below the detection limit of culture, so
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_11, Springer Science+Business Media New York 2015
153
154
false-negative culture results may be obtained. Chemical decontamination of the tissues (typically with oxalic acid) prior to culture on
solid and liquid media is known to significantly reduce the viability
of M. bovis in vitro [2], which may also lead to false-negative culture
results. The time to confirm the presence or absence of M. bovis in
clinical samples by culture can be up to 8 weeks, and any delay in
confirmation, as well as false-negative culture results for NVL tissues, has consequences for disease control programs [3].
Immunomagnetic separation (IMS) is a sample preparation
technique that aids the selective separation and concentration of
target bacterial cells from complex food and veterinary sample
matrices such as milk, feces, blood, and tissues [4]. It effectively
selects the desired bacterium out of the sample background microflora and sample components by use of microscopic paramagnetic
beads coated with antibodies (or other appropriate binders) specific for the bacterium of interest. Numbers of target bacterial cells
may also be concentrated from the test sample by resuspending
beads in a smaller volume after IMS. IMS has a longer history of
being employed for food testing [5] than in the veterinary diagnostic laboratory context, but it is becoming more widely used for
veterinary testing. Several IMS methods for M. bovis have been
published in recent years [69]. However, Stewart et al. [9]
attempted to optimize the IMS method by evaluating a range of
novel M. bovis binders (polyclonal and monoclonal antibodies, as
well as phage display-derived 12-mer M. bovis-specific peptide
binders) they had produced, in order to identify the best beadbinder combination to achieve maximal M. bovis cell capture
(Fig. 1). Dynabeads MyOne Tosylactivated dually coated with a
Fig. 1 Mycobacterium bovis cells captured by monoclonal antibody and biotinylated 12-mer peptide-coated Dynabeads MyOne Tosylactivateda large clump
of M. bovis cells and an individual cell are clearly seen attached to the beads. Bead
suspension after IMS was stained by auramine O fluorescent acid-fast stain
155
Materials
1. Purified mouse anti-Mycobacterium bovis IgM monoclonal
antibody (11G3) (see Note 1).
2. N-terminally biotinylated 12-mer peptide (EEA302) with
amino acid sequence NFRVSIDVVKSR (10 mg/ml in sterile
distilled water), custom synthesized by any peptide synthesis
company.
3. Dynabeads MyOne Tosylactivated (Life Technologies):
bead diameter 1.08 m, bead concentration 100 mg beads/
ml (approx. 1012 beads/g), surface area 8 m2/g (see Note 2).
4. Coating buffer: 0.1 M sodium borate buffer pH 9.5, prepared
by dissolving 6.183 g H3BO3 (MW 61.83) in 800 ml distilled
water, adjusting pH to 9.5 using 5 M NaOH and then adjusting volume to 1,000 ml with distilled water.
5. 3 M ammonium sulfate: prepared in coating buffer by dissolving 39.6 g (NH4)2SO4 (MW 132.1) in 0.1 M sodium borate
(pH 9.5), adjusting pH (if necessary) and then adjusting volume to 100 ml.
6. Phosphate buffered saline pH 7.4 (PBS): 0.01 M phosphate
buffered saline composed of 0.138 M sodium chloride and
0.0027 M potassium chloride, pH adjusted to 7.4.
7. Washing and storage buffer for beads (PBS/BSA/T20): phosphate buffered saline pH 7.4 (PBS) with 0.1 % (w/v) bovine
serum albumin and 0.05 % (w/v) Tween 20 added.
8. Wash buffer for IMS (PBS/T20): phosphate buffered saline
pH 7.4 (PBS) with 0.05 % (w/v) Tween 20 added.
9. Tris-EDTA buffer (pH 8.0): 10 mM TrisHCl and 1 mM
disodium EDTA, pH adjusted to 8.0.
10. Reagents for conventional PCR reactions (for post-IMS detection of M. bovis by PCR): DNA polymerase enzyme and
156
Methods
3.1 Coating
Dynabeads MyOne
Tosylactivated
with Phage DisplayDerived Peptide
and Monoclonal IgM
Antibody
MyOne
157
For health and safety reasons all procedures involving bovine tissues containing, or potentially containing, M. bovis must be carried
out in a class 1 biological safety cabinet located in a containment
level 3 laboratory facility.
1. Transfer approx. 3 g of cubed lymph node material to a sterile
mortar, add sterile sand, and grind thoroughly with pestle.
2. Add 4.5 ml of sterile phosphate buffered saline pH 7.4 (PBS)
and grind further (=1:1.5 dilution).
3. Transfer sample to centrifuge tube and centrifuge at 300 g for
3 min to sediment sand and tissue particulates.
4. Make a 1:10 dilution of clarified lymph node tissue supernatant in sterile PBS (see Note 5) and use 1 ml of this dilution
(=1:15 dilution of original lymph node sample) for immunomagnetic separation (IMS).
1. Add 10 l of dually coated (Mab 11G3 and biotinylated peptide EEA302) Dynabeads MyOne Tosylactivated to first
well of BeadRetriever tube strip, 1 ml of PBS/T20 buffer in
the next 2 wells, and an appropriate volume of buffer, depending on subsequent endpoint detection method (PCR or culture; see later), to 4th well.
2. Transfer 1 ml of the 1:10 dilution of the clarified lymph node
tissue supernatant to the 1st well of the BeadRetriever tube
strips. Fifteen samples can be processed at the same time.
3. Transfer tube rack containing samples to BeadRetriever
machine.
4. Pre-wet BeadRetriever tip strips (magnet covers) by dipping
each tip briefly in PBS/T20 and slide into place in
BeadRetriever machine (see Note 6).
5. Perform automated IMS using environmental program.
This preloaded IMS program consists of a 35-min incubation
period in well 1 of the bead retriever strip with mixing at a
medium speed throughout. The beads are then captured and
transferred to well 2 and washed for 1 min at a medium speed.
During the next stage, the beads are transferred to well 3 and
washed again for 1 min at medium speed. Finally, the beads are
transferred to well 4 (see Note 7) where they are released using
a 10-s high-speed mix into an appropriate buffer, depending
on which endpoint detection method is going to be applied
(see below).
158
3.4 M. bovis
Detection Options
Post-IMS
3.4.1 Touchdown IS6110
PCR (See Note 12)
159
Notes
1. The monoclonal antibody (11G3) was jointly produced by
Queens University Belfast and the Agri-Food and Biosciences
Institute for Northern Ireland [9]. Other anti-M. bovis polyclonal or monoclonal antibodies may be available commercially
or may have been generated in-house by other research groups.
Any such antibody could potentially be used to coat Dynabeads
MyOne Tosylactivated for IMS purposes; however, there is
no guarantee that capture of M. bovis will be as good as when
monoclonal antibody 11G3 is used in conjunction with the
12-mer peptide. An experiment to assess the detection sensitivity of such coated beads should be carried out, as described by
Stewart et al. [9], to verify good M. bovis capture capability
before adoption of an alternative IMS protocol.
2. Other types and sizes of surface-activated magnetic beads are
commercially available. If a different type of bead is employed,
then the bead coating protocol may differ, so follow the manufacturers instructions accordingly.
160
161
Acknowledgement
This work was supported by a grant from the UK Department of
Environment, Food and Rural Affairs (Project SE3262).
References
1. Office International des Epizooties (2011)
Chapter 2.4.7. Bovine tuberculosis. Manual of
diagnostic tests and vaccines for terrestrial animals 2009. https://www.oie.int/fileadmin/
Home/eng/Health_standards/tahm/2.04.07_
BOVINE_TB.pdf. Accessed 15 Aug 2013
2. Corner LA, Trajstman AC, Lund K (1995)
Determination of the optimum concentration
of decontaminants for the primary isolation of
Mycobacterium bovis. N Z Vet J 43:129133
3. de la Rua-Domenech R, Goodchild T,
Vordermeier M et al (2006) Ante mortem
diagnosis of Bovine Tuberculosis: the significance of unconfirmed test reactors. Gov Vet J
16:6571
4. Stevens KA, Jaykus LA (2004) Bacterial separation and concentration from complex sample
matrices: a review. Crit Rev Microbiol 30:724
5. Benoit PW, Donahue DW (2003) Methods for
rapid separation and concentration of bacteria
in food that bypass time-consuming cultural
enrichment. J Food Prot 66:19351948
6. Sweeney FP, Courtenay O, Ul-Hassan A et al
(2006)
Immunomagnetic
recovery
of
Mycobacterium bovis from naturally infected environmental samples. Lett Appl Microbiol 2:460462
7. Sweeney FP, Courtenay O, Hibberd V et al (2007)
Environmental monitoring of Mycobacterium
8.
9.
10.
11.
12.
Chapter 12
Detection of Fish Pathogens by Loop-Mediated Isothermal
Amplification (LAMP) Technique
Hatem Soliman, Mona Saleh, and Mansour El-Matbouli
Abstract
Rapid detection of fish pathogens is mandatory for applying the crucial preventive and control measures to
reduce fish losses and, consequently, minimize the economic impact of diseases on the fish farm owners. The
currently used molecular diagnostic tools of fish infectious agents, such as PCR and RT-PCR, are sensitive
and specific but still have some drawbacks. These tools are usually time consuming and laborious, need
skilled persons, and require sophisticated devices to be performed. Therefore, next-generation tools for
rapid diagnosis of fish infectious diseases were developed to conquer these shortages. One of these novel
tools is the loop-mediated isothermal amplification (LAMP) technique. LAMP is considered a more advantageous tool than PCR because it needs only a heating block or a thermostatically controlled water bath as
a source of constant temperature. It is considered to be more specific than the PCR assay as it uses 46
primers, which may diminish the occurrence of false-positive results. The time required for the amplification
process by LAMP is ranging from 30 min to 1 h comparing to 35 h in the case of PCR. The visual detection methods coupled with the LAMP assay eliminates the post-run processing for detection of the amplification products. Its sensitivity is either comparable with the PCR or better than it. A variety of LAMP
assays were developed for simple and rapid detection of a diversity of fish pathogens. Herein, we describe
how to perform a LAMP assay and troubleshoot any potential problem arising during the process.
Key words Diagnosis of fish infectious diseases, Bst polymerase enzyme, Visual detection, Lateral flow
strips, Loop primers, SYBR Green I stain, Fluorescent detection reagent (FDR), FITC-labeled DNA
probe, Biotin-labeled primer
Introduction
In the past centuries, diagnosis of fish infectious diseases was conventional and based on clinical signs, postmortem examination,
and isolation of the etiological agents, followed by phenotypic and
serological confirmation or histopathological investigations [1, 2].
However, these techniques have some drawbacks, such as lack of
specificity and inadequate sensitivity to detect pathogens present in
low numbers or in the absence of the disease clinical signs [3].
Therefore, a new generation of diagnostic techniques was in
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_12, Springer Science+Business Media New York 2015
163
164
165
processing time to 1530 min but also the cost of the sample preparation step as there is no need for nucleic acid extraction kits.
LAMP yields a large amount of DNA and a pyrophosphate byproduct, which enabled the development of simple visual detection
methods of the amplicons without costly specialized equipments
[17]. In addition to SYBR Green I, fluorescent detection reagent,
and cationic polymers, nucleic acid lateral flow assays have been
used for the visual detection of the LAMP products [14, 18].
LAMP and RT-LAMP have thus been used successfully for the
detection of a variety of fish pathogens. Herein, we describe how
can LAMP and RT-LAMP assays be performed correctly in order
to help the veterinarians to reach a rapid and accurate field diagnosis with reduced costs.
2
2.1
Materials
Primers
166
Table 1
Sequences of LAMP primers targeting relevant viral fish pathogens
Target virus
References
VHSa
[13]
[14]
[16, 18]
167
Methods
LAMP reaction mixtures should be prepared on ice. After thawing,
LAMP reagents and primers should be thoroughly mixed by vortexing and then spin-down and kept on ice until used. Carefully
follow all waste disposal regulations when disposing waste
materials.
3.1 Amplification
of DNA Templates
by LAMP
168
3.3 Detection
of LAMP Products
3.3.1 Using SYBR Green
I Nucleic Acid Stain
169
Fig. 1 Visual detection of LAMP products using SYBR Green I nucleic acid stain.
In tube A, the color of the reaction mixture changed to green, indicating the presence of the LAMP product (positive reaction). In tube B, the color of the reaction
mixture remained orange, indicating the absence of LAMP products (negative
reaction)
170
Fig. 3 Detection of LAMP products using nucleic acid lateral flow strips. The strip
labeled as pos is the positive control showing two purple bands (positive reaction). Strips 16 correspond to positive samples, demonstrating two purple
bands at both the control and test lines. Strips 79 correspond to negative samples showing only one purple band on the control line. Strip labeled as neg is
the no-template control (Color figure online)
Notes
1. Usually, four primers (F3, B3, FIP, and BIP) must be used to
perform the LAMP assay. No LAMP products will be produced in the absence of these primers. One or two additional
loop primers (FLP and BLP) may be used, depending on the
target sequence used to construct the primers. Sometimes the
target sequence does not enable the design of any loop primer
or will enable the design of one (forward or reverse) or two
loop primers. These primers, if present, will usually accelerate
171
172
173
References
1. Bernardet JF, Campbell AC, Buswell JA (1990)
Flexibacter maritimus is the agent of black
patch necrosis in Dover sole in Scotland. Dis
Aquat Org 8:233237
2. Pazos F, Santos Y, Macas AR et al (1996)
Evaluation of media for the successful culture
of Flexibacter maritimus. J Fish Dis 19:
193197
3. Ambrosia RE, De Wall DT (1990) Diagnosis
of parasitic disease. Rev Sci Tech 9:759778
4. Craw P, Balachandran W (2012) Isothermal
nucleic acid amplification technologies for
point-of-care diagnostics: a critical review. Lab
Chip 12:24692486
5. Gill P, Ghaemi A (2008) Nucleic acid isothermal
amplification technologies: a review. Nucleosides
Nucleotides Nucleic Acids 27:224243
6. Cunningham CO (2002) Molecular diagnosis
of fish and shellfish diseases: present status and
potential use in disease control. Aquaculture
206:1955
7. Walker P, Subasinghe R (2000) DNA-based
molecular diagnostic techniques: research
needs for standardization and validation of the
detection of aquatic animal pathogens and diseases. In: Report and proceedings of the joint
FAO/NACA/CSIRO/ACIAR/DFID expert
workshop, Bangkok, Thailand, 79 February
1999. FAO fisheries technical paper, no. 395.
FAO, Rome, pp 93
8. Parida M, Sannarangaiah S, Dash PK et al
(2008) Loop mediated isothermal amplification (LAMP): a new generation of innovative
gene amplification technique; perspectives in
clinical diagnosis of infectious diseases. Rev
Med Virol 18:407421
9. Tomita N, Mori Y, Kanda H et al (2008) Loopmediated isothermal amplification (LAMP) of
gene sequences and simple visual detection of
products. Nat Protoc 3:877882
Chapter 13
Direct Detection of Theileria annulata in Bovine
Blood Samples Using Standard and Isothermal
DNA Amplification Approaches
Jacinto Gomes and Joo Incio
Abstract
Tropical theileriosis is a tick-borne disease responsible for important health problems in cattle, caused by
the hemoprotozoan Theileria annulata. Traditionally, detection of Theileria pathogens in infected animals
requires the microscopic examination of stained-blood smears and serological methods. Molecular diagnostic
assays have been developed for the detection of Theileria parasites, including PCR-based and reverse line
blotting approaches, but these methods usually demand qualified personnel, complex instrumentation,
and expensive materials. Loop-mediated isothermal amplification (LAMP) can facilitate the design of
molecular assays independent of the use of sophisticated equipment. In this chapter we describe the application of two molecular assays for the direct detection of T. annulata in bovine blood samples, based in
real-time PCR and LAMP, both targeting the Tams1-encoding gene of this parasite.
Key words Theileria annulata, Theileriosis, Molecular diagnostics, Real-time PCR, Loop-mediated
isothermal amplification
Introduction
Tropical theileriosis is a tick-borne hemoprotozoan disease
responsible for important health problems in cattle (Bos taurus and
Bos indicus) and in Asian buffalo (Bubalus bubalis). The etiological
agent is the apicomplexan parasite Theileria annulata, which
occurs around the Mediterranean basin, Middle East, and Southern
Asia [13]. The animals that survive the acute disease become carriers of T. annulata piroplasms and play an important role as a
reservoir for the maintenance of the parasite life cycle [4].
Traditionally, detection of Theileria pathogens in infected animals
requires the microscopic examination of stained-blood smears,
which has low sensitivity and specificity. Serological methods are
also available, but cross-reactions are common, current infections
and previous exposures are not generally distinguished, and
antibodies tend to disappear in long-term animal carriers [2, 5].
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_13, Springer Science+Business Media New York 2015
175
176
Materials
1. Genomic DNA extracted from bovine blood samples to test
(see Note 1).
2. Positive control of amplification: DNA extracted from a
T. annulata positive sample (see Note 2).
3. PCR specific primers for T. annulata targeting the Tams1encoding gene: Tams1_forw (5-CAA ATT CGA GAC CTA
CTA CGA TG-3) and Tams1_rev (5-CCA CTT RTC GTC
CTT AAG CTC G-3) (see Notes 35) [9].
4. LAMP specific primers for T. annulata targeting the Tams1encoding gene: Tams1_F3 (5-CCG TTA ATG CTG CAA
ATG AGG-3), Tams1_B3 (5-CCA CTT ATC GTC CTT
AAG CTC G-3), Tams1_FIP (5-GCT TAA GTT TGA ATG
CCT KTA CTG GCC CTT AAG GTC GGA GAC AAG-3),
and Tams1_BIP (5-GAT GTT CAA GAA GAA GGG AGA
CAA GCC CTT GAA CAA GAC WTC ATC G-3) (see Notes
3, 6, and 7) (Fig. 1).
177
5
3
F3
F3c
F1c
F3 primer
FIP
F2
F2c
3
F1c
B1
B2
B3
B1c
B2c
B3c
Target
DNA
F3
F2
F1
B2
B1c
BIP
B3
B3 primer
Fig. 1 Generic location of the six segments in the target DNA used to design
LAMP primers. Forward (F3) and backward (B3) outer primers and forward (FIP)
and backward (BIP) inner primers are indicated. The specially designed FIP and
BIP primers contain two distinct sequences (F1c plus F2 and B1c plus B2,
respectively) corresponding to sense and antisense segments of the target DNA,
one for priming in the first stage and the other for self-priming in a subsequent
amplification reaction stage
Methods
3.1 Detection of
T. annulata with
Real-Time PCR
178
179
Notes
1. Blood samples may be collected from animals into sterile tubes
with EDTA and the total genomic DNA extracted using
commercially available kits or automated systems (e.g., we use
a BioSprint96 automated workstation and the BioSprint96
Blood kit, Qiagen) [9]. The average total DNA yield in samples is around 3540 ng/l. Extracted DNA should be stored
at 20 C until further use.
2. As positive controls of amplification, we may use DNA samples
extracted from T. annulata-infected macrophage cultures or
directly from bovine blood samples for which T. annulata
DNA was detected using other molecular tests such as reverse
line blotting [9].
3. Primers are frequently delivered lyophilized and need to be
diluted with sterile water according to the manufacturers
instructions. Stock solutions can be prepared at a standard
concentration of 100 pmol/l and stored at 20 C. Aliquots
of working solutions of each primer are prepared from stock
solutions in water and stored also at 20 C. Prepare small aliquots of working solutions (up to 100 l) and avoid continuous freeze and thaw of the solutions.
180
181
182
References
1. Gubbels JM, de Vos AP, van der Weide M et al
(1999) Simultaneous detection of bovine
Theileria and Babesia species by reverse line
blot hybridization. J Clin Microbiol
37:17821789
2. Garca-Sanmartn J, Nagore D, Garca-Prez
AL, Juste RA, Hurtado A (2006) Molecular
diagnosis of Theileria and Babesia species
infecting cattle in Northern Spain using
reverse line blot macroarrays. BMC Vet
Res 2:16
3. Gomes J, Soares R, Santos M et al (2013)
Detection of Theileria and Babesia infections
amongst asymptomatic cattle in Portugal. Tick
Tick Borne Dis 4:148151
4. DOliveira C, van der Weide M, Habela MA,
Jacquiet P, Jongejan F (1995) Detection of
Theileria annulata in blood samples of carrier
cattle by PCR. J Clin Microbiol 33:
26652669
5. Altay K, Aydin MF, Dumanli N, Aktas M
(2008) Molecular detection of Theileria and
Babesia infections in cattle. Vet Parasitol
158:295301
6. Criado-Fornelio A, Buling A, Asenzo G et al
(2009) Development of fluorogenic probebased PCR assays for the detection and quantification of bovine piroplasmids. Vet Parasitol
162:200206
7. Georges K, Loria GR, Riili S et al (2001)
Detection of haemoparasites in cattle by reverse
line blot hybridisation with a note on the distribution of ticks in Sicily. Vet Parasitol
99:273286
8. Bilgic HB, Karagen T, Shiels B et al (2010)
Evaluation of cytochrome b as a sensitive target
for PCR based detection of T. annulata carrier
animals. Vet Parasitol 174:341347
9. Santos M, Soares R, Costa P et al (2013)
Revisiting the Tams1-encoding gene as a
species-specific target for the molecular detection
of Theileria annulata in bovine blood samples.
Tick Tick Borne Dis 4:7277
10. Notomi T, Okayama H, Masubuchi H et al
(2000) Loop mediated isothermal amplification of DNA. Nucleic Acids Res 28:e63
11. Alhassan A, Thekisoe OM, Yokoyama N et al
(2007) Development of loop-mediated isothermal amplification (LAMP) method for
diagnosis of equine piroplasmosis. Vet Parasitol
143:155160
Chapter 14
Reverse Line Blot Hybridization with Species-Specific
Oligonucleotide Probes: Application to Piroplasm Detection
Ana Hurtado
Abstract
Reverse line blot (RLB) hybridization has become a well-established and widely used method for the
multiplex identification of several Babesia and Theileria species in hosts and tick vectors. The procedure is
based on the simultaneous PCR amplification of a polymorphic region of the 18S rRNA gene from different piroplasms followed by identification of the individual species by hybridization to species-specific
oligonucleotide probes covalently linked to a nylon membrane in a macroarray format.
Key words PCR, RLB hybridization, Babesia, Theileria, Piroplasms, Ticks, Tick-borne disease
Introduction
Piroplasmosis are worldwide-distributed diseases caused by tickborne intracellular apicomplexan parasites of the genera Theileria
and Babesia [1, 2]. These diseases are of serious health and economic concern in extensive or semi-extensive production systems
where livestock spend most of the year grazing in mountain pastures in contact with ticks. Specific and sensitive identification of
piroplasms in infected animals and ticks is crucial in disease investigations and epidemiological surveys.
Reverse line blot (RLB) allows the simultaneous detection and
identification of piroplasms in three steps: DNA purification (not
described here), simultaneous PCR amplification of a polymorphic
region of the 18S rRNA gene from the different piroplasm species,
and identification of the individual species by hybridization to
species-specific oligonucleotide probes bound to a nylon membrane. Simultaneous PCR amplification of the different Babesia
and Theileria species is achieved by using generic primers (one of
them biotinylated) that target conserved regions of the 18S rRNA
gene. Species-specific oligonucleotide probes designed within variable regions of the amplicons are covalently linked to the carboxyl
groups of a negatively charged nylon membrane through a C6
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_14, Springer Science+Business Media New York 2015
183
184
Ana Hurtado
90
Chemiluminescence
ECL
Probes
HRP
Streptavidin
Biotin
Probes
PCR product
PCR products
Probe
PCR products
Membrane
Fig. 1 Schematic representation of the RLB hybridization process. (a) Oligoprobes are covalently coupled to the
membrane in lines using a line-blotter apparatus; (b) the membrane is then rotated 90; (c) the PCR amplicons
are added perpendicularly to the probes; (d) amplicons come into contact with all the probes in a macroarray
format; (e) schematic representation of the hybridization and detection reactions; (f) example of the RLB
hybridization signal on an autoradiography film
185
2
2.1
Materials
Equipment
1. PCR thermocycler.
2. Miniblotter 45 line-blotter apparatus and foam cushions.
3. Hybridization oven, glass cylinders, and hybridization mesh
sheets.
4. Shaking water bath.
5. Centrifuge.
6. Vacuum pump.
7. Orbital shaker.
8. Exposure cassette.
9. Plastic container (e.g., Tupperware).
2.2
PCR
2.3
RLB
186
Ana Hurtado
Methods
187
Table 1
Sequence and concentration of oligonucleotide probes used for piroplasm species identification
by RLB
Target species
Sequence (53)a
Concentration
(M)
References
[7]
B. bigemina
[3]
B. bovis
[3]
B. caballi (genotype B)
32
[5]
B. caballi (genotype A)
[5]
[11]
[11]
B. crassa
[8]
B. divergens
[3]
B. gibsoni
32
[11]
B. major
[7]
B. microti
16
[11]
B. motasi
16
[4]
B. occultans
16
[17]
B. ovis
32
[4]
T. annae
[11]
T. annulata
[7]
T. buffeli
[3]
T. equi
10
[18]
T. equi (genotype A)
16
[5]
T. equi (genotype B)
[5]
T. lestoquardi
[8]
T. luwenshuni/OT1
[4]
T. ovis
[4]
32
[4]
32
[12]
Note: cross-reaction between the T. lestoquardi probe [8] and the amplicon generated from T. annulata and between
the T. annulata probe [7] and the amplicon generated from T. lestoquardi can occur since targeted sequences in both
species differ only in one nucleotide within the sequence of the probes
a
Oligonucleotide probes contain a N-(trifluoroacetamidohexyl-cyanoethyl-N,N-diisopropyl phosphoramidite [TFA])C6 amino linker at 5 end
188
Ana Hurtado
2. Cycling conditions: PCR conditions consist of an enzyme activation step of 4 min at 94 C and 40 cycles of 30 s at 94 C,
35 s at 51 C, and 35 s at 72 C.
3. Gel electrophoresis (optional): Run 5 l of the PCR product in a
1.5 % agarose gel to visualize bands of 385450 bp.
3.2 Covalent
Coupling
of Oligonucleotide
Probes
to the Membrane
3.3 RLB
Hybridization
with the PCR
Products
and Detection
189
5. Fill the slots with the denatured PCR products avoiding air
bubbles (see Note 12) and incubate at 42 C for 1 h in the prewarmed hybridization oven (do not shake).
6. Remove the PCR products from the slots by aspiration using
the vacuum pump before removing the membrane from the
miniblotter.
7. Cover the membrane (DNA side up) with the hybridization
mesh and gently roll them into the shape of a cylinder. Place
the covered membrane inside a hybridization glass cylinder.
8. Wash the membrane with 2 SSPE/0.5 % SDS into the glass
cylinder while rotating it in the hybridization oven at 52 C for
10 min (see Note 13).
9. Do a second wash.
10. Incubate the membrane with streptavidin-peroxidase in 2
SSPE/0.5 % SDS into the glass cylinder while rotating it in the
hybridization oven at 42 C for 30 min (see Note 14).
11. Wash the membrane with 2 SSPE/0.5 % SDS into the glass
cylinder while rotating it in the hybridization oven for 10 min
at 42 C twice.
12. Remove the membrane from the glass cylinder, place it in a
plastic container, and wash it with 2 SSPE for 5 min at RT
twice agitating the fluid gently using an orbital shaker.
13. Blot the edge of the membrane against filter paper to remove
excess liquid and add the mix of chemiluminescent substrate
as per manufacturers instructions. Incubate for 1 min
(see Note 15).
14. Wrap the membrane in plastic wrap and place it in the expose
cassette (see Note 16).
15. In the dark room place an autoradiography film on top of the
membrane (see Note 17) and expose it as desired (see Note 18).
16. Develop the film.
3.4 Stripping
of Hybridized
Amplicons
190
Ana Hurtado
Notes
1. Units of Taq polymerase can vary according to type and brand
used.
2. Extraction controls and PCR negative (water) controls are
included in each PCR run as negative controls.
3. Oligonucleotide
probes
are
synthesized
with
a
N -(trifluoroacetamidohexyl-cyanoethyl- N , N -diisopropylphosphoramidite [TFA])-C6 amino linker at the 5 end.
Resuspend lyophilized oligoprobes in water at a concentration
of 100 M upon arrival and store the stock solutions at 20 C.
4. Always prepare fresh EDAC before use and incubate the membrane for just 10 min, not more.
5. Fill slots 1 and 45 with Indian ink diluted 1/100 in 2 SSPE
for membrane orientation reference; the ink will indicate the
direction of probes in the membrane.
6. To avoid membrane drying never leave empty slots, fill with 2
SSPE all the slots that are not filled with probes or ink.
7. Incubations longer than 10 min will produce weaker signals.
8. Pre-warm the 2 SSPE/0.1 % SDS at 60 C in the oven or
water bath before using it for membrane inactivation.
9. Seal membrane in plastic wrap or keep it in a closed container
to avoid dehydration while stored.
10. Always include PCR negative and positive controls, the former
to control contamination and the latter to control the
process.
11. Quick spin the tubes after denaturing to pull down
condensation.
12. To avoid membrane drying, never leave empty slots; fill empty
slots with 2 SSPE/0.1 % SDS.
13. Make sure to set oven temperature in advance as required in
each step and keep solutions pre-warmed at the required
temperature.
14. When using ECL (Amersham) as chemiluminescent substrate,
mix 3.8 l streptavidin-peroxidase with 15 ml of 2 SSPE/0.5 %
SDS (1:4,000 dilution); if using SuperSignal West Dura
(Pierce), mix 1 l streptavidin-peroxidase with 75 ml of 2
SSPE/0.5 % SDS (1:75,000 dilution).
15. Warm both detection reagents to room temperature before
mixing them as per manufacturers instructions. Make sure the
membrane is well covered. This step does no need to be carried
out in the dark.
191
Acknowledgments
This work was supported by the Department of Agriculture of the
Basque Country Government, the Spanish National Institute for
Agricultural and Food Research and Technology (INIA), and the
European Regional Development Fund (ERDF).
References
1. Preston PM (2001) Theilerioses. In: Service
MW (ed) Encyclopedia of arthropodtransmitted infections of man and domesticated animals. CABI Publishing, Wallingford,
UK, pp 487502
2. Uilenberg G (2001) Babesiosis. In: Service
MW (ed) Encyclopedia of arthropodtransmitted infections of man and domesticated animals. CABI Publishing, Wallingford,
UK, pp 5360
3. Gubbels JM, de Vos AP, van der Weide M et al
(1999) Simultaneous detection of bovine
Theileria and Babesia species by reverse line blot
hybridization. J Clin Microbiol 37:17821789
4. Nagore D, Garca-Sanmartn J, Garca-Prez
AL et al (2004) Identification, genetic diversity
and prevalence of Theileria and Babesia species
in a sheep population from Northern Spain.
Int J Parasitol 34:10591067
5. Nagore D, Garca-Sanmartn J, Garca-Prez
AL et al (2004) Detection and identification of
equine Theileria and Babesia species by reverse
line blotting: epidemiological survey and phylogenetic analysis. Vet Parasitol 123:4154
192
Ana Hurtado
11. Garca-Sanmartn J, Barandika JF, GarcaPrez AL, Hurtado A (2008) Distribution and
molecular detection of Theileria and Babesia in
questing ticks from Northern Spain. Med Vet
Entomol 22:318325
12. Garca-Sanmartn J, Aurtenetxe O, Barral M
et al (2007) Molecular detection and characterization of piroplasms infecting cervids and
chamois in Northern Spain. Parasitology
134:391398
13. Kouam MK, Kantzoura V, Masuoka PM et al
(2010) Genetic diversity of equine piroplasms
in Greece with a note on speciation within
Theileria genotypes (T. equi and T. equi-like).
Infect Genet Evol 10:963968
14. Oura CA, Bishop RP, Wampande EM et al
(2004) Application of a reverse line blot assay
to the study of haemoparasites in cattle in
Uganda. Int J Parasitol 34:603613
Chapter 15
DNA Microarray-Based Detection of Multiple Pathogens:
Mycoplasma spp. and Chlamydia spp.
Christiane Schnee and Konrad Sachse
Abstract
Rapid detection of slow-growing or non-culturable microorganisms, such as Mycoplasma spp. and
Chlamydia spp., is still a challenge to diagnosticians in the veterinary field. In addition, as epidemiological
evidence on the frequency of mixed infections involving two and more bacterial species has been emerging,
detection methods allowing simultaneous identification of different pathogens are required. In the present
chapter, we describe DNA microarray-based procedures for the detection of 83 Mollicutes species
(Mycoplasma assay) and 11 Chlamydia spp. (Chlamydia assay). The assays are suitable for use in a routine
diagnostic environment, as well as in microbiological research.
Key words DNA microarray, Rapid detection, Mycoplasma spp., Chlamydia spp., Multiple-species
infection
Introduction
1.1 Mycoplasmas:
Etiological Importance
and Diagnosis
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_15, Springer Science+Business Media New York 2015
193
194
195
196
Rapid detection of bacterial pathogens remains a challenge to diagnosticians in veterinary medicine, particularly when slow-growing
or non-culturable microorganisms are involved, as is the case with
Mycoplasma spp. and Chlamydia spp. Early diagnosis of infection is
crucial for the effectiveness of control measures to contain the
spread of the infectious agent and prevent economic losses.
In the past two decades, the most significant advances in diagnostic technology were due to the broad application of nucleic acid
amplification techniques, notably the polymerase chain reaction
(PCR). Now that demands on quality, quantity, and rapidity of
microbiological diagnosis are steadily increasing, the inherent limitations of PCR are becoming evident. The parallelity of PCR assays
is very limited, i.e., usually a single agent is targeted, and even in
multiplex assays the number of targets is lower than 10. This is a
serious drawback as diagnosticians are increasingly becoming aware
of dual and multiple infections. Furthermore, as soon as one has to
deal with more subtle differences between species or strains, such
as minor sequence variations up to single-nucleotide polymorphisms or multi-locus sequence features, PCR alone cannot provide the necessary resolution.
In this situation, a DNA microarray-based approach, which is
capable of verifying the exact nucleotide sequence of a target
region through hybridization, appears a promising alternative.
Moreover, a DNA microarray assay can be designed to target a
large number of genomic loci (limited only by the size of the array)
to ensure discrimination between microbial species, strains, genotypes, serotypes, resistance types, etc.
The commercially available ArrayStrip (AS) platform represents an efficient system for processing low- and high-density DNA
arrays (Alere Technologies, http://alere-technologies.com/products). An AS unit consists of eight connected plastic vessels in
microtiter format, each one carrying a microarray chip of 3 3 mm
size with an active area of 2.4 2.4 mm on the bottom.
Hybridization and analysis can be conducted in an easy, rapid, and
parallel fashion, largely using standard laboratory equipment (see
Fig. 1 and Note 1).
Validated protocols for differentiation among 37 Mollicutes
species [10] and nine Chlamydia spp. [28] have been published by
our group. Other applications based on AS assays include DNA
serotyping of Salmonella enteric [29], genotyping of methicillinresistant Staphylococcus aureus [30], enterohemorrhagic Escherichia
coli [31], and many more.
In the present chapter, we describe the procedure for detection
of 83 Mollicutes species (Mycoplasma assay, see Table 1) and 11
Chlamydia spp. (Chlamydia assay). Basic principles of assay design
and experimental features are given in Notes 2 and 3.
