17.11.2012 Views

WAVLD Symposium Handbook_V4.indd - csiro

WAVLD Symposium Handbook_V4.indd - csiro

WAVLD Symposium Handbook_V4.indd - csiro

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Symposium</strong> Proceedings<br />

HOSTS GOLD SPONSOR<br />

13TH INTERNATIONAL WORLD ASSOCIATION OF<br />

VETERINARY LABORATORY DIAGNOSTICIANS SYMPOSIUM<br />

11–14 November 2007<br />

Crown Promenade Melbourne AUSTRALIA<br />

www.wavld2007.com


WELCOME<br />

It is our pleasure to welcome you to the 2007 World Association of Veterinary Laboratory Diagnosticians biennial symposium<br />

held in conjunction with the World Animal Health Organisation (OIE). The timing of this meeting could not be more<br />

appropriate given the challenges brought by Avian Infl uenza, Bluetongue and Foot and Mouth Disease in Europe, Africa and<br />

Asia and, here in Australia, Equine Infl uenza. Equally, the attention given by regulatory authorities to biosafety, biosecurity,<br />

bio-terrorism and select agent management issues continues to occupy us.<br />

The organising committee have brought together a wide array of internationally renowned speakers. We would like to thank<br />

our generous sponsors for enabling us to achieve this.<br />

The fi rst day focuses on trans-boundary animal diseases with specialised sessions on avian infl uenza, foot and mouth<br />

disease and bluetongue. Prompted by the current epidemic in Australia, there is also a special session on equine infl uenza.<br />

Day Two belongs to the OIE and, chaired by Professor Steve Edwards, provides an insight into a wide array of new approaches<br />

to disease diagnosis. On Day Three we focus on issues that are critical to current laboratory management,<br />

including the use of new and developing technologies.<br />

Of course no meeting of this size can proceed without a social program and we hope that you will be well entertained.<br />

We have great pleasure in inviting you to our conference dinner, highlighted by a presentation by the Nobel Laureate<br />

Professor Peter Doherty. But let’s not forget that Melbourne is a beautiful city, well known for its many attractions and vast<br />

range of restaurants for you to enjoy.<br />

Whether a visitor from overseas or Australia, we welcome you to Melbourne for what we hope will be a most memorable<br />

Scientifi c and Social Program.<br />

Martyn Jeggo Peter Kirkland<br />

Joint Chair <strong>WAVLD</strong> 2007 Joint Chair <strong>WAVLD</strong> 2007<br />

Director President<br />

CSIRO Livestock Industries AAVLD<br />

Australian Animal Health Laboratory<br />

CONTENTS<br />

WELCOME & CONTENTS ..............................................................................................................................................3<br />

SPONSOR ACKNOWLEDGEMENT & COMMITTEE..........................................................................5<br />

INVITED SPEAKERS ..................................................................................................................................................8 – 9<br />

PROGRAM .......................................................................................................................................................................10 – 13<br />

POSTER LISTING ....................................................................................................................................................16 – 17<br />

EXHIBITION FLOORPLAN ........................................................................................................................................18<br />

EXHIBITOR LISTING .......................................................................................................................................................19<br />

SOCIAL PROGRAM & DINING IN MELBOURNE .................................................................20 – 21<br />

<strong>WAVLD</strong> ORAL PRESENTATIONS ......................................................................................................22 – 122<br />

<strong>WAVLD</strong> POSTER PRESENTATIONS ...........................................................................................123 – 225<br />

<strong>WAVLD</strong> 2007, 11 – 14 November 2007 | 3 4


SPONSORSHIP<br />

SPONSORS<br />

<strong>WAVLD</strong> 2007 extends its appreciation to the following sponsors for their support:<br />

GOLD SPONSOR<br />

SILVER SPONSORS<br />

Includes Sponsorship of:<br />

SYMPOSIUM DINNER<br />

MORNING OR AFTERNOON TEAS<br />

DELEGATE SPONSORSHIP<br />

Welcome Reception<br />

ORGANISING COMMITTEE<br />

Dr Martyn Jeggo<br />

Co-Convenor<br />

Dr Peter Kirkland<br />

Co-Convenor<br />

Dr Peter Daniels<br />

Secretary<br />

Dr Russell Graydon<br />

Local Arrangements Committee Chair<br />

Delegate Name<br />

Badge & Lanyard<br />

Poster Area &<br />

Internet Cafe<br />

<strong>Symposium</strong> Cafe<br />

<strong>WAVLD</strong> 2007 SYMPOSIUM MANAGERS<br />

The Meeting Planners<br />

91–97 Islington Street<br />

Collingwood Victoria 3066<br />

tel: +61 3 9417 0888<br />

fax: +61 3 9417 0899<br />

email: wavld2007@meetingplanners.com.au<br />

W W W . M E E T I N G P L A<br />

N E R S . C O M . A U<br />

<strong>WAVLD</strong> 2007, 11 – 14 November 2007 | 5<br />

The pace of leadership.<br />

Institut Pourquier and IDEXX Laboratories are proud to join forces to sponsor<br />

the World Association of Veterinary Laboratory Diagnosticians.<br />

Renowned medical research and<br />

development since 1878.<br />

A wholly owned subsidiary<br />

of IDEXX Laboratories, Inc.<br />

© 2007 IDEXX Laboratories, Inc. All rights reserved. • 7406-00


The science of innovation.<br />

Institut Pourquier joined IDEXX Laboratories in March 2007 and together will<br />

continue unparalleled research, development and support of the most advanced<br />

animal health diagnostics, services and solutions in the world.<br />

Innovative production animal<br />

services since 1984. A worldwide<br />

leader in veterinary, food<br />

and water biotechnology.<br />

INVITED SPEAKERS<br />

Dr Tammy Beckham<br />

Dr Beckham received her Doctorate of Veterinary<br />

Medicine from Auburn University in 1998. While still<br />

at Auburn, she furthered her education through a<br />

Doctoral program, which allowed her to conduct<br />

research in support of her dissertation at the U.S.<br />

Army Medical Research Institute for Infectious<br />

Diseases (USAMRIID), in Frederick, MD. Dr Beckham received her<br />

Doctorate in 2001. Following completion of her Doctorate,<br />

Dr Beckham fostered her interest in diagnostic assay<br />

development and validation through positions with the<br />

Department of Homeland Security and the USDA Animal and<br />

Plant Health Inspection Service (APHIS), Foreign Animal Disease<br />

Diagnostic Laboratory (FADDL). Dr Beckham recently became<br />

the Head of the Profi ciency and Validation Services Section at<br />

FADDL. In this position, Dr Beckham coordinates all aspects of<br />

FADDL’s role as a reference laboratory for the National Animal<br />

Health Laboratory Network (NAHLN). This includes overseeing<br />

the validation of assays; identifying, evaluating, and developing<br />

new and emerging technologies; and developing and organizing<br />

training and profi ciency testing programs.<br />

Professor Corrie Brown<br />

Corrie Brown received her B.Sc. in Animal<br />

Behavior from McGill University and her DVM<br />

from Ontario Veterinary College at the University<br />

of Guelph (1981). She completed a combined<br />

residency/PhD in Comparative Pathology at<br />

the University of California at Davis. Board<br />

certifi cation (ACVP) and PhD were both attained in 1986. She was<br />

an assistant professor of pathology at Louisiana State University<br />

briefl y before joining the U.S. Department of Agriculture at Plum<br />

Island, where, as Head of the Pathology Section, she specialized<br />

in the diagnosis and pathogenesis of foreign animal diseases.<br />

In 1996, she joined the University of Georgia College of Veterinary<br />

Medicine as Professor and Head of the Department of Veterinary<br />

Pathology. She currently serves as Coordinator of International<br />

Veterinary Medicine for the College of Veterinary Medicine. In<br />

2003, she was honored with the university’s highest teaching<br />

award, being named a Josiah Meigs Distinguished Teaching<br />

Professor. Her professional interests are in infectious diseases<br />

of food-producing animals, emerging diseases, and international<br />

veterinary medicine. She has published or presented over 250<br />

scientifi c papers and has testifi ed to Congress on issues involving<br />

agroterrorism. She has served on many industrial and federal<br />

panels, and been a technical consultant to numerous foreign<br />

governments on issues involving infectious diseases and animal<br />

health infrastructure.<br />

8 | <strong>WAVLD</strong> 2007, 11 – 14 November 2007<br />

Dr Ilaria Capua<br />

Dr Ilaria Capua graduated in Veterinary Medicine<br />

with honours (110/110 e lode ), University of<br />

Perugia , in February 1989. Obtained post<br />

graduate qualifi cations as a specialist in Animal<br />

Health and hygiene from Pisa University in 1991<br />

and PhD from University of Padua. She is currently<br />

Head of the Virology Department at Istituto Zooprofi lattico<br />

Sperimentale delle Venezie, Padova, Italy and Head of the<br />

National, FAO and OIE Reference Laboratories for Avian Infl uenza<br />

and Newcastle disease. Between 1999 and 2006 she has been<br />

directly involved in managing several AI and ND epidemics and<br />

in 2000 developed the “DIVA”-Differentiating Vaccinated from<br />

Infected Animals strategy, based on heterologous vaccination, to<br />

combat AI. This strategy, the fi rst ever developed to combat AI by<br />

vaccination still enabling trade of products resulted in eradication<br />

of AI at that time in Italy and was approved by the EC and by OIE.<br />

During her career as a veterinary virologist her work has been<br />

recognized with her nomination as OIE and FAO expert for AI and<br />

ND. Since 1995 she has been involved with the EU Commission,<br />

participating in meetings and working groups on viral diseases of<br />

poultry and mammals, including AI, ND, FMD, CSF and Crimean-<br />

Congo Haemorrhagic Fever. In 2005 she has been nominated<br />

the Chairman of OFFLU- the newly established OIE/FAO network<br />

on Avian Infl uenza. This network has the role of supporting<br />

developing countries in managing the AI crisis and offering<br />

veterinary expertise to complement the international efforts of<br />

the medical community in managing the pandemic threat posed<br />

by AI.<br />

Laureate Professor Peter Doherty<br />

Laureate Professor Peter Doherty shared the<br />

Nobel Prize in Physiology or Medicine in 1996<br />

with Swiss colleague Rolf Zinkernagel, for their<br />

discovery of how the immune system recognises<br />

virus-infected cells. He was Australian of the Year<br />

in 1997, and has since been commuting between<br />

St Jude Children’s Research Hospital in Memphis and the<br />

Department of Microbiology and Immunology at the University<br />

of Melbourne. His research is mainly in the area of defence<br />

against viruses. He regularly devotes time to delivering public<br />

lectures, writing articles for newspapers and magazines and<br />

participating in radio discussions. Peter Doherty graduated from<br />

the University of Queensland in Veterinary Science and became a<br />

veterinary offi cer. Moving to Scotland, he received his PhD from<br />

the University of Edinburgh Medical School. He is the fi rst person<br />

with a veterinary qualifi cation to win a Nobel Prize.


INVITED SPEAKERS CONTINUED<br />

Professor Steve Edwards<br />

Steve qualifi ed as a veterinarian from Cambridge<br />

University, England, in 1972. He spent four years<br />

in clinical farm animal practice in the Welsh<br />

border country before returning to university<br />

at Edinburgh to study for an MSc in Tropical<br />

Veterinary Science. This stimulated his continuing<br />

passion for laboratory-based diagnostic investigation work. He<br />

then spent two years working in the UK Government’s overseas<br />

aid programme in El Salvador, Central America, working<br />

alongside local counterparts to identify the disease constraints<br />

to animal production. Steve joined the Central Veterinary<br />

Laboratory (now VLA) at Weybridge in 1980 working on virology<br />

research. From 1991 Steve brought his interest and expertise<br />

in laboratory diagnostics to the OIE Standards Commission,<br />

eventually becoming President of that group (now renamed<br />

Biological Standards Commission) in 2003, a position he still<br />

holds. He took over as Director and Chief Executive of the VLA in<br />

2000 on the retirement of his predecessor. He has led the Agency<br />

through a period of consolidation, with an expansion of its external<br />

collaborative links, as well as coping with an apparently neverending<br />

succession of animal disease issues, including classical<br />

swine fever, foot and mouth disease, tuberculosis, BSE, rabies,<br />

Newcastle disease, and avian infl uenza.<br />

Dr Luis Rodriguez<br />

Dr Rodriguez obtained his DVM in 1979 at the<br />

National University of Costa Rica. From 1980 to<br />

1985 he attended graduate school at the University<br />

of Wisconsin-Madison. In 1982 obtained a M.Sc.<br />

and in 1985 a Ph.D. in veterinary science with<br />

emphasis in Animal Virology working in molecular<br />

virology of bovine herpesvirus 1. From 1985 to 1995 worked a the<br />

School of Veterinary Medicine, National University of Costa Rica<br />

where he taught veterinary virology and carried out research<br />

focusing on vesicular stomatitis. He established the Tropical<br />

Disease Research Program and obtained several grants from<br />

international organizations to carry out research in pathogens of<br />

human and veterinary importance. From 1995 –1997 worked at<br />

the Special Pathogens Branch, Division of Viral and Rickettsial<br />

Diseases of the Centers for Disease Control and Prevention, in<br />

Atlanta Georgia. In 1997 joined the USDA – ARS Foot-and Mouth<br />

Disease Research Unit Plum Island as lead scientist in charge<br />

of vesicular stomatitis research working on genomics and<br />

pathogenesis of VSV and in 2002 Dr Rodriguez became Research<br />

Leader of the Foot and Mouth Disease Research Unit. In 2004 he<br />

also established the Foreign Animal Disease Research Unit. Dr<br />

Rodriguez personal current research focuses on studying the early<br />

phases of FMDV infection and pathogenesis and insecttransmission<br />

of VSV in cattle.<br />

Professor Mark Rweyemamu<br />

A Tanzanian veterinarian. Former Head of the<br />

Infectious Diseases Programme of the Food and<br />

Agriculture Organization (FAO) of the United<br />

Nations at its Headquarters in Rome. From 1994<br />

to his retirement from FAO in 2002, he was the<br />

inaugural Head of the FAO special programme on<br />

infectious diseases, known as the Emergency Prevention System<br />

for Animal and Plant Pests and Diseases (EMPRES) which<br />

included the coordination of the Global Rinderpest Eradication<br />

Programme (GREP). Before moving to FAO Headquarters, he<br />

had set up the Pan African Veterinary Vaccine Centre in Addis<br />

Ababa, on behalf of FAO and the Organisation of African Unity<br />

(now African Union). He has worked in both governmental<br />

and industrial settings. In government: as Virologist and Chief<br />

Veterinary Research Offi cer for the Tanzanian Government<br />

and as Head of the Virus Diseases at the then East African<br />

Veterinary Research Organisation, Muguga, Kenya. In industry<br />

he has been the Director of Veterinary Vaccine Research for<br />

Pfi zer International and Head of FMD Vaccine Research for<br />

the Wellcome Foundation. His area of interest is on the major<br />

infectious diseases of animals, known as transboundary animal<br />

diseases. He is now a private consultant and a Visiting Professor<br />

at the Royal Veterinary College, University of London.<br />

Dr Linfa Wang<br />

Dr Linfa (Lin-Fa) Wang is a Senior Principal<br />

Research Scientist at the CSIRO Australian Animal<br />

Health Laboratory (AAHL), and a project leader in<br />

the Australian Biosecurity Cooperative Research<br />

Centre (AB-CRC) for Emerging Infectious Diseases.<br />

Dr Wang is a member of the WHO SARS Scientifi c<br />

Research Advisory Committee and participated<br />

in the WHO-FAO-Chinese Government joint mission to China on<br />

animal reservoir of the SARS-CoV and potential transmission<br />

to humans in August 2003. His research group played key<br />

roles in the discovery and molecular analysis of Hendra and<br />

Nipah viruses, which led to the establishment of a new genus<br />

Henipavirus to accommodate the newly discovered highly lethal<br />

zoonotic paramyxoviruses of bat origin. Dr Wang completed his<br />

science degree in 1982 at the East China Normal University,<br />

Shanghai, followed by a PhD in Biochemistry (Molecular Biology)<br />

from the University of California, Davis, USA in 1986, where he<br />

went on to become a Postdoctoral Research Fellow with the<br />

Department of Biochemistry. After moving to Australia in 1989,<br />

Dr Wang spent 18 months working at Monash University and<br />

then joined CSIRO in 1990. Dr Wang has more than 140 scientifi c<br />

publications to his name along with four patents and numerous<br />

conference abstracts. He is currently serving on fi ve international<br />

editorial boards for publications in the areas of virology,<br />

biotechnology and immunotechnology.<br />

Crown Promenade, Melbourne, Australia | 9<br />

PROGRAM<br />

Monday 12 November<br />

0700 Registration Open<br />

0815 – 1030 Opening Session<br />

Room: Promenade 1 & 2<br />

Welcome Address<br />

Martyn Jeggo & Peter Kirkland<br />

Opening<br />

Minister Joe Helper<br />

Plenary Sessions – Global Risks and Challenges<br />

Chair: Peter Daniels<br />

Prof Mark Rweyemamu<br />

Future Risks from Infectious Disease of Animals<br />

Prof Corrie Brown<br />

New & Emerging Diseases<br />

Dr Ilaria Capua<br />

Avian Infl uenza – Challenges and Opportunities for the Veterinary Profession<br />

Dr Linfa Wang<br />

Zoonotic Viruses of Bat Origin – Discovery and Diagnosis<br />

1030 – 1100 Morning Tea & Exhibition/Poster Viewing<br />

Location: Exhibition Area<br />

1100 – 1230 Room: Promenade 1 & 2 Room: Promenade 3<br />

Concurrent Session 1.1<br />

New & Emerging Diseases/Wildlife<br />

Chair: Dr Linfa Wang Chair: Dr Simone Warner<br />

Dr Kim Halpin<br />

Experimental models for Henipavirus infection: bats, cats and<br />

pseudo-rats<br />

Prof Paul Gibbs<br />

Emergence of canine infl uenza in the USA: developments since<br />

discovery in 2004 and the diagnostic challenge<br />

Ms Deborah Finlaison<br />

Investigations of viral myocarditis in pigs associated with a novel<br />

pestivirus.<br />

Mrs Melinda Frost<br />

Characterisation of a novel pestivirus associated with an outbreak<br />

of stillbirths and pre-weaning in pigs due to myocarditis<br />

Prof Stefan Vilcek<br />

How many pestiviruses circulate in nature? Pestiviruses in<br />

free-living animals: present status and problems<br />

Dr Julia Ridpath<br />

Characterization and detection of BVDV-related reproductive<br />

disease in white tail deer<br />

1230 – 1330 Lunch<br />

Location: Exhibition Area<br />

1330 – 1455 Room: Promenade 1 & 2 Room: Promenade 3<br />

Concurrent Session 1.2<br />

FMD<br />

Concurrent Session 2.1<br />

Molecular Diagnostic Assays – I (Bacterial diseases)<br />

Mrs Melissa Higgins<br />

Development of an improved identifi cation matrix for<br />

Aeromonas salmonicida<br />

Dr Ian Marsh<br />

An improved PCR-based test for QX disease confi rmation in<br />

Sydney Rock Oysters<br />

Dr Adrian Whatmore<br />

The development of SNP discrimination assays on a real-time<br />

PCR platform as a tool for the rapid identifi cation and speciation<br />

of Brucella.<br />

Dr Graeme Eamens<br />

Detection of Mycoplasma hyopneumoniae in pig samples using<br />

polymerase chain reaction tests<br />

Ms Kgomotso Sibeko<br />

Development and evaluation of a real-time PCR test for the<br />

detection of Theileria parva infections in cape buffalo<br />

(Syncerus caffer) and cattle<br />

Dr Wendy McDonald<br />

Detection and strain typing of Coxiella burnetii using Molecular<br />

Beacons in a real-time PCR<br />

Concurrent Session 2.2<br />

Avian Infl uenza<br />

Chair: Prof Mark Rweyemamu Chair: Dr Ilaria Capua<br />

Dr Divakar Hemadri<br />

Changing faces of foot-and-mouth disease type O Pan Asia strain<br />

India<br />

Dr Paul Kitching<br />

The 2001 UK foot-and-mouth disease outbreak:<br />

an update<br />

Speaker TBA<br />

FMD in the UK, 2007<br />

Ms Janine Muller<br />

Development and evaluation of a recombinant antibody-based<br />

competitive ELISA for FMDV<br />

antibody detection<br />

Mr Chris Morrissy<br />

International validation of a new 3abc C-ELISA for FMD serology<br />

10 | <strong>WAVLD</strong> 2007, 11 – 14 November 2007<br />

Mrs Janice Pedersen<br />

Results of 2006 wild bird surveillance for the detection of highly<br />

pathogenic avian infl uenza in the United States<br />

Dr Ian K. Barker<br />

Canada’s inter-agency wild bird infl uenza surveillance<br />

programme 2005 – 2007<br />

Dr Tze-Hoong Chua<br />

Performance evaluation of fi ve detection tests<br />

Dr Simone Warner<br />

Optimised sampling and processing for enhanced detection of<br />

avian infl uenza virus from fi eld samples<br />

Dr Michael Johnson<br />

Analysis of the sensitivity of H5N1 Isolates from the South East<br />

Asian region to Oseltamivir and Zanamivir


Monday 12 November (continued)<br />

1455 – 1525 Afternoon Tea<br />

Location: Exhibition Area<br />

1525 – 1720 Room: Promenade 1 & 2 Room: Promenade 3<br />

Concurrent Session 1.3<br />

Bluetongue & orbiviruses<br />

Concurrent Session 2.3<br />

Zoonoses, Pox viruses & other TADS<br />

Chair: Dr Ian Gardner Chair: Prof Corrie Brown<br />

Dr Carrie Batten<br />

An update on bluetongue virus in Europe<br />

Dr Heather Elliott<br />

From testing to trade: the response to the outbreak of BTV 8 in<br />

NW Europe<br />

Gudrun Delbridge<br />

Evaluation of ELISAs to detect antibodies to bluetongue virus in<br />

individual and tank milk samples<br />

Dr Sushila Maan<br />

Molecular epidemiology of bluetongue virus type 8 from the<br />

Netherlands 2006<br />

Prof Peter Mertens<br />

Sequencing and RT-PCR assays for genome segment 2 of the 24<br />

bluetongue virus serotypes: identifi cation of exotic serotypes in<br />

the Southeastern USA (1999-2006)<br />

Ms Karin Darpel<br />

Replication of bluetongue virus in the skin of<br />

infected sheep<br />

Dr Hume Field<br />

Emerging zoonoses and wildlife – a multi-disciplinary challenge<br />

Prof. Tin Tin Myaing<br />

Wild life as a source of zoonotic diseases<br />

Mr Keith Jahans<br />

Surveillance of Mycobacterium bovis infection in cattle in Great<br />

Britain during 2006<br />

Mr Claude Sabeta<br />

Spread of canid rabies into the Free State province of South Africa<br />

Dr Tim Bowden<br />

Capripoxvirus tropism and shedding: a quantitative time-course<br />

study in experimentally infected sheep and goats<br />

Dr David Boyle<br />

An indirect ELISA, based upon recombinant capripoxvirus<br />

antigens, for serological detection of sheeppox, goatpox and<br />

lumpy skin disease<br />

Mrs Eva Veronesi<br />

Validation of a diagnostic technique for the detection and<br />

quantifi cation of bluetongue virus in adult biting midges<br />

(Culicoides spp: Diptera: Ceratopogonidae)<br />

1745 – 1915 Special Plenary Sessions – Equine Infl uenza<br />

Room: Promenade 1 & 2<br />

Chair: Dr Bob Biddle<br />

Dr Peter Timoney<br />

Changing dynamics in the global distribution of equine diseases<br />

Richard Newton<br />

Global Overview<br />

Graeme Garner<br />

Australian Situation<br />

Dr Peter Daniels<br />

Diagnosis and Characterisation of Equine Infl uenza (EI) in Australia: A/equine/Sydney/2888-8/2007 H3N8<br />

Dr Peter Kirkland<br />

Equine infl uenza in NSW – challenges and highlights from a laboratory perspective<br />

1915 – 2115 Welcome Reception<br />

Location: Exhibition Area<br />

Crown Promenade, Melbourne, Australia | 11<br />

Tuesday 13 November<br />

0830-0945 OIE Biotechnology <strong>Symposium</strong><br />

Room: Promenade 1 & 2<br />

Co-Chairs: Prof Steve Edwards & Dr Gideon Brückner<br />

Welcome Address<br />

Prof Steve Edwards<br />

Dr Yi-Seok Joo<br />

Simple rapid, on-site (pen-side) detection of animal pathogens<br />

Dr Deborah Middleton<br />

Application of Biotechnology to Infections within Wildlife Hosts<br />

0945 – 1015 Morning Tea<br />

Location: Exhibition Area<br />

1015 – 1215 Dr Gideon Bruckner<br />

The OIE Concept of Twinning between Laboratories<br />

Dr Michael Macintosh<br />

Diagnostics of Emerging Infectious Diseases by Microarray<br />

Prof Noboru Inoue<br />

Development of Loop-Mediated Isothermal Amplifi cation (LAMP) Technique for Diagnosis of African Trypanosomosis<br />

Dr Alex Hyatt<br />

Diagnostic Electron Microscopy: Historical review and future<br />

1215 – 1315 Lunch<br />

Location: Exhibition Area<br />

1315-1445 Dr Norbert Bannert<br />

Overview of electron microscopy and its role in infectious disease diagnosis<br />

Dr Yves Robinson<br />

Telepathology: its role in disease diagnosis in meat hygiene<br />

Dr Paul Monaghan<br />

Understanding Host-Pathogen Interactions: Confocal and Live Cell Imaging in Veterinary Science<br />

1445 – 1515 Afternoon Tea<br />

Location: Exhibition Area<br />

1515-1645 Dr Brad J Marsh<br />

Multi-Resolution Spatio-Temporal Analysis of Mammalian Cells Reconstructed in 3D by Electron Microscope Tomography<br />

Dr Simon Carpenter<br />

Detecting and interpreting arboviral infections in insects<br />

Dr Hez Hird<br />

Molecular methods for speciation<br />

1900 – 2400 <strong>Symposium</strong> Dinner<br />

Location: RACV Club<br />

Wednesday 14 November<br />

0815 – 1000 Plenary Sessions – Preparing for the Future<br />

Room: Promenade 1 & 2<br />

Chair: Dr Russell Graydon<br />

Dr Martyn Jeggo<br />

Labs of the Future<br />

Dr Luis Rodriguez<br />

Biosafety and Biosecurity Concerns Of High Security Animal Pathogen Laboratories<br />

Dr Tammy Beckman<br />

Diagnostic Technologies for Veterinary Laboratories of the Future<br />

Prof Steve Edwards<br />

Can we believe Laboratory Test Reports?<br />

1000 – 1030 Morning Tea<br />

Location: Exhibition Area<br />

1030 – 1230 Room: Promenade 1 & 2 Room: Promenade 3<br />

Concurrent Session 1.4<br />

Concurrent Session 2.4<br />

New Technologies & Platforms<br />

Quality Assurance<br />

Chair: Dr Tammy Beckham Chair: Dr Luis Rodriguez<br />

Mrs Barbara Martin<br />

The United States National Animal Health Laboratory Network<br />

(NAHLN)<br />

Dr Bill Colston<br />

Developing an approach for rapid identifi cation of emerging<br />

biological threats<br />

Dr Juliet Dukes<br />

Viral identifi cation using microarrays<br />

Dr. Julia Ridpath<br />

Assay platforms for the rapid detection of viral pathogens by the<br />

ultrahigh sensitivity monitoring of antigen-antibody binding<br />

Prof Lawrence Wangh<br />

LATE-PCR with PrimeSafeTM – maximum diagnostic information<br />

from a closed-tube<br />

Dr Xingwang Fang<br />

A complete workfl ow for nucleic acid based high throughput<br />

pathogen detection<br />

Dr Carmelo Volpe<br />

An advanced fi eld deployable sample preparation and PCR system<br />

Dr Rod Reece<br />

The Australian Animal Pathology Standards program:<br />

preservation and digitisation of the collection, and access via<br />

‘new’ technologies<br />

12 | <strong>WAVLD</strong> 2007, 11 – 14 November 2007<br />

Dr. Paul Kitching<br />

Labs of the future – a cultural shift in Canada for foreign animal<br />

disease (FAD) detection<br />

Dr Craig Carter<br />

Laboratory-based early animal disease detection utilizing a<br />

prospective scan statistic<br />

Dr Philip Wakeley<br />

Development, validation and implementation of molecular<br />

diagnostics tests for important pathogens of farmed animals and<br />

wildlife – a pragmatic approach<br />

Dr Jane Oakey<br />

Quality assurance and control issues with PCR as a diagnostic tool<br />

Mr Lindsay Pritchard<br />

A review of training in molecular diagnostic techniques for avian<br />

infl uenza in South East Asia<br />

Dr Kim Halpin<br />

Development and consolidation of an EQA programme for the<br />

molecular diagnosis of avian infl uenza<br />

Dr. Hermann Unger<br />

How useful are test kits when applied in different target<br />

population: producer and end-user responsibilities from a<br />

developing country perspective<br />

Dr Mark M Williamson<br />

Laboratory management techniques for responding to client<br />

demands: diagnosis of swine pneumonia.


Wednesday 14 November (continued)<br />

1230 – 1330 Lunch<br />

Location: Exhibition Area<br />

1330-1500 Room: Promenade 1 & 2 Room: Promenade 3<br />

Concurrent Session 1.5<br />

Diagnostic assays – ELISA & molecular<br />

Concurrent Session 2.5<br />

Biosecurity, IT, & LIMS<br />

Chair: Dr Lorna Melville Chair: Dr Michael Johnson<br />

Dr Simon Anthony<br />

Complete sequence analyses of the genome of epizootic<br />

haemorrhagic disease virus (EHDV): the design of<br />

diagnostic assays.<br />

Dr Jovita Fernández<br />

New advances in the molecular diagnosis of African horse<br />

sickness (AHS)<br />

Dr Carmina Gallardo<br />

New approaches in molecular and serological diagnosis of<br />

African swine fever (ASF) on new emerging variants<br />

Dr Karen Harmon<br />

Rapid high-throughput real-time RT-PCR testing for PRRSV:<br />

problems and solutions<br />

Dr Roger Ayling<br />

Development and initial evaluation of a lateral fl ow device for the<br />

rapid detection of contagious bovine pleuropneumonia.<br />

1500 – 1530 Afternoon Tea<br />

Location: Exhibition Area<br />

1530 – 1700 Room: Promenade 1 & 2 Room: Promenade 3<br />

Concurrent Session 1.6<br />

Molecular Diagnostic assys- II (Viral)<br />

Dr Scott Fitzgerald<br />

Benefi ts realized from the fi rst biosafety level 3 large animal<br />

necropsy facility in a state diagnostic laboratory in North America<br />

Dr Steve Weber<br />

Linking multiple veterinary laboratory information<br />

management systems to support the National Animal Health<br />

Laboratory Network<br />

Dr Paul Collery<br />

Implementation, function and benefi ts of a laboratory information<br />

management system in a national veterinary laboratory service<br />

Dr Peter Durr<br />

Beyond the laboratory gate – enhancing a laboratory information<br />

management system (LIMS) for exotic animal disease<br />

preparedness<br />

Dr Paul-Michael Agapow<br />

The ReLaIS project: an epi-informatics constellation<br />

Concurrent Session 2.6<br />

Epidemiology & Surveillance<br />

Chair: Dr Gary Horner Chair: Dr Peter Daniels<br />

Mrs Katarzyna/Kasia Bachanek-Bankowska<br />

High throughput diagnostic PCR for bluetongue virus<br />

Dr William C. Wilson<br />

Nucleic acid diagnostic tools for early detection of<br />

arthropod-borne animal viral diseases.<br />

Dr Dennis Gomez<br />

Molecular detection of betanodaviruses from subclinically<br />

infected aquarium fi shes and invertebrates<br />

Dr Frank Wong<br />

Purifi cation and characterisation of a herpes-like virus infecting<br />

Australian populations of abalone, Haliotis spp.<br />

Dr Giovanni Cattoli<br />

Development of a real-time duplex TaqMan-PCR assay for the<br />

detection of equine rhinitis A and B viruses in clinical specimens<br />

1715 – 1730 <strong>Symposium</strong> Close<br />

Ms Kerri Tyrrell<br />

Effects of dynamic ‘communal’ interactions on disease processes<br />

and individual agent diagnosis where multiple infectious agents<br />

are present in sheep<br />

Dr Fraser Hill<br />

Use of molecular and epidemiological information for the costeffective<br />

diagnosis of BVD infection in New Zealand dairy cattle<br />

Eng Sandra Benevides<br />

Assessing the within herd prevalence of antibody positive cows to<br />

bovine herpevirus 1 using an Indirect ELISA on bulk tank milk<br />

Mr Ronald Coilparampil<br />

Evaluation of a commercially available ELISA for detection<br />

antibodies to liver fl uke (Fasciola hepatica) in pooled bovine serum.<br />

Dr Ashraf Ahmed<br />

Zoo animals as a potential reservoir of gram-negative bacteria<br />

harboring integrons and antimicrobial resistance genes<br />

Dr Nguyen Van Long<br />

Porcine reproductive & respiratory syndrome (PRRS) –<br />

a new challenge<br />

Crown Promenade, Melbourne, Australia | 13 14<br />

A new direction for animal health.<br />

Introducing animal health solutions from a proven technology leader.<br />

With traditional procedures, simplicity, speed and precision are often compromised. That’s why more labs<br />

are switching to superior PCR-based reagents for everyday veterinary and agricultural applications.<br />

PCR helps simplify procedures and accelerate time to results with unparalleled sensitivity and specifi city.<br />

Only Applied Biosystems, with over 25 years of experience, provides one source for a complete molecular<br />

solution. With automated sample prep, superior molecular reagents and master mixes, and easy-to-use<br />

instruments for PCR analysis, we’ve got you covered — from sample to signal.<br />

Go molecular.<br />

For a free AgPath-ID One-Step<br />

RT-PCR Reagent, go to<br />

appliedbiosystems.com/animalhealth.<br />

For research use only. Not for use in diagnostic procedures. Not licensed by the USDA.<br />

Applied Biosystems, and AB (Design) are registered trademarks and AgPath-ID is a trademark<br />

of Applera Corporation or its subsidiaries in the US and/or certain other countries. The<br />

AgPath-ID OneStep RT-PCR Kit is compatible with the 5’ nuclease detection and ds<br />

DNA-binding dye processes covered by patents owned or licensable by Applied Biosystems.<br />

No license under these patents is conveyed expressly, by implication, or by estoppel to the<br />

purchaser by the purchase of this product. Further information on purchasing licenses may<br />

be obtained by contacting the Director of Licensing, Applied Biosystems, 850 Lincoln Centre<br />

Drive, Foster City, California 94404, USA. © 2007 Applied Biosystems. All rights reserved.


<strong>WAVLD</strong> POSTER PRESENTATIONS<br />

Poster No. Author Poster Title<br />

1 Ann Nordengrahn Proximity ligation assay – detection of individual microbial pathogens<br />

2 Wayne Jorgensen Improved detection of bovine reproductive disease pathogens<br />

3 Marinda Oosthuizen Identifi cation of a novel Babesia species from sable, roan and giraffe by means of the Reverse Line Blot (RLB)<br />

hybridization assay<br />

4 Maria Jose Dus Santos Detection of BLA in frozen semen samples by PCR assay<br />

5 Ian Marsh A simple internal PCR control for Mycobacterium avium subsp. paratuberculosis constructed by PCR techniques<br />

6 Graeme Eamens Investigating shedding rates and the proportion of animals shedding Mycobacterium avium subsp paratuberculosis in<br />

cattle and goat herds<br />

7 Darcy Myers A streamlined workfl ow for rapid and sensitive detection of Mycobacterium avium subsp. paratuberculosis (MAP) in<br />

bovine faecal samples by real-time PCR<br />

8 Jose Blanco Automated DNA isolation: an advantageous tool in molecular diagnostic of dermatophytosis<br />

9 Sarah North Development of real time PCRs to detect important bacterial pathogens of the horse.<br />

10 Masoud Ghorbanpoor<br />

Najafabadi<br />

Enhanced cultural sensitivity of Staphylococcus aureus from bovine mastitis milk by sample freezing<br />

11 Lourdes Corpus Standardisation of PCR for identifi cation of Listeria monocytogenes in fresh cheese<br />

12 Lone Hoj Molecular ID of vibrio isolates<br />

13 Samantha Gibbs Indonesian veterinary laboratory capacity building project for highly pathogenic avian infl uenza<br />

14 Manuela Dalla Pozza Management of a DIVA vaccination programme for the control of low pathogenicity avian infl uenza in Italy<br />

15 Adam Foord Adaptation of real time PCR assays for detection of new and evolving avian infl uenza virus strains<br />

16 Hans Heine Identifi cation of a novel hemagglutinin cleavage site in a highly pathogenic avian infl uenza H7 from North Korea<br />

17 Anna Lecoq Preliminary validation of a commercial avian infl uenza N1 antibody competitive ELISA kit that can be used as part of a<br />

DIVA strategy<br />

18 Giovanni Cattoli Inactivation of avian infl uenza viruses<br />

19 Huaguang Lu A rapid laboratory diagnosis of H5N1 avian infl uenza in Saudi Arabia<br />

20 Masoud Hosseini Seroprevalence of H9N2 antibody in poultry farm and slaughter-house workers of Iran using HI test<br />

21 Ross Lunt Advancing the role of a regional reference laboratory for foot–and-mouth disease in South East Asia<br />

22 Melanie Chitray Heterogeneity in the outer capsid-coding region of foot-and-mouth disease virus A and O serotypes in Africa<br />

23 Claro Mingala Quantifi cation of water buffalo (Bubalus bubalis) cytokine expression in response to inactivated foot-and-mouth<br />

disease vaccine using real-time PCR assay<br />

24 Chris Morrissy Development of an improved capability in support of national biosecurity for the surveillance and control of foot &<br />

mouth disease in cattle and pigs in Vietnam.<br />

25 Simon Anthony Nucleotide sequence analyses of epizootic haemorrhagic disease virus genome segments encoding the outer capsid<br />

proteins: a serological and genetic re-evaluation of serotypes.<br />

26 Rahana Dwarka Phylogenetic analysis of African swine fever viruses from South Africa, Mozambique and Tanzania for the period<br />

2001 – 2007<br />

27 Scott Fitzgerald Efforts to control and eradicate bovine tuberculosis in a wildlife reservoir in Michigan, USA: a preliminary success story<br />

28 Ji-hyung Kim Detection of major bacterial and viral pathogens from trash fi sh for cultured fl ounder<br />

29 Amir Hamir Experimental transmission of transmissible spongiform encephalopathies<br />

30 Elizabeth Houston TSE surveillance in Australia: a regional laboratory perspective<br />

31 Francisco Blanco Viera BSE surveillance in Argentina<br />

Crown Promenade, Melbourne, Australia | 15<br />

<strong>WAVLD</strong> POSTER PRESENTATIONS (continued)<br />

Poster No. Author Poster Title<br />

32 Michael Zalunardo Complete automation of a BSE post mortem diagnostic test<br />

33 Sanjay Kapil Emergence of canine parvovirus 2c in North America: 2006<br />

34 Tin Tin Myaing Prevalence study of parasitic infestation in Myanmar timber elephants<br />

35 Hermann Unger The concept of early detection and rapid response to TAD’s by APH, IAEA<br />

36 Emma Stubberfi eld Brucellosis in marine mammals: Veterinary Laboratories Agency involvement<br />

37 Magdalena Dunowska Prevalence of methicillin resistant Staphylococcus aureus (MRSA) in a veterinary teaching hospital<br />

38 Ross Lunt A virus neutralization test for the detection of antibody to Australian bat lyssavirus<br />

39 Chris Morrissy Development of a porcine circovirus diagnostic capability at AAHL allowing the detection and nucleotide sequence<br />

analysis of PCV from disease outbreaks.<br />

40 Babak Asghari Study of brucellosis among abattoir workers in Isfahan,Iran<br />

41 Mihai Turcitu Genetic variability analysis of rabies virus serotype 1 on Romanian territory<br />

42 Jianning Wang Inter-laboratory evaluation of a real-time PCR assay for detection of bovine herpesvirus 1 in bovine semen<br />

43 Petrina Young The Australian National Quality Assurance Program<br />

44 James Watson LESTER: The use of a simulation tool to enable detailed evaluation of laboratory capacity to respond to an outbreak.<br />

45 Jacek Gwozdz Reproducibility of results in batches of three ELISA kits for the diagnosis of Johnes’s disease in cattle:<br />

recommendations for kit evaluation criteria<br />

46 Ann Nordengrahn Lateral fl ow technology – pen-side test for the detection of foot-and-mouth disease<br />

47 Kerri Tyrrell Development of a high throughput, robotics-based, ELISA for the detection of antibodies specifi c for parasitic worm<br />

species in sheep<br />

48 Rohit Mistry LATE-PCR detection of foot and mouth disease virus using the BioSeeq II<br />

49 Lawrence Wangh A highly multiplexed RT-LATE-PCR assay for detection of H5N1 and other subtypes of infl uenza virus<br />

50 Mikhayil Hakhverdyan One-step primer-probe energy transfer (PriProET) real-time RT-PCR detection of SVDV using Rotor-Gene 6000<br />

51 Giovanni Cattoli Development and validation of a real time PCR assay for the simultaneous detection of avian infl uenza viruses<br />

belonging to the H5, H7 and H9 subtypes<br />

52 John White An evaluation of commercially available nucleic acid extraction methods for the processing and subsequent qPCR<br />

analysis of samples infected with avian infl uenza virus.<br />

53 Angela Burrell High throughput isolation and detection of North American and European porcine reproductive and respiratory virus by<br />

qRT-PCR<br />

54 Songhua Shan Multiplex PCR for NDV & AI<br />

55 Songhua Shan Detection of CSFV in meat by qPCR<br />

56 Richard Hodgson Sample preparation methods for high quality nucleic acid isolation from a variety of veterinary samples<br />

57 Richard Hodgson Evaluation of real-time PCR based assays for the detection and quantitation of different diseases in animals<br />

58 Luis Samartino Advances in fl uorescence polarization for diagnosis of bovine brucellosis in milk<br />

59 Jing Zhang Effects of different populations on an assessment of the sensitivity and specifi city of an antigen ELISA for BVDV<br />

60 Michael Zalunardo Development of a new avian infl uenza antibody blocking ELISA<br />

61 Mirabos Aminjonov Diagnostics of parasitic diseases of agricultural animals in Uzbekistan<br />

62 Ann Nordengrahn Targeted selective treatment of Ostertagia ostertagi – the use of an ELISA as a diagnostic tool for the control of the<br />

parasite in cattle.<br />

63 Irene Alvarez Validation of an agar gel immunodiffusion and western blot assays for the diagnosis of equine infectious anaemia using<br />

a recombinant p26 protein<br />

64 Wayne Jorgensen New diagnostic assays for the detection and monitoring of coccidiosis in poultry.<br />

65 Gerónimo Gutiérrez Accurate detection of bovine leukaemia virus infection using recombinant P24-ELISA and PCR<br />

66 Annie Rodolakis Is the Coxiella burnetii Nine Mile strain a suitable antigen for detection of Q fever antibodies of ruminants by ELISA?<br />