Fig. 1 General workflow and timeline of the Mycoplasma and Chlamydia assays
Table 1
Selection of important pathogenic animal and human mycoplasmas and cell culture contaminants
that can be identified using the Mycoplasma microarray assay
Animal mycoplasmas
Human mycoplasmas
M. agalactiae
M. genitalium
A. laidlawii
M. bovigenitalium
M. hominis
M. arginini
M. bovis
M. pneumoniae
M. fermentans
M. californicum
U. parvum
M. hyorhinis
U. urealyticum
M. orale
M. conjunctivae
M. salivarium
M. dispar
M. felis
M. gallisepticum
M. hyopneumoniae
M. mycoides subsp. mycoidesa
M. ovipneumoniae
M. suis
M. synoviae
a
Members of the M. mycoides cluster are identified at species but not at subspecies level
198
Materials
2.1
DNA Extraction
2.2
Biotinylation PCR
Table 2
Primers for biotinylation PCR
Name
Sequence 53a
Target gene
Amplicon size
References
F1388
Mycoplasma
23S rDNA
594 bp
[10]
R1982
tuf-064F
Mycoplasma tuf
614 bp
[10]
tuf-681R
U23F-19
Chlamydia
23S rDNA
171 bp
[28]
23R-22
EGFP-11F
Internal
control
276 bp
[33]
EGFP-10R
2.3
Electrophoresis
199
2.5 General
Equipment and
Consumables
200
3
3.1
Methods
DNA Extraction
3.2 Pre-hybridization
Biotinylation PCR
3.2.1 Mycoplasma Assay
Table 3
Composition of the reaction mixtures for pre-hybridization real-time PCR in the Mycoplasma assay
Target
Components
Working
concentration
Final
concentration
23S rDNA
PCR-grade water
7 l
DyNAmoq PCR
Mastermix
10 l
Primer F1388
10 pmol/l
1 l
500 nM
Primer R1982
10 pmol/l
1 l
500 nM
DNA template
1 l
PCR-grade water
5.9 l
DyNAmoq PCR
Mastermix
10 l
Primer tuf-064F
10 pmol/l
1 l
500 nM
Primer tuf-681R
10 pmol/l
1 l
500 nM
10 pmol/
0.5 l
250 nM
10 pmol/l
0.5 l
250 nM
IC: pGEM-EGFP2reva
105 copies/l
0.1 l
104 copies/rxn
DNA template
1 l
tuf
IC internal control
201
Components
Working
concentration
Final concentration
PCR-grade water
11.1 l
PCR buffer
10
2 l
MgCl2
50 mM
0.6 l
1.5 mM
dNTP
2 mM each
2 l
200 nM
Primer U23F-19
10 pmol/l
1 l
500 nM
Primer 23R-22
10 pmol/l
1 l
500 nM
10 pmol/l
0.5 l
250 nM
10 pmol/l
0.5 l
250 nM
IC: pGEM-EGFP2reva
105 copies/l
0.1 l
104 copies/rxn
5 U/l
0.2 l
2U
DNA template
1 l
IC internal control
202
203
3.4.1 Manual
Identification
of Mollicutes Species
3.4.2 Manual
Identification
of Chlamydia spp.
204
Fig. 2 Examination of a cell culture sample using the Chlamydia assay. The species C. psittaci was identified.
The bar diagram reveals the following specific hybridization signals (from left to right ): family Chlamydiaceae
(two probes reacting), genus Chlamydia (Chlamydophila probes reacting), species C. psittaci (all four probes
reacting). Control reactions on the right-hand margin: background control for spotting buffer negative, internal
amplification/hybridization controls positive (three probes reacting), staining control positive. All other signals
represent cross-hybridization or are below cutoff. As the microarray carries three more sectors of spotted
Chlamydia probes, only the relevant parts are shown here
205
Notes
1. The AS technology is easy to handle as it mainly involves standard laboratory equipment and, unlike in more expensive
microarray platforms, no sophisticated devices, such as hybridization chamber and fluorescence reader, are needed. In a
positive reaction, oligonucleotide probes covalently linked to a
glass surface will form a duplex with complementary target
DNA strands carrying a biotin label. To visualize the hybridization signal, a streptavidin-horseradish peroxidase conjugate
is added and allowed to cleave a tetramethylbenzidine-like substrate. As a result, dark blue precipitates will be formed at each
reactive spot.
The expenses for consumables are moderate, particularly
when the high degree of parallelity is taken into account. This
means that, for instance, a single AS test for Chlamydia spp. is
equivalent to 11 different PCR tests for the individual species.
Detection of mycoplasmas or chlamydiae using the AS assay
can be accomplished within a working day (68 h). This technology is manageable in a high-throughput environment, with
up to 40 samples per day and technician.
2. For mycoplasma identification, the 23S ribosomal (r) RNA
and elongation factor Tu (tuf) gene loci were shown to be suitable target regions with a high discriminatory potential at species level but also with high intraspecific stability [10, 34]. The
necessary discriminatory capacity and robustness of the assay
have been attained through this combinatorial two-target
approach. The current version of the mycoplasma microarray
(no. 5) carries five genus-specific (Mycoplasma and Ureaplasma)
and 142 species-specific oligonucleotide probes derived from
the 23S rDNA target, as well as 157 species-specific probes
from the tuf site, thus enabling identification of 83 Mollicutes
species from animals, birds, man, and cell cultures. The major
selection criterion for the hybridization probes was the specificity of the target sequence, i.e., at least two nucleotides difference to the second-best matching target. In rare cases where
this criterion is not fulfilled, species assignment will be possible
at the alternative target or by a combinatorial approach via analysis of more complex hybridization patterns. The selected oligonucleotides are sized between 20 and 36 nt, have an average
206
207
7.
8.
9.
10.
denaturing gradient gel electrophoresis; a single generic test for detecting and differentiating Mycoplasma species. J Med Microbiol
54:731739
Rebelo AR, Parker L, Cai HY (2011) Use of
high-resolution melting curve analysis to identify Mycoplasma species commonly isolated
from ruminant, avian, and canine samples.
J Vet Diagn Invest 23:932936
Volokhov DV, George J, Liu SX et al (2006)
Sequencing of the intergenic 16S-23S rRNA
spacer (ITS) region of Mollicutes species and
their identification using microarray-based
assay and DNA sequencing. Appl Microbiol
Biotechnol 71:680698
Tonelli A, Sacchini F, Krasteva I et al (2012)
One test microbial diagnostic microarray for
identification of Mycoplasma mycoides subsp.
mycoides and other Mycoplasma species. Mol
Biotechnol 52:285299
Schnee C, Schulsse S, Hotzel H et al (2012) A
novel rapid DNA microarray assay enables
identification of 37 Mycoplasma species and
highlights multiple Mycoplasma infections.
PLoS One 7:e33237
208
11. OIE (2012) Avian chlamydiosis. World organisation for animal health: manual of diagnostic
tests and vaccines for terrestrial animals (chapter
2.3.1), vol. 1, 7th edn. Paris
12. OIE (2012) Enzootic abortion of ewes (ovine
chlamydiosis). World organisation for animal
health: manual of diagnostic tests and vaccines
for terrestrial animals (chapter 2.7.7), vol. 1,
7th edn. Paris
13. Sachse K, Vretou E, Livingstone M et al
(2009) Recent developments in the laboratory
diagnosis of chlamydial infections. Vet
Microbiol 135:221
14. Everett KD, Bush RM, Andersen AA (1999)
Emended
description
of
the
order
Chlamydiales, proposal of Parachlamydiaceae
fam. nov. and Simkaniaceae fam. nov., each
containing one monotypic genus, revised taxonomy of the family Chlamydiaceae, including
a new genus and five new species, and standards for the identification of organisms. Int J
Syst Bacteriol 49:415440
15. Stephens RS, Myers G, Eppinger M, Bavoil
PM (2009) Divergence without difference:
phylogenetics and taxonomy of Chlamydia
resolved. FEMS Immunol Med Microbiol
55:115119
16. Kuo C-C, Stephens RS, Bavoil PM, Kaltenboeck
B (2011) Genus Chlamydia Jones, Rake and
Stearns 1945, 55. In: Krieg NR, Staley JT,
Brown DR, Hedlund BP, Paster BJ, Ward NL,
Ludwig W, Whitman WB (eds) Bergeys manual of systematic bacteriology, vol 4, 2nd edn.
Springer, Heidelberg, pp 846865
17. Anonymous (2002) Avian chlamydiosis as a
zoonotic disease and risk reduction strategies.
SCAHAW (Scientific Committee on Animal
Health and Animal Welfare) European
Commission, Health & Consumer Protection
Directorate-General, SANCO/AH/R26/2002
18. Kaleta EF, Taday EM (2003) Avian host range
of Chlamydophila spp. based on isolation, antigen detection and serology. Avian Pathol
32:435461
19. Andersen AA, Vanrompay D (2000) Avian
chlamydiosis. Rev Sci Tech 19:396404
20. Pantchev A, Sting R, Bauerfeind R, Tyczka J,
Sachse K (2010) Detection of all Chlamydophila
and Chlamydia spp. of veterinary interest
using species-specific real-time PCR assays.
Comp Immunol Microbiol Infect Dis 33:
473484
21. Lenzko H, Moog U, Henning K et al (2011)
High frequency of chlamydial co-infections in
clinically healthy sheep flocks. BMC Vet Res
7:29
Chapter 16
In Situ Hybridization with Labeled Probes: Assessment
of African Swine Fever Virus in Formalin-Fixed
Paraffin-Embedded Tissues
Maria Ballester and Fernando Rodrguez
Abstract
In situ hybridization (ISH) has become a very valuable molecular diagnostic tool to detect specific DNA
or RNA sequences in biological samples through the use of complementary DNA- or RNA-labeled probes.
Here, we describe an optimized in situ hybridization protocol to detect African swine fever virus (ASFV)
DNA in formalin-fixed, paraffin-embedded tissues using digoxigenin-labeled probes.
Key words In situ hybridization, Nucleic acid, Formalin-fixed paraffin-embedded tissues, Digoxigenin,
Molecular diagnostic tool, African swine fever virus
Introduction
In situ hybridization is a method that allows the precise location
and/or quantification of specific DNA or RNA sequences in morphologically preserved chromosomes, tissues, or cell preparations.
The principle of the technique is based on the remarkable stability
of the double DNA helix through the hydrogen bonding of complementary strands, which can be broken (DNA denaturation or
melting) with heat or chemicals and rejoined (DNA hybridization)
under normal conditions. Thus, specific nucleotide-labeled probes
are annealed to complementary sequences and visualized using different detection systems depending on the type of label used to tag
the probe [1].
Since the first ISH experiment in the late 1960s [2], in which
the method was applied to localize DNA/RNA hybrids in cytological preparations, the method has undergone many improvements to become an important tool for both research and diagnostic
purposes.
Nowadays, there are a variety of ISH procedures with different
applications [35] although the main steps are basically the same.
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_16, Springer Science+Business Media New York 2015
209
210
Materials
Prepare all the stock solutions using ultrapure water- and analyticalgrade reagents and store them at room temperature (unless otherwise indicated). Please follow the lab security issues governed by
your national regulations on good laboratory practices, check how
to securely work with the chemicals described in this protocol, and
follow all waste disposal regulations diligently.
2.1
Probes
2.2
ISH
211
212
2.4 Special
Equipment
Methods
Carry out all the procedures at room temperature unless otherwise
specified.
3.1 Probe
Preparation
1. Digest total viral DNA, isolated from purified virions, with the
appropriate restriction enzyme following the manufacturer
instructions (see Note 11).
2. Purify the DNA after digestion using a DNA purification kit
and quantify the concentration of digested DNA (see Note 12).
3. Run an agarose gel to assure the complete digestion of viral
DNA by determining the length of the DNA fragments (Fig. 1)
(see Note 13).
4. Label the DNA fragments using a DNA labeling system,
following the instructions of the fabricant (see Note 14).
5. After DNA labeling, purify probes using a DNA purification
kit and quantify the concentration of the labeled DNA (see
Note 12).
6. Mix 300 ng of DNA-labeled probes (see Note 15) with 30 g
of competitor DNA and add water to obtain a precipitable
volume (minimum volume of 30 l).
Fig. 1 ASFV viral DNA digested with restriction enzyme. (a) Lane 1: DNA ladder.
Lane 2: two microliters (400 ng) of non-digested ASFV viral DNA. (b) Lane 1: DNA
ladder. Lane 2: ten microliters (763 ng) of MboI digested ASFV viral DNA
213
7. Precipitate the mixed DNA using a standard ethanol precipitation protocol (see Note 16).
8. Resuspend the DNA probes in 33 l of hybridization buffer
(see Note 17) and store at 20 C (see Note 18).
3.2 In Situ
Hybridization
214
Fig. 2 ASFV-DNA detection using DIG-labeled probes in formalin-fixed paraffin-embedded tissues. (a) Negative
lymph node. (bf) Positive tissues, where dark-blue dots in the preparation correspond to the presence of viral
nucleic acids (examples of positive labeling are indicated with arrows). (b) Spleen. Intense positive detection
in red pulp macrophages and trabecular medullary areas. (c) Lymph node. Moderate positive detection in
perifollicular macrophages (marginal zone). (d) Liver. Moderate to intense positive detection in sinusoidal and
alveolar macrophages and Kupffer cells. (e) Kidney. Intense positive detection in circulating macrophages and
renal endothelium. (f) Lung. Intense positive detection in pulmonary alveolar, vascular, and parenchymal
macrophages
Notes
1. ISH experiments can be performed with different types of
probes (DNA, cDNA, RNA, oligonucleotides, etc.). One of
the limitations to overcome during the ISH procedure is the
probe size. When using probes of dsDNA, the optimal probe
size is between 200 and 500 bp. In our case, we are using all
the ASFV viral genome (approx. 180 kb) and, for that reason,
we choose a restriction enzyme, MboI, with a recognition site
equal to 4 bases long (GATC) to cut ASFV genome very frequently. This enzyme generates medium ASFV-DNA fragment
lengths of 300500 bp (Fig. 1).
2. Any method or commercial kit which recovers small dsDNA
fragments can be used here. We use Qiagen Nucleotide
Removal Kit (Qiagen).
3. dsDNA probes prepared by random priming present higher
yield of labeled probe compared with nick-translation method.
We use DIG-High Prime system (Roche) for labeling dsDNA
probes.
215
216
217
Acknowledgment
This work has been fully supported by the AGL2010-22229-C03-01
Project from the Ministerio de Economa y Competitividad,
Spanish Government.
References
1. Jin L, Lloyd RV (1997) In situ hybridization:
methods and applications. J Clin Lab Anal
11:29
2. Gall JG, Padue ML (1969) Formation and
detection of RNA-DNA hybrid molecules in
cytological preparation. Proc Natl Acad Sci U S
A 63:378383
3. Ballester M, Rodrguez-Cario C, Prez M et al
(2011) Disruption of nuclear organization dur-
218
Chapter 17
Fluorescence InSitu Hybridization fortheTissue Detection
ofBacterial Pathogens Associated withPorcine Infections
HenrikElvangJensen, LouiseKruseJensen, KristianeBarington,
SusanneElisabethPors, ThomasBjarnsholt, andMetteBoye
Abstract
Fluorescence in situ hybridization (FISH) is an efficient technique for the identification of specific bacteria
in tissue of both experimental and spontaneous infections. The method detects specific sequences of
nucleic acids by hybridization of fluorescently labeled probes to complementary target sequences within
intact cells. FISH allows direct histological localization of the bacteria in the tissue and thereby a correlation between the infection and the histopathological changes present. This chapter presents protocols for
FISH identification of bacterial pathogens in fixed deparaffinized tissue samples mounted on glass slides.
Two different methods are presented: one is illustrated with the use of peptide nucleic acid (PNA) that is
carried out directly on glass slides (Method I), whereas the other is exemplified by using a DNA probe in
a Shandon rack (Method II). In the two methods, both PNA and DNA probes can be used.
Key words Bacteria, DNA, PNA, FISH, Fluorescence, Hybridization, In situ, Porcine, rRNA, Tissue
1 Introduction
Fluorescence in situ hybridization (FISH) has proven effective for
detecting specific species of bacteria in both experimental and
spontaneous infections in pigs (Table1). The method detects specific sequences of nucleic acids by hybridization of fluorescently
labeled oligonucleotide probes to complementary target sequences
within intact cells (Figs.1 and 2) [1]. FISH can be used for the
detection of DNA and RNA sequences [2]. For the identification
of bacteria by the FISH technique, ribosomal RNA (rRNA)-
targeting probes are most commonly applied because of the natural amplification of rRNA molecules [3]. Bacterial rRNA is present
in all cells and highly conserved between species, but still contains
variable sequences which serve as the target for identification at
species level. Bacterial 70S ribosomes consist of a 50S subunit and
a 30S subunit. The 50S subunit consists of two RNA molecules
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_17, Springer Science+Business Media New York 2015
219
GTCATTCCATCGAAACATA
CTCACGATGAACCTTCGAC
CCGCCTTACGGCAAACCT
GCTCATCGTGAAGCGAAA
TGGTTGAATGATGATGCC
TGTACTGGCTCACCTTTG
CTTTCCGCCTACACGCCC
TAGCTGATATCACATAGA
CTAACTTTCCTTTCCGCC
GTGTCTTCCACATTTCGT
L-G-Serp-1410-a-A-19
L-S-S.hyo-1210-a-A-19
L-S-B.murdoc-1219-a-A-18
S-S-S.pilo-0209-a-A-18
S-S-Cperfring-0185-a-A-18
S-S-Cl.diff-0193-a-A-18
S-O-Chls-0523-a-A-18
S-F-Cla-0574-a-A-18
S-G-Chla-0232-a-A-18
S-G-Chlph-0583-a-A-18
L-S-Efaecium-0347-a-A-18
Intestine
Intestine
Intestine
Intestine
Intestine
Intestine
Intestine
Intestine
Intestine
Intestine
Intestine
Genus Brachyspira
Brachyspira
hyodysenteriae
Brachyspira
murdochii
Brachyspira pilosicoli
Clostridium
perfringens
Clostridium difficile
Chlamydiales
Chlamydiaceae
Chlamydia
Chlamydophila
Enterococcus faecium
CCTCCGTATTACCGCAGC
CCCACCCTTTAATCCATAG
S-S-Ap-0185-a-A-19
Actinobacillus
pleuropneumoniae
Systematic namea
Tissue sample
Target species
[20]
[21]
[21]
[21]
[21]
[19]
[19, 20]
[1517]
[17]
[15, 17,
18]
[1517]
[1114]
References
Table 1
Panel of rRNA-targeting probes used for the detection of pathogenic bacteria by fluorescence in situ hybridization in porcine tissue samples
220
Henrik Elvang Jensen et al.
CCCAGTCCTCATGACCAG
S-G-Leptospira-1414-a-A-18
S-S-L.intrac-1148-a-A-20
S-S-M.hyop-0466-a-A-18
S-S-M.hyos-0466-a-A-18
S-S-M.hyor-0193-a-A-18
S-S-Pmul-0449-a-A-20
L-S-Sal-1713-a-A-18
S-S-S.suis-0183-a-A-18
S-S-Treponema-0833-a-A-18
Intestine
Intestine
Lung, heart
Lung, heart,
joints
Lung, heart
Lung, heart,
kidney
Intestine
Heart, brain
Intestine
Leptospira
interrogans
Lawsonia
intracellularis
Mycoplasma
hyopneumoniae
Mycoplasma
hyosynoviae
Mycoplasma
hyorhinis
Pasteurella
multocida
Salmonella spp.
Streptococcus suis
Treponema spp.
CCATGCGGTAAATACTGT
L-S-Kpneu-1705-a-A-18
Intestine
Klebsiella
pneumoniae
AATCACTTCACCTACGTG
CTATTTAACAACATCCCTTC
GCTGTGAAGCTCCTTTCT
CCGTCAGTTCAGTTGCAT
CCGTCAAGACTAGAGCAT
AACCGGAGCAGTCTCTCTAG
CGGGTGCTCCCCACTCAG
TACACACCAGCGTGCCTT
CACCGTAGTGCCTCGTCATCA
L-S-Eco-1531-a-A-21
Intestine
Escherichia coli
TCCCTCTTCCTATCGTTC
S-S-E.rhusiopathiae-0449-a-A-18
Heart
Erysipelothrix
rhusiopathiae
[24]
[22, 34]
[32, 33]
[30, 31]
[29]
[29]
[29]
[6, 2528]
[24]
[20]
[23]
[22]
Fig. 1 FISH using oligonucleotide probes. FISH probes can be classified as DNA or PNA probes depending on
the structure of the backbone. PNA probes are non-charged which allow a tighter and more specific binding to
RNA nucleic acid targets. DNA probes must overcome a destabilizing electrostatic repulsion during hybridization. The fluorescently labeled probes target RNA of the bacterial ribosomes (rRNA). The 16S and 23S rRNA
gene sequences contain hyper variable regions that can provide species-specific signature sequences useful
for bacterial identification, which most often is utilized in FISH. A adenine, T thymine (DNA specific), G guanine,
C cytosine, and U uracil (RNA specific)
223
Fig. 3 A bone with osteomyelitis is fixated in formalin and afterward embedded in paraffin. The block of paraffin
is cut with a microtome and the tissue is mounted on glass slides. It is necessary to remove the paraffin before
the application of the FISH probe. Following hybridization, it is possible to see the fluorescent bacteria within
the tissue using a microscope or a scanner equipped for fluorescence
(5S and 23S), while the 30S subunit consists of a single RNA
molecule (16S). The 16S rRNA is the preferred target for the identification of specific bacteria due to its combination of conserved
sites and variable regions [4]. The 23S rRNA subunit may be used
as well; however, as it has not been sequenced in a number of bacteria, most probes are targeting specific 16S rRNA.The FISH
technique is easy to apply and of a low cost [1]. When the probes
have been designed and evaluated, the FISH technique can be
completed in a few hours [5]. In addition, the technique allows the
direct localization of the bacteria in the tissue and thereby a histological correlation between the bacteria the tissue morphology and
the pathological changes present (Fig.2). Predominantly, viable
cells are detected as rRNA is fragile and rapidly disintegrates if the
cell has suffered irreversible damage [6]. Because the identification
of bacteria by FISH is dependent on the sequence of the probe, the
technique will not identify unknown species [4]. However, universal (pan-bacterial) probes are useful for the initial screening of sections for contents of bacteria [1].
The FISH technique consists of four steps: (1) fixation and
permeabilization of the sample, (2) hybridization, (3) removing
unbound probe, and (4) detection of labeled cells (Figs.2 and 3).
224
Fig. 4 (a) Direct labeling of the 3 tail of the probe. (b) Direct labeling of the 5 tail
of the probe. (c) Labeling of the probe with a reporter molecule which is detected
by a fluorescent antibody. (d) Labeling of the probe with horseradish peroxidase
(HRP) using a substrate for an enzymatic signal amplification
225
2 Materials
2.1 Fixation
ofTissue (Fig.3)
1. Tissue sample.
2. 10% neutral-buffered formalin, pH6.8 (formaldehyde,
3740%: 100mL/L; distilled water 900mL/L; sodium
phosphate, monobasic 4.0g/L; sodium phosphate, dibasic
(anhydrous) 6.5g/L).
3. 70%, 96%, and 99% ethanol.
4. Xylene.
226
5. Paraffin wax.
6. Bowl of water.
7. Plastic container with a tight-fitting lid.
8. Tissue cool plate.
9. Microtome.
10. Adhesive glass slides, e.g., SuperFrost Plus slides (Gerhard
Menzel GmbH, Braunschweig, Germany), poly-l-lysine-coated
glass (Ted Pella, Inc., Redding, CA, USA), or Biobond Tissue
Section Adhesive (AX-LAB, Copenhagen, Denmark).
2.2 Deparaffinization
(Fig.3)
2.4 Method I:
Hybridization
withPNA or DNA
Probes onTissue
Sections Mounted
onGlass Slides (Fig.3)
227
3 Methods
3.1 Fixation
ofTissue (Fig.3)
228
Fig. 6 Detection of bacteria with FISH in tissue mounted on glass slide using a
Shandon rack
3.2 Deparaffinization
(Fig.3)
229
rRNA at, e.g., GenScript USA Inc., Piscataway, NJ, USA; Tag
Copenhagen, Copenhagen, Denmark; Eurofins MWG Operon
AG, Ebersberg, Germany; and AdvanDx Inc., Woburn,
Massachusetts, USA (see Note 6). Although a probe seems specific
according to the software used, it is mandatory to have the specificity verified on a panel of well-characterized mono-bacterial tissue
infections. One limiting issue when designing probes for FISH is
that not all sites within the rRNA molecules are readily accessible
for hybridization [9].
The method should be repeated with EUB338 and non-EUB338
as positive and negative controls, respectively, in hybridization experiments [10].
1. PNA probes are typically received ready for use and are stored
at 4C (see Note 7).
2. DNA probes are stored at 20C or 80C (see Note 7).
If the probe is delivered as lyophilized, it should be dissolved
in nuclease-free sterile water, e.g., to a stock concentration of
500ng/L.
3. Thaw the probes and keep them on ice within a Styrofoam box
in the dark until preparing the probe solution. For each slide,
100L solution of probe should be used. Calculate with one
extra slide in each hybridization solution. Calculate the total
volume (Vtotal) of probe solution needed by using the following
formula (where nslide=number of slides):
230
m probe
C probe in stock
The mprobe is the mass of the probe needed in ng, while the Cprobe in
stock is the concentration of the probe in stock in ng/L.Calculate
the volume of hybridization buffer (Vhybridization buffer) by using the
following formula:
V hybridization buffer = V total - V probe in stock
The calculated volumes of hybridization buffer (Vhybridization buffer)
and probe in stock (Vprobe in stock) are mixed in a microcentrifuge
tube.
For example, preparing a probe solution for four slides and an
extra slide:
V total = 100 m L (1 + 4 ) = 500 m L
The concentration of the probe solution should be 5ng/L.If the
concentration of the probe solution in stock is 13.5ng/L:
2, 500 ng
= 185.2 m L
13.5ng / m L
231
1. Open the Shandon rack by lifting the top rack. Pour 23mL
deionized water in the bottom container (see Note 12).
2. Place the rack lid bottom up on the bench. Lay the cover plate
horizontally across the rack lid, front side up. Place a few drops
of washing buffer on the cover plate.
3. Place the tissue slide on the cover plate; tissue section must
face the front of the cover plate (see Note 13).
4. The tissue slide should be placed within the six stop notches on
the cover plate. A hybridization chamber of about 80L is
thereby formed between the cover plate and the slide.
5. Place the hybridization chamber upright in the Shandon rack
chamber slot so that the rectangular stop notch in the slot fits
exactly (clicks in place) into the rectangular hole in the middle of the spring clamp.
6. Apply 100L of probe solution to the hybridization chambers top well.
7. Place the lid on the Shandon rack, wrap it in tinfoil, and incubate overnight at 55C.Place the buffers in the incubator
together with the Shandon rack in order to equilibrate them to
the washing temperature of 55C (see Notes 1 and 9).
8. On the second day, remove the tinfoil and the lid from the rack
and place it back in the incubator. Apply warm (55C) hybridization buffer to the hybridization chambers top well
(see Notes 1 and 14). Let the buffer run through and repeat
the procedure two times. Repeat this with the washing buffer.
9. Fill a Coplin jar with MilliQ water, remove the slide from the
cover plate, and place it in the jar for 12min at room
temperature.
10. Remove the slides from the Coplin jar and place them in the
incubator 55C on a clean lab napkin or in an open slide box
until they air-dry (see Note 1).
11. Apply antifading mounting media and add a coverslip to each
slide. Gently press with a pencil to spread the antifading
mounting media under the slide and to remove bubbles.
232
4 Notes
1. The temperature of the hybridization buffer, washing buffer,
heating block, and incubator when using DNA probes depends
on the melting temperature (Tm) of the probe. This may vary
from 35C to 60C.Preliminary specificity tests should be
carried out at 10C below the Tm of the actual probe. PNA
probes are always run at 55C.
2. Formalin is carcinogenic; therefore, only handle it when wearing nitrile gloves and in a fume hood. For ensuring maximum
retention of nucleic acids, fixation should be carried out
promptly. Apart from formalin, which is a cross-linking fixative, a number of other fixation techniques may be used, e.g.,
freezing, formaldehyde based on paraformaldehyde, and Carnoys
fixative, but also acetic acid and ethanol can be used before
cryostat sectioning.
3. Paraffin-embedded tissue sections must not be folded when
mounted on a glass slide. Changing the temperature of the
water in the bowl can help unfold the tissue sections. For most
tissues, 47C is optimal, but5C may solve folding problems. When mounting brain and skin tissue, a lower temperature of approximately 37C is often required.
4. Slides with tissue sections treated through steps 1 to 3 in the
deparaffinization process can be removed directly from 99.9%
ethanol (deparaffinization step 3) and left to air-dry on a clean
paper napkin before hybridization.
5. In order to facilitate the penetration of the probe, pretreatment
is necessary for some Gram-positive species before the FISH procedure. Lysozyme is used on streptococci and on staphylococci
in FISH.This enzyme helps the oligonucleotide with the fluorescent dye molecule to enter the bacterial cell. Lysozyme pretreatment is done on the slide. We routinely use 3mg/ml lysozyme
solution in 100mM TrisHCL, 50mM EDTA, pH7.2, for
10min. Lysozyme treatment is done after the ethanol series.
6. When a probe has been designed in silico, its specificity should
be verified by looking up the sequence in other databases
afterward.
233
234
Chapter 18
Identification of Animal Pasteurellaceae
by MALDI-TOF Mass Spectrometry
Joachim Frey and Peter Kuhnert
Abstract
Species of the family Pasteurellaceae play an important role as primary or opportunistic, predominantly
respiratory, pathogens in domestic and wild animals. Some of them cause severe disease with high economic losses in commercial animal husbandry. Hence, rapid and accurate differentiation of Pasteurellaceae
is important and signifies a particular challenge to diagnostic laboratories. Identification and differentiation of Pasteurellaceae is mostly done using phenotypic tests or genetic identification based on sequence
similarity of housekeeping genes, such as the rrs gene encoding the 16S ribosomal RNA (16S rRNA). Both
approaches are time consuming, laborious, and costly, therefore often delaying the final diagnosis of disease or epidemics. Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry represents an alternative rapid and reliable method for the differentiation of most members of the
family Pasteurellaceae. It is able to differentiate within a few minutes the currently known 18 genera and
most of the over 60 species and subspecies of Pasteurellaceae including many members encountered in
veterinary diagnostic laboratories. A few closely related species and subspecies that cannot be discriminated
by MALDI-TOF are easily identified further by complementary simple tests, such as hemolysis done simultaneously or routinely during pathogen isolation.
Key words MALDI-TOF MS, Bacterial identification, Veterinary diagnostics, Molecular identification, Pasteurellaceae, New taxa, Rapid diagnosis
Introduction
The family Pasteurellaceae represents Gram-negative, aerobic, coccoid- or rod-shaped, nonspore-forming, and nonmotile bacteria,
currently consisting of 18 genera and more that 60 validly named
species (J.P. Euzeby, www.bacterio.net) [13]. While most taxa
appear as commensal bacteria on mucosal surfaces of animals, several species are of particular concern, either as persistent opportunistic or as primary pathogens in farm animals, such as Actinobacillus
pleuropneumoniae in pigs, Pasteurella multocida and Avibacterium
paragallinarum in poultry, Mannheimia haemolytica in cattle, or
Actinobacillus equuli subsp. haemolyticus in horses. Phenotypic
identification based on special growth media, detection of
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_18, Springer Science+Business Media New York 2015
235
236
237
Materials
Prepare all solutions using ultrapure water and analytical grade
reagents. Preparation and storage of reagents is at room temperature unless otherwise indicated.
238
2.1
Chemicals
2.2
Equipment
Methods
Procedures can be carried out at room temperature unless otherwise specified. They are given for the Bruker Daltonik Microflex
LT (Bruker Daltonik GmbH, Bremen, Germany), using the Flex
control software, and might vary slightly with other instruments.
3.1 Generation
of Reference Spectra
for the Establishment
of a New or
for Upgrading
a Database
239
6. Remove all liquid with a pipette and air-dry the bacterial pellet
for 2030 min. Suspend the pellet in 50 l of formic acid 70 %
and verify the full suspension of the bacteria.
7. Add 50 l of acetonitrile and mix well.
8. Centrifuge for 2 min in a micro-centrifuge at full speed.
9. Keep the supernatant for the production of MALDI-TOF
spectra.
10. Spot 1 l aliquots of the supernatant onto the steel target support of the MALDI-TOF mass spectrometer in eight replicas
(see Note 2).
11. Let the samples on steel target support air-dry (25 min).
12. Cover each dried sample with 1 l of -cyano-4hydroxycinnamic acid matrix solution (see Notes 2 and 3).
13. Let the samples on steel target support air-dry (25 min).
14. Include a calibration with a bacterial test standard (BTS),
which is, in general, a specific strain of Escherichia coli provided by the supplier of the MALDI-TOF instrument.
15. Produce reference spectra with the MALDI-TOF mass spectrometer by measuring each spot three times with standard
settings resulting in a total of 24 spectra for each strain (Fig. 1).
Run one to three BTS calibration spots with each plate read.
16. Import the spectra into the Flex analysis software and process it according to the manufacturers recommendations.
17. Check the quality of the 24 spectra, generate a main spectrum
(MSP), and register this as reference for the species in the
database of the MALDI-TOF mass spectrometer according to
the manufacturers instructions (see Notes 4 and 5).
18. Use the MSP (reference spectra) to compile a dendrogram as
shown in Fig. 2 using the Biotyper 3.0 software (Bruker) and
the correlation distance measure with the average linkage
algorithm.
3.2 Diagnostic
Identification
of Colonies by Direct
Transfer
240
1000
800
600
400
Distance Level
200
Fig. 2 Dendrogram derived from similarity matrices based on MALDI-TOF MS reference spectra from species
of the family Pasteurellaceae that are relevant in diagnostics of animal infectious diseases or represent important genera of this family. The distance level is normalized to a maximum value of 1,000. Type species of the
currently 18 genera are indicated in bold. Species misclassified in the corresponding genus are indicated
informally in brackets [11]
241
Fig. 3 Simple application of material from individual bacterial colonies onto the
steel target support for MALDI-TOF mass spectrometry
Notes
1. If using growth liquid media, removal of the media components from the bacterial samples may be important for successful MALDI-TOF analysis. Cell suspension may be centrifuged,
and the pellet washed several times with water before performing the analysis.
2. When spotting MALDI-TOF plates, allow the wicking action
to pull the sample or matrix solution aliquots off from the
pipette rather than touching the surface with the pipette tip, in
order to achieve a better homogeneity of the spot.
3. Samples are overlaid with a matrix solution and air-dried. The
crystallized organic acid forms the matrix, consisting of laserabsorbing small organic molecules in large excess over the bacterial proteins to be analyzed. Several organic acids are suitable
for the matrix solution, but cinnamic acid is adequate for
examining proteins (usually -cyano-4-hydroxycinnamic acid,
which enables highly sensitive MALDI-TOF measurement of
peptides and proteins from 0.7 to 20 kDa). The solvent acetonitrile leads to co-crystallization of matrix and sample molecules when evaporating.
4. Reference spectra databases can also be transferred from one
instrument to another, if the instruments are compatible.
However, best results are obtained with reference spectra generated on the proper instrument.
5. For certain bacterial species, reference spectra created on
the proper instrument might show differences compared to
242
243
7. Fenselau
C,
Demirev
PA
(2001)
Characterization of intact microorganisms by
MALDI mass spectrometry. Mass Spectrom
Rev 20:157171
8. Tamura H, Hotta Y, Sato H (2013) Novel
accurate bacterial discrimination by MALDITime-of-Flight MS based on ribosomal proteins coding in S10-Spc-Alpha operon at strain
level S10-GERMS. J Am Soc Mass Spectrom
24:11851193
9. Clark AE, Kaleta EJ, Arora A et al (2013)
Matrix-assisted laser desorption ionization-time
of flight mass spectrometry: a fundamental shift
in the routine practice of clinical microbiology.
Clin Microbiol Rev 26:547603
10. Kuhnert P, Bisgaard M, Korczak BM et al
(2012) Identification of animal Pasteurellaceae
by MALDI-TOF mass spectrometry. J
Microbiol Methods 89:17
11. Schaller A, Djordjevic SP, Eamens GJ et al
(2001) Identification and detection of
Actinobacillus pleuropneumoniae by PCR based
on the gene ApxIVA. Vet Microbiol 79:4762
12. Christensen H, Bisgaard M (2008) Taxonomy
and biodiversity of members of Pasteurellaceae.
In: Kuhnert P, Christensen H (eds)
Pasteurellaceae: biology, genomics and molecular aspects. Caister Academic Press, Norfolk,
UK, pp 126
13. Kuhnert P, Berthoud H, Straub R et al (2003)
Host cell specific activity of RTX toxins from
haemolytic
Actinobacillus
equuli
and
Actinobacillus suis. Vet Microbiol 92:161167
Chapter 19
Gold Nanoparticles as a Potential Tool
for Diagnosis of Fish Diseases
Mona Saleh, Hatem Soliman, and Mansour El-Matbouli
Abstract
Infectious diseases are a serious problem and a major contributor to severe economic losses in intensive fish
culture. Therefore, rapid and sensitive detection of fish pathogens is extremely important. Although various assays for determination of fish pathogens have been developed, most of these diagnostic methods are
time-consuming and laborious. To overcome these limitations, functional nanomaterials have been actively
investigated to improve detection ability and rapidity of diagnostic assays. Gold nanoparticles (AuNPs)
have been widely studied for their unique optical properties arising from their surface plasmon resonance,
which is responsible for their large absorption and scattering properties. These unique properties are four
to five orders of magnitude larger than those of conventional dyes and can be controlled by varying their
sizes, shapes, and compositions. Moreover, AuNPs can be easily synthesized and functionalized with different biomolecules, including pathogen-specific oligonucleotides or antibodies. Recently, nanoparticlebased assays have been introduced as a tool for laboratory diagnosis. They have been used for the direct
detection of unamplified nucleic acids in hybridization assays. Single- and double-stranded oligonucleotides can be adsorbed on AuNPs in colloidal solution under certain conditions. The result of the hybridization process can be visually detected within 1 min after addition of AuNPs, when the color of the
reaction mixture changes from red to blue (positive reaction) or remains red (negative). The development
of such nanoparticle-based strategies holds the potential to become powerful approaches for diagnosis of
fish pathogens.
Key words Diagnosis, Fish pathogens, Gold nanoparticles, Label-free, Colorimetric detection,
Unamplified nucleic acids, Nucleic acid probes, Hybridization assay
Introduction
Methods conventionally used for the diagnosis of fish diseases
mostly require expensive laboratory facilities, preventing their
wide-scale use. Additionally, these methods need extensive sample
preparation steps and have long readout times, which delay a timely
response and hamper effective disease control [1]. Recently, a
powerful emerging technology based on the unique properties of
nanoscale materials was introduced, which presents a great
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_19, Springer Science+Business Media New York 2015
245
246
247
Complementary RNA
Probe
RNA
Denaturation
Annealing
Addition
of
AuNPs
Fig. 1 Schematic diagram of a colorimetric assay based on unmodified AuNPs for detection of unamplified
nucleic acids (SVCV RNA). First, the target RNA is denatured and the complementary probe hybridizes to the
target forming double strands. Adding AuNPs causes its aggregation since the probe is not free to stabilize the
AuNPs, and the solution color changes from red to blue. In the presence of a noncomplementary target RNA,
the probe will be free to adsorb onto and stabilize the AuNPs consequently preventing their aggregation and
the solution color remains red
248
Materials
2.1 Gold
Nanoparticles
2.2 Oligonucleotide
Probes
1. Oligonucleotide probes targeting specific sequences of a particular gene of interest of the pathogen. For example, we have
recently described the development of a 26 bp specific probe
(5-GTC TAT CAT CAG CTA CAT CGC ATT CC-3)
designed to target the spring viremia of carp virus (SVCV) glycoprotein gene [29]. Assess the specificity of the probe against
other common aquatic pathogens sequences deposited in
GenBank.
2. Stock solutions of the probes: order the designed probes commercially and prepare them according to the supplied specification sheet that usually gives instructions needed to rehydrate
the probes. For each probe, add the recommended amount of
purified, double-distilled, deionized, and autoclaved water
(PCR grade water), and mix well to get a stock solution of
100 pmol/l. Prepare several aliquots from these stock solutions to avoid degradation by repeated freezing and thawing,
and keep at 20 C until required.
2.3 Colorimetric
Detection of Nucleic
Acids
Methods
3.1 Preparation
of Gold Nanoparticles
Prepare gold nanoparticles of 13 nm diameter by the citrate reduction method according to Grabar et al. [32] or purchase commercially (e.g., from Strem Chemicals. Newburyport, MA, USA).
1. Add 10 ml of 38.8 mM sodium citrate solution rapidly to
100 ml of vigorously stirred boiling 1 mM HAuCl4 aqueous
solution (see Note 3).
2. The mixture should be boiled for 10 min and stirred for an
additional 15 min.
249
250
Notes
1. Inoculate SVCV onto EPC cell line maintained in Eagles minimal essential medium (EMEM) buffered to pH 7.6 with
sodium bicarbonate, supplemented with 2 % of fetal bovine
serum (FBS) and standard concentrations of antibiotic.
Incubate the inoculated cultures at 15 C.
2. Optimum concentrations of NaCl and of oligonucleotide
probe need to be determined for each assay, allowing the visual
detection of the color change of the solutions and, at the same
time, an effective annealing of the probe to its complementary
target. For the detection of SVCV, the concentration of NaCl
sufficient for both aggregation of AuNPs and proper annealing
of the probe to its complementary target was 0.4 M. Use different concentrations of the probe to determine the optimum
probe concentration sufficient to stabilize the AuNPs in the
presence of salt. We found that a final probe concentration of
more than 3 M was too high for any aggregation to occur in
the presence of the target, leading to false negative results. In
contrast, a probe concentration of less than 0.2 M did not
prevent aggregation of AuNPs in the absence of the target and
led to false-positive results.
3. A change in the color of the solution from pale yellow to deep
red will be observed.
4. The specificity of the assays should be previously assessed using
nucleic acids extracted from closely related organisms as template. Our SVCV-targeted assay showed no false-positive
results when tested with nucleic acids extracted from epizootic
hematopoietic necrosis virus (EHNV), infectious hematopoietic necrosis virus (IHNV), infectious salmon anemia virus
(ISAV), koi herpes virus (KHV), viral hemorrhagic septicemia
virus (VHSV), pike fry rhabdovirus (PFRV), zander rhabdovirus (ZRV), and, also, the EPC cells.
5. The lower detection limit of the AuNPs-based assay was determined using a tenfold serial dilution of RNA extracted from
SVCV grown in EPC culture with a known titer estimated
according to the method described by Reed and Muench [33]
(Fig. 3). The detection limit is the lowest RNA concentration
able to change the solution color from red to blue upon aggregation of AuNPs.
6. The ability of the AuNPs-based assay to detect SVCV RNA
directly from fish specimens can be evaluated by testing RNA
samples extracted from SVCV-infected and noninfected fish
tissue homogenates. Test outcomes can be compared with
those of virus isolation or PCR amplification [34].
251
Fig. 3 Lower detection limit of the gold nanoparticle (SVCV-AuNP) assay was
estimated in cell culture system using a tenfold dilution series of the nucleic acid
tested (SVCV RNA). Tubes 110 contain 105, 104, 103, 102, 10, 101, 102, 103,
104, and 105 TCID 50 ml1, respectively. SVC-AuNPs assay detection limit was
assessed as about 103 TCID 50 ml1 (Tube 8; blue color)
References
1. Kaittanis C, Santra S, Perez JM (2010)
Emerging nanotechnology-based strategies for
the identification of microbial pathogenesis.
Adv Drug Deliv Rev 62:408423
2. Jain KK (2005) Nanotechnology in clinical
laboratory diagnostics. Clin Chim Acta
358:3754
3. Rosi NL, Mirkin CA (2005) Nanostructures in
biodiagnostics. Chem Rev 105:15471562
4. Kreibig U, Genzel L (1985) Optical absorption spectra of pairs of small metal particles.
Surf Sci 156:678700
5. Quinten M, Kreibig U (1986) Optical properties of small metal particles. Surf Sci
172:557577
6. Link S, El-Sayed MA (2000) Shape and size
dependence of radiative, nonradiative, and
photothermal properties of gold nanocrystals.
Int Rev Phys Chem 19:409453
7. Nath N, Chilkoti A (2002) A colorimetric gold
nanoparticle sensor to interrogate biomolecular interactions in real time on a surface. Anal
Chem 74:504509
8. Nath N, Chilkoti A (2004) Label free
colorimetric biosensing using nanoparticles. J
Fluoresc 14:4
9. Jain PK, Lee KS, El-Sayed IH, El-Sayed MA
(2006) Calculated absorption and scattering
properties of gold nanoparticles of different
size, shape, and composition: applications in
10.
11.
12.
13.
14.
15.
16.
252
26.
27.
28.
29.
30.
31.
32.
33.
34.
Chapter 20
Nucleic-Acid Testing, New Platforms and Nanotechnology
for Point-of-Decision Diagnosis of Animal Pathogens
Fernando Teles and Lus Fonseca
Abstract
Accurate disease diagnosis in animals is crucial for animal well-being but also for preventing zoonosis
transmission to humans. In particular, livestock diseases may constitute severe threats to humans due to
the particularly high physical contact and exposure and, also, be the cause of important economic losses,
even in non-endemic countries, where they often arise in the form of rapid and devastating epidemics.
Rapid diagnostic tests have been used for a long time in field situations, particularly during outbreaks.
However, they mostly rely on serological approaches, which may confirm the exposure to a particular
pathogen but may be inappropriate for point-of-decision (point-of-care) settings when emergency
responses supported on early and accurate diagnosis are required. Moreover, they often exhibit modest
sensitivity and hence significantly depend on later result confirmation in central or reference laboratories.
The impressive advances observed in recent years in materials sciences and in nanotechnology, as well as in
nucleic-acid synthesis and engineering, have led to an outburst of new in-the-bench and prototype tests for
nucleic-acid testing towards point-of-care diagnosis of genetic and infectious diseases. Manufacturing,
commercial, regulatory, and technical nature issues for field applicability more likely have hindered their
wider entrance into veterinary medicine and practice than have fundamental science gaps. This chapter
begins by outlining the current situation, requirements, difficulties, and perspectives of point-of-care tests
for diagnosing diseases of veterinary interest. Nucleic-acid testing, particularly for the point of care, is
addressed subsequently. A range of valuable signal transduction mechanisms commonly employed in
proof-of-concept schemes and techniques born on the analytical chemistry laboratories are also described.
As the essential core of this chapter, sections dedicated to the principles and applications of microfluidics,
lab-on-a-chip, and nanotechnology for the development of point-of-care tests are presented. Microdevices
already applied or under development for application in field diagnosis of animal diseases are reviewed.
Key words Lab-on-a-chip, Microfluidics, Nanotechnology, Nucleic-acid testing, Point of care,
Veterinary diagnosis
Abbreviations
AIDS
BTRP
CNT
DNA
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_20, Springer Science+Business Media New York 2015
253
254
EIDSS
FIA
FMD
FMDV
FRET
GNP
HIV
HPAI
LAMP
LATE-PCR
MB
MNP
MWCNT
NASBA
NP
PCR
PDMS
PMMA
PNA
POC
QCM
QD
RNA
rPCR
RT-PCR
SARS
S/N
S/V
SELEX
SPR
SWCNT
TADR
Current Situation
255
viral origin. Animal or human contamination with a veterinaryrelevant pathogen may occur through infected animals (live or
dead) and infected animal products (e.g., bush meat, unpasteurized milk) or, to a lesser extent, through direct contact with the
pathogen itself. In developing countries and regions, the impact of
livestock diseases and outbreaks goes far beyond animal welfare
and food safety. Too often, farm animals are also the only way of
human and cargo transportation and also of income, as feedstock,
as a source of manure and draft power, and as raw material for
other industries (e.g., leather, shoes, and clothing) [1].
The advent of human immunodeficiency virus (HIV)/acquired
immunodeficiency syndrome (AIDS) and other human immune
depressing conditions has been a relatively neglected factor of
enhanced susceptibility of humans to animal diseases, especially
with respect to pets. Immune-depressed individuals or other vulnerable groups, such as children or the elderly, may even become
susceptible to subtypes of zoonotic microorganisms that are usually harmless for healthy humans [2]. Interaction between domestic and wild animals may also provide an important via for indirect
human contamination [3]. A particularly important group of zoonotic agents is that of enteric pathogens that are transmitted to and
between humans and animals through the oral-fecal route, with
subsequent dissemination to wastewater effluents. Outbreaks may
occur upon contamination of surface waters and groundwater used
for recreational and irrigation purposes and insufficient microbial
removal of drinking water and/or of treated wastewaters, since
many of these enteric pathogens are resilient to classical treatment
and disinfection procedures [4]. They often contaminate water
supplies in very small concentrations, hence hindering final disinfection and complicating detection as well [5]. Screening of enteric
zoonotic agents has been proposed for pets cohabiting with
immune-susceptible humans [6]. Very often, the presence of
enteric viruses is detected indirectly, through bacterial indicators of
fecal contamination, namely, coliforms and enterococci [5]. On
the other hand, emerging wild-type zoonosis have been transmitted from animals to humans (e.g., SARS and West Nile virus)
or after specific mutations that facilitate the species jump (e.g.,
HIV and H5N1 or H1N1 influenza) [7]. Concerning bioterrorism issues and depending on the nature of the infectious pathogen,
the detection of potential zoonotic agents in animals should alert
for an intentional release act [8]. A successful anti-bioterrorism
strategy must not only account for the detection of the known
pathogen but also of genetically modified forms. However, only a
few potential veterinary pathogens fulfill the criteria for being considered effective bioterrorist agents. The group is composed by the
viral agents of rinderpest, classical swine fever, African swine fever,
avian influenza, Newcastle disease, Rift Valley fever, and foot-andmouth disease (FMD) [1]. Among them, FMD and avian
256
257
258
259
260
been also used for field differentiation between vaccine and field
strains, especially in endemic regions where vaccination is permitted [12]. Suitable application of such differential testing at slaughtering settings to confirm prior livestock vaccination has been
argued [1].
1.4 Challenges
for Point-of-Care
Testing
261
262
Nucleic-Acid Testing
2.1 Advantages
for Disease Diagnosis
263
264
Most of the current POC tests for human and animal medicine still
rely on antigen/antibody bioaffinity reactions, but they are usually
handicapped by relatively small sensitivity. Thus, there is a trend for
shifting towards more sensitive nucleic-acid detection, usually
based on DNA/DNA hybridization schemes. A major challenge
for detection of pathogen nucleic acids in host body fluids, apart
their scarcity compared to antigenic proteins, is the confinement
inside pathogen cells, surrounded by hard biomembranes and cell
walls. Consequently, the nucleic-acid levels present in host fluids
may be too low for successful diagnosis without prior target amplification by PCR-based techniques. For blood infections in humans,
the amount of human genomic DNA can be 1014 times higher than
that of pathogen DNA [39], an important challenge in terms of
sensitivity and selectivity. For POC testing, this constitutes a challenge for the miniaturization of the blood sample preparation step,
especially in the case of gram-positive bacteria, whose cell walls are
thicker and more rigid than those of gram-negative bacteria. The
majority of available POC systems for clinical and veterinary applications require off-device sample preparation (including microorganism concentration and nucleic-acid extraction). This is a
traditionally cumbersome and time-consuming step, especially for
in-the-field testing, but newer processes, including filter paper capture of nucleic acids and automated extraction procedures and kits,
have been developed to simplify such task [40]. In parallel with
POC device development, considerable efforts have been spent in
the simplification and robustness of sample preparation, in order to
minimize manual handling, thus reducing cross-contamination
and the effects from potentially interfering substances present during the nucleic-acid amplification assay. Of note are the technical
difficulties inherent to the analysis of matrices such as feces, semen,
and decomposing tissue or the detection of Mycobacterium tuberculosis from saliva [12]. For these cases, it is necessary to incorporate, in POC devices, systems that are able to suppress such
interfering substances and that reduce handling prior to the
nucleic-acid amplification step.
PCR has been, by far, the most widely employed technique for
nucleic-acid amplification in laboratory analysis but also in POC
tests. Many similar techniques have been originated from the basic
PCR principles, including nucleic-acid sequence-based amplification (NASBA), targeted for direct RNA amplification, thus precluding the need for a previous reverse-transcription step. The
development of this panoply of techniques for pathogen detection
has been possible due to the increasing availability of whole
genomic sequences; a remarkable case in veterinary science is the
current possibility of rapidly distinguishing severe acute respiratory
265
266
267
Optical detection has been the most widely employed transducing principle in biosensing, partially driven by the unending
advances of optic applications in telecommunications and information systems. Furthermore, the very high frequency of optical signals is appropriated for the enormous amount of information that
can be carried by optical systems and devices [24]. Conventional
optical microscopy has been reliably used for detection and imaging of infectious pathogens (at both cellular and molecular levels),
integrated with research and development of new POC diagnostic
devices [50]. Within optical detection methods, chemiluminescence and fluorescence have been the most widely used for their
versatility of designs and applications. Imaging methods usually
require bulky and expensive microscopes and camera-equipped
microscopy systems, i.e., in off-device formats. Such equipment is
obviously unsuitable for POC diagnostics. The innovative technique of optofluidic seems thus promising in this regard, as this
high-throughput and high-resolution on-device technique does
not make use of such components [51]. In optofluidic devices, the
sample flows through a metal film-etched array with submicron
apertures for imaging, onto a plastic platform.
An intrinsic limitation of conventional optical detection is the
interference between closely spaced light waves. Understandably,
this is especially limiting for the design and performance of miniaturized devices for multi-analyte detection and identification.
However, optical transmission through minuscule structures
gained a new impulse with the technique of surface plasmon resonance (SPR), in which light waves are directed to the interface
between a metal and a dielectric. The technique is label-free and
the immobilized probe can be easily reused, although sensitivity
must still be improved [24]. Moreover, it faces the challenges of
other optical techniques for POC diagnosis, namely, the high cost,
instrumental complexity, expensiveness of optical components,
and difficulty for miniaturization/portability and mass production.
The advent of integrated photodiode detectors is promising for the
development of cheap, sensitive, easy-to-use, and easy-to-fabricate
fluorescence-based diagnostic microdevices [52]. A remarkable
milestone might be the shift from current fluorescent-based detection methods to naked-eye (visual) recognition, which would considerably reduce complexity and costs. These readouts can be
recorded as digital images and then transmitted to remote clinicians through trivial and inexpensive telecommunication devices
(e.g., digital cameras and scanners); some current examples include
the detection of glucose, pH, and proteins [53]. It is expected that
future POC diagnostic devices will become connected to wireless
communication and information systems through camera-equipped
mobile phone networks coupled to web databases, for diagnostic
imaging and telemedicine, especially in remote and resourcelimited regions and settings [54]. Remote monitoring will not
268
4
4.1
Multiplex PCR and DNA microarrays have been among the most
commonly used techniques for the detection and identification of
infectious pathogens. Multiplex PCR is frequently limited to the
analysis of only a few target genes, owing to interferences between
different primer sets in the same reaction vessel. DNA microarrays
are usually too expensive, labor intensive, and complex for POC
applications; moreover, the microarray assays are usually lengthy
due to the slow diffusion-limited hybridization kinetics. On the
other hand, lateral-flow devices employed for POC diagnosis usually have relatively low sensitivity; moreover, in the most common
format (immunochromatographic format, corresponding to the
well-known test strips), they do not provide genetic information,
thus hindering the assessment of eventual pathogen subtypes and/
or drug resistance markers. In general, the number of targets that
can be simultaneously probed in lateral-flow assays is low, thus
increasing the cost per assay and limiting the throughput. Also,
their degree of multiplexing usually does not reach that of microarrays [55].
The last decade has witnessed tremendous advances in microfluidic sciences and applications for bioanalysis. Microfluidic
devices basically consist in a set of microchambers interconnected
through microchannels, imprinted onto a suitable solid platform.
After injection into the system, the liquid biological sample flows
throughout the hydrophilic inner walls of the chambers and channels for final analysis. The flow is usually controlled automatically
by mechanical valves. A primary advantage of microfluidics for the
development of miniaturized diagnostic devices, especially for
POC usage, is the requirement for only very small sample volumes,
which permits to save reagents (viz., probes and labels), but also
gaining time and sensitivity, as a consequence of the enhanced
binding kinetics (unlike in bulky reaction media). It also provides
lower power consumption and lower operating costs. Moreover,
since the overall analytical process is automated, sample losses and
contamination due to human handling can be minimized, in parallel with improved multiplexing capabilities. The confinement and
automation of the process also reduces errors due to human
manipulation and increases biosafety. In microfluidic devices, the
occurrence of nucleic-acid hybridization in solution and the
enhanced aqueous mixing allow surpassing the kinetic barriers that
slow affinity reactions due to limited diffusion in traditional
269
270
Lab-on-a-Chip
271
hence increases sensitivity [53]. In lab-on-a-chip devices, minimization of the chemical interferences that result from tight spatial
confinement is usually envisaged, enabling miniaturization and
highly sensitive detection. The high mass transfer rate thus achieved
is due to the low diffusional distance and high surface-to-volume
ratio (S/V) [59]. Traditional cell analysis and processing, including cell sorting, cell/serum separation (for immunoassays), and
cell lysis (for immunoassays or nucleic-acid amplification), can be
adapted to POC schemes [53]. Some principles commonly used to
separate and concentrate cells within POC devices are cell size,
labeling (fluorescent or magnetic), electrophoretic mobility, and
cell adhesion [60]. For analysis of real, biological samples, the
detection of highly virulent pathogens presumes the ability of
detecting very few cells from large sample volumes and, concurrently, washing out sample contaminants (e.g., other cells), in
order to prevent, for instance, downstream PCR inhibition and
microchannel clogging [61]. In particular, environmental water
samples may contain certain substances, like humic acids, that
inhibit PCR reactions, able to drastically decrease the resulting signals. The presence of contaminants in biological samples may be
particularly troublesome in microfluidic assays for veterinary
applications.
A particular advantage of bioanalysis with lab-on-a-chip devices
concerns the processing of pathogen RNA. Owing to the confined
environment of the purification steps and to the requirement for
minimal user handling, the probability for RNA contamination
and degradation by ubiquitous RNases is highly decreased [62].
Schemes employing nucleic-acid extraction through detergents
rather than high temperature-operating lytic enzymes coupled
with isothermal (e.g., LAMP) instead of PCR and RT-PCR amplification processes are also preferred in this regard. In this way, the
concept of microfluidics might be able to address a recurrent gap
in veterinary literaturethe lack of information about RNA quality and integrity, which further complicates the standardization of
RT-PCR procedures [34]. Several commercial multiplexed POC
tests for diagnosis of influenza and other respiratory viruses are
already available, as the FilmArray RP (BioFire Diagnostics, Salt
Lake City/UT, United States), the xTAG RVP (Luminex, Toronto,
Canada), and the Xpert Flu A Panel (Cepheid, Sunnyvale/CA,
United States), consisting in closed systems that include sample
preparation and detection by integrated RT-PCR [63, 64].
However, these systems may not be able to detect low viral load
specimens, as is the case of the Xpert Flu A Panel with some important avian influenza viruses. In addition, economic constraints still
restrict full applicability of some of these tests in POC testing. For
the Xpert Flu A panel, it was estimated a cost of 45 euros per test,
compared to 15 euros for the antigenic tests [63]. Current lab-ona-chip devices, despite being simpler than conventional laboratory
272
Table 1
Illustrative works described in the literature employing nucleic-acid amplification methods
for the detection of animal pathogens. POC tests are those claiming, at least, one of the following
characteristics: lab-on-a-chip; microfluidics; portability
POC
Target pathogen(s)
Yes
Amplification
No method
LOD or sensitivity
References
LAMP
330 copies
[67]
RT-PCR
[10]
rRT-PCR
98 %
[42]
Influenza virus
LAMP
90.9 %
[43]
Influenza A viruses
RT-PCR
4005,000 viral
particles/ml
[63]
RT-PCR
[62]
FMDV
rPCR
109 dilution
[9]
FMDV
RT-LAMP
10 copies
[13]
FMDV
RT-LATE-PCR
10 copies (100 %)
[38]
Respiratory viruses
RT-PCR
82.2100 %
[64]
RT-LAMP
[41]
LAMP
<10 copies/l
[65]
PCR
0.2 CFU/l
[61]
RT-LAMP
17 copies
[40]
SARS-CoV
RT-PCR
10 to 10 CFU/ml [66]
rPCR
10100 fg
[8]
LAMP
20 copies
[68]
273
Nanotechnology
An emergent topic in the development of new bioanalytical procedures, structures, and systems is nanotechnology, particularly for
the generation of useful nanostructures for diagnostic applications;
this is the so-called field of nanobiotechnology. Novel and
improved electronic devices and biosensor platforms have emerged
as a consequence of the inherent small size, enlarged surface area,
and unusual optical, magnetic, catalytic, and mechanical properties
of nanomaterials, unlike those of bulk materials [24]. Depending
on their specific nature, for biosensing, nanomaterials may act as
labels (including signal amplification), as biomolecule immobilization supports, or even as probes for specific biotarget anchoring.
Certain nanomaterials can also be used for pre-concentration of
biological targets. Among these applications, labeling has been the
most commonly employed. Label-based detection methods are
usually more time-consuming and labor intensive than label-free
methods due to the labeling steps. Labels have also limited shelf
lives and are subjected to leakage from sensing surfaces. However,
label-based methods usually provide superior performance, especially in terms of sensitivity, than label-free ones. Moreover, standardized protocols with labeling procedures are already available.
Fluorescence labeling has been, by far, the most common approach
in this regard, although suffering from pH sensitivity and photobleaching over time [53]. Such handicaps and the advent of nanoengineering have propelled the search and development of new
and improved labels.
Nanoparticles (NPs) have been the most widely employed type
of nanomaterials for biosensing, especially metallic NPs. Metallic
NPs are inorganic NPs that exhibit improved physicochemical
characteristics compared to fluorescent labels, including higher
sensitivity. In general, they are suitable for construction of highdensity bioanalytical devices, taking advantage of their high signalto-noise ratio (S/N). They are easily synthesizable and
functionalized (by simple mixing at room temperature) and have a
controlled, self-assembled surface structure [24]. Gold nanoparticles (GNPs), in particular, are already used frequently in molecular
diagnosis; some of their advantages include low toxicity and versatility for many specific biorecognition applications and schemes.
One common way to enhance the GNP signal even further, and
thus the sensitivity of detection, is the inclusion of a final step of
silver staining (silver enhancement), yielding detection schemes
able to preclude the use of a prior PCR amplification step. The
high sensitivity exhibited by many NP-based detection layouts,
especially in the form of microarrays, has enabled to avoid a prior
step of nucleic-acid amplification [69].
Quantum dots (QDs) constitute another class of metallic NPs,
able for fluorescence tagging. They are much brighter and more
274
275
276
277
Table 2
Illustrative works described in the literature employing nanostructures for the detection of animal
pathogens
Target
pathogen(s)
Nanostructuresa
Transduction
mechanism
LOD or
sensitivity
Canine
parvovirus
PNA
Fluorescence
40
[105]
2,000 copies/
l (89.8 %)
Influenza virus
(H5)
MB
Fluorescence
0.6 nM
[37]
Influenza virus
(H5N1)
Light scattering
103 TCID50
units
[69]
Influenza virus
(H5N1)
DNA aptamer
SPR
1.28 HAU
[87]
Influenza virus
(H1N1)
GNPs
Fluorescence and
surface-enhanced
Raman scattering
[93]
Influenza virus
(H5N1)
Complementary oxide
semiconductor (CMOS)
Impedance
spectroscopy
5 nM (1011 F)
[98]
Influenza virus
(H5N1)
DNA aptamer/hydrogel
QCM
0.0128 HAU
[102]
16 avian
influenza
viruses
Magnetic beads
Feline
calicivirus
Liposomes
Fluorescence
[5]
Magnetic beads
Pestiviruses
(Classical
swine fever
virus; Border
disease virus;
Bovine viral
diarrhea virus
1 and 2)
Optic (visual;
microscopy; chip
reader)
[55]
Alexandrium
sp. complex
Colorimetry
[89]
B. anthracis
SWCNT
Raman spectroscopy
[97]
B. anthracis
Cyclic voltammetry
0.01 ng/l
[94]
B. anthracis
GNPs
QCM
B. anthracis; S.
enteritidis
50 pg/ml
References
[90]
(continued)
278
Table 2
(continued)
Target
pathogen(s)
Nanostructuresa
Transduction
mechanism
LOD or
sensitivity
References
E. coli
DNA aptamer
Impedance
spectroscopy
107 M
[79]
E. coli
Alginic acid-coated Co
magnetic beads
[86]
E. coli
Amperometry
5 CFU/ml
[99]
E. coli
O157:H7
Cyclic voltammetry
and impedance
spectroscopy
0.5 nM
[91]
E. coli
O157:H7
Fluorescence
250 zM
[104]
F. tularensis
MB
Fluorescence
[84]
M. avium
GNPs
Colorimetry
1.875 ng/l
[100]
(87.5100 %)
M. tuberculosis;
M. bovis
GNPs
Colorimetry
5 108 M
[85]
S. aureus
GNPs/poly-3,4ethylenedioxythiophene
(PEDOT) film
Chronoamperometry
150 pM
[96]
S. aureus
GNPs/PANI nanofibers
Cyclic voltammetry
pM range
[101]
S. aureus
(MRSA)
PNA
Impedance
spectroscopy
10 pM
[103]
1.76 1012 M
[92]
C. perfringens; GNPs
C. tetani; S.
pneumoniae;
P. aeruginosa;
E. coli
QCM
Salmonellae
Colorimetry
104 cells
[88]
Conclusions
The advances observed on the past decades in proteomics and
genomics have led to the discovery of novel diagnostic biomarkers
for pathogens relevant in human medicine. In parallel, cuttingedge developments in materials science and in nanotechnology
279
have also been registered. Veterinary science and practice took the
train and, as such, have been greatly favored from such advances.
Microfluidic technologies and nanoengineered structures, especially when coupled together, have led to an unprecedented degree
of high-throughput, large-scale genetic analysis, even at wholegenome levels. Ultimately, the high-throughput and multiplexing
abilities of in vivo (implanted or swallowed) nanosensor arrays
should be able to monitor animals physiology and health status
during their entire lifetime, and even beyond, intended to track
and assess the quality of animal products for human feeding. In this
way, the current shortcomings related with the limited number of
sanitary surveillance resources that can be allocated to guarantee
proper product origin, stocking, and shipping could be circumvented. So far, only few POC schemes and devices have reached the
exquisite sensitivity thresholds required for detection of nucleicacid traces in unamplified biological samples, which is the ultimate
goal of nucleic-acid testing. As for human diagnostics, the veterinary medicine and practice still lack the commercial availability of
more POC devices, more probably as a result of manufacturing and
commercial cost-effectiveness constraints than to a shortage of fundamental knowledge. While remaining too expensive for single
testing, many tests targeted for POC diagnosis will ultimately prove
to be cost-effective when savings with unnecessary laboratory manpower are taken into account. Other major challenges are the need
for initial investments that are often prohibitive for small companies and the usual difficulties for obtaining regulatory approvals for
testing and commercialization. For effective improvement of
human health, more adequate and coordinated actions to face animal diseases are needed, especially concerning livestock. This
includes a more effective technology transfer from developed countries to those where diseases are prevalent and where disease preventive measures may be crucial to avoid or contain epidemics.
Probably still without meritorious examples in the world, more
effective communication and coordination among public health,
animal health, and wildlife disease surveillance authorities will be
necessary to tackle the problems posed by common and hazardous
veterinary diseases, especially in situations of outbreaks endangering animal and human health as well.
References
1. Breeze RG (2006) Technology, public policy
and control of transboundary livestock diseases in our lifetimes. Rev Sci Technol
25:271292
2. Pieniazek NJ, Bornay-Llinares FJ, Slemenda
SB et al (1999) New Cryptosporidium genotypes in HIV-infected persons. Emerg Infect
Dis 5:444449
3. Robertson ID, Irwin PJ, Lymbery AJ et al
(2000) The role of companion animals in the
280
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
281
282
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
283
Part III
Molecular Profiling of Veterinary Relevant Microbial
Pathogens
Chapter 21
Molecular Typing Tools: From Pattern Recognition
to Genome-Based Algorithms
Konrad Sachse and Petra Moebius
Abstract
In the present chapter, we discuss DNA-based typing methods for microbial pathogens that were frequently
used in the past two decades and their essential features, as well as virtues and downsides. We conclude
with an outlook on the fundamental changes that can be expected in the era of high-throughput
genomics.
Key words Review, RAPD, RFLP, AFLP-PCR, PFGE, VNTR/MLVA, MIRU-VNTR, Spoligotyping,
Single-locus sequence typing, MLST, Genomotyping
Introduction
Ever since diagnosticians have been trying to identify the origin of
disease outbreaks, there was a demand for characterizing and classifying the microbial strain involved. Strain typing can help to
reveal important information about the infection, i.e., (1) transmission pathways and patterns, (2) possible association of the strain
with clinical manifestations and pathogenicity, (3) specific tissue or
organ affinity, and (4) the status of the infection, e.g., new infection, reinfection, or persistent infection.
Historically, the first typing schemes were based on phenotypes,
e.g., surface morphology (e.g., gram stain), serotype, biotype, or
intrinsic phenotypic properties, such as fermentation of substrates,
auxotrophy, or expression of specific antigens. The broad use of
DNA amplification and sequencing technology in the past two
decades prompted the gradual phasing out of most of these methods and a shift toward genetic typing schemes.
Most of the currently recognized approaches are based on
variations in specific genomic loci that are exploited in a direct (i.e.,
through sequence analysis) or indirect (e.g., through cleavage patterns) fashion. Since the target region for typing is usually selected
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_21, Springer Science+Business Media New York 2015
287
288
2
2.1
289
Table 1
Selection of published genotyping studies on bacterial pathogens
Agent
Genotyping method
References
Bacillus anthracis
VNTR/MLVA
SNP analysis
[9193]
[94, 95]
Borrelia spp.
[54, 96]
[97]
Brucella spp.
SNP analysis
[98]
VNTR/MLVA
[99101]
MLST
[102, 103]
Burkholderia mallei
MLST
VNTR/MLVA
PFGE
[104106]
[107, 108]
[109]
Campylobacter spp.
PFGE
MLST
PCR-RFLP genotyping (flaA)
Microarray DNA hybridization
assay
[110]
[111, 112]
[11, 113]
[114, 115]
PFGE
[25, 116]
AFLP
MLST
[21, 117]
[118, 119]
Chlamydia spp.
MLST
[61, 120]
Chlamydia psittaci
Chlamydia abortus
VNTR/MLVA
[123]
Clostridium botulinum
PFGE
VNTR/MLVA
[26, 27]
[124]
Clostridium perfringens
PFGE
VNTR/MLVA
[125128]
[129, 130]
Clostridium difficile
VNTR/MLVA
[131]
Clostridium septicum
MLST
[132]
Coxiella burnetii
VNTR/MLVA
10-locus multi-spacer sequence
typing (MST)
[133135]
[68, 69]
Escherichia coli
[136139]
[122]
[62]
[140]
[48, 49, 141, 142]
[143]
[144146]
(continued)
290
Table 1
(continued)
Agent
Genotyping method
References
Microarray genotyping
MLST
[147149]
[150]
PFGE
AFLP
VNTR/MLVA
SNP genotyping (RT-PCR,
microarray)
VNTR + INDELs + SNP
[151]
[151]
[152, 153]
[154]
Reviews
PFGE
IS900-RFLP
MIRU-VNTR
MLSSR
[157, 158]
[159161]
[16, 17]
[33]
[3, 162164]
IS901-RFLP
MIRU-VNTR
[165]
[33]
IS1245-RFLP, IS1311-RFLP
MIRU-VNTR
[166168]
[33]
Mycobacterium tuberculosis
complex
IS6110-RFLP
[13]
MIRU-VNTR
[3538]
M. bovis
M. caprae
MIRU-VNTR
Spoligotyping, microarray
spoligotyping
[169]
[41, 44]
VNTR/MLVA
MLST
[170]
[171173]
Mycoplasma bovis
RAPD
AFLP
PFGE
VNTR/MLVA
MLST
[5]
[5]
[5]
[174, 175]
[176]
Salmonella enterica
Reviews
PFGE
VNTR/MLVA
S. Typhimurium
VNTR/MLVA
S. Enteritidis
VNTR/MLVA
S. Newport
MLST
[177179]
[28, 179, 180]
[181]
Francisella tularensis
Taylorella equigenitalis
[155, 156]
[182184]
[185]
[59, 186]
(http://mlst.
ucc.ie/mlst/
dbs/Senterica)
[187]
[188]
[77, 189, 190]
PFGE
[191194]
291
RFLP
292
AFLP-PCR
2.4
PFGE
The acronym stands for macro-restriction fragment length polymorphism analysis using pulsed-field gel electrophoresis. The
method utilizes specific restriction sites throughout the microbial
genome for differentiation below the species level. Bacterial cultures are embedded in agarose blocks, lysed in situ, and digested
with rare-cutter restriction endonucleases. The resulting macrorestriction fragments sized up to 10 Mb can only be separated
using agarose gel electrophoresis with an alternating electric field
[23]. The final fingerprint patterns typically consist of 1020 fragments. For an update on recent developments in PFGE technology, the reader is referred to an exhaustive review [24].