67 Katsuhiko Fukai Differentiation of foot-and-mouth disease virus (FMDV) infected pigs from vaccinated pigs using a western blotting<br />

assay based on baculovirus expressed FMDV 2C and 3D antigens<br />

68 Ross Lunt Development of a commercial ELISA for the detection of serum antibody to Akabane virus<br />

69 Anna Lecoq Preliminary validation of a new multi-species competitive ELISA kit for the detection of antibodies directed against<br />

West Nile virus: ID Screen ® West Nile Competition<br />

70 Anna Lecoq Validation of a new commercial kit for the detection of antibodies directed against Leishmania infantum in canine sera:<br />

ID Screen ® Leishmaniasis indirect<br />

16 | <strong>WAVLD</strong> 2007, 11 – 14 November 2007


<strong>WAVLD</strong> POSTER PRESENTATIONS (continued)<br />

Poster No. Author Poster Title<br />

71 Maria Jose Dus Santos Development of an ELISA kit to detect and evaluate bovine viral diarrhoea virus antigen in the critical stages of<br />

vaccine production<br />

72 Hinrich Voges Fine-tuning a bulk milk EBL ELISA as primary screening tool for all EBL-free herds in New Zealand<br />

73 Hinrich Voges Adapting a DNA tissue sampler as an aid for effi cient BVDV antigen screening<br />

74 Hinrich Voges Validation of an indirect bulk milk BVDv antibody ELISA for sero-prevalence investigations amongst New Zealand<br />

dairy herds<br />

75 Hinrich Voges Preliminary evaluation of Neospora caninum ELISA kits for use with New Zealand milk samples<br />

76 Seong-in Lim ELISA based on recombinant Erns protein mixtures for serelogical diagnosis of classical swine fever<br />

77 Misako Konishi Development and evaluation of ELISA for detecting antibodies against CAEV<br />

78 Jody Hobson-Peters A diagnostically useful peptide to detect West Nile virus infection in horses<br />

79 Maria Virginia Meikle Novel antigens for the diagnosis of bovine tuberculosis<br />

80 Anna Lecoq Preliminary validation of a new commercial ELISA kit for the detection of antibodies directed against Chlamydia abortus<br />

81 Anna Lecoq Preliminary validation of a new commercial diagnostic test for the detection of anti-equine infectious anaemia virus<br />

antibodies in horse sera<br />

82 James Conland Application of immunomagnetic bead technology for improved diagnosis of classical swine fever virus in a low<br />

technology setting<br />

83 Oday Sami Beyon Laboratory evaluation of the appropriate vaccination programs against Newcastle disease in broiler chickens under<br />

fi eld conditions in Mosul province (Iraq).<br />

84 Xingnian Gu Australian strains of bovine herpesvirus 1 are genetically homogenous<br />

85 Richard Clough Interference in disease differentiation in pigs by pestivirus cross-reactions<br />

86 Rodney Davis Venereal disease and infertility in cattle due to bovine herpesvirus type 5.<br />

87 Bruce Corney An unusual case of infectious laryngotracheitis virus detection in a backyard poultry fl ock<br />

88 Georgina Ibata Detection of bovine lymphotropic herpesvirus in cases of non-responsive post partum metritis in dairy herds in the UK<br />

89 Andrew Cheung Reproduction of PMWS of high mortality with a porcine circovirus type 2-group 1 isolate<br />

90 Jasbir Singh Seroprevalence of porcine reproductive and respiratory syndrome in Malaysia<br />

91 Edla Arzey Investigations of nodavirus infections of Australian bass<br />

92 Masao Kamiya Diagnosis and countermeasure of alveolar echinococcosis in red foxes utilizing local resources in Japan<br />

93 Jose Trinipil Lagapa Smart fi eld sampling for diagnosis of Echinococcosis in wildlife defi nitive host using GIS-based maps<br />

94 Laura Hernández<br />

Andrade<br />

Mycoplasma mycoides subsp.capri associated with goat respiratory disease and high fl ock mortality<br />

95 Sandra Romero Reservoir of West Nile virus and another encephalitis virus in Mexican bats (Desmodus rotundus)<br />

96 Jianming Tan Purifi cation of a herpes-like virus in abalone<br />

97 Gian Mario De Mia Genetic heterogeneity of bovine viral diarrhoea virus isolates from Italy: identifi cation of new BVDV-1 genotypes<br />

98 Stefan Vilcek Genetic characterisation of border disease virus isolated from chamois<br />

99 Mihai Turcitu Molecular characterisation of Newcastle disease viruses isolated from recent outbreaks in Romania<br />

100 Claude Sabeta Molecular epidemiology of rabies in bat-eared foxes (Otocyon megalotis) in South Africa<br />

101 Claude Sabeta Identifi cation of a new dog rabies lineage in northern South Africa<br />

Crown Promenade, Melbourne, Australia | 17<br />

<strong>WAVLD</strong> 12 – 14 NOVEMBER 2007<br />

LEVEL 1, CROWN PROMENADE MELBOURNE<br />

18 | <strong>WAVLD</strong> 2007, 11 – 14 November 2007


<strong>WAVLD</strong> 2007 EXHIBITOR LIST<br />

Booth Company<br />

1 Enfer Diagnostics<br />

2 Corbett Life Science<br />

3 Laboratorios Hipra<br />

4 Lab Diagnostics<br />

5 Pathtech Pty Ltd<br />

6 Bio-X Diagnostics<br />

7 Department Primary Industries Research Victoria<br />

(DPI) Silver Sponsor<br />

8 National Association of<br />

Testing Authorities (NATA)<br />

9–10 Gribbles Vet Pathology<br />

11 InterAct<br />

12 Veterinary Laboratories Agency<br />

Silver Sponsor<br />

Booth Company<br />

13 QIAGEN Pty Ltd<br />

14 Smiths Detection Silver Sponsor<br />

15 Australian Biosecurity CRC<br />

16 Applied Biosytems<br />

Silver Sponsor<br />

17 Animal Genetics Inc<br />

18 Svanova<br />

19 Inverness Medical<br />

20 ID Vet<br />

21 CSIRO Livestock Industries<br />

22–23 The Department of Agriculture, Fisheries<br />

and Forestry (DAFF) Gold Sponsor<br />

24 IDEXX Laboratories<br />

25 Ai Scientifi c<br />

Crown Promenade, Melbourne, Australia | 19<br />

SOCIAL PROGRAM<br />

SYMPOSIUM WELCOME RECEPTION<br />

Proudly Sponsored by<br />

Date: Monday, 12th November 2007<br />

Time: 7.15pm – 9.15pm<br />

Venue: Crown Promenade, Pre Function and Exhibition Area<br />

Additional Tickets: A$80.00 per guest<br />

Inclusive of <strong>Symposium</strong> Full and Day Registration<br />

Delegates are invited to attend the <strong>WAVLD</strong> 2007<br />

<strong>Symposium</strong> Welcome Reception to be held at Crown<br />

Promenade. It will be a great chance to come together with<br />

old friends and colleagues and make new acquaintances,<br />

whilst enjoying canapés and drinks. Additional tickets are<br />

available for partners and work colleagues.<br />

DEPARTMENT OF<br />

PRIMARY INDUSTRIES<br />

Science<br />

SYMPOSIUM DINNER<br />

Proudly Sponsored by<br />

Date: Tuesday, 13th November 2007<br />

Time: 7.00pm – 12.00am<br />

Venue: RACV Club – Level 17, 501 Bourke Street, Melbourne<br />

Tickets: A$100.00 per guest<br />

The <strong>Symposium</strong> Dinner will be held at RAVC Club, level<br />

17 – offering spectacular views of Melbourne skylines and<br />

Port Phillip Bay . Come and enjoy an evening of delicious<br />

food, good wine and great company. You will be able to<br />

socialise and network with old and new friends alike.<br />

Please note: The <strong>Symposium</strong> Dinner is not included in<br />

the registration fee for delegates. If you wish to purchase<br />

tickets, please visit the registration desk.<br />

How to get to the venue: Directions have been provided in<br />

the delegate satchel and are posted on the message board.<br />

Directions are also available at the registration desk.<br />

DPI’s science assists<br />

Victoria’s primary<br />

industries to improve their<br />

productivity and respond<br />

to the challenges of climate<br />

change and drought.<br />

DPI employs more<br />

scientists than any other<br />

organisation in Victoria.


DINING IN MELBOURNE<br />

Melbourne’s melting pot of cultures is refl ected in its<br />

variety of restaurants, cafes, bistros and bars. Fashionable,<br />

eclectic and eccentric – Melbourne’s dining spots offer<br />

a dizzying spread of the world’s great cuisines, serving<br />

meals from the substantial and classic to the truly exotic.<br />

In the city, you can enjoy afternoon tea in the genteel<br />

surroundings of a nineteenth-century hotel, watch and be<br />

watched in buzzing laneway cafés and bars, or handpick<br />

a bottle of Yarra Valley chardonnay at the latest überchic<br />

hangout. Head out a little further and explore one of<br />

Melbourne’s specialist eating destinations – Carlton for<br />

Italian classics or Fitzroy for tantalising Spanish tapas.<br />

LYGON STREET, CARLTON<br />

Enticing aromas of Italy lead you along the footpaths of<br />

Carlton as you enjoy some of Australia’s’ best hospitality<br />

in one of the friendliest cities in the world. Lygon Street is<br />

a must-see part of Carlton, one of the favourite suburbs of<br />

inner Melbourne. Casual or otherwise, the choice is yours<br />

and the restaurateurs of Lygon Street are great at providing<br />

terrifi c food, quickly almost any time of the day or night.<br />

What’s attracting<br />

all the attention on<br />

stand 14?<br />

Maybe it’s the opportunity to see the worlds fi rst<br />

truly portable, veterinary diagnostic laboratory?<br />

Perhaps it’s the prospect of meeting the most<br />

innovative and advanced manufacturer<br />

of detection equipment?<br />

Or is it the chance to win the latest GPS<br />

Navigation system?<br />

Make sure you visit us on stand 14 and see for<br />

yourself how we may have the answer to the world’s<br />

fi ght against virulent diseases such as Avian Flu and<br />

Foot and Mouth.<br />

www.smithsdetection.com/vet<br />

BRUNSWICK STREET, FITZROY<br />

If you were after one area of Melbourne that best refl ects<br />

the city’s soul, it could be Brunswick Street, Fitzroy. This<br />

cafe and food precinct bordering on the CBD is very much<br />

like Melbourne in that it is tolerant and accommodating of<br />

all types of people. Here you can be Bohemian, poor, rich,<br />

alternative, trendy, young or old. Brunswick Street is art<br />

with a capital A; the people who live here ooze art. It’s the<br />

best selection of small, unpretentious cafes serving tasty<br />

food of any food precinct in Melbourne<br />

SOUTHGATE, MELBOURNE CITY<br />

Southgate is home to 17 of Melbourne’s favourite and<br />

most awarded restaurants, bars and cafes. Where better<br />

to sit and relax with world class food, wine and the most<br />

spectacular views of the Melbourne skyline and the Yarra<br />

River. We have more awards than any other precinct in<br />

Melbourne; making Southgate arguably Melbourne’s<br />

premier dining destination.<br />

21<br />

13th International World Association of Veterinary<br />

Laboratory Diagnosticians Syposium<br />

11 - 14 November 2007<br />

Monday 12 November


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

0815 - 1030 Plenary Session - Global Risks and Challenges Mon<br />

FUTURE RISKS FROM INFECTIOUS DISEASES OF ANIMALS<br />

12<br />

Mark M. Rweyemamu - Tanzania and Former FAO<br />

Visiting Professor of Virology, Department of Pathology and Infectious Diseases,<br />

The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, England, UK. mark.rweyemamu@btinternet.com<br />

November<br />

Abstract:<br />

During the last 2 decades there have been incidents of high profile epidemics of human and animal diseases.<br />

These reflect trends in the global distribution of the major infectious diseases. The UK driven Foresight<br />

global study 1 on infectious diseases of humans, animals and plants identified common underlying risk drivers<br />

as (i) Culture and governance, including legislation and systems of government; (ii) Technology and<br />

innovation; (iii) Conflict and law; (iv) Human activity and social pressures; (v) Economic factors – including<br />

globalisation; and (vi) Climate change. Probably the most acute human activity and social pressure with<br />

respect to animal agriculture is that which the study by FAO, IFPRI and ILRI had termed the livestock<br />

revolution. This is characterised by an unprecedented demand for livestock products, a rapid growth in the<br />

livestock sector and an increased risk for animal disease epidemics. Animal agriculture now represents<br />

the fastest growing component of agriculture. An aspect of social pressures is the increasing human<br />

settlement encroachment on previously separated wildlife habitats resulting in an increasing humanlivestock-wildlife<br />

interface.<br />

Several risk assessment studies lead to a conclusion that major infectious diseases are normally absent from<br />

highly industrialised countries while they remain endemic in developing countries. The pattern of disease<br />

spread is no longer limited to episodic spread in localities or to neighbouring countries but has also led to<br />

wider global dissemination. Thus the recent patterns of international spread of foot-and-mouth disease,<br />

classical swine fever, avian influenza and bluetongue have illustrated the importance of transboundary<br />

animal diseases to the global livestock industry and to trade in livestock commodities beyond their endemic<br />

settings. Another trend has been the association of the majority of emerging human diseases with the<br />

animal source. Most such diseases have also originated from developing countries.<br />

Thus the overall conclusion of the Foresight study was that:<br />

• Many existing diseases will remain important, but new diseases will emerge in the<br />

future – noting that in the last 25 to 30 years some 80% of new/emerging infectious<br />

diseases of humans had originated from animals;<br />

• Major infectious diseases are endemic in Africa and Asia;<br />

• Substantial advances in infectious disease prevention and management will be made<br />

through integration of research across sectors (human, animal, plant) and disciplines<br />

(natural and social science);<br />

• New technological systems for early detection, identification and monitoring of<br />

infectious diseases have the potential to transform our capabilities in managing future<br />

disease risks, especially if challenges of international development are met;<br />

• Societal contexts will be crucial in realising the benefits of the new technological<br />

systems.<br />

Therefore, future research on the Detection, Identification and Monitoring (DIM) of infectious diseases needs<br />

to have a special focus on developing countries, preferably through inter-institutional partnerships between<br />

those in highly industrialised countries, where expertise resides but major infectious diseases are not<br />

endemic and those in developing countries, where such diseases are endemic and thereby the risk to global<br />

animal agriculture and human health. This research should be viewed as a contribution to the international<br />

pubic good and essential to the concept of ONE WORLD, ONE HEALTH, ONE MEDICINE.<br />

1 Foresight DIID study www.foresight.gov.uk. Professor Rweyemamu was the Lead Coordinator for the African strand of the study<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NEW AND EMERGING DISEASES<br />

Corrie Brown, DVM, PhD, University of Georgia, Athens, Georgia, USA<br />

Emerging disease has become almost a household term. Fully three-quarters of all the emerging diseases<br />

in the last decade came from animals. The world is averaging at least one new extensive emerging disease<br />

every year. Today, the unrelenting forces of globalization, fueled effectively by the expanding human<br />

population and the institution of the World Trade Organization, have ensured that pathogens can travel<br />

easily from one species to another and one continent to another. No health issue can remain local for long.<br />

The steady parade of new pathogens, beginning with HIV, moving from a primate reservoir into a few and<br />

then millions of humans globally, mobilized the field of infectious diseases in a massive way. The almost<br />

simultaneous emergence of Salmonella DT104 and E. coli O157:H7 underscored large gaps in<br />

understanding between human medicine and animal husbandry. Subsequently, the march of bovine<br />

spongiform encephalopathy through Europe in the Trojan cow of exported meat and bone meal further<br />

emphasized the need for connectedness between animal and human health. More recently, the emergence<br />

of Nipah virus and avian influenza, to the intense consternation of both agricultural and public health<br />

communities, has mobilized synergistic efforts in earnest. Ebola and SARS are two other headline agents<br />

that have sparked panic in humans after moving out of their animal reservoirs.<br />

Bioterror threats have received star billing recently with extensive funding provided to the biomedical<br />

community for directed study. Of the bioterror agents on the US Department of Health and Human Services<br />

List, almost all are pathogens of, or have their origins in, animals. One medicine reigns here as well. The list<br />

of emerging zoonotic disease and targeted bioterror pathogens has so many interconnections that<br />

classifying the two individually is a challenge. They are as connected as the figures in a Jackson<br />

Pollock drawing.<br />

Elucidating the pathogenesis of some of these emerging and bioterror pathogens will help greatly in devising<br />

effective control and intervention measures. Veterinarians have studied many of these diseases, e.g.,<br />

anthrax, smallpox, Nipah, SARS, and avian influenza, using suitable animal models, with resulting expansion<br />

of available information. Findings from a few of these studies will be presented.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AVIAN INFLUENZA – CHALLENGES AND OPPORTUNITIES FOR THE VETERINARY PROFESSION<br />

I. Capua OIE, FAO and National Reference Laboratory for Newcastle Disease and Avian Influenza<br />

Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10 35020 – Legnaro, Padova, Italy<br />

D. Alexander Unaffiliated Consultant Virologist<br />

Notifiable Avian influenza is an OIE listed disease that has become a disease of great importance both for<br />

animal and human health. The increased relevance of AI in the fields of animal and human health, has<br />

highlighted the lack of scientific information on several aspects of the disease, which has hampered the<br />

adequate management of some of the recent crises. Millions of animals have died, and there is growing<br />

concern over the loss of human lives and over the management of the pandemic potential.<br />

Until 1999, Highly Pathogenic Avian Influenza (HPAI) was considered a rare disease, with only 18 primary<br />

outbreaks reported since 1959. At the turn of the millennium, HPAI initiated a series of outbreaks with<br />

different characteristics to what had been previously seen. The Italian H7N1 1999-2000, and Dutch H7N7<br />

2003 caused outbreaks of unprecedented magnitude were only a prelude to the ongoing H5N1 crisis. The<br />

latter appears to represent the possibly the greatest threat to animal health, with very serious implications for<br />

public health that the veterinary community has ever been called to face.<br />

Avian influenza has become widespread in vast areas including Asia, the Middle East, Europe and Africa.<br />

This opportunity given to the virus has greatly increased its potentials, affecting the health of wild and<br />

domestic animals and of humans. Currently, human health is affected both in terms of the reduction of food<br />

security and of the infection of humans as a prelude to the emergence of a new pandemic virus.<br />

Highly Pathogenic avian influenza is believed to originate from an H5 or H7 precursor of low pathogenicity<br />

harboured in wild birds, primarily Anseriformes. The introduction of this progenitor into domestic poultry has<br />

caused, on several occasions the mutation of the low pathogenicity virus to a highly pathogenic mutant<br />

containing multiple basic aminoacids at the cleavage site of the haemagglutinin molecule. This mutation<br />

enables the virus to become systemic, replicate in vital organs and bring about the death of the bird.<br />

Historically, highly pathogenic avian influenza caused a self-limiting disease as most birds affected died as a<br />

result of systemic infection. The virus infected poultry, did not infect wild birds and was apathogenic for<br />

waterfowl. The latter was only very rarely infected.<br />

In the Asian context, the persistence of a highly pathogenic H5N1 virus in mixed rural poultry, in an area with<br />

a high population density has generated a unique set of opportunities for the virus. It has closely<br />

encountered avian and mammalian species, to which it has been able to adapt and is in a continuous effort<br />

to be more efficient. The rural farming environment, typically developed in the open has determined the spillover<br />

of the virus to wild birds. So far, only few species have been shown to be infected but the<br />

epidemiological consequences of this unique situation are impossible to predict.<br />

The co-existence of susceptible species is inducing the development of mutations which often result in a<br />

greater capability of causing clinical disease resulting in death of the host.<br />

The virus is actively circulating in birds reared for agricultural purposes in Asia, the Middle East and Africa<br />

and in the Eurasian wild bird population. It is now able to cause a 50% fatality rate in humans and has<br />

acquired the capability of killing domestic and wild waterfowl. It has also affected other atypical hosts such<br />

as felines.<br />

The crucial issue in resolving this situation is to limit the circulation of the virus in the animal reservoir, as this<br />

represents a never-ending source of virus. Although specific tools are available, the infrastructure and<br />

economic conditions of most of the affected areas are insufficient to react to the emergency. At the rural<br />

level, basic hygienic measures are rarely respected in animal husbandry and no concept of disease control<br />

measures is known. The social behaviour of the rural human population includes habits which facilitate the<br />

spread of infection, within the same village and, through trade, to other villages. International interventions<br />

are therefore to be focused primarily on education programmes and on veterinary support to farmers.<br />

The scientific veterinary community has the duty and responsibility to address the complex issue of AI<br />

ecology and epidemiology in the countries it affects – which in turn is essential to the development of<br />

adequate control strategies.<br />

The medical, veterinary and agricultural scientific communities are challenged with a virus that is moving in a<br />

tri-dimensional fashion, modifying itself as it adapts to different species and reassorting with other influenza<br />

viruses of avian and potentially mammalian origin, as it infects new species. A significant collaborative and<br />

financial effort in a transparent scientific environment are required to generate data and ideas contributing to<br />

the eradication effort.<br />

Until the extensive circulation of the virus is limited in the avian reservoir, avian influenza will continue to<br />

remain an issue for food security and a global threat for animal and human health.<br />

The veterinary community should take ownership of this responsibility as it has the knowledge and<br />

understanding to offer sustainable solution for the management of this infection.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ZOONOTIC VIRUSES OF BAT ORIGIN – DISCOVERY AND DIAGNOSIS<br />

Linfa Wang<br />

CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Victoria 3220<br />

Bats, probably the most abundant, diverse and geographically dispersed vertebrates on earth, have recently<br />

been shown to be the reservoir hosts of a variety of zoonotic viruses responsible for severe human and<br />

animal disease outbreaks, some with very high mortality. These include some of the most deadly viruses to<br />

emerge in recent history, such as Hendra and Nipah viruses, SARS coronaviruses and Ebola virus. For the<br />

last decade, our group has been working on bat zoonotic viruses belonging to three different virus families:<br />

Paramyxoviridae, Coronaviridae and Reoviridae. Examples will be given for each of the three groups of<br />

viruses, emphasizing on different technology platforms used in virus discovery and development of novel<br />

diagnostic strategies.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1100 - 1230 Concurrent Session 1.1 - New & Emerging Diseases/Wildlife Mon<br />

EXPERIMENTAL MODELS FOR HENIPAVIRUS INFECTION: BATS, CATS AND PSEUDO-RATS.<br />

12<br />

Bruce A. Mungall, Deborah J. Middleton, Kim Halpin, Peter Daniels, and John Bingham.<br />

Australian Animal Health Laboratory, CSIRO Livestock Industries, 5 Portarlington Rd, Geelong, Australia<br />

Introduction:<br />

November<br />

Hendra virus (HeV) and Nipah virus (NiV), comprising the genus Henipavirus, family Paramyxoviridae, have<br />

been responsible for numerous outbreaks of zoonotic disease in livestock and humans. Bats of the genus<br />

Pteropus have been identified as the reservoir hosts for these viruses. However, how virus spills over from<br />

the reservoir host has remained a mystery. Nipah virus has continued to re-emerge in Asia, specifically<br />

Bangladesh, since 2001, with case fatality rates as high as 92%. As there are no currently available vaccines<br />

or antivirals indicated for Henipavirus infection, possible therapeutic and prophylactic options in the treatment<br />

and prevention of Nipah virus disease has been seen as a priority in this field of research. In addition to<br />

novel emerging pathogens, Henipaviruses are also BSL4 agents that possess several biological features<br />

that make them highly adaptable for use as bioterror agents. Clearly, there is a high level of public health<br />

importance attributed to Henipaviruses, and by inference, a considerable risk associated with new<br />

emergence events by other unknown or as yet, uncharacterised paramyxoviruses.<br />

Material & methods:<br />

Understanding the amplifying host, the routes of transmission, the type of susceptible human hosts, and the<br />

epicentres for zoonotic and human transmissions is crucial in the control of zoonotic infections. There have<br />

been multiple recent paramyxovirus emergence events, many of these involving bats. Some of these newly<br />

emerged viruses are highly pathogenic (henipaviruses), some are moderately pathogenic (Menangle, PoRV,<br />

PPMV) while many are of unknown pathogenicity (Tioman, Mapuera, SalV, TPMV). The development or<br />

characterization of animal models to study these newly identified viral zoonoses is important for<br />

understanding their pathogenic features and in the development of therapeutics or vaccines. In order to<br />

address the different angles of Henipavirus research, we have developed a range of experimental animal<br />

models including bats, pigs, cats, guinea pigs and ferrets, all amenable to BSL4 conditions. This<br />

presentation describes these models.<br />

Results:<br />

While the human symptoms of NiV infection range from fever and headache to severe acute febrile<br />

encephalitis [1], the porcine disease is primarily a febrile respiratory illness with or without neurological signs<br />

[3]. In both humans and pigs, NIV pathogenesis appears to be related to a systemic vasculitis [3, 5] and<br />

experimental infection of cats, hamsters and ferrets has revealed a similar underlying pathology [3, 4, 6]<br />

(Middleton, unpublished data). While guinea pigs have also been experimentally infected with HeV [2]<br />

(Halpin et al., Manuscript in preparation) and NiV (Halpin et al., Manuscript in preparation), the pathology<br />

differed significantly in several respects from the human cases and from naturally and experimentally<br />

infected horses. NiV and HeV do not cause disease in mice even after subcutaneous administration, and<br />

there is no serological evidence for NiV in rodents in Malaysia. However, and not surprisingly, HeV and NiV<br />

will kill mice if administered intracranially but the lack of natural infection precludes the establishment of<br />

useful rodent models.<br />

Discussions & conclusions:<br />

While the cat represents the animal model in which the pathology most closely resembles the lethal disease<br />

course in humans, and will provide an excellent model for some studies, the ferret provides an excellent<br />

more economical model for preliminary therapeutic testing. The high level of similarity between the two<br />

viruses enables therapeutic studies to be carried out for both pathogens in parallel in ferrets followed by cats.<br />

References:<br />

1. Goh et al., Clinical features of Nipah virus encephalitis among pig farmers in Malaysia. N Engl J<br />

Med, 2000. 342(17): p. 1229-35.<br />

2. Hooper et al., Lesions of experimental equine morbillivirus pneumonia in horses. Veterinary<br />

Pathology, 1997. 34: p. p312-322.<br />

3. Middleton etal., Experimental Nipah virus infection in pigs and cats. J Comp Pathol, 2002. 126(2-3):<br />

p. 124-36.<br />

4. Mungall et al., Feline model of acute nipah virus infection and protection with a soluble glycoproteinbased<br />

subunit vaccine. J Virol, 2006. 80(24): p. 12293-302.<br />

5. Wong et al., Nipah virus infection: pathology and pathogenesis of an emerging paramyxoviral<br />

zoonosis. Am J Pathol, 2002. 161(6): p. 2153-67.<br />

6. Wong et al., A golden hamster model for human acute Nipah virus infection. Am J Pathol, 2003.<br />

163(5): p. 2127-37.<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE EMERGENCE OF CANINE INFLUENZA IN THE USA: THE RELATIONSHIP TO<br />

EQUINE INFLUENZA AND DEVELOPMENTS SINCE DISCOVERY IN 2004.<br />

P.C. Crawford 1 , T.L. Anderson 1 , W.L. Castleman 1 , M.T. Long 1 , R.O. Donis 2 , T.E. Chambers 3 ,<br />

E.P.J. Gibbs 1* .<br />

1 College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.<br />

2 Division of Viral and Rickettsial Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.<br />

3 OIE International Reference Laboratory for Equine Influenza, Gluck Equine Research Center, University of Kentucky, Lexington, KY,<br />

USA.<br />

In 2004, an influenza virus was isolated from an outbreak of severe respiratory diseases in racing<br />

greyhounds at a track in Florida. Several dogs died of pulmonary haemorrhage. An influenza virus was<br />

isolated and the entire genome was sequenced. It was concluded that all eight genes had originated from<br />

equine influenza virus (H3N8), consistent with cross-species transfer from horse to dog. The details of the<br />

discovery of canine influenza and the characterization of the virus have been published (Crawford et al.<br />

2005, Science 310, 482-485). This paper reviews the features of subsequent outbreaks of canine influenza<br />

in Florida and other states within the USA, and reports on the susceptibility of horses to the canine isolate.<br />

Since 2004, canine influenza has been confirmed in multiple outbreaks of respiratory disease at greyhound<br />

tracks throughout the USA and the virus has spread into the pet dog population, particularly those dogs<br />

housed in shelters. The disease has been confirmed by serology and virus isolation in pet dogs in 25 states<br />

in the contiguous USA. Molecular characterizations of recent isolates indicate that the virus is continuing to<br />

evolve. Horses experimentally inoculated with the canine virus were infected as evidenced by<br />

seroconversion and virus replication in pulmonary tissues, but clinical disease was mild.<br />

The paper will discuss the relevance of this discovery to the recent epidemic of equine influenza in Australia.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INVESTIGATIONS OF VIRAL MYOCARDITIS IN PIGS ASSOCIATED WITH A NOVEL PESTIVIRUS<br />

*D.S. Finlaison, P.D. Kirkland, R.W. Cook, M. Srivastava, K.R. King, M.J. Frost<br />

Virology Laboratory, Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries<br />

Introduction<br />

In June 2003 a syndrome of sudden death in sucker pigs, elevated stillbirth percentages, increased<br />

preweaning losses and to a lesser extent increased mummification rates were recognised on a property in<br />

NSW, Australia 1 . Pathological changes included acute to subacute multifocal, nonsuppurative myocarditis<br />

and myonecrosis. In addition, elevated immunoglobulin G (IgG) was identified in almost 50% of stillborn<br />

piglets indicating an in utero infection of viral aetiology was the likely cause. The term porcine myocarditis<br />

syndrome (PMC) was used to describe this disease. A wide range of recognised viral and bacterial<br />

pathogens were excluded during diagnostic investigations and numerous attempts to isolate a virus in cell<br />

culture were unsuccessful. The aim of this study was to produce infected tissues suitable for identification<br />

and characterisation studies, to reproduce the lesions of the PMC, and to investigate the pathogenesis of the<br />

disease.<br />

Materials & methods<br />

Direct foetal inoculation following exposure of the uterus by laparotomy was used to inoculate a proportion of<br />

foetuses in each of 22 litters with tissue extracts collected from foetuses and stillborn piglets on the affected<br />

farm. Three gestational ages were chosen for inoculation and the effects of 6 different inocula were<br />

examined. Gilts were euthanased between 7 and 28 days post-inoculation. Foetuses were screened for<br />

evidence of exposure to an infectious agent by measurement of serum IgG and a section of heart was<br />

examined for histopathological lesions consistent with PMC. A peroxidase linked assay and real-time RT-<br />

PCR were used to examine the antibody response and quantify viral replication in the dam and foetus.<br />

Results<br />

Three inocula were identified as infectious on the basis of elevated IgG in inoculated foetuses. An increase<br />

in IgG was observed in those foetuses greater than 70 days of gestation at inoculation and euthanased at<br />

least 17 days post-inoculation. There was evidence of in utero spread of the agent based on IgG results.<br />

Virus-like crystalline arrays were observed in the tissues from which infectious inocula were derived and in<br />

the hearts of infected foetuses. Unequivocal lesions of PMC were not reproduced. Serum collected from<br />

foetuses in one infected litter was utilised for sequence independent single primer amplification and a novel<br />

pestivirus identified 2 . PCR results provide additional evidence of in utero viral transmission with most<br />

foetuses in litters inoculated with infectious inocula positive for Bungowannah virus RNA by 17 days postinoculation.<br />

Virus was still detected in foetuses at 28 days post-inoculation despite the presence of antibody.<br />

Viraemia in dams receiving infectious inocula resolved with the development of antibody.<br />

Discussion & conclusions<br />

Direct foetal inoculation proved to be a valuable method for amplification and identification of a novel<br />

pestivirus that had previously not been detected by cell culture systems. This study also provides preliminary<br />

information on the replication of this novel pestivirus in the porcine foetus. Further studies will be required to<br />

reproduce the disease.<br />

References<br />

1. McOrist, S., Thornton, E., Peake, A., Walker, R., Robson, S., Finlaison, D., Kirkland, K., Reece, R., Ross,<br />

A., Walker, K., Hyatt, A. and Morrissy, C., 2004. An Infectious myocarditis syndrome affecting late-term and<br />

neonatal piglets. Aust. Vet. J. 82, 509-511.<br />

2. Kirkland, P.D., Frost, M.J., Finlaison, D.S., King, K.R., Ridpath, J.F., Gu, X., 2007. Identification of a novel<br />

virus in pigs-Bungowannah virus: A possible new species of pestivirus. Virus Res. 129, 26-34.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CHARACTERISATION OF A NOVEL PESTIVIRUS ASSOCIATED WITH AN OUTBREAK OF<br />

STILLBIRTHS AND PRE-WEANING DEATHS IN PIGS DUE TO MYOCARDITIS.<br />

*M.J. Frost 1 , D.S. Finlaison 1 , P.D. Kirkland 1 , R. Cook 1 , M. Srivastava 1 , K.R. King 1 , J.F. Ridpath 2 , X.Gu 1 .<br />

1 Virology Laboratory, Elizabeth Macarthur Agricultural Institute, Camden, NSW, Australia<br />

2 USDA, National Animal Disease Center, Ames, Iowa, USA.<br />

Introduction<br />

A syndrome of stillbirths and preweaning losses with myocarditis occurred on 2 Australian pig farms in<br />

2003 1 . While extensive investigations excluded a wide range of known agents, a foetal inoculation study 2<br />

confirmed an infectious agent was present and likely to be viral. This paper describes the<br />

identification/characterisation of this agent.<br />

Material & methods<br />

Sequence independent single primer amplification (SISPA) 3 was undertaken on nucleic acid extracted from<br />

a pool of foetal serum. PCR primers matching terminal sequences of RNA fragments detected by SISPA<br />

were used to amplify intervening RNA segments. PCR products were cloned, sequenced and compared with<br />

data in Genbank. Viral replication in PK-15A cells was detected by PCR and immunoperoxidase (IPX)<br />

staining using convalescent pig serum. Antigenic characterisation was undertaken by IPX using a large<br />

collection of monoclonal antibodies and polyclonal antisera.<br />

Results<br />

Viral RNA sequences identified by SISPA and PCR included the 5’UTR, N pro and E2 coding regions of a<br />

pestivirus 4 . There was also limited sequence from the p7, E rns , NS5A and NS5B regions of the genome. The<br />

entire genome was subsequently sequenced. Genetic analyses and the construction of dendrograms show<br />

that this novel virus is genetically distinct from all of the recognised pestiviruses. Sequence homology with<br />

the four currently defined pestivirus species ranges from 45% to 64.91%. In addition it does not cluster with<br />

other divergent viruses such as the giraffe, HoBi or pronghorn strains of pestivirus. Antigenic<br />

characterisation studies showed that this virus is also antigenically remote from pestiviruses in each of the<br />

recognised species. No IPX staining was observed with any of the pan-reactive monoclonal antibodies.<br />

There was, however, limited cross reactivity between Bungowannah virus antigens and polyclonal antisera<br />

against some strains of BVDV2 and CSFV in IPX and virus neutralisation assays.<br />

Discussion & conclusions<br />

The viral RNA sequences identified are consistent with a novel pestivirus that has a low degree of genetic<br />

identity with recognised and proposed pestivirus species. This virus is also antigenically very different from<br />

most pestiviruses, showing no reactivity with any of the widely used pan-reactive monoclonal antibodies.<br />

Collectively these results suggest that this virus is the most divergent of the pestiviruses that has been<br />

described and warrants classification as a new species in the genus. The failure of this virus to react with<br />

commonly used monoclonal antibodies and its divergent nucleotide sequence have implications for the<br />

design and use of diagnostic assays for pestiviruses. At present it is probable that the virus would escape<br />

detection by existing diagnostic procedures.<br />

References<br />

1. McOrist S, Thornton E, Peake A, Walker R, Robson S, Finlaison D, Kirkland P, Reece R, Ross A, Walker<br />

K, Hyatt A, Morrissy C. An infectious myocarditis syndrome affecting late-term and neonatal piglets. Aust Vet<br />

J. 2004 Aug;82(8):448.<br />

2. Finlaison D.S, Kirkland P.D, Cook R.W, Sirastava M, King K.R, Frost M.J. Investigations of viral<br />

mycoarditis in pigs associated with a novel pestivirus. <strong>WAVLD</strong> 13 th International <strong>Symposium</strong> 2007<br />

3. Allander T, Emerson S.U, Engle R.E, Purcell R.H and Bukh J. A virus discovery method incorporating<br />

DNase treatment and its application to the identification of two bovine parvovirus species, Proc. Natl. Acad.<br />

Sci. 98 (2001), pp. 11609–11614.<br />

4. Kirkland P.D, Frost M.J, Finlaison D.S, King K.R, Ridpath J.F, Gu X., 2007 Identification of a novel virus in<br />

pigs—Bungowannah virus: A possible new species of pestivirus. Virus Research 129, 26–34


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

HOW MANY PESTIVIRUSES CIRCULATE IN NATURE? PESTIRUSES IN FREE-LIVING ANIMALS:<br />

PRESENT STATUS AND PROBLEMS<br />

S.Vilcek 1* , P.F. Nettleton 2<br />

1 University of Veterinary Medicine, Kosice, Slovakia; 2 Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian EH26<br />

0PZ, UK<br />

Four pestivirus species infect domestic animals. BVDV-1 and BVDV-2 mostly infect cattle, CSFV – swine,<br />

BDV – sheep. Pestiviruses are not strictly host specific because they can infect many other animals. Genetic<br />

analysis of pestiviruses by rapid sequencing of PCR products coupled with phylogenetic analysis revealed<br />

new viral genotypes, some of them in free-living animals.<br />

Mainly BVDV specific antibodies have been reported in captive and free-living animals. Pestivirus specific<br />

antibodies were detected in over 40 animal species but the isolation of viruses from wild animals is relatively<br />

rare. It is generally known that CSFV infects not only domestic pigs but also wild boars which are significant<br />

reservoirs of this virus. The BVDV-1 isolates were already detected for example in deer, roe deer, mouse<br />

deer, lama, yak, eland, buffalo, bison. Recently, a pestivirus isolated from reindeer has been typed as BDV-2<br />

genotype (Becher et al., 2003). Pestivirus identified in chamois has been typed as BDV-4 genotype causing<br />

significant problems in chamois population in Pyrenees (Arnal et al., 2004). The first giraffe pestivirus strain<br />

was identified in 60-ties, the second giraffe strain just several years ago. Both strains belong to the<br />

unclassified pestivirus species and in the phylogenetic analysis form a giraffe pestivirus genotype (Avalos-<br />

Raminez et al., 2001). Pestivirus isolated from a pronghorn antelope found dead in Wyoming (Vilcek et al.,<br />

2005) as well as recently identified Bungowannah pestivirus in pig (Kirkland et al., 2007) are representants of<br />

two most distinct pestivirus genotypes so far.<br />

All together six pestivirus genotypes (CSFV, BVDV-1, BDV-2, BDV-4, giraffe and pronghorn antelope) have<br />

been identified in wild animal population. Viruses of BVDV-2, BDV-1 or BDV-3 genotype infecting mostly<br />

cattle and sheep have not been found in wild animals yet.<br />

Most data presented in scientific literature suggest that pestivruses are more likely transmitted from domestic<br />

animals to wildlife population. The transmission in opposite way is not exactly proved but theoretically it can<br />

not be excluded that transmission of highly virulent pestivirus strain from domestic to wild animals or vice<br />

versa could have a devastating effect on the eradication programmes as well as on wildlife population.<br />

There are potential problems with the identification of new pestiviruses:<br />

i/ selection of the correct virus for use in serological studies<br />

ii/ use of the correct cells for the isolation of new pestiviruses<br />

iii/ proper selection of RT-PCR primers for the detection of new pestivirus genomes<br />

All data suggest that pestiviruses are highly efficient viruses infecting domestic and many free-living animal<br />

species and they evolved well-adapted strategy to survive in nature. Taking into account present status with<br />

the identification of new pestiviruses we can conclude that wild animals may be infected with more<br />

pestiviruses than we have identified at present. No doubt that there is a need for more systematic research<br />

focused on the occurrence of pestiviruses in free-living animals.<br />

References<br />

Arnal, M., et al.: J. Gen. Virol., 85, 3653, 2004.<br />

Avalos-Raminez, R., et al.: Virology 286, 456, 2001.<br />

Becher, P., et al.: Virology 311, 96, 2003.<br />

Kirkland, P.D. et al. : Virus Res., 129, 26, 2007.<br />

Vilcek S., et al.: Virus Res., 108, 187, 2005.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CHARACTERIZATION AND DETECTION OF BVDV RELATED<br />

REPRODUCTIVE DISEASE IN WHITE-TAILED DEER<br />

J.F. Ridpath 1* , E.A. Driskell 2 , C.C. Chase 3 , J.D. Neill 1<br />

1 NADC/ARS/USDA, Ames, IA, 2 University of Georgia, Athens, GA, 3 South Dakota State University, Brookings, SD<br />

Introduction<br />

Bovine viral diarrhea viruses (BVDV) are the causative agent of reproductive and respiratory disease in<br />

cattle resulting in significant economic loss to the beef and dairy industries. The primary consequences of<br />

reproductive infection are due to the direct infection of the fetus and the outcome depends on the stage of<br />

gestation in which the fetal infection occurs 1 . Although abortions and weak calves have been attributed to<br />

BVDV infection in late gestation 6 , infections occurring earlier in gestation generally have greater impact.<br />

Fetal infections in cattle, occurring during the first trimester, result in fetal reabsorption, mummification,<br />

abortion or the establishment of persistently infected (PI) animals. PI cattle are considered the main vector<br />

for introduction of the virus to naïve herds.<br />

BVDV also replicates in white-tailed deer (Odocoileus virginianus) 2-5 . Free ranging white-tailed deer<br />

populations are frequently in contact with domestic cattle in the U.S., therefore, possible transfer of BVDV<br />

between cattle and deer has significant implications for proposed BVDV control programs. The goal of this<br />

study was to examine the effects of BVDV infection in pregnant white-tailed deer<br />

Material & methods<br />

Eleven white-tailed does were purchased from a commercial breeder and housed in BSL2 containment.<br />

Pregnancy status was confirmed and the calculated stage of pregnancy was based on date of contact with<br />

buck. Does were inoculated with one of two BVDV previously recovered from wild white-tailed deer. Levels<br />

of BVDV neutralizing serum antibodies were determined prior to inoculation and 21 or 35 days post<br />

inoculation. Virus isolation was performed on tissues from aborted fetuses, tissues from does that died and<br />

from blood samples from live fawns. Additionally, ear notches of live fawns were tested for BVDV antigen by<br />

antigen capture ELISA (ACE).<br />

Results<br />

Two of the does had serum antibody titers against BVDV (>512) prior to inoculation; the remaining 9 does<br />

were seronegative. Both seropositive animals gave birth to normal fawns. Of the remaining 9 seronegative<br />

animals, one was not pregnant at the start of the study, four died (death between 8 to 79 days post<br />

inoculation), one apparently readsorbed its fetus, two aborted and one gave birth to two probable PI fawns.<br />

BVDV was isolated from fetuses, maternal tissues and PI fawns. Ear notches of PI fawns were positive by<br />

ACE.<br />

Discussions & conclusions<br />

Following BVDV infection of deer we confirmed infection of fetal tissues within the first 7 – 8 days of<br />

infection. We observed abortion and mummification following infection and the birth of apparently<br />

persistently infected fawns. The fawns of does that had serum neutralizing antibodies against BVDV at the<br />

time of inoculation were protected. These observations are consistent with the clinical presentation of BVDV<br />

associated reproductive disease in cattle following exposure before 125 days gestation. Further research<br />

needs to be done to determine if the similarities between BVDV associated reproductive disease in whitetailed<br />

deer and cattle hold at later stages of gestation and to determine, more exactly, the window of fetal<br />

vulnerability for development of persistent BVDV infection in white-tailed deer.<br />

References<br />

1 Brock KV, Grooms DL, Givens MD: 2005, Reproductive disease and persistent infections. In: Bovine viral<br />

diarrhea virus: diagnossis, management and control, eds. Goyal SMRidpath JF, pp. 145-156.<br />

Blackwell Publishing, Ames, IA.<br />

2 Chase CCL, Braun LJ, Leslie-Steen P, et al.: 2007, Evidence of bovine viral diarrhea virus persistent<br />

infection in two white-tailed deer in southeastern South Dakota. Journal of Wildlife Diseases In<br />

press.<br />

3 Passler T, Walz PH, Ditchkoff SS, et al.: 2007, Experimental persistent infection with bovine viral diarrhea<br />

virus in white-tailed deer. Vet Microbiol 122:350-356.<br />

4 Ridpath JF, Mark CS, Chase CCL, et al.: 2007, Febrile response and decrease in circulating lymphocytes<br />

following acute infection of white tail deer fawns with either a BVDV1 or a BVDV2 strain Journal of<br />

Wildlife Diseases 43:In press.<br />

5 Van Campen H, Williams ES, Edwards J, et al.: 1997, Experimental infection of deer with bovine viral<br />

diarrhea virus. J Wildl Dis 33:567-573.<br />

6 Ward GM, Roberts SJ, McEntee K, Gillespie JH: 1969, A study of experimentally induced bovine viral<br />

diarrhea-mucosal disease in pregnant cows and their progeny. Cornell Vet 59:525-538.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1100 - 1230 Concurrent Session 2.2 - Molecular Diagnostic Assays - I (Bacterial disease) Mon<br />

DEVELOPMENT OF AN IMPROVED IDENTIFICATION MATRIX FOR AEROMONAS SALMONICIDA<br />

12<br />

*Melissa Higgins<br />

November<br />

1 , Jeremy Carson 1 , and Nick Gudkovs 2<br />

1<br />

Department of Primary Industries and Water,<br />

2<br />

Australian Animal Health Laboratory, CSIRO, Geelong<br />

Introduction<br />

Members of the genus Aeromonas are widely distributed in aquatic environments and have been described<br />

in association with disease in both aquatic animals and humans. The genus comprises 16 species and a<br />

number of subspecies and biovars. As the number of validly described Aeromonas species is increasing,<br />

more is being revealed about the importance of these bacteria as pathogens of aquatic animals. A number<br />

of Aeromonas species are capable of causing disease in fish. One of the more notable species is<br />

Aeromonas salmonicida, the aetiological agent of furunculosis in salmonids causing significant economic<br />

losses in aquaculture worldwide. It is essential that diagnostic laboratories have the capacity to identify the<br />

subspecies and biovars of A. salmonicida. The aim of this work was to phenotypically characterise A.<br />

salmonicida including subspecies and biovars and incorporate the information into the MicroSys A24<br />

miniaturised biochemical identification system developed by The Department of Primary Industries & Water,<br />

Tasmania.<br />

Material & methods<br />

A library of 148 A. salmonicida isolates was complied and characterised for numerical taxonomy using 27<br />

tests in miniaturised format targeted for use on the Aeromonads. Phenotyping was undertaken at 25 °C for<br />

48 hours. Relationships based on similarities were assessed by cluster analysis using ClustanGraphics<br />

software. The phenotypic characteristics of each of the clusters was then used to construct a data matrix to<br />

be used in probabilistic identification using PIBWin software package<br />

Results<br />

The characterisation and subsequent analysis of the A. salmonicida library resulted in the generation of 12<br />

defined clusters. Aeromonas salmonicida ssp. salmonicida formed a discrete cluster that contained strains<br />

that were all PCR positive when tested with the Miyata primer set. The Tasmanian biovar Acheron also<br />

formed a very homogeneous cluster as did those of greenback flounder and goldfish ulcer disease strains, A.<br />

salmonicida spp achromogenes and A. salmonicida spp masoucida. The remaining five clusters<br />

represented distinct biovars of exotic atypical strains.<br />

Two identification matrices were developed from the data generated. A complete Aeromonas matrix ,<br />

AeroMat-1 capable of identifying isolates to the species level and a second matrix, AsalMat-1 for the<br />

identification of A. salmonicida subspecies and biovars<br />

Discussions & conclusions<br />

The MicroSys A24 identification panel and revised probability provide the means to identify potentially<br />

enzootic and exotic isolates of A. salmonicida and allocate them to defined biovars and subspecies. These<br />

matrices do not aim to replace the role of PCR in the primary identification of A. salmonicida rather provides<br />

a means of identifying biovars of relevance to Australia.<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AN IMPROVED PCR-BASED TEST FOR QX DISEASE CONFIRMATION IN SYDNEY ROCK OYSTERS<br />

I. Marsh 1 *, J. Go 2 , S. Austin 1 , S. Fell 1 , V. Saunders 1 and L. Reddacliff 1<br />

New South Wales Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Australia 1<br />

Faculty of Veterinary Science, University of Sydney, Camden, NSW, Australia 2<br />

Introduction<br />

Marteilia sydneyi (M. sydneyi) is the causative agent of QX disease, a parasitic disease of Sydney rock<br />

oysters (Saccostrea glomerata - SRO) that results in mass mortality events along the east coast of<br />

Australia 1 . QX disease can be diagnosed using a range of laboratory tests including: histology 2, 3 , indirect<br />

fluorescent antibody testing 4 and DNA hybridization with fluorochrome-labeled probes 5 . However, routine<br />

diagnosis is achieved using cytology 2, 3 and polymerase chain reaction (PCR) 5-7 . During recent surveillance<br />

studies of QX disease on the New South Wales coast we identified a number of issues with using a single M.<br />

sydneyi specific PCR. Firstly, without in vitro culture or experimental transmission techniques for M. sydneyi<br />

we are lacking a consistent source of positive control tissue to include in routine diagnostic testing for this<br />

disease. Secondly it is not possible to distinguish between true negative results and a reaction that has<br />

failed to amplify due to the presence of PCR inhibitory substances originating from the digestive gland or<br />

from neighboring tissues that have been included in the sample for DNA extraction. Finally, we wanted to<br />

introduce a third PCR assay to differentiate between SRO and Pacific oysters (Crassostrea gigas - PO) that<br />

may accidentally be included in a submission of SRO. Our aim was to develop a modified PCR-based QX<br />

disease testing regime to overcome these problems.<br />

Methods<br />

We developed two PCR assays that target the 16sRNA genes of the SRO and PO and examined these, plus<br />

the M. sydneyi PCR, on digestive glands of infected SRO and healthy PO. We also examined the effects of<br />

neighboring tissues, stomach and gonad, on these assays.<br />

Results<br />

The 16sRNA assays were successfully used, individually and multiplexed, to confirm the DNA extraction and<br />

differentiate between SRO and PO. We also found that the inclusion of neighboring tissues has no adverse<br />

effect on all three assays.<br />

Conclusions<br />

Here we recommend a modified PCR-based QX disease testing regime for when PCR confirmation is<br />

required to determine the presence of M. sydneyi in SRO. This testing regime overcomes much of the<br />

ambiguity of a negative PCR result when the M. sydneyi specific PCR assay is used on its own.<br />

Furthermore, it can also differentiate between SRO and PO thus increasing the overall quality control of this<br />

diagnostic test. We have attempted to multiplex the M. sydneyi and 16sRNA assays but the results indicated<br />

that the reaction was biased towards the amplification of the 16sRNA products even when M. sydneyi was<br />

present. Therefore, these assays are currently run separately until further research can be undertaken to<br />

overcome the stoichiometry conditions of the combined assay. Future work includes modifying this PCR test<br />

to a real-time PCR format.<br />

References<br />

1. Wolf, P.H. 1979. Life Cycle and Ecology of Marteilia sydneyi in the Australian Oyster, Crassostrea<br />

commercialis. Marine Fisheries Review. 41: 70 – 72.<br />

2. Bower, S.M. and Kleeman, S.N. 2003. Synopsis of Infectious Diseases and Parasites of Commercially<br />

Exploited Shellfish: Marteilia sydneyi of oysters. (www.pac.dfompo.gc.ca/sci/shelldis/pages/marsydoy)<br />

3. ProMed. 2003. Fisheries investigate Sydney rock oyster parasite. Hoover News.<br />

4. Newton, K., Peters, R. and Raftos, D. 2004. Phenoloxidase and QX disease resistance in Sydney rock<br />

oysters (Saccostrea glomerata). Developmental and Comparative Immunology. 28: 565 – 569. 2004.<br />

5. Kleeman, S.N., Le Roux, F., Berthe, F. et. al. 2002. Specificity of PCR and in situ hybridisation assays<br />

designed for detection of Marteilia sydneyi and M. refringens. Parasitology. 125: 131 – 141.<br />

6. Nell, J.A. and Hand, R.E. 2003. Evaluation of the progeny of second-generation Sydney rock oyster<br />

Saccostrea glomerate (Gould, 1850) breeding lines for resistance of QX disease Marteilia sydneyi.<br />

Aquaculture. 228: 27 – 35.<br />

7. Wesche, S. J., Adlard, R.D. and Lester, R.J.G. 1999. Survival of spores of the oyster pathogen Marteilia<br />

sydneyi (Protozoa, Paramyxea) as assessed using fluorogenic dyes. Disease of Aquatic Organisms.<br />

36: 221 – 226.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE DEVELOPMENT OF SNP DISCRIMINATION ASSAYS ON A REAL-TIME PCR PLATFORM AS A<br />

TOOL FOR THE RAPID IDENTIFICATION AND SPECIATION OF BRUCELLA.<br />

K.K. Gopaul, C.J. Smith and A.M. Whatmore*<br />

Department of Statutory and Exotic Bacterial Disease, Veterinary Laboratories Agency, Woodham Lane, Addlestone, Surrey, United<br />

Kingdom, KT15 3NB. [a.whatmore@vla.defra.gsi.gov.uk]<br />

Introduction. Brucellosis, caused by members of the genus Brucella, remains a major zoonotic problem in<br />

much of the world. Six distinct species are identified within the genus; B. abortus (bovine), B. melitensis<br />

(caprine, ovine), B. suis (porcine, rangiferine, leporine), B. canis (canine), B. ovis (ovine) and B. neotomae<br />

(desert wood rat) with evidence of additional species, yet to be formally named, associated with various<br />

marine mammal species [1]. Currently the most widely recognised tool for Brucella speciation is biotyping but<br />

this method is slow, can be very subjective and involves hazardous culture of a pathogen readily acquired by<br />

laboratory workers. The aim of this work is therefore to develop a rapid molecular alternative that can initially<br />

identify to species level but will ultimately be extended, if possible, to include markers for subgroups (e.g.<br />

biovars) and live vaccine strains.<br />

Materials & Methods. Single nucleotide polymorphisms (SNPs) specific for each Brucella species were<br />

identified on the basis of an extension of multilocus sequencing studies previously described by us [2]. Some<br />

21 distinct genomic fragments, representing >10 Kb of sequence, were sequenced from >300 Brucella<br />

isolates representing the known diversity of the group. Based on this work, that comprises the most<br />

extensive study of the population structure of the genus undertaken to date, pairs of MGB allele<br />

discrimination probes were designed and tested. Probes corresponded to species-specific SNPs and the<br />

alternative ‘common’ allelic state, and were designed for use in real-time PCR reactions to discriminate each<br />

of the six Brucella species and the marine mammal Brucella.<br />

Results. The performance of various probe pairs designed to interrogate SNP sites with a change specific<br />

for each of the individual species was assessed and those that provided most promising discrimination were<br />

selected. Probe pairs were all optimised to function under identical PCR conditions allowing development of<br />

a multiplex assay. Once optimised the performance of the resulting ‘speciation’ assay was assessed by<br />

testing over 300 field and reference isolates of Brucella. Sensitivity of the assay was determined by testing<br />

serial dilutions of purified genomic DNA while specificity was assessed by testing organisms phylogenetically<br />

related to Brucella.<br />

Discussion and Conclusions. Use of multilocus sequence data from several hundred strains facilitated the<br />

confident design of a diagnostic assay based on a robust phylogenetic framework. The assay can ‘speciate’<br />

an isolate in around 2 hours and has many advantages over biotyping being much easier to perform, less<br />

labour intensive, less subjective and avoiding extensive culturing of this hazardous agent. Furthermore, the<br />

principle of the assay can now be extended to further subtype beyond the species level by incorporating<br />

additional markers to identify subgroups, such as biovars, and live vaccine strains. The assay will also be<br />

amenable to future expansion should additional Brucella species be identified – suitable markers for<br />

inclusion should readily be identified by undertaking the same multilocus sequencing approach.<br />

References.<br />

[1] Groussaud, P., Shankster, S.J., Koylass, M.S., and Whatmore, A.M. 2007. Molecular typing divides<br />

marine mammal strains of Brucella into at least three groups with distinct host preferences. Journal of<br />

Medical Microbiology. In press.<br />

[2] Whatmore, A.M., Perrett, L.L., and Macmillan, A.P. 2007. Characterisation of the genetic diversity of<br />

Brucella by multilocus sequencing. BMC Microbiology 7:34.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DETECTION OF MYCOPLASMA HYOPNEUMONIAE IN PIG SAMPLES<br />

USING POLYMERASE CHAIN REACTION TESTS<br />

G. J. Eamens, NSW Department of Primary Industries<br />

Introduction<br />

Mycoplasma hyopneumoniae (Mhp) is a major respiratory pathogen of pigs with worldwide distribution. It is<br />

responsible for lost production through its ability to induce pneumonia but particularly to predispose pigs to<br />

secondary respiratory infections with other bacterial and viral pathogens. As Mhp is difficult to culture from<br />

field material, diagnostic testing has relied on serological and PCR-based assays to indicate herd infection.<br />

Numerous reports describe PCR tests for the specific and sensitive detection of Mhp, varying from<br />

conventional one step and two step (nested) assays to real time PCR, and targeting numerous DNA<br />

sequences considered specific for Mhp. Few have been examined concurrently, and thus diagnostic<br />

laboratories tend to rely on data collected with limited head-to-head evaluation.<br />

Material & methods<br />

This study reviewed 25 PCR assays reported in the literature since 1993. It targeted conventional PCR<br />

assays reported with sufficient information to score the features of each. The factors considered of interest<br />

were: sensitivity < 10 fg, prior evaluation in lung tissue, nasal swabs and other tissues, as well as a proven<br />

ability to detect M. hyopneumoniae in nasal swabs, and prior evaluation with commercial extraction kits. In<br />

the initial phase, issues affecting test evaluation in “beta testing” assays were identified. Cultures of Mhp and<br />

related mycoplasmas (M. flocculare, M. hyorhinis) were also obtained to assist in test optimisation prior to<br />

application to field samples from several herds with high and low disease status for mycoplasmal pneumonia<br />

(nasal swabs from weaner and finisher pigs, lung samples from affected slaughter pigs).<br />

Results and discussion<br />

Among nine conventional PCR assays which were well described and for which analytical sensitivity and<br />

application data was available, five assays with reported high sensitivity were able to be selected for<br />

concurrent evaluation (Table 1). Among nine commercial DNA capture kits applied to M. hyopneumoniae<br />

PCR tests on tissues, two currently used in Australian diagnostic laboratories (Instagene matrix, BioRad;<br />

DNeasy, Qiagen) were selected for comparative evaluation. Optimisation studies using DNA from previously<br />

characterised mycoplasmal strains showed some published protocols required adjustment of annealing<br />

temperatures and some assays showed cross-reactivity or failure to detect some Mhp strains. Efforts to<br />

clone some cultured strains and apply multiplex assays were also required to validate the source strains.<br />

Table 1. Comparative literature evaluation of conventional PCR for M. hyopneumoniae<br />

Type Test Analytical<br />

sensitivity<br />

PCR<br />

nested<br />

PCR<br />

Artiushin 1 1-10 pg 1 ;1-10 pg 7<br />

Mattsson 8<br />

6 fg 8 ; 100 fg 7<br />

Blanchard 3<br />

0.5 pg 3 ; 10 pg 7<br />

Baumeister 2 5-18.5 fg 5<br />

Caron 6 50 pg 6 ; 1 pg 7<br />

Stark 9 1.2 fg 9 ; 2.5 fg 7<br />

Calsamiglia 4 96 fg 4 ; 1 fg 7<br />

Verdin 10 1 fg 10 , 1 fg 7<br />

Kurth 7 0.5 – 1 fg 7<br />

High<br />

sensitivity<br />

< 10 fg<br />

Tested<br />

on lung<br />

tissue<br />

Tested on<br />

nasal<br />

swabs<br />

Tested<br />

on other<br />

tissue<br />

Detects in<br />

nasal<br />

swabs<br />

DNA kit<br />

used<br />

- - - - - -<br />

- + + + + -<br />

- - - + - -<br />

+/- + + + - +<br />

- + - + - +<br />

+ + + + + +<br />

+ + + + + +<br />

+ + - + - -<br />

+ + + + + +<br />

Conclusions<br />

A significant investment can be required in the laboratory introduction of diagnostic Mhp PCR, and may<br />

require extensive in-house validation to optimise the assay and confirm specificity.<br />

References<br />

1. Artiushin S, Stipkovits L, Minion FC (1993). Molec Cel Probes 7, 381-385.<br />

2. Baumeister AK, et al H (1998) J Clinical Microbiol 36, 1984-1988.<br />

3. Blanchard B, et al (1996). Molec Cell Probes 10, 15-22.<br />

4. Calsamiglia M, Pijoan C, Trigo A (1999). J Vet Diag Invest 11, 246-251.<br />

5. Carew D (2004). PhD thesis, Swinburne Univ of Technology 56-63; 97-111.<br />

6. Caron J, Ouardani M, Dea S (2000). J Clin Microbiol 38, 1390-1396.<br />

7. Kurth KT, et al (2002). J Vet Diag Invest 14, 463-469.<br />

8. Mattsson JG, et al (1995). J Clin Microbiol 33, 893-897.<br />

9. Stark KDC, Nicolet J, Frey J (1998). Appl Environ Microbiol 64, 543-548.<br />

10. Verdin E, et al (2000). Vet Microbiol 76, 31-40.<br />

Supported by Australian Pork Limited


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT AND EVALUATION OF A REAL-TIME PCR TEST FOR THE DETECTION OF<br />

THEILERIA PARVA INFECTIONS IN CAPE BUFFALO (SYNCERUS CAFFER) AND CATTLE<br />

*1 KP Sibeko, 1 MC Oosthuizen, 1 NE Collins, 2 D Geysen, 3 A Latif, 4 HT Groeneveld, 1 JAW Coetzer,<br />

3 F Potgieter<br />

1 Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort,<br />

0110, South Africa<br />

2 Department of Animal Health, Institute of Tropical Medicine, 155 Nationalestraat, Antwerp B-2000, Belgium<br />

3 Agricultural Research Council-Onderstepoort Veterinary Institute, Private Bag X5, Onderstepoort, 0110, South Africa<br />

4 Department of Statistics, School of Mathematical Science, University of Pretoria, Pretoria, 0002, South Africa<br />

Introduction<br />

Corridor disease, caused by the tick-borne protozoan parasite Theileria parva, is a controlled disease in<br />

South Africa. The Cape buffalo is the reservoir host and uninfected buffalo have become sought-after by the<br />

game industry in South Africa, particularly for introduction into Corridor disease-free areas. A real-time PCR<br />

test for detection of T. parva DNA was developed to improve the sensitivity and specificity of the official<br />

diagnostic tests.<br />

Material & methods<br />

A set of Theileria genus-specific primers was designed to amplify a 230 bp region of the 18S ribosomal RNA<br />

(rRNA) gene and a hybridization probe set was designed for specific detection of T. parva. Furthermore, a<br />

T. parva-specific forward primer was designed, to increase the specificity of the assay. DNA extracted from<br />

cattle and buffalo blood samples collected from different areas in South Africa was used in the evaluation of<br />

the real-time PCR test. These included T. parva positive, negative and field samples.<br />

Results<br />

The two sets of primers designed for the real-time PCR assay successfully amplified T. parva DNA under the<br />

conditions optimised for this assay. In addition to T. parva amplicons, the T. parva-specific hybridization<br />

probes recognised T. taurotragi and T. annulata PCR products generated by the Theileria-genus specific<br />

primers. These amplicons could be distinguished by melting curve analysis, with the T. parva Tm at 63<br />

±0.62°C, T. annulata Tm at 48 ±0.09°C and T. taurotragi at 45 ±0.19°C. The use of the T. parva-specific<br />

forward primer eliminated amplification of all other Theileria species, except for Theileria sp. (buffalo). In<br />

samples infected with both of these species, only the T. parva amplicon was detected by the T. parva probe<br />

set. No amplification was observed from any of the other Theileria species or other blood parasites and<br />

bacterial DNA samples tested. The real-time PCR assay was ~27% more sensitive than the conventional<br />

PCR/probe and RLB assays and 16% more sensitive than the coxI assay in detecting T. parva in field<br />

samples.<br />

Discussions & conclusions<br />

The T. parva-specific probe set also detected T. taurotragi and T. annulata when the Theileria genus-specific<br />

primers were used, because of the similarities in the 18S rDNA sequence of these species in the region from<br />

which the probe sequence was derived. However, this did not influence the specificity of the assay because<br />

the different products were easily discriminated by melting curve analysis. The sensitivity of the test was<br />

improved by designing a T. parva-specific forward primer. Although the Theileria sp. (buffalo) DNA could still<br />

be amplified when the T. parva-specific forward primer because of the high similarity of the Theileria sp.<br />

(buffalo) 18S rRNA gene sequence with that of T. parva, the test still remains specific in detecting only T.<br />

parva as the T. parva-specific hybridization probe set only detects T. parva amplicons. In conclusion, the<br />

real-time PCR assay reported here is specific for T. parva and more sensitive and faster than other<br />

molecular assays currently used in T. parva diagnostics. The assay is highly reproducible and has been<br />

shown to be reliable in the detection of T. parva at piroplasm parasitaemia levels as low as 8.79x10 -4 %.<br />

Acknowledgements<br />

The authors are grateful to the FAO-EMPRES for the funding granted to attend the <strong>Symposium</strong>.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DETECTION AND STRAIN TYPING OF COXIELLA BURNETII USING MOLECULAR BEACONS IN A<br />

REAL-TIME PCR<br />

W.L. McDonald* 1 , S. Humphrey 1 , K. Fernando 1 , J. Fitzmaurice 1 , A.M.J. McFadden 1 , J. E. Samuel 2 , K.E. Russell-Lodrigue 2<br />

and J. O’Keefe 1<br />

1. Investigation and Diagnostic Centre, MAF Biosecurity New Zealand, Upper Hutt, NEW ZEALAND, 2. Department of Veterinary<br />

Pathobiology, Texas A&M University System Health Science Center, College Station, USA.<br />

Introduction<br />

Q fever is an exotic disease to New Zealand (NZ), but is endemic in the livestock of all of our trading partners<br />

and is a significant zoonotic disease. In humans Q fever manifests in two forms; acute, which is<br />

characterised by flu-like symptoms, pneumoniae or hepatitis and chronic, which is characterised by<br />

prolonged fever and endocarditis. In animals Q fever is usually asymptomatic, but in some cases can cause<br />

abortions and reproductive disorders. Transmission of C. burnetii is primarily by inhalation of infectious<br />

aerosols; ingestion, skin trauma and sexual contact are secondary routes. Q fever has a world-wide<br />

distribution, except for NZ with freedom being established by serological surveys (1). The NZ Import Health<br />

Standards require importers to serologically test donor animals of germplasm. The use of a validated<br />

sensitive PCR assay to screen germplasm would reduce the risk of a border incursion. Furthermore, it is<br />

hypothesised that cattle may harbour different strains of C. burnetii to that of goats and sheep. Strain<br />

differentiation is also important in further understanding the pathogenesis and epidemiology of C. burnetii.<br />

The presentation will provide background information on Q fever and describe the development of a rapid,<br />

real-time PCR employing molecular beacons for the detection and strain differentiation of C. burnetii.<br />

Material & methods<br />

Oligonucleotide primers and molecular beacons were designed to discriminate a SNP at position 745 in the<br />

icd gene of C. burnetii. Strains of C. burnetii that cause persistent Q fever in cattle, ‘acute’ Q fever in<br />

humans and isolated from ticks contain a guanine nucleotide at the SNP site (probe AP). Strains of<br />

C. burnetii that cause abortions in goats and sheep, ‘chronic’ Q fever in humans and isolated from rodents<br />

have an adenine nucleotide (probe C). The analytical sensitivity was evaluated by testing reference strains of<br />

C. burnetii, replicates of ten-fold dilutions of purified C. burnetii DNA and replicates of ten-fold dilutions of<br />

irradiated C. burnetii spiked in extended bovine semen. Preliminary diagnostic sensitivity was conducted by<br />

testing a range of samples from sheep experimentally-infected with C. burnetti at the Texas A&M University.<br />

The analytical specificity was evaluated by testing other bacteria and viruses that may be present in semen<br />

or cause similar disease syndromes. Diagnostic specificity testing was conducted on extended semen from<br />

NZ bulls.<br />

Results<br />

Probe AP detected 100% (8/8) of C. burnetii isolates from ticks, chiggers, cows, and humans with acute<br />

symptoms of Q fever. Probe C detected 100% (10/10) of isolates from rodents, a goat and humans with<br />

chronic symptoms of Q fever. The analytical sensitivity using purified DNA was 100 ag of strain RSA329 with<br />

probe AP and 10 fg of strain Q177 with probe C. The analytical sensitivity for detection of non-viable, whole<br />

cell C. burnetii in semen was 4.2 ± 0.7 CFU/mL. Preliminary diagnostic sensitivity was 100% (12/12) for<br />

placenta, 60% (3/5) for vaginal swabs and 25% (1/4) for milk samples from sheep experimentally-infected<br />

with C. burnetii. The PCR had an analytical specificity of 100% for 50 non-target microorganisms and a<br />

diagnostic specificity of 100% (95% CI: 97 – 100%) when tested against negative controls consisting of 145<br />

semen straws from NZ bulls.<br />

Discussions & conclusions<br />

The real-time PCR is sensitive and specific, and whilst it was developed for use in screening animal<br />

germplasm for C. burnetii, it also has application in investigation of exotic disease notifications and for<br />

surveillance testing in NZ. In countries with Q fever the PCR could provide a tool for the identification of<br />

infected animals facilitating control programs. The icd gene was targeted for the design of the molecular<br />

beacons as it has been previously described to differentiate strains of C. burnetii (2). The animal host range<br />

of strains of C. burnetii has not been conclusively determined and the real-time PCR could be used as a tool<br />

for testing large numbers of samples from infected animals to explore this question. The PCR could be used<br />

as an aid for diagnosis of Q fever in humans and to explore causes of the different disease manifestations.<br />

References<br />

1.. Hillbink, F. 1993. Surveillance. 20:39-40.<br />

2. Van Nguyen, S. and K. Hirai. (1999). FEMS Microbiol: 180:249-254.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1330 - 1455 Concurrent Session 1.2 - FMD Mon 12<br />

CHANGING FACES OF FOOT-AND-MOUTH DISEASE TYPE O PANASIA STRAIN IN INDIA<br />

D. Hemadri, A. Sanyal, C.Tosh, R. P. Tamilselvan, J. K. Mohapatra, S. Saravanan T. J. Rasool, S. K. Bandyopadhyay and B. Pattnaik<br />

Project Directorate on Foot-and-Mouth Disease,<br />

IVRI Campus, Mukteswar-Kumaon, Nainital 263 138, (Uttarakhand) India<br />

November<br />

Foot and mouth disease (FMD) is an acute vesicular disease of wild and domestic ungulates. The causative<br />

agent, foot and mouth disease virus (FMDV, family Picornaviridae, genus Aphthovirus) exists in seven<br />

serologically and genetically distinct types, viz., O, A, C, Asia 1, South African Territories (SAT) 1-3. In India,<br />

the disease is endemic and outbreaks are recorded due to O, A and Asia 1 every year with type O<br />

accounting for nearly 75-80% of total outbreaks. Our earlier studies have shown that a particular lineage,<br />

named PanAsia strain (Knowles et al., 2000) to be predominantly involved in type O outbreaks in India<br />

(Hemadri et al., 2000). Notably, this strain was responsible for an explosive pandemic in Asia, parts of Africa<br />

and Europe during 2000-01 (Knowles et al., 2000). Surprisingly, this strain was overtaken by a strain (new<br />

strain/Ind 2001strain; Knowles et al., 2005) that emerged in 2001 (Hemadri et al., 2002). In this study we<br />

have analyzed type O FMD situation in the aftermath of the emergence of this strain.<br />

Field viruses, collected during 2000-05, either in the form of infected cell culture supernantant or infected<br />

tongue epithelium were used for RNA extraction using RNAeasy (qiagen) mini kit. Genomic region was<br />

successfully amplified by RT and PCR as described previously (Hemadri et al., 2000). A minimum of 450 nt<br />

bases from each of these isolates were used for phylogenetic reconstruction using the programme MEGA 4.<br />

The sequences generated in this study were also compared to the previously published sequences.<br />

The neighbour-joining tree constructed from these isolates showed four major clusters: Cluster I represented<br />

isolates of PanAsia strain. Cluster II was represented by isolates of the recent origin and the sublineage,<br />

new train (NS)/Ind 2001, that caused maximum oubreaks during 2001-02 formed cluster III. Fourth cluster<br />

was represented by fewer isolates recovered during 2000-05.<br />

The study shows following interesting epidemiological patterns; for example there was an upsurge of type A<br />

outbreaks which started in the later part of 1999, continued till mid 2000. Type A outbreaks rose from an<br />

average 10-12% to 33.0% and that of type O fell from an average 75% to 55%. Curiously, despite decrease<br />

in type O outbreaks, there was no change in predominance of PanAsia strains. They caused nearly 50% of<br />

total type O outbreaks (11 of 22 sequences belonged to PanAsia strain). Interestingly, year 2001 saw<br />

outbreak patterns returning to normal with type O being responsible nearly 75% of the outbreaks, but not<br />

with out the emergence of NS/Ind 2001 strain. That year NS/Ind 2001 strain caused nearly 63% of type O<br />

outbreaks, while PanAsia caused 31%.<br />

In the year 2002, outbreaks due to NS/Ind 2001 strain declined sharply (19%), and PanAsia regained<br />

supremacy causing 57% of the type O outbreaks. In 2003, outbreaks due to strain Ind 2001/NS declined<br />

further and we could find only one sequence out of the 30 sequences to be of this strain. Interestingly, the<br />

PanAsia strain, which regained predominance in the previous year, could not hold its position (13.3%) and<br />

was overtaken by strain (PanAsia II?) that originated from it. Year 2004 saw continued dominance of<br />

these strains.<br />

Thus the study shows the continuous evolution of PanAsia strain, which appear to be driven not only by the<br />

multiple rounds of replication, which normally happens in the endemic settings, but also by the competition<br />

among the strains and the serotypes.<br />

Acknowledgements<br />

Indian Council of Agricultural Research for infrastructural facilities and financial support to carry out this work.<br />

Participation of DH in <strong>WAVLD</strong> symposium is supported by FAO, Rome and CSIRO, AAHL, Australia.<br />

References<br />

Hemadri et al. (2000), Epidemiology and infection, 125: 729-736.<br />

Hemadri et al., (2002) Virus genes, 25:23-34.<br />

Knowles et al. (2000) Report of EUFMD, Borovets, Bulgaria, Appendix 1. p. 20–31.<br />

Knowles et al. (2005), Emerging infectious diseases, 11:1887-1893<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE 2001 UK FOOT-AND-MOUTH DISEASE OUTBREAK: AN UPDATE<br />

R. Paul Kitching, Canadian Food Inspection Agency, *presenting author<br />

Following the outbreak of foot-and-mouth disease (FMD) in the United Kingdom (UK) in 2001, the official<br />

inquiry which reported the following year on the lessons to be learned (1) made 80 recommendations, in<br />

addition to the central recommendation that the Government should develop a national strategy for animal<br />

health and disease control. The 80 recommendations were under three headings; a strategy for disease<br />

avoidance and control, appropriate contingency plans and effective preparedness, and managing an<br />

outbreak of disease.<br />

Most of these recommendations are self evident to anyone who has been involved with disease control, and<br />

almost all had appeared in previous reports from previous outbreaks, not only related to FMD. Therefore the<br />

first question could be, why had so little been adopted from previous experience with major disease<br />

outbreaks? Over time husbandry methods change, farming becomes more intensive, species become more<br />

productive and possibly more susceptible to disease, but the basic fundamentals of disease transmission<br />

remain the same. And the obvious statement that if transmission is prevented, the disease will disappear<br />

remains valid.<br />

At an early stage in the outbreak, before the true extent of the spread had been ascertained, a group of<br />

modellers were brought together to interpret the available evidence and produce predictions as to the likely<br />

outcome. Since the outbreak there have been numerous publications showing the flaws in the modellers’<br />

assumptions and consequent predictions (see 2 and 3 for review). Regrettably, because many of the models<br />

were published at the time in reputable scientific journals, these papers are quoted as foundation to support<br />

further propositions, like building on sand. That the outbreak was brought under control and FMD virus<br />

eliminated was inevitable, but at considerable cost. The level of slaughter that took place in the UK outbreak<br />

would no longer be tolerated by the public and consequently by the politicians. Whether this slaughter was<br />

actually necessary is almost now irrelevant. Policies have now been changed in Europe to make vaccination<br />

more acceptable (4). What was a standard and reasonable response to control an outbreak of disease was<br />

distorted by a policy driven by unvalidated models, so as to make this option no longer acceptable, but<br />

without a credible alternative being created.<br />

1. Anderson I. (2002). Foot and mouth disease 2001: lessons to be learned, inquiry. The Stationary<br />

Office, London.<br />

2. Kitching R.P., Thrusfield M.V. and Taylor N.M. (2006). Use and abuse of mathematical models; an<br />

illustration from the 2001 foot and mouth disease epidemic in the United Kingdom. Rev. Sci. Tech.<br />

Off. Int. Epiz., 25, 293-311.<br />

3. Haydon D.T., Kao R.R. and Kitching R.P. (2004). The UK foot-and-mouth disease outbreak - the<br />

aftermath. Nat. Rev. Microbiol., 2, 675-681.<br />

4. European Union (EU) (2003) - Council Directive 2003/85/EC of 29 September 2003 on Community<br />

measures for the control of foot-and-mouth disease repealing Directive 85/511/EEC and Decisions<br />

89/531/EEC and 91/665/EEC and amending Directive 92/46/EEC. Off. J. Eur. Union, L 306 of<br />

22.11.2003, 1-87.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PRODUCTION OF FMDV DIAGNOSTIC REAGENTS TO IMPROVE<br />

OUTBREAK PREPAREDNESS - RECOMBINANT AGS<br />

Jef Hammond<br />

Not available at time of printing.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT AND EVALUATION OF A RECOMBINANT ANTIBODY-BASED COMPETITIVE ELISA<br />

FOR FMD<br />

Introduction<br />

J.D. Muller 1, 2, * , A.J. Foord 1 , H.G. Heine 1 , M. Yu 1 , C.R. Wilks 2 , and L-F. Wang 1<br />

1 Australian Biosecurity CRC, CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong Victoria.<br />

2 The University of Melbourne, School of Veterinary Science, Parkville Victoria<br />

The ability to respond to an outbreak of foot and mouth disease (FMD) relies heavily on our capacity to make<br />

a confident and timely diagnosis. The detection of antibodies to FMD virus (FMDV) is not a definitive<br />

diagnostic tool because it may not differentiate those antibodies generated during a natural infection from<br />

those due to vaccination. FMDV vaccines are composed of whole inactivated virus and hence primarily<br />

induce antibodies to viral structural proteins whereas replicating virus stimulates host antibodies specific for<br />

both structural and non-structural proteins. The availability of quality diagnostic tests that can differentiate<br />

infected from vaccinated animals (DIVA) is a challenge for serological diagnosis. Such tests are crucial for<br />

proving freedom from disease after an outbreak and allowing resumption of trade in livestock products. All<br />

current FMDV DIVA tests rely on polyclonal or monoclonal hybridoma derived antibody reagents, which can<br />

be difficult to prepare and maintain in a quality-assured manner and in the quantities required for postoutbreak<br />

surveillance. The current preferred DIVA test is a competitive ELISA (C-ELISA) designed to detect<br />

antibodies to the non-structural protein 3ABC. The principle of the C-ELISA involves competition between a<br />

known 3ABC-binding antibody and antibodies present in test sera. If 3ABC specific antibodies are present in<br />

the test serum inhibition will be detected. We have improved this assay by the adoption of entirely<br />

recombinant detecting antibody and coating antigen that can be produced in large quantities in E. coli,<br />

making them safe, consistent and more economical to produce without the requirement for infectious virus or<br />

animals.<br />

Material & methods<br />

Chickens were immunised with 3ABC produced in E. coli. Spleens were harvested and libraries were<br />

constructed for phage display. Libraries were panned against the E. coli produced 3ABC and positive clones<br />

were identified, characterized and organised into three distinctive genetic sequences known as recombinant<br />

antibodies (RecAb) 26, 27 and 29 1 .<br />

Western blot and ELISA were used to characterize and define binding specificities.<br />

Two antibodies were further engineered to incorporate different epitope tags for simple and more economical<br />

production and were applied in the FMDV C-ELISA.<br />

Results<br />

The recombinant antibodies bound a specific and well defined epitope region of the non-structural protein<br />

3B. When evaluated in the C-ELISA using sera derived from cattle, sheep and pigs representing either<br />

naïve, FMDV-vaccinated or FMDV-infected animals; RecAb-27 performed the best and was an ideal<br />

replacement for the monoclonal antibodies and FMDV-specific serum currently used.<br />

The generation of different epitope tags for detection has improved the C-ELISA by removing extra<br />

incubation steps; a one-step direct detection can now be performed due to the direct conjugation of alkaline<br />

phosphatase to the RecAb.<br />

Discussions & conclusions<br />

The generation of recombinant antibodies to FMDV-3ABC and the application of these reagents in a C-<br />

ELISA to differentiate infected from vaccinated animals represents the first FMDV DIVA assay in which the<br />

detecting antibody and the coating antigen are recombinant and derived from in vitro expression platforms.<br />

Through engineering of these RecAb we have refined this technology and significantly reduced the time<br />

needed for DIVA C-ELISA.<br />

References<br />

1. Foord, A.J., Muller, J.D., Yu, M., Wang, L.F., Heine, H.G. 2007. Production and application of recombinant<br />

antibodies to foot-and-mouth disease virus non-structural protein 3ABC. J. Immunol. Methods.<br />

321(1-2),142-51.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INTERNATIONAL VALIDATION OF A NEW 3 ABC COMPETITIVE ELISA FOR FMD SEROLOGY<br />

C. J. Morrissy 1 , J. McEachern 1 , A. Colling 1 , L. Wright 1 , Ngo Thanh Long 3 , W. Goff 1 , J. Hammond 1 , M. Johnson 1 , J. Crowther 2 , 3 Dong<br />

Manh Hoa 3 and P. Daniels 1<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory (AAHL), Geelong, Victoria, Australia.<br />

2 International Atomic Energy Agency (IAEA), Vienna, Austria<br />

3 Regional Animal Health Centre, Ho Chi Minh City (RAH0 - 6, HCMC), South Vietnam<br />

Introduction<br />

Foot and Mouth Disease (FMD) is an economically important disease of cloven-hoofed species. It is<br />

prevalent throughout Asia and Africa and is present in South America. North America, Japan, Australasia,<br />

Indonesia and Europe are considered free. FMD virus occurs as seven main serotypes; O, A, Asia 1, C, and<br />

SAT 1, 2 and 3. The disease is usually controlled by vaccination against the prevailing serotypes where it is<br />

endemic, or by stamping out where introduced into disease free areas. Infected animals produce antibodies<br />

to both the serotype specific structural proteins and also the non-structural (NS) proteins of the virus, which<br />

are conserved among the serotypes. Vaccinated animals will produce antibodies predominantly to structural<br />

proteins, depending on the purity of the vaccine. A diagnostic assay based on the non-structural polyprotein,<br />

3ABC, was developed to differentiate animals infected with FMDV from vaccinates, the DIVA strategy.<br />

Routine FMD ELISAs measure antibodies to virus structural proteins, and if positive do not indicate whether<br />

serum is from an animal that has been infected or vaccinated. Another disadvantage of these ELISAs is that<br />

a separate assay is required to measure antibody to each of the seven serotypes of FMD virus. The assay<br />

described here is a competitive ELISA (c-ELISA) that detects antibody to 3ABC regardless of the infecting<br />

serotype. The use of DIVA tests will be important in FMD free countries such as Australia if vaccines are<br />

used to control any future FMD outbreaks.<br />

Materials & methods<br />

Recombinant 3ABC protein was produced in both a baculovirus expression system and an E. coli expression<br />

system. Although the baculovirus system is considered to yield a protein with a more natural conformation<br />

than the E coli expressed protein, the E.coli expressed 3ABC was easier and quicker to grow up in large<br />

amounts than the baculovirus expressed protein, and was more easily purified. Laying chickens were<br />

inoculated with the purified E. coli expressed 3ABC to obtain antibodies from the egg yolk. The more purified<br />

protein was chosen for immunisation to help reduce non-specific reactions in the ELISA. The unpurified<br />

baculovirus expressed 3ABC protein was used as the antigen to coat ELISA plates. The polyclonal antibody<br />

obtained from the chickens egg yolk was used as the competing antibody in the assay. A buffer containing<br />

skim milk powder and normal horse serum was also used to reduce non-specific reactions in the ELISA.<br />

Field sera from FMD disease cases necessary to validate FMD diagnostics were obtained from international<br />

projects and collaborations. The FMD CARD AusAID project has been valuable in improving AAHL<br />

diagnostic capability for detection of antibody and antigen. This project in Vietnam has used the NS c-ELISA<br />

for serosurveillance.<br />

Results<br />

The assay has been validated using a panel of experimental and field sera derived from naïve, vaccinated<br />

and post-infected animals of various species. All vaccinated and naïve animals tested thus far are negative<br />

for antibodies to 3ABC and antibodies to 3ABC have been detected as early as seven days in experimentally<br />

infected animals. The preliminary results indicate that the c-ELISA is sensitive and highly specific.<br />