Due to its high discriminatory power, PFGE analysis evolved
as a gold standard for typing of many bacteria, such as Campylobacter
293
spp. [25], Clostridium spp. [26, 27], and Salmonella [28]. There
are publicly accessible databases having thousands of individual
strain patterns of food-borne pathogens, such as Salmonella
serovars, Listeria monocytogenes, and others (e.g., http://www.
pulsenetinternational.org/).
The main reason why the use of the procedure has been confined to a limited circle of specialized laboratories lies in its sophisticated and technically demanding work flow. For instance, the in
situ digestion of genomic DNA in the agarose block may prove
difficult for certain pathogens. Therefore, it remains a challenge to
attain satisfactory interlaboratory reproducibility.
3
3.1
294
Table 2
VNTR loci recommended for MLVA typing of Mycobacterium bovis, Mycobacterium caprae, and other
MTC isolatesa
Locusb
Synonym
VNTR 424
Specificity
MTC, M. bovis + M. caprae, M. caprae
VNTR 580
ETR D
VNTR960
MIRU 10
VNTR 1644
MIRU 16
M. bovis + M. caprae
VNTR 1955
VNTR 2163a
QUB 11a
VNTR 2163b
QUB 11b
VNTR 2165
ETR A
VNTR 2461
ETR B
M. bovis
VNTR 3232
QUB 3232
VNTR 3690
VNTR 4052
QUB 26
VNTR 4156
QUB 4156
Bold print means that the respective loci are particularly suitable for differentiating among M. bovis and
M. caprae strains, respectively
a
Compiled by the National Reference Laboratory for Bovine Tuberculosis at Friedrich-Loeffler-Institut Jena, Germany
(Head: Dr. Irmgard Moser)
b
All loci have a discriminatory index larger than 0.6 according to Hunter and Gaston [195]
3.2
MIRU-VNTR
3.3
SSR Typing
295
Genomic regions harboring this kind of repeats are often the most
variable targets in a bacterial genome, whereas longer repeats are
generally less diverse. SSR typing was suggested for differentiation
and subtyping of Mycoplasma spp., Mycobacterium spp., and other
bacteria [29].
The method can be efficient in discriminating between similar
strains. However, mononucleotide repeats of more than ten units
can give rise to slipped-strand mispairing or replication slippage
events affecting the DNA polymerase during the amplification
reaction. This leads to inaccurate results as shown in a study on M.
avium subsp. paratuberculosis [40].
3.4
Spoligotyping
296
297
298
299
300
301
Conclusion
The development of microbial typing over the last two decades has
significantly contributed to enhanced surveillance and outbreak
management [7, 63, 83, 87], as well as to ever more detailed characterization of field strains in terms of virulence properties, antimicrobial resistance, phylogenetic position, etc. As outlined in the
previous sections, the evolution of tools shows a clear tendency
toward sequence-based methods, with the nascent WGS-based
algorithms holding great promise.
Given the diversity in the various methodological approaches
and the multitude of typing protocols, it is obvious that there is no
single method that would be universally applicable. The choice of
the methodology rather depends on the objective of the study, the
concrete epidemiological situation, and the budget available.
For instance, in an outbreak scenario, rapid methods are preferable, such as VNTR, partial sequencing, and RAPD. As shortterm delivery of relevant strain characteristics is crucial to inform
decision makers, it can be more important to use a fast and robust
tool rather than aspiring to the highest resolution. In monitoring
and surveillance schemes, the overall cost, practical feasibility, and
availability of a standardized protocol are likely to be the essential
parameters. Therefore, MLST and VNTR schemes are frequently
used for this purpose.
Finally, retrospective studies and epidemiological research
projects will be demanding ever higher information contents to be
obtained on the strains involved, as well as maximum resolution,
all of which can be delivered using genome-wide typing methods.
References
1. de Beer JL, van Ingen J, de Vries G et al
(2013) Comparative study of IS6110 restriction fragment length polymorphism and
variable-number tandem-repeat typing of
Mycobacterium tuberculosis isolates in the
Netherlands, based on a 5-year nationwide
survey. J Clin Microbiol 51:11931198
2. Pitondo-Silva A, Santos AC, Jolley KA, Leite
CQ, Darini AL (2013) Comparison of three
molecular typing methods to assess genetic
diversity for Mycobacterium tuberculosis. J
Microbiol Methods 93:4248
3. Fritsch I, Luyven G, Kohler H, Lutz W,
Mobius P (2012) Suspicion of Mycobacterium
avium subsp. paratuberculosis transmission
between cattle and wild-living red deer
(Cervus elaphus) by multitarget genotyping.
Appl Environ Microbiol 78:11321139
4. Welsh J, McClelland M (1990) Fingerprinting
genomes using PCR with arbitrary primers.
Nucleic Acids Res 18:72137218
302
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
303
304
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
305
306
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
138.
139.
140.
141.
142.
143.
144.
145.
146.
147.
307
308
170.
171.
172.
173.
174.
175.
176.
177.
178.
179.
180.
181.
182.
183.
184.
185.
186.
187.
188.
189.
190.
191.
309
310
restricted genomic DNA of strains of Taylorella equigenitalis isolated in Ireland and in the
United States. Vet Res Commun 22:217224
192. Kagawa S, Moore JE, Murayama O, Matsuda
M (2001) Comparison of the value of pulsedfield gel electrophoresis, random amplified
polymorphic DNA and amplified rDNA
restriction analysis for subtyping Taylorella
equigenitalis. Vet Res Commun 25:261269
193. Kagawa S, Klein F, Corboz L et al (2001)
Demonstration of heterogeneous genotypes
of Taylorella equigenitalis isolated from horses
Chapter 22
Characterization of Campylobacter jejuni
and Campylobacter coli Genotypes in Poultry Flocks by
Restriction Fragment Length Polymorphism (RFLP) Analysis
Ana Cludia Carreira and Mnica V. Cunha
Abstract
We describe a simple, rapid, and discriminatory methodology that allows the routine molecular characterization of Campylobacter jejuni and Campylobacter coli isolates. The proposed approach is built on one of
the earliest and simplest molecular typing methods ever, consisting on the analysis of the fragments of
different lengths generated by digestion of homologous DNA sequences with specific restriction endonucleases, a process known as restriction fragment length polymorphism (RFLP) analysis. The strategy
underneath the workflow reported here is meant to explore the polymorphisms of Campylobacter spp. flaA
gene (flaA-RFLP) that allows the local investigation of the genetic diversity and distribution of C. coli and
C. jejuni isolates from different sources, namely, chickens caeca. Although not appropriate for global and
long-term epidemiological studies as a single approach, flaA-RFLP analysis can be very useful in surveys
limited in space and time and, for specific epidemiological settings, an alternative to more modern and
resource-demanding techniques.
Key words Genotyping, Restriction fragment length polymorphism, flaA-RFLP, Campylobacter
jejuni, Campylobacter coli, Molecular epidemiology
Introduction
Campylobacteriosis caused by Campylobacter spp. is considered
one of the most prevalent zoonotic enteric infections occurring
worldwide [1], Campylobacter jejuni being associated to the vast
majority of human campylobacteriosis cases (about 90 %), followed
by C. coli (510 %) [2, 3]. The main reservoirs of these species are
the gastrointestinal tracts of poultry (e.g., chickens, turkeys, ducks,
and geese). Handling and consumption of chicken concur as
important risk factors in pathogen transmission to humans [47].
Isolation, identification, and phenotypic and genetic characterization of pathogens are fundamental to understand the
epidemiology of infectious diseases. Using suitable genotyping
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_22, Springer Science+Business Media New York 2015
311
312
313
Materials
2.1 Growth of
Campylobacter spp.
and Maintenance
2.2 Isolation
of Genomic DNA
314
315
3. Molecular weight markers: Select up to three ladders producing regularly spaced 100 bp, 50 bp, and 25 pb fragments.
4. Commercial DNA loading and staining dye or homemade
solution (see Note 6).
5. Standard equipment for performing agarose gel electrophoresis and for detecting nucleic acids under UV light.
2.5 Digestion of
Amplified DNA with
Restriction Enzymes
and Analysis of
flaA-RFLP Profiles
Methods
3.1 Growth of
Thermotolerant
Campylobacter Strains
3.2 Phenol/
Chloroform Extraction
of Genomic DNA
316
3.4 Amplification
of flaA Gene by PCR
317
3.5 Detection of
Amplification Products
by Agarose Gel
Electrophoresis
3.6 Endonuclease
Restriction
of Amplification
Products (flaA-RFLP)
318
Notes
1. To save on gas generation systems, select an incubation jar or
an incubation bag taking into consideration the number of
Petri dishes. According to size, incubation bags are normally
appropriate for two dishes only.
2. SDS precipitates at 4 C. Should this occur, warm the solution
in a water bath up to 20 C.
3. The glass bottle should have twice the capacity of the volume
of the prepared gel. This prevents spillage of gel upon
heating.
4. Staining the gel about 30 min in 1 TBE or in water containing 0.3 g/mL of ethidium bromide before UV visualization
319
320
321
References
1. FAO/WHO
[Food
and
Agriculture
Organization of the United Nations/World
Health Organization] (2009) Risk assessment
of Campylobacter spp. in broiler chickens:
Technical Report. Microbiological Risk
Assessment Series No 12. Geneva. pp 132
2. Miller WG, Mandrell RE (2005) Prevalence of
Campylobacter in the food and water supply:
incidence, outbreaks, isolation and detection.
In: Ketley JM, Konkel M (eds) Campylobacter:
molecular and cellular biology. Horizon
Bioscience, Norfolk, UK, pp 101108
3. Wilson DJ, Gabriel E, Leatherbarrow AJH
et al (2008) Tracing the source of campylobacteriosis. PLoS Genet. doi:10.1371/journal.
pgen.1000203
4. Wingstrand A, Neimann J, Engberg J et al
(2006) Fresh chicken as main risk factor for
campylobacteriosis, Denmark. Emerg Infect
Dis 12:280285
5. Humphrey T, OBrien SJ, Madsen M (2007)
Campylobacters as zoonotic pathogens: a food
production perspective. Int J Food Microbiol
117:237257
6. Dasti JI, Tareen AM, Raimond L et al (2010)
Campylobacter jejuni: a brief overview on
pathogenicity associated factors and diseasemediating mechanisms. Int J Med Microbiol
300:205211
7. Silva J, Leite D, Fernandes M et al (2011)
Campylobacter spp. as a foodborne pathogen: a
review. Front Microbiol 2:112
8. Wassenaar T, Newell D (2000) Genotyping of
Campylobacter spp. Appl Environ Microbiol
66:19
9. Klena JD, Konkel E (2005) Methods for epidemiological analysis of Campylobacter jejuni. In:
Ketley JM, Konkel M (eds) Campylobacter:
molecular and cellular biology. Horizon
Bioscience, Norfolk, UK, pp 166175
10. Fitzgerald C, Sails AD, Fields PI (2005)
Campylobacter jejuni strain variation. In: Ketley
JM, Konkel M (eds) Campylobacter: molecular
and cellular biology. Horizon Bioscience,
Norfolk, UK, pp 5972
11. Jagannathan A, Penn C (2005) Motility. In:
Ketley JM, Konkel M (eds) Campylobacter:
12.
13.
14.
15.
16.
17.
18.
19.
20.
Chapter 23
Pulsed-Field Gel Electrophoresis (PFGE): Application
inPopulation Structure Studies ofBovine
Mastitis-Causing Streptococci
IldaSantos-Sanches, LliaChambel, andRogrioTenreiro
Abstract
Pulsed-field gel electrophoresis (PFGE) separates large DNA molecules by the use of an alternating electrical field, such that greater size resolution can be obtained when compared to normal agarose gel electrophoresis. PFGE is often employed to track pathogens and is a valuable typing scheme to detect and
differentiate strains. Particularly, the contour-clamped homogeneous electric field (CHEF) PFGE system
is considered to be the gold standard for use in epidemiological studies of many bacterial pathogens. Here
we describe a PFGE protocol that was applicable to the study of bovine streptococci, namely, Streptococcus
agalactiae (group B Streptococcus, GBS), Streptococcus dysgalactiae subsp. dysgalactiae (group C
Streptococcus, GCS), and Streptococcus uberiswhich are relevant pathogens causing mastitis, a highly prevalent and costly disease in dairy industry due to antibiotherapy and loss in milk production.
Key words Bovine mastitis, Streptococcus agalactiae, Streptococcus dysgalactiae subsp. dysgalactiae,
Streptococcus uberis, Molecular typing, Molecular epidemiology, Pulsed-field gel electrophoresis,
PFGE, Alternating electrical field, CHEF
1 Introduction
Pulsed-field gel electrophoresis (PFGE) is generally considered the
most discriminatory method to infer relationships between strains
[1, 2]. Briefly, PFGE is based on enzymatic DNA restriction of
bacterial genome (by using rare-cutting restriction endonucleases)
to generate DNA fragments, which are subsequently separated on
a gel, due to molecular reorientation produced by periodic changes
in the electric field, in order to separate the large DNA fragments.
The banding pattern obtained from each isolate is then compared
with the remaining patterns in order to infer epidemiological relation between the isolates. When two isolates show indistinguishable DNA patterns, it is assumed that they are of the same strain,
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_23, Springer Science+Business Media New York 2015
323
324
IldaSantos-Sanches et al.
2 Materials
Prepare all solutions using distilled (or purified) water and analytical
grade reagents. Prepare and store all reagents at room temperature
(unless indicated otherwise). Sterilize the buffers and solutions by
autoclaving at 121C for 20min (or by filtration through a
0.22m filter unit, when indicated).
2.1 Solutions
forStreptococcal
Chromosomal DNA
Agarose Discs
(for PFGE)
325
326
IldaSantos-Sanches et al.
10. SmaI restriction buffer: mix 15ml of pre-SmaI restriction buffer and 6.3l of mercaptoethanol (prepare on the day of use).
11. 10 TBE running buffer: 1M TrisHCl, 0.9M boric acid, and
0.01M EDTA.Dilute to 0.5 TBE with double-distilled (or
ultrapure) water (see Note 4).
12. Solution of ethidium bromide (1g/ml): add 25l of a
10mg/ml stock solution in 300ml of autoclaved distilled
water. The stock solution should be stored away from light
at 4C.
13. Loading buffer with bromophenol blue (6): 0.25% bromophenol blue and 40% (w/v) sucrose in double-distilled (or
ultrapure) water to a final volume of 10ml. Store at 4C.
14. Low melting temperature agarose solution for the DNA discs:
weigh out 0.15g of low melting temperature agarose (we use
SeaPlaque agarose), add 10ml of PIV, and heat at the microwave to dissolve completely. Transfer dissolved agarose to a
42C water bath where it should stay for at least 10min before
use (prepare just before use).
15. Low melting temperature agarose solution to seal the DNA
discs in the gel: to prepare sealing agarose, weigh out 0.075g
of low melting temperature agarose (we use SeaPlaque
agarose), add 10ml of 0.5 TBE, and boil on the microwave.
Keep at 42C until use.
16. SeaKem LE Agarose solution for the gel: 150ml of 1% SeaKem
LE Agarose. Weigh out 1.5g of agarose, and add 7.5ml of
10 TBE and 130ml of distilled water. Boil in microwave for
12min and mix in between. Adjust the final volume to 150ml
with distilled water. Put a magnetic stirrer bar into agarose; stir
slowly at room temperature, until not too hot for hands (about
50C); and pour immediately.
17. Running buffer: add 100ml of 10 TBE in a measuring cylinder and adjust the volume to 2L with distilled water.
2.2 Other
Materials, Equipments,
and Software
327
5. Digital gel documentation system to capture and image scanning after the pulsed-field gel electrophoresis (PFGE) (e.g.,
Gel Doc XR system and Quantity One 1-D analysis software,
BioRad) or conventional camera photography and a
UV-sensitive film.
6. Software for the analysis of DNA banding profiles and construction of dendrograms (e.g., BioNumerics, version 6.6,
Applied Maths, Belgium).
3 Methods
Carry out all procedures at room temperature unless otherwise
specified. Wear gloves during the protocol to avoid nuclease contamination of the DNA samples from the operators skin. Use sterile materials throughout (e.g., tubes, tips, etc.).
3.1 Growth
ofBacterial Cultures
3.2 Harvest
andWash ofCells
3.3 Adjustment
ofCell Concentration
328
IldaSantos-Sanches et al.
3.5 Lysis
andTreatment
withProteinaseK
1. Remove the agarose discs one by one (from the previous step)
into 15ml plastic tubes containing 1ml of EC lysis solution in
each (use disposable sterile loops to transfer the agarose discs
into the tubes; all 12 or more agarose discs containing the
same DNA go into the same tube) (see Note 7).
329
Side view
Deposit 20 l
agarose droplets
Microscope
slides
Side view of
the droplets:
Final presentation:
Microscope
slides
Glass plate coated
(wrapped) with parafilm
Fig. 1 Schematic drawing of DNA agarose discs preparation (adapted from Chung etal. [11]). Source: H. de
Lencastre, ITQB. Portugal)
330
IldaSantos-Sanches et al.
1. Transfer 1 DNA agarose disc into a 1.5ml microtube containing 150l of pre-SmaI restriction buffer.
2. Incubate at room temperature for 3040min and remove the
buffer with a pipette.
3. Add 45l of SmaI restriction buffer plus 20 units of SmaI
enzyme stock per DNA disc (check the equivalent volume in
l) (see Note 3).
4. Incubate at 25C overnight.
5. Add 5 l of loading buffer (the preparation stays at room
t
emperature while the gel is prepared; if the discs are not
immediately used, add 10l of ES to the preparation and store
the discs at 4C for up to 2 days).
3.7 Preparation
oftheGel andPFGE
Apparatus
331
it on the glass slip, and blot off excess liquid with a soft hand
paper towel; with the help of the loop, slide the agarose disc
into the first well. Make sure the disc is positioned at the bottom and front part of the well (see Note 8).
8. Repeat the process for other agarose DNA discs.
9. The PFGE lambda marker is used as a molecular weight
standard.
10. At the end, when all the wells are filled, seal them with the sealing agarose (be careful not to leave bubbles inside wells).
11. Put the assembled gel into the PFGE chamber and cover the
chamber with the lid.
12. Start running the program.
3.8 Staining
andPhotography
oftheGel
3.9 Computer-
Fingerprinting
Analysis
332
IldaSantos-Sanches et al.
Fig. 2 Representative image of SmaI and Cfr9I PFGE patterns of bovine streptococci (S. dysgalactiae subsp.
dysgalactiae). Capital letters B, C, D, E, F, G, I, and J on top of PFGE images are farm codes where isolates were
collected. Capital letters and numeral suffix under farm code represent the PFGE typesubtype patterns.
LADDER corresponds to lambda ladder PFG marker. (a) All PFGE profiles obtained among the 18 bovine
isolates using the endonuclease SmaI (one isolate from farm E was resistant to cleavage by endonuclease
SmaI). (b) PFGE profile of Cfr9I-digested genomic DNA of one isolate (of PFGE F-1) which was resistant to
cleavage by SmaI
Fig. 3 Representative image of a dendrogram of PFGE patterns of bovine streptococci (S. dysgalactiae subsp.
dysgalactiae) (adapted from Rato et al. (http://wwwnc.cdc.gov/eid/article/16/1/09-0632-f1)) [7]. The dendrogram was produced by using Dice coefficients and an unweighted pair group method using arithmetic averages
(UPGMA). Default clustering settings of 0% optimization (i.e., the relative distance an entire lane is allowed to
shift in matching attempts) and 1.5% band position tolerance were used
333
4 Notes
1. Solutions prepared from powdered pancreatic RNase (ribonuclease A from bovine pancreas, Sigma-Aldrich) products can be
made free of DNase by boiling. Prepare a 10mg/ml stock
solution in 10mM sodium acetate buffer, pH5.2. Heat to
100C for 15min, allow to cool to room temperature, and
then adjust to pH7.4 using 0.1 volumes of 1M TrisHCl,
pH7.4. Aliquot and store at 20C.
2. The deproteinization solution should be freshly prepared
before use.
3. The Cfr9I (XmaI) restriction enzyme (C^CCGGG) can be
used instead of SmaI (CCC^GGG) when DNAs are uncut by
SmaI.If Cfr9I is used, do the equilibration step of the protocol
as referred but with pre-Cfr9I restriction buffer (10mM Tris
HCl, pH7.2, 5mM MgCl2, 200mM sodium glutamate,
100 g/ml BSA) at 37C.For the restriction, add 45l of
Cfr9I restriction buffer plus 20 units of Cfr9I enzyme stock
per DNA disc and incubate at 37C overnight.
4. TBE buffer is prone to precipitation over time.
5. Remove supernatants carefully using a 5ml pipette with an
automatic pipette aid.
Caution: Pellets are easy to lose! Be careful when taking tubes
out of centrifuge. Remove the last amounts of supernatant
fluid using 1ml tips and corresponding pipette.
6. The formula is derived as follows:
OD620 200 Vadd + 210
=
5.0
210
where:
334
IldaSantos-Sanches et al.
Acknowledgments
The PFGE data mentioned in this chapter were obtained by Mrcia
Rato as part of her research leading to Ph.D. thesis. Fundao para
a Cincia e a Tecnologia (FCT) is acknowledge for the financial
support for the accomplishment of Mrcia Rato Doctoral Project
(SFRH/BD/32513/2006). The work was supported by the
grants Pest-OE/BIA/UI0457/2011-CREM (Centro de Recursos
Microbiolgicos) and PROC 60839, funded by Fundao para a
Cincia e Tecnologia, Portugal and Fundao Calouste Gulbenkian,
Portugal (respectively).
References
1. Tenover FC, Arbeit RD, Goering RV etal
(1995) Interpreting chromosomal DNA
restriction patterns produced by pulsed-field
gel electrophoresis: criteria for bacterial strain
typing. J Clin Microbiol 33:22332239
2. Van Belkum A, Tassios PT, Dijkshoorn L etal
(2007) Guidelines for the validation and application of typing methods for the use in bacterial epidemiology. Clin Microbiol Infect
13(suppl 3):146
3. Ericsson U, Lindberg A, Persson W etal
(2009) Microbial aetiology of acute clinical
mastitis and agent-specific risk factors. Vet
Microbiol 137:9097
4. Barkema HW, Green MJ, Bradley AJ, Zadoks
RN (2009) Invited review: the role of contagious disease in udder health. J Dairy Sci 92:
47174729
5. Muellner P, Zadoks RN, Perez AM etal
(2011) The integration of molecular tools into
veterinary and spatial epidemiology. Spat
Spattemporal Epidemiol 2:159171
6. Rato MG, Bexiga R, Nunes SF etal (2008)
Molecular epidemiology and population structure of bovine Streptococcus uberis. J Dairy Sci
91:45424551
Chapter 24
Multiple-Locus Variable-Number Tandem Repeat (VNTR)
Analysis (MLVA) Using Multiplex PCR and Multicolor
Capillary Electrophoresis: Application to the Genotyping
of Brucella Species
Giuliano Garofolo
Abstract
The multiple-locus variable-number tandem repeat (VNTR) analysis (MLVA) is a genetic typing method
based on the evaluation of the number of repeated sequences in multiple selected loci of microbial
DNA. Although several MLVA typing panels have been proposed for brucellae, the 16-loci panel is recognized as the standard genotyping method, also used for the Brucella international online database. This
chapter describes a high-throughput MLVA-16 protocol using multiplex PCRs and multicolor capillary
electrophoresis.
Key words MLVA, Capillary electrophoresis, Multiplex PCR, Brucella spp, Genotyping
Introduction
Brucellosis is a widespread animal infectious disease and probably
the worlds most common zoonosis. The Brucella genus currently
comprises six classical species (B. abortus, B. melitensis, B. suis, B. ovis,
B. canis, and B. neotomae) and four novel species (B. ceti, B. pinnipedialis, B. inopinata, and B. microti) [1]. New species have
been proposed in recent years. The highly monomorphic nature of
this genus, presenting few polymorphisms not only within species
but between species as well, makes strains particularly difficult to
differentiate using most genetic typing techniques. This makes
crucial public health tasks, such as diagnosis and strain typing, a
challenge for veterinary laboratories testing livestock and wildlife.
To date, multiple-locus variable-number tandem repeat (VNTR)
analysis (MLVA) [2] is recognized as one of the best genotyping
approaches for Brucella. Mutation rate variability between loci in
Brucella allows this method to discriminate closely related strains
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_24, Springer Science+Business Media New York 2015
335
336
Giuliano Garofolo
Materials
2.1 Laboratory
Plasticware
2.2 Laboratory
Reagents
337
338
Giuliano Garofolo
Table 1
MLVA primers sequences
Multiplex
PCR mixa
Locus
M1
Bruce30
119199
Bruce08
312384
Bruce11
2571076
Bruce45
133187
Bruce19
79205
Bruce06
312714
Bruce42
164914
Bruce12
302452
Bruce18
130186
Bruce55
193553
Bruce21
431463
Bruce04
313473
Bruce07
134246
Bruce09
124292
Bruce43
170194
Bruce16
227353
M2
M3
M4
Allele size
range (bp)c
a
For primer mixes including the forward primers targeting the Bruce18, Bruce42, and Bruce55 loci, equal concentrations of both labelled and unlabelled primers targeting these loci were added to the mix in order to avoid the posterior
production of excessive fluorescence during the capillary electrophoresis [6]
b
Fw, forward primer; Rw, reverse primer
c
Expected allele size ranges are given in base pairs. The test has been designed so that fragments would differ from one
another by either size, fluorescence, or both, to exclude the possibility of overlap in capillary electrophoresis results
339
340
Giuliano Garofolo
Software
3
3.1
Methods
PCR Setup
341
Table 2
PCR master mixes composition
Volume per reaction (l)a
Component
Final concentration
2 Type-it microsatellite
PCR Master Mix
Q solution
0.5
1050 ng
10
The volume of each component to add to the master mix corresponds to the volume per reaction of that component
multiplied by the total number of samples (including any positive and negative controls) plus 10 % of the original volume to compensate for eventual pipetting losses (DNA template solutions are not added to master mixes)
b
Primer mix for the specific multiplex PCR being prepared, primer solutions should be paired as follows: M1fw and
M1rw (for multiplex master mix 1M1), M2fw and M2rw (for multiplex master mix 2M2), M3fw and M3rw (for
multiplex master mix 3M3), and M4fw and M4rw (for multiplex master mix 4M4)
3.3 Preparation
of the Samples
for Capillary
Electrophoresis
342
Giuliano Garofolo
The following steps are adapted for ABI PRISM genetic analyzer
instruments.
1. Power the ABI PRISM genetic analyzer.
2. Run the data collection software.
3. Ensure that consumables (e.g., polymers, buffers) are not
expired and that buffer levels are at the fill lines.
4. Set the oven temperature to 60 C by clicking preheat.
5. Check for and remove any bubbles from the pump assembly.
6. Create a plate record and assign plate contents by entering the
following information:
(a) Sample names: be sure to name the samples in a way that
reflects their order in the 96-well plate.
(b) Assay, set run module parameters as follows: oven temperature (C) 60, run voltage (kV) 8.5, pre-run voltage (kV)
15, injection voltage (kV) 1.6, run time (sec.) 4900, prerun time (sec.) 180, injection time (sec.), and data delay
(sec.) 480.
(c) Assign file name convention to each sample: sample name,
wells, and capillary number.
(d) Results group: create a MLVA_Brucella folder.
7. Save the plate record.
8. Load the plate onto the instrument, access the plate record
created, and link the plate to its plate record.
343
10
553
91
85
79
11
887
12
13
15
220
331
134 142 150 158 166 174 182 190 198 206 214 222 230
124 132 140 148 156 164 172 180 188 196 204 212
227 235 243 251 259 267 275 283 291 299 307 315 323
119 127 135 143 151 159 167 175 183 191 199
Bruce07
Bruce09
Bruce16
Bruce30
425
422
1013 1076
14
Bruce04 313 321 329 337 345 353 361 369 377 385 393 401 409 417
Panel 2B
Bruce21
Bruce19
Bruce18
Panel 2A
Bruce45
635 698
Bruce12
Bruce11
509
Bruce08
17
19
20
21
22
23
18
24
337
353
246
437 452
16
205
25
529
26 27 28
Data pertaining to B. melitensis biovar 1 str. 16 M is underlined and given in bold; the 16 markers can be split into two groups: one comprises 8 minisatellite loci (panel 1), and the second group
contains 8 microsatellite loci (panel 2A and panel 2B). In this approach, the loci are divided into different panels containing increasing levels of genetic resolution: the MLVA-8 (panel 1), MLVA-11
(panels 1 and 2A), and MLVA-16 (panels 1, 2A, and 2B) (for the original source of the table, go to http://mlva.u psud.fr/brucella/IMG/pdf/bru_table_allele_assignement_version3.6.pdf)
Panel 1
Allelic
value
Table 3
Expected VNTR sizes per locus of Brucella sequences currently known and correspondent allelic valuea
345
Table 4
VNTR size by locus
Locus
Bruce06
134
Bruce08
18
Bruce11
63
Bruce12
15
Bruce42
125
Bruce43
12
Bruce45
18
Bruce55
40
Bruce18
Bruce19
Bruce21
Bruce04
Bruce07
Bruce09
Bruce16
Bruce30
346
Giuliano Garofolo
Cluster Analysis
Notes
1. The preparation of primer mix solutions should be performed
in a DNA-free room. Thaw primer stock solutions and place
on ice until use. Avoid the exposure of primers to light. In the
absence of amber-colored tubes, use aluminum foil to cover
the vials.
2. To minimize repetitive freeze-thaw cycles of primer stock solutions, prepare 10 batches of 10 primer mixes (volume 110 l)
every couple of months or if a drop in fluorescence is observed
in your old batch. This amount should be sufficient for 1000
PCRs, allowing MLVA on 200250 strains.
3. Other capillary electrophoresis machines may be used, but it is
recommended that fragment analysis be set to 1401,100 bp
and the proper reagents be used, as suggested by the
manufacturer.
347
4. In the first few runs, verify the dilution factor by running one
column at a time (M1M2, M3, and M4 fragments, respectively). If peak fluorescence intensity does not fall between 400
and 6,000 nm, correct the dilution to optimize your electrophoresis. Further PCRs will generate a similar pattern of fluorescence for all samples, provided that equal amounts of
purified DNAs are used.
5. For example, for a total of 32 DNA samples: 1082.4 l of
formamide [10.25 l 3 injections 32 (=984 l) + 10 % of this
volume (=98.4 l)] and 31.6 l of Liz 1,200 size standard
[0.3 l 3 injections 32 (=28.8 l) + 10 % of this volume
(=2.8 l)].
6. Each injection takes about 1.5 h. The analysis of a single plate
involves 12 injections and will thus take about 17 h.
7. A cooperative public online database, based on the MLVA-16
panel, was established with the aim of promoting the creation
of a global epidemiological map of Brucella and is freely accessible at http://mlva.u-psud.fr/brucella/.
8. MLVA genotypes are considered to be closely related if they
share 70 % or more of their VNTR alleles. For MLVA-16,
strains having between 11 and 16 alleles in common are said to
belong to the same species.
9. Further analysis may be recommended. Both free (e.g., Phyloviz
1.0) and paid (e.g., PAUP 4.0, Bionumerics) software are available for this purpose. UPGMA, neighbor-joining analyses, and
minimum spanning trees can be generated to assess fine and
broad relationships between the strains analyzed.
References
1. Pappas G (2010) The changing Brucella ecology: novel reservoirs, new threats. Int J
Antimicrob Agents 36:S8S11
2. Whatmore AM (2009) Current understanding
of the genetic diversity of Brucella, an expanding
genus of zoonotic pathogens. Infect Genet Evol
9:11681184
3. Al Dahouk S, Flche PL, Nckler K et al (2007)
Evaluation of Brucella MLVA typing for human
brucellosis. J Microbiol Methods 69:137145
4. Whatmore AM, Shankster SJ, Perrett LL et al
(2006) Identification and characterization of variable-number tandem-repeat markers for typing of
Brucella spp. J Clin Microbiol 44:19821993
Chapter 25
Multilocus Sequence Typing (MLST): Markers
for the Traceability of Pathogenic Leptospira Strains
Ahmed Ahmed, Ana S. Ferreira, and Rudy A. Hartskeerl
Abstract
Leptospirosis is a major zoonosis with worldwide distribution. Conventional serological typing is arduous
and time consuming. Genotyping is increasingly applied for the typing and identification of leptospires and
contributes to genetic and virulence divergence and molecular epidemiological characteristics such as host
versus leptospires population interactions and dynamics. Presently, multilocus sequence typing (MLST) is
the most robust approach. In this chapter, we describe the practical steps of two major multilocus sequence
typing methods for leptospires. The first method (denoted as the 6 L scheme) is based on genotyping by
phylogeny using concatenated sequences derived from six loci, including genes that encode outer membrane proteins and rrs and can be used for typing pathogenic species and strains of intermediate species.
The second method (referred to as the 7 L scheme) uses seven loci on housekeeping genes and allows the
analysis of seven major Leptospira pathogenic species. The 7 L scheme is web based and includes the option
to analyze sequence types (STs).
Key words Leptospira, Leptospirosis, Genotyping, MLST, Multilocus typing
Introduction
Leptospirosis is a hemorrhagic zoonotic disease with a worldwide
distribution presenting a major burden for the veterinary and public health. Leptospires, the causative agents, are long (620 m)
and thin (0.10.2 m) helically coiled gram-negative bacteria with
typically hooked ends from which each of the two endoflagella is
extruding. These bacteria belong to the family Leptospiraceae and
to the genus Leptospira. There are free-living saprophytic and
pathogenic leptospires, the latter requiring a host for survival.
Traditionally, saprophytic and pathogenic leptospires have been
denoted as Leptospira biflexa and Leptospira interrogans, respectively. There is a wide variety of sero- and genotypes of leptospires,
which are generally associated with a certain host. Hence, the availability of effective tools to identify infecting leptospires is of utmost
importance in both animal and human health fields, not only for
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_25, Springer Science+Business Media New York 2015
349
350
the diagnosis of the disease but also for assessing its epidemiology
and defining intervention strategies.
Traditionally, the serovar is the basic taxon of leptospires.
The serovar is a serological entity determined by the cross agglutinin absorption test (CAAT). To date more than 300 serovars, both
saprophytic and pathogenic, have been identified, separated into 26
serogroups on the basis of similarity in serological features. Since
the 1990s, speciation of leptospires based on genomic DNA homology has been introduced. This technique, involving heterologous
DNA hybridization, allows the speciation of members of Leptospira
based on the established criterion that >70 % homology under
defined stringency conditions corresponds to conspecificity. The
method is laborious and tedious, and presently it is only executed
at the Centers for Disease Control and Prevention (CDC), Atlanta,
USA. Currently, molecular approaches have identified 21 Leptospira
species, consisting of three clades comprising nine pathogenic, five
intermediate, and seven saprophytic species. The molecular and
serological classification of leptospires show little correlation.
In the past years, numerous restriction enzyme-generated or
PCR-based DNA fingerprinting methods have been described.