Conclusions<br />

Validation of new FMD tests such as this C-ELISA for detection of NS antibodies is difficult in Australia<br />

without access to live virus. Samples required for validation of serology tests are obtained through a number<br />

of international collaborations This ELISA is simple to perform and can differentiate post-vaccination sera<br />

from post-infection sera. The current validation data suggests that this c-ELISA will prove to be a reliable<br />

diagnostic assay for post-FMD outbreak surveillance and the assay also offers an additional advantage for<br />

such surveillance in that antibodies to all seven serotypes can be detected in the one test. Hence it can be<br />

used as a simple sero-surveillance tool in FMD free countries. More importantly, because of its DIVA<br />

potential, it could also allow vaccination to be used to control the disease in traditionally disease free areas<br />

to avoid mass slaughtering of animals during an outbreak.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1330 - 1455 Concurrent Session 2.2 - Avian Influenza<br />

RESULTS OF 2006 WILD BIRD SURVEILLANCE FOR THE DETECTION OF HIGHLY PATHOGENIC<br />

AVIAN INFLUENZA IN THE UNITED STATES<br />

Janice Pedersen 1 , Dennis Senne 1 , Mary Lea Killian 1 , Nichole Hines 1 , Brundaban Panigrahy 1 , Seth Swafford 2 , Tom Deliberto 2 , Hon Ip 3<br />

1 U. S. Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services,<br />

National Veterinary Services Laboratories, Ames, IA, USA<br />

2 U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services,<br />

National Wildlife Disease Program, Fort Collins, CO, USA<br />

3 U.S. Geological Survey, National Wildlife Health Center, Madison, WI, USA<br />

The potential role played by migratory waterfowl and water birds in the spread of H5N1 highly pathogenic<br />

avian influenza (HPAI) virus from Asia to Europe, the Middle East, and Africa prompted a large-scale<br />

surveillance to detect HPAI H5N1 in wild aquatic birds in the United States. The program was a cooperative<br />

effort between the U.S. Departments of Agriculture (USDA) and Interior (DOI) and State Wildlife Agencies.<br />

Between April 2006 and March 2007, more than 148,000 cloacal (CL) and fecal swabs were collected from<br />

dead birds as well as apparently healthy and hunter-killed birds in all 50 states and tested for presence of<br />

avian influenza virus (AIV) by the real-time reverse transcriptase-polymerase chain reaction (rRT-PCR)<br />

assay at National Animal Health Laboratory Network (NAHLN) laboratories, the National Wildlife Research<br />

Center, and the National Wildlife Health Center. Cloacal swab pools were screened for AI virus by the matrix<br />

rRT-PCR assay. Positive matrix pools were subsequently tested by the H5 and H7 rRT-PCR subtyping<br />

assays, and individual specimens from H5 and H7 positive pools were shipped to the National Veterinary<br />

Services Laboratories (NVSL) for confirmatory testing, virus isolation (VI), and characterization.<br />

A total of 2210 specimens were submitted to the NVSL for rRT-PCR screening or confirmatory testing. Of<br />

these 1413 specimens, 340 CL pools and 21 fecal pools were H5 presumptive positive (PP), 745 were<br />

surveillance and mortality event swabs, and 31 were viral isolates. Of the 340 H5 PP pools, 250 were<br />

confirmed as H5 rRT-PCR positive, giving the H5 subtyping assay a diagnostic sensitivity and specificity of<br />

74% and 100%, respectively for wild bird CL swab specimens when compared to the AI matrix rRT-PCR<br />

assay. When compared to VI, the H5 assay showed a diagnostic sensitivity and specificity of 79% and 89%,<br />

respectively (64/304 H5 rRT-PCR positive specimens yielded H5 virus and 17 H5 rRT-PCR negative<br />

specimens yielded H5 virus). A total of 97 H5 subtype AI viruses, including 5 H5N1, were isolated and<br />

characterized as low pathogenicity AI (LPAI) of North American lineage by nucleotide sequence analysis.<br />

The H5 subtypes identified were H5N2 (80), H5N1 (5), H5N3 (5), H5N8 (3), H5N9 (3), and H5N4 (1). Other<br />

subtypes isolated included H1-H6, as well as H10 and H11. No highly pathogenic viruses were found and<br />

no H7 rRT-PCR positive specimens were detected, although H7 AI subtype viruses have subsequently been<br />

isolated from rRT-PCR negative swabs.<br />

Diagnostic challenges posed by the surveillance included presence of PCR inhibitors, low virus recovery rate<br />

from rRT-PCR positive specimens, mixed viral infections and the 0% detection rate of H7 wild bird North<br />

American lineage viruses by the USDA H7 rRT-PCR assay.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CANADA’S INTER-AGENCY WILD BIRD INFLUENZA SURVEILLANCE PROGRAMME 2005-2007<br />

*I.K. Barker, Canadian Cooperative Wildlife Health Centre, University of Guelph, Guelph, ON; S. Lair, Centre canadien coopératif de la<br />

santé de la faune, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, QC; P.-Y. Daoust, Canadian Cooperative<br />

Wildlife Health Centre, University of Prince Edward Island, Charlottetown PEI; J. Pasick, National Centre for Foreign Animal Diseases,<br />

Canadian Food Inspection Agency, Winnipeg MB; P.A. Buck, Foodborne, Waterborne and Zoonotic Infections Division, Public Health<br />

Agency of Canada, Ottawa, ON; E.J. Parmley, Centre for Coastal Health, Nanaimo, BC; E.J. Jenkins, Science and Technology Branch,<br />

Environment Canada, Saskatoon, SK; C. Soos, Science and Technology Branch, Environment Canada, Saskatoon; F.A. Leighton,<br />

Canadian Cooperative Wildlife Health Centre, University of Saskatchewan, Saskatoon, SK.<br />

Introduction<br />

Stimulated by a major outbreak of avian influenza (AI) in British Columbia poultry in 2004, and by the<br />

dissemination of H5N1 highly pathogenic avian influenza virus from Asia to Europe and Africa in 2005,<br />

Canada initiated an avian influenza surveillance programme in wild birds in 2005. Organized, under the<br />

oversight of a federal-provincial inter-agency committee, by the Canadian Cooperative Wildlife Health Centre<br />

(CCWHC), it initially involved active surveillance for influenza A viruses in populations of wild birds, mainly<br />

waterfowl, supplemented by scanning surveillance in dead wild birds beginning in September 2005. Active<br />

surveillance was to determine the influenza A viruses circulating in wild bird populations; monitor for those of<br />

animal or human health concern; and establish in Canada an integrated multiagency field, laboratory,<br />

regulatory and communications capacity to carry out Influenza A virus sampling, identification, and molecular<br />

characterization on many specimens under emergency conditions, from any species. Formally initiated in<br />

2006, passive surveillance for highly pathogenic avian influenza in dead wild birds was aimed mainly at<br />

detection of H5N1 influenza virus, but it also contributes to monitoring its global and continental<br />

dissemination, and to the science base for risk assessment.<br />

Materials & methods<br />

Oropharyngeal and/or cloacal swabs were collected into transport medium from live and hunter-harvested<br />

waterfowl and selected other species, sampled at multiple regional sites across Canada in 2005, 2006 and<br />

2007, and in Iceland in 2006. For scanning surveillance oropharyngeal and cloacal swabs were collected at<br />

necropsy from birds of a wide array of species found dead across Canada, emphasizing those using wetland<br />

or aquatic habitat, or involved in mortality events unusual in their circumstances, size or species composition.<br />

In one of seven collaborating regional veterinary laboratories, swabs were initially tested by real-time reverse<br />

transcriptase PCR (RRT-PCR) for Influenza M1 (matrix protein) gene sequences; positive samples were<br />

then tested by RRT-PCR for H5 and H7 protein gene sequences, and any samples positive for these were<br />

forwarded immediately to the National Centre for Foreign Animal Disease, CFIA for further identification,<br />

characterization and virulence testing. In 2005 and 2006, samples testing matrix protein positive and H5/H7<br />

negative were cultured in regional laboratories to attempt virus isolation; in 2007 they were retained frozen.<br />

Isolates were deposited in a national influenza strain archive at NCFAD CFIA. Data of all types were<br />

recorded in the CCWHC national Internet web-interfaced relational database, for rapid communication of<br />

results among laboratories and agencies and to decision-makers.<br />

Results<br />

Through Canada’s Inter-agency Wild Bird Influenza Survey, over 15,000 live wild birds have been sampled<br />

and tested for avian influenza since the fall of 2005. Of these, over 3,000 (>20%) have tested positive for<br />

avian influenza by RRT-PCR. About 3% of over 5400 dead wild birds have been AI matrix protein sequence<br />

positive. The survey results have revealed wide variation in detection of AI viruses among regions and<br />

between species sampled in different years. To date all AI viruses detected have been of low pathogenicity<br />

North American strains. In addition to the Canadian birds tested, over 700 trans-Atlantic migrants (Eastern<br />

High Arctic Brant and Red Knot) were sampled in Iceland and tested in Canada for the presence of avian<br />

influenza in 2006/07. Only 1 has been positive for avian influenza by RRT-PCR.<br />

Discussion & conclusions<br />

The infrastructure and laboratory capacity-building objectives of the programme largely have been met. A<br />

firm base for understanding the species in which AI viruses predominantly circulate has been established,<br />

though highly pathogenic AI viruses have yet to be identified in wild birds in Canada.<br />

References<br />

Parmley, E.J., et al. Influenza viruses in wild ducks in Canada: PCR results from the 2005 Wild Bird<br />

Influenza Survey. Emerging Infectious Diseases. Accepted for publication, 2007.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PERFORMANCE EVALUATION OF FIVE DETECTION TESTS FOR AVIAN<br />

INFLUENZA ANTIGEN WITH VARIOUS AVIAN SAMPLES<br />

Tze-Hoong Chua, AB* Trevor M. Ellis, AC Chun W. Wong, D Yi Guan, EF Sheng Xiang Ge, F Geng Peng, F<br />

Chinta Lamichhane, G Con Maliadis, H Sze-Wee Tan, I Paul Selleck, J and John Parkinson K<br />

(A)School of Veterinary and Biomedical Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia<br />

(B) Agri-Food and Veterinary Authority, 5 Maxwell Road, Singapore 069110<br />

(C) Senior author<br />

(D)Tai Lung Veterinary Laboratory, Agriculture Fisheries and Conservation Department, Lin Tong Mei, Sheung Shui,<br />

Hong Kong SAR, China<br />

(E) Department of Microbiology, The University of Hong Kong, Hong Kong SAR, China<br />

(F) National Institute of Diagnostics and Vaccine Development in Infectious Disease, Xiamen University, Xiamen, Fujian, China<br />

(G) Synbiotics Corporation, 11011 Via Frontera, San Diego, CA 92127<br />

(H) Bio-Mediq DPC (Diagnostic Products Corporation) Pty. Ltd., 1 Williamson Road, Doncaster, Victoria, 3108, Australia<br />

(I) Rockeby Biomed (Singapore) Pte. Ltd., 350 Orchard Road, Shaw House, Singapore 238868<br />

(J) CSIRO Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, 3220, Australia<br />

(K) Department of Agriculture and Food, 3 Baron Hay Court, South Perth, Western Australia, 6151, Australia<br />

INTRODUCTION. In this paper, we report on the evaluation of five influenza antigen detection tests by avian<br />

influenza H5N1 virus–positive swab samples to estimate their diagnostic sensitivity. The tests included two<br />

chromatographic immunoassays, an H5 avian influenza–specific antigen detection enzyme-linked<br />

immunosorbent assay (ELISA), an influenza antigen detection ELISA, and anH5 rapid immunoblot assay.<br />

METHODS & RESULTS. The results showed that the overall sensitivities of these tests ranged from 36.3%<br />

to 51.4% (95% confidence interval ranging from 31.0% to 57.0%), which were comparable to Directigen_Flu<br />

A antigen detection tests but substantially lower than genome detection methods. Diagnostic sensitivity<br />

performance is a function of the concentration of antigens in samples and the analytical sensitivity of the<br />

individual test. The test sensitivities were significantly higher for sick and dead birds by cloacal, tracheal, or<br />

tissue swabs than for fecal swabs from apparently healthy birds, and these tests would not be suitable for<br />

surveillance testing of clinically healthy birds. Furthermore, the sensitivity for testing tracheal and cloacal<br />

swabs from waterfowl and wild birds was not as good as for chickens. This was most likely to be associated<br />

with variation in virus titers between specimens from different bird species. However, the tests showed good<br />

sensitivities for testing brain swabs from clinically affected waterfowl species.<br />

CONCLUSION. The results indicate that these antigen detection tests could be used for preliminary<br />

investigations of H5N1 outbreaks as a low-cost, simple flock test in sick and dead birds for the rapid<br />

detection of H5N1 infection. However, the relatively low sensitivity of the tests as individual bird tests means<br />

that they should be used on optimal clinical specimens from diseased birds, testing birds on a flock basis, or<br />

testing samples as close to the onset of disease as possible before viral titers diminish. They should be<br />

followed up by confirmatory tests, such as reverse transcription polymerase chain reaction or viral culture,<br />

wherever possible but could assist in facilitating rapid investigations and control interventions.<br />

*Presenting author email: T.Chua@murdoch.edu.au


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

OPTIMISED SAMPLING AND PROCESSING FOR ENHANCED DETECTION<br />

OF AVIAN INFLUENZA VIRUS FROM FIELD SAMPLES<br />

S. Warner*, K. O’Riley, A. Welch & D. Grix<br />

Department of Primary Industries,<br />

475 Mickleham Rd, Attwood,VIC 3049, Australia<br />

Introduction<br />

Critical in the expeditious diagnosis of any disease is the ability to collect good clinical specimens and to<br />

transport them to the laboratory in a timely manner. This is particularly important in the diagnosis of an<br />

exotic or emergency disease, and when viable organisms are required for culture. In addition, the<br />

introduction of molecular-based tests has significantly increased the detection of viruses in field samples,<br />

however it is not always possible to grow virus from the same sample in order to confirm the result.<br />

Material & methods<br />

This project was designed to optimise the sampling procedure for enhanced detection of live virus from field<br />

samples. This included the investigation of three viral transport media based on glycerol, Hanks or Brain<br />

Heart Infusion broth, and six storage temperatures including room temperature (approx 20 o C), chilled with<br />

ice packs, 4 o C, –20 o C, dry ice and liquid nitrogen. All three media were inoculated with either high or low<br />

concentrations of AIV, and then assayed by culture in embryonated eggs and by Real time PCR.<br />

Another factor considered in this project was the time delay between collection of field samples and<br />

processing by PCR. This component was made possible since DPI Victoria owns a powered, mobile<br />

diagnostic van fitted with laboratory equipment that can be driven to the site of choice. Fresh faecal samples<br />

were collected from birds and RNA extracted without delay using individual columns in the mobile laboratory.<br />

Samples were also extracted and tested back at the laboratory on days 1, 3, 7 and 14 after storage of the<br />

viral transport media sample at 4 o C. Samples were assayed for the presence of AIV genetic material using<br />

the DPI Type A PCR test.<br />

Results<br />

Overall, the transport media study showed that either the BHI-based or the Hanks-based media can be<br />

considered good transport media when stored at a range of temperatures for the optimal detection of AIV by<br />

culture or PCR.<br />

The mobile van experiment showed that whilst there may be an advantage to processing samples<br />

immediately for viral culture, a delay in the setup time does not have a deleterious effect on PCR results, and<br />

even samples that were stored for two weeks at 4 o C still had strongly detectable PCR results at low dilutions<br />

of virus. These results were also encouraging in the fact that there was no significant decline in sensitivity of<br />

PCR when performing a technique such as RNA extraction in the field.<br />

Discussions & conclusions<br />

Diagnostic samples are often assessed for the presence of both genetic material and live virus. These<br />

experiments were designed to test the ability to detect both genetic material and live virus by using three VTM at<br />

six different temperatures. When all factors were considered, the BHI VTM would be the transport media of<br />

choice for the detection of virus by culture and PCR. The use the mobile diagnostic van is recommended in<br />

situations where rapid results are required and there is an advantage to performing the work on site, such as in<br />

the event of an exotic outbreak index case investigation.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ANALYSIS OF THE SENSITIVITY OF H5N1 ISOLATES FROM THE SOUTH EAST ASIAN REGION TO<br />

OSELTAMIVIR AND ZANAMIVIR<br />

JL McKimm-Breschkin 1 , P Selleck 2 , T Usman 3 , M Johnson 2<br />

1 CSIRO Molecular and Health Technologies, Parkville, Australia; 2 CSIRO Livestock Industries, The Australian Animal Health Laboratory<br />

Geelong, Australia; 3 Disease Investigation Centre, Yogjakarta, Indonesia<br />

Introduction Two different strains of highly pathogenic H5N1 avian influenza have been circulating since<br />

2003. Clade 1 has been fou<br />

nd in Vietnam, Thailand, Cambodia, Lao People’s Democratic Republic, and Malaysia. Clade 2 subsequently<br />

emerged and spread from China to Indonesia, Europe and Africa in 2004-2005. Two influenza specific drugs<br />

oseltamivir and zanamivir are available for the treatment and prevention of influenza. Due to its systemic<br />

availability oseltamivir is the drug of choice for treating humans infected with H5N1 viruses.<br />

Materials and Methods Clade 1 viruses were isolated from chickens, ducks, geese and quail from Vietnam<br />

(2004), Malaysia (2004), Cambodia (2004, 2005) and clade 2 2005 viruses were isolated from Indonesia.<br />

Samples were irradiated and their NAs were tested for sensitivity to zanamivir and oseltamivir. In the<br />

absence of a validated cell culture assay, the 50% inhibitory concentration was measured in the MUNANA<br />

fluorescent based enzyme inhibition assay (1).<br />

Results Despite their origins from different countries and different avian species, all clade 1 and clade 2<br />

isolates had a similar sensitivity to zanamivir, with IC50’s ranging from 1.2 to 1.96 nM, compared to the<br />

human reference strain, mean IC50 of 1.3 nM. Sensitivities of the NAs to oseltamivir fell into three groups<br />

compared to the human H1N1 control. The 2004 clade 1 viruses from Vietnam, Cambodia, and Malaysia<br />

were all more sensitive to oseltamivir, with a mean IC50 of 0.5 nM, compared to 2.2 nM for the human H1N1<br />

control. This is consistent with the findings of Rameix-Welti et al . (2). However, the 2005 Cambodian<br />

isolates were all specifically less sensitive to oseltamivir than the2004 isolates, with a mean IC50 of 2.9 nM,<br />

around 6- fold higher. Of more concern was the third group. The NAs from the clade 2 Indonesian isolates<br />

demonstrated a 15-30-fold increase in IC50, specifically to oseltamivir, with a mean IC50 of 11.5 nM.<br />

Zanamivir and oseltamivir differ by substitutions at the equivalent of the 4 and 6 positions on the sugar ring,<br />

with oseltamivir having a bulky hydrophobic pentyl ether group at the 6’ position. Reorientation of E276 in<br />

the active site is required to accommodate oseltamivir. We therefore tested the sensitivities of a subset of<br />

viruses from each group to 4’ aminoNeu5Ac2en, which is similar to oseltamivir at the 4’ position, but<br />

zanamivir at the 6’ position. All viruses had a similar sensitivity to this inhibitor, with a mean IC50 of 2.5 WM.<br />

This means that the decreased binding to oseltamivir was specifically due to altered interactions around the<br />

6-pentyl ether group, thus explaining why no altered interaction to zanamivir was seen. We have recently<br />

analysed more isolates from Indonesia from 2005, and shown that all have a similar shift in sensitivity, again<br />

specifically only to oseltamivir. Consistent with our earlier findings all viruses had a similar sensitivity to<br />

zanamivir as the control H1N1 virus.<br />

Discussions and Conclusions Analysis of sequences of H5N1 NAs in the public data bases shows that<br />

there is 1 mutation at position 252 in the globular head which varies between all clade 1 and clade 2 NAs<br />

which may decrease oseltamivir binding(2,3). There are also a further 3 amino acid differences between<br />

most clade 1 and clade 2 NAs, and the Indonesian clade 2 isolates have a further amino acid change. Since<br />

none of the sequence variations correlates with any mutation known to confer oseltamivir resistance (3) this<br />

suggests that the decrease in sensitivities may be due to drift mutations rather than from exposure to<br />

oseltamivir. While H5N1 infection remains primarily an infection of birds, the emergence of resistant viruses<br />

in clade 1 infected, H5N1 patients treated with oseltamivir has been suggested to be due to suboptimal<br />

dosing. Since the clade 2 Indonesian isolates have a further decrease in sensitivity compared to the clade 1<br />

isolates, this suggests the standard dosing of oseltamivir may be even less effective. These findings stress<br />

the importance of phenotypic testing of drug sensitivities of avian isolates, since the changes in sensitivities<br />

would not have been detected by genotypic analysis.<br />

References<br />

1 Wetherall NT, Trivedi T, Zeller J, Hodges-Savola C, McKimm-Breschkin JL, Zambon M et al. Evaluation<br />

of Neuraminidase Enzyme Assays Using Different Substrates To Measure Susceptibility of Influenza<br />

Virus Clinical Isolates to Neuraminidase Inhibitors: Report of the Neuraminidase Inhibitor Susceptibility<br />

Network. J Clin Microbiol 2003; 41: 742-50.<br />

2 Rameix-Welti MA , Agou F, Buchy P, Mardy S, Aubin JT, Veron M et al. Natural variation can<br />

significantly alter sensitivity to oseltamivir of Influenza A(H5N1) viruses. Antimicrob Agents Chemother<br />

2006; 50: 3809-15.<br />

3 Russell RJ, Haire LF, Stevens DJ, Collins PJ, Lin YP, Blackburn GM et al. The structure of H5N1 avian<br />

influenza neuraminidase suggests new opportunities for drug design. Nature 2006; 443: 45-9.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1525 - 1720 Concurrent Session 1.3 - Bluetongue & orbiviruses Mon<br />

AN UPDATE ON BLUETONGUE VIRUS IN EUROPE<br />

12<br />

Carrie A. Batten*, Kasia Bankowska, Andrew Shaw, Anna Swain, Abid Bin-Tarif, Narender Maan, Simon Anthony, Chris Oura and<br />

Peter P. C. Mertens<br />

Institute for Animal Health, Ash Road, Pirbright, UK, GU24 0NF.<br />

Bluetongue virus (BTV) is the ‘type species’ of the genus Orbivirus, within the family Reoviridae. BTVs are<br />

November<br />

transmitted between their vertebrate hosts almost exclusively by adult females of certain Culicoides species<br />

(biting midges). They can infect most ruminants, but cause economically important disease (known as<br />

‘bluetongue’ (BT)) primarily in sheep. Clinical signs include fever, hyperaemia, coronitis, oedema,<br />

haemorrhage, erosions and ulcerations of the dermis, and death may occur after 8-10 days. Significant<br />

clinical signs have also been observed in cattle, although these are usually milder, and are seen in a smaller<br />

percentage of animals, particularly after introductions into naive populations (Darpel et al 2007).<br />

Between 1998 and 2005, seven strains of BTV (from five different serotypes: 1, 2, 4, 9 &16) invaded<br />

southern and central Europe. The virus appeared for the first time in northern Europe during August 2006,<br />

with the recognition of typical clinical signs of BT in sheep in the Netherlands, 5 degrees further north within<br />

Europe than ever before. Antibodies to BTV were detected in sera by C-ELISA, and BTV RNA was identified<br />

(at IAH Pirbright) in blood samples from infected animals, using an RT-PCR assay targeting genome<br />

segment 1 (Shaw et al 2007). This confirmed the identification of the BT outbreak by the Netherlands<br />

authorities. The virus was subsequently identified as BTV-8 by serotype-specific RT-PCR (Mertens et al<br />

2007) and sequence analysis of genome segment 2 (Maan et al 2007), showing that it was an entirely new<br />

incursion into Europe. During 2006 the outbreak spread across the Netherlands, Belgium, Germany,<br />

Luxemburg and north-east France, indicating that the whole of Europe is now at risk from this disease.<br />

New cases of BT were reported in Northern Europe up to December 2006. At this time the colder weather<br />

had almost completely removed the population of adult Culicoides in the region, making transmission of the<br />

virus almost impossible. However, BTV has been known to overwinter under similar conditions in other parts<br />

of the world, reappearing when the adult vectors re-emerge in the following spring/summer. On the 6 th July<br />

2007 new cases of BTV started to re-appear in Germany, and subsequently on the 17 th July in Belgium, then<br />

France (19 th July), Netherlands (25 th July) and on 17 th August in Luxemburg. Sequence analyses of genome<br />

segment 2 from German and Netherlands isolates, confirmed that this was a re-emergence of the 2006<br />

strain of BTV-8. These data also demonstrate that the virus had managed to overwinter successfully in<br />

many different locations across Northern Europe.<br />

Until 2007 BT had never been seen in the field within the UK. However, on the 21 st of September 2007<br />

samples from a Highland cow (born in the UK) showing excessive salivation and lameness, were submitted<br />

to the European Community Reference Laboratory for BTV, at Pirbright, UK (IAH reference collection<br />

number: UKG2007/01). The animal was shown to have antibodies to BTV by C-ELISA and BTV RNA was<br />

detected in blood samples by RT-PCR (Shaw et al 2007). The virus was identified as BTV-8 by serotype<br />

specific RT-PCR (Mertens et al 2007). Sequence comparisons to the 24 reference strains of BTV (Maan et al<br />

2007) showed a close relationship with other strains of BTV-8. Detailed comparison with these BTV-8<br />

strains showed that UKG2007/01 clusters with the other isolates of BTV-8 from northern Europe (with ~99%<br />

identity in Seg-2 with strains from Belgium and the Netherlands from 2006 or 2007), confirming that the virus<br />

was derived directly from the continuing outbreaks of disease in northern Europe.<br />

Further testing identified a second infected cow on the same farm, and another infected cow was found ~ 50<br />

kilometres away on a farm at Lowestoft on the east-coast, in the same region of England (both also identified<br />

as BTV-8). Studies of the regional weather patterns in northern Europe, in collaboration with the UK Met<br />

Office, show that a plume of air from the near continent travelled across East Anglia on the 2-4 August. This<br />

plume is likely to have brought the BTV-infected midges. On the 27 th of September The UK Department of<br />

Food and Rural Affairs (Defra) concluded that the virus was being transmitted locally within the UK and<br />

officially declared an outbreak of disease, imposing movement restrictions up to 150km from the infected<br />

premises. Currently, surveillance into the extent of this outbreak is underway within a 10km zone of the<br />

infected premises. Further information will be presented<br />

References<br />

IAH Reference Collection: www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ReoID/BTV-isolates.htm<br />

Darpel, KE, Batten, CA, Veronesi, E, & 11 other authors (2007) Clinical signs and pathology shown by British sheep and cattle infected<br />

with bluetongue virus serotype 8 derived from the 2006 outbreak in northern Europe Vet Rec.161: 253 - 26<br />

Maan S, Maan NS, Samuel AR, Rao S, Attoui H, Mertens PP. (2007) Analysis and phylogenetic comparisons of full-length VP2 genes<br />

of the 24 bluetongue virus serotypes. J Gen Virol. 88, 621-630.<br />

Shaw AE, Monaghan, P, Alpar, HO, & 10 other authors (2007). Development and validation of a real-time RT-PCR assay to detect<br />

genome bluetongue virus segment 1. J Virol Methods (in press) doi:10.1016/j.jviromet.2007.05.014.<br />

Mertens, P.P.C. Maan N. S., Prasad, G.,& 5 other authors (2007). The design of primers and use of RT-PCR assays for typing<br />

European BTV isolates: Differentiation of field and vaccine strains. J Gen Virol 88, 2811-2823.<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

RESPONSE TO THE OUTBREAK OF BTV 8 IN NW EUROPE - TESTING AND TRADE WITHIN THE EU<br />

H. Elliott*, S. Hall, L. Raw, M.Sabirovic<br />

International Trade Department of Environment, Food and Resources, 1A Page St. London. SW1 4PQ<br />

Introduction<br />

NW Europe has experienced an outbreak of bluetongue virus serotype 8 (BTV 8) since 17 August 2006. This<br />

commenced in the Maastricht region and extended over Belgium, The Netherlands, Luxemburg, Germany<br />

and Northern France throughout 2006 and 2007 and on 22 nd September reached Great Britain.<br />

The EU response to the outbreak of bluetongue has been balanced between “control” of the disease and the<br />

economic penalties of restricted movement of animals on trade within and between Member State (MSs). As<br />

bluetongue is a vector born pathogen, the traditional approaches to disease control are limited in<br />

effectiveness. The Council Directive 2000/75/EC of laying down specific provisions for the control and<br />

eradication of bluetongue and the Decision 2005/393/EC on protection and surveillance zones in relation to<br />

bluetongue and conditions applying to movements from or through these zones provided clear guidance but<br />

additional measures were undertaken by the UK in order to prevent bluetongue entering the UK.<br />

Material & methods<br />

The UK identified the pathways by which BTV could enter the country – wind born incursion of infected<br />

midges and by movement of animals and genetic material. Within the provisions of the Decision<br />

2005/393/EC and domestic legislation, the UK would not accept ruminants from the BTV restricted zones.<br />

Through monitoring of the outbreak on the Continent from test results provided by MSs to Commission on<br />

the Animal Diseases Notification System (ADNS) , the UK was able to undertake risk assessments, which<br />

underpinned a compliance based testing regime. Thus post-import testing was implemented on all ruminants<br />

from outside the non-BTV restricted zones. ELISA and PCR tests were carried 7-10 days post arrival and the<br />

animals were restricted to the first destination holding until results were known. Prior advice of the arrival of<br />

an animal to the Division Animal Health Office was provided electronically through the TRAde Control and<br />

Expert System (TRACES. Route plans were checked if transit of a restricted zone has occurred.<br />

Measures to detect bluetongue through the wind born incursion of midges in the UK were based on a<br />

combination of passive and active surveillance. Clinical investigations and testing was undertaken when<br />

there was a suspicion of disease. Sentinel herds were not set up.<br />

Active surveillance of the weather conditions and midge population was undertaken. Meteorological<br />

assessment of weather patterns and conditions favourable to the incursion of midges from the continent<br />

were provided daily in the form of plume maps (Gloster et al, 2007). If a high alert state arose, Defra would<br />

advise the Animal Health Office which would cascade information to local veterinary surgeons and farmers.<br />

Midge traps had been set up and were sampled regularly as part of an ongoing national midge monitoring<br />

project (S Carpenter, personal communication)<br />

A public awareness campaign on the clinical signs of bluetongue was conducted by Defra through the<br />

stakeholders and veterinary profession and to farmers directly through web-based information sites.<br />

Discussions & conclusions<br />

The UK successfully remained free of BTV 8 during 2006 and until September 2007. This was potentially<br />

achieved through a combination of the banning of import of ruminants from BTV restriction zones and<br />

application of a 100% testing regime. It provided not only assurance that animals were virus-free, but<br />

encouraged compliance by importers. Over 4000 animals were tested. The testing strategy was robust but<br />

costly in trade but enabled the UK to remain free of BTV for over 1year.<br />

References<br />

1. Council Directive 2000/75/EC<br />

http://eur lex.europa.eu/pri/en/oj/dat/2000/l_327/l_32720001222en00740083.pdf ;<br />

2. Commission Décision 2005/393/EC<br />

http://eur-lex.europa.eu/LexUriServ/site/en/oj/2005/l_130/l_13020050524en00220028.pdf;<br />

3. Gloster J, Mellor PS, Manning AJ, Webster HN and Hort MC, Assessing the risk of windborne spread of<br />

bluetongue in the 2006 outbreak of disease in northern Europe Vet Rec. 2007 Jan 13;160(2):54-6


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EVALUATION OF ELISAS TO DETECT ANTIBODIES TO BLUETONGUE VIRUS IN<br />

INDIVIDUAL AND TANK MILK SAMPLES.<br />

G Delbridge* 1 , RA Hawkes 1 , A Jugow 1 , B Hoffmann 2 and PD Kirkland 1<br />

1 Virology Laboratory, Elizabeth Macarthur Agricultural Institute, NSW DPI, Menangle, NSW Australia<br />

2 Friederick Loeffler Institute, Insel Reims, Germany<br />

Introduction<br />

Milk samples have been used for the identification of individual animals that have been infected with viruses,<br />

either by the detection of antibodies or direct agent detection. Further, testing of tank (bulk) milk samples has<br />

been used as a very efficient and cost effective method for the identification of infected herds and may be<br />

used to define the geographical distribution of an agent. This study describes the evaluation of ELISAs to<br />

detect antibodies to bluetongue virus (BTV) in both individual and tank milk samples and the subsequent use<br />

to map geographical areas where there has been BTV infection.<br />

Material & methods<br />

BTV group reactive ELISAs using monoclonal antibodies (mAbs) in either competitive or blocking formats<br />

have been used for the detection of antibodies to viruses from the BTV serogroup for many years 1 . In this<br />

project, a range of antigens derived from different BTV serotypes and different monoclonal antibodies were<br />

compared to establish a combination that would provide an assay with optimal sensitivity and specificity. A<br />

limited number of reagent combinations were later compared by testing a panel of serum samples from cattle<br />

and sheep infected with one of the 23 serotypes of BTV. Several thousand samples from naturally infected<br />

cattle, sheep and goats from Australia, China, Italy and Germany and sheep and cattle sera from a BTV free<br />

country were tested to confirm the sensitivity and specificity of the preferred reagent combination. An indirect<br />

ELISA was also developed using cell culture derived antigen and a peroxidase conjugated antibovine IgG.<br />

Matching individual serum and milk samples from naturally infected cattle were tested in both the blocking<br />

and indirect ELISAs. Positive milk samples were titrated to determine the limits of detection and as a means<br />

of estimating herd prevalence. The performance of the blocking ELISA was also compared with all of the<br />

commercial ELISA kits available. Finally, tank milk samples from dairy herds in NSW were tested to evaluate<br />

the use of these assays to map the distribution of BTV.<br />

Results<br />

A blocking assay employing antigen purified BTV23 infected cell cultures was found to provide optimal<br />

sensitivity and specificity and was shown to reliably detect antibodies to each serotype of BTV, both soon<br />

after the onset of infection and also after several years in the absence of re-infection with another serotype.<br />

This blocking ELISA gave superior results in correctly identifying naturally infected cattle, sheep and goats<br />

from Australia, China, Italy and Germany to any of the commercially available assays and also had higher<br />

analytical sensitivity. There was a very high level of agreement between the results for matching serum and<br />

milk samples from individual animals. The indirect ELISA also had good diagnostic performance and was<br />

able to reliably identify BTV infected cattle with any of the serotypes. Both assays could detect antibodies in<br />

tank milk samples from herds where there was a moderate to low prevalence of infection, usually giving<br />

positive results with a prevalence of 5-10%. There was a high specificity when testing either individual or<br />

tank milk samples.<br />

Discussion & conclusions<br />

The results of this study show that these assays are suitable for the detection of BTV antibodies in both milk<br />

and serum samples from individual animals and can detect antibodies to any of the 23 serotypes of BTV. In<br />

regions where there are dairy cattle, the capacity to be able to detect antibodies in tank milk samples<br />

provides a useful tool to define areas in which there has been BTV transmission and to identify BTV free<br />

areas. These should be valuable assays for surveillance in regions where the distribution of BTV is<br />

expanding. Both assays provide advantages. A blocking assay can be used to test specimens from any<br />

animal species while the indirect ELISA can be used in combination with the blocking ELISA when testing<br />

cattle as a confirmatory assay. Further, because of the inclusion of a control antigen, the indirect assay can<br />

be used to clarify the BTV status of suspicious reactors in the blocking ELISA.<br />

References<br />

1. Jeggo, M., Wright, P., Anderson, J., Eaton, B., Afshar, A., Pearson, J., Kirkland, P., and Ozawa, Y. 1992.<br />

Standardization of the competitive ELISA test and reagents for the diagnosis of Bluetongue. In: Bluetongue,<br />

African Horse Sickness, and Related Orbiviruses, Eds Walton, T.E. and Osborne, B.I., CRC, Boca Raton, pp<br />

547-560.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MOLECULAR EPIDEMIOLOGY OF BLUETONGUE VIRUS TYPE 8 FROM THE NETHERLANDS 2006<br />

S. Maan*, N.S. Maan, S.J. Anthony, K.E. Darpel, A.E. Shaw, C.A. Batten, H. Attoui & P.P.C. Mertens<br />

Arbovirology Dept., Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey, GU24 0NF (UK)<br />

Introduction: Bluetongue virus (BTV) (genus Orbivirus, family Reoviridae) is an ‘arbovirus’ with a ten<br />

segmented dsRNA genome that is transmitted between its ruminant hosts by Culicoides biting midges. Since<br />

1998, eight strains of six BTV serotypes (types-1, 2, 4, 8, 9 and 16) have invaded Europe, collectively<br />

representing the largest outbreak of the disease on record, with the deaths of > 2 million animals. Since<br />

1998, the virus has spread further west and north into Europe, initially reflecting changes in the distribution of<br />

Culicoides imicola, the major vector species for BTV in southern Europe and the influence of climate change<br />

[9]. In the summer of 2006, BTV-8 caused a major outbreak of disease in northern Europe, which was<br />

transmitted by novel vector species (C. obsoletus and C. pulicaris groups) [3, 5, 8]. The virus arrived in the<br />

UK (for the first time ever) in September 2007, infecting a Highland cow at a rare-breeds farm in Great<br />

Blakenham, Near Ipswich, in the East Anglian region of England.<br />

BTV serotype is controlled primarily by the outer coat protein VP2 (encoded by genome segment 2 (Seg-2).<br />

Sequencing studies of Seg-2 from different BTV strains, were used to create a sequence database for<br />

molecular epidemiology studies [4, 5, 6, 7, 8], which showed that most serotypes can be divided in eastern<br />

and western strains (topotypes). Live attenuated ‘vaccine’ strains of BTV-2, 4, 9 (western topotype) and<br />

BTV-16 (eastern topotype) were used in attempts to minimise the circulation of BTV in Europe. The South<br />

African ‘Group B’ vaccine strains (types 3, 8, 9, 10 and 11) were also used briefly, in Bulgaria. The release of<br />

these ‘attenuated’ strains, has added genetic diversity to the pool of field strains circulating in southern<br />

Europe, generating an unprecedented mix of eastern and western viruses, leading to reassortment [2].<br />

RT-PCR assays were developed for identification of the 24 BTV serotypes [7, 8] and used to identify the<br />

strains circulating in Europe, including the most recent isolates from the UK (UKG2007/01). The entire<br />

genome (segments 1-10) of BTV-8 from the Netherlands 2006 (NET2006/04) was sequenced and compared<br />

to other European field and vaccine strains, to help determine the origin of the outbreak. This is the first<br />

report of the full genome sequence of the BTV-8 strain from northern Europe.<br />

Material & methods: RNA was extracted from cell-free supernatants, or EDTA treated blood samples, using<br />

the QIAamp Viral RNA Mini Kit (QIAGEN), for serogroup or serotype specific RT-PCR assays [1, 7]. RNA for<br />

synthesis of full length cDNAs and sequencing was purified from cell cultures using Trizol® (Invitrogen), [5,<br />

6]. Sequences were aligned and subjected to phylogenetic analysis.<br />

Results: Phylogenetic analyses of nucleotide (nt) sequence data for all ten genome segments of<br />

NET2006/04 showed up to 98% identity with other viruses derived from a western origin (from the reference<br />

collection at IAH Pirbright), although the most closely related strain varied for each segment. Seg-2 from<br />

NET2006/04 showed >93% sequence identity with the reference strain of BTV-8 (identifying its serotype)<br />

and was most closely related (> 97%) to BTV-8, from Nigeria 1982 (NIG1982/07) showing that it originated in<br />

sub-Saharan Africa. None of the genome segments of NET2006/04 was identical to corresponding segments<br />

of other field or vaccine strains, indicating that it had not exchanged genome segments (reassorted) with<br />

other European viruses. The most recent BTV-8 strain from the UK (UKG2007/01) clusters with other BTV-<br />

8s from northern Europe (~99% nt identity in Seg-2, with strains from Belgium or the Netherlands,<br />

2006/2007), confirming that the UK strain was derived from the northern European outbreak.<br />

Discussions & conclusions: The BTV-8 strain from northern European (2006) was not derived either<br />

directly or by reassortment, from field or vaccine strains already present in the Mediterranean region, but<br />

represents a new introduction to the region. Although its route of entry to northern Europe has not been<br />

established, it originated in Sub-Saharan Africa and is distinct from the BTV-8 vaccine strain. The most<br />

recent strain of BTV-8 from the UK (UKG2007/01) is derived from the outbreak in Belgium or the Netherlands<br />

(2006-2007).<br />

References<br />

1. Anthony S, Jones H, Darpel KE, Elliott H, Maan S, Samuel A, Mellor PS & Mertens PPC (2007). J Virol Methods 141, 188-<br />

197.<br />

2. Batten CA, Shaw AE, Maan S, Maan NS and Mertens PPC (2007). J Gen Virol (Submitted).<br />

3. Darpel KE, Batten CA, Veronesi E, and 11 other authors (2007). Vet Rec 161, 253-261.<br />

4. IAH reference collection: http://www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ReoID/BTV-isolates.htm<br />

5. Maan S, Maan NS, Samuel AR, Rao S, Attoui H, Mertens PPC. (2007a). J Gen Virol 88, 621-630.<br />

6. Maan S, Rao S, Maan NS, Anthony SJ, Attoui H, Samuel AR & Mertens PPC (2007b). J Virol Methods 143,132-139.<br />

7. Mertens PPC Maan NS, Prasad G, Samuel AR, Shaw AE, Potgieter AC, Anthony SJ & Maan S (2007a). J Gen Virol 88,<br />

2811–2823.<br />

8. Mertens PPC, Maan S, Maan NS, Bankowska K, Swain A, Batten C, Carpenter S, Gloster J, Mellor PS & Oura C (2007b).<br />

Promed report Archive Number 20070926.3196, 26-SEP-2007.<br />

9. Purse BV, Mellor PS, Rogers DJ, Samuel AR, Mertens PPC & Baylis M (2005). Nat Rev Microbiol 3, 171–181.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SEQUENCING AND RT-PCR ASSAYS FOR GENOME SEGMENT 2 OF THE 24 BLUETONGUE VIRUS<br />

SEROTYPES: IDENTIFICATION OF EXOTIC SEROTYPES IN THE SOUTHEASTERN USA (1999-2006)<br />

P. P.C. Mertens* 1 , N. S. Maan 1 , D. J. Johnson 2 , E. N. Ostlund 2 , and S. Maan 1<br />

1 Arbovirology Dept., Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey, GU24 0NF (UK)<br />

2 USDA, Animal and Plant Health Inspection Service, Veterinary Services Laboratories (NVSL), Ames, IA<br />

Introduction: Bluetongue (BT) is non-contagious arthropod-borne viral-disease of ruminants that occurs<br />

almost worldwide between latitudes 35 o S and 50 o N. The bluetongue virus (BTV) is transmitted by adult<br />

females of certain species of Culicoides midge (Diptera: Ceratopogonidae). The BTV genome is composed<br />

of ten linear segments of dsRNA, which code for ten distinct viral proteins (VP1-VP7 and NS1-NS3). Twentyfour<br />

BTV serotypes of have been identified world-wide, that can be distinguished in serum neutralisation<br />

assays based on the specificity of reactions between the neutralising antibodies generated during BTV<br />

infection of the mammalian host, and outer-capsid protein VP2 (encoded by Seg-2) [3, 5]. In an area where<br />

multiple strains of different BTV types are co-circulating (e.g. the USA, Australia, Africa, India and Europesince<br />

1998) a full understanding of the epidemiological situation depends on assays that rapidly and reliably<br />

detect and identify the different virus types and strains involved.<br />

Material & methods: RNA for RT-PCR assays was extracted from BTV infected tissue-culture supernatants<br />

or EDTA blood samples, using QIAamp Viral RNA Mini Kit (QIAGEN). Serogroup-specific RT-PCR assays<br />

were as described previously [1, 7]. Details of serotype-specific RT-PCR assays are described by Mertens<br />

et al [5 and in preparation]. Sequencing methods and data for phylogenetic comparisons of BTV genome<br />

segment 2 (Seg-2) from the 24 BTV reference strains have been reported by Maan et al [3, 4].<br />

Results: A nucleotide (nt) sequence database has been developed for Seg-2 of multiple isolates of the 24<br />

BTV serotypes, showing that variations in Seg-2 correlate perfectly with virus type (see link to phylogenetic<br />

trees) [3]. These assays can detect and ‘type’ any of the BTV isolates that were tested from different BTV<br />

serotypes or different geographical origins (i.e. different Seg-2 topotypes within the individual serotypes).<br />

These serotype specific assays (and primers) showed no cross-amplification when evaluated with multiple<br />

isolates of the most closely related BTV types (same nucleotype [3]), or with reference strains of the<br />

remaining 24 BTV serotypes [5]. These ‘type’ specific primers are listed on the internet and will be<br />

periodically updated to maintain their relevance to current BTV distribution and epidemiology. These RT-<br />

PCR assays have been used at IAH Pirbright to identify BTV-8 in northern Europe (2006-2007) (e.g.<br />

UKG2007/01), and BTV-4 / 15 in Israel 2006, BTV-1 in North Africa in 2006 (see link to phylogenetic trees).<br />

Isolations and identification of BTV in the USA are routinely performed at the National Veterinary Services<br />

Laboratories (NVSL), Ames, IA. Prior to 1999, five BTV serotypes (BTV-2, BTV-10, BTV-11, BTV-13, and<br />

BTV-17) were considered endemic in the USA. However, several isolates from Florida (1999-2005), failed to<br />

‘type’ as members of these existing U.S. The serotypes of these isolates were subsequently identified by RT-<br />

PCR assays and sequence analysis of Seg-2. These include: BTV-3 (multiple isolates from Florida 1999-<br />

2003); BTV-5 (USA2003/05); BTV-6: from Florida 2006. BTV-14 (USA2003/03): BTV-19 (USA2003/04):<br />

BTV-22 (USA2002/02). More details concerning each isolate can be obtained from links at:<br />

www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ReoID/virus-nos-by-country.htm#USA<br />

Discussions & conclusions: Oligonucleotide primers and RT-PCR assays, targeting Seg-2, have been<br />

developed for the rapid identification (within 24h) of the 24 BTV types. These assays have been used to<br />

identify the 6 current European types (1, 2, 4, 8, 9 & 16) [5]. These assays have also been used to identify 6<br />

BTV types that are new introductions / exotic to USA (type- 3, 5, 6, 14, 19 and 22). The assays are fast,<br />

sensitive and specific, showing perfect agreement with results of conventional virus neutralisation assays.<br />

They can also positively identify BTV serotype, even if the animal has been infected with several different<br />

BTV virus types, leading to a non-specific serological response [2]. BTV serotype and virus strain (topotype)<br />

can be confirmed by sequence analyses and phylogenetic comparisons of the Seg-2 cDNA products. Since<br />

1998, several exotic BTV serotypes (types have spread into the USA and Europe at approximately the same<br />

time. These events in Europe have been linked to changes in climate and their effects on vector insect [6].<br />

References:<br />

Global distribution of BTV types: www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/btv-serotype-distribution.htm<br />

Phylogenetic trees for Seg-2 of BTV types: www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/btv-seg-2.htm<br />

Seg-2 specific primers: www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ReoID/rt-pcr-primers.htm<br />

BTV accession numbers: www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/orbivirus-accession-numbers.htm<br />

Reports concerning recent BTV outbreaks www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/outbreaks.htm#top<br />

BTV reference collection: http://www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ReoID/BTV-isolates.htm<br />

1. Anthony S, Jones H, Darpel KE, & 5 other authors (2007). J Virol Methods 141, 188-197.<br />

2. Jeggo MH, Gumm ID, & Taylor WP. (1983) Res Vet Sci. 34, 205-211.<br />

3. Maan S, Maan NS, Samuel AR, Rao S, Attoui H, Mertens PP. (2007a).J Gen Virol. 88, 621-630.<br />

4. Maan S, Rao S, Maan NS, & 4 other authors (2007b). J Virol Methods 143,132-139.<br />

5. Mertens, P.P.C. Maan N. S., Prasad, G.,& 5 other authors (2007). J Gen Virol 88, 2811-2823.<br />

6. Purse BV, Mellor PS, Rogers DJ, & 3 other authors (2005). Nat Rev Microbiol 3, 171–181.<br />

7. Shaw AE, Monaghan, P, Alpar, HO, & 10 other authors (2007). J Virol Methods 145,115-126.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

REPLICATION OF BLUETONGUE VIEUS IN THE SKIN OF INFECTED SHEEP<br />

Karin E. Darpel 1 , Paul Mongahan 1 , Jennifer Simpson 1 , Harriet W. Brooks 2 , Simon J. Anthony 1 , Eva Veronesi 1 , Joe Brownlie 2 , Natalie<br />

Ross-Smith 1 , Haru H. Takamatsu 1 , Philip Mellor 1 and Peter P.C. Mertens 1<br />

1<br />

Institute for Animal Health, Pirbright Laboratory, Ash Road, Woking, Surrey, GU24 0NF (UK)<br />

2<br />

Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Herts, AL9 7TA<br />

Introduction:<br />

Previous studies of BTV pathogenesis and tissue tropism in the infected mammalian host have been<br />

primarily based on pathology and the time course of virus isolation, targeting different organs (Pini 1976;<br />

Lawman 1979; MacLachlan et. al. 1990). However, these methods are unable to identify which cell types<br />

within each organ are infected and do not differentiate infectious virus present in the bloodstream, from virus<br />

which is actively replicating in the cellular components of the organs itself.<br />

The separation between active virus replication and the simple physical presence of virus in infected animal<br />

cells can be achieved using immunolabelling and confocal microscopy. Detection of BTV NS proteins, which<br />

are not carried into the cell by the infecting virus particles, represents a clear indication of virus replication.<br />

However, earlier immunofluoresence microscopy studies of BTV infection in ruminants gave relatively poor<br />

detection of viral proteins and therefore often also gave inconclusive or unsatisfactory results (Mahrt and<br />

Osburn 1986; MacLachlan et.al. 1990). Here the replication of BTV in ruminants was further investigated,<br />

successfully using modern confocal-microscopy to detect viral proteins in different organs.<br />

Material and Methods:<br />

Tissues were collected during the necropsy of BTV-2 (RSA1971/03) infected sheep (at 3,4,6,8,and 9 d.p.i.),<br />

BTV-8 (IAH reference collection number NET2006/01) infected sheep (8 d.p.i.) and cattle (10 d.p.i.),<br />

processed as described previously by Mongahan et. al. (2005). BTV proteins were stained using anti-NS2<br />

antibodies (ORAB 01 and ORAB0268) that had been generated using bacterially expressed and purified<br />

NS2 protein from BTV-1 (RSArrrr/01), the A3 monoclonal antibody against VP7 (ORAB036) and antibodies<br />

raised against purified BTV-1 core particles (ORAB06). Antibody binding was detected with species-specific<br />

fluorescent conjugate. Skin samples from other BTV infected sheep (several serotypes) were investigated for<br />

BTV persistence by establishing primary fibroblast cultures, which showed cytopathic effect (CPE) if BTV<br />

was present (confirmed by RT-PCR – Anthony et.al. 2007/ Maan 2004)<br />

Results:<br />

Viral antigens were found in two major cell populations: micro-vascular endothelial cells and leukocytes<br />

(lymphocytes, dendritic cells and monocytes). Infected endothelial cell were detected very early postinfection<br />

(3 dpi), in the head lymph-nodes, tonsils, tongue, lips and skin. A time-course study of sheep<br />

infected with BTV-2 (RSA1971/03) suggested that leukocytes and endothelial cells are infected<br />

simultaneously. Early manifestation of BTV replication (and clinical signs) may depend on the susceptibility<br />

and damage to endothelial cells in different organs. Viral proteins were found in local foci within certain<br />

organs, suggesting local spread, possibly within the capillary bed of the organs themselves.<br />

Infection and replication of BTV was observed in sheep skin, soon after infection (3 dpi), particularly in<br />

endothelial cells of the smaller blood-vessels. Infection of the skin intensified, peaking at 6-8 dpi. At this<br />

stage some skin-glands contained BTV proteins. The virus occasionally persisted in the skin, even after the<br />

viraemia had subsided.<br />

Conclusion:<br />

The skin is the ‘transmission-organ’ for BTV, in both directions between the ruminant-host and insect-vector.<br />

It was demonstrated that BTV replicates in the vascular endothelial cells (and to a lesser extend in<br />

leukocytes) of the skin. The skin was also one of the organs (along with head lymph nodes and tonsil) were<br />

viral proteins were detected in the largest amounts. The skin is the largest single organ of the body,<br />

suggesting that it may also be the major organ involved in BTV replication.<br />

BTV isolation from skin samples of BTV infected sheep does not appear to correlate with the detected levels<br />

of systemic viraemia. It may therefore be possible that feeding midges (Culicoides vectors) can take up and<br />

become infected by virus produced locally in the skin, even if the level of circulating viraemia is low or<br />

absent. Ruminants with a low viraemia may still be important for virus transmission. Lymphatic organs like<br />

head lymph nodes and tonsil not only contained viral proteins in infected leukocytes but endothelial cells of<br />

capillaries and small blood vessels were also strongly infected.<br />

References:<br />

BTV reference collection: http://www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ReoID/BTV-isolates.htm<br />

Anthony et.al. 2007 J.Virol.Methods 141, 188-197.<br />

Lawman 1979 Thesis University of Surrey.<br />

Maan 2004 Thesis University of London.<br />

MacLachlan et.al. 1990 Vet.Path. 27, 233-229.<br />

Mahrt and Osburn 1986 Am. J. Vet.Res. 47,1198-1203.<br />

Mongahan et. al. 2005 J.Virol. 79, 6410-6418.<br />

Pini 1976 Oond.J.Vet.Res. 43,159-164.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

VALIDATION OF A DIAGNOSTIC METHOD FOR THE DETECTION AND QUANTIFICATION OF<br />

BLUETONGUE VIRUS IN ADULT CULICOIDES BITING MIDGES (DIPTERA: CERATOPOGONIDAE).<br />

*Eva Veronesi , Peter P.C. Mertens, Philip S. Mellor and Simon Carpenter<br />

Institute for Animal Health, Ash Road, Pirbright, Surrey, GU24 ONF, UK.<br />

Introduction: Bluetongue (BT) is a non-contagious arthropod-borne viral disease of domesticated and wild<br />

ruminants that is transmitted by the bite of adult females, from certain species of Culicoides biting midge<br />

(Diptera: Ceratopogonidae). Screening of field caught midges for the presence of bluetongue virus (BTV)<br />

can provide valuable information concerning BTV transmission during an outbreak of the disease.<br />

In this study we describe the validation of an efficient method to quantify BTV from adult midges. We present<br />

results for virus isolation from adult C. sonorensis (derived from the colony at IAH Pirbright) that were<br />

intrathoracically infected with BTV, then homogenised using a conventional mortar and pestle method.<br />

These data were compared with results obtained using a TissueLyser® machine (Qiagen), which<br />

homogenises samples by high frequency shaking in the presence of a ball-bearing. This machine has<br />

already been used in several laboratories to extract viral DNA or RNA from infected tissues or insects (e.g. to<br />

isolate West Nile Virus from wild caught mosquitoes) but has not previously been validated for quantification<br />

of virus in an arboviral/vector system. A quantitative assessment of viral load in the vector is thought to be<br />

particularly important for BTV, since a titre of ≥3.0 log10TCID50/midge represents a fully disseminated<br />

infection (Fu et al. 1999) and is required for virus transmission to the ruminant host during a blood meal.<br />

Material & Methods: In order to ensure the presence of the virus, adult C. sonorensis were individually<br />

inoculated (intrathoracically) with BTV serotype 9 (BTV-9, IAH reference collection number KOS2001/03).<br />

Immediately after inoculation, equal numbers of these insects were homogenised (individually) using either a<br />

mortar pestle, or TissueLyser® method. The TissueLyser® homogenisation parameters that were optimised,<br />

include: 1) Duration of shaking; 2) Frequency of shaking; 3) Material of the ball-bearing (polyethylene /<br />

stainless steel); and 4) Size of ball-bearing (1, 2, 3, 4, 5 mm diameter). The efficiency of virus recovery was<br />

assessed by titrating a ten fold dilution series of each homogenised insect on BHK-21 cells. Adult C.<br />

sonorensis were also infected via an oral route, then incubated at ±25ºC for 10 days to allow virus<br />

replication. Equal numbers of these midges were individually homogenised using either a mortar and pestle<br />

or optimised TissueLyser® method, and virus isolation was carried out as before. Real time RT-PCR (Shaw<br />

et al 2007) was used to confirm the identity of BTV infected insects.<br />

Results: The optimum TissueLyser® homogenization program, involved shaking insects in 100 µl of<br />

Glasgow MEM for 1 min at 25 Hz with a 2 or 3 mm stainless steel ball bearing. The virus titres obtained<br />

using oral or intrathoracic infection techniques and the optimised program, did not differ significantly from<br />

those obtained using a polypropylene motor-driven pestle (a method that is currently in common use for<br />

studies of vector competence). Although a significant difference in the amount of virus obtained was<br />

observed between different homogenisation programs (H = 57.73; df = 7; P


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

WILD LIFE AS A SOURCE OF ZOONOTIC DISEASES<br />

Tin Tin Myaing and Tay Zar Aye Cho<br />

Despite the discovery of cooking last 1.9 million years ago, the risk of zoonotic diseases emerging from<br />

hunting and eating wildlife is still of global importance. World landscape and habitat of the wild animals has a<br />

kaleidoscope of niches, leading to the diseases transmitted from animals to man. Infectious pathogens of<br />

wild animals'origin have become increasingly important on substantial impacts of human health, agricultural<br />

production, international trade, wildlife-based economies, native wildlife populations, wildlife conservation,<br />

and the health of ecosystems. Expanded demand for bush meat will likely lead to changes in the exposure of<br />

humans to potentially zoonotic microbes.The ancient wild life epidemic zoonotic disease was bubonic<br />

plague, emerged in 14 th century, killed approximately one third of Europe’s population transmitted by fleas<br />

and the reservoir was Ratus ratus. Rabies was described in Mesopotamia in hunting dogs as early as<br />

2,300BC. Alexander the Great passed in 323 BC, died of encephalitis caused by West Nile Virus of a wild<br />

bird reservoir as the modern hypotheses suggestion. HIV/AIDs, SARs, Nipah virus, Rabies and Avian<br />

Influenza H5N1 are some zoonotic diseases of interest, originated from wild life reservoir. House rat (Rattus<br />

rattus robustulus), fruit bats and flying fox (Pteropus vampyrus) and Civet cat(Paguma larvata) were the<br />

source of some zoonotic diseases originated from wild animals, populated in Asian countries and Myanmar.<br />

There were evidences of warble fly larvae from Asian Elephant (Elephas maximus) transmitted to mahout’s<br />

skin at arm and legs, in Myanmar. A 2/50(4%) cases of warble fly transmitted to mahout's skin ( palm and<br />

toe) was observed in 2005. Mechanical removal of the embedded larva under the skin had been observed.<br />

There was no further systematic treatment as well as no clinical signs except slight itching. Transmission of<br />

Anthrax disease 1% (1/100) (cutaneous form) to a worker who cut off the internal organs of the elephant’s<br />

dead body was investigated in 2006 but infected person took antibiotic therapy and saved. Tuberculosis<br />

infected in persons closed contact with elephants have been observed but not yet been identified. Civet cat<br />

(Paguma larvata) was valuable in Myanmar due to its perfume like smell produced from anal gland. Civet cat<br />

was said to be the reservoir of SARs disease. Understandings the emergence of zoonotic agents requires<br />

knowledge of pathogen biodiversity in wildlife, human-wildlife interactions, anthropogenic pressures on<br />

wildlife populations, human behavior and demographic factors. To reduce risk for emerging zoonoses, the<br />

public should be educated about the risks associated with wildlife, bush meat, and exotic pet trades, better<br />

understanding of how deforestation and associated hunting leads to the emergence of novel zoonotic<br />

pathogens. Control of emerging zoonotic diseases of wild life origin should be relies on national,<br />

international, regional and cross sectional networks, public health and veterinary services should be more<br />

strengthened, especially the services can become more prepared to respond to endemic and epidemic<br />

diseases. Veterinarian diagnosticians should have wide knowledge and skillful in their field of interest. On the<br />

other hand wild life conservation and animal welfare should also be considered as an important theme.<br />

Key words: wild life based economy, emerging zoonoses, bush meat hunting,<br />

public health, animal welfare<br />

corresponding author: tintinmyaing@mail4u.com.mm<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SURVEILLANCE OF MYCOBACTERIUM BOVIS INFECTION IN CATTLE<br />

IN GREAT BRITAIN DURING 2006<br />

K.L.Jahans*, D.R.Worth, J.Brown, J.M.Gunn, M.Okker, E. Wood and L. Potts<br />

Veterinary Laboratories Agency, New Haw, Addlestone, Surrey KT15 3NB.<br />

Introduction<br />

Despite an intensive test and slaughter programme to prevent cattle-to-cattle transmission, Great Britain has<br />

one of the highest animal and herd incidences of bovine TB in Europe. The Veterinary Laboratories Agency<br />

has seen an increase in isolates submitted for TB diagnosis since 2002 following the occurrence of Foot and<br />

Mouth Disease in 2001. During most of that year (mid February to November) routine testing of cattle for<br />

bovine tuberculosis was suspended. Up to 2005, the number of incidents in Great Britain has increased by<br />

an average of 16% each year since the mid 1980s.<br />

Cattle and other bovine species, such as buffaloes and bison, are the natural hosts but nearly all warmblooded<br />

animals, including humans can be affected. Species are not all equally susceptible or share the<br />

same ability to act as reservoirs of infection for other species: some are considered spill-over hosts, whereas<br />

others (e.g. cattle and badgers) act as true maintenance hosts.<br />

Materials & methods<br />

Bovine tissue samples (approx 20g) were received in sterile plastic pots with a standard form. Samples from<br />

other species were also received but tended to be smaller and arrived with a similar form or letter. Details<br />

from both submission types were entered on a database. Lesioned tissue was selected over<br />

non-lesioned material.<br />

Approximately 1cm 3 of tissue containing lesions was placed in 50ml buffered formaldehyde solution for<br />

histopathology. The remainder was homogenized with a solution of 5% oxalic acid in order to destroy any<br />

contaminating organisms, centrifuged at 1100g for 10 minutes and the resulting deposit washed and resuspended<br />

in 0.85% sterile saline. Then approximately 300 µl were sown onto selective media and<br />

incubated for 6 weeks at 37 o C.<br />

Identification of M. bovis was usually made by its appearance on solid media slopes. PCR and spoligotyping<br />

was used in addition for more accurate differentiation using the methods of Wilton and Cousins (1992) and<br />

Kamerbeek et al (1997). Molecular typing of Mycobacterium species other than members of the TB<br />

complex, M. avium or M. intracellulare was carried out using a kit produced by Hain Lifescience<br />

(http://www.hain-lifescience.com).<br />

Results<br />

In 2006 the TB Diagnosis Section at the Veterinary Laboratories Agency - Weybridge received 13,907<br />

bovine tissue submissions for confirmation of diagnosis by culture. The number of submissions from other<br />

species (excluding badgers) was 690 and consisted mainly of a non-random sample of deer, cat, dog, pig,<br />

sheep, goat and camelids. 4,710 (34%) of the cattle submissions and 83 (12%) of submissions from these<br />

other species were found to be positive for M. bovis. There has been no significant increase in the number<br />

of confirmed bovine tuberculosis incidents in Great Britain in 2006 compared to 2005 (2.49% and<br />

2.45% respectively).<br />

Discussion & conclusions<br />

Spoligotypes have become more widspread and some of this may have been due to the restocking of herds<br />

with cattle from areas of high TB prevalence. Veterinarians are gradually making more use of molecular<br />

typing data, in conjunction with cattle movement data, as an aid to tracing the origins of infection because<br />

spoligotypes are generally found in geographical clusters. The available evidence suggests that cases in<br />

farmed and companion animals were the result of infection spill-over from a cattle or wildlife reservoir.<br />

References<br />

Kamerbeek J, Schouls L ,Kolk A, van Agterveld M, van Soolingen D, Kuijper S, BunschoteA, Molhuizen H, Shaw R, Goyal M and van<br />

Embden J (1997). Simultaneous detection and strain differentiation of Mycobacterium tuberculosis for diagnosis and epidemiology.<br />

Journal of Clinical Microbiology, Apr 1997, 907-914, Vol 35, No. 4.<br />

Wilton S and Cousins D (1992). Detection and identification of multiple mycobacterial pathogens by DNA amplification in a single tube.<br />

PCR Methods Appl. 1992 May;1(4):269-73.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CANINE RABIES IN SOUTH AFRICA: IDENTIFICATION OF A NEW LINEAGE IN LIMPOPO AND A<br />

RECENT SPREAD INTO THE FREE STATE PROVINCE<br />

Introduction<br />

Chuene Ernest Ngoepe a, * , Gugulethu Zulu a , Claude Sabeta a , Louis Nel b<br />

a Rabies Unit, Onderstepoort Veterinary Institute, Private Bag X05, Onderstepoort, 0110<br />

b University of Pretoria, Microbiology and Plant Pathology, 0002 Pretoria, South Africa<br />

In 2005, there was a drastic and unexpected increase of dog rabies cases in the Limpopo province.<br />

Laboratory confirmed cases increased from 5 in 2004 to 35 in 2005 and to 100 in 2006. The outbreak of<br />

rabies in domestic dogs was followed by a human rabies outbreak in which at least 30 human deaths were<br />

confirmed between 2005 and 2006. In contrast, the Free State province has been historically associated with<br />

endemic rabies in the yellow mongoose Cynictis penicillata (Snyman, 1940; Swanepoel et al. 1993). Due to<br />

spillover events, rabies viruses of the mongoose biotype were recovered from domestic dogs. More recently,<br />

there has been increased number of rabies cases of the canid biotype reported in domestic dogs in this<br />

province. The objectives of this study were; 1. to establish the exact source of infection in the human rabies<br />

cases and the origin of the rabies virus lineage responsible for the recent rabies outbreak in Limpopo, and 2.<br />

to trace the origin of canid rabies and assess the public health threat of mongoose rabies (in dogs) in the<br />

Free State province.<br />

Material & methods<br />

A molecular epidemiological study was therefore performed on a cohort of 98 rabies viruses recovered from<br />

domestic dogs between 1995 and 2007 from the Free State province and 52 rabies viruses recovered from<br />

domestic dogs, jackals and humans from Limpopo and southern Zimbabwe. The cytoplasmic domain of the<br />

glycoprotein and the G-L intergenic region of the rabies viruses in this study sample were amplified and<br />

sequenced. Phylogenetic trees were reconstructed from an alignment of a 592-bp region of the genome<br />

under investigation.<br />

Results<br />

Case 1: Phylogenetic analysis revealed that human rabies viruses were closely related to those obtained<br />

from domestic dogs in the same locality and the rabies viruses from Limpopo were closely related to viruses<br />

obtained from southern Zimbabwe.<br />

Case 2: The phylogenetic analyses segregated the rabies viruses in this study sample into two main<br />

clusters; the genetically compact canid rabies biotype and a second group of heterogeneous viruses of the<br />

mongoose rabies biotype. From the data, it could be demonstrated that viruses of the canid rabies group<br />

were recently introduced into this part of South Africa.<br />

Discussions & conclusions<br />

The reasons for the emergence and rapid dissemination of the new dog rabies strain in Limpopo are not very<br />

clear. It appears that common rabies infection cycles persist between domestic dogs and jackals in southern<br />

Zimbabwe and northern South Africa. It is evident that the new canid group belongs to the same<br />

epidemiological cycle circulating in dogs in both the Free State province and across the international border<br />

with Lesotho. Our results confirm that spillover of the mongoose biotype into domestic dogs lead to dead-end<br />

infections. In comparison to mongoose rabies that is endemic here, canid rabies has emerged to become of<br />

much greater importance to the public and veterinary health sectors of this region.<br />

References<br />

1. Swanepoel, R., Barnard, B. J. H., Meredith, C. D., Bishop, G. C., Bruckner, G. K., Fogging, C. M.,<br />

Hubschle, O. J. B., 1993. Rabies in Southern Africa. Onderstepoort Journal of Veterinary Research, 60: pp.<br />

325-346.<br />

2. Snyman, P.S., 1940. The study and control of vectors of rabies in South Africa. Onderstepoort Journal of<br />

Veterinary Science Animal Husbandry, 66: pp. 296-307<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CAPRIPOXVIRUS TROPISM AND SHEDDING: A QUANTITATIVE TIME-COURSE STUDY IN<br />

EXPERIMENTALLY INFECTED SHEEP AND GOATS<br />

T R Bowden 1,* , S L Babiuk 2,3 , M P Anderson 1 , G R Parkyn 2 , R P Kitching 2 , J S Copps 2 and D B Boyle 1<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia<br />

2 Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg,<br />

Manitoba R3E 3M4, Canada<br />

3 University of Manitoba, Department of Immunology, Basic Medical Sciences Building 730 William Avenue, Winnipeg,<br />

Manitoba R3E 0W3, Canada.<br />

Introduction<br />

Sheeppox virus (SPPV) and goatpox virus (GTPV), which are members of the genus Capripoxvirus in the<br />

family Poxviridae, cause systemic disease in sheep and goats that is characterized by fever, generalized<br />

skin nodules, lesions in the respiratory and gastrointestinal tracts and lymph node enlargement. SPPV and<br />

GTPV are endemic in north and central Africa, the Middle East, central Asia and the Indian subcontinent,<br />

where they are a cause of significant morbidity and mortality in susceptible sheep and goats. Despite the<br />

considerable threat that these viruses pose to small ruminant production and to global trade in sheep, goats<br />

and their products, knowledge of the underlying pathogenesis of sheeppox and goatpox has long been<br />

deficient. To increase understanding of the pathogenesis of these diseases, we undertook quantitative timecourse<br />

studies in sheep and goats, utilizing real-time PCR and traditional virological assays, following<br />

intradermal inoculation of Nigerian SPPV or Indian GTPV in their respective homologous hosts.<br />

Materials & methods<br />

Nine sheep and nine goats were inoculated intradermally with either 10 4.9 TCID50 of Nigerian SPPV (sheep)<br />

or 10 4.4 TCID50 of Indian GTPV (goats). To enable absolute quantitation of viral DNA concentrations in blood,<br />

swabs and tissue samples, we developed a quantitative real-time PCR TaqMan assay to amplify and detect<br />

a short product from within ORF074 of capripoxvirus genomes. Multiplexed detection of either an<br />

endogenous or exogenous control, depending on the sample type, provided a means of verifying the<br />

absence of PCR inhibitors in samples tested. Isolation of infectious virus from clinical materials was routinely<br />

attempted by inoculation of samples, in duplicate, onto 80-90% confluent ovine testis cell monolayers.<br />

Capripoxvirus-positive specimens were subsequently titrated on replicate cell monolayers to determine<br />

sample infectivity.<br />

Results<br />

Viral DNA concentrations in blood, swabs, urine, faeces and solid tissues varied by as many as six orders of<br />

magnitude. Viraemia, determined by real-time PCR and virus isolation, cleared within two to three weeks<br />

post inoculation. Peak shedding of viral DNA and infectious virus in nasal, conjunctival and oral secretions<br />

occurred between days 10 and 14 post inoculation, and persisted at low levels for up to an additional three to<br />

six weeks. The highest infectious titres in nasal, conjunctival and oral swabs were observed in goats, and<br />

correlated with the greater severity of disease observed in these animals. Although gross lesions were<br />

evident in multiple organs at necropsy, highest viral titres were detected in skin as well as in lung and<br />

discrete sites within oronasal tissues and gastrointestinal tract. Of all tissues tested, skin was consistently<br />

positive throughout the sampling period.<br />

Discussion & conclusions<br />

Our studies have provided novel insights regarding replication, dissemination and shedding of<br />

capripoxviruses in their natural hosts, the pathogenesis of which is similar to smallpox and monkeypox<br />

where greatest viral replication occurs in the skin. The consistently high concentrations of viral DNA in skin,<br />

along with its high infectivity, suggest that mechanical transmission by biting insects might occur more<br />

frequently than previously thought. Furthermore, long term shedding of virus from mucosal surfaces<br />

illustrates the potential for some animals to remain infectious for up to two months after the onset of fever.<br />

These findings should facilitate development of improved strategies for detection and control of capripoxvirus<br />

infections in sheep and goats.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

An indirect ELISA, based upon recombinant capripoxvirus antigens, for serological detection of<br />

sheeppox, goatpox and lumpy skin disease<br />

David B Boyle 1 , Timothy R Bowden 1 , Vicky Boyd 1 , Christine J Duch 1 , Mary-Ann P Anderson 1 , Vicky Stevens 1 , John R White 1 , Barbara E.<br />

Coupar 1 , Shawn L Babiuk 2,3 , Geoff R Parkyn 2 , R Paul Kitching 2 and John S Copps 2<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia<br />

2 Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba,<br />

Canada R3E 3M4<br />

3 University of Manitoba, Department of Immunology, Basic Medical Sciences Building 730 William Avenue, Winnipeg MB,<br />

Canada. R3E 0W3<br />

Background:<br />

Sheeppox, goatpox and lumpy skin disease are the most serious poxvirus diseases of production animals,<br />

and are caused by viruses that comprise the genus Capripoxvirus in the family Poxviridae. Australia’s<br />

livestock export industries would face major restrictions should an outbreak of capripoxvirus disease occur.<br />

Furthermore, our ability to respond to such an outbreak would be seriously compromised by the lack of<br />

modern serological surveillance capabilities for these diseases.<br />

Aim:<br />

To develop an indirect ELISA, based on recombinant antigens, suitable for detection of antibodies to<br />

sheeppox, goatpox and lumpy skin disease viruses.<br />

Methods:<br />

Selected open reading frames were amplified from the genome of a Nigerian sheeppox isolate and<br />

expressed in Escherichia coli as N-terminal His-tagged fusion proteins. Recombinant proteins were<br />

characterised initially by Western blot to confirm the presence of full-length His-tagged constructs and<br />

subsequently by ELISA for reactivity to sera from sheep, goats and cattle experimentally infected with<br />

capripoxvirus or parapoxvirus isolates.<br />

Results:<br />

By screening approximately 40 candidate virus antigens, we have identified two that are readily purified<br />

using affinity chromatography and which react with sequential sera from capripoxvirus-infected animals. Most<br />

sheep and goats vaccinated with an attenuated vaccine isolate did not develop detectable antibodies to<br />

either antigen.<br />

Conclusion:<br />

We have established reliable methods for large scale expression and purification of two recombinant<br />

proteins for use as antigens in an indirect ELISA. Our data using sera from experimentally infected sheep<br />

and goats suggest that these antigens will facilitate capripoxvirus serosurveillance in countries that can not<br />

work with live virus.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NEW ADVANCES IN THE MOLECULAR DIAGNOSIS OF AFRICAN HORSE SICKNESS (AHS)<br />

J Fernández 1* , P Fernández 1 , B Rodríguez 2 , E Sotelo 1 , A Robles 1 , M Arias 1 , JM Sánchez-Vizcaíno 2<br />

1 CISA-INIA, Valdeolmos, Madrid, Spain<br />

2 Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Spain<br />

OIE Reference Laboratory for African Horse Sickness<br />

Introduction<br />

African Horse Sickness (AHS) is a non-contagious artropod-borne infectious disease that afects equidae,<br />

caused by a double-stranded RNA orbivirus. It is a notifiable disease, and it is endemic in many sub-Saharan<br />

countries. Previously, our laboratory developed rapid and sensitive ELISA tests for AHS virus (AHSV)<br />

antigen and antibody detection, as well as a conventional RT-PCR method, that are included in the OIE<br />

Terrestrial Manual 5 . Our objective was the development of new RT-PCR systems for the improved diagnosis<br />

of AHS using novel technologies. A real-time RT-PCR format using a TaqMan-MGB probe, but also a highly<br />

sensitive conventional RT-PCR to assist laboratories with limited resources 2 , have been standardised.<br />

Material and methods<br />

The nine serotypes of AHSV were used in the study. For specificity assays, a collection of spleen<br />

homogenates from horses proceeding from Spanish AHS outbreaks (1987-1990), as well as Bluetongue<br />

Virus (BTV), Vesicular Stomatitis Virus (VSV), West Nile Virus (WNV), Equine Influenza Virus (EIV), and<br />

non-infected Vero and BHK-21 cell lines cultures, were also employed. Total RNA was extracted from<br />

samples using commercial High Pure Viral Nucleic Acid Kit, following manufacturer’s instructions (Roche).<br />

Genome sequences from all viral serotypes were compared. A specific primer set and a TaqMan-MGB probe<br />

were designed from conserved regions of the VP7 viral genome region using the Primer Express TM 2.0<br />

program (Applied Biosystems) to amplify the different viral serotypes, delimiting an amplicon of 102 bp.<br />

One-step RT-PCR kit and QuantiTect Probe RT-PCR kit (Qiagen) were used for AHSV conventional and<br />

real-time amplification assays, respectively. The real-time RT-PCR was adapted for its use both in capillary<br />

(LightCycler, Roche) and 96-well plate (MX3005, Stratagene) thermocyclers.<br />

Results<br />

The analytical sensitivity of conventional and real-time RT-PCR assays was determined using three<br />

replicates of 10-fold dilutions of viral suspensions of AHSV-3, AHSV-4 and AHSV-5 with known titres, grown<br />

in Vero cell line cultures. The detection limit of the real-time method was 0.008 TCID , 0.001 TCID , and<br />

50 50<br />

0.007 TCID for each of the mentioned serotypes. Standard curves were then constructed based on the<br />

50<br />

observed Ct values for each viral dilution, showing a linear relationship of six-seven orders of magnitude for<br />

each serotype. The sensitivity of the developed conventional RT-PCR using the same primer set was also<br />

established applying the same approach, and showed to be 0.8 TCID for AHSV-3, 0.01 TCID for AHSV-4,<br />

50 50<br />

and 0.07 TCID for AHSV-5. In comparative studies, the sensitivity of the real-time and conventional RT-<br />

50<br />

PCR methods showed to be in a range from 10 1<br />

to 10 3<br />

-fold higher than that obtained when performing<br />

conventional RT-PCR using the primer set recommended in OIE Terrestrial Manual. Similar analytical<br />

sensitivity testing were further performed with all the other AHSV serotypes: 1, 2, 6, 7, 8, and 9, obtaining a<br />

detection limit ranging from 0.001 to 0.15 TCID50 per reaction in all of them using the two presented assays.<br />

Specificity of the RT-PCR tests was also evaluated using a collection of spleen homogenates from field<br />

AHSV-infected and non-infected horses, VSV, BTV, WNV, EIV, and non infected Vero and BHK-21 cell lines.<br />

Specific amplified product was observed only in AHSV positive field samples.<br />

Discussion and Conclusions<br />

Several RT-PCR methods have been reported in the past for the AHSV detection, though all of them are gelbased<br />

assays 1,3-7 . This work presents the development and standardisation of two new RT-PCR methods for<br />

the improved AHSV detection to be used in Animal Health Laboratories 2 . One system is a simple and robust<br />

conventional RT-PCR test, that improves the existing diagnostic one described in the OIE Terrestrial<br />

Manual 5 . The other method is a real-time RT-PCR assay, described for the first time for AHS diagnosis. Both<br />

procedures have proved to be rapid, highly sensitive and specific tools for the detection of all the nine AHSV<br />

serotypes. Furthermore, the real-time protocol has shown as a versatile and reproducible assay that can be<br />

used in any format of real-time equipment.<br />

References<br />

1. Díaz-Laviada, M; Sánchez-Vizcaíno, JM; Roy, P; Sobrino, F. (1997) Invest. Agr. SA., 12: 97–102.<br />

2. Fernández, J; Fernández, P; Rodríguez, B; Sotelo, E; Robles, A; Arias, M; Sánchez-Vizcaíno, JM. (2007), submitted.<br />

3. Koekemoer, JJ; Dijk, AA. (2004) J Virol Methods., 122 (1): 49-56.<br />

4. Sailleau, C; Hamblin, C; Paweska, JT; Zientara S. (2000) J Gen Virol., 81 (Pt 3): 831-837.<br />

5. Sánchez-Vizcaíno, J.M. (2004) OIE Terrestrial Manual, chapter 2.1.11.<br />

6. Stone-Marschat, M; Carville, A; Skowronek, A; Laegreid, WW. (1994) J Clin Microbiol., 32: 697-700.<br />

7. Zientara, S; Sailleau, C; Moulay, S; Cruciere, C. (1994) J Virol Methods., 46 (2): 179-188.<br />

This work has been funded by INIA project OT01-002 and EU project SSPE-CT2004-513645.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1745 - 1915 Special Plenary Sessions - Equine Influenza Mon<br />

CHANGING DYNAMICS IN THE GLOBAL DISTRIBUTION OF EQUINE DISEASES<br />

12<br />

P.J. Timoney* Department of Veterinary Science 108 Gluck Equine Research Center Lexington, KY 40546-0099<br />

In an era of ever increasing globalization both with respect to travel and trade, the risk of spread of infectious<br />

diseases of humans and animals has never been greater. Countries historically free of certain diseases of<br />

public health or veterinary importance can no longer consider themselves remote from the risk of incursion of<br />

November<br />

those diseases. The frequency with which diseases are disseminated either within or between countries<br />

continues to escalate. As a consequence, fewer and fewer diseases can be considered geographically<br />

restricted as was formerly the case.<br />

Apart from humans, equids best exemplify the ease with which infectious diseases can be spread through<br />

international movement. Horses are unique not only because of the longevity of the species, considerable<br />

financial value of individual animals, but perhaps most importantly because of the frequency with which they<br />

are shipped between countries for commercial and other reasons. Various industry initiatives over the past<br />

30 to 40 years have contributed very significantly to the evolving nature of international trade in equids and<br />

equine germplasm (semen and embryos), with the inevitable consequence of increasing the risk of global<br />

spread of various diseases. These include continued proliferation in the number of prestigious and lucrative<br />

racing and competition events, dual-hemisphere breeding of stallions, acceptance of artificial insemination by<br />

the vast majority of breed registries, and the growing popularity of embryo transfer in certain horse breeds.<br />

The objectives of this presentation are twofold: firstly, to increase awareness of what has been responsible<br />

for the unprecedented growth in international trade in equids and equine germplasm and the resultant<br />

enhanced risk of dissemination of certain diseases and secondly, to identify the major factors that can<br />

influence the geographic distribution of those diseases.<br />

Movement of equids has been confirmed as the single most important factor responsible for the spread of<br />

equine diseases. This has been borne out by the numerous times specific diseases have been introduced or<br />

re-introduced into countries or regions of the world through the importation of equids either incubating,<br />

acutely infected, or persistently infected with particular pathogens. The countries at greatest risk of such<br />

incursions are those with a significant import trade in equids and equine germplasm. Of the various modes<br />

of disease transmission, spread by the respiratory route is widely accepted as the most rapid and efficient<br />

means of dissemination of a pathogen. Spread of respiratory-borne diseases such as equine influenza,<br />

equine rhinopneumonitis and strangles between countries through international movement has been<br />

extensively documented. Equine influenza is the most important of these and the one that has resulted in<br />

widespread epidemics of disease in naïve or inadequately protected horses. Over the past 40 to 50 years,<br />

equine influenza virus has been responsible for a greater number of epidemics in equine populations<br />

worldwide than any other pathogen, some of which have had a highly significant economic impact on the<br />

affected country or countries. In individual instances, this has totaled many millions of dollars and caused<br />

major disruption, especially of the racing and performance sectors of the industry. Aside from short-term<br />

financial losses, the introduction of certain diseases into a country’s resident equine population can have a<br />

considerable and sometimes long-term effect on international trade. This is exemplified by the restrictions<br />

many countries impose on the importation of equids and equine germplasm from countries known to be<br />

affected with African horse sickness.<br />

Apart from international movement of equids and trade in equine germplasm, multi-national trade<br />

agreements, emergent diseases, mutation of recognized equine pathogens, climate-related phenomena,<br />

migration of amplifying/reservoir hosts or vectors of specific pathogens, availability of new vectors, vaccine<br />

contamination and agroterrorism, have all been shown to affect or have the potential to alter the global<br />

distribution of a wide range of equine infectious diseases.<br />

While historically, countries have responded to the threat of introduction of infectious diseases by formulating<br />

import controls that maximize disease exclusion measures, such overly restrictive policies are no longer<br />

tenable in today’s global economic climate. This is especially pertinent to the equine industry worldwide, the<br />

economic viability and success of which is critically dependant on the ability to ship horses within and<br />

between countries without excessive restrictions on movement. Import policies should be based on the<br />

principal control standards for preventing the international spread of equine diseases specified in the<br />

Terrestrial Animal Health Code of the OIE. In view of the risks inherent in international trade in horses and<br />

semen, countries need to ensure the adequacy of their post-entry disease containment and riskmanagement<br />

control measures.<br />

Disease surveillance and reporting at a national level and the timely exchange of accurate up-to-date<br />

information on specific disease outbreaks at an international level are critical to reducing the risks of global<br />

spread of the more important equine infectious diseases in the future.<br />

Timoney PJ: Factors influencing the international spread of equine diseases. Vet Clin N Am Equine Pract 16:536-551, 2000.<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EQUINE INFLUENZA: LEARNING LESSONS FROM OUTBREAKS<br />

J R Newton, Animal Health Trust, Lanwades Park, Newmarket, Suffolk, United Kingdom, CB8 7UU<br />

Among naïve horses equine influenza is a highly contagious respiratory disease which is characterized by<br />

pyrexia, associated depression and anorexia, harsh dry cough, nasal discharge and secondary bacterial<br />

respiratory infection. A novel H3N8 equine influenza A virus subtype, which first emerged in Miami, Florida in<br />

1963, initiated a worldwide pandemic of equine respiratory disease and was the stimulus for development of<br />

multivalent, adjuvanted influenza vaccines for horses. This early work, based on experience from human<br />

vaccines, led to development of the now broadly standardized schedules for equine influenza vaccination.<br />