Among others, pulsed-field gel electrophoresis (PFGE), bacterial
endonuclease DNA analysis (BRENDA), and arbitrarily primed
PCR (AP-PCR) have been explored to genotype leptospires and,
eventually, to achieve molecular speciation, but these methods usually yield complex patterns and lack the ability to directly generate
digital data, show low reproducibility, and/or require cultures of
viable leptospires. Considering the increasing complexity and associated costs for the international shipment of pathogens, there is a
need for typing methods that generate electronically portable data
and online databases that can be easily accessed for data insertion
and comparison.
Phylogeny-based genotyping has been described for leptospires using sequences of several genes such as rrs (16S ribosomal
RNA), secY (translocase preprotein secY), gyrB (DNA gyrase subunit B), flaB (flagellar protein B), and rpoB (RNA polymerase
beta-subunit), some of these presenting a high discriminative
power. However, horizontal DNA transfer has been described for
Leptospira, and thus limitation to one locus holds the risk of misclassification. Therefore, the use of multiple loci for the genotyping of Leptospira is imperative. Multilocus sequence genotyping is
one of such methods and currently the most robust one. The first
Leptospira multilocus sequence genotyping system has been developed in the early 2000s [1] and was hampered at that time by a
scarcity of available working sequences from public databases. This
scheme uses six loci (6 L), including the rrs gene and two genes,
lipL32 and lipL41, coding for outer membrane proteins. This 6 L
scheme for leptospires was followed by additional MLST
approaches using seven loci (7 L) [24], while a theoretical
351
Materials
2.2 Multilocus
Sequence Typing
(6 L and 7 L Schemes)
ATCTCCGTTGCACTCTTTGC
ACCATCATCATCATCGTCCA
LipL32R
CCGTCCCTTAATTTTAGACTTCTTC
(alternative: CCTTCCTTTAATTTTAGACTTTTTC)f
secYR
LipL32F
ATGCCGATCATTTTTGCTTC
AGTTGAGCCCGCAGTTTTC
rrsR
secYF
CATGCAAGTCAAGCGGAGTA
GCATCGAGAGGAATTAACATCA
LipL41R
rrsF
TAGGAAATTGCGCAGCTACA
LipL41F
TTTTTTGAGATCCGCAGCTTT
(alternative: CTTTTTTGAGATCTCCGGCTTT)f
icdAR
1.5
1.5
1.5
1.5
1.5
1.5
MgCl2
(mM)c
474
548
549
541
520
674
674
531
557
Size of PCR
product (bp)
1667072166641
34594023458902
18625351862984
36036443604120
39809263980372
34587893458361
Location of the
sequence used to
define MLST locusd, e
432
501
450
477
555
429
Size of MLST
locus (bp)d
When using these alternative primers, the annealing temperature should be adjusted to 54 C in the PCR cycling conditions
adk (adenylate kinase), icdA (isocitrate dehydrogenase), lipL41 (outer membrane lipoprotein LipL41), rrs (16S rRNA), secY (preprotein translocase SecY protein), and lipL32
(outer membrane lipoprotein LipL32)
b
Original primers according to Ahmed et al. [1] and additional alternative primers for some loci according to Ahmed et al. [5]
c
Concentration of MgCl2 in the PCR mixture as adjusted for each primer pair
d
The location of the sequence used to define MLST locus and the size of MLST locus (bp) as modified in Ahmed et al. [5]
e
Nucleotide positions based in the published genome sequence of L. interrogans serovar Copenhageni strain Fiocruz L1-130 chromosome I (GenBank Accession Number
AE016823)
lipL32
secY
rrs
lipL41
GGGACGAGATGACCAGGAT
icdAF
ACGCAAGCTCCTTTTGAATC
(alternative: TTACACAAGCTCCCTTTGAAT)f
adkR
icdA
GGGCTGGAAAAGGTACACAA
(alternative: ACATTATCTTCATGGGACCTCC)f
adkF
adk
Sequence (5 to 3)b
Primer
Locus/
genea
Table 1
Loci and respective flanking oligonucleotide primers for the Leptospira 6 L MLST scheme
352
Ahmed Ahmed et al.
CAACTTGCGGAYATAGGAGGAG
ATTATGTTCCCCGTGAYTCG
caiB-R
TCCRTAACTCATAAAMGACAAAGG
mreA-RM
caiB-F
GGCTCGCTCTYGACGGAAA
AGAACACCCGCCGCAAAACAAT
pfkB-RM
mreA-FM
CGGAGAGTTTTATAARAAGGACAT
GTTTTACRGAACCHCCGTAGAGAAT
tpiA-RM
pfkB-FM
TTGCAGGAAACTGGAAAATGAAT
TCTTTTTTGAATTTTTGACG
sucA-RM
tpiA-FM
TCATTCCACTTYTAGATACGAT
sucA-FM
AAGAAGCAAGATCCACAAYTAC
pntA-RM
1.5
2.0
1.5
3.5
2.5
1.5
1.5
MgCl2
(mM)c
650
719
588
639
640
621
650
Size of PCR
product (bp)
15628451563246
27345502734116
13865531386984
16946731694248
12274741227920
5634756871
37849553784512
Location
of MLST locusd
402
435
432
426
447
525
444
Size of MLST
locus (bp)
Nucleotide positions based in the published genome sequence of L. interrogans serovar Lai strain 56601 chromosome I (GenBank Accession Number NC004342)
glmU (UDP-N-acetylglucosamine pyrophosphorylase), pntA (NADP transhydrogenase subunit alpha), sucA (2-oxoglutarate dehydrogenase decarboxylase component), tpiA
(triosephosphate isomerase), pfkB (ribokinase), mreA (rod-shape-determining protein rodA), and caiB (carnitine dehydratase)
b
Primers according to Boonsilp et al. [4]
c
Concentration of MgCl2 in the PCR mixture as adjusted for each primer pair
caiB
mreA
pfkB
tpiA
sucA
TAGGAAARATGAAACCRGGAAC
pntA-FM
AGTTTTTTTCCGGAGTTTCT
glmU-RM
pntA
AGGATAAGGTCGCTGTGGTA
glmU-FM
glmU
Sequence (5 to 3)b
Primer
Locus/
genea
Table 2
Loci and respective flanking oligonucleotide primers for the Leptospira 7 L MLST scheme
354
Methods
3.1 6 L MLST
Scheme
355
356
Notes
1. The 6 L MLST scheme was originally developed by Ahmed
et al. [1], for genotyping a broad range of pathogenic leptospires. This scheme does not conform to the original concept
of MLST as it includes a non-housekeeping gene (rrs) and
genes that encode cell surface proteins (LipL32 and LipL41).
Later, in 2011, Ahmed et al. [5] designed four additional
alternative primers for use with the 6 L scheme (see Table 1).
These primers should be used in a repeat PCR reaction in the
event that the original primers fail to generate an amplicon.
2. The 7 L MLST scheme was originally described by
Thaipadungpanit et al. [2], in 2007, primarily for genotyping
isolates of L. interrogans. The scheme followed a conventional
strategy for MLST by selecting seven housekeeping genes that
were distributed around the genome and were not under positive selection. However, the primers used for this scheme
worked variably for other leptospiral species. This scheme was
recently further expanded by Boonsilp et al. [4], by updating
primers sequences and replacing one of the analyzed locus.
Therefore, as presented in this chapter, the modified 7 L
MLST scheme can be used to genotype the seven pathogenic
species of Leptospira linked to the overwhelming majority of
disease: L. interrogans, L. kirschneri, L. borgpetersenii, L. noguchii, L. santarosai, L. weilii, and L. alexanderi.
3. Defrost the components needed for preparing the PCR mixtures at room temperature but maintain the DNA polymerase
at 20 C (or in ice) and add it to the mixtures in the last.
Prepare the required master PCR mixtures (one for each
357
358
359
6.
7.
8.
9.
Chapter 26
Single-Nucleotide Polymorphism Discrimination
Using High-Resolution Melting Analysis
for the Genotyping of Bacillus anthracis
Sylviane Derzelle
Abstract
High-resolution melting (HRM) is a post-PCR technique that determines with high precision the melt
profile of PCR products using a new generation of double-stranded DNA-binding dyes and accurate fluorescence data acquisition over small temperature increments. The method can be used to interrogate small
sets of single-nucleotide polymorphisms (SNPs). Here, we describe a simple and cost-effective HRMbased method for the screening of 14 phylogenetically informative SNPs within the genome of Bacillus
anthracis that subtype the species into 13 major sublineages or subgroups. Fourteen monoplex and seven
duplex SNP-discrimination assays have been designed. We detail the parameters most important for the
successful application of HRM for B. anthracis genotyping.
Key words High-resolution melting (HRM), Single-nucleotide polymorphism (SNP), Genotyping,
Phylogeny, Discrimination, Diversity, Bacillus anthracis
Introduction
Bacillus anthracis, the etiological agent of anthrax, is a sporeforming, Gram-positive bacterium belonging to the B. cereus
group. Although all mammals are known to be susceptible to
anthrax, the bacterium primarily affects wild and domesticated herbivores, causing acute, often fatal disease. Transmission to animals
typically occurs through the gastrointestinal tract. Ruminants
become infected by ingestion of soil-borne spores while browsing
or grazing [1]. Anthrax has been one of the infectious diseases with
major mortality among livestock for centuries. It is nowadays considered as a sporadic disease in much of Western Europe, Northern
America, and Australia, but anthrax remains endemic in many
countries and particularly in Africa [24].
B. anthracis is a monomorphic pathogen with extremely low
genetic variability [5, 6]. Due to this lack of diversity, only modern
molecular characterization techniques with high discrimination
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_26, Springer Science+Business Media New York 2015
361
362
Sylviane Derzelle
B - lineage
B.Br CNEVA
B.Br 001/002
A - lineage
B.BrKruger
A.Br 005/006
A.Br 003/004
A.Br Vollum
A.Br Aust94
A.Br 001/002
A.Br Ames
A.Br 008/011
A.Br 011/009
A.Br 008/009
A.Br WNA
C - lineage
C.Br A1055
Fig. 1 Phylogeny of the major groups (in bold) and sublineages (marked by stars)
of B. anthracis (modified from [8] and [9])
363
2
2.1
Materials
DNA Preparation
2.2
PCRHRM Mix
BA2
BA3
BA4
BA5
BA6
BA7
BA8
BA9
BA10
BA11
BA12
BA13
BA14
A.Br.002
A.Br.003
A.Br.004
A.Br.006
A.Br.007
A.Br.008
A.Br.009
B.Br.001
B.Br.002
B.Br.003
B.Br.004
A/B.Br.001
A.Br.011
2552486
3697886
69952
1494269
1056740
1455279
2589823
3947248
266439
162509
3600659
1493157
947760
182106
G to A
A to G
T to C
G to A
G to T
T to C
A to G
T to G
T to C
C to A
T to C
A to G
G to A
T to C
Base
change
CGAATTCCCGCTGAAAATAA
AAAATCGGAATTGAAGCAGGA
ATTCCAATCGCTGCACTCTT
CCCCGATAATTTTCACAAAGC
TGCTTGGGTAACCTTCTTTACTT
AGAATAAAATGAAGATAATGACAAACG
ATTCGCATAGAAGCAGATGAGC
TCAAGTTCATAACGAACCATAACG
GCACCTTCTGTGTTCGTTGTT
TTCACCGAATGGAGGAGAAG
GCACGGTCATAAAAGAAATCG
TGTTCAAAAGGTTCGGATATGA
AATCGGCCACTGTTTTTGAAC
AGGTATATTAACTGCGGATGAT
CCAAACGGTGAAAAAGTTACAAA
GCAACTACGCTATACGTTTTAGATGG
TTACAAGGTGGTAGTATTCGAGCTG
TTGGTAACGAGACGATAAACTGAA
GCGTTTTTAAGTTCATCATACCC
ATGTTGTTGATCATTCCATCG
ATCGCCGTCATACTTTGGAA
GGAATTGGTGGAGCTATGGA
AAAGGAATTTAGATTTTCGTGTCG
ATAAAAACCTCCTTTTTCTACCTCA
GCAGAAGGAGCAAGTAATGTTATAGGT
CCTAAAATCGATAAAGCGACTGC
GTGGTAAGGCAAGCGGAAC
ACGGTTTCCCTTTATCATCG
Localization on the Bacillus anthracis str. Ames Ancestor chromosome (accession no. NC_003997.3)
BA1
A.Br.001
canSNP name
canSNPa
location
Table 1
Set of 14 canonical SNPs and primer sequences
50
59
62
59
68
75
55
80
67
54
53
58
62
76
Size
(bp)
Pair n
1
2
3
3
4
4
5
5
2
1
6
6
7
7
Duplex
concentration
0.2 M
0.15 M
0.2 M
0.15 M
0.2 M
0.2 M
0.2 M
0.25 M
0.2 M
0.15 M
0.2 M
0.3 M
0.2 M
0.2 M
364
Sylviane Derzelle
365
Equipment
Methods
3.1 Preparation
of DNA Samples
and Biosafety
Procedures
3.2
Assay Design
366
Sylviane Derzelle
A.Br.001/002
B.Br.CNEVA
A.Br.005/006
B.Br.CNEVA
A.Br.005/006
Temperature (C)
A.Br.001/002
BA12
B.Br.CNEVA
A.Br.005/006
A.Br.001/002
Difference
post-melt Area
pre-melt Area
p
Fluorescence
Temperature (C)
A.Br.001/002
Temperature (C)
Normalized Fluorescence (%)
Temperature (C)
Difference Plot
Difference
post-melt Area
Fluorescence
pre-melt Area
p
B.Br.CNEVA
A.Br.005/006
BA11
Temperature (C)
Temperature (C)
Fig. 2 Representative melt curves and HRM difference plots obtained for three canSNP genotypes (B.Br.CNEVA,
A.Br.005/006, and A.Br.001/002) by HRM using the canSNP B.Br.004 (BA12) singleplex assay (a) or B.Br.003
and B.Br.004 (pair BA11 and BA12) duplex assays (b). Data and plots were produced by the ViiA7 instrument
using the ViiA7 v1.2. HRM software (Life Technologies)
367
368
Sylviane Derzelle
Table 2
canSNP signatures defining B. anthracis sublineages and subgroups and representative melting
temperatures (Tm) for each SNP allele obtained by HRM
Mean
Tm (C)
C.Br.A1055
B.Br.KrugerB
B.Br.001/002
B.Br.CNEVA
A.Br.Ames
A.Br.001/002
A.Br.Aust94
A.Br.003/004
A.Br.Vollum
A.Br.005/006
A.Br.008/009
A.Br.008/011
A.Br.011/009
A.Br.WNA
A.Br.001 BA1
75.6 76.4
A.Br.002 BA2
77.5 78.3
A.Br.003 BA3
72.0 72.9
A.Br.004 BA4
76.6 77.6
A.Br.006 BA5
76.4 77.3
A.Br.007 BA6
73.9 74.9
A.Br.008 BA7
75.0 75.9
A.Br.009 BA8
78.3 nd
B.Br.001 BA9
74.6 nd
canSNP sublineage
C/G allele
A/T allele
canSNP name
tT
A/B.
BA13 74.5 nd
Br.001
canSNPs that define a particular sublineage or subgroup are underlined and the corresponding allele is indicated in
bold. nd: not determined
a
Tm value of 71.5 C (tT) instead of 72.5 C (cT) due to a second SNP (C to T) located 5 to the B.Br.004 canSNP in
strains affiliated to the A.Br.005/006 subgroup
Notes
1. The following points should be considered when preparing
DNA for HRM:
(a) Use ideally the same extraction procedure to prepare all
DNA samples to be analyzed via HRM. This eliminates
any subtle differences that might be introduced by the
reagent components in the final elution buffers.
(b) Quantify all DNA samples and adjust to the same
concentration.
369
370
Sylviane Derzelle
371
Chapter 27
Clustered Regularly Interspaced Short Palindromic
Repeats (CRISPRs) Analysis of Members
of the Mycobacterium tuberculosis Complex
Ana Botelho, Ana Canto, Clia Leo, and Mnica V. Cunha
Abstract
Typical CRISPR (clustered, regularly interspaced, short palindromic repeat) regions are constituted by
short direct repeats (DRs), interspersed with similarly sized non-repetitive spacers, derived from transmissible genetic elements, acquired when the cell is challenged with foreign DNA. The analysis of the structure, in number and nature, of CRISPR spacers is a valuable tool for molecular typing since these loci are
polymorphic among strains, originating characteristic signatures. The existence of CRISPR structures in
the genome of the members of Mycobacterium tuberculosis complex (MTBC) enabled the development of
a genotyping method, based on the analysis of the presence or absence of 43 oligonucleotide spacers separated by conserved DRs. This method, called spoligotyping, consists on PCR amplification of the DR
chromosomal region and recognition after hybridization of the spacers that are present. The workflow
beneath this methodology implies that the PCR products are brought onto a membrane containing synthetic oligonucleotides that have complementary sequences to the spacer sequences. Lack of hybridization
of the PCR products to a specific oligonucleotide sequence indicates absence of the correspondent
spacer sequence in the examined strain. Spoligotyping gained great notoriety as a robust identification and
typing tool for members of MTBC, enabling multiple epidemiological studies on human and animal
tuberculosis.
Key words Tuberculosis, Bovine tuberculosis, Molecular typing, Direct repeats, Spoligotyping,
Epidemiology
Introduction
Clustered regularly interspaced short palindromic repeat (CRISPR)
regions are present in the genome of almost all archaea and many
bacteria, in association with cas (CRISPR-associated system) genes,
which are involved in DNA recombination and repair [1]. The
origin of cas in prokaryotic genomes is related with the acquisition
of heritable immunity against mobile nucleic acid elements, limiting phage infection and horizontal gene transfer of plasmids [2, 3].
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_27, Springer Science+Business Media New York 2015
373
374
Cas genes
Leader sequence
DVR 5
DR
DRa
DR
DRb DRa
DR
DRb DRa
DR
DRb DRa
DR
DRb DRa
DRb
DR
DRa
DRb
c
Multi-sized PCR products
IS
Strain I
Strain III
Strain II
Strain III
Strain IV
Spacers 1 to 43
Strains
1 2 3 4 5............43
Strain I
Strain II
Strain III
Strain IV
Fig. 1 Schematic structure of a CRISPR/Cas region in MTBC strains and application of the CRISPR array, to the
development of the spoligotyping method, using 43 selected spacer sequences. (a) General structure of a
CRISPR/Cas region; (b) structure of DR region: 056 copies of 36 bp direct repeats, separated by variable
spacer sequences of 3651 bp, whose order is well conserved in the genome of MTBC. Spacers, here numbered from 4 to 8. The DRs together with the adjacent spacers form the direct variant repeats (DVRs); (c) PCR
amplification of the DR flanking spacers, using primers DRa and DRb, one of them biotin labeled, complementary to the conserved direct repeats; (d) five pretend patterns in the DR region and possible formation of strain
III pattern by insertion of IS6110 sequence in DVR 5, hampering the amplification of spacer 5 by loss of primer
DVRb hybridization site. (e) Simplified spoligotyping membrane results obtained for the five strains and example of a real spoligotyping membrane. Note that the genome spacer order/number does not correspond to the
membrane spacer order/number
375
376
Membrane
Membrane
Multi-sized PCR
products biotinlabelled
NH3
COOH
Membrane
Spacers 1 to 43
1 2 3 4 5 .......................................43
. Hybridization
2. Hybridization
PCR products-biotin
Membrane
. Detection
Streptavidin-peroxidase
Spacers 1 to 43
Strains
Membrane
O2+H2O
Oxidized product
Light
Membrane
Fig. 2 Schematic basic steps of spoligotyping method. See text for details
reverse line blot hybridization, gained wide diffusion, due to its easy
execution and high throughput, to assess global MTBC strain
diversity. This technique consists in the amplification of the DR
region using two inversely orientated primers, one of them labeled
with biotin, complementary to the flanking conserved DR sequence
(Figs. 1 and 2). DNA spacer sequences in between adjacent DRs
and in between more distantly positioned DRs are amplified [8].
Evaluation of the presence or absence of spacers is done by reverse
hybridization of the obtained PCR products with a set of selected
43-spacer oligonucleotides covalently linked to a nylon membrane
(Table 1, Fig. 2), designed according to the sequences of M. tuberculosis H27Rv and M. bovis BCG P3 strains. The PCR products are
applied onto the membrane in reverse orientation of the rows with
the synthetic oligonucleotides. Since one of the DR primers is
labeled with biotin, the detection of hybridization is done by
chemiluminescence, through addition of streptavidin-peroxidase
conjugate and a substrate (Fig. 2).
Spoligotyping applied to bacterial isolates is simple, robust,
and highly reproducible. On the other hand, for MTBC strains
containing few copies of the IS6110 element, such as M. bovis,
spoligotyping is generally more discriminatory than IS6110 typing
methods, and the DR region is, consequently, also more stable.
377
Table 1
Sequences and concentrations of the oligonucleotide probes covalently linked to the Biodyne C
membrane [8]
Oligonucleotide
number order
Concentration
(pmol/150 l)
ATAGAGGGTCGCCGGTTCTGGATCA
12.5
CCTCATAATTGGGCGACAGCTTTTG
30.0
CCGTGCTTCCAGTGATCGCCTTCTA
12.5
ACGTCATACGCCGACCAATCATCAG
12.5
TTTTCTGACCACTTGTGCGGGATTA
12.5
CGTCGTCATTTCCGGCTTCAATTTC
12.5
GAGGAGAGCGAGTACTCGGGGCTGC
25.0
CGTGAAACCGCCCCCAGCCTCGCCG
50.0
ACTCGGAATCCCATGTGCTGACAGC
12.5
10
TCGACACCCGCTCTAGTTGACTTCC
15.0
11
GTGAGCAACGGCGGCGGCAACCTGG
30.0
12
ATATCTGCTGCCCGCCCGGGGAGAT
60.0
13
GACCATCATTGCCATTCCCTCTCCC
12.5
14
GGTGTGATGCGGATGGTCGGCTCGG
30.0
15
CTTGAATAACGCGCAGTGAATTTCG
30.0
16
CGAGTTCCCGTCAGCGTCGTAAATC
12.5
17
GCGCCGGCCCGCGCGGATGACTCCG
100.0
18
CATGGACCCGGGCGAGCTGCAGATG
12.5
19
TAACTGGCTTGGCGCTGATCCTGGT
12.5
20
TTGACCTCGCCAGGAGAGAAGATCA
12.5
21
TCGATGTCGATGTCCCAATCGTCGA
25.0
22
ACCGCAGACGGCACGATTGAGACAA
12.5
23
AGCATCGCTGATGCGGTCCAGCTCG
50.0
24
CCGCCTGCTGGGTGAGACGTGCTCG
50.0
25
GATCAGCGACCACCGCACCCTGTCA
25.0
26
CTTCAGCACCACCATCATCCGGCGC
12.5
27
GGATTCGTGATCTCTTCCCGCGGAT
25.0
28
TGCCCCGGCGTTTAGCGATCACAAC
12.5
29
AAATACAGGCTCCACGACACGACCA
12.5
30
GGTTGCCCCGCGCCCTTTTCCAGCC
12.5
(continued)
378
Table 1
(continued)
Oligonucleotide
number order
Concentration
(pmol/150 l)
31
TCAGACAGGTTCGCGTCGATCAAGT
12.5
32
GACCAAATAGGTATCGGCGTGTTCA
25.0
33
GACATGACGGCGGTGCCGCACTTGA
34
AAGTCACCTCGCCACACCGTCGAA
25.0
35
TCCGTACGCTCGAAACGCTTCCAAC
12.5
36
CGAAATCCAGCACCACATCCGCAGC
12.5
37
CGCGAACTCGTCCACAGTCCCCCTT
12.5
38
CGTGGATGGCGGATGCGTTGTGCGC
25.0
39
GACGATGGCCAGTAAATCGGCGTGG
25.0
40
CGCCATCTGTGCCTCATACAGGTCC
12.5
41
GGAGCTTTCCGGCTTCTATCAGGTA
12.5
42
ATGGTGGGACATGGACGAGCGCGAC
25.0
43
CGCAGAATCGCACCGGGTGCGGGAG
50.0
100.0
379
Table 2
Spoligotyping patterns and signatures of members of the MTBC [14, 21, 22]
Materials
All the solutions should be freshly prepared, with ultrapure water
(18.2 M cm at 25 C) and analytical grade reagents, and stored
at room temperature. The stock solutions should be sterilized in
autoclave at 121 C for 15 min. The nylon membrane can be
regenerated and reused up to ten times.
2.1
Solutions
1. 0.5 M EDTA: For 1,000 ml, weight 186.12 g of ethylenediaminetetraacetic acid (EDTA) and add half the water. Mix
until complete dissolution using a magnetic stirrer and adjust
to pH 8.0. Complete the volume to 1,000 ml (see Note 1).
2. 20 SSPE (w/v): Weight 35.6 g of Na2HPO4.2H2O, 210.24 g
of NaCl, and 7.4 g of EDTA for a final volume of 1,000 ml in
water. Mix well and adjust to pH 7.4.
380
For Hybridization
2.2 Membrane
Preparation
381
2.4 DNA
Hybridization
382
2.5 Detection
of Hybridizing DNA
Methods
Always use gloves and forceps to manipulate the membrane.
3.1 Preparation
of In-House Membrane
with the 43
Oligonucleotides
Covalently Linked
(See Note 7)
1. Dilute each oligonucleotide probe in the recommended optimized concentrations (see Table 1) in 150 l of 500 mM
NaHCO3 (pH 8.3).
2. Incubate the nylon membrane in a laboratory tray containing
10 ml of 16 % (v/v) EDAC solution for 10 min at room temperature to activate the membrane.
3. Discard the EDAC solution and wash the membrane with
ultrapure water for 2 min, with orbital shaking.
4. With the forceps, place the membrane on top of the foam
cushion of the Miniblotter system and hand-tight the screws.
5. Remove all the residual water of the membrane by aspiration
with the vacuum pump (see Note 8).
6. Fill the slots of the Miniblotter with 150 l of the 43 diluted
oligonucleotide probes, in the order displayed on Table 2,
leaving the first and the last slots empty (see Note 9).
7. Fill the first and the last slots of the Miniblotter with the diluted
China ink.
8. Incubate for 2 min at room temperature.
9. Remove all the solutions of the slots of the Miniblotter by aspiration with the vacuum pump, by the same order in which they
were filled.
383
10. Remove the membrane from the Miniblotter using forceps and
incubate in a laboratory tray, with 250 ml of 100 mM NaOH
for 8 min at room temperature with shaking.
11. Discard the solution and wash the membrane with ultrapure
water at room temperature.
12. Wash the membrane with 250 ml of 2 SSPE/0.1 % SDS with
gentle shaking for 5 min at 60 C.
13. The membrane is ready to use or to store.
14. Before storing the membrane, wash it in a laboratory tray, with
100 ml of 20 mM EDTA (pH 8.0), for 15 min at room temperature with gentle shaking.
15. Remove the membrane with forceps and store at 4 C, in a
sealed plastic bag to avoid dehydration, with 10 ml of 20 mM
EDTA (pH 8.0) until being used.
3.2 Sample
Preparation
(In a Biosafety
Level 3 Facility)
3.3 DR Region
Amplification by PCR
1. Prepare PCR reaction mix in a total volume of 50 l per reaction. Mycobacterium tuberculosis H37rv (CCUG 37357) and
Mycobacterium bovis BCG (CCUG 27863) are used as positive
controls and ultrapure water as negative control.
384
3.5 Detection
of Hybridized DNA
385
3.6 Interpretation
of Results
386
Clear sharp signals are easy to interpret, but some weak signals
may occur with some spacers, mainly 14, 26, and 33, whose presence or absence is more critical to evaluate. Comparing with
well-known control patterns and checking for the presence of the
expected spots can clarify some doubts. The negative control
should be absolutely clear and with no contamination or leaks.
Very faint dots should, in principle, not be considered, unless the
positive control gives otherwise information. Obtaining a clear
negative control and a correct positive control profile is critical for
the assessment of membrane quality and for the interpretation of
results.
3.7 Regeneration
of the Nylon
Membrane for Reuse
Notes
1. To prepare 1,000 ml of 0.5 M EDTA, weight 186.12 g of
EDTA to a glass bottle and add 800 ml of water. It is convenient to dissolve the powder using a magnetic stirrer with maximum speed. Add NaOH pellets to adjust the solution at a
pH 8.0. Only a pH 8.0 solution, at 25 C, completely dissolves
the EDTA. Adjust the volume to 1,000 ml with water and mix
again. Store at room temperature.
2. To prepare 100 ml of 10 % SDS, weight 10 g of sodium lauryl
sulfate to a glass bottle and add 80 ml of water. Dissolve by
gentle shaking (to avoid the formation of foam) in a magnetic
stirrer with heating up to 68 C. Adjust the volume to 100 ml
with water and mix again. Store at room temperature.
3. Prepare the solutions of 2 SSPE/0.1 % SDS and 2
SSPE/0.5 % SDS firstly mixing the water with the 2 SSPE.
After complete homogenization, add the SDS and homogenize again. The addition of the solutions should be as indicated
to avoid the formation of a precipitate.
387
388
14. Carefully wrap the membrane and the mesh, with the size of
the membrane, together and place them inside the rolling
bottle. Unroll the membrane and the mesh in the opposite
rotation direction to adhere completely at the rolling bottle
walls, avoiding bubbles and promoting the contact of the membrane with the streptavidin solution. Place the rolling bottle
in the hybridization oven, in the proper rotation direction, in
order to prevent the curl of the membrane during the incubation period.
15. To prepare the autoradiography cassette, place a sheet of blotting paper 3 MM and acetate sheet inside the cassette and hold
them individually with adhesive tape.
16. Incubate the membrane at room temperature, in the dark
room, with the ECL mixture with gentle shaking in order to
cover the entire membrane surface.
17. Exposure time can be adjusted according to the results obtained
and the intensity of signals. Shorter or longer exposures may
vary from 5 to 20 min, depending on the results from the first
film.
18. Leave the hyperfilm in the X-ray developer solution the time
enough (could be a few seconds) to visualize positive signals,
avoiding darkness of the hyperfilm. Once the hyperfilm starts
to get dark, wash immediately in water.
Acknowledgments
This work was supported by Fundaco para a Cincia e Tecnologia
(FCT) through project PTDC/CVT/117794/2010 and in the
framework of Projecto 3599Promover a Produo Cientfica e
Desenvolvimento Tecnolgico e a Constituio de Redes Temticas.
References
1. Jansen R, van Embden J, Gaastra WL, Schouls
M (2002) Identification of genes that are associated with DNA repeats in prokaryotes. Mol
Microbiol 43:15651575
2. Barrangou R, Fremaux C, Deveau H et al
(2007) CRISPR provides acquired resistance
against viruses in prokaryotes. Science 31:
17091712
3. Deveau H, Garneau JE, Moineau S (2010)
CRISPR/Cas system and its role in phagebacteria interactions. Annu Rev Microbiol
64:475493
4. Pourcel C, Salvignol G, Vergnaud G (2005)
CRISPR elements in Yersinia pestis acquire
new repeats by preferential uptake of bacte-
8.
9.
10.
11.
12.
13.
14.
389
Chapter 28
Rapid Microarray-Based Genotyping of Chlamydia spp.
Strains from Clinical Tissue Samples
Konrad Sachse and Anke Ruettger
Abstract
Pathogenic Chlamydia (C.) psittaci and C. trachomatis strains can be genotyped based on variations in the
ompA genomic locus. In the present chapter, we describe rapid genotyping assays for both chlamydial
agents using the ArrayStrip (AS) microarray platform. The test is targeting multiple discriminatory sites
in the variable domains of the ompA gene by using 35 (C. psittaci) and 61 (C. trachomatis) oligonucleotide
probes representing genotype-specific polymorphisms. In addition to discrimination among the established genotypes, this approach allows identification of atypical strains that were not accessible to typing
using previously established techniques, such as PCR-RFLP or serotyping.
The present DNA microarray assay can be conducted directly on clinical tissue samples and is suitable
for tracing epidemiological chains and exploring the dissemination of particular genotypes. The procedure
is easy to handle and economically affordable, and it allows genotyping of up to 32 clinical samples per day,
thus lending itself for routine diagnosis as well.
Key words Chlamydia psittaci, Chlamydia trachomatis, ompA genotyping, Diagnostic DNA microarray
test, Direct testing of clinical samples
1
1.1
Introduction
History of Typing
Members of the family Chlamydiaceae represent obligate intracellular bacteria that are distinguished by a characteristic biphasic
developmental cycle. There are several well-established pathogens
among them. For instance, Chlamydia (C.) psittaci, the causative
agent of psittacosis in birds and humans, is responsible for outbreaks in psittacine birds and domestic poultry [1], as well as cases
of atypical pneumonia in exposed individuals [2, 3]. The fact that
C. psittaci is a heterogeneous taxon in terms of host range and
virulence entailed the development of a serotyping scheme in the
1990s [4]. Strains of the agent were assigned to serovars A, B, C,
D, E, and F on the basis of their immune reaction with a panel of
monoclonal antibodies (MAbs) recognizing specific epitopes of
the major outer membrane protein (MOMP). Later on, Sayada
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_28, Springer Science+Business Media New York 2015
391
392
393
Table 1
ompA genotypes and subtypes of Chlamydia psittaci [Adapted from reference [14]]
Genotype
Known hosts
Subtypes
A-VS1
VS1, MN Zhang
AF269281
A-6BC
6BC
X56980
A-8455
84-55
Y16561
CP3
AF269265
L25436
Turkey, human
D-NJ1
NJ1
AF269266
D-9 N
9N
EF375557
X12647
EB-E30
WS/RT/E30
AY762613
EB-859
06-859/1
EU159263
EB
Duck, human
EB-KKCP KKCP-1
AB284062
Psittacine
VS225
AF269259
M56
M56
AF269268
WC
Cattle
WC
AF269269
1 Va
Crow
1V
EF028916
Crow
6N
EF197820
Mat116
Psittacine, budgerigar
Mat116
AB284058
R54a
Antarctic skua
R54
AJ243525
Psittacine
Daruma-1981
AB284065
Stork
CPX0308
AB284064
6N
a
a
YP84
a
a
CPX0308
a
394
2
2.1
Materials
DNA Extraction
2.2 Biotinylation
PCR and Agarose Gel
Electrophoresis
395
Table 2
Primers for ompA genotyping of C. psittaci and C. trachomatis
Designation
C. psittaci
VD1-fw
VD2-rev
201CHOMP
ompA-rev
418 bp
570 bp
C. trachomatis
Trach-VD1-fw 5-Bio-ACC AAG CCT TAT GAT CGA C-3
326 bp
572 bp (with
Trach-VD1-fw)
231 bp
2.4 General
Equipment
and Consumables
396
Methods
3.1
DNA Extraction
3.2
Biotinylation PCR
397
3.4
Data Processing
398
Notes
1. Genotyping analysis using the AS assay can be accomplished
within a working day (68 h). This renders the procedure
faster than PCR-RFLP and ompA sequencing. Furthermore,
AS genotyping is easy to handle as it mainly involves standard
laboratory equipment and requires less hands-on time than
PCR-RFLP and less experience than sequencing. The expenses
for consumables are moderate, and the equipment costs considerably less (about 25 %) than that for DNA sequencing. The
microarray technology is manageable in a high-throughput
environment, with up to 32 samples per day and technician.
2. Amplification can also be accomplished using standard PCR
reagents and preparing the reaction mixes in the usual way.
The main arguments in favor of using the present kit are that it
is convenient to use as it contains all reagents and that the
commercial mix is optimized for high yield.
3. Alternatively, buffers can be prepared manually according to
the protocols published previously [14, 15].
4. For the sake of high specificity and stringency of the hybridization reaction, it is extremely important to ensure that the prescribed temperatures are actually attained in the AS vessel itself.
This requires heat transfer from the heatable shaker to the
liquid contents of the AS vessel to be very rapid and efficient.
Most of the commercially available shakers are too slow, so that
too much time (and, consequently, specificity) is lost until the
necessary in-tube temperature has been reached. In our hands,
the Bioshake iQ fulfills the criteria to be used in the hybridization process.