These schedules recommend that a primary course of two doses of vaccines be given approximately four to<br />

six weeks apart, followed by a booster vaccination six months after the end of the primary course and annual<br />

boosters thereafter. The same schedules are still adopted today for the product datasheet recommendations<br />

for the latest vaccines and are the basis for the regulatory rules for most international equine competitions.<br />

It was recognized during several influenza outbreaks in the United Kingdom during the 1970’s especially in<br />

the 1979 outbreak, that vaccinated horses generally suffered less severe disease than those that were<br />

unvaccinated. Influenza vaccination of Thoroughbred racehorses in Great Britain became mandatory under<br />

the Jockey Club Rules of Racing at the start of the flat racing season in March 1981 and was implemented<br />

soon after in Ireland and France. Since 1981, British racing has not been cancelled because of equine<br />

influenza but there have been continued seasonal peaks of infection among unvaccinated non-<br />

Thoroughbred horses associated with increased mixing at shows in the summer months. Due to the absence<br />

of systematic, consistent and long term surveillance data it is not possible to provide absolutely conclusive<br />

evidence of the true impact of mandatory influenza vaccination on reducing the incidence of influenza virus<br />

infection and associated disease. However, it is the widely held belief of many that this is indeed the case<br />

and the markedly differing financial and welfare consequences of significant influenza outbreaks in<br />

Thoroughbreds in 2003 in Great Britain and South Africa in which mandatory vaccination was and was not<br />

respectively adopted, serve to highlight the benefits of vaccination in horses at risk of exposure to this highly<br />

contagious disease agent. The outbreaks in Japan and Australia in 2007 perhaps provide a similar contrast.<br />

With changes in equine H3N8 influenza A viruses due to antigenic drift, influenza outbreaks have, however,<br />

caused periodic disruption to the training schedules of vaccinated Thoroughbreds in individual yards or<br />

training centres in the UK, thereby bringing mandatory vaccination under periodic scrutiny. However,<br />

investigations of these largely clinically mild and geographically limited outbreaks have permitted closer<br />

assessment of factors associated with disease occurrence in vaccinated populations. Outbreaks of influenza<br />

virus infection among racehorses vaccinated according to Jockey Club Rules, were investigated in<br />

Newmarket in 1995 and 1998 to establish reasons for vaccine failure. Investigations showed that in 1995<br />

horses with antibody levels above a threshold equivalent to that observed in previous experimental challenge<br />

infections using nebulised aerosol, were protected from infection. Investigations in 1998 demonstrated no<br />

such protective threshold. Subsequent characterization showed that the 1998 H3N8 virus was antigenically<br />

distinct from the 1995 virus and those in available vaccines. These outbreaks highlighted the need for potent<br />

vaccines to maintain protective antibody and inclusion of epidemiologically relevant viral strains in vaccines.<br />

A more recent outbreak in racehorses in the UK in 2003 did, however, serve to confound some of the widely<br />

held beliefs regarding vaccine breakdown in young horses and again highlighted the benefits of detailed<br />

epidemiological and microbiological investigation. Between March and May 2003, equine influenza virus<br />

infection was confirmed as the cause of clinical respiratory disease among both vaccinated and nonvaccinated<br />

horses in at least 12 locations in the UK. In the largest outbreak, at least 21 training yards in<br />

Newmarket, comprising more than 1300 racehorses, were variously affected with horses showing signs of<br />

coughing and nasal discharge during a 9-week period. An American sub-lineage H3N8 equine influenza<br />

virus, previously not identified in the UK, was responsible for the outbreak. On the basis of accepted criteria<br />

there did not appear to be significant antigenic differences between the infecting virus and Newmarket/1/93,<br />

the American lineage virus representative in the most widely used vaccine, to explain the vaccine failure.<br />

Two-year-old horses were apparently less susceptible to infection than three-year-olds and older animals,<br />

despite broadly equivalent antibody levels. However, multivariable analyses comparing infected and noninfected<br />

animals showed that the apparently counter intuitive inverse age effect was explained by elements<br />

in the vaccine history of these animals. Among factors in the vaccine history associated with influenza<br />

infection, there was significantly increased risk of infection associated with male gender, and a period >3<br />

months since the last vaccination. There was a significantly reduced risk of infection associated with an<br />

increasing pre-infection antibody level and being first vaccinated after 6 months of age. There was also<br />

significant variation in risk according to the last type of vaccine administered. Findings highlight the benefits<br />

of relating vaccine history with risk of infection among vaccinated horses suffering influenza infection.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EQUINE INFLUENZA: THE AUSTRALIAN SITUATION<br />

Graeme Garner, Office of the Chief Veterinary Officer, Dept Agriculture, Fisheries and Forestry, Canberra, Australia<br />

Australia is currently experiencing a major outbreak of equine influenza. This presentation will review the<br />

epidemiology of the infection in Australia and the management of the outbreak.<br />

From 17 to 20 August, several horses in Eastern Creek Quarantine Station (ECQS) developed mild fever<br />

and some respiratory symptoms. Results of initial laboratory tests suggested the possibility of EI, which was<br />

supported by polymerase chain reaction (PCR) test results on 23 August. On 22 August, two horses at<br />

Centennial Park Equestrian Centre (CPEC) in the Sydney metropolitan area were noticed to be ill. Samples<br />

were taken and tested positive to EI by PCR on 24 August. The diagnosis of an exotic disease (EI) was<br />

confirmed by Australia’s Consultative Committee on Emergency Animal Disease (CCEAD) on 25 August.<br />

During the ensuing days a number of other horses in the CPEC facility became ill and cases were identified<br />

in a number of locations in NSW and at Warwick in Qld, many of them linked to a two-day horse event held<br />

near Maitland, over the weekend of 18–19 August. Both the Maitland event and CPEC were subsequently<br />

identified as key locations from which EI was disseminated to many sites in NSW and Qld. Although initially<br />

confined to the recreational sector, the infection soon spread to the racing sector with racing precincts in<br />

Sydney and Brisbane affected despite stringent biosecurity protocols being in place.<br />

In response to the finding of EI, Animal health authorities held an emergency meeting on 25 August and a<br />

range of actions were implemented in accordance with AUSVETPLAN, including declaration of Restricted<br />

and Control Areas around the infected premises (IPs), tracing of horse movements and establishment of a<br />

Local Disease Control Centre, State Disease Control Headquarters and a National Disease Control Centre in<br />

Canberra. All horse racing was cancelled in NSW and Qld and a national 72-hour movement standstill on<br />

horses instituted. Governments and industry agreed to implementation of cost sharing under Australia’s<br />

Emergency Animal Disease Response Agreement. An extensive public awareness and communications<br />

program was undertaken and contingency arrangements for the supply and registration of vaccines for<br />

emergency use commenced.<br />

In the next few weeks EI was confirmed in horses at a range of locations in NSW and Qld and there was a<br />

rapid increase in the areas affected. On 17 September, there was national agreement to the use of<br />

vaccination to assist containment and revised NSW and QLD response plans to establish buffer zones<br />

based on natural barriers, horse-free areas and vaccination were endorsed. Subsequently, wider user of<br />

vaccination for mitigation of impact in infected areas and for business risk mitigation in infected and noninfected<br />

areas, was approved. The only vaccine being used is the canary pox recombinant vaccine as it<br />

rapidly induces immunity and enables DIVA methods to be used. It has been recognised that use of other<br />

vaccines will significantly compromise the ability to demonstrate freedom from EI infection.<br />

By 22 October, EI had been reported on more than 6000 premises (4919 in NSW and 1384 in Qld). Despite<br />

a substantial increase in the number of IPs, infection has largely remained contained, with relatively little<br />

increase in the number of clusters of infection and the total area affected, especially since 30 August.<br />

Although the disease has continued to spread most of this has been in-filling of already infected areas,<br />

associated with ‘local spread’. The outbreak has had a major impact on both the racing and recreational<br />

horse sectors.<br />

The national goal has been, and remains, to eliminate the disease. The agreed process is to progressively<br />

eliminate infection within declared areas whilst protecting non-infected areas. A range of strategies are being<br />

used to reduce disease transmission and to alleviate the financial and social cost of the outbreak by<br />

facilitating return to some normal activities. These include:<br />

1. Multiple barriers to spread, including biosecurity measures in infected areas, zoning and vaccination<br />

buffers, movement restrictions between States and awareness/biosecurity in non-infected<br />

jurisdictions.<br />

2. Strategic vaccination in infected areas.<br />

3. Risk-based approach to movements, with progressive freeing up of movements within defined<br />

infected areas, authorisation for the aggregation of horses and development of protocols and<br />

timelines with industry to restore normal business operations.<br />

4. Risk-based movements between areas and jurisdictions, movement between non-infected<br />

jurisdictions, development of protocols for movement between zones in infected jurisdictions and<br />

protocols for movement from infected to non-infected jurisdictions.<br />

5. Strategic vaccination in non-infected areas as a business risk insurance.<br />

Mon 12 November<br />

Mon 12 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIAGNOSIS AND CHARACTERIZATION OF EQUINE INFLUENZA (EI) IN AUSTRALIA:<br />

A/EQUINE/SYDNEY/2888-8/2007 H3N8<br />

Peter Daniels and all Staff, CSIRO Australian Animal Health Laboratory, Geelong, Australia<br />

The index case of equine influenza (EI) in Australia in 2007, at the AQIS Eastern Creek Quarantine Station,<br />

Sydney, New South Wales (NSW), was confirmed on the 22 August, followed quickly by diagnosis of<br />

infected horses at Centennial Park in metropolitan Sydney. The emergency response in NSW to EI<br />

commenced 24 August with declaration of a Control Area which prohibited movement of horses, horse<br />

products, fittings and vehicles except under permit. Shortly after this, the disease was confirmed in southern<br />

Queensland, where it had already been moved in infected horses. Through the rapid application of<br />

movement restrictions, linked to a range of other control measures, the disease has been restricted to NSW<br />

and Queensland. The objective has been containment leading to eradication.<br />

In an outbreak of an exotic disease in Australia the initial diagnosis, isolation and characterisation of the<br />

agent are undertaken by the Australian Animal Health Laboratory (AAHL). In its role as the national<br />

reference laboratory for foreign animal diseases, AAHL has transferred diagnostic tests for some of the main<br />

transboundary animal diseases to the network of State Laboratories around the country. This is particularly<br />

the case with newer technologies such as real time PCR (qPCR). A test for influenza A viruses developed in<br />

support of avian influenza preparedness and targeting the matrix gene has proved particularly useful in the<br />

current outbreak.<br />

Whilst the initial confirmatory diagnosis at the Eastern Creek Quarantine Station was made at AAHL, the<br />

provisional diagnosis at Centennial Park was made at the NSW State Veterinary Laboratories at EMAI, using<br />

the influenza A qPCR test developed at AAHL. Samples were submitted to AAHL for confirmation, but<br />

action in terms of a standstill on horse movement was made on the basis of these initial qPCR results. Much<br />

of the rapid testing following identification of suspect premises has been conducted in the State Laboratories<br />

using the same qPCR assay. Confirmatory testing is done at AAHL, particularly in cases of epidemiologically<br />

significant cases outside the current restricted (infected) area.<br />

At AAHL the qPCR for influenza A is used as a rapid test and is backed up by conventional PCR which<br />

detects a segment of the EI H3N8 haemagglutinin gene. This confirms the H type of the agent and sequence<br />

analysis allows preliminary assessment of the lineage of the strain. Samples are processed for virus isolation<br />

in either embryonated chicken eggs or MDCK cells. Several isolates of the outbreak virus have been made,<br />

from the main epidemiologically significant infected properties in NSW and Qld. Sequence analysis shows it<br />

to be closely aligned with A/Equine/Wisconsin/1/2003.<br />

Serology has been used as an integral part of diagnosis and management of the outbreak. A<br />

haemagglutination inhibition (HI) test is available and detected a seroconversion in the horses at the Eastern<br />

Creek Quarantine Station. A C-ELISA detecting antibodies to influenza A nucleoprotein (NP) had been<br />

validated in Australia for avian influenza antibody detection. AAHL rapidly undertook an exercise to validate<br />

this test for EI. Standard reagents have been prepared for distribution to State laboratories for routine use.<br />

The C-ELISA will allow differentiation of infected from vaccinated animals since the vaccine chosen for use<br />

in the control program is the canarypox vectored vaccine that does not induce antibodies to NP.<br />

Apart from providing confirmatory testing for suspect and new infected premises, a significant effort at AAHL<br />

has gone into generating and analysing data for further test validation, for both the q PCR and the C-ELISA.<br />

These tests were validated for avian influenza and so dossiers for assessment of performance<br />

characteristics of the tests with equine samples have been prepared.<br />

AAHL has also conducted experimental reproduction of the disease using clinical material from Sydney.<br />

Infected animals showed clinical signs two days post exposure, and were sampled to allow further<br />

assessment of agent detection tests such as the PCR tests and virus isolation.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EQUINE INFLUENZA IN AUSTRALIA – A HIGH THROUGHPUT LABORATORY RESPONSE:<br />

ACHIEVEMENTS AND CHALLENGES.<br />

PD Kirkland* 1 ,<br />

1 Virology Laboratory, Elizabeth Macarthur Agricultural Institute, NSW DPI, Menangle, NSW Australia<br />

On 24 August 2007, the Chief Veterinary Officer of New South Wales was advised of clinical signs that were<br />

consistent with influenza in horses in a large equestrian centre. Over the next 2 days, a multi-focal outbreak<br />

was identified on properties scattered over a 700 km range extending from the Sydney region through the<br />

Central Coast and Hunter Valley regions and north along the NSW northern tablelands and slopes into<br />

southern Queensland. The Virology Laboratory at EMAI provided the laboratory support for NSW during this<br />

outbreak with confirmatory testing of selected samples at the Australian Animal Health Laboratory (AAHL) at<br />

Geelong, Victoria.<br />

Nasal swabs collected into viral transport medium and clotted blood were submitted to the laboratory for the<br />

confirmation of influenza virus infection in horses with suggestive clinical signs or from those that had had<br />

possible contact with clinical cases. Samples were also collected from horses during surveillance in buffer<br />

zones at the time of vaccination and for movement testing once the initial standstill/movement restrictions<br />

were eased. Influenza A group reactive assays previously developed at AAHL were used for both the<br />

detection of viral RNA and antibodies to the virus. The viral transport medium was tested in an Influenza A<br />

group reactive real time reverse transcriptase PCR (qPCR) assay to detect a sequence of the matrix gene<br />

while serum samples were tested in an Influenza A group reactive blocking ELISA (bELISA) and in a<br />

haemagglutination inhibition (HI) assay to detect antibodies specific to the H3 subtype.<br />

The qPCR assay had been established as a broadly reactive test for influenza viruses in birds but had not<br />

been applied to testing of equine samples. Similarly, the bELISA had not been validated for testing of horse<br />

sera. Nevertheless, these assays were successfully applied to testing of equine samples with parallel testing<br />

taking place to support validation of the assays. The existing high throughput PCR capability in the EMAI<br />

Virology Laboratory was further refined during this rapidly evolving outbreak in response to variable but<br />

increasing workloads driven by a rapidly changing field situation. The test procedures were streamlined to<br />

minimise repetitive tasks and maximise reproducibility from day to day. The system being employed is based<br />

on a 96 well plate format utilising magnetic bead based total nucleic acid extraction chemistry (MagMAx<br />

Viral, Ambion) in conjunction with a magnetic particle processing system (Kingfisher 96, Thermo). The qPCR<br />

chemistry consisted of a complete master-mix to which the primers and probe had been previously been<br />

added. The only component added at the time of set-up of the assay was the enzyme mix and the sample<br />

RNA. Assays were run on either an ABI 7500 Fast or an ABI 7900HT Fast thermocycler, both run in standard<br />

mode. This combination of RNA extraction and qPCR equipment allowed a throughput of more than 1,000<br />

samples per day and a “turn-around” time of about 3 hours for a batch of samples. Urgent samples could be<br />

completed in about 2 hours. Over the first 2 months of the outbreak the assay system has proven to have<br />

extremely high sensitivity and specificity, combined with ‘same day’ completion of testing.<br />

While sample test procedures were streamlined and a large number of samples could be tested with a<br />

relatively small number of staff, numerous challenges arose in other areas. These rarely involved complex<br />

technical issues and usually were associated with the timely delivery and receipt of specimens. Due to the<br />

diverse nature of the horse industry, there were many small properties and hence individual laboratory<br />

accessions, resulting in an extremely heavy workload and delays in specimen receival and cataloguing.<br />

From time to time, delays in specimen delivery also caused frustration. Similarly, sub-optimally collected or<br />

packaged specimens had a major impact on throughput, sometimes doubling the number of specimens to be<br />

handled and resulting in significant additional processing in the laboratory. Availability of consumables for<br />

specimen collection (eg swabs and sample vials) often caused problems and the high throughput placed the<br />

manufacturers of PCR reagents under pressure on occasions.<br />

While the need for a sustained output placed demands on staff, additional pressure arose when investigating<br />

sites with a high public profile (eg thoroughbred racing stables) due to the intense media interest. At times<br />

there was public monitoring of progress with specimen receival and anticipated times for release of results.<br />

At the scientific level, the application of new assays also raised other questions. In particular, the application<br />

of qPCR and the inevitable detection of residual virus and RNA at times longer than previously described<br />

generated concern when a property was due for release from quarantine. Finally, the detection of infection in<br />

dogs, questions about the role that birds might play and the potential for human infections all added extra<br />

dimensions to providing laboratory support for this outbreak. And, of course, the test needs for the influenza<br />

outbreak have been fully met on a daily basis in a virology laboratory that routinely has a high workload<br />

which has been maintained throughout this outbreak<br />

Mon 12 November<br />

13th International World Association of Veterinary<br />

Laboratory Diagnosticians Syposium<br />

11 - 14 November 2007<br />

Tuesday 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

0830 - 1645 OIE Biotechnology <strong>Symposium</strong><br />

OIE BIOTECHNOLOGY SYMPOSIUM: INTRODUCTORY REMARKS<br />

Steven Edwards, President of the OIE Biological Standards Commission<br />

The OIE (World Organisation for Animal Health) was established in 1924 in order to facilitate an<br />

internationally harmonised approach to animal disease control and reporting, particularly for transboundary<br />

diseases. Since then the organisation has expanded both its remit and its membership to include all<br />

countries with a significant livestock sector (current 170 Member Countries). The OIE is an<br />

intergovernmental organisation in its own right, with responsibility for improving animal health worldwide.<br />

The key missions of the OIE are:<br />

• To ensure transparency on animal diseases and zoonoses<br />

• To collect, analyse and disseminate veterinary scientific information<br />

• To provide expertise and encourage international solidarity on animal disease control<br />

• To publish animal health standards for international trade in animals and their products<br />

(this underpins OIE’s official mandate under the sanitary and phytosanitary agreement of the WTO)<br />

• To improve the legal framework and resources of national veterinary services<br />

• To promote safety for foods of animal origin and to promote science-based animal welfare<br />

As part of its science-based approach the OIE has a long tradition of developing standards for laboratory<br />

procedures used in animal disease diagnosis. The responsible bodies for this activity within OIE are the<br />

Biological Standards Commission (dealing with terrestrial animal diseases) and the Aquatic Animal Health<br />

Standards Commission. Among their other activities, these two commissions advise the OIE on the<br />

designation of Reference Laboratories for specified diseases, and produce laboratory manuals giving details<br />

of recognised and validated diagnostic tests.<br />

These commissions are supported by a variety of working and ad hoc groups, including the Biotechnology<br />

Ad hoc Group. It has been a tradition for some years now for the OIE to sponsor and organise a one day<br />

Biotechnology <strong>Symposium</strong> within the International Symposia of the <strong>WAVLD</strong>. This gives the opportunity for<br />

OIE to bring to the diagnosticians’ community a persective on emerging technologies with a potential for<br />

future application in laboratory-based diagnosis. Some of these are novel developments within established<br />

methodologies, while others present totally new approaches. Some are “near market” in that they are already<br />

or soon will be appearing in the armoury of tests offered by diagnostic laboratories. Others remain<br />

speculative and of uncertain potential, but we certainly should not ignore the developments happening in the<br />

research community.<br />

It is my pleasure to introduce the 2007 OIE Biotechnology <strong>Symposium</strong>, and I hope you find the presenations<br />

stimulating and informative.<br />

Introduction:<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

Simple rapid, on-site detection for diagnosis of animal disease<br />

K.N. Lee, Y. J. Lee, J. W. Kim, J. H. Park, J. G. Choi, Y. J. Kim, 1 J. S. Oh, 2 C. H. Kim, Y. S. Joo<br />

National Veterinary Research and Quarantine Service, Anyang, Korea,<br />

1 Animal Genetics, Inc., Suwon, Korea,<br />

2 Princeton BioMeditech Cooperation, NJ, USA<br />

An immunochromatographic assay is a representative pen-side test relevant to simple, rapid and on-site<br />

detection of etiological agents or antibodies in economically important animal diseases[1]. We developed six<br />

rapid tests using this principle for the diagnosis of foot-and-mouth disease(FMD), avian influenza(AI) and<br />

canine and bovine Brucellosis.<br />

Material & methods:<br />

Two different combinatorial monoclonal antibody pairs, targeting structural protein(SP) and non-structural<br />

protein(NSP) of FMDV respectively, were selected and used as key components for the FMD antigen kit.<br />

Recombinant proteins of 2C and 3ABC of serotype O FMDV were used as captured antigens for detecting<br />

FMDV antibodies in the sera of infected animals. For AI, type A(H1~H16) or subtype(H5) specific monoclonal<br />

antibodies were selected and used respectively for two different pen-side tests to detect viral antigens. For<br />

the serodiagnosis of bovine and canine Brucellosis, the purified antigen containing lipopolysaccharide of B.<br />

canis or B. abortus origin was used as a capture probe for the respective pen-side test.<br />

Results:<br />

The FMD antigen kit could detect viral antigens of four serotype(O, A, Asia 1, C) on SP detection line in<br />

epithelial samples, and the FMD antibody kit showed diagnostic sensitivity comparable to conventional 3ABC<br />

ELISA and could differentiate the infected from the vaccinated. H5 HA antigen was detected specifically with<br />

subtype(H5) specific pen-side kit, while type A specific kit detected all subtypes of type A virus in avian<br />

cloaca swab or feces with the detection limit of 10 5.6 EID50/ 0.1ml approximately. In case of Brucellosis<br />

antibody kits, they showed accuracies comparable to or higher than other conventional serological or<br />

bacteriological tests.<br />

Discussions & conclusions:<br />

The rapid and self-performing immunochromatographic assay has been established for the routine diagnosis<br />

of many viral or bacterial diseases worldwide. These tools have a potential to play important role in rapid<br />

confirmation of infection on infected farms or in the laboratory in case of epidemic. During the recent<br />

outbreaks of FMD or AI by a pandemic strain or a highly pathogenic virus in chicken, the development and<br />

use of these pen-side tests enabled the early diagnosis and prompt stamping out of infected farms, which is<br />

regarded as the key factor in limiting the number of cases.<br />

References :<br />

[1]Reid SM, Ferris NP, Bruning A, Hutchings GH, Kowalska Z, Akerblom L. Development of a rapid<br />

chromatographic strip test for the pen-side detection of foot-and-mouth disease virus antigen. Journal of<br />

virological methods 2001 Aug;96(2):189-202.<br />

Tues 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

APPLICATION OF BIOTECHNOLOGY TO INFECTIONS WITHIN WILDLIFE HOSTS<br />

D J Middleton<br />

CSIRO Australian Animal Health Laboratory, PB24 Geelong 3220<br />

“Wildlife enters the frame”<br />

Wildlife health is complicated and essentially unexplored, while intervention strategies are even less clear.<br />

This is the case even for infectious diseases that affect wildlife hosts although they are of special<br />

significance for diverse reasons. Firstly, their control is necessary for the protection of human health: of the<br />

1415 recognised human pathogens, 61% are zoonotic and wildlife is often a link in the chain of emergence.<br />

Secondly, wildlife pathogens influence domestic animal health and by association the human food supply,<br />

with spillover of known emergency animal diseases providing particular political, conservation and<br />

commercial challenges. Thirdly, they may be the sign of a stressed ecosystem and represent a risk to<br />

biodiversity, especially where keystone species such as a top predator are affected leading, for example, to<br />

a population explosion of herbivores and overexploitation of plants. Ultimately, the casualties of this process<br />

will be loss of ecosystem services such as tourism and “existence value”. Climate change, with the potential<br />

for emergence of new diseases as well as changed wildlife and vector competencies and distribution,<br />

generates additional levels of complexity. Finally, infectious disease control is essential across the broader<br />

national canvas to safeguard trade, the wider economy and for national security.<br />

Deployment of late 20 th C to 21 st C biotechnology is essential for the purpose of enhancing the assessment<br />

and management of infectious disease threats in wildlife. Specific applications include the discovery of<br />

elusive wildlife reservoirs using high sensitivity, high throughput testing systems, such as those employed in<br />

the discovery of fruit bats as the reservoir of Ebola virus. Contemporary biotechnology also has enabled<br />

incidence and prevalence studies, on which epidemiologic data effective disease management relies, to be<br />

carried out in reservoir animals, including surveillance and diagnosis for serious zoonotic diseases such as<br />

Nipah virus infections in Pteropus lylei where Biosafety Level 3 and 4 laboratories are not available. Modern<br />

technologies have contributed greatly to our understanding of the mechanism of host-switching by<br />

pathogens and post-spillover host adaptation, most notably for viral agents such as SARS-CoV from<br />

Rhinolophus spp. to civet cats to people, but also for bacteria including M.bovis in badgers to cattle to<br />

people. They will increasingly be employed in pathogenesis research of intra-reservoir transmission to<br />

provide decision support tools for preventing spillover to humans or livestock through discovery of novel<br />

targets for diagnosis or control. In particular, new whole genome sequencing technologies will lead to rapid<br />

sequencing of novel emerging microorganisms. Optimisation of in silico design of detection tests or<br />

countermeasures will depend upon the availability of accurate genomic surveys of the natural microbiota<br />

within the reservoirs from which they have emerged. Although currently limited, primarily due to lack of<br />

definition of critical intervention points, there are also examples of biotechnology applications to disease<br />

control in wildlife. The most notable of these is the live recombinant oral rabies vaccine targeting wild<br />

carnivores that has been used extensively and effectively in both Europe and the United States.<br />

There is developing acceptance within certain governments of a level of responsibility for the identification<br />

and management of risks from wildlife to human and livestock health as well as on wildlife populations,<br />

although it is less clear who should share ownership of this. Also unclear is the optimum mechanism for<br />

robust collection, amalgamation, and assessment of surveillance data. The notion that detection of an<br />

unusual event may be the first sign of a major epidemic should always be towards the front of our minds. It is<br />

almost certain that modern laboratory methods will play a central role in characterization, assessment and<br />

management of the next such event.<br />

One world, one health, one medicine<br />

Introduction<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE OIE CONCEPT OF TWINNING BETWEEN LABORATORIES<br />

G.K. Brückner, World Organisation for Animal Health (OIE)<br />

The OIE acknowledge that the most effective way to effectively detect, diagnose, control and respond to<br />

animal disease and zoonotic incursions, is to ensure good veterinary governance in Member Countries and<br />

by assisting and enabling them to move towards compliance with the international standards of the OIE.<br />

The OIE has in response to this need secured substantial donor support to embark on a unique strategic<br />

initiative for the assessment and evaluation of the veterinary services of developing and transitional countries<br />

by identifying weaknesses in their system that hinders compliance, prevent the early detection and diagnosis<br />

of diseases and limiting access to expertise to provide scientific justification for certification of animals and<br />

animal products for trade.<br />

A critical prerequisite in achieving this ideal would also be to facilitate and encourage a more even global<br />

geographical spread and access to available scientific expertise and veterinary diagnostics.<br />

Discussion<br />

There are currently 170 OIE Reference Laboratories in 30 countries with 146 designated experts covering 93<br />

diseases. The total number of OIE Collaborating Centres is 24 in 14 countries covering 22 functional aspects<br />

related to diseases and OIE activities. More than 70% of the 170 Member Countries of the OIE are from<br />

developing countries whilst the majority of OIE Reference Laboratories and Collaborating Centres are<br />

clustered within developed countries within the northern hemisphere. During the First International<br />

Conference for OIE Reference Laboratories and Collaborating Centres held in Florianopolis, Brazil in<br />

December 2006, delegates unanimously supported a recommendation to establish closer relationships<br />

between OIE Reference Laboratories and Collaborating Centres and candidate laboratories in developing<br />

and transitional countries. The main thrust would be to encourage a more even global geographical spread<br />

of diagnostic expertise and by that, giving developing and transitional countries more ready access to<br />

scientific expertise enable them to become scientifically competent and to debate on equal footing on the<br />

scientific justification of standards.<br />

To enhance this request for a closer relationship between OIE Reference Laboratories and laboratories such<br />

as national laboratories with a potential of eventually becoming a Reference Laboratory on their own, the<br />

concept of twinning between laboratories was initiated. The main objective of twinning is to assist<br />

laboratories in developing or in-transition countries to build their capacity and scientific expertise with the<br />

eventual aim that some of them could become OIE Reference Laboratories in their own right. To practically<br />

apply this concept, a link between an existing OIE Reference Laboratory with another laboratory in a<br />

developing or in- transition country must be established in a medium to long-term relationship for exchange<br />

of scientific expertise and capacity building. Taking into consideration the current geographical spread and<br />

actual localities of OIE Reference Laboratories and Collaborating Centres, the twinning concept could imply<br />

a transfer of knowledge, training and expertise from the ‘North’ to the ‘South’ or from an existing OIE<br />

Reference Laboratory or Collaborating Centre of the South to another less advanced laboratory in the South<br />

applying for such assistance.<br />

References<br />

1. OIE, Performance, Vision and Strategy: A tool for governance of Veterinary Services, OIE 2007<br />

(World Organisation for Animal Health), 12, rue de Prony, 75017, Paris, France.<br />

2. E. Erlacher-Vindel, G.K. Brückner, B. Vallat. The OIE Concept of Laboratory Twinning in<br />

Lombard M, Dodet B (eds): First International Conference of the OIE Reference Laboratories<br />

and Collaborating Centres. Dev Biol (basel). Basel, Karger, 2007, vol 128, pp 115-119.<br />

Tues 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIAGNOSTICS OF EMERGING INFECTIOUS DISEASES BY MICROARRAY.<br />

R. W. Barrette, S. A. Metwally, and M. T. McIntosh*<br />

Foreign Animal Disease Diagnostic Laboratory, Animal and Plant Health Inspection Services, United States Department of Agriculture,<br />

Plum Island Animal Disease Center, P.O. Box 848, Greenport, NY 11944, USA.<br />

Introduction<br />

Foreign animal and emerging infectious diseases represent threats to public and animal health and present a<br />

significant challenge to the diagnosis of disease. A classical diagnostic approach is to assess<br />

epidemiological findings to limit the differential list of suspect diseases and then run specific diagnostic tests<br />

for those pathogens. Such an approach relies heavily on previously identified disease manifestations and<br />

antigenic or genetic properties of known pathogens. In emerging diseases, pathogens have diverged<br />

significantly from known agents such that they may manifest different pathologies and/or may no longer be<br />

recognized by antigen detection assays, PCR primers, or serological tests. This is likewise true for foreign<br />

animal disease pathogens which may not be recognized by traditional methods used to detect domestic<br />

strains. While such instances are rare, they are quite confounding, and there exists a need for broader<br />

methods of detection in order to identify divergent or emerging pathogens. To aid in such complex animal<br />

disease investigations, we have designed pan-viral DNA microarrays capable of detecting emerging viruses<br />

and foreign animal disease viruses.<br />

Materials & methods<br />

A multi-tiered bioinformatics search of the more than 540,000 viral nucleotide sequences present in<br />

GenBank was used to design a comprehensive pan-viral microarray consisting of approximately 12,000<br />

different virus family, genus or species specific oligonucleotide features. The pan-viral microarray (FADDL<br />

PanVira4) was then commercially synthesized by Combimatrix Co. or Agilent Technologies Inc. A variety of<br />

samples infected with known or unknown viral agents were randomly amplified by RT-PCR, indirectly<br />

labelled with Cy3 and Cy5 dyes and microarrays were hybridized using modifications to previously described<br />

methods (Wang et al., 2002). Microarrays were scanned and results recorded using a GenPix 4200AL<br />

scanner and GenPix Pro software.<br />

Results<br />

Probing of microarrays with randomly amplified cDNA from infected (Cy5 labelled) and uninfected (Cy3<br />

labelled) samples revealed differential hybridization patterns as seen in the simulated scatter plot (Fig. 1).<br />

Using a simple analysis, preliminary identification of the infecting agent could often be achieved by checking<br />

the identity of the features that differentially hybridized to the Cy5 or “infected sample” (Fig. 1 List of Selected<br />

Identities). For example, Figure 1 lists some of the selected<br />

identities of hybridized features from a vesicular case of unknown<br />

etiology used to probe FADDL PanVira1. Using a more<br />

complex analysis that compares the theoretical or expected<br />

hybridization pattern for each candidate viral genome to the<br />

observed hybridization pattern, we were able to accurately<br />

discriminate between closely related species of foot-and-mouth<br />

disease virus using a FMDV array. More than 5,000 viral taxaspecific<br />

domains were identified in the design of FADDL<br />

PanVira4 which consisted of 12,000 different olignoucleotide<br />

features theoretically capable of detecting all animal, human,<br />

avian, and marine viruses with sequence representation<br />

in GenBank.<br />

Cy5 Infected Fluorescence<br />

Selected Identities<br />

Bovine viral diarrhea virus 2<br />

Bovine viral diarrhea virus 2<br />

Bovine viral diarrhea virus 2<br />

Bovine viral diarrhea virus 2<br />

Border disease virus<br />

Bovine viral diarrhea virus<br />

Bovine viral diarrhea virus 2<br />

Border disease virus<br />

Ovine pestivirus<br />

Border disease virus<br />

Bovine viral diarrhea virus 2<br />

Border disease virus<br />

Bovine viral diarrhea virus 1<br />

Cy3 Uninfected Fluorescence<br />

Discussions and conclusions<br />

Genomic approaches to pathogen detection represent a powerful new trend in diagnostics and broadspecificity<br />

techniques such as microarrays have the potential to significantly simplify otherwise complex and<br />

lengthy diagnostic analyses. These methods have the further potential to aid in discovery of previously<br />

unknown pathogens, identify contaminating or endogenous viruses in vaccines and cell lines, and when<br />

applied to a specific genus, can be valuable tools for rapid genotyping.<br />

References<br />

Wang D, Coscoy L, Zylberberg M, Avila PC, Boushey HA, et al. (2002) Microarray-based detection and genotyping of viral pathogens. Proc Natl<br />

Acad Sci USA 99: 15687–15692.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF LOOP-MEDIATED ISOTHERMAL AMPLIFICATION (LAMP) TECHNIQUE FOR<br />

DIAGNOSIS OF AFRICAN TRYPANOSOMOSIS<br />

N. Inoue and O. M. M. Thekisoe<br />

National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-<br />

8555, Japan<br />

Introduction<br />

The low levels of parasitemia usually hamper parasitological diagnosis of trypanosomes in humans or<br />

animals. Although antibody detection tests are useful for screening purposes, they do not distinguish<br />

between past and present infections, and the current reliability of antigen detection tests is limited.<br />

Polymerase Chain Reaction (PCR) has developed as one of the most specific and sensitive molecular<br />

methods for diagnosis of infectious diseases and has been widely applied for detection of pathogenic<br />

microorganisms. However, in spite of the excellent specificity and sensitivity, these molecular biology<br />

techniques are not commonly used in the diagnosis of trypanosomosis in countries lacking resources where<br />

the disease is endemic. This is due to lack of skilled personnel and expensive automated thermal cyclers for<br />

PCR that are not easily available in these countries. Loop-mediated isothermal amplification (LAMP) is a<br />

new DNA amplification method that is performed under isothermal conditions. This unique characteristic of<br />

LAMP allows us to use simple and inexpensive heating device for incubating LAMP reaction mixture. In<br />

addition, LAMP is a rapid and simple technique since it can be carried out within 30 min. Therefore, LAMP<br />

has great potential of being used for diagnosis of trypanosomosis in the laboratory and the field, especially in<br />

countries that lack sufficient resources needed for application of molecular diagnostic techniques. To further<br />

enhance specific trypanosome detection by LAMP, the current study aimed at developing LAMP for<br />

specifically detecting African trypanosome species and sub-species including T. brucei gambiense, T. b.<br />

rhodesiense, T. congolense, and T. evansi.<br />

Material & methods<br />

The LAMP primer sets were designed from 18S rRNA gene for T. congolense, the TgsGP gene for T. b.<br />

gambiense, SRA gene for T. b. rhodesiense, and VSG RoTat1.2 gene for T. evansi. All the primer<br />

sequences were designed using the software program Primer Explorer V4 (Fujitsu, Japan). LAMP reaction<br />

was conducted such that each reaction mixture (25 μl total volume) contained 12.5 μl of the reaction buffer<br />

(40 mM Tris-HCl (pH 8.8), 20 mM KCl, 16 mM MgSO4, 20 mM (NH4)2SO4, 0.2% Tween 20, 1.6 M Betaine,<br />

2.8 mM of each dNTP), 1 μl (8 units) of Bst DNA polymerase, 0.9 μl primer mix with FIP and BIP at 40 pmol<br />

each and F3 and B3 at 5 pmol each), 2 μl of template DNA and 8.6 μl of distilled water.<br />

Results<br />

The LAMP primer sets for T. b. gambiense, T. b. rhodesiense, T. congolense, and T. evansi were tested for<br />

their species specificity, and they showed high specificity. The genomic DNA of T. b. gambiense, T. b.<br />

rhodesiense, T. congolense, and T. evansi was quantified from 100 ng down to 1 fg by serial dilution and<br />

used to assess the sensitivity of the LAMP primers. The primers showed high sensitivity while detecting<br />

trypanosome DNA range from 1 fg to 1 pg. A volume of 10 pg of DNA represents approximately 100<br />

trypanosomes, implying that 1 fg is equivalent to 0.01 trypanosome.<br />

Discussions & conclusions<br />

LAMP primer sets developed in this study are highly sensitive, and are capable of detecting as little as 100 fg<br />

to 1 pg of trypanosomal DNA, which is equivalent to 1 to 10 trypanosomes. Although our LAMP system<br />

should be further improved and standardized for field application, we will discuss possibility and rationale of<br />

LAMP as a field molecular diagnostic technique.<br />

References<br />

Thekisoe, O. M. M. et al. (2007) Acta Tropica, 102: 182-189.<br />

Kuboki, N. et al. (2003) J. Clin. Microbiol., 41: 5517-5524.<br />

Tues 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIAGNOSTIC ELECTRON MICROSCOPY: Historical review and future.<br />

Alex D. Hyatt BSc(Hons), DipEd, PhD.<br />

Senior Principal Research Scientist, Projec Leader.<br />

President Australian Microscopy and Microanalysis Society (AMMS).<br />

CSIRO, Livestock Industries, Australian Animal Health Laboratory, 5 Portarlington Road, Geelong Vic 3220.<br />

Email: alex.hyatt@<strong>csiro</strong>.au<br />

Historical perspective.<br />

Electron microscopy (EM) emerged in the mid 1930s and the ability to magnify images far beyond that of the<br />

light microscope with improved resolution made this instrument popular amongst virologists who were<br />

working in an exciting era of virus discovery and characterization. The ability to observe the actual size and<br />

shape of these filterable and previously invisible agents together with the development of specialized<br />

techniques (e.g. negative contrast staining, the preparation of ultrathin sections from infected tissues and the<br />

use of antibodies) led to the development of new criteria for virus classification (Tyrrell and Almeida, 1967).<br />

Development of specialized techniques. Ultramicrotomy evolved from the histological microtome in the late<br />

1930s. Advances came in the 1950s and from there thermal- and mechanical-advance, in addition to<br />

automated instruments, appeared. Today ultramicrotomes offer reliability in terms of routinely cutting ultrathin<br />

sections (refer to Dykstra and Reuss, 1992). These instruments were used together with classical techniques<br />

such as negative contrast electron microscopy (Brenner and Horne, 1959) to identify, differentiate and<br />

ultrastructurally characterize viruses and their morphogenesis (Biel and Gelderblom, 1999).<br />

Identification of new viruses. During the early years the use of known antibodies were also introduced to<br />

specifically identify infectious agents (e.g. Anderson and Stanely, 1941). The early techniques involved the<br />

forming of antibody mediated clumps, virus decoration and then progressed to the use of ferritin and gold<br />

probes to confirm the presence of antibodies. As per other techniques immuno-electron microscopy has<br />

evolved further to become a critical tool in virus identification, study of virus morphogenesis and<br />

pathogenesis of specific diseases.<br />

Current applications<br />

The classical techniques (refer above) continue to be used in major international diagnostic laboratories.<br />

They are still used to quickly differentiate between putative viruses responsible for diseases where vesicular<br />

fluids are present and to identify new viruses that are emerging into our modern society. Of the latter, in<br />

Australia and South East Asia viruses such as Hendra virus, Nipah virus, bat lyssavirus, Menangle virus,<br />

Pulau virus, Melaka virus, Tioman virus, Pilchard herpesvirus and a range of yet unclassified arboviruses<br />

(e.g. orbiviruses, bunyaviruses and rhabdoviruses) have been identified by transmission electron<br />

microscopy. The above viruses are of varying importance to human, veterinary, aquatic and wildlife health<br />

and arguably until comprehensive viral identification arrays are developed and used routinely and easily (i.e.<br />

user friendly) in major laboratories, EM will continue to be a front-line diagnostic tool.<br />

Future applications<br />

It is apparent that diagnostic electron microscopy, when used in accordance with associated quality<br />

assurance programs, is an important tool in the rapid and accurate identification of viruses, particularly those<br />

which have not been previously identified and associated reagents generated. It is also apparent that skill<br />

bases and academic knowledge in this important area of diagnostics is declining. There is therefore a critical<br />

need to form national and international networks whereby these resources and databases can co-exist in<br />

real time. Activities into forming these networks have begun.<br />

References<br />

Anderson TF and Stanley WM. J. Biol. Chem. 139:339-44.<br />

Tyrrell DAJ and Almeida J, 1967. Arch Gesamte Virusforsch.;22(3):417-25<br />

Dykstra and Reuss, 1992. Biological electron microscopy : theory, techniques, and troubleshooting (Plenum Press)<br />

Brenner S and Horne RW. 1959. Biochim Biophys Acta. Jul;34:103-10.<br />

Biel SS and Gelderblom HR. 1999. J Clin Virol. Jun;13(1-2):105-19. Review.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

OVERVIEW OF ELECTRON MICROSCOPY AND ITS ROLE IN INFECTIOUS DISEASE DIAGNOSIS<br />

Norbert Bannert, Centre for Biological Security 4, Robert Koch Institute, Berlin, Germany<br />

Michael Laue, Centre for Biological Security 4, Robert Koch Institute, Berlin, Germany<br />

Introduction<br />

Electron microscopy (EM) is well suited to directly visualize virus particles at high magnification. The first<br />

images showing the size and morphology of viruses were published in the late 1930s (von Borries et al.<br />