5. The PatternMatch algorithm compares the experimentally
obtained signal pattern with those of reference strains representing the genotypes of C. psittaci or C. trachomatis, respectively.
It provides combined bar diagrams of sample and reference (see
an example in Fig. 1), as well as two numerical parameters to
assess the similarity of two patterns and the accuracy of identification. The matching score (MS) represents the sum of numerical differences between corresponding signal intensities of
sample and theoretical and/or practical reference hybridization
patterns. Thus, the MS value is a measure of the overall dissimilarity between two hybridization patterns. An ideal match of
two patterns based on the same set of oligonucleotide probes
would yield MS = 0, but practical values can be in the order of
100. Prior to the examination of field samples, the user should
run a series of samples of known genotypes in order to determine the practical MS range and set the upper threshold,
above which the best match is no more valid for genotyping.
399
Fig. 1 Identification of the ompA genotype of the C. psittaci strain from the feces sample of a diseased pigeon
(sample ID 11G1376). Using the PatternMatch function of the software, the experimentally obtained hybridization pattern of the sample (black bars) is compared to the closest match of a panel of reference strains, i.e., type
strain CP3 of genotype B (gray bars). The matching score (MS) of 154.12 is a measure of dissimilarity between
sample and reference, and the Delta MS value of 1.2 represents the numerical difference between best and
second best match. These values confirm that the assignment of the present sample to genotype B is valid
400
Chapter 29
Multiplexed Genotyping ofBacillus anthracis by Luminex
xMap Suspension Array
SimonThierry andSylvianeDerzelle
Abstract
The Luminex xTAG technology is a medium to high throughput, open methodology able to test many single
nucleotide polymorphisms (SNPs) in a single reaction and a minimum time. Multiplex SNPs interrogation are
conducted on the Luminex xMAP system, which uses lasers to read universal tag, color-coded microspheres
that attach to specific nucleic acid sequences. The present method describes a Multiplex Oligonucleotide
Ligation-PCR procedure (MOL-PCR) for the simultaneous interrogation of 13 phylogenetically informative
SNPs within the genome of Bacillus anthracis. The reported 13-plex assay enables efficient B. anthracis genotyping into major sublineages and groups. While cost-effective compared to other monoplex methods, the
present MOL-PCR method also offers a high degree of flexibility and scalability. It can easily accommodate
newly identified SNPs to increase resolving power to the canSNP typing of B. anthracis.
Key words Bacillus anthracis, Genotyping, SNP, MOL-PCR, Suspension microarray, Luminex
1 Introduction
Single nucleotide polymorphisms (SNPs) represent a major source of
genetic variation in Bacillus anthracis. Recently, a set of 14 representative SNPs that define major clades within the B. anthracis species
have been selected and used for assigning an isolate to one sublineages or subgroups [13]. These canonical SNPs (canSNPs) subdivided all B. anthracis isolates into three major lineages (A, B, and C),
with further subdivisions into seven distinct sublineages (C.Br.
A1055, B.Br.KrugerB, B.Br.CNEVA, A.Br.Ames, A.Br.Australia94,
A.Br.Vollum, A.Br.Western North America) and six subgroups (B.
Br.001/002, A.Br.001/002, A.Br.003/004, A.Br.005/006, A.
Br.008/011, A.Br.011/009). CanSNP analysis is currently considered the reference method in B. anthracis genotyping.
Several platforms and methods exist for SNP-based genotyping
[4, 5], including ultra-high-throughput array-based technologies,
such as those offered by Illumina [6] and Affymetrix [7]. But only
few of them are at the same time flexible, rapid (<1day), cost-effective,
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_29, Springer Science+Business Media New York 2015
401
402
SimonThierry andSylvianeDerzelle
403
404
SimonThierry andSylvianeDerzelle
2 Materials
2.1 DNA Preparation
andBiosafety
Procedures
2.2 MOL-PCR
2.3 Luminex
Equipment
andReagents
BA5 A.Br.006
BA4 A.Br.004
BA3 A.Br.003
A015
A028
B008
A027
A026
A022
A025
A021
A014
BA2 A.Br.002
B007
BA1 A.Br.001
P2
P1
P1
P2
P1
P1
P2
P1
P1
P2
P1
P1
P2
P1
P1
Allele MOLigo
xTAG
canSNP
5'P-AGGCGATGGAATGATCAACAACATATTGA TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT GTTGTAAATTGTAGTAAAGAAGTA
CGTTTTTAAGTTCATCATACCIIIIICATGCACG-3'
5'-ACTCGTAGGGAATAAACCGT TGTAAGTGAAATAGTGAGTTATTT
CGTTTTTAAGTTCATCATACCIIIIICATGCACT-3'
5'P-CCCTAATCCTTCCATAGCTCCACCA TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT GATAGATTTAGAATGAATTAAGTG
GACATCGCCGTCATACTTIIIIITGGAATGC-3'
5'-ACTCGTAGGGAATAAACCGT AAGATGATAGTTAAGTGTAAGTTA
GACATCGCCGTCATACTTIIIIITGGAATGT-3'
(continued)
5'P-TTGAGGTAGAAAAAGGAGGTTTTTATACAATGACA TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT TTTGATTTAAGAGTGTTGAATGTA
TGTATAAAAACCTCCTTTTTCTIIIIIACCTCAAA-3'
5'-ACTCGTAGGGAATAAACCGT GTATGTTGTAATGTATTAAGAAAG
TGTATAAAAACCTCCTTTTTCTIIIIIACCTCAAG-3'
5'P-TGGGCGGCAGTCGCTTTATC TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT GATTGATATTTGAATGTTTGTTTG
AGAAGGAGCAAGTAATGTTATAGIIIIIGTTTAGGT-3'
5'-ACTCGTAGGGAATAAACCGT ATTAAGTAAGAATTGAGAGTTTGA
AGAAGGAGCAAGTAATGTTATAGIIIIIGTTTAGGC-3'
5'P-TTGAAGTCGATGATAAAGGGAAACCGTATTATA TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT ATTGTGAAAGAAAGAGAAGAAATT
CAAATTTAATCTTTAAAGGAAAIIIIIACCGAAAT-3'
5'-ACTCGTAGGGAATAAACCGT AAATTGTGAAAGATTGTTTGTGTA
CAAATTTAATCTTTAAAGGAAAIIIIIACCGAAAC-3'
Sequence (5'-3')a
Table 1
Oligonucleotide sequences of all probes and primers used
A038
A036
A034
A035
A033
A018
BA9 B.Br.001
BA8 A.Br.009
B009
A030
BA7 A.Br.008
A029
BA6 A.Br.007
P1
P1
P2
P1
P1
P2
P1
P1
P2
P1
P1
P2
P1
P1
Allele MOLigo
xTAG
canSNP
Table 1
(continued)
5'-ACTCGTAGGGAATAAACCGT AGTAAGTGTTAGATAGTATTGAAT
ACCTTCTGTGTTCGTTGTTAAIIIIICGTTACTG-3'
5'-ACTCGTAGGGAATAAACCGT TGTATATGTTAATGAGATGTTGTA
ACCTTCTGTGTTCGTTGTTAAIIIIICGTTACTT-3'
5'P-TCTTCTATTGTACCGATTTCTTTTATGACCG TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT TTGTGTAGTTAAGAGTTGTTTAAT
CGGATATGATACCGATACCTIIIIITCTTATCT-3'
5'-ACTCGTAGGGAATAAACCGT AATAAGAGAATTGATATGAAGATG
CGGATATGATACCGATACCTIIIIITCTTATCC-3'
5'P-AAAGCCGTTCAAAAACAGTGGCC TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT TGATATAGTAGTGAAGAAATAAGT
AGGTATATTAACTGCGGATGIIIIIATGCAAGT-3'
5'-ACTCGTAGGGAATAAACCGT TATTAGAGTTTGAGAATAAGTAGT
AGGTATATTAACTGCGGATGIIIIIATGCAAGC-3'
5'P-CCGCTTGTTAAACGTATATTTGTAACTTTTTCAC TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT GTAATTGAATTGAAAGATAAGTGT
TATACGTTTTAGATGGAGATAIIIIIATTCTTCG-3'
5'-ACTCGTAGGGAATAAACCGT GAATTGTATAAAGTATTAGATGTG
TATACGTTTTAGATGGAGATAIIIIIATTCTTCT-3'
5'P-ATTCAGCTCGAATACTACCACCTTGTAATTC TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT GTGTTATAGAAGTTAAATGTTAAG
AGACGATAAACTGAATAATACCIIIIIATCCTTAC-3'
5'-ACTCGTAGGGAATAAACCGT TTTAAGTGAGTTATAGAAGTAGTA
AGACGATAAACTGAATAATACCIIIIIATCCTTAT-3'
Sequence (5'-3')a
406
SimonThierry andSylvianeDerzelle
P2
P1
P1
P2
P1
P1
P2
P1
P1
5'-ACTCGTAGGGAATAAACCGT-3'
5'BIOT-CGCGGTAGTAAGAAGTGAGA-3'
5'-P-ATTATTTTCAGCGGGAATTCGTTTCTTTTTAG TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT ATTTGTTATGATAAATGTGTAGTG
TTGAAGCAGGAIIIIIGCGCCCCC-3'
5'-ACTCGTAGGGAATAAACCGT TTGTGATAGTAGTTAGATATTTGT
TTGAAGCAGGAIIIIIGCGCCCCT-3'
5'P-TAGAAGTAAAGAAGGTTACCCAAGCACTTG TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT AAATTAGTTGAAAGTATGAGAAAG
TAAAATGAAGATAATGACAAAIIIIICGGGATGG-3'
5'-ACTCGTAGGGAATAAACCGT AGTGAATGTAAGATTATGTATTTG
TAAAATGAAGATAATGACAAAIIIIICGGGATGA-3'
5'P-CTTCACGTTATGGTTCGTTATGAACTTGAG TCTCACTTCTTACTACCGCG-3'
5'-ACTCGTAGGGAATAAACCGT GTGTGTTATTTGTTTGTAAAGTAT
GCATAGAAGCAGATGAGCTTAIIIIICATATCCA-3'
5'-ACTCGTAGGGAATAAACCGT AGTAGAAAGTTGAAATTGATTATG
GCATAGAAGCAGATGAGCTTAIIIIICATATCCG-3'
5'P-CTGTTCCTTTTGCAACTTCTCCTCCA TCTCACTTCTTACTACCGCG-3'
Sequence (5'-3')a
Primer, xTAG, and DNA target sequences of MOLigos are indicated, respectively, by underlined, italic and bold, and light gray sequences. The forward primer is biotinylated
at its 5 end and at two internal T nucleotides (positions are underlined and indicated in bold). The 5-end of each MOLigoP2 probe is phosphorylated
Reverse
A042
Forward
A020
A019
P2
Allele MOLigo
xTAG
canSNP
408
SimonThierry andSylvianeDerzelle
3 Methods
Carry out all procedures at room temperature.
Each experiment must include two controls for the calculation
of signal-to-noise ratio: a bead-only control that reports background fluorescence obtained from the microspheres alone and a
no template PCR control (H2O) that reports cross reactivity
between MOLigo pairs in the absence of any DNA template.
3.1 MOLPCRMethod
3.2 Hybridization
onBeads andSA-PE
Incubation
409
410
SimonThierry andSylvianeDerzelle
(1)
4 Notes
1. Viability testing can be performed by spreading an aliquot of
each DNA preparation on blood agar Petri Dish and incubation at 37C for 1824h. In case of growth, perform microfiltration and repeat viability step.
2. Assay limit of detection was determined to be 2ng of genomic
DNA per ligation reaction.
3. The supplier of DNA ligase (Epicentre) and streptavidin-
phycoerythrin (Bio-Rad) is critical for the method. Use the
specified products.
4. MOLigoP1 probes contains three functional components that
allow (a) detection of target sequences, (b) universal amplification of successfully ligated probes, and (c) capture of amplified products onto a microsphere array. Each pair contains a
5-end universal PCR Reverse primer sequence common for
all the different MOLigoP1, an internal 24bpxTAG sequence
unique for each MOLigoP1 probe for capture to conjugated
xTAG microsphere, and a 3-end sequence complementary to
the specific target DNA including the allele-specific nucleotide
(SNP) at its 3 end. To improve allele specificity, the 3 specific
target sequence is separated into two distinct priming regions
by a polydeoxyinosine (Poly(I)) linker, according to the DPO
principle [21]. The longer 5 segment initiates stable priming.
The shorter 3 segment determines target-specific extension
and SNP discrimination).
411
Acknowledgments
412
SimonThierry andSylvianeDerzelle
References
1. Pearson T, Busch JD, Ravel J etal (2004)
Phylogenetic discovery bias in Bacillus anthracis using single-nucleotide polymorphisms
from whole-genome sequencing. Proc Natl
Acad Sci U S A 101:1353613541
2. Marston CK, Allen CA, Beaudry J etal (2011)
Molecular epidemiology of anthrax cases associated with recreational use of animal hides and
yarn in the United States. PLoS One 6:e28274
3. Van Ert MN, Easterday WR, Huynh LY etal
(2007) Global genetic population structure of
Bacillus anthracis. PLoS One 2:e461
4. Kim S, Misra A (2007) SNP genotyping: technologies and biomedical applications. Annu
Rev Biomed Eng 9:289320
5. Kwok PY (2001) Methods for genotyping single nucleotide polymorphisms. Annu Rev
Genomics Hum Genet 2:235258
6. Gunderson KL, Steemers FJ, Ren H etal
(2006) Whole-genome genotyping. Methods
Enzymol 410:359376
7. Kennedy GC, Matsuzaki H, Dong S etal
(2003) Large-scale genotyping of complex
DNA.Nat Biotechnol 21:12331237
8. Price EP, Matthews MA, Beaudry JA etal
(2010) Cost-effective interrogation of single
nucleotide polymorphisms using the mismatch
amplification mutation assay and capillary electrophoresis. Electrophoresis 31:38813888
9. Bruse S, Moreau M, Azaro M, Zimmerman R,
Brzustowicz L (2008) Improvements to bead-
based oligonucleotide ligation SNP genotyping assays. Biotechniques 45:559571
10. Dunbar SA (2006) Applications of Luminex
xMAP technology for rapid, high-throughput
multiplexed nucleic acid detection. Clin Chim
Acta 363:7182
11. Dunbar SA, Vander Zee CA, Oliver KG, Karem
KL, Jacobson JW (2003) Quantitative, multiplexed detection of bacterial pathogens: DNA
and protein applications of the Luminex
LabMAP system. J Microbiol Methods
53:245252
12. Janse I, Bok JM, Hamidjaja RA, Hodemaekers
HM, van Rotterdam BJ (2012) Development
and comparison of two assay formats for parallel
Part IV
Integrative Omics and High-Throughput Platforms to Unravel
the Biology of Pathogens and Their Interaction with the Host
Chapter 30
Next-Generation Sequencing in Veterinary Medicine: How
Can the Massive Amount of Information Arising from HighThroughput Technologies Improve Diagnosis, Control,
and Management of Infectious Diseases?
Steven Van Borm, Sndor Belk, Graham Freimanis, Alice Fusaro,
Fredrik Granberg, Dirk Hper, Donald P. King, Isabella Monne,
Richard Orton, and Toon Rosseel
Abstract
The development of high-throughput molecular technologies and associated bioinformatics has dramatically
changed the capacities of scientists to produce, handle, and analyze large amounts of genomic, transcriptomic, and proteomic data. A clear example of this step-change is represented by the amount of DNA
sequence data that can be now produced using next-generation sequencing (NGS) platforms. Similarly,
recent improvements in protein and peptide separation efficiencies and highly accurate mass spectrometry
have promoted the identification and quantification of proteins in a given sample. These advancements in
biotechnology have increasingly been applied to the study of animal infectious diseases and are beginning to
revolutionize the way that biological and evolutionary processes can be studied at the molecular level. Studies
have demonstrated the value of NGS technologies for molecular characterization, ranging from metagenomic characterization of unknown pathogens or microbial communities to molecular epidemiology and
evolution of viral quasispecies. Moreover, high-throughput technologies now allow detailed studies of hostpathogen interactions at the level of their genomes (genomics), transcriptomes (transcriptomics), or proteomes (proteomics). Ultimately, the interaction between pathogen and host biological networks can be
questioned by analytically integrating these levels (integrative OMICS and systems biology). The application
of high-throughput biotechnology platforms in these fields and their typical low-cost per information content
has revolutionized the resolution with which these processes can now be studied.
The aim of this chapter is to provide a current and prospective view on the opportunities and challenges associated with the application of massive parallel sequencing technologies to veterinary medicine,
with particular focus on applications that have a potential impact on disease control and management.
Key words Next-generation sequencing, Animal infectious disease management, OMICS
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_30, Springer Science+Business Media New York 2015
415
416
Introduction
Genetic characterization of infectious agents plays a central role in
the diagnosis, monitoring, and control of infectious diseases. The
development of rapid DNA sequencing methods based on the
selective incorporation of chain-terminating dideoxynucleotides
([1]; later termed first-generation sequencing technologies) and
the polymerase chain reaction (PCR) DNA amplification technologies ([2]; reviewed in [3]) has paved the way for the study of biological and evolutionary processes at the molecular level. Such
technologies have been extensively applied to the diagnosis and
molecular epidemiology of infectious diseases of livestock and
become important tools for targeted research on host-pathogen
interactions. The most recent versions of these first-generation
sequencing technologies are widely accessible and provide highquality data. However, their application to projects such as whole
genome sequencing is expensive and time-consuming, often
requiring prior knowledge of the target genome for specific template amplification. These limitations have been particularly problematic for large sequencing projects and have motivated the
development of alternative, post-Sanger sequencing technologies
(next-generation sequencing or NGS).
Next-generation sequencing platforms provide unprecedented
throughput, generating hundreds of gigabases of data in a single
experiment. Although the initial capital investment and cost per
experiment remain high, the price per information unit (nucleotide) has been dramatically reduced in comparison with firstgeneration sequencing. Moreover, these technologies allow
unbiased sequencing without prior knowledge of the complete
DNA content in a sample while retaining the flexibility to allow for
targeted sequencing.
This paradigm shift in the scale of DNA sequence data has
revolutionized the way biological and evolutionary processes can
be studied at the molecular level, enabling genome projects previously restricted to high profile model organisms and human
pathogens to target pathogens of lesser economic and medical
significance.
Such advancements are now being increasingly applied to veterinary medicine. As a result, the increasing availability of these
technologies combined with the rapid development of applied
tools and protocols has provided a diverse array of applications for
use in genomics and transcriptomics and even routine diagnostics.
In this chapter, we review recent advances in NGS technologies that are becoming commonplace in many laboratories, with an
emphasis on the applications that have the potential to significantly
impact on diagnosis, prevention, and control of infectious diseases
in animals.
2
2.1
417
418
Challenges
Fig. 1 Steps in next-generation sequencing and data analysis workflows where error and bias introduction
may occur
419
Fig. 2 High-throughput technologies can be applied to numerous aspects of animal infectious diseases
420
Furthermore, reagent contamination has been reported to interfere with metagenomic analyses [14, 15]. Errors can also be made
during the sequencing process itself via base miscalls by NGS
machines. For example, the loss of synchronicity (dephasing) in a
percentage of the clonally amplified DNA template [16] results in
increased noise and sequencing errors [17, 8]. Each of the different NGS platforms available has its own distinct characteristics in
terms of read and error profiles, with the Illumina platform often
regarded as having the lowest error rate, while other platforms can
produce longer reads [7, 18, 19]. In addition, certain biases and
errors can be introduced during the analysis of NGS datasets, due
to the limitations of the algorithms or reference data used [20, 21].
For example, genomic repeat regions are a well-known problem
for sequence assembly algorithms [22]. However, software
advancements combined with platform and chemistry developments are expected to further reduce operational costs, error rates,
and input DNA quantities, allowing more careful sampling strategies and reducing experimentally introduced error and bias in the
future.
As a result of errors and bias introductions, NGS data needs to
be cleaned. This includes sequence filtering (removing lowquality sequences) and alignment followed by variant calling and
error correction. Discriminating true biological variants from those
due to experimental noise is an important issue when trying to
identify low-frequency variants in a population, for example, in
viral quasispecies or metagenomic analyses, and there are currently
a number of bioinformatics tools to aid in this (e.g., [2326, 19]).
Currently, a multitude of software has been developed to address
different aspects of NGS analyses [27, 28]. However, the available
algorithms for both genome assembly and amplicon analysis can
present some limitations [29], meaning that custom-made scripting and in-house resolution of bioinformatic problems are often
needed to investigate novel datasets and specific hypotheses. In this
context, researchers are frequently faced with the need to acquire
computer skills and bioinformatics expertise. To evaluate the
potential of NGS for a wider group of scientists and diagnosticians,
there is a real need to develop flexible and practical bioinformatics
workflows that can provide user-friendly tools for the analysis of
massive datasets and that become publicly available. Although
some software with a menu-driven approach is available (e.g.,
Geneious, CLC Workbench, Galaxy), most applications are optimized on UNIX-based operating systems and require some bioinformatics expertise. Although less user-friendly, UNIX-based
pipelines are typically freely available to the NGS user community
and are equipped with algorithms that track the high pace of innovation in the NGS field.
A further issue is the scale of genetic data produced by NGS
technologies which presents a physical constraint in terms of data
421
422
related to pathogen resistance and interaction with microbial communities in poultry [36]. Others have used novel sequencing technologies for the targeted study of specific gene families occupying
key roles in host immunology (e.g., Toll-like receptor (TLR) gene
family [37]).
The high variability and large size of the mitochondrial genome
(mtDNA) of eukaryotic parasites have been recently explored using
NGS (reviewed by [38]). mtDNA sequences proved very informative in epidemiological studies [38] but also include comparative
mtDNA sequencing of parasites with low and high zoonotic potential [39]. Targeting specific polymorphic genes in the Cryptosporidium
parvum genome using NGS, extensive intra-host genetic diversity
was documented [40]. Studies of the transcriptome (all mRNA
transcripts in an organism, tissue, or cell; also called RNA-Seq) of
different parasite species and/or developmental stages provide
insights into aspects of gene expression, regulation, and function,
which are major steps to understanding their biology (reviewed in
[41]). Examples include the characterization of the transcriptome
from Eimeria sp. from chicken [42] and Taenia sp. from sheep
[43]. In addition, RNA-Seq data have been used to predict potential drug targets [44] and to identify key genes involved in anthelmintic resistance [45].
Over the last five years, NGS has been used as an extremely
important tool in the tracing of transmission, genome characterization, and outbreak management of both viral and bacterial diseases.
The sequencing of these two pathogen types poses very different sets
of challenges and issues, where the large data output expressed typically in the Mb (megabase) to Gb (gigabase) range [4] is particularly
suited for the sequencing of larger bacterial genomes. The high plasticity of some microbial genomes, with large mobile elements, genecoding plasmids, chromosomal genes, and regions of extensive
genetic variability, can frequently complicate genome assembly [46].
While most viral genomes are significantly smaller than their bacterial
counterparts, the viral replication biology (particularly that of RNA
viruses) poses its own unique problems. These involve the inherent
variability of many viral genomes due to replication machinery lacking efficient proofreading mechanisms. This, combined with a short
generation time and high replication rate, results in a complex mix of
differing genomes (a swarm of closely related viruses) within a
single host that are often termed as quasispecies, reviewed in [47].
In addition, recombination and reassortment of segmented viral
genomes frequently occur. NGS techniques offer an unprecedented
step-change increase in the amount of sequence data that can be
generated from both types of these samples.
Figure 3 (different scales of sequence analysis) highlights
where genetic analyses can target different biological scales and
whether these are within an individual host, or between hosts,
423
Fig. 3 The differing levels of intra- and inter-host variation that can be explored using NGS technologies range
from intracellular dynamics to epidemiological applications
424
and resulting quasispecies population after inoculation of foot-andmouth disease virus (FMDV) into a single animal and identified
genetically distinct populations originating from different lesions
[50]. Morelli and colleagues [51] studied the evolution of FMDV
intra-sample sequence diversity during serial transmission in bovine
hosts, providing novel insights into the fine-scale evolution of an
RNA virus. NGS can also provide insights on microevolutionary
processes of viruses at different scales, including the fine-point resolution molecular epidemiology analysis of outbreaks [52].
Recent studies on influenza A viruses have demonstrated that
minority variants present in the donor population can be successfully
transmitted to the recipient host and become prevalent with unpredictable impact on the virus biological properties [53, 54]. These
findings suggest that the use of NGS approaches in RNA virus surveillance will be strategic to promptly detect biologically relevant
viral quasispecies and will help in expanding our understanding of
viral dynamics and emergence and the possible implications of mutation emergence for studies done using isolated viruses [55, 56].
The study of the viral swarm within individual hosts also has
implications for understanding the evolutionary dynamics of viral
populations under selection pressures, e.g., antiviral drugs. This
has been a particularly active field in human medicine, e.g., with
regard to human immunodeficiency virus (HIV) antiviral drugs
response, drug resistance, and viral tropism (reviewed in [5759])
and human influenza A (e.g., [60]) studies. The technologies
application to personalize antiviral treatment as a function of
genetic marker makeup in human medicine is just around the corner [61]. Although at present only an emerging field in veterinary
science, the development of antiviral drugs has the potential to
translate into efficient animal infectious disease control strategies
(e.g., [62, 63]).
The majority of the papers using NGS to investigate animal
infectious disease focus specifically upon the level of animal-toanimal transmission and the characterization of pathogens within a
single host, as this yields the most useful data in terms of outbreak
management and identifying mechanisms/sources of disease transmission. For example, Lefbure and colleagues [64] used NGS to
study genome complexity and horizontal gene transfer in foodborne Campylobacter spp. Biek et al. [65] studied local transmission patterns of Mycobacterium bovis in cattle and wildlife reservoirs
using whole genome sequences from 31 samples originating on
five farms. These demonstrated enough diversity between individual outbreaks to determine evolutionary variation down to herd
level. The identification of novel antimicrobial resistance genes in
the foodborne pathogen Campylobacter coli [66] was possible
using NGS, and the application of NGS technologies during a
recent crisis involving foodborne enterohemorrhagic Escherichia
coli O104:H4 allowed a swift genomic identification [67] that was
425
426
427
428
Conclusions
Next-generation sequencing technologies have the potential to
revolutionize our understanding of the complex dimensions of animal infectious disease and infection biology (Fig. 2), ranging from
the intracellular interactions to disease epidemiology. The application of high-throughput biotechnology platforms in these fields
and their typical low-cost per information content has increased
the resolution with which these processes can now be studied.
We now have high-resolution tools that provide veterinary
diagnostic laboratories with the ability to undertake swift and flexible responses to emerging infectious diseases and unexpected
pathogen variants. Moreover, these tools provide an increased resolution for the characterization of pathogens and provide important assets to improve our understanding. Fundamental research
on pathogen evolution, adaptation, and virulence determinants
can now be studied on a scale allowing within and between host
dissections of genetic variability. Moreover, high-throughput tools
429
430
Acknowledgments
The collaboration between the authors was supported by EpiSEQ: a research project supported under the 2nd joint call for
transnational research projects by EMIDA ERA-NET (FP7 project
nr 219235). Additional support for this work in the United
Kingdom was obtained from the Department of Environment,
Food and Rural Affairs (Defra project SE2940) and BBSRC (BB/
I014314/1).
References
1. Sanger F, Nicklen S, Coulson AR (1977) DNA
sequencing with chain-terminating inhibitors.
Proc Natl Acad Sci U S A 74:54635467
2. Mullis K, Faloona F, Scharf S et al (1986)
Specific enzymatic amplification of DNA
in vitro: the polymerase chain reaction. Cold
Spring Harb Symp Quant Biol 51:263273
3. Bartlett JM, Stirling D (2003) A short history
of the polymerase chain reaction. Methods
Mol Biol 226:36
4. Glenn TC (2011) Field guide to nextgeneration DNA sequencers. Mol Ecol
Resour 11:759769
5. Radford AD, Chapman D, Dixon L et al
(2012) Application of next-generation sequencing technologies in virology. J Gen Virol 93:
18531868
6. Pareek CS, Smoczynski R, Tretyn A (2011)
Sequencing technologies and genome
sequencing. J Appl Genet 52:413435
7. Loman NJ, Misra RV, Dallman TJ et al (2012)
Performance comparison of benchtop highthroughput sequencing platforms. Nat
Biotechnol 30:434439
8. Schadt EE, Turner S, Kasarskis A (2010) A
window into third-generation sequencing.
Hum Mol Genet 19(R2):R227240
9. Eisenstein M (2012) Oxford Nanopore
announcement sets sequencing sector abuzz.
Nat Biotechnol 30:295296
10. Brodin J, Mild M, Hedskog C et al (2013)
PCR-induced transitions are the major source
of error in cleaned ultra-deep pyrosequencing
data. PLoS One 8:e70388
11. Kim KH, Bae JW (2011) Amplification methods bias metagenomic libraries of uncultured single-stranded and double-stranded
DNA viruses. Appl Environ Microbiol
77:76637668
12. Rosseel T, Van Borm S, Vandenbussche F et al
(2013) The origin of biased sequence depth in
13.
14.
15.
16.
17.
18.
19.
20.
21.
sequence-independent nucleic Acid amplification and optimization for efficient massive parallel sequencing. PLoS One 8:e76144
Pettengill JB, McAvoy E, White JR et al
(2012) Using metagenomic analyses to estimate the consequences of enrichment bias for
pathogen detection. BMC Res Notes 5:378
Lysholm F, Wetterbom A, Lindau C et al
(2012) Characterization of the viral microbiome in patients with severe lower respiratory
tract infections, using metagenomic sequencing. PLoS One 7:e30875
Naccache SN, Greninger AL, Lee D et al
(2013) The perils of pathogen discovery: origin of a novel parvovirus-like hybrid genome
traced to nucleic acid extraction spin columns.
J Virol 87:1196611977
Whiteford N, Skelly T, Curtis C et al (2009)
Swift: primary data analysis for the Illumina
Solexa sequencing platform. Bioinformatics
25:21942199
Metzker
ML
(2010)
Sequencing
technologiesthe next generation. Nat Rev
Genet 11:3146
Quail MA, Smith M, Coupland P et al (2012)
A tale of three next generation sequencing
platforms: comparison of Ion Torrent, Pacific
Biosciences and Illumina MiSeq sequencers.
BMC Genomics 13:341
Zagordi O, Klein R, Daumer M,
Beerenwinkel N (2010) Error correction of
next-generation sequencing data and reliable
estimation of HIV quasispecies. Nucleic
Acids Res 38:74007409
Archer J, Rambaut A, Taillon BE et al (2010)
The evolutionary analysis of emerging low
frequency HIV-1 CXCR4 using variants
through timean ultra-deep approach. PLoS
Comput Biol 6:e1001022
Degner JF, Marioni JC, Pai AA et al (2009)
Effect of read-mapping biases on detecting
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
431
432
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
433
434
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
106.
134.
135.
136.
137.
138.
139.
140.
141.
142.
143.
144.
145.
435
rus serotype 10 detected in rockhopper penguins from the Falkland Islands. J Virol
84:1149611504
Nordentoft S, Molbak L, Bjerrum L et al
(2011) The influence of the cage system and
colonisation of Salmonella Enteritidis on the
microbial gut flora of laying hens studied by
T-RFLP and 454 pyrosequencing. BMC
Microbiol 11:187
Qu A, Brulc JM, Wilson MK et al (2008)
Comparative metagenomics reveals host specific metavirulomes and horizontal gene
transfer elements in the chicken cecum microbiome. PLoS One 3:e2945
Glass EJ, Crutchley S, Jensen K (2012) Living
with the enemy or uninvited guests: functional genomics approaches to investigating
host resistance or tolerance traits to a protozoan parasite, Theileria annulata, in cattle.
Vet Immunol Immunopathol 148:178189
Leon AJ, Banner D, Xu L et al (2013)
Sequencing, annotation, and characterization
of the influenza ferret infectome. J Virol
87:19571966
Maceachern S, Muir WM, Crosby S, Cheng
HH (2011) Genome-wide identification of
allele-specific expression (ASE) in response to
Mareks disease virus infection using next generation sequencing. BMC Proc 5(Suppl 4):S14
Miller LC, Fleming D, Arbogast A et al
(2012) Analysis of the swine tracheobronchial
lymph node transcriptomic response to infection with a Chinese highly pathogenic strain
of porcine reproductive and respiratory syndrome virus. BMC Vet Res 8:208
Wang F, Hu S, Liu W et al (2011) Deepsequencing analysis of the mouse transcriptome response to infection with Brucella
melitensis strains of differing virulence. PLoS
One 6:e28485
Wang CR, Xu MJ, Fu JH et al (2012)
Characterization of microRNAs from
Orientobilharzia turkestanicum, a neglected
blood fluke of human and animal health significance. PLoS One 7:e47001
Kong BW (2011) Identification of virus
encoding microRNAs using 454 FLX sequencing platform. Methods Mol Biol 733:8191
Ma M, Huang Y, Gong Z et al (2011)
Discovery of DNA viruses in wild-caught
mosquitoes using small RNA high throughput sequencing. PLoS One 6:e24758
Cullen BR (2006) Viruses and microRNAs.
Nat Genet 38(Suppl):S2530
Eulalio A, Schulte L, Vogel J (2012) The
mammalian microRNA response to bacterial
infections. RNA Biol 9:742750
436
155.
156.
157.
158.
159.
160.
161.
162.
Chapter 31
Impact of Next-Generation Technologies on Exploring
Socioeconomically Important Parasites and Developing
New Interventions
Cinzia Cantacessi, Andreas Hofmann, Bronwyn E. Campbell,
and Robin B. Gasser
Abstract
High-throughput molecular and computer technologies have become instrumental for systems biological
explorations of pathogens, including parasites. For instance, investigations of the transcriptomes of different
developmental stages of parasitic nematodes give insights into gene expression, regulation and function in
a parasite, which is a significant step to understanding their biology, as well as interactions with their
host(s) and disease. This chapter (1) gives a background on some key parasitic nematodes of socioeconomic importance, (2) describes sequencing and bioinformatic technologies for large-scale studies of the
transcriptomes and genomes of these parasites, (3) provides some recent examples of applications and (4)
emphasizes the prospects of fundamental biological explorations of parasites using these technologies for
the development of new interventions to combat parasitic diseases.
Key words Parasitic nematodes, Genomics, Transcriptomics, Bioinformatics, Next-generation
sequencing, Post-genomics, Anthelmintic resistance, Drug targets, Diagnostic markers
Introduction
Parasitic nematodes (roundworms) of humans and other animals are
of particular significance as pathogens [15]. For example, the soiltransmitted helminths (STHs) Ascaris spp. (large roundworm),
Ancylostoma duodenale, Necator americanus (hookworms) and
Trichuris trichiura (whipworm) are estimated to infect almost one
sixth of all humans [6, 7], and parasitic nematodes of livestock,
including species of Haemonchus, Ostertagia, and Trichostrongylus,
cause substantial losses estimated at billions of dollars per annum, due
to poor productivity, failure to thrive, the costs of anthelmintic treatment and deaths [810]. In addition to their socioeconomic impact,
anthelmintic resistance in nematodes of livestock [1113] has stimulated research towards developing alternative intervention and control
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_31, Springer Science+Business Media New York 2015
437
438
2.1 Selected
Examples
of Nematodes (Order
Strongylida) of Major
Socioeconomic
Importance
2.1.1 Trichostrongyles
2.1.2 Hookworms
439
Within the superfamily Trichostrongyloidea, Haemonchus contortus (barbers pole worm) and Trichostrongylus spp., for example,
are responsible for substantial production losses in the livestock
industries worldwide [10, 28]. H. contortus is the most important
nematode of small ruminants in subtropical and tropical (summer
rainfall) areas, whereas some Trichostrongylus spp. are often dominant in winter rainfall areas due to their ability to develop and
survive at lower temperatures than H. contortus does [29]. The life
cycles of H. contortus and T. colubriformis are similar and direct,
with eggs being laid by females in the abomasum (H. contortus) or
small intestine (Trichostrongylus) of the host [26, 30, 31]. Under
suitable environmental conditions [30, 32], first-stage larvae (L1s)
hatch from eggs to develop, via the second-stage larvae (L2s), to
infective, third-stage larvae (L3s). The cuticle of the L2 is retained
as a sheath around the L3 and protects it from desiccation [14, 30,
32]. Small ruminants acquire the infection by ingesting L3s from
contaminated pastures. The L3s pass through the forestomachs
and undergo an exsheathment process to then establish, via the
parasitic fourth-stage larvae (L4s), as adult males and females in
the abomasum (H. contortus) or small intestine (Trichostrongylus
spp.) within ~3 weeks [14, 26, 30, 32]. The exsheathment process
is triggered by stimuli within the host and may include (depending
on the species of nematode) dissolved gaseous CO2 and undissociated carbonic acid (H. contortus) or hydrochloric acid and pepsin
(T. colubriformis) in the abomasum. L3s respond to these stimuli
by producing an exsheathment fluid which determines the detachment of the sheath from the bodies of the larvae [5, 3335].
The adults of H. contortus feed on blood from vessels in the
gastric wall. Consequently, the main clinical signs of acute haemonchosis are anemia, variable degrees of edema, as well as lethargy,
decreased live-weight gain, impaired wool/milk production, and
decreased reproductive performance, often leading to death in
severely affected animals [36, 37]. Trichostrongylosis is triggered
by the presence of adult parasites in mucus-covered tunnels in the
epithelial surface of the small intestine [38], usually associated with
extensive villous atrophy, combined with hyperplasia of the submucosal glands, mucosal thickening, and erosion as well as infiltration
of lymphocytes and neutrophils into affected areas [3842]. Clinical
signs of trichostrongylosis include malabsorption, weight loss, progressive emaciation, and diarrhea (scouring or black scour).
The hookworms N. americanus and A. duodenale of humans are
estimated to infect ~1 billion people in rural regions of the subtropics and tropics [1], with the highest prevalence (~17 %)
recorded in areas of sub-Saharan Africa and China [1, 43, 44], and
cause an estimated disease burden of 22 million disability-adjusted
life years (DALYs) [45]. Although N. americanus is the most
widely distributed hookworm of humans globally [1], a related
440
441
442
443
444
445
446
SSH-based microarray analysis [183] showed a substantial upregulation in the numbers and levels of transcripts encoding ASPs in
serum-activated L3s [183].
To date, molecular studies of hookworms have mainly involved
A. caninum, because of its use as a model for species infecting
humans [182, 183, 190192]. Clearly, detailed knowledge and
understanding of the molecules transcribed in all stages of different
species of hookworms, including N. americanus and A. duodenale
of humans, should facilitate the identification of conserved pathways linked to development, survival, reproduction, parasite interactions and disease, and could assist in the discovery of new
intervention strategies.
3.2 High-Throughput
Sequencing
Techniques
Table 1
Technical features of next-generation sequencing platforms (i.e., 454/Roche, Illumina/Solexa, and SOLiD)a
Description
454/Roche
Illumina/Solexa
SOLiD
Platform
Sequencing
method
Isothermal bridge
amplification on flow
cell
Emulsion PCR of
bead-bound oligos
Sequencing
chemistry
Pyrosequencing using
polymerase
Ligation (dual-base
encoding octamers)
Reversible terminator
using polymerase
~1 million
Up to 3 billion
Read length
1,000 bp
50250 bp
100 bp
Run time
~12 h
~29 days
~3 days
Peer-reviewed
manuscripts
++++
+++
++
Examples of
applications
De novo sequencing,
metagenomics, targeted
sequencing
Resequencing, RNA-Seq,
DNA methylation
studies
Resequencing, RNA-Seq
447
sequences simultaneously, these platforms have been at the forefront of the genomic and transcriptomic research [197199] and
are powerful tools for investigating the transcriptomes of parasitic
nematodes on an unprecedented scale.
The 454/Roche platform [193] uses a sequencing-by-synthesis
approach. For transcriptomic studies, cDNA is randomly fragmented (by nebulization) into sections of variable size; adaptors
are ligated to each end of these fragments, which are then mixed
with a population of agarose beads whose surfaces anchor oligonucleotides complementary to the 454-specific adapter sequence,
such that each bead is associated with a single fragment. Each of
these complexes is transferred into individual oilwater micelles
containing amplification reagents and is then subjected to an emulsion PCR (emPCR) step, during which ~10 million copies of each
cDNA are produced and bound to individual beads. Subsequently,
in the sequencing phase, the beads anchoring the cDNAs are
deposited on a picotiter plate, together with other enzymes
required for the pyrophosphate sequencing reaction (i.e., ATP sulfurylase and luciferase), and sequencing is carried out by flowing
the reagents (nucleotides and buffers) over a plate [200].
Following the introduction of the 454 technology, the first
Illumina (formerly Solexa) sequencer became available [194]. This
technology involves fragmentation of cDNA sample into a shotgun
library, followed by the in vitro ligation of Illumina-specific adaptors to each cDNA template; the termini of the template are covalently attached to the surface of a glass slide (or flow cell). Attached
to the flow cell are primers complementary to the other end of the
template, which bend the cDNAs to form bridge-like structures.
During the amplification step (bridge-PCR), clonal clusters, each
consisting of ~1,000 amplicons, are generated; subsequently, the
cDNAs are linearized, and the sequencing reagents are directly
added to the flow cell, with four types of fluorescently labeled
nucleotides. After the incorporation of a fluorescent base, the flow
cell is interrogated with a laser in several locations, which results in
several image acquisitions at the end of a single synthesis cycle
[200]. This technology is considered ideal for both de novo and
resequencing projects, targeted sequencing, single-nucleotide
polymorphism (SNP) analyses and gene transcription studies.
The sequencing process of the SOLiD platform [195] employs
the enzyme DNA ligase, instead of a polymerase [200]. Briefly, after
an emPCR step, the adaptor sequences of the cDNA templates bind
to complementary primers that are covalently anchored to a glass
slide. Subsequently, a set of four fluorescently labeled di-probes
(octamers of random sequence, except known dinucleotides at the
3-terminus) are added to the sequencing reaction. In case an
octamer is complementary to the template, it will be ligated, and the
two specific nucleotides can be called; subsequently, an image is acquired
448
Bioinformatics
3.3.1 Assembly
449
Following assembly, the contigs and single reads (or singletons) are
compared with known sequence data available in public databases,
in order to assign a predicted identity to each query sequence if
significant matches are found [206, 215]. In addition, assembled
nucleotide sequences are usually conceptually translated into predicted proteins using algorithms that identify protein-coding
regions (open reading frames, ORFs) from individual contigs.
Examples of such algorithms are OrfPredictor [236], ESTScan
[213], DECODER [237] and ORFcor [238]. Once peptide
sequences are predicted, they are compared with amino acid
sequence data available in public databases to identify protein
domains [206, 215]. For instance, the software InterProScan
[216] provides an integrated tool for the characterization of a protein family or an individual protein sequence, domain and/or
functional site by comparing sequences with information available
in the databases PROSITE [239], PRINTS [240], Pfam [241],
ProDom [242], SMART [243] and/or Gene Ontology (GO;
244). In addition, other programs are available for the prediction
of transmembrane domains (e.g., TMHMM; 245) and/or signal
peptide motifs (e.g., SignalP; 246).
Different types of the Basic Local Alignment Software Tool
(BLAST; 247) are used for comparing the nucleotide sequence data
with DNA or cDNA (BLASTn) or amino acid (BLASTx) sequences
or conceptually translated peptides with protein sequences
(BLASTp), available in databases [206, 215]. Public databases represent comprehensive collections of nucleotide and amino acid
sequences. Due to the rapid progress in the discovery and character-
450
451
452
453
454
455
456
457
458
459
460
461
Acknowledgments
Funding from the Australian Research Council, the National
Health and Medical Research Council, the Australian Academy of
Science, the Alexander von Humboldt Foundation, and Melbourne
Water Corporation is gratefully acknowledged (RBG). Our
research program was also supported by the Victorian Life Sciences
Computation Initiative (grant number VR0007) on its Peak
Computing Facility at the University of Melbourne, an initiative of
the Victorian Government (RBG).
462
References
1. de Silva NR, Brooker S, Hotez PJ et al (2003)
Soil-transmitted helminth infections: updating the global picture. Trends Parasitol
19:547551
2. Artis D (2006) New weapons in the war on
worms: identification of putative mechanisms
of immune-mediated expulsion of gastrointestinal nematodes. Int J Parasitol 36:723733
3. Bethony J, Brooker S, Albonico M et al
(2006) Soil-transmitted helminth infections:
ascariasis, trichuriasis, and hookworm. Lancet
367:15211532
4. Brooker S, Clements AC, Bundy DA (2006)
Global epidemiology, ecology and control of
soil-transmitted helminth infections. Adv
Parasitol 62:221261
5. Nikolaou S, Gasser RB (2006) Prospects for
exploring molecular developmental processes
in Haemonchus contortus. Int J Parasitol
36:859868
6. Hotez PJ, Fenwick A, Savioli L et al (2009)
Rescuing the bottom billion through control
of neglected tropical diseases. Lancet
373:15701575
7. OHarhay MO, Horton J, Olliaro PL (2010)
Epidemiology and control of human gastrointestinal parasites in children. Expert Rev Anti
Infect Ther 8:219234
8. Newton SE, Munn EA (1999) The development of vaccines against gastrointestinal nematode parasites, particularly Haemonchus
contortus. Parasitol Today 15:116122
9. Newton SE, Meeusen EN (2003) Progress
and new technologies for developing vaccines
against gastrointestinal nematode parasites of
sheep. Parasite Immunol 25:283296
10. Roeber F, Jex AR, Gasser RB (2013) Advances
in the diagnosis of key gastrointestinal nematode infections of livestock, with an emphasis
on small ruminants. Biotechnol Adv
31(8):11351152.
doi:10.1016/j.biotechadv.2013.01.008,
pii:
S0734-9750(13)00010-4
11. Wolstenholme AJ, Fairweather I, Prichard R
et al (2004) Drug resistance in veterinary helminths. Trends Parasitol 20:469476
12. Gilleard JS (2006) Understanding anthelmintic resistance: the need for genomics and
genetics. Int J Parasitol 36:12271239
13. Wolstenholme AJ, Kaplan RM (2012)
Resistance to macrocyclic lactones. Curr
Pharm Biotechnol 13:873887
14. Anderson RC (2000) Nematode parasites of
vertebrate. Their development and transmission, 2nd edn. CABI Publishing, Wallingford
463
464
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73. Miller
JE,
Horohov
DW
(2006)
Immunological aspects of nematode parasite
control in sheep. J Anim Sci 84:E124E132
74. Larsen RH, Christensen CM, Lind P (1997)
Serological assays for the identification of
Oesophagostomum dentatum infections in
pigs. Res Vet Sci 63:510
75. Joachim A, Ruttkowski B, Daugschies A
(1998) Changes in antigen and glycoprotein
patterns during the development of
Oesophagostomum dentatum. Int J Parasitol
28:18531860
76. Joachim A, Ruttkowski B, Christensen CM
et al (1999) Identification, isolation and characterisation of a species-specific 30 kDaantigen of Oesophagostomum dentatum.
Parasitol Res 85:307311
77. Stockdale PH (1970) Necrotic enteritis of
pigs caused by infection with Oesophagostomum
spp. Br Vet J 126:526530
78. Haussler M (1996) Immunhistologische
Untersuchungen an der Dickdarmschleimhaut
bei der Oesophagostomum dentatum-infektion
des
Schweines
zu
unterschiedlichen
Zeitpunkten nach experimenteller infection.
Dr Med. Vet. Thesis, Tierartzliche
Hochschule, Hannover, Germany
79. McSorley HJ, Loukas A (2010) The immunology of human hookworm infections.
Parasite Immunol 32:549559
80. Pearson MS, Tribolet L, Cantacessi C et al
(2012) Molecular mechanisms of hookworm
disease stealth, virulence, and vaccines. J
Allergy Clin Immunol 130:1321
81. Holden-Dye
L,
Walker
RJ
(2007)
Anthelmintic drugs. WormBook, ed. The C.
elegans Research Community, doi:/10.1895/
wormbook.1.143.1,
http://www.wormbook.org
82. Robertson AP, Clark CL, Martin RJ (2010)
Levamisole and ryanodine receptors. I: A
contraction study in Ascaris suum. Mol
Biochem Parasitol 171:17
83. Keiser J, Utzinger J (2008) Efficacy of current
drugs against soil-transmitted helminth infections: systematic review and meta-analysis.
JAMA 299:19371948
84. Stepek G, Buttle DJ, Duce IR et al (2006)
Human gastrointestinal nematode infections:
are new control methods required? Int J Exp
Pathol 87:325341
85. Epe C, Kaminsky R (2013) New advancement in anthelmintic drugs in veterinary
medicine. Trends Parasitol 29:129134
86. Gilleard JS, Beech RN (2007) Population
genetics of anthelmintic resistance in parasitic
nematodes. Parasitology 134:11331147
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
465
466
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
125.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
159.
160.
161.
467
468
176.
177.
178.
179.
180.
181.
182.
183.
184.
185.
186.
469
470
231.
232.
233.
234.
235.
236.
237.
238.
239.
240.
241.
242.
243.
244.
245.
246.
247.
248.
249.
250.
251.
252.
253.
254.
255.
256.
257.
258.
259.
260.
261.
262.
263.
264.
265.
266.
267.
268.
269.
270.
271.
272.
273.
274.
275.
276.
277.
278.
279.
471
472
280. Morris KV (2008) RNA-mediated transcriptional gene silencing in human cells. Curr Top
Microbiol Immunol 320:211224
281. Rosso MN, Jones JT, Abad P (2009) RNAi
and functional genomics in plant parasitic
nematodes.
Annu
Rev
Phytopathol
47:207232
282. Serkirk ME, Huang SC, Knox DP et al (2012)
The development of RNA interference
(RNAi) in gastrointestinal nematodes.
Parasitology 139:605612
283. Stinchcomb DT, Shaw JE, Carr SH et al
(1985) Extrachromosomal DNA transformation of Caenorhabditis elegans. Mol Cell Biol
5:34843496
284. Fire A (1986) Integrative transformation of
Caenorhabditis
elegans.
EMBO
J
5:26732680
285. Chalfie M, Tu Y, Euskirchen G et al (1994)
Green fluorescent protein as a marker for
gene expression. Science 263:802805
286. Lok JB, Artis D (2008) Transgenesis and neuronal ablation in parasitic nematodes: revolutionary new tools to dissect host-parasite
interactions. Parasite Immunol 30:203214
287. Hobert O, Loria P (2006) Uses of GFP in
Caenorhabditis elegans. Methods Biochem
Anal 47:203226
288. Reinke V, Smith HE, Nance J et al (2000) A
global profile of germline gene expression in
C. elegans. Mol Cell 6:605616
289. Kim SK, Lund J, Kiraly M et al (2001) A gene
expression map for Caenorhabditis elegans.
Science 293:20872092
290. Jiang M, Ryu J, Kiraly M et al (2001)
Genome-wide analysis of developmental and
sex-regulated gene expression profiles in
Caenorhabditis elegans. Proc Natl Acad Sci U
S A 98:218223
291. Chilton NB (2004) The use of nuclear ribosomal DNA markers for the identification of
bursate nematodes (order Strongylida) and
for the diagnosis of infections. Anim Health
Res Rev 5:173187
292. Brglin TR, Lobos E, Blaxter ML (1998)
Caenorhabditis elegans as a model for parasitic
nematodes. Int J Parasitol 28:395411
293. Mitreva M, Jasmer JP, Zarlenga DS et al
(2011) The draft genome of the parasitic
nematode Trichinella spiralis. Nat Genet
43:228243
294. Williamson AL, Lecchi P, Turk BE et al
(2004) A multi-enzyme cascade of hemoglobin proteolysis in the intestine of bloodfeeding
hookworms.
J
Biol
Chem
279:3595035957
320.
321.
322.
323.
324.
325.
326.
327.
328.
329.
330.
331.
332.
473
474
346.
347.
348.
349.
350.
351.
352.
353.
354.
355.
356.
357.
Chapter 32
Functional Genomics of Tick Vectors Challenged
with the Cattle Parasite Babesia bigemina
Ana Domingos, Sandra Antunes, Margarita Villar, and Jos de la Fuente
Abstract
Ticks are obligate hematophagous ectoparasites considered as vectors of animal diseases, having a huge
economic impact in cattle industry. Babesia spp. are tick-borne pathogens that cause a disease called babesiosis in a wide range of animals and in humans. Control of tick infestations is mainly based on the use of
acaricides, which have limited efficacy reducing tick infestations, mostly due to wrong usage, and is often
accompanied by the selection of acaricide-resistant ticks, environmental contamination, and contamination
of milk and meat products. Vaccines affecting both vector and pathogens constitute new control strategies
for tick and tick-borne diseases and are, therefore, a good alternative to chemical control.
In this chapter we describe the identification of Rhipicephalus (Boophilus) annulatus genes differentially expressed in response to infection with B. bigemina by using suppression-subtractive hybridization
(SSH), which allows the identification of differentially expressed genes. The results of the SSH studies are
validated by real-time reverse transcription (RT)-PCR. Functional analyses are conducted by RNAi on
selected R. annulatus genes to determine their putative role in B. bigeminatick interactions. Gathered
data may be useful for the future development of improved vaccines and vaccination strategies to control
babesiosis.
Key words Tick, Genomics, Babesia, Rhipicephalus, Boophilus, Subtractive hybridization, RNA
interference, Vaccines
Introduction
Tick-borne pathogens of the genus Babesia are Apicomplexan parasites responsible for a disease called babesiosis that affects a wide
range of animals and, occasionally, humans. The major economic
impact of babesiosis is on cattle industry, caused mainly by Babesia
bovis and B. bigemina. Rhipicephalus (Boophilus) spp. ticks are their
principal vectors, being considered one of the most important cattle ectoparasites due to the direct impact affecting leather quality,
meat and milk production and, most importantly, pathogen transmission capacity [1].
In the last decades, new tools have been developed in the
molecular biology field, leading to a better knowledge of genes
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_32, Springer Science+Business Media New York 2015
475
476
477
478
2
2.1
Materials
Organisms
2.2 General
Laboratory
Consumables,
Materials,
and Equipment
2.3 Suppression
Subtractive
Hybridization (SSH)
Library Construction
479
2.4 Sequence
Analysis and
Database Search
Spin
Kit
(GE
Healthcare,
480
2.5 Validation
of SSH Results
dsRNA Synthesis
1. R. annulatus RNA.
2. Specific primers for target genes with T7 extension.
3. Access RT-PCR system (Promega, Madison, USA).
4. PCR products purification kit (e.g., PureLink PCR
Purification Kit, Life Technologies, Carlsbad, CA, USA).
5. MEGAscript T7 Kit (Ambion Life Technologies, Carlsbad,
CA, USA).
Injection in Ticks
1. Specific dsRNA.
2. Hamilton syringe, with 1 in., 33 G needle.
3. Forceps/tweezers.
4. Double face duck tape.
5. Dental wax plates.
6. Control buffer: 10 mM TrisHCl, pH 7, 1 mM EDTA.
Tick Feeding
1. Infected B. bigemina calf.
2. Freshly molted female R. annulatus.
3. Contact glue.
4. Cotton sleeves.
5. Razor blade.
Gene Knockdown Assessment and Infection Assessment
1. R. annulatus ticks treated with dsRNA.
2. iScript One-Step RT-PCR Kit with SYBR Green (Biorad,
Hercules, CA, USA).
3. Gene-specific and housekeeping primers.
3
3.1
Methods
Ticks Processing
3.1.1 Uninfected
and Babesia bigeminaInfected Ticks for SSH
Library Construction
481
3.2.2 Detection
of Babesia bigemina
in Ticks by PCR
3.2.3 Complementary
DNA (cDNA) Library
Construction and SSH
1. Check the RNA integrity before proceeding with cDNA synthesis (see Note 5).
2. Create two pools corresponding to the infected and uninfected
tick populations. Seven to ten ticks are enough to obtain sufficient RNA for each pool.
482
483
1. Purify plasmids from the bacterial culture using Illustra plasmidPrep Mini Spin Kit, following the manufacturers
instructions.
2. Randomly select and sequence clones obtained from the
SSH library using traditional Sanger sequencing approaches
(see Note 8).
3.3 Sequence
Analysis and
Database Search
1. Use the available means for automated sequence clean-up, assembly, and BLAST analysis against nonredundant sequence database
(nr) and databases of tick-specific sequences (see Note 9).
3.3.1 Sequencing
and Analysis of SSH Clones
2. Analyze protein/gene ontology using a protein reference database (see Note 9).
3. Align nucleotide and protein sequences using widely available
software like CLUSTAL 2.1 multiple sequence alignment tool.
3.3.2 Confirmation
of Differential Gene
Expression by
Real-Time RT-PCR
1. Design appropriate primers manually or using available programs for the selected differentially expressed candidate genes.
2. Use total RNA from uninfected and B. bigemina-infected
R. annulatus female ticks to construct cDNA using the
iScript cDNA Synthesis Kit following the manufacturers
instructions.
3. Use the previously synthesized cDNA to perform quantitative
real-time PCR. iQ SYBR Green Supermix can be used for the
real-time PCR following the manufacturers recommendations, including forward and reverse primers at a final concentration of 500 mM and cDNA at a final concentration of
2025 ng (500 ng).
4. PCR conditions include an initial denaturation and enzyme
activation step at 95 C for 2 min, followed by 40 cycles of 15
s at 95 C, 15 s at the primers annealing temperature, and 15 s
at 62 C for extension.
484
1. Based in the SSH clone sequences, design specific primers containing T7 promoter sequences (5-TAA TAC GAC TCA CTA
TAG GGT ACT-3) at the 5-end. Long fragments are desired.
2. Use RNA as template to amplify the fragments of interest by
RT-PCR, using the Access RT-PCR system according to
instructions given by the manufacturer, including 1 M of
each primer and 200 ng of template RNA. Conditions of the
RT-PCR should be: first-strand synthesis performed at 45 C
for 45 min and AMV reverse transcriptase inactivation performed at 94 C for 3 min, followed by PCR cycling for 40
cycles of 30 s at 94 C, 30 s at 55 C, and 90 s at 68 C with a
final extension step of 7 min at 68 C.
3. Analyze amplification products on a 0.5 TBE, 1.2 % agarose gel.
4. Purify the PCR products using an appropriated kit like the
PureLink PCR Purification Kit.
5. Use the MEGAscript T7 Kit to synthesize dsRNA according
to the manufacturers instructions.
6. Purify and quantify by spectrometry the resulting dsRNA and
check it on a 1.2 % TBE agarose gel.
3.4.2 dsRNA
Injection in Ticks
485
486
7. Normalize against the tick 16S rDNA gene using the ddCT
method [27, 28].
8. Compare the B. bigemina infection levels in ticks between
dsRNA and control ticks using t-student test (P = 0.05).
Notes
General note: A project having as target the identification of new
vaccine candidates has to be carefully designed and has to surpass
different milestones. Experiments have to be properly designed
and the obtained biological samples must be in good conditions.
1. Ticks must be reared in a certified laboratory according to animal welfare guidelines. Individual features as tick life cycle
must be considered, for instance, if its a one, two, or three
hosttick species. When dealing with cattle ticks like R. microplus and R. annulatus, some constrains can arise due to their
host specificity [30]. Plus, cattle are most of the times a requirement for the conduction of laboratorial experiments. Other
species are less selective and smaller vertebrates like rabbits can
be used to establish tick colonies. The tick infection can be
obtained in different ways such as artificial feeding or even
injection of pathogen, but the best way of copying natural
infections is to infect the host and promote ordinary feeding.
2. DNA derived from nontarget organisms present on the surface
of ticks may yield amplicons with some primers. To overcome
this problem, the ticks may be rinsed two times individually in
distilled water, once in 75 % ethanol and once more in water.
3. The TRI Reagent is a mixture of guanidine thiocyanate and
phenol that uses a convenient single-step liquid phase separation resulting in the simultaneous isolation of RNA, DNA, and
proteins. After adding chloroform or 1-bromo-3-chloropropane
and centrifuging, the mixture separates into 3 phases: an aqueous phase containing the RNA, the interphase containing
DNA, and an organic phase containing proteins. Each component can then be isolated after separating the three phases.
This reagent extracts total RNA, but when poly A + RNA is the
target, it is necessary to separate this last type of RNA from the
remaining existent RNA in a cell (rRNA and tRNA).
4. A one step PCR can be used to detect the presence of B. bigemina in sampled ticks. The example shown here uses primers
Bbi400F: 5-AGC TTG CTT TCA CAA CTC GCC-3 and
Bbi400R: 5-TTG GTG CTT TGA CCG ACG ACA T-3 that
amplify a 400 bp fragment within the conserved region of the
five rap-1a paralogous genes [31, 32], but the detection of
pathogen infection in ticks can be done using different protocols
487
488
References
1. Bock R, Jackcon L, de Vos A, Jorgensen W
(2004) Babesiosis of cattle. Parasitology 129:
S247S269
2. de la Fuente J, Moreno-Cid JA, Canales M
et al (2011) Targeting arthropod subolesin/
akirin for the development of a universal vaccine for control of vector infestations and
pathogen transmission. Vet Parasitol 181:
1722
3. Macaluso KR, Mulenga A, Simser JA et al
(2003) Differential expression of genes in
uninfected and rickettsia-infected Dermacentor
variabilis ticks as assessed by differentialdisplay PCR. Infect Immun 71:61656170
4. Mulenga A, Macaluso KR, Simser JA et al
(2003) Dynamics of Rickettsia-tick interactions: identification and characterization of
differentially expressed mRNAs in uninfected
and infected Dermacentor variabilis. Insect
Mol Biol 12:185193
5. Nene V, Lee D, Kanga S, Skilton R et al
(2004) Genes transcribed in the salivary glands
of female Rhipicephalus appendiculatus ticks
infected with Theileria parva. Insect Biochem
Mol Biol 34:11171128
6. Rudenko N, Golovchenko M, Edwards MJ
et al (2005) Differential expression of Ixodes
ricinus tick genes induced by blood feeding or
Borrelia burgdorferi infection. J Med Entomol
42:3641
7. de la Fuente J, Blouin EF, Manzano-Roman
NR et al (2007) Functional genomic studies of
tick cells in response to infection with the cattle pathogen, Anaplasma marginale. Genomics
90:712722
8. de la Fuente J, Kocan KM, Almazan C et al
(2007) RNA interference for the study and
genetic manipulation of ticks. Trends Parasitol
23:427433
9. Villar M, Torina A, Nunez Y et al (2010)
Application of highly sensitive saturation labeling to the analysis of differential protein
expression in infected ticks from limited samples. Proteome Sci 8:43
10. Zivkovic Z, Esteves E, Almazan C et al (2010)
Differential expression of genes in salivary
glands of male Rhipicephalus (Boophilus)
microplus in response to infection with
Anaplasma marginale. BMC Genomics
11:186
11. Mercado-Curiel RF, Palmer GH, Guerrero FD
et al (2011) Temporal characterisation of the
organ-specific Rhipicephalus microplus transcriptional response to Anaplasma marginale
infection. Int J Parasitol 41:851860
26.
27.
28.
29.
30.
31.
32.
33.
489
Chapter 33
Metagenomic Approaches to Disclose Disease-Associated
Pathogens: Detection of Viral Pathogens in Honeybees
Fredrik Granberg, Oskar E. Karlsson, and Sndor Belk
Abstract
Metagenomic approaches have become invaluable for culture-independent and sequence-independent
detection and characterization of disease-associated pathogens. Here, the sequential steps from sampling
to verification of results are described for a metagenomic-based approach to detect potential pathogens in
honeybees. The pre-sequencing steps are given in detail, but due to the rapid development of sequencing
technologies, all platform-specific procedures, as well as subsequent bioinformatics analysis, are more generally described. It should also be noted that this approach could, with minor modifications, be adapted
for other organisms and sample matrices.
Key words Metagenomics, Sequencing, Unknown viruses, Virus detection, Diagnosis, Unknown
etiology
Introduction
Metagenomics based on high-throughput sequencing (HTS)
allows for cell culture-independent and nucleotide sequenceindependent detection of pathogens as well as characterization of
the entire microbial flora in a sample. It thus has the potential to
detect the full spectrum of emerging new pathogens, including
novel viruses and fastidious bacteria, as demonstrated and reviewed
[14]. Viral metagenomics (or metaviromics) focuses on the viral
fraction of a metagenomic sample, and several HTS-based
approaches have been developed for unbiased characterization of
the viral populations in various organisms and environments. These
approaches are usually comprised of several typical steps including
sample homogenization, target enrichment, sequence-independent
amplification, sequencing library construction, HTS, and bioinformatics analysis [5, 6].
The pre-sequencing steps are similar to the method outlined by
Allander et al. [7]. Following homogenization and filtration, nuclease treatment with both DNase and RNase is used to reduce the host
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_33, Springer Science+Business Media New York 2015
491
492
493
Materials
If solutions are not obtained from a commercial supplier, they
should be prepared using ultrapure water (prepared by purifying
deionized water to attain a sensitivity of 18 M cm at 25 C) and
analytical grade reagents. Prepare and store reagents according to
494
2.2 Enrichment by
Nuclease Treatment
and Nucleic Acid
Extraction
495
4. 1.5 ml tubes.
5. Nuclease-free water (not DEPC treated).
6. Tag labeling primer FR26RV-N (GCCGGAGCTCTGCAG
ATATCNNNNNN): Dilute to 10 M with nuclease-free
water.
7. Amplification primer FR20RV (GCCGGAGCTCTGCAGAT
ATC): Dilute to 10 M with nuclease-free water.
8. dNTP mix (10 mM each).
9. dNTP mix (2.5 mM each).
10. Exo() Klenow DNA polymerase (5 U/l; e.g., New England
Biolabs or Thermo Fisher Scientific).
11. Exo() Klenow Buffer (10; supplied with Exo() Klenow
DNA polymerase).
12. DTT (0.1 M).
13. RNase inhibitor (40 U/l), e.g., RNaseOUT (Life
Technologies), RiboLock (Thermo Fisher Scientific), or
Protector (Roche Applied Science).
14. Reverse transcriptase (200 U/l), e.g., SuperScript III (Life
Technologies) or Maxima (Thermo Fisher Scientific).
15. Reverse transcriptase (RT) buffer (5; supplied with reverse
transcriptase).
16. MgCl2 (25 mM).
17. DNA polymerase (5 U/l), e.g., AmpliTaq Gold with Buffer
II (Life Technologies).
18. PCR buffer (10; supplied with DNA polymerase).
19. Column-based PCR purification kit, e.g., QIAquick PCR
Purification Kit (Qiagen) or PureLink PCR Purification Kit
(Life Technologies).
20. Elution buffer (EB): 10 mM TrisHCl (pH 8.5).
21. EcoRV (20 U/l; e.g., New England Biolabs).
22. Restriction enzyme buffer (10; supplied with EcoRV).
23. Instrument for quantification of nucleic acids, e.g., Qubit 2.0
Fluorometer with Qubit Assay Kits for DNA and RNA (Life
Technologies) or NanoDrop ND-1000 spectrophotometer
(NanoDrop Technologies).
24. Fluorescence-based capillary electrophoresis instrument, 2100
Bioanalyzer (Agilent Technologies).
25. TBE buffer (0.5): 45 mM TrisHCl (pH 8.3), 45 mM boric
acid, 1 mM EDTA.
496
26. Agarose (type LE) gel (1 %): Prepare using your standard
protocol in 0.5 TBE buffer. Add GelRed or ethidium bromide for visualization.
27. 1 kb Plus DNA Ladder (e.g., Life Technologies or Thermo
Fisher Scientific).
2.4 Library
Preparation
and Sequencing
2.5 Data Handling
and Bioinformatics
(ftp://ftp.ncbi.nlm.nih.gov/
1. Thermocycler.
2. 0.2 or 0.5 ml tubes: Should fit the thermocycler.
3. 1.5 ml tubes.
4. Column-based DNA extraction kit with proteinase K, e.g.,
QIAamp DNA Mini Kit (Qiagen) or PureLink Genomic DNA
Mini Kit (Life Technologies).
5. TRIzol LS Reagent (Life Technologies) or other acidguanidinium-phenol-based reagents, e.g., TRI Reagent (SigmaAldrich), TriPure (Roche Applied Science), or QIAzol (Qiagen).
6. Column-based RNA extraction kit, e.g., RNeasy Mini Kit
(Qiagen) or PureLink RNA Mini Kit (Life Technologies).
7. First-strand cDNA synthesis kit, e.g., SuperScript III FirstStrand Synthesis System (Life Technologies) and RevertAid
First-Strand cDNA Synthesis Kit (Thermo Fisher Scientific)
or Transcriptor First-Strand cDNA Synthesis Kit (Roche
Applied Science).
497
Methods
The sequential steps in the metagenomic-based approach outlined
below are summarized in Fig. 1. Carry out all laboratory procedures at room temperature unless otherwise specified.
3.1 Sample
Preparation
3.2 Enrichment by
Nuclease Treatment
and Nucleic Acid
Extraction
498
Enrichment by nuclease
treatment
Extraction of DNA & RNA
Amplification
Generation of platformspecific libraries
Sequencing
Sequence analysis and
assembly
Homology searches
against databases
Verification of results
499
500
Table 1
Reagent mixture for tag labeling DNA
Component
Volume (l)
FR26RV-N (10 M)
DNA template
10
1.5
1.5
Total volume
15
Table 2
Reaction mix 1 for tag labeling RNA
Component
Volume (l)
RNA template
10
1.5
Total volume
13.5
501
Table 3
Reaction mix 2 for tag labeling RNA
Component
Volume (l)
13.5
RT buffer (5)
DTT (0,1 M)
20.5
Table 4
Reaction mix for PCR amplification
Component
Volume (l)
0.5
2.5
Nuclease-free H2O
29
Total volume
50
502
Table 5
Thermal cycling conditions
Cycle step
Temperature (C)
Duration
Number of cycles
Initial denaturation
95
10 min
Denaturation
95
60 s
Annealing
58
60 s
Extension
72
60 s
Final extension
72
10 min
40
Table 6
Restriction reaction mixture
Component
Volume (l)
26
0.5
Total volume
29.5
The generation of sequencing libraries as well as the actual sequencing is commonly performed by engaging a commercial provider or
an academic core facility. This paragraph will therefore be kept
brief and not outline a detailed protocol.
1. Since the preparation of sequencing libraries are platform specific, it is first necessary to select a sequencing platform. It has
been demonstrated that HTS data in the range of 1050 Mbp
is enough to identify viruses with the described approach [24].
Most current platforms have a capacity in this range or higher.
There are therefore other factors that might influence the
503
Fig. 2 Successfully amplified random products analyzed using (a) the 2100 Bioanalyzer DNA 1000 Assay and
(b) separation on a 0.8 % agarose gel. The Bioanalyzer produces a graph where the y-axis represents fluorescence units [FU] and the x-axis is time, which translates to fragment size in base pair (bp). This information is
also used to create a gel-like densitometry plot. On the agarose gel, sizes were determined using a 1 kb Plus
DNA Ladder. A smear is expected due to the randomness of the amplification procedure and the DNA size typical range from 200 to 1,000 bp, with the mean size ~375 bp
choice, such as read length, cost (both per run and per Mb of
DNA sequence), and ease of availability. For comparison,
major commercial HTS platforms suitable for metagenomicbased detection and their characteristics are summarized in
Table 7. A comprehensive review about HTS platforms has
also been made by Glenn et al. [25].
2. The necessary reagents for sequencing library generation can
be obtained as platform-specific commercial kits. It should
therefore be straightforward to prepare a sequencing library
according to the kit manufacturers protocol. In addition, the
use of specific short sequence tags (indexes) is supported by
most platforms. They can be added during the construction of
multiple libraries to allow multiplexing for increased throughput and reduced costs (for other considerations regarding
preparation of sequencing libraries, see Note 3).
3. Perform sequencing according to the platform manufacturers
protocol.