1938). In the following decades EM was used to characterize a variety of viruses at the ultrastructural level<br />

and for routine diagnosis of human and non-human viruses. The introduction of the negative staining method<br />

in 1959 increased the diagnostic speed and throughput of the application. Today routine diagnostic EM is<br />

predominantly used in cases of suspected skin associated viral infections and viral gastroenteritis. The open<br />

view and the fact that no sequence or protein information are require for a diagnosis, makes the method<br />

indispensable in emergent situation where the etiologic agent is difficult to identify with standard methods or<br />

a new infectious agent appears. EM has also been adopted in many countries as a front line method in the<br />

examination of samples from suspected bioterroristic (BT) attacks with relevant viral and bacterial<br />

pathogens. With improved negative staining protocols and rapid embedding techniques, the impact of EM in<br />

the diagnostic arsenal increased significantly. Moreover, EM is still an attractive tool in viral research. In our<br />

laboratory it is applied for the characterization of ancient human endogenous retroviral particles (HERV) and<br />

the recently identified Koala Retrovirus (KoRV).<br />

Material and Methods<br />

Before regular negative staining EM, all samples are inactivated with 2% paraformaldehyde. BT-suspected<br />

samples are treated with 10% paraformaldehyde, 0.05% glutaraldehyde in 0.05 M Hepes buffer (pH 7.2) for<br />

2 hours in a BSL3 laboratory, to inactivate potentially present bacterial spores. Hydrophilic carbon reinforced<br />

support grids are placed on top of a drop with the agent containing suspension to allow adhesion of particles.<br />

Following short washing steps in deionised water, the particles on the grid are stained with uranyl acetate or<br />

related solutions of heavy metal salts. Following this simple contrasting method, the grids can be inspected<br />

in the EM. For rapid embedding, a microwave-assisted chemical fixation followed by LR White embedding<br />

has been developed (Laue et al. 2007). Cell lines producing retroviral particles are processed using<br />

conventional fixation and epon embedding protocols.<br />

Results<br />

EM can be used for rapid identification of causative agents of infectious diseases in humans and animals.<br />

Although the routine screening application of EM decreased in recent years in many laboratories, its value<br />

for the inspection of new or emerging zoonotic diseases did increase. This was very well demonstrated in<br />

critical diagnostic situations including the SARS epidemics and cases of monkeypox virus in the USA when<br />

diagnostic EM gave the first indications of the causative virus (Ksiasek et al. 2003).<br />

Regarding the diagnosis of bacterial spores and other pathogens in BT-suspected samples, the development<br />

and introduction of rapid embedding techniques and immuno EM complement the quick and simple negative<br />

staining techniques. It increases the reliability and specificity of the diagnosis by providing ultrastructural<br />

information and allows differentiation of morphologically identical pathogens with specific antibodies for<br />

internal antigens.<br />

Besides diagnostic applications, electron microscopy is indispensable in ongoing own research projects<br />

including those investigating aspects of the recently described endogenous Koala Retrovirus (KoRV) and<br />

related retroviruses. KoRV has been identified by EM in specimens from Koala bears suffering from<br />

lymphomas and leukaemia. Analogous to human endogenous Retroviruses it is suspected to play a role in<br />

the induction or progression of malignancies. Recently we have shown that it is able to infect human cells.<br />

Discussion<br />

Based on its inherent features, including the simple and rapid preparation (less than 15 min), the direct “open<br />

view” allowing the visualisation and potential identification of all viruses and bacteria in the clinical or<br />

environmental specimen, diagnostics negative stain EM has its advantages over routinely used PCR and<br />

antibody techniques in clinically or epidemiologically critical situations. Together with rapid embedding<br />

protocols it should be considered as a front line method in such cases.<br />

References<br />

1. von Borries et al. Klin. Wochenschr. 17 (1938)<br />

2. Ksiasek et al. New Engl. J. of Med. 348 (2003)<br />

3. Laue et al. J. of Microbiol. Methods 70 (2007)<br />

Tues 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

TELEPATHOLOGY: ITS ROLE IN DISEASE DIAGNOSIS IN MEAT HYGIENE.<br />

Y. Robinson<br />

Saint-Hyacinthe Laboratory, Canadian Food Inspection Agency<br />

Saint-Hyacinthe, Québec, Canada<br />

Introduction<br />

Telepathology is defined as a branch of telemedicine and pathology based on the exchange of patient’s data<br />

and images through means of telecommunications for diagnosis, consultation, education and research (1).<br />

Static telepatholoy is of low cost and simple method with capture of digital images and their electronic<br />

transmission (2). It allows quick and timely access to an expert opinion at a distance.<br />

The first publication of this new field appeared in 1986 (3) and until today a variety of modifications has<br />

developed, including those with a simple offline transmission of images for second opinion or educational<br />

purposes. The availability of digital cameras at reasonable prices created new training and teaching tools for<br />

meat inspection veterinarians and plant employees.<br />

Material and methods<br />

This is a study of static telepathology in which gross pathology images were taken in 10 federally inspected<br />

slaughter houses in Canada, sent electronically to the pathologist along with a case history. In the majority of<br />

cases formalin fixed tissues were submitted to the laboratory for histopathology. Three hundred seventeen<br />

cases in a period of 4 years (August 2003 – July 2007) were evaluated with this system. All images were<br />

submitted through the Internet to the pathologist using a Canon PowerShot A80, 4.0 Megapixels with a 3x<br />

Optical Zoom Lens. For some cases in this study, representative microscopic hematoxilin-phloxine-safran<br />

(HPS) digital images along with other special technique images such as special staining were sent to 1 or 2<br />

pathologists for a second opinion. Images for histopathology were taken using a Leica DM4000 B<br />

microscope with HC Fluotar lenses and a Leica 490 digital camera.<br />

Results<br />

The number of images per case ranged from 3 to 19. The optimal time for that type of consultation was<br />

usually within one hour. Such fastest diagnosis insured an early initiation of specific processes regarding the<br />

disposition of carcasses or a more extensive investigation on the case. The use of telemicroscopy facilitated<br />

a more accurate diagnosis in some cases. Images linked to explanatory texts were added to pathology<br />

reports.The images from six cases are presented at this conference.<br />

Discussions and conclusions<br />

The long-distance transmission of digitised images was used in a pilot project in 10 Canadian<br />

slaughterhouses to evaluate the feasibility of creating rapid communication pathways with an expert<br />

pathologist. Our experience, based on over 300 cases, suggests that static telepathology is useful in<br />

simplifying and expanding access to specific remote pathology expertise, rapid intervention as well as<br />

improving continuous education in pathology. . Pathology reports which featured images linked to<br />

explanatory text had greater clarity, greater teaching value, and held more useful information than traditional<br />

text only reports. Another application for the teletransmission is the generation of a database of<br />

reference material specifically for teaching purposes.<br />

References<br />

1. La Rosa F (2000). TelePathology Consultants. 6 th Internet World Congress for Biomedical Sciences<br />

2. Weinstein RS (1996). Static image telepathology in perspective. Hum Pathol 27:99-101.<br />

3. Weinstein RS (1986). Prospects for telepathology. Hum Pathol 17:433-434.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

UNDERSTANDING HOST-PATHOGEN INTERACTIONS: CONFOCAL AND LIVE CELL IMAGING IN<br />

VETERINARY SCIENCE<br />

P. Monaghan, P. Hawes, J. Simpson, E. Brooks, A. Banyard, T. Jackson<br />

Institute for Animal Health, Ash Road, Pirbright, Surrey, GU24 0NF, UK<br />

Introduction<br />

At the Institute for Animal Health we are using multiple microscopy approaches to study host-pathogen<br />

interactions. These include receptor binding, internalisation and intracellular replication. The combination of<br />

molecular techniques including in vitro transfection, fluorescent proteins and siRNA with confocal and livecell<br />

microscopy is proving particularly effective in these studies. Pathogens under investigation include<br />

parasites, such as Eimeria tenella, and bacterial and virus pathogens of farmed animals. Examples from<br />

these studies will be used to illustrate how these techniques give new insights into the cell biology of these<br />

economically important pathogens.<br />

Materials and methods<br />

Samples of cells and tissues were prepared for imaging by a variety of techniques including confocal, livecell<br />

and electron microscopy.<br />

Results and Discussion<br />

We have improved our understanding of several stages in the replication cycle of Foot-and-Mouth Disease<br />

Virus (FMDV). In vitro evidence suggests that the virus can use a number of receptors for binding and<br />

internalisation, but that in vivo, the virus uses the integrin αvβ6. We have mapped for the first time, the<br />

expression of the integrin αvβ6 in epithelia from susceptible animals and confirmed that it is expressed in<br />

epithelial tissues known to be infected by the virus (Monaghan et al, 2005a). Further verification was gained<br />

from localisation of viral proteins in tissues from infected animals by confocal microscopy and this data was<br />

supported by quantitative information on viral RNA copy numbers by RT-PCR (Monaghan et al, 2005b).<br />

Using a tissue culture model, we have shown that the virus uses this integrin receptor and gains entry to the<br />

cell via clathrin-mediated endocytosis (Berryman et al, 2005).<br />

Many viruses interact with the cytoskeleton (eg Jouvenet et al 2006) and we have highlighted the role of the<br />

cytoskeleton in the cytopathic effect induced by FMDV infection. We have investigated this interaction by<br />

transfecting individual FMDV non-structural proteins into uninfected cells and identified the viral nonstructural<br />

protein responsible. FMDV is a member of the picornaviridae – a group of viruses which includes<br />

poliovirus (PV) and whilst all viruses of this group require cytoplasmic membranes on which to replicate, the<br />

source of these membranes is not clear. We have investigated the possible involvement of cytoplasmic<br />

components including ER, Golgi or lysosomes as well a possible role for autophagy, in FMDV infection.<br />

Autophagy is a normal cellular process initiated in response to nutrient deprivation and recent reports of the<br />

replication strategy of PV (Suhy et al, 2000 ) indicate that this virus may use the autophagy pathway as an<br />

aid to replication and cell exit.<br />

Incorporating GFP into a pathogen genome enables the replication of the virus to be studied in real-time<br />

using live-cell imaging giving new insights into the dynamics of infection. A GFP-Rinderpest virus is currently<br />

under development for both in vitro and in vivo experiments.<br />

These microscopical techniques are not only improving our understanding of the complex cell biological<br />

interactions between host and pathogen at the cellular and sub-cellular level but will also open new avenues<br />

for the development of novel control measures.<br />

References<br />

Berryman S, Clark S, Monaghan P, Jackson T. The early events in αvβ6-mediated cell entry of foot-and- mouth disease virus. J Virol<br />

79: 8519-8534 2005<br />

Jouvenet N, Windsor M, Rietdorf J, Hawes P, Monaghan P, Way M, Wileman T. African swine fever virus induces filopodia-like<br />

projections at the plasma membrane. Cell Microbiol 8:1803-1811 2006<br />

Monaghan P, Gold S, Simpson J, Zhang Z, Alexandersen S, Jackson T. The αvβ6 integrin receptor for foot-and-mouth disease virus is<br />

constitutively expressed on the epithelial cells targeted in a natural infection of cattle. J Gen Virol 86: 2769-2780 2005a<br />

Monaghan P, Simpson J, Murphy C, Durand S, Quan M Alexandersen S Localisation of Viral Non-Structural Proteins and Potential<br />

Sites of Replication by Confocal Immuno-Fluorescence Microscopy in Pigs Experimentally Infected with Foot-and-Mouth Disease Virus.<br />

J Virol. 79: 6410-6418 2005b<br />

Suhy DA, Giddings TH Jr, Kirkegaard K. Remodeling the endoplasmic reticulum by poliovirus infection and by individual viral proteins:<br />

an autophagy-like origin for virus-induced vesicles.<br />

J Virol. 74:8953-8965. 2000<br />

Tues 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MULTI-RESOLUTION SPATIO-TEMPORAL ANALYSIS OF MAMMALIAN CELLS RECONSTRUCTED IN<br />

3D BY ELECTRON MICROSCOPE TOMOGRAPHY<br />

Brad J Marsh W Group Leader/Senior Research Fellow, Institute for Molecular Bioscience, Centre for Microscopy & Microanalysis and<br />

School of Molecular & Microbial Sciences, The University of Queensland, St Lucia, QLD 4072, AUSTRALIA<br />

SUMMARY<br />

The beta cell - the sole source of the hormone insulin in mammals - resides within the islets of Langerhans in<br />

the pancreas. We are focused on understanding the basic mechanisms that underpin normal beta cell<br />

function, so that we can elucidate the steps that lead to beta cell/islet dysfunction and ultimately, diabetes.<br />

To this end, we combine fast-freezing techniques with electron microscope tomography (ET) to conduct<br />

comparative structure-function studies of pancreatic islets isolated from mice and humans. To complement<br />

insights from these high-resolution 3D reconstructions (“tomograms”) of parts of cells, and to move toward a<br />

more integrated or “holistic” approach to understanding the mammalian cell as a unitary example of an<br />

ordered complex system, we have undertaken a multi-scale/multi-resolution approach to reconstructing<br />

mammalian (beta) cells in toto in 3D by ET at both high (≤5nm) and intermediate (15-20nm) resolutions. By<br />

providing complete sets of 3D spatio-temporal coordinates for cells at a range of resolutions that will uniquely<br />

inform advanced in silico studies of 3D cell and molecular organization, we are working to develop the<br />

world's first navigable ‘Visible Cell atlas’ within the broader framework of the Visible Cell project<br />

(http://www.visiblecell.com/). Such an interactive high-resolution map of the 3D landscape of an entire<br />

mammalian cell imaged and reconstructed by ET at the EM level will serve as a unique international<br />

resource for protein and organelle annotation, database integration and 3D visualization, and as a framework<br />

for 4D animations and computational simulations of cells at pseudo-molecular resolution.<br />

KEY REFERENCES (IN CHRONOLOGICAL ORDER)<br />

2001 Marsh BJ, Mastronarde DN, Buttle KF, Howell KE, McIntosh JR. Organellar relationships in the Golgi<br />

region of the pancreatic beta cell line, HIT-T15, visualized by high resolution electron tomography. Proc<br />

Natl Acad Sci USA. 98:2399-2406<br />

2001 Marsh BJ, Mastronarde DN, McIntosh JR, Howell KE. Structural evidence for multiple transport<br />

mechanisms through the Golgi complex in the pancreatic beta cell line, HIT-T15. Biochem Soc Trans.<br />

29:461-467<br />

2002 Marsh BJ, Howell KE. Timeline: The mammalian Golgi - complex debates. Nat Rev Mol Cell Biol.<br />

3:789-795<br />

2004 Marsh BJ, Volkmann N, McIntosh JR, Howell KE. Direct continuities between cisternae at different<br />

levels of the Golgi complex in glucose-stimulated mouse islet beta cells. Proc Natl Acad Sci USA.<br />

101:5565-5570<br />

2005 Marsh BJ. Lessons from tomographic studies of the mammalian Golgi. Biochimica et Biophysica<br />

Acta. 1744:273-29<br />

2006 Marsh BJ. Toward a “Visible Cell”…and beyond. Australian Biochemist. 37:5-10<br />

2007 LR Brunham, JK Kruit, TD Pape, JM Timmins, AQ Reuwer, Z Vasanji, BJ Marsh, B Rodrigues, JD<br />

Johnson, JS Parks, CB Verchere and MR Hayden. Beta-cell ABCA1 influences insulin secretion, glucose<br />

homeostasis and response to thiazolidinedione treatment. Nature Medicine. 13:340-347<br />

2007 P van der Heide, X Xu, BJ Marsh, D Hanein and N Volkmann. Efficient automatic noise reduction of<br />

electron tomographic reconstructions based on iterative median filtering. J Struct Biol. 158:196-204<br />

2007 BJ Marsh, C Soden, C Alarcón, BL Wicksteed, K Yaekura, AJ Costin, GP Morgan and CJ Rhodes.<br />

Regulated autophagy controls hormone content in secretory-deficient pancreatic endocrine beta-cells. Mol<br />

Endocrinol. 21:2255-2269<br />

2007 BJ Marsh. Reconstructing mammalian membrane architecture by large area cellular tomography.<br />

Methods in Cell Biology. 79C:193-220<br />

2007 AB Noske, AJ Costin, GP Morgan and BJ Marsh. Expedited approaches to whole cell electron<br />

tomography and organelle mark-up in situ in high-pressure frozen pancreatic islets. J Struct Biol. In press<br />

ACKNOWLEDGEMENTS<br />

This work has been supported by grants from the Juvenile Diabetes Research Foundation International (2-<br />

2004-275) and the National Institutes of Health (DK-71236) in the USA. BJM is a Senior Research Affiliate<br />

of the ARC Special Research Centre for Functional and Applied Genomics, and is a Chief Investigator of the<br />

ARC Centre of Excellence in Bioinformatics.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DETECTING AND INTERPRETING ARBOVIRAL INFECTIONS IN INSECTS.<br />

Dr Simon Carpenter<br />

Arbovirology programme, Institute for Animal Health, Pirbright Laboratory, Ash Road, Woking, Surrey. GU24 0BN. UK.<br />

simon.carpenter@bbsrc.ac.uk<br />

Vector-borne arboviruses have a significant detrimental impact on both human health and animal health and<br />

production world wide. A vital key to understanding the epidemiology of the diseases caused by these<br />

viruses lies in the correct identification of the insect vectors responsible for their spread. While this appears<br />

at first sight to be a relatively simple task, the complexities of virus-vector interactions commonly lead to<br />

results that are difficult to interpret. In this talk I will examine the ways in which vectors have traditionally<br />

been implicated, including studies of their ecology, the presence of virus in field collected samples, PCR<br />

positives and laboratory-based infection trials.<br />

I will begin by distinguishing between different modes of arboviral transmission and their implications for<br />

studying the epidemiology of each particular virus. I will then examine the process by which vectors become<br />

infected when taking a blood-meal from a viraemic host. This will include a discussion of the various barriers<br />

that may inhibit dissemination of the virus through the body of the insect vector, with examples drawn from<br />

work carried out upon biting flies. I will then briefly describe the influence of environmental factors upon this<br />

process and examine the evidence for likely impacts with regard to climate change.<br />

The commonly used techniques of arbovirus detection in insects will then be described in detail and their<br />

advantages and disadvantages examined. I will critically explore a case study of ongoing bluetongue virus<br />

outbreaks in Europe, (spread via Culicoides midges, which are true biological vectors), to illustrate how<br />

these techniques are commonly implemented in practice. This will include a discussion of where vector<br />

diagnostics can be integrated into other diagnostic methods to monitor arboviral circulation, and suggestions<br />

as to how these can be easily expanded into standard laboratory set-ups.<br />

Finally, I will discuss the application of novel and high-throughput diagnostic techniques to virus-vector<br />

systems and some of the methodological difficulties that need to be addressed to ensure their correct<br />

application. I will discuss the problems intrinsic in attempting to combine entomological and molecular<br />

expertise emphasising that input is required from all parties to facilitate the incorporation of new technologies<br />

into mainstream diagnostics.<br />

Tues 13 November


Tues 13 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MOLECULAR METHODS FOR SPECIATION<br />

Dr Hez Hird, Senior Molecular Biologist<br />

Biotechnology and Molecular Genetics<br />

Central Science Laboratory<br />

Sand Hutton<br />

York<br />

YO41 1LZ<br />

United Kingdom<br />

E-mail h.hird@csl.gov.uk<br />

The advent of the polymerase chain reaction (PCR) has revolutionised the diagnosis of diseases over the<br />

last 2 decades. The challenge though has been to develop practical routine methods, which could be used<br />

in diagnostic laboratories, where reliable and simple amplicon detection would allow the objective analysis of<br />

results. The main breakthrough in achieving this aim came at the end of the 1990s with the development of<br />

real-time PCR using TaqMan technology. In TaqMan PCR, the exponential amplification of target specific<br />

DNA is measured by the use of an oligonucleotide probe complementary to sequence internal to the PCR<br />

primers and labelled at each end with fluorescence reporter and quencher molecules. When the probe is<br />

intact, the reporter dye fluorescence is absorbed by the quencher molecule by fluorescence resonance<br />

energy transfer (FRET). During amplification, the 5’ nuclease activity of Taq polymerase causes the probe to<br />

be cleaved and a signal is generated from the free fluorescence reporter dye. The increase in fluorescence<br />

is related to the amount of template amplified and can be measured using a fluorimeter. TaqMan PCR offers<br />

a number of advantages over conventional PCR, including closed tube assay format with no need for post-<br />

PCR manipulations, increased sensitivity, capacity for high throughput and the objective analysis of results.<br />

This presentation will focus on the application of real-time PCR based methods for the detection and<br />

identification of zoonotic diseases and will be illustrated using Mycobacterium bovis and Bacillus anthracis.<br />

M. bovis is a problem in the UK where eradication of the disease from the national cattle heard has been<br />

unsuccessful. This is probably due to the presence of M. bovis in wildlife reservoirs, primarily in the badger,<br />

which is highly susceptible to the disease. One strategy to eradicate the disease is the vaccination of both<br />

badgers and cattle using M. bovis BCG, however differentiation between M. bovis shed by infected animals<br />

and M. bovis BCG shed by vaccinated animals is impossible using current PCR based assays. Ongoing<br />

work at the Central Science Laboratory, to develop a rapid, robust real-time PCR based assay, to<br />

differentiate M. bovis and M. bovis BCG, will be discussed.<br />

Bacillus anthracis is an example of a zoonotic disease which causes the rapid death of humans after<br />

inhalation exposure and as such, is a possible biological warfare agent. This powerful driver has led to the<br />

development of real-time PCR assays for the rapid identification of B. anthracis in the field, and which has<br />

necessitated the development of robust portable real-time PCR equipment. There are a number of portable<br />

real-time PCR machines available, however the Cephaid Smart cycler appears to be currently the most<br />

popular for non-military uses. The Smart cycler has sixteen cycling modules which can be independently<br />

programmed, via a controlling computer, to perform four colour, real-time fluorometric detection. The system<br />

software enables one or more operators to define and simultaneously carry out separate experiments, each<br />

with a unique set of cycling protocols, threshold criteria and data analysis. The data can be viewed in realtime<br />

and when used with assays optimised to return results in 30-40 minutes, the time to positive<br />

identification for a sample can be kept to a minimum. An additional advantage of the Smart cycler over the<br />

laboratory based real-time machines is the much reduced price: making real-time PCR more accessible.<br />

The application of field based portable real-time PCR methods for disease diagnosis will also be discussed.<br />

In summary, this presentation will demonstrate the use of real-time PCR for the identification of zoonotic<br />

diseases, which for the first time offers the opportunity for field-based PCR testing.<br />

13th International World Association of Veterinary<br />

Laboratory Diagnosticians Syposium<br />

11 - 14 November 2007<br />

Wednesday 14 November


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

0815 - 1000 Plenary Sessions - Preparing for the Future<br />

LABS OF THE FUTURE<br />

Dr. Martyn Jeggo<br />

Director, Australian Animal Health Laboratory, Geelong, Victoria,<br />

Australia<br />

Today, veterinary laboratories are faced with a range of challenges that constantly threaten their resource<br />

base, yet globally the need for veterinary diagnostic services and the underpinning research activities has<br />

never been greater. The occurrence and risks from infectious diseases has steadily been increasing during<br />

the past 20 years. Whilst many endemic production diseases have been on the wane, infectious disease e.g.<br />

avian influenza, foot and mouth disease, West Nile, bluetongue continues to threaten livestock and in a<br />

growing number of cases, man.<br />

How best can veterinary laboratories utilize current and new technologies to provide an effective diagnostic<br />

service and do so in a growing regulatory environment that makes the delivery of these services evermore<br />

complex and resource hungry?<br />

During the past few years, the development of molecular technologies based around the polymerase chain<br />

reaction (PCR) offer a quantum leap in diagnostic levels of sensitivity and specificity, in a timely and<br />

increasingly, quality assured manner. But where will this take us in the next ten years and can we genuinely<br />

equate the presence of genetic material with that of an infectious agent? Can the laboratory of the future<br />

dispense with causative agent isolation and characterization and rely on these newer gene based<br />

approaches? On the one hand, the promises of multiplex assays for multiple agent detection in a single test<br />

provides an opportunity to check for an enormously wide range of potential pathogens, whilst at the other<br />

end of the spectrum, the development of pen-side assays offers the opportunity to dispense with the need to<br />

even submit samples to a laboratory. How will these contrasting approaches impinge on activities<br />

undertaken by a veterinary laboratory in ten year time?<br />

In considering the regulatory and operating environment that laboratories now find themselves, a wide range<br />

of health, safety and environmental priorities must be taken into account for every activity undertaken.<br />

Invariably processes involving gene based technologies are now carefully regulated and activities involving<br />

the use of animals overseen by welfare and ethic committees. But beyond this, a new biosecurity regulatory<br />

environment is being created for anyone working with a range of pathogens, and increasingly this deals not<br />

only with biosafety and bio-containment but the potential element of bioterrorism. This increasing regulation<br />

of the operational environment of a veterinary laboratory must lead to changes in the way they operate, but<br />

what is the likely outcome?<br />

The biggest challenge though, maybe in the area of bio-informatics, information exchange and the decision<br />

making processes that this will underpin. The technologies embraced today have an ability to generate large<br />

amounts of data, that need to be collated, processed and re-organized to assist the veterinary diagnostician<br />

and the disease management services he supports. What will be the impact of the continuing information<br />

technology explosion on the activities of the veterinary laboratory in the future?<br />

This paper attempts to answer the questions posed above, taking examples from a number of recent disease<br />

outbreaks both in Australia and elsewhere, illustrating trends taking place today, that are likely to shape the<br />

veterinary laboratory of the future.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

Biosafety and Biosecurity Concerns Of High Security Animal Pathogen Laboratories<br />

Luis L. Rodriguez, D.V.M., Ph.D.<br />

Overview:<br />

In response to the global spread of emerging infectious diseases and the threat of bioterrorism, highcontainment<br />

biosafety laboratories (BSL)--specifically biosafety level (BSL)-3 and BSL-4--have been<br />

proliferating in the United States as well as throughout the world. Specific incidents such as the anthrax<br />

terrorist attacks in the USA in 2001; the emergence of SARS and the infection of humans with H5N1 Avian<br />

influenza in Asia generated great concern and interest in around the world and in particular in the USA. A<br />

new industry; biodefense has emerged with academia, government and industry adapting and responding to<br />

this new niche for biodefense-related research, diagnostics, laboratory equipment, high-containment<br />

construction and pharmaceuticals. This multi-billion dollar industry ($5.24 billion in 2007 in the US alone)<br />

places great demand for BL3 and BL4 labs in government, academia and private sector.<br />

High-consequence human and animal pathogens, which traditionally have been studied in Public Health or<br />

Agriculture government laboratories, have become potential targets for bioterrorists. For example, foreign<br />

animal diseases such as Foot-and-Mouth Disease were determined to be high threats for US agriculture and<br />

food security. This increased concern for protecting an infrastructure critical to national security resulted in<br />

the transfer of the Administration and Operations of the Plum Island Animal Disease Center facility from the<br />

US Department of Agriculture (USDA) to the newly created Department of Homeland Security (DHS) in<br />

2003, while maintaining the research and diagnostics mission in USDA. Laws were revised and or created<br />

in 2002 to ensure the proper handling, storage and tracking of high consequence agents for both human and<br />

animal health.<br />

Although investment in biodefense research has been significant over the last 5 years, particularly in the<br />

area of human health, some of the critical infrastructure in animal health has lagged behind and only recently<br />

have plans been made to replace or renovate high-containment laboratories such as the Pirbright laboratory<br />

in the UK and the Plum Island laboratory in the US. These aging facilities have come under close scrutiny<br />

after a recent FMD outbreak was linked to the Pirbright site. This incident as well as recent incidents in US<br />

labs handling select agents motivated ongoing investigations at the congressional level. Similar inquiries are<br />

likely to follow in the UK as well as other countries with high-security laboratories.<br />

This lecture will present an overview of the current biosafety and biosecurity concerns, including the real as<br />

well as perceived risks from the construction and operation of high-security animal disease laboratories.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIAGNOSTIC TECHNOLOGIES FOR VETERINARY LABORATORIES OF THE FUTURE<br />

T. R. Beckham<br />

Foreign Animal Disease Diagnostic Laboratory, Animal and Plant Health Inspection Services, United States Department of Agriculture,<br />

Plum Island Animal Disease Center, P.O. Box 848, Greenport, NY 11944, USA.<br />

Veterinary diagnostic laboratories of the future will be challenged to provide rapid, accurate diagnostics that<br />

are capable of detecting and identifying emerging and re-emerging diseases. There will be an emphasis on<br />

rapid identification and subsequent characterization of high consequence agricultural and/or zoonotic agents.<br />

Over the past two decades, there has been a substantial growth in molecular diagnostic capabilities within<br />

these laboratories. More specifically, conventional PCR, real time PCR and sequence analysis have<br />

become routine tools used to detect and characterize isolates from diseased animals. Although these tools<br />

are capable of providing a rapid and accurate diagnosis, each has their limitations. First, the conventional<br />

and real time PCR assays are typically designed to detect a specific agent and therefore, their design<br />

requires a readily available source of sequence information. In addition, multiplexing capabilities (the<br />

simultaneous testing of multiple agents) of the real time based PCR method is limited.<br />

Recently, other more sophisticated technologies have begun to be developed. These new technologies<br />

(Luminex bead based assays, Triangulation Genetic Evaluation of Risk (TIGER, Ibis Therapeutics), and<br />

microarrays) offer additional capabilities that will enhance surveillance programs and the ability to detect and<br />

identify an unknown or emerging pathogen.<br />

The Luminex xMap technology is a bead-based system that allows for deep multiplexing of diagnostic<br />

signatures. This technology is extremely versatile in that it can be utilized for detection of antibody and/or<br />

nucleic acids. Current efforts to utilize this technology include the development of multiplexed “syndromic<br />

panels” (i.e. vesicular, swine fever and swine respiratory) and serological panels designed to differentiate<br />

vaccinated and infected animals.<br />

The TIGER biosensor and microarray are both unique in that they enable identification of an unknown agent<br />

from a clinical and/or environmental sample. The TIGER system utilizes both PCR and mass spectrometry<br />

to detect and identify known and/or unknown agents from a sample. Unlike the microarray and other<br />

detection devices (PCR, rRT-PCR and Luminex), the TIGER system does not rely on previous knowledge of<br />

sequence information for design of oligonucleotides, primers and probes. The powerful bioinformatics<br />

package that accompanies this tool allows for sophisticated differentiation and identification of multiple<br />

known and/or unknown pathogens in a single sample.<br />

As demonstrated in 1999, with the introduction of West Nile virus into North America and more recently with<br />

outbreak of monkeypox virus in the U.S. (2003), veterinarians and veterinary diagnostic laboratories of the<br />

future must and will play a central role in the early detection and identification of emerging and/or zoonotic<br />

pathogens. Veterinary laboratories (both state and federal) will be expected to be prepared for and respond<br />

to animal and public health issues. Preparation for these animal/public health emergencies will involve<br />

building on newly available technologies (expanding our diagnostic arsenal with assays that are “fit for<br />

purpose’), developing high throughput sample processing capabilities and maintaining and evaluating the<br />

current portfolio of diagnostic tests. Although each laboratory will face significant challenges in implementing<br />

many of the new, more sophisticated molecular techniques (identifying and training qualified staff, start-up<br />

costs, cost to maintain equipment, reagents, etc), it is the implementation of these tools, that can be utilized<br />

along side of the “gold-standard” assays, that will define veterinary diagnostics and its’ role in animal and<br />

public health issues in the future.<br />

References<br />

Perkins J, Clavijo A, Hindson BJ, Lenhoff RJ, McBride MT. (2006) Multiplexed detection of antibodies to<br />

nonstructural proteins of foot-and-mouth disease virus. Anal Chem. Aug 1;78(15):5462-8.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

OIE BIOTECHNOLOGY SYMPOSIUM: INTRODUCTORY REMARKS<br />

Steven Edwards, President of the OIE Biological Standards Commission<br />

The OIE (World Organisation for Animal Health) was established in 1924 in order to facilitate an<br />

internationally harmonised approach to animal disease control and reporting, particularly for transboundary<br />

diseases. Since then the organisation has expanded both its remit and its membership to include all<br />

countries with a significant livestock sector (current 170 Member Countries). The OIE is an<br />

intergovernmental organisation in its own right, with responsibility for improving animal health worldwide.<br />

The key missions of the OIE are:<br />

• To ensure transparency on animal diseases and zoonoses<br />

• To collect, analyse and disseminate veterinary scientific information<br />

• To provide expertise and encourage international solidarity on animal disease control<br />

• To publish animal health standards for international trade in animals and their products<br />

(this underpins OIE’s official mandate under the sanitary and phytosanitary agreement of the WTO)<br />

• To improve the legal framework and resources of national veterinary services<br />

• To promote safety for foods of animal origin and to promote science-based animal welfare<br />

As part of its science-based approach the OIE has a long tradition of developing standards for laboratory<br />

procedures used in animal disease diagnosis. The responsible bodies for this activity within OIE are the<br />

Biological Standards Commission (dealing with terrestrial animal diseases) and the Aquatic Animal Health<br />

Standards Commission. Among their other activities, these two commissions advise the OIE on the<br />

designation of Reference Laboratories for specified diseases, and produce laboratory manuals giving details<br />

of recognised and validated diagnostic tests.<br />

These commissions are supported by a variety of working and ad hoc groups, including the Biotechnology<br />

Ad hoc Group. It has been a tradition for some years now for the OIE to sponsor and organise a one day<br />

Biotechnology <strong>Symposium</strong> within the International Symposia of the <strong>WAVLD</strong>. This gives the opportunity for<br />

OIE to bring to the diagnosticians’ community a persective on emerging technologies with a potential for<br />

future application in laboratory-based diagnosis. Some of these are novel developments within established<br />

methodologies, while others present totally new approaches. Some are “near market” in that they are already<br />

or soon will be appearing in the armoury of tests offered by diagnostic laboratories. Others remain<br />

speculative and of uncertain potential, but we certainly should not ignore the developments happening in the<br />

research community.<br />

It is my pleasure to introduce the 2007 OIE Biotechnology <strong>Symposium</strong>, and I hope you find the presenations<br />

stimulating and informative.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1030 - 1230 Concurrent Session 1.4 - New Technologies & Platforms<br />

The United States National Animal Health Laboratory Network (NAHLN)<br />

B. M. Martin*, United States Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, National<br />

Veterinary Services Laboratories,<br />

1800 Dayton Ave., Ames, Iowa, 50010, USA;<br />

T. F. McElwain, Washington Animal Disease Diagnostic Laboratory and Animal Health Research Center, College of Veterinary<br />

Medicine, Washington State University,<br />

155N Bustad Hall, Pullman, Washington 99164, USA<br />

Introduction<br />

The United States National Animal Health Laboratory Network (NAHLN) was established in 2002 to enhance<br />

the early detection of, response to, and recovery from animal health emergencies, including bioterrorist<br />

events, newly emerging diseases, and foreign animal disease outbreaks that threaten the Nation’s food<br />

supply and public health. The NAHLN is a collaborative effort between the United States Department of<br />

Agriculture (USDA) and the American Association of Veterinary Laboratory Diagnosticians (AAVLD). From<br />

an initial group of 12 laboratories the NAHLN has expanded to 54 laboratories in 45 states.<br />

Elements upon which the NAHLN was founded included:<br />

• Standardized, rapid diagnostic techniques<br />

• A secure communications, alert mechanism, and reporting system<br />

• Modern equipment and trained personnel<br />

• Training, proficiency testing, and quality assurance programs<br />

• Facilities that meet biocontainment and security requirements<br />

• Scenario testing in support of regional and national training exercises<br />

Discussions & conclusions<br />

A program review of the NAHLN was initiated in 2007 to identify stakeholder perspectives concerning the<br />

objectives of the network, how well those objectives were being met, and whether changes in objectives are<br />

needed. The report indicates that the original objectives are appropriate and valid and that the NAHLN has<br />

made significant progress. Key accomplishments are summarized below.<br />

• Standardized, rapid diagnostic assays have been validated and deployed to NAHLN laboratories for<br />

avian influenza (AI), exotic Newcastle disease (END), and classical swine fever (CSF). NAHLN laboratories<br />

are also participating in USDA supported surveillance efforts for bovine spongiform encephalopathy (BSE),<br />

chronic wasting disease (CWD), and scrapie.<br />

• A centralized national data system has been developed that receives standardized data sets from<br />

participating NAHLN laboratories and supports automated transmission of data via Health Level Seven (HL7)<br />

messaging. Efforts are now focused on ensuring that NAHLN laboratories routinely transmit electronic test<br />

result messages.<br />

• A “Train the Trainer” program for foot and mouth disease (FMD), CSF, AI, and END rapid assays was<br />

developed and implemented. Not only has the program increased the number of laboratory personnel<br />

prepared to respond to a national animal health emergency, but it provides a cadre of trainers available to<br />

teach others. Successful implementation of this program was a significant step for the NAHLN and its<br />

mission of ensuring sufficient diagnostic capability and capacity to address an animal health emergency.<br />

• USDA’s National Veterinary Services Laboratories (NVSL) serves as the reference laboratory for NAHLN<br />

and has provided training and proficiency testing programs for NAHLN laboratory personnel. The NVSL also<br />

provides support through production and distribution of reagent standards and quality control panels.<br />

• One of the major successes of the NAHLN has been the collaboration with other Federal and state<br />

organizations to achieve common goals. The NAHLN has become the animal health laboratory backbone of<br />

the United States emergency response and recovery program, and has enabled implementation of national,<br />

standardized surveillance for high priority diseases.<br />

References<br />

1. American Association of Veterinary Laboratory Diagnosticians Website:<br />

http://www.aavld.org/mc/page.do<br />

National Animal Health Laboratory Network Website: http://aphis.usda.gov/animal_health/nahln<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPING AN APPROACH FOR RAPID IDENTIFICATION OF EMERGING BIOLOGICAL THREATS<br />

Bill Colston, Ray Mariella, Reginald Beer, Klint Rose, Kevin Ness, Elizabeth Wheeler, Tom Slezak, Shea Gardner, Peter Williams, Amy<br />

Hiddessen, Monica Borucki, Ben Hindson, Chris Bailey, and Crystal Jaing (UC Lawrence Livermore National Laboratory)<br />

In our era of rapid, easy world wide travel, infectious diseases, both naturally occurring and<br />

intentionally introduced, hold increasing potential to cause disease, disability and death. Their<br />

prevention and control is fundamental to individual, national and global health and security.<br />

Lessons learned from the recent SARS outbreak teach us that without the ability to rapidly identify<br />

and characterize a previously unknown or emerging pathogen, our country’s ability to mount a<br />

timely and effective response to a nationwide bioterrorism event is unlikely. In recent years the<br />

biodefense community has focused on short-term production of assays for the specific<br />

identification of a short list of known pathogens. While necessary, this focus has created a gap in<br />

our defensive and public health arsenal. We are currently ill-prepared to deal with novel pathogens<br />

(natural or engineered), complex mixtures of organisms, or detection of virulence regardless of the<br />

organism conferring it. Particularly in the case of viral agents, persistent technology gaps exist in<br />

this process, including sample handling and preparation and highly-multiplexed assays that can<br />

detect and identify both known and unknown viruses. Also, in the case of viral infectious agents,<br />

this problem is compounded by our near-total lack of knowledge of “normal” viral backgrounds in<br />

environmental, human, and agricultural samples.<br />

To address this challenge, we have begun developing an approach to create a translational<br />

measurement capability that will allow rapid, high-throughput viral screening. This approach<br />

includes (1) Sample preparation capabilities to isolate virus particles from the numerous other<br />

inter- and intra-cellular materials present in a nasopharyngeal sample. Viruses, by their very<br />

nature, cannot live and replicate by themselves, and must rely on the complex biochemical<br />

machinery of the host cells. Thus, detection of viral signatures will rely on removing the great<br />

number of interfering cellular components of the host cells, including proteins, organelles, and the<br />

cells’ own genetic materials. (2) A long-term detection capability to isolate and individually test<br />

every viral particle in a sample in a high-throughput, massively parallel microfluidic system. This<br />

will be done by isolating each virus particle in its own picoliter size ‘individual biochemical<br />

laboratory’ or microdroplet for sensitive PCR analysis. (3) Development of comprehensive, specific<br />

multiplex PCR assays of known organisms that can be carried out in a microdroplet and will be<br />

capable of family level identification following amplicon analyisis. The goal is to ultimately be able<br />

to screen all known viral genomes to develop a minimum set of signatures that can be used to<br />

characterize an unknown virus.<br />

We will present new developments in microfluidic engineering, highly multiplexed biological assays<br />

and advances in bioinformatics aimed at providing a broad capability for identification and<br />

characterization of previously known and unknown viruses. Specifically, we have currently<br />

demonstrated:<br />

• Application of acoustic, electrophoretic and electrophoretic techniques to demonstrate a<br />

continuous size selective sorting of particles in a flowing microfluidic stream. We have<br />

developed a 3D theoretical simulation for a multi-field separator that combines acoustic<br />

focusing capability with electrophoresis, and have used this simulation to design and test a<br />

physical prototype.<br />

• Development of preliminary viral discovery assays using theoretical calculations coupled<br />

with wet-bench validation. To accomplish this, we designed novel algorithms and built<br />

software to select highly conserved primer sets, optimized to work in multiplex format. We<br />

have demonstrated reproducible, specific PCR amplification of predicted fragments from a<br />

viral DNA genome (vaccinia virus, lister strain) using 10 bp primer sets.<br />

•<br />

Demonstration of Real-time, Taq-man-based sub-nanoliter PCR using digital microfluidics. Our<br />

system detected a single copy of viral genomic DNA encapsulated in ten-picoliter droplets, with a<br />

much earlier cycle threshold than conventional devices.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

VIRAL IDENTIFICATION USING MICROARRAYS<br />

J.P. Dukes 1 , A. Abu-Median 2 , M. Watson 2 , R. Card 3 , D. Stone 4 , M. Banks 3 ,<br />

P. Britton 2 and D.P. King 1<br />

1 Institute for Animal Health, Pirbright Laboratory, Ash Road, Pirbright, Woking, Surrey, UK; 2 Institute for Animal Health, Compton<br />

Laboratory, Newbury, Berkshire, UK; 3 Veterinary Laboratories Agency, New Haw, Addlestone, Surrey, United Kingdom, 4 Cefas<br />

Weymouth Laboratory, The Nothe, Barrack Road, Weymouth, Dorset<br />

Key words: Virus; characterisation; detection; microarray<br />

The identification of viruses using standard molecular techniques is often obfuscated by the existence of<br />

closely related isolates. The capacity of microarrays to perform numerous assays on the same sample<br />

increases experimental range by many orders of magnitude. Microarrays enable both broad-spectrum<br />

detection, and finer resolution fingerprinting. A microarray consisting of probes covering 300+ viral ‘species’<br />

has been developed. The array is being validated to serotype and subtype veterinary viruses using Foot and<br />

Mouth Disease Virus (FMDV), Infectious Bronchitis Coronavirus (IBV), and other livestock and fish diseases<br />

as models to optimise protocols for probe design, sample labelling, hybridisation and analysis.<br />