3.5 Data Handling
and Bioinformatics
~24 h
MiSeq
90 min
11 days
27 h
10 h
23 h
Run
timea
HiSeq 2500
(rapid)
35 h
26 h
GS Junior
Illumina
26 h
Library
preparation time
GS FLX Titanium
XL+
Roche/454c
Platforms
2 250
2 100
2 100
~400
~700
7.58.5 Gb
600 Gb
180 Gb
3550 Mb
700 Mb
Throughput
per run
~$1 K
~$23 k
~$6 k
~$1 k
~$6 k
Reagent
cost/run
~$0.13
~$0.04
~$0.04
~$20
~$8.6
Reagent
cost/Mb
Utility gradeb
Table 7
Characteristics and utility grades of HTS platforms suitable for metagenomic-based detection of potential pathogens
504
Fredrik Granberg et al.
24 h
24 h
Chip I
Chip II
~200
~200
~400
~400
~400
~100 Gb
~10 Gb
~1 Gb
400 Mb
1040 Mb
~$1 k
~$1 k
$750
$550
$350
~$0,01
~$0,1
~$0,75
~$1.38
~$8.5
23 h
318 Chip
46 h
23 h
316 Chip
Ion Proton
23 h
46 h
314 Chip
506
507
Notes
1. On the use of bioinformatics, centers, and in-house solutions.
One of the major obstacles to using HTS methodologies for
detection of viruses is the need for server solutions for bioinformatics analysis as well as for storage of data. Most sequencing centers provide basic bioinformatics as part of their
services, but for more in-depth analysis, the baseline service
might not be sufficient. The best cost/benefit solution is to
attach a bioinformatician to the group and access either a
national high-performance center for computing or a local-/
university-based solution. The second best alternative is to
build the capacity yourself by expanding the group with bioinformaticians and server solutions for handling the data; there,
our recommendations are the minimum recommendations.
The third option is to outsource the analysis to a commercial
partner and/or another group with the needed expertise. This
is the least favored option due to the loss of control over a
major part of the project.
2. Kits from other manufacturers can be used, but it is preferable
to use nucleic acid extraction systems incorporating a spin or
vacuum column. This allows for efficient removal of inhibitors
since nucleic acid trapped in a silica gel membrane can be very
effectively washed before elution [28].
3. For each sample, the SISPA products (from DNA and cDNA,
respectively) can be pooled (unless there is a specific reason not
to). Additionally, when sequencing SISPA material on HTS platforms that permit longer read lengths (300600 bp), an alternative to fragmentation is to perform direct size selection since the
majority of the SISPA products already are within this size range,
as illustrated in Fig. 2. This can be performed by traditional gel
electrophoresis or by an automated preparative gel electrophoresis system, such as the Pippin Prep (Sage Science).
4. Most sequencing platforms can deliver the data in a variety of
output formats, including FASTQ or FASTA with a corresponding
quality (QUAL) file. One noticeable exception is the Roche/454
508
509
Acknowledgments
The authors would especially like to thank Professor Jos Manuel
Snchez-Vizcano (Animal Health Department, Complutense
University of Madrid, Madrid, Spain) and his research group for
their contribution to the original paper on which this chapter was
based. This work was supported by Epi-SEQ, a research project
supported under the 2nd joint call for transnational research projects by EMIDA ERA-NET (FP7 project nr 219235); by the Award
of Excellence (Excellensbidrag), provided to SB by the Swedish
University of Agricultural Sciences (SLU); and by/executed in the
510
511
Chapter 34
Proteomics Characterization of Tick-Host-Pathogen
Interactions
Marina Popara, Margarita Villar, and Jos de la Fuente
Abstract
Ticks are blood-feeding arthropod ectoparasites of wild and domestic animals that transmit disease-causing
pathogens to humans and animals worldwide and a good model for the characterization of tick-host-pathogen
interactions. Tick-host-pathogen interactions consist of dynamic processes involving genetic traits of hosts,
pathogens, and ticks that mediate their development and survival. Proteomics provides information on the
protein content of cells and tissues that may differ from results at the transcriptomics level and may be
relevant for basic biological studies and vaccine antigen discovery. In this chapter, we describe various
methods for protein extraction and for proteomics analysis in ticks based on one-dimensional gel electrophoresis to characterize tick-host-pathogen interactions. Particularly relevant for this characterization is
the use of blood-fed ticks. Therefore, we put special emphasis on working with replete ticks collected after
feeding on vertebrate hosts.
Key words Tick-host-pathogen interactions, Proteomics, Protein extraction, Mass spectrometry,
LC-MS/MS, Electrophoresis
Introduction
Ticks are blood-feeding arthropod ectoparasites of wild and domestic animals that transmit disease-causing pathogens to humans and
animals worldwide [13]. Tick-host-pathogen interactions consist
of dynamic processes involving genetic traits of hosts, pathogens,
and ticks that mediate their development and survival [24]. In the
early 1990s, a cost-effective alternative for tick control became
commercially available with the development of vaccines reducing
the use of acaricides and the problems associated with them such as
selection of acaricide-resistant ticks and the contamination of the
environment and animal products with pesticide residues [5].
However, new vaccines are needed for efficient control of vector
infestations and pathogen infection and transmission [6].
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4_34, Springer Science+Business Media New York 2015
513
514
Materials
All reagents used for buffer preparations need to be of analytical
grade. The solutions are prepared with ultrapure water and stored
at 4 C, except for the solutions containing SDS that are stored at
20 C to avoid detergent precipitation. Reagents for protein digestions and mass spectrometry analysis need to be of liquid
chromatography-mass spectrometry (LC-MS) grade.
2.1
Tick Samples
Tick Proteomics
2.2 Protein
Extraction Buffers
515
2.3 Laemmli
Sample Buffer
0.125 M TrisHCl, pH 6.8, 4 % SDS, 20 % glycerol, 0.004 % bromophenol blue, and 10 % -mercaptoethanol. Mix 1.51 g of Tris
HCl, 4 g of SDS, 20 ml of glycerol, and 0.004 g of bromophenol
blue and bring up the volume to 100 ml with water. Make aliquots
of 1 ml and store at 20 C. Supplement the buffer with 10 % final
of -mercaptoethanol before use.
1. Resolving gel buffer: 1.5 M TrisHCl, pH 8.8. Weigh 181.7 g
of Tris, add water to a volume of 900 ml, adjust to pH 8.8 with
HCl, and make up to 1 L.
2. Stacking gel buffer: 1.5 M TrisHCl, pH 6.5. Weigh 181.7 g
of Tris, add water to a volume of 900 ml, adjust to pH 6.5 with
HCl, and make up to 1 L.
3. Resolving gel: Mix 2.5 ml of resolving gel buffer, 4 ml of 30 %
acrylamide/bis-acrylamide solution (Bio-Rad, Hercules, CA,
USA), 100 l of 10 % SDS, and 4 ml of water and mix well.
Add 100 l of 10 % ammonium persulfate and 5 l of TEMED
and cast gel.
516
1. Ice-cold acetone.
2. Trifluoroacetic acid.
3. 0.1 % formic acid.
4. 10 % (v/v) acetonitrile.
5. Methanol.
6. Chloroform.
7. GelCode Blue Stain Reagent (Thermo Scientific, San Jose,
CA, USA).
8. BCA Protein Assay (Thermo Scientific, San Jose, CA, USA).
9. Liquid nitrogen.
10. Sterilized mortar and pestle.
11. Syringe equipped with a 21 gauge needle.
12. OMIX pipette tips C18 (Agilent Technologies, Santa Clara,
CA, USA).
13. Ultrasonic cooled bath.
14. SDS-PAGE apparatus.
15. Centrifuge.
16. Freezer (20 C).
17. Vertical rotating shaker.
18. Confocal microscope.
Tick Proteomics
517
Methods
3.1 General
Considerations
Carry out all the procedures at 4 C until the SDS-containing buffer is used, requiring 20 C to avoid detergent precipitation:
1. Remove the ticks from the ethanol storage and leave for
12 min in a fume hood to evaporate excess of ethanol.
2. Remove the cuticle by dissecting the ticks using a confocal
microscope and add 10 mM PBS for constant hydration of the
tissues during dissection (see Note 3).
3. Homogenize the tick internal organs by quick-freezing in liquid
nitrogen and pulverizing with a sterilized mortar and pestle.
4. Add at least 2 ml of protein extraction buffer 1, 2, 3, or 4 per
100 g of tissue. Homogenize in a glass homogenizer with a
minimum of 10 strokes of a glass rod.
5. Sonicate the sample for 1 min in an ultrasonic cooled bath
followed by 10 s vortex and leave to rest on ice for 1 min.
Repeat these cycles 23 times or until the sample becomes
518
Buffer 1
Buffer 2
Buffer 3
Buffer 4
Ixodida
62.9 %
48.2 %
44.4 %
82.0 %
Pecora
72.4 %
44.2 %
40.2 %
72.7 %
a
The efficacy of different detergents on tick protein extraction is compared by using
protein extraction buffers 14. We have observed that extraction with buffer 4 gives the
highest yield for both tick and host proteins identification, compared to the detergentbased buffers 13. Of the three detergent-based buffers, SDS-containing buffer 1 shows
the best efficiency in proteins extraction, whereas the nonionic detergents in buffers 2
and 3, Triton X-100 and Tween 20, show similar values for protein extraction. Proteins
are expressed as percentages of the total proteins identified
Fig. 1 Representative one-dimensional SDS-PAGE gel showing protein band patterns in extracts prepared using different protein extraction buffers. (1) Proteins
extracted with buffer 1. (2) Proteins extracted with buffer 2. (3) Proteins extracted
with buffer 3. (4) Proteins extracted with buffer 4. MW: molecular weight markers
(PageRuler Plus Prestained Protein Ladder, Scientific, San Jose, CA, USA). Twenty
micrograms of soluble fractions are run on a 5 mm wide conventional 12 % SDSPAGE gel performing electrophoresis at 180 V of constant voltage. Bands are visualized by staining with GelCode Blue Stain Reagent by manufacturers protocol
Tick Proteomics
3.3 Protein
Fractionation Method
519
3.4 Proteome
In-Gel Digestion
520
Buffer 1
Buffer 4
b
300
Supernatant proteins
250
150
100
50
Fig. 2 The subcellular distribution of the extracted proteins depends on the buffer used. (a) Subcellular distribution of identified tick proteins after extraction with buffer 1 and buffer 4. We observed that buffer 4, a more
astringent buffer, allows the extraction of a greater number of internal organelle and membrane proteins. (b)
Number of identified tick and host proteins in the different fractions obtained using the buffer 1. An average of
41 % increase in the number of tick proteins and up to 72 % for host proteins is detected when crude plasma
membrane fraction is included in the analysis compared to the supernatant fraction only. Therefore, it is necessary to process both fractions, cytoplasmatic soluble, and plasma membrane pellet, obtained after detergent
extraction and separated by ultracentrifugation, in order to characterize the entire proteome. Identification is
performed using the SEQUEST algorithm of Proteome Discoverer 1.3 against Ixodida and Ruminantia databases for tick and host proteins identification, respectively. An FDR<0.05 for tick and an FDR<0.01 for host
proteins identification are considered as cutoff. Abbreviations: Cyt, cytoplasmic soluble protein fraction; PM,
plasma membrane protein fraction
Tick Proteomics
521
3.6 Proteomics
Data Analysis
3.7 Hemoglobin
Removal
522
RI: 2.51E7
55.16
100
Number of proteins
90
Relative Abundance
80
70
60
Hemoglobins
55.40
50
99.51
54.62
40
45.37
30
20
52.90
67.66
59.47
74.18 77.25
51.98
60.51
10
6.05 11.60
0
100
12.40
18.01
67.32
40.67 41.65
24.66 27.82
90.07
92.77
93.31
93.82
Tick
proteins
101.30
86.64
68.75
72.45
79.34 83.95
101.81
104.01 109.38 114.33 121.22
97.63
Host
proteins
128.23
54.00 57.18
90
80
70
RI: 1.66E7
100.74
60
50
40
99.52
73.62
30
49.45
43.57
35.12 41.34 46.11
39.88
20
10
9.94
7.83 9.41 11.60 17.74 20.91
10
20
59.12
30.04 34.31
30
40
50
60
68.27
74.57 81.99
67.72
87.91 91.79 94.45
67.07 73.14 75.10
71.45
96.34
75.83
70
Time (min)
80
90
101.33
100
102.37 109.68
110
116.54 120.27
120
125.28
130
Number
of proteins
51.98
Hemoglobins
after acetone
precipitation
Hemoglobins
after
chloroform
precipitation
Fig. 3 Removal of host hemoglobin from tick protein samples. (a) Representative protein distribution in
engorged tick samples. (b) Mass spectra comparing the MS/MS fragment peptides of two precipitation methods: acetone- (above) and chloroform-based method (below). The same profile is observed with a slight
increase in the intensity of the spectra using the acetone precipitation method. (c) Presence of hemoglobins in
the sample before and after treatment. Chloroform-methanol precipitation decreases the number of detected
hemoglobins in a total sample where hemoglobins represented 14 % of total of proteins identified compared
to 23 % of hemoglobins present after acetone precipitation. Additionally, the hemoglobins that remain in the
sample are identified with a lower number of peptides. This method is therefore very helpful in treating the
engorged tick samples, but it does not completely eliminate host hemoglobins
Tick Proteomics
523
Fig. 4 Identification of tick, host, and pathogen proteins in a single sample with a high level of confidence.
Protein extract of engorged ticks was obtained by total protein extraction method with buffer 4. (a) Database
search is performed against a database composed of Uniprot-Ruminantia.fasta, Uniprot-Ixodida.fasta, and
Uniprot-Alphaproteobacteria.fasta. Data is analyzed using the SEQUEST algorithm of Proteome Discoverer 1.3
software applying a 1 % FDR as criteria for assignations. (b) The application of de novo sequencing software
PEAKS Studio 6.0 significantly increases the number of identified proteins, primarily tick and pathogen proteins that are usually masked by host proteins in engorged ticks
3.9 De Novo
Sequencing and
Homology Analyses
PEAKS Studio v 6.0 software (Bioinformatics Solutions Inc.) permit protein identification by generating de novo peptide sequences,
which is especially useful when working with organisms such as
ticks with limited sequence information available. These analyses
may be done with the same general parameters as for the routinely
applied software Proteome Discoverer 1.3 (as described in
Subheading 3.6). Additionally, in PEAKS a special algorithm is
used to generate de novo sequences of the input spectrum, and the
SPIDER module is used to identify variations from sequences
using a homology match query. The filtering of the scores for all
identified peptides is done by assigning a 10lg P value of 30 that
524
Proteomics analyses are intended not only to obtain the identification of as many proteins as possible in a given sample but also to
allow focusing on the differences between several samples of interest, which is possible through the application of quantitative proteomics techniques. Quantitative proteomics approaches can be
divided into label-based or label-free methodologies depending
on the use or not of stable isotopic or isobaric labeling reagents to
mark the proteins or peptides under comparison before mass
spectrometry analysis. Additionally, gel-based and gel-free
approaches using multidimensional chromatography can be developed for quantitative proteomics analyses (a good review of these
quantitative methodologies can be found in [19]). The combination of more than one technique is the best way to obtain greater
proteome coverage and to identify a bigger number of differentially represented proteins between samples due to the complementarity of the quantitative approaches. The application of
quantitative proteomics to the characterization of the tickpathogen interface is still in its infancy [2], but its development is
crucial for the identification of proteins involved in pathogen
infection, multiplication, and transmission, as well as possible
antigens for vaccine development.
Notes
1. When working in field experiments such as epidemiology
studies or vaccine trials, collected ticks are generally stored in
ethanol and not deep-frozen because it is easier under field
conditions and makes their shipment from one lab to another
cheaper. Therefore, this work focused on optimizations on this
kind of samples.
2. Triton X-100 and Tween 20 detergents are diluted to a 10 %
stock solution to reduce their viscosity and facilitate pipetting.
SDS is also prepared in a 10 % stock solution in order to work
in a similar manner.
3. Removing the cuticle from a tick enables a better detection of
low-abundance tick proteins. Our previous studies showed
that processing the entire tick results in the detection of over
Tick Proteomics
525
526
Acknowledgments
We thank the CMBSO proteomics facility (Centro de Biologa
Molecular Severo Ochoa, Madrid, Spain) for technical assistance.
This research was supported by grants BFU2011-23896 and the
EU FP7 ANTIGONE project number 278976. M. Popara is an
early-stage researcher supported by the POSTICK ITN (postgraduate training network for capacity building to control ticks and
tick-borne diseases) within the FP7-PEOPLEITN program
(EU Grant No. 238511).
References
1. de la Fuente J, Estrada-Pea A, Venzal JM et al
(2008) Overview: Ticks as vectors of pathogens that cause disease in humans and animals.
Front Biosci 13:69386946
2. Villar M, Popara M, Bonzn-Kulichenko E
et al (2012) Characterization of the tickpathogen interface by quantitative proteomics.
Ticks Tick Borne Dis 3:154158
3. de la Fuente J, Kocan KM, Blouin EF et al
(2010) Functional genomics and evolution of
tick-Anaplasma interactions and vaccine development. Vet Parasitol 167:175186
4. Galindo RC, de la Fuente J (2012)
Transcriptomics Data integration reveals JakSTAT as a common pathway affected by pathogenic intracellular bacteria in natural reservoir
hosts. J Proteomics Bioinform 5:108115
5. de la Fuente J, Almazn C, Canales M et al
(2007) A ten-year review of commercial vaccine performance for control of tick infestations on cattle. Anim Health Res Rev 8:
2328
6. de la Fuente J (2012) Vaccines for vector control: exciting possibilities for the future. Vet J
194:139140
7. Villar M, Torina A, Nuez Y et al (2010)
Application of highly sensitive saturation labeling to the analysis of differential protein
8.
9.
10.
11.
12.
13.
Tick Proteomics
14.
15.
16.
17.
527
INDEX
A
Accreditation ........................................................ 80, 81, 119
Accuracy .............................................................. 4, 8, 13, 20,
27, 28, 31, 40, 49, 50, 56, 58, 61, 62, 84, 86, 90,
91, 9597, 110, 134, 145, 147, 151, 165, 207, 237,
246, 257259, 270, 288, 295, 296, 298, 313, 357,
362, 369, 398, 429, 448, 459, 492
Actinobacillus
A. equuli subsp. haemolyticus ................................ 235, 242
A. pleuropneumoniae .................................... 235, 236, 242
A. seminis ....................................................................... 22
Aerial animals .......................................................................3
AFLP-PCR......................................................................292
African swine fever virus (ASFV) .................... 209217, 255
Agglutination test .................................................................4
Alkaline phosphatase .......................................... 24, 211, 215
Allantoic fluid.....................................................................71
Alternating electrical field ........................................ 236, 323
Amblyomma ....................................................................... 514
Amplified fragment length polymorphism .......................292
Analytical accuracy .............................................................96
Analytical sensitivity ....................................... 11, 9193, 261
Analytical specificity...........................................................93
Ancylostoma
A. caninum ....................440, 442, 443, 445, 446, 453455
A. duodenale ..........................................437, 439, 442, 446
Animal health ...................................................... 3, 7, 15, 35,
37, 55, 56, 78, 110, 126, 153, 193, 254, 279
Animal welfare ........................................3, 15, 254, 255, 486
Anthelmintics .................... 145, 422, 429, 437, 441, 442, 458
Anthrax .................................................................... 109, 361
Anthropic ...........................................................................28
Antibiotic ........................................................ 115, 195, 250,
299, 300, 425, 427, 429, 478, 482, 487
Antifading mounting media ............................. 226, 227, 231
Antimicrobial resistance ..........................4, 15, 301, 424, 427
Antimicrobial therapy......................................... 4, 7, 10, 256
Aptamers ..................................................................275276
Aquatic animals ................................................................258
Ascaris
A. suum ................................................................ 448, 452
Assay performance................................................. 12, 21, 24,
77, 8385, 87, 94, 99102, 104, 151, 274
Asymmetric PCR .............................................................133
B
Babesia
B. bigemina .......................................................... 475487
B. bovis ........................................................................ 475
Babesiosis ......................................................... 475, 480, 481
Bacillus anthracis .........................118, 298, 361371, 401411
Backward inner primer (BIP) .......................... 165, 170172,
176178, 180
Barbers pole worm ...........................................................439
Basic local alignment software tool (BLAST) ......... 449, 483,
487, 493, 496, 506, 508, 509
Beta-actin gene ...................................................................26
Biobanking ...................................................................4359
Biochip .............................................................................117
Biocontainment .......................................33, 3740, 365, 404
Biodiversity...................................................................15, 56
Biohazard ............................................................... 27, 36, 45
Bioinformatics ................................................23, 27, 56, 318,
420, 438, 448453, 457, 459461, 479, 491, 493,
496, 503507, 509, 517, 523
Biological resources .................................... 4446, 48, 5457
Biosafety ...................................................28, 3141, 72, 135,
260, 268, 269, 365, 383, 404
Biosecurity .................................................................. 38, 257
Biosensors....................................14, 256, 266268, 273, 274
Bioterrorism .....................................................................255
BLASTn .................................................. 449, 506, 508509
BLASTp...........................................................................449
BLASTx ........................................................... 449, 506, 509
Blocking solution.............................................. 211, 213, 217
Blood samples.............................. 6, 7, 50, 175181, 264, 522
Bluetongue .......................................... 3, 7, 25, 125131, 425
Borrelia burgdorferi ............................................................ 296
Bos
B. indicus ...................................................................... 175
B. taurus ....................................................................... 175
Mnica V. Cunha and Joo Incio (eds.), Veterinary Infection Biology: Molecular Diagnostics and High-Throughput Strategies,
Methods in Molecular Biology, vol. 1247, DOI 10.1007/978-1-4939-2004-4, Springer Science+Business Media New York 2015
529
C
Caenorhabditis elegans ........................................ 438, 451452
C6 amino linker.................................183184, 186, 187, 190
Campylobacter
C. coli ............................................297, 300, 311320, 424
C. fetus ......................................................................... 292
C. jejuni ........................ 291, 297, 300, 311320, 375, 427
Campylobacteriosis...........................................................311
Canonical SNPs (canSNPs) .................................... 362368,
401, 402, 404, 410, 411
Capillary electrophoresis.......................................... 292, 293,
335347, 363, 494, 495
Carbon nanotube (CNTs) ................................................ 275
Cas genes .................................................................. 373, 374
cDNA library cloning ....................................... 479, 482483
cDNA microarray technology ..........................................445
Cell cultures......................................................20, 26, 46, 48,
5051, 53, 194, 197, 204, 205, 251, 425, 482, 491
Chabertia ovina .................................................. 146, 148, 149
Chemiluminescence...................184, 186, 189191, 267, 376
Chlamydia
C. abortus ..............................................194, 195, 204, 297
C. avium .............................................................. 195, 204
C. gallinacea ......................................................... 195, 204
C. pecorum ............................................................ 195, 204
C. pneumoniae .............................................. 194, 195, 204
C. psittaci .............................................194, 195, 202, 204,
206, 291, 296, 297, 391393, 395399
C. suis................................................................... 195, 204
C. trachomatis.................... 5, 194, 195, 204, 392, 394398
Chlamydiaceae ................................. 46, 51, 194, 204, 206, 391
Chlamydophila ........................................... 194, 203204, 206
Classical diagnostic methods ................................ 8, 111, 426
Classical swine fever ............................ 25, 114, 255, 258, 425
Clinical accuracy ..................................................... 49, 96, 97
Clonorchis sinensis............................................................... 448
Clostridium difficile ........................................................ 5, 114
Clustered regularly interspaced short
palindromic repeats (CRISPR) ............... 373388
D
Data recording ........................................................ 45, 46, 58
de Bruijn graph......................................................... 448, 449
Deparaffinization.............................................. 225229, 232
Dephasing ........................................................................420
Dermacentor ....................................................................... 514
Diagnosis ........................................ 4, 19, 31, 44, 61, 77, 110,
125, 134, 145, 153, 163, 176, 184, 193, 209, 236,
245, 254, 298, 335, 350, 362, 393, 402, 416, 438
sensitivity ..................................................... 8, 12, 15, 26,
90, 9394, 110, 151, 268, 275
specificity ................................................8, 12, 15, 26, 90,
9394, 110, 151, 275
4,6-Diamidino-2-phenylindole (DAPI) ......... 226, 231, 232
Dice coefficient......................................................... 331, 332
Digoxigenin ......................................................................210
Direct repeats (DRs) .........................295, 374376, 383, 387
Direct variable repeat polymerase
chain reaction (DVR-PCR) ............................375
DNA arrays ................................................ 25, 115116, 196
DNA Data Bank of Japan (DDBJ) ..................................450
Domestic animals ................ 15, 109, 133, 255, 426, 513, 514
DR region......................................................... 374376, 383
Dual-labeled probe ........................................... 113, 133142
Dual priming oligonucleotide system (DPO) .......... 140, 402
E
Echinococcus multilocularis .................................................. 456
Eimeria .............................................................................. 422
Electrophoresis .................................................14, 22, 24, 39,
112, 118, 125, 141, 156, 158, 177179, 181, 188,
194, 199, 202, 206, 288, 291293, 312, 314319,
326, 336, 339, 345, 347, 354, 355, 357, 363, 381,
383, 394397, 481, 482, 487, 507, 514, 517, 518
F
False negative .................................................. 1113, 20, 25,
26, 72, 90, 91, 98, 101, 154, 161, 250, 259, 262
False positive ................................................8, 24, 25, 72, 90,
98, 101, 112, 130, 164, 250, 258, 259, 265
Farm .......................14, 27, 145, 256, 257, 324, 332, 424, 426
Farmed animals ................... 3, 51, 56, 61, 235, 254, 255, 324
Fasciola
F. gigantica ................................................................... 448
F. hepatica .................................................................... 448
Fast Green solution .................................................. 211, 213
Fecal flotation ...................................................................147
Feces samples....................................................................399
Feline immunodeficiency virus (FIV)...........................11, 12
Field tests ......................................................... 256, 260, 264
Fish infectious diseases .....................................................163
Fit-for-purpose...................................................... 13, 22, 77,
78, 80, 84, 85, 90, 99, 100, 104
Fixation of tissue ......................................................225228
flaA-RFLP.........................................312, 313, 315, 317318
Flagellin genes ..................................................................291
Flotation of nematode eggs ...................................... 147, 149
Fluorescein isothiocyanate (FITC).................. 167, 170172,
231, 232
Fluorescence in situ hybridization (FISH) ................ 25, 115,
219, 222, 223, 225, 228, 229, 232
Fluorescence resonance energy transfer (FRET) ..............276
Fluorescent detection reagent (FDR) ............... 165, 167169
Fluorescent reporter .........................................................126
G
Gel-based PCR ........................................................ 112, 113
GenBank ......................................................... 229, 248, 358,
392, 450, 459, 506
Genomics .............................................23, 56, 263, 278, 288,
296, 416, 421, 427, 429, 444, 450, 452455,
475487, 514
partitioning ......................................................... 115, 116
wide typing .........................................................298301
Genomotyping ......................................................... 298, 299
Gold nanoparticles (AuNPs) ............................ 245251, 273
Gold standard ...................................................12, 20, 25, 26,
8788, 90, 94, 96, 134, 292293, 312
Good laboratory practices (GLPs) ................. 25, 47, 58, 210
H
Haemonchosis ..................................................................439
Haemonchus
H. contortus ..................................................146, 149, 151,
439, 440, 442445, 452454, 456, 458460
Hazard identification and assessment...........................3234
Hazard prevention and control .....................................3234
Helicase-dependent amplification ....................................114
HelmDB...........................................................................451
Hematoxylin .........................................................................4
Herd ............................................................ 74, 78, 257259,
266, 291, 324, 419, 423, 424
Highly pathogenic avian influenza (HPAI) ......................261
High resolution melting (HRM) ...................... 194, 361371
High throughput sequencing (HTS) ................. 27, 288, 299,
417, 446448, 450, 452454, 458, 461, 491493,
496, 502504, 507
Histophilus somni ......................................................... 22, 242
Hookworm ................................437441, 443, 446, 453, 455
Hot-start Taq DNA polymerase....................... 369, 404, 408
Housekeeping gene ........................................... 26, 238, 288,
296298, 312, 356, 359, 480
Humic acids.............................................................. 151, 271
Hybridization buffer .................................178, 199, 202, 210,
213, 226, 227, 230233, 248, 249, 395, 397, 404
Hybridoma ...................................................................46, 52
Hydrolysis probes .............................................................126
I
Immunofluorescence ....................................................4, 111
Immunomagnetic separation (IMS) .........................153161
K
k-mers...............................................................................449
L
Lab-on-a-chip (LOC)................................ 14, 117, 270272
Large sequence polymorphisms (LSPs)..............................23
Lateral flow strips ....................................... 14, 167, 170, 172
Leptospira
L. alexanderi ........................................................ 351, 356
L. biflexa ...................................................................... 349
L. borgpetersenii ........................................... 351, 356358
L. interrogans ....................................... 349, 351, 356359
L. noguchii............................................................ 351, 356
L. santarosai ......................................................... 351, 356
L. weilii ....................................................... 351, 356, 358
Leptospirosis ....................................................................349
Levey Jennings charts .........................................................82
LightUp probes ................................................................113
Limit of detection (LOD) ...............82, 9193, 118, 275, 410
Linearity ..........................................82, 94, 95, 276, 370, 447
Liner-after-the-exponential
PCR (LATE-PCR) ................................ 113, 265
Liposome ..........................................................................275
Listeria monocytogenes ........................................................ 293
Livestock ..................................................5, 25, 56, 134, 139,
145, 183, 254261, 279, 335, 361, 416, 421, 427,
429, 437, 439, 442
Loading buffer.......................................................... 326, 330
Loop-mediated isothermal
amplification (LAMP)....................7, 14, 27, 111,
114, 163172, 176181, 265, 266, 271
Loop primers .....................................114, 164, 165, 170172
Louping ill virus ...............................................................425
Low melting temperature agarose ............................ 326, 328
LSPs. See Large sequence polymorphisms (LSPs)
Luminex array .................................................. 185, 401411
LUX primers ....................................................................113
Lysozyme ................................................................. 232, 325
M
Macroarray ............................................................... 115, 184
Magnetic beads................................................ 118, 154, 155,
159, 274, 275, 277, 278, 409
Magnetic nanobeads .........................................................274
Magnetic particles ............................................ 274, 275, 278
MALDI-TOF MS ...................................................235243
Management ..............................................15, 3234, 36, 37,
4359, 80, 81, 85, 86, 97, 101, 256, 260, 265, 268,
301, 324, 336, 415429, 450, 459, 496
Mannheimia haemolytica .................................................... 235
Manual grinder .............................................................64, 69
Mareks disease .................................................................428
Massive parallel sequencing ......................................417421
Mass spectrometry (MS) ..........................235243, 274, 429,
514, 516, 521525
Mastitis..................................................6, 194, 323334, 429
Matrix-assisted laser desorption/ionization
time-of-flight (MALDI-TOF)...............235243
Measurement range ......................................................89, 95
Mechanical grinder..................................... 6465, 69, 72, 73
Melt curve analysis ........................................... 363, 366367
Melting temperature (Tm)............................... 113, 126, 148,
180, 181, 206, 232, 236, 357, 362, 365, 367, 368,
370, 371, 394, 404
Melt profile .......................................362, 365, 367, 369371
Metabolomics ...................................................................444
Metagenomics ........................................8, 23, 420, 425427,
429, 446, 491510
Metaviromics ....................................................................491
Microarrays...............................................7, 25, 52, 115116,
193207, 263, 268, 269, 273, 289, 290, 295299,
391399, 402, 445, 446, 452, 477
Microbiome ...................................................... 426, 427, 429
Microfluidics ............................................... 10, 14, 117118,
268272, 276, 279, 299
microRNA (miRNA) ....................................... 263, 427, 428
MIQE ........................................................................ 87, 102
Mismatch amplification
mutation assays (MAMA) ..............................363
Molecular beacons (MBs)................................. 113, 276278
Molecular epidemiology .................................... 23, 297, 312,
324, 375, 416, 424, 425, 429
N
Nanoarrays........................................................................263
Nanobiotechnology ..........................................................273
Nanomagnetic beads ........................................................274
Nanomagnets....................................................................274
Nanomaterials .................................................. 118, 119, 273
Nanoscale sensors ............................................. 118, 119, 245
Nanospheres .....................................................................274
O
Oesophagostomum
O. dentatum ..................................440, 445, 452455, 458
O. venulosum ........................................................ 146, 149
OIE listed diseases ......................................... 8, 19, 110, 193
ompA gene .................................291, 296, 391395, 398, 399
Open reading frames (ORFs) ...........................................449
Opisthorchis viverrini ......................................................... 448
Optofluidic devices ...........................................................267
Orientobilharzia turkestanicum........................................... 428
Ornithosis.........................................................................195
Ostertagia ................................................................... 437, 444
Overlap-layout-consensus ................................................448
Ovine chlamydiosis ..........................................................195
P
Padlock probe (PLP) ........................................ 115, 116, 120
Pandemic H5N1 avian influenza ......................................258
Paraffin-embedded tissues .....................4, 213, 226228, 232
Parasitic nematodes ...................437452, 455, 456, 458461
Pasteurellaceae ............................................................ 235243
Pasteurella multocida................................................... 235, 242
PCR. See Polymerase chain reaction (PCR)
PDB. See Protein Data Bank (PDB)
Pen side testing ...................................................................27
Peptide binders .................................................................154
Peptide nucleic acid (PNA) ...................................... 224, 276
PFGE. See Pulsed-field gel electrophoresis (PFGE)
Photodiode detectors ........................................................267
Piroplasm.................................................. 175, 180, 183191
Piroplasmosis ....................................................................183
PLA. See Proximity ligation assay (PLA)
Plexor primers ..................................................................113
Q
Quality assurance (QA) ............ 26, 40, 47, 48, 57, 58, 7273,
77104, 261, 369
Quality control (QC)........................................12, 20, 25, 40,
4548, 51, 58, 74, 77, 7984, 98, 99, 101, 261,
379, 427, 493, 506, 508
Quality management system (QMS)...................... 4551, 81
Quantification of target sequences (qPCR) ................ 25, 198
Quantitative assay .........................................................81, 94
Quantitative proteomics analyses ............................. 517, 524
Quantum dots (QDs) ....................................... 119, 273, 274
Quartz crystal microbalance (QCM)................................266
Quasispecies .............................................................420425
Quencher ...........................................113, 126, 127, 136, 276
R
Rabies ................................................................. 93, 109, 428
Random amplified polymorphic
DNA (RAPD) ................................ 288292, 301
T
Taenia ................................................................................ 422
Tams1-encoding gene .......................................................176
TaqMan .............................................................113, 126, 134,
135, 137, 138, 140, 363
Taq polymerase .................. 126, 130, 185, 186, 190, 198, 408
Teladorsagia circumcincta ............................................ 146, 445
Telemedicine ............................................................ 260, 267
Terrestrial animals ............................................................258
Texas red........................................................... 136, 231, 232
Theileria
T. annulata ............................................... 7, 175181, 187
T. lestoquardi ........................................................ 180, 187
T. parva ....................................................................... 180
T. taurotragi ................................................................. 180
Theileriosis .......................................................................175
Threshold cycle (Ct) ............ 25, 126, 129131, 146, 206, 369
Tick-borne disease ...........................................................476
Time-of-flight (TOF) mass spectra .................................236
Tissue samples ....................................................... 5253, 62,
6973, 134, 135, 137138, 141, 153, 160, 198,
220, 224, 225, 227, 233, 391399
UniGene ...........................................................................450
Unweighted pair group method using arithmetic
averages (UPGMA) ........................ 331, 332, 347
Uracil-DNA glycosylase (UNG) ......................................369
Urine samples ........................................................... 4, 48, 74
Usutu virus .......................................................................425
W
Westgard rules ..............................................................82, 94
West Nile virus (WNV) ........................................... 255, 425
Whipworm .......................................................................437
Whole genome sequencing (WGS) .......................... 23, 264,
298, 300, 301, 416, 424
Wild animals ....................................................... 3, 133, 254,
255, 426, 514
World Organisation for
Animal Health ................. 7, 37, 78, 110, 126, 193
WormBase ........................................................ 450, 451, 460
Y
Yersinia pestis ............................................................. 374375