Using sequence data available both in the public domain and generated in-house, we have designed 70base<br />

oligonucleotide probes covering 35 virus families. Oligo probes were synthesised commercially and<br />

arrays spotted in-house using a previously published protocol. Nucleic acid is isolated, amplified, labelled<br />

and hybridised by established methods, but we have developed bespoke software to analyse the<br />

fluorescence data generated (‘DetectiV’) providing statistical support for viral identification.<br />

The microarray is capable of discriminating different viruses (IBV, FMDV). Furthermore, different serotypes<br />

of FMDV and different topotypes within a serotype also give different ‘signatures’. Viral signatures are also<br />

detectable in ‘real’ samples from infected animals, and normalisation to an uninfected control reduces the<br />

majority of noise. Current work at IAH Pirbright is focused on validating this array and determining the<br />

resolution that can be achieved using further topotypes of FMDV, related picornaviruses, and other vesicular<br />

viruses.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ASSAY PLATFORMS FOR THE RAPID DETECTION OF VIRAL PATHOGENS BY THE ULTRAHIGH<br />

SENSITIVITY MONITORING OF ANTIGEN-ANTIBODY BINDING<br />

A Fabricate capture antibody chip<br />

Au<br />

B Expose chip to sample, capturing antigens<br />

Au<br />

Antibodies Antigens Reporters<br />

Figure 1. Chip design and assay<br />

scheme. A) Immobilize capture<br />

antibodies on smooth gold surface. B)<br />

Expose chip to sample. C) Expose<br />

chip to SERS reagent, i.e., gold<br />

nanoparticle coated with Raman<br />

reporter molecules and antibodies.<br />

Lastly, read chip by acquiring Raman<br />

spectrum.<br />

Au<br />

Au<br />

J. Uhlenkamp, K. Kwarta, and B. Yakes<br />

Center for Combinatorial Sciences and Department of Chemistry and Biochemistry<br />

Arizona State University, Tempe, AZ USA<br />

R. J. Lipert<br />

Institute for Combinatorial Discovery and Ames Laboratory-USDOE, Iowa State University, Ames, IA USA<br />

J. Ridpath and J. Neill<br />

USAD/ARS/National Animal Disease Center, P.O. Box 70, Ames, IA USA<br />

M. D. Porter*<br />

Departments of Chemistry and of Chemical Engineering, University of Utah, Salt Lake City, UT USA<br />

Introduction<br />

The drive for early disease detection and growing threat of bioterrorism has markedly amplified the demand<br />

for ultrasensitive, high-speed diagnostic tests for viral pathogens. This presentation describes innovations in<br />

the development of platforms and readout methodologies that potentially address demands in this arena<br />

through a coupling of gold nanometric particle labelling, surface enhanced Raman spectroscopy (SERS),<br />

and high speed fluidics.<br />

Figure 1 briefly overviews our strategy, which uses gold nanoparticle labels and surface enhanced Raman<br />

spectroscopy (SERS) as a highly sensitive detection methodology that can be applied in a high throughput<br />

format by rapidly reading several pathogens from multiple addresses on a single biochip. As such, this<br />

strategy exploits the strong SERS signal for organic dyes (i.e., Raman reporter molecules) that are<br />

immobilized on gold nanoparticles and subsequently coupled to the appropriate biospecific species, thus<br />

acting as a label. Using a sandwich immunosorbent assay, the identity of each antigen is determined from<br />

the characteristic SERS spectrum of the nanoparticle-bound reporters linked to the tracer antibody, with<br />

each antigen then quantified by the spectral intensity of reporter species.<br />

Au<br />

The main advantage of our extrinsic Raman labels (ERLs)<br />

reflects the strength of the scattering intensity, which has enabled<br />

the detection of individual particles with only a few seconds of signal<br />

acquisition. This capability translates to the data exemplified by an<br />

assay of feline calici virus<br />

(FCV) in Figure 2. As<br />

evident, the spectral signature<br />

of the Raman reporter<br />

increases with FCV<br />

concentration, yielding a<br />

detection limit at low<br />

femtomolar levels.<br />

Figure 2. SERS-based immunoassay<br />

for FCV on gold film capture surface.<br />

SERS spectra (1-s integration and<br />

offset for visualization) as a function of<br />

FCV concentration: a) blank (cell<br />

culture media only), b) 5.0 x 10 5 , c) 5.0<br />

x 10 6 , d) 5.0 x 10 7 , e) 1.0 x 10 8 , f) 2.5 x<br />

10 8 viruses/mL.<br />

In extending this<br />

ability, this presentation<br />

details the fabrication,<br />

incubation, and rapid readout<br />

of chip-scale platforms that<br />

realize virus detection limits at<br />

low femtomolar levels at total<br />

development times of 10 min<br />

or less. Examples will focus<br />

on the use of protein arrays<br />

as platforms targeted for virus<br />

immunoassays. Each<br />

example will also highlight<br />

issues related to sensitivity,<br />

non-specific adsorption, fluid<br />

manipulation, and assay<br />

speed.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LATE-PCR WITH PRIMESAFE - MAXIMUM DIAGNOSTIC INFORMATION FROM A CLOSED-TUBE<br />

L.J. Wangh, C. Hartshorn, K.E. Pierce, J.A. Sanchez, J.E. Rice, and A.H. Reis, Jr.<br />

Department of Biology, Brandeis University, Waltham MA, USA, 02454-9110<br />

(email: Wangh@brandeis.edu)<br />

Introduction: In veterinary and human medicine there is a growing need for diagnostic assays that can<br />

rapidly detect and analyze numerous species and strains of infectious organisms and viruses. RNA viruses<br />

are particularly challenging because they evolve rapidly. In the case of veterinary medicine, tests have to be<br />

done at pen-side or in the field and the findings can have great economic impact, including slaughter of<br />

many animals. Currently the use of RT-PCR is limited in scope and few, if any assays provide results from<br />

multiplex data in the field. We have overcome these limitations by constructing assays based on LATE-PCR<br />

and (RT)-LATE-PCR. We are collaborating with Smiths Detection to implement these assays on an<br />

automated “pen side” sample preparation and PCR system (see C.Volpe et. al. this volume).<br />

Materials & Methods: Linear-After-The-Exponential (LATE)-PCR, invented in our laboratory, is an advanced<br />

form of asymmetric PCR in which each amplicon is generated using a Limiting Primer whose initial,<br />

concentration-adjusted melting temperature (Tm L ) is at least as high as that of the Excess Primer (Tm X ).<br />

Under these conditions amplification begins with efficient exponential amplification of double-stranded DNA<br />

and then abruptly switches to linear amplification of one strand, as the limiting primer is depleted from the<br />

reaction. Each single-stranded amplicon can be quantitatively detected in real-time or at end-point using<br />

fluorescent probes of different color. Moreover, in LATE-PCR, product detection is carried out separately<br />

from primer annealing in the thermal cycle, thereby making it possible to use both sequence-specific and<br />

mis-match tolerant fluorescent probes that hybridize to their target strands over a broad temperature range<br />

below the annealing temperature of the reaction. In addition, each single-strand can be sequenced by a<br />

convenient Dilute-‘N’-Go procedure, even when multiple sequences are generated in the same reaction.<br />

PrimeSafe TM , also invented in our laboratory, is a PCR additive that suppresses mis-priming throughout all<br />

thermal cycles, thereby simplifying construction of multiplexed LATE-PCR assays. (RT)-LATE-PCR assays<br />

use random hexamers to generate cDNA molecules (two-step protocol), or use the LATE-PCR primers<br />

themselves (one-step protocol). In either case, cDNA synthesis and amplification can be carried out in the<br />

same tube.<br />

Results: Using LATE-PCR we are constructing assays that are quantitative for the number of target<br />

molecules present at the start over seven orders of magnitude and down to single DNA or cDNA molecules.<br />

In this conference we present new results using multiplexed LATE-PCR assays for Foot and Mouth Disease<br />

(see Pierce et al. ) or Avian Flu (see Hartshorn et al.) are described in this volume. These assays are being<br />

implemented on Smiths Detections “pen side” device . Conserved sequences characteristic of particular<br />

pathogens can be reliably detected with sequence-specific probes that form probe-target hybrids at<br />

characteristic temperatures. Alternatively, variable sequences that differ slightly from one pathogenic strain<br />

to another can be distinguished using mis-match tolerant probes that allow for probe-target hybridization<br />

over a range of low temperatures. In either case, the complete nucleotide-by-nucleotide sequence of the<br />

amplicon within which the probe sequence lies can be determined conveniently and cost effectively using the<br />

Dilute-‘N’-Go dideoxy sequencing protocol. Thus, when fully implemented, LATE-PCR assays installed in a<br />

Portable Veterinary PCR Laboratory currently under development by Smiths Detection, Inc. will generate<br />

sophisticated information in the field, as well as the amplicons needed for thorough sequence analysis in the<br />

laboratory.<br />

Discussion & Conclusions: LATE-PCR and its related novel technologies promise a new generation of<br />

highly informative diagnostic assays for use in the laboratory or in the field. Smiths Detections “pen side”<br />

system is the only portable instrument specifically designed to take full advantage of these improvements.<br />

References:<br />

All of our publications on LATE-PCR are available at: http://www.brandeis.edu/projects/wanghlab/<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A COMPLETE WORKFLOW FOR NUCLEIC ACID BASED<br />

HIGH THROUGHPUT PATHOGEN DETECTION<br />

XW Fang*, A Burrell, R C Willis, Q Hoang, W Xu, M Bounpheng, W Ge and J El-Attrache<br />

Ambion, Inc. An Applied Biosystems Business. 2130 Woodward St., Austin, TX 78744<br />

Introduction<br />

Nucleic acid-based technology is increasingly used for pathogen detection and classification, as well as for<br />

differentiation of infected animals from vaccinated animals. Nucleic acid isolation is the crucial step for this<br />

technology. The variety of sample matrix and pathogen types poses great challenges in developing a<br />

method for universal sample preparation easily amendable for all applications. An additional challenge is to<br />

automate the sample preparation to minimize human exposure to the infectious pathogens and to reduce the<br />

variation of the process. Moreover, qRT-PCR buffer optimization and careful primer/probe design are critical<br />

to assure analytic sensitivity and detection sensitivity and specificity.<br />

Materials & methods<br />

Pathogen DNA/RNA is isolated using MagMAXTM kits on KingFisher Magnetic Particle Processors. RNA<br />

quality is evaluated for purity (by A260/A280 ratio and RT-PCR inhibition), intactness (with gelelectrophoresis).<br />

Pathogen detection is performed on ABI 7500 Fast and 7900HT real-time instruments<br />

using AgPath-IDTM Reagent Kits.<br />

Results<br />

High quality DNA/RNA can be isolated with MagMAX Kits from a variety of sample matrices, such as swabs,<br />

feces, growth media, serum, plasma, cultured cells, tissue and blood. The purified RNA/DNA is ready for<br />

quantification by real-time PCR and sequencing. Samples can be process in microfuge tubes and 96-well<br />

plates. The process can be easily automated.<br />

KingFisher Magnetic Particle Processors automate magnetic bead-based processes with the coordinated<br />

move of the permanent magnetic rods and disposable tip combs. This unique approach makes washing and<br />

elution more efficient, accelerates processing, and also makes walk-away automation feasible. The<br />

processors can do up to 15, 24 or 96 samples per run depending on the model of the instrument. A typical<br />

nucleic acid isolation on a KingFisher machine takes


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AN ADVANCED FIELD DEPLOYABLE “PEN SIDE” SAMPLE PREPARATION AND PCR SYSTEM<br />

Doug Green 1 , C. Volpe 2 *, John Czajka 1 , Jason Betley 2 and Jay Lewington 2<br />

1 Smiths Detection, 2202 Lakeside Blvd. Edgewood, MD 21040, USA<br />

2 Smiths Detection, 459 Park Avenue, Bushey, Watford, WD23 2BW, United Kingdom.<br />

Introduction<br />

Exotic and Zoonotic diseases pose a threat to the world’s wildlife, commercial livestock and the population in<br />

general. Efforts to control the spread of naturally occurring highly infectious pathogens, for example Highly<br />

Pathogenic Avian Influenza, have been complicated by the need to transport samples to the lab for ultimate<br />

identification. This is especially problematic in remote locations. This abstract describes a briefcase sized<br />

portable sample preparation and PCR system, which has been designed, from the outset, with the field<br />

veterinarians needs and mode of operation in mind, including the ability to sanitise the unit with disinfectant.<br />

The system automatically purifies nucleic acids from a wide range of sample types and carries out PCR<br />

analysis, reporting either a positive or negative result or a strain level identification where applicable. The<br />

system uses a number of novel technologies and approaches to provide a fully automated portable on-site<br />

identification capability in a wide range of weather conditions, by a person with no knowledge of PCR.<br />

Material & methods<br />

Operation of the device is extremely simple. A veterinarian suspecting the presence of disease takes a<br />

sample from the animal. The nature of the sample is dependant on the disease under suspicion and is not<br />

limited by the instrument. For example they may take blood samples or vesicular tissue as appropriate. The<br />

veterinarian then places that sample in to a single use sample preparation device and places the device on<br />

the instrument. At this point the assay to be performed is automatically selected and the automated sample<br />

preparation process begins. The purified nucleic acid is automatically mixed with PCR reagents and the PCR<br />

process begins. The instrument then performs the appropriate data analysis and reports the result as a<br />

positive and negative test result for the test carried out, and reports the strain level identification of the<br />

pathogen where applicable. This entire process is performed with no user intervention and is designed to be<br />

performed by a person with no knowledge of molecular biology techniques.<br />

Results<br />

The overall performance of the system depends on the individual performance of the instrument, sample<br />

preparation and assays. Recent analysis of the performance of the PCR instrument has shown a good<br />

correlation with selected lab based PCR instruments, indicating a similar level of performance can be<br />

expected in the field. The sample preparation device was designed to take the lab based process into the<br />

field and automate it. Initial results indicate that the device performs as well as the original bench process.<br />

There are currently 2 assays being developed and validated on this platform, one to detect all seven<br />

serotypes of Foot and Mouth Viruses in a single tube, and one to differentiate high and low pathogenic H5N1<br />

Avian Influenza Virus. These assays are in development/validation using real samples in collaborating labs<br />

to determine their efficiency in a lab setting prior to their transfer to our field deployable platform (see<br />

abstracts in this volume). The platform uses a novel PCR chemistry called LATE-PCR which is able to<br />

identify many pathogens or strains of a pathogen in a single tube. The highly multiplexable nature of LATE-<br />

PCR means that a wide range of pathogens can be tested for simultaneously resulting in a simplified mode<br />

of operation. The system is currently being prepared for field based trials.<br />

Discussions & conclusions<br />

“Pen side” detection systems offer the promise of rapid detection of disease allowing a more resilient<br />

response and more effective outbreak management. The challenge in deploying PCR based analysis<br />

systems in the field is the requirement for a simple, sample preparation system capable of producing good<br />

quality nucleic acid from a wide range of animal samples. Systems such as Smiths Detection’s “Pen Side”<br />

testing system may provide the means to rapidly detection disease outbreaks.<br />

References<br />

For more information please visit www.smithsdetection.com/vets<br />

or e-mail vets@smithsdetection.com<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE AUSTRALIAN ANIMAL PATHOLOGY STANDARDS PROGRAM: PRESERVATION AND<br />

DIGITISATION OF THE COLLECTION, AND ACCESS VIA ‘NEW’ TECHNOLOGIES<br />

<strong>WAVLD</strong> SYMPOSIUM 2007<br />

C. Lenghaus, Australian Animal Pathology Standards Program; R.L. Reece*, L. Simmons NSW Department of Primary Industries.<br />

BACKGROUND<br />

The Australian Animal Pathology Standards Program (AAPSP) is managed and maintained by Animal Health<br />

Australia. It encompasses the former National Registry of Domestic Animal Pathology of approximately<br />

20,000 glass slides from 4,000 indexed cases.<br />

AIMS<br />

AAPSP is creating an immortalised library of veterinary pathology digitised images; with readily accessible<br />

subsets focused on particular species, tissues and lesions; and annotated individual images for selfteaching.<br />

RESTORATION<br />

A subset has been selected for digitisation but many chosen slides require renovation and repair: others<br />

need to be replaced.<br />

OUTCOMES<br />

CURRENT<br />

ROLE IN CONTINUING PROFESSIONAL DEVELOPMENT<br />

It is mandatory for veterinarians to undertake Continuing Professional Development, and NATA requires<br />

participation in professional competency assessments. For veterinary pathologists in Australia this is<br />

provided by AAPAP via:<br />

ANNUAL TRAINING COURSES prepared and delivered by Australian veterinary pathologists, and by USA<br />

specialists with financial assistance from the C.L.Davis Foundation for Comparative Veterinary Pathology.<br />

They take the format of a 2-3 days intensive workshop held in several accessible locations and deal with a<br />

particular subject such as dermatopathology or neuropathology.<br />

COMPETENCY. A quarterly Histopathology Proficiency test is available through the AAPSP. For one round,<br />

unstained histological slides are distributed, these slides are stained HE, examined and described, and then<br />

the slides and a standardised report returned for evaluation; on the alternate round a cases is scanned onto<br />

a DVD at multiple magnifications and it is examined and reported on.<br />

THE FUTURE<br />

Due to dispersal of veterinary pathologists around Australia and their reduced numbers and relative paucity<br />

of ‘free’ diagnostic cases for teaching and learning, especially by trainees, a web based system allowing<br />

access from office or laboratory is crucial. For NATA purposes, trainees are required to document their<br />

training. Options are:<br />

Individual cases for QA. A few digitised slides examined remotely by many sites and users. The best<br />

answers to judicious questions can be selected from proffered options and the answers stored and collated<br />

centrally.<br />

Teaching modules with self-instruction. Expressions of interest to prepare these have been made and some<br />

are in process: one on-line module prepared for a post-graduate degree course has been purchased<br />

already. They require enormous time and materials for preparation. It is hoped that recently retired or near to<br />

retirement veterinary pathologists can prepare these based on their experience and personal or employer<br />

archives.<br />

Continuing Professional Development and Help. Access to a collection of indexed digitised images of a<br />

particular species, disease or syndrome, so that help is obtained for a current case requiring further<br />

elucidation, or knowledge base is expanded.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1030 - 1230 Concurrent Session 2.4 - Quality Assurance<br />

LABS OF THE FUTURE<br />

A CULTURAL SHIFT IN CANADA FOR FOREIGN ANIMAL DISEASE (FAD) DETECTION<br />

D. Ridd & A. Copps, National Centre for Foreign Animal Disease, Canadian Food Inspection Agency (CFIA)<br />

Presenter: Dr. R.P. Kitching, Director, National Centre for Foreign Animal Disease, CFIA<br />

The success of a country’s capability for rapid detection of potentially epidemic animal diseases depends on<br />

many factors, one of which is laboratory diagnostic capacity and capability. Canada’s lab strategy under the<br />

Avian Influenza (AI) Pandemic Preparedness Plan brought forward the priority in 2006 to accelerate the<br />

plans and timelines for laboratory interoperability and to establish a national early warning system for<br />

perceived risks of Avian Influenza. Within 6 months, 4 federal and 7 provincial laboratories in the Canadian<br />

Animal Health Surveillance Network (CAHSN) were trained, equipped and approved for testing the presence<br />

of AI virus. The CAHSN was developed to consolidate the efforts across provincial, territorial and federal<br />

jurisdictions to respond to animal disease threats that could affect animal health, the food supply, or public<br />

health.<br />

Laboratory interoperability is a means towards meeting Canada’s requirements of an early warning system<br />

for outbreak management and emergency preparedness, planning for breaches of biosecurity and threats of<br />

bioterrorism. Quality Assurance (QA) Support and Laboratory Support although separate programs within<br />

CAHSN, are working together in achieving these goals and objectives. These programs’ are designed to<br />

deliver and harmonize quality assurance and laboratory systems through collaborations between<br />

federal/provincial animal health laboratories across Canada. The provincial laboratories as first responder<br />

laboratories will have the capacity and capability to rapidly diagnose foreign animal diseases, having been<br />

trained in FAD test methods and receiving the necessary supports towards lab infrastructure.<br />

The CAHSN Quality Assurance Support Program delivers support to the development of internal quality<br />

systems for FAD within the network laboratories with the goal of the labs achieving ISO/IEC 17025<br />

accreditation. Based on site evaluations, funding has been allocated to establish and aid the implementation<br />

and maintenance of a standardized quality management program and to support the achievement of<br />

ISO/IEC 17025 accreditation. Resources were provided for QA Officers, ISO training, and ongoing<br />

accreditation costs. The CAHSN QA Support Program Manager liaises with the Network Laboratory<br />

Directors, Laboratory Managers/Supervisors and Quality Assurance Officers in a consulting, advisory<br />

capacity and provides oversight regarding all QA aspects in network laboratories relating to FAD testing.<br />

The CAHSN Laboratory Support Program ensures the animal health laboratories sharebest diagnostic<br />

practice, particularly for the major animal diseases such as Foot-and-Mouth Disease, Classical Swine Fever,<br />

Avian Influenza and Newcastle Disease, and establishes common diagnostic protocols and reagents, while<br />

ensuring diagnostic competence is maintained by the regular distribution and evaluation of proficiency<br />

panels. Where necessary, new equipment is provided to allow all network laboratories to make use of the<br />

most sensitive and specific diagnostic tests available. The CAHSN Laboratory Support Manager liaises with<br />

the Network Laboratory Directors and Laboratory Managers/Supervisors in a consulting, advisory capacity<br />

regarding all aspects of training/certification for network labs, encompassing the areas of equipment advice<br />

and purchases, reagents/supplies, surge capacity issues and is directly involved in training and certification<br />

of analysts.<br />

Conclusion:<br />

Canada’s jurisdictional requirements for diagnostic surge capacity are being achieved through harmonization<br />

and standardization of quality assurance and lab interoperability. The CAHSN Quality Assurance Support<br />

Program and the Laboratory Support Program are integral in aiding the network laboratories in the detection<br />

of emerging animal disease threats which could have zoonotic potential. This capability of early diagnosis<br />

and control is essential to minimizing the human health and economic consequences to the country.<br />

Ann Copps, CAHSN QA Support Manager coppsa@inspection.gc.ca<br />

Deidre Ridd, CAHSN Lab Support Manager dridd@inspection.gc.ca<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LABORATORY-BASED EARLY ANIMAL DISEASE DETECTION UTILIZING A<br />

PROSPECTIVE SPACE-TIME PERMUTATION SCAN STATISTIC<br />

*C.N. Carter, University of Kentucky; A. Odoi, University of Tennessee; J. Riley, Hensley and Associates, Lexington, Kentucky; J. Smith,<br />

University of Kentucky; R. Stepusin, University of Kentucky;<br />

T. Cattoi, University of Kentucky; S. McCollum, University of Kentucky<br />

Introduction<br />

Veterinary diagnostic laboratories are in a unique position to analyze data from large numbers of clinical<br />

cases and to help with the early detection of health problems. The aim of this study is to develop a system<br />

for early detection of clusters of health events in animal populations using diagnostic laboratory data. The<br />

system provides near-real-time cluster alarms/alerts and medical situational awareness. Thus, it aids in the<br />

decision-making process related to conducting field investigations and/or mounting appropriate and timely<br />

medical responses. The system could, as well, be used to detect clusters of animal health events of public<br />

health significance (e.g. bioterrorist attacks).<br />

Material & methods<br />

Kulldorff’s prospective space-time permutation scan statistic was utilized in this project. The method uses a<br />

cylindrical window of varying size to scan for potential clusters in space and time. The statistic only requires<br />

case health events (e.g. confirmed diagnoses, deaths), thereby eliminating the need for population-at-risk<br />

data. We developed an engine that automatically performs statistical analysis at the close of business each<br />

day. A likelihood ratio test is used to test statistical significance of potential clusters. Identified clusters are<br />

automatically mapped for daily epidemiological decision-making. Here we present the methodology that was<br />

used to detect outbreaks of bovine blackleg and equine leptospirosis which occurred in 2006-2007.<br />

Results<br />

The system successfully detected significant clusters of bovine blackleg and equine leptospirosis cases<br />

during the 2006-2007 outbreaks. The detection of these clusters allowed the laboratory epidemiology<br />

section to mount a timely awareness campaign that included email alerts, field investigations, and direct<br />

consults with practicing veterinarians. Although not verifiable, this campaign likely led to enhanced<br />

vaccination efforts against bovine blackleg and aggressive MAT titer screening of horse farms for possible<br />

prophylactic treatment of mares. Blackleg losses for the 2006-2007 season are estimated at over $500,000<br />

USD for eastern Kentucky alone. In addition, economic losses reported by 13 horse farms that experienced<br />

equine leptospiral abortion amounted to over $3 million USD for the 2006-2007 season alone. Earlier<br />

medical responses to outbreaks should result in reducing economic losses.<br />

Discussions & conclusions<br />

The appropriate analysis of laboratory-detected animal health events can be successfully used to generate<br />

medical alerts which can lead to timely and appropriate responses that can improve overall health outcomes<br />

and reduced economic losses. Further studies will be conducted to assess the sensitivity and specificity of<br />

medical alerts generated by this system.<br />

References<br />

Carter CN, Odoi A: Diagnostic Laboratory Surveillance and Epidemiology: Serving Agriculture and Public<br />

Health, Proceed 143 rd AVMA Convention, CD-ROM, July, 2006.<br />

Carter CN: Equine Disease Surveillance in Kentucky, Equine Disease Quarterly, Oct, 2005, Vol. 14, No. 4,<br />

pp 5-6.<br />

Bradley CA, Rolka H, et al. BioSense: implementation of a national early event detection and situational<br />

awareness system. MMWR Morb Mortal Wkly Rep. 2005 Aug 26; 54 Suppl: 11-9.<br />

Kulldorff M, Heffernan R, Hartman J, Assuncao R, Mostashari F. A space-time permutation scan statistic for<br />

disease outbreak detection. PLoS Medicine 2005 March; 2(3): 216-224.<br />

Norström M, Pfeiffer DU, Jarp J. A space-time cluster investigation of an outbreak of acute respiratory<br />

disease in Norwegian cattle herds. Preventive Veterinary Medicine, 47: 107-119, 2000.<br />

Perez AM, Ward MP, Torres P, Ritacco V. Use of spatial statistics and monitoring data to identify clustering<br />

of bovine tuberculosis in Argentina. Preventive Veterinary Medicine, 56: 63-74, 2002.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT, VALIDATION AND IMPLEMENTATION OF MOLECULAR DIAGNOSTIC TESTS FOR<br />

IMPORTANT PATHOGENS OF FARMED ANIMALS AND WILDLIFE – A PRAGMATIC APPROACH<br />

*P.R. Wakeley, J. Errington, N. Johnson, C. Fearnley, M. J. Slomka, F. Yong and J. Sawyer<br />

Veterinary Laboratories Agency – United Kingdom<br />

Introduction<br />

We have developed a panel of molecular diagnostic tests for the detection of pathogens of farmed animals<br />

and wildlife to support diagnosis and surveillance activities. The tests developed include those to detect<br />

pathogens of farmed food animals such as a bovine viral diarrhoea virus/border disease virus (BVDV/BDV)<br />

and an assay to detect pathogenic leptospires. We have also developed and validated assays for the<br />

detection of pathogens of wildlife with zoonotic potential such as lyssavirus 1 and avian influenza, as well as<br />

assays to support the horse industry e.g. for the detection of Taylorella equigenitalis 2 the causative agent of<br />

contagious equine metritis (CEM). Molecular diagnostics offer the advantage over traditional “gold standard”<br />

technologies of viral or bacterial culture in that they are rapid, allow differentiation of closely related species<br />

and are not reliant on the presence of viable pathogen.<br />

Material & methods<br />

The tests developed make use of real-time polymerase chain reactions using fluorescence detection, either<br />

TaqMan or FRET. Where possible the assays have been validated against the accepted “gold standard” test<br />

which generally involves culture methods. However, we have found that culture techniques are either<br />

expensive to perform and can be substituted by inexpensive tests such as antigen ELISA e.g. for BVDV, or<br />

the confirmatory test is extremely lengthy to perform such as culture of leptospires (36 weeks) requiring other<br />

confirmatory tests of positive PCRs e.g. nucleic acid sequencing. Assays have been characterised with<br />

respect to analytical and diagnostic sensitivity and specificity as well as repeatability and robustness in<br />

comparison to the standard assays and have also been field trialled. In most instances we have taken a<br />

pragmatic approach to field validation as sufficient positive samples have not been available. One of the key<br />

limitations to successful performance of a PCR test is the efficient extraction of nucleic acids. Control assays<br />

have been developed to detect either RNA transcripts from the host, based on β-actin expression or for<br />

bacterial pathogens, such as leptospires and T.equigenitalis, real time detection assays based on the<br />

generic 16S rDNA of eubacteria. These controls are crucial for assays where the specimen cannot be resampled<br />

e.g. cloacal swabs from live captured wild birds for avian influenza surveillance, and provide<br />

confidence that the extraction component of the assay is performing optimally. Automated extraction of<br />

nucleic acids has been implemented to reduce contamination and “hands on time” for both the BVDV/BDV<br />

assay where we have used the lower throughput MagNApure (Roche) and avian influenza virus assays<br />

where we have used the higher throughput BioRobot 96 (QIAGEN).<br />

Results<br />

Field validation of several assays has led to novel and interesting findings. The first BVDV genotype 2 in the<br />

UK, BVDV in alpaca in the UK and the first border disease virus in cattle were discovered during the<br />

validation process of the BVDV/BDV TaqMan assay. Using the lyssavirus assay a number of European bat<br />

lyssavirus genotype 2 have been found in bats in the UK, including in an urban area close to the VLA.<br />

Implementation of the CEM assay has not only allowed detection of the reportable bacteria in infected<br />

horses in transit but due to the ability of the assay to discriminate T.equigenitalis from a closely related<br />

Taylorella species has led to changes in the controls used for culture purposes in our laboratories. The<br />

leptospire PCR has been successfully used to detect and trace pathogenic leptospire in a human case of<br />

leptospirosis and has also led to discussion of the impact of these bacteria on bovine abortion in the UK due<br />

to the low numbers of positives detected using the PCR.<br />

Discussions & conclusions<br />

The design and development of molecular tests for animal pathogens is not time consuming and costly; the<br />

validation of such tests most definitely is. Due to time constraints, the non-availability of appropriate<br />

confirmatory tests and the lack of suitable field samples, a pragmatic approach must be adopted to<br />

validation. Wherever possible internal controls should be implemented for not only the control of the<br />

amplification process but also the extraction. Validation processes in our experience can frequently lead to<br />

the discovery of novel scientific findings.<br />

References<br />

(1) Wakeley et al. J. Clin. Microbiol. 2005. 43 (6):2786-2792<br />

(2) Wakeley et al. Vet. Microbiol. 2006. 118 (3-4):247-254<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

QUALITY ASSURANCE AND CONTROL ISSUES WITH PCR AS A DIAGNOSTIC TOOL<br />

J. Oakey, Biosecurity Queensland, Tropical and Aquatic Animal Health Laboratory, Department of Primary Industries and Fisheries<br />

This aims to be a discussion of the quality assurance principles and current issues with the use of molecular<br />

biology techniques as tools for veterinary diagnosis.<br />

Detection of identifiable nucleic acid rapidly has become commonplace in veterinary diagnostic laboratories,<br />

presenting a number of significant advantages over conventional techniques. These methods are often<br />

faster, and more sensitive and specific. However, the routine use of what were so recently considered as<br />

research tools may be prone to over-reliance and lack of adequate quality management, quality control and<br />

overall quality assurance. It is essential that such techniques are demonstrated to be at least as reliable and<br />

“fit-for-purpose” as conventional methods if the test results are to be universally accepted.<br />

Standard regulations, manuals and official guidelines are increasingly being implemented. These will greatly<br />

assist in ensuring the reliability of the test results through standardisation of procedures, techniques, level of<br />

facilities and control of this powerful technology. It is thought that these guidelines will continue to form the<br />

basis for accreditation, validation and demonstration of proficiency of testing laboratories.<br />

This presentation will describe some of the current and draft guidelines in the context of using nucleic acid<br />

amplification (in the form of polymerase chain reaction) as diagnostic tools in veterinary laboratories and as<br />

prescribed tests for international trade. Practical issues that may affect quality assurance, application and<br />

standardisation of these techniques, and are yet to be addressed by standard documentation, will be<br />

identified from a laboratory implementation perspective. These concerns include availability of reagents and<br />

equipment on an international basis, inter-changeability of reagents and equipment, interpretation of<br />

methodology, geographical variations in target organisms and reactivity, and variations in accreditation<br />

standards.<br />

The processes of PCR will be discussed with application of a HACCP-like approach to identifying critical QA<br />

points affecting accuracy, reproducibility, and contamination control. Critical control points will be identified<br />

that may contribute to problematic standardisation between laboratories. In this context PCR will be split into<br />

pre-processing; sample processing / nucleic acid extraction; reaction mix preparation; adding template<br />

material to reaction mix; running a reaction; post-amplification processes; and post-amplification analyses.<br />

Each of these stages will be discussed with identification of perceived critical control points that may<br />

contribute towards reproducibility and standardisation concerns between laboratories.


Introduction<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A REVIEW OF TRAINING IN MOLECULAR DIAGNOSTIC TECHNIQUES<br />

FOR AVIAN INFLUENZA IN SOUTH EAST ASIA<br />

L. I. Pritchard 1 , A. Foord 1 , T. Taylor 1 , A. Axel 1 , R. Lunt 1 , J. Hammond 1 ,<br />

C. Groocock 3 , L.H. Lauerman 2 and P.W. Daniels 1<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory (AAHL) Geelong, Australia<br />

2 P.O. Box 189 East Olympia, WA 98540-0189, USA<br />

3 1030 West Creek Ave Cutchogue NY 11935, USA<br />

The ability to control avian influenza (AI) is critically dependant on the speed and accuracy of both<br />

epidemiology and diagnosis. For this reason AusAid and APHIS/USDA funded projects were initiated to<br />

provide training for diagnosis and epidemiology to scientists from South East Asian countries. Starting in<br />

early 2006, training courses for molecular diagnosis of AI were developed and conducted in Australia,<br />

Indonesia, Philippines and Taiwan. Under these projects diagnostic staff were trained in epidemiological,<br />

serological and molecular techniques (including real-time qRT-PCR and conventional RT-PCR). The<br />

scientific participants involved came from Indonesia, Cambodia, Thailand, Philippines, Vietnam, Sri Lanka,<br />

India and Taiwan.<br />

Methods<br />

The setup and workflows for real-time PCR laboratories were the major focus of the technology transfer and<br />

training, however advice was also given with regards to other procedures such as biosecurity. The training<br />

was both in-house and multiple visits to each Indonesian DIC lab. The real-time RT-PCR techniques were<br />

designed at AAHL and run on Applied Biosystems 7500 real-time machines as described by Heine et al,<br />

2007.<br />

Results<br />

During this short period we were able to;<br />

1. Provide training to about 80 scientific staff from South East Asian countries, including Indonesia,<br />

Cambodia, Thailand, Philippines, Vietnam, Sri Lanka, India and Taiwan.<br />

2. Develop a proficiency test program in AI real-time RT-PCR for use in Indonesian labs.<br />

3. Implement a novel purchasing system for Indonesian labs to enable them to access reagents<br />

for molecular diagnosis of AI.<br />

4. Evaluate different real-time PCR methodologies and machines for AI diagnosis.<br />

Discussion and Conclusions<br />

This AusAid/USDA-funded project for the technology transfer of Avian Influenza (AI) real-time PCR tests was<br />

developed at the Australian Animal Health laboratory (AAHL). The project involved; an initial gap analysis of<br />

the facilities at eight laboratories in Indonesia, and training of South East Asian scientific staff at AAHL, in<br />

Indonesia and in Taiwan.<br />

Training for scientific staff from other South East Asian countries was also sponsored by the USDA – FAS, -<br />

APHIS, American Institute in Taiwan (AIT), AAHL/CSIRO and Animal Health Research Institute in Taiwan. In<br />

Indonesia a program was carried out between February and June 2007 for the diagnosis by real-time PCR of<br />

AI Type A, H5 and H7 viruses. Initially training was done at AAHL for scientific staff from Indonesia and<br />

proficiency test programs were developed and dispatched. Many factors were shown to affect the accuracy<br />

of molecular diagnosis with laboratory setup, workflows, quality controls, difficulties with reagents and the<br />

competency of the staff being the major focus areas. We also undertook a limited evaluation of the methods<br />

used for AI training carried out in Taiwan between May and June 2007. Scientists from Indonesia, India, Sri<br />

Lanka, Vietnam, Cambodia, Thailand, Philippines and Taiwan undertook an intensive, two week course<br />

which involved both real-time and conventional RT-PCR, serology and virus isolation.<br />

Findings revealed that the accuracy of molecular diagnosis can be significantly affected by a number of<br />

factors including sample workflows and storage conditions, but especially the methods used for nucleic acid<br />

extraction, cDNA synthesis and PCR amplification. We present the results of this evaluation and training.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT AND CONSOLIDATION OF AN EQA PROGRAMME FOR THE MOLECULAR<br />

DIAGNOSIS OF AVIAN INFLUENZA<br />

Halpin, K., Heine, H., Johnson M., Stevens, V., Trinidad, L., Davies, K. and Selleck, P.<br />

CSIRO<br />

Australian Animal Health Laboratory<br />

Background: In 2006, avian influenza (AI) TaqMan RT-PCR assays which were developed at the Australian<br />

Animal Health Laboratory were transferred from AAHL to several international and state veterinary<br />

diagnostic laboratories. In order to support the implementation of these assays and provide the laboratories<br />

with the ability to assess their capability to perform the assay to an outcome consistent with other<br />

laboratories performing the same test, a proficiency testing program was established.<br />

Methods: Participating laboratories were provided with protocols, technical information and support to<br />

implement the AI TaqMan assays. All laboratories implemented the Influenza Type A TaqMan assay and the<br />

H5 TaqMan assay. Over the next 8 months, four proficiency testing panels were distributed to ten<br />

laboratories, including state, national and international laboratories. Each panel contained blind coded<br />

samples representing a range of concentrations of the pathogen, as well as material derived from an<br />

uninfected source for test evaluation. Test results were analysed and critical parameters from the different<br />

instrument platforms were identified in consultation with the participating laboratories.<br />

Results: A national proficiency testing program containing high and low positive, negative and replicate<br />

samples has been implemented to monitor the performance of AI TaqMan RT-PCR. Management of this<br />

proficiency program has been transferred to the Australian National Quality Assurance Program (ANQAP).<br />

Standard operating procedures for these assays and test validation data collected from this program have<br />

been submitted to the Australian Subcommittee on Animal Health Laboratory Standards (SCAHLS).


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

HOW USEFUL ARE TEST KITS WHEN APPLIED IN DIFFERENT TARGET POPULATION: PRODUCER<br />

AND END-USER RESPONSIBILITIES FROM A DEVELOPING COUNTRY PERSPECTIVE<br />

Diagnostic kits provide the advantage of quality assured reagents combined in a package with proper quality<br />

assurance and result evaluation tools. Their defined fitness for purpose and diagnostic sensitivity/specificity<br />

help to design appropriate (field) studies and to calculate a predictive value for an individual result.<br />

In order to obtain valid results several prerequisites have to be met. It is the obligation of the producer to<br />

design the assay in a robust and rugged way, i.e. taking transport and storage problems, but as well<br />

laboratory and equipment conditions into account. Manuals must be suitable for proper test performance and<br />

include background information to help trouble shouting. The result interpretation strategy should be decided<br />

before the final test design, should be validated in the field and data made available and must compensate<br />

for intrinsic variations.<br />

The end user has to define the needed fitness parameters before selecting a kit, check whether he can<br />

collect specimen as required, is technically capable to perform accurately all steps, monitors continuously the<br />

performance and finally evaluate the results by statistic means to verify the validity of results.<br />

Unfortunately it is not easy for end users to obtain the necessary information and for producers to meet all<br />

requirements.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LABORATORY MANAGEMENT TECHNIQUES FOR RESPONDING TO CLIENT DEMANDS:<br />

DIAGNOSIS OF SWINE PNEUMONIA<br />

Mark M Williamson<br />

Gribbles Veterinary Pathology,<br />

1868 Dandenong Rd, Clayton, Victoria, Australia, 3168<br />

Clients require value from all diagnostic laboratory submissions. Accordingly, the two most important<br />

objectives of a diagnostic laboratory should be to provide accurate repeatable results and written reports that<br />

can be interpreted, understood and valued by the client. This talk discusses biosafety, biosecurity, education<br />

and communication techniques to improve sample quality and client satisfaction using examples of swine<br />

pneumonias as a case study.<br />

Clients submitting samples for Hemophilus parasuis, Actinobacillus pleuropneumonia (APP), Mycoplasma<br />

hyopneumoniae and viral pneumonias, from field necropsies, need clear instructions on the criteria for good<br />

quality sample collection, packaging, transportation and if transported by air, IATA regulations. Submitters<br />

are counselled in collection of potential zoonotic agents, such as Streptococcus suis type 2, which should be<br />

handled with care. Submitters are encouraged to contact the laboratory before submitting urgent samples or<br />

to discuss specific requirements for difficult cases. There needs to be discussion between the client and the<br />

laboratory about the purpose of the diagnostic submission: disease investigation, specific disease exclusion<br />

or disease surveillance. Communication between laboratory and client is essential to generate realistic<br />

expectations and understanding of the test requirements, purpose and test limitations. In particular,<br />

difficulties in communicating issues such as sensitivity, specificity, disease prevalence and measurement of<br />

uncertainty must be addressed. In swine pneumonias, examples of these differences include Mycoplasma<br />

hyopneumoniae antibody ELISA has a sensitivity of 47-60% and specificity of 97-100% and APP antibody<br />

ELISA, approximate sensitivity of 95% and specificity of 99%.<br />

Diagnostic laboratories should be able to provide the following: 1. The single and most important objective of<br />

a laboratory is to make a rapid and accurate diagnosis of pneumonia. 2. The ability to investigate cases of<br />

atypical or unusual clinical presentations of pneumonia beyond the initial diagnosis. Possibly identifying new<br />

agents or disease processes. 3. A prime objective of laboratories is to provide technical and strategic advice<br />

to clients, professional colleagues in other laboratories and to Government regulatory bodies. 4. Classical<br />

swine fever can cause an interstitial pneumonia, but has lesions in other organs. It is essential to consider<br />

that pneumonia cases should not be considered in isolation alone and that important systemic diseases can<br />

lead result in lung lesions. 5. A laboratory needs to be continually adopting new diagnostic methods and<br />

tests for the diagnosis of swine pneumonias. 6. Longevity and retention of staff should be an aim of the<br />

laboratory and is often a barometer of staff morale. Therefore recruitment of staff with the appropriate<br />

qualifications and with the attitude of working in a co-operative team environment is essential. A high<br />

caseload of swine pneumonia cases allows staff to develop and maintain their diagnostic skills. The<br />

provision of continuing professional development can be used to provide non-salary rewards to professional<br />

staff. 7. Laboratories should where applicable contribute to national surveillance programs that provide<br />

market assurance, export testing certification, and swine meat product integrity. 8. Laboratories need to hold<br />

accreditation by national and, where appropriate, international accreditation schemes, working with good<br />

laboratory practice and participating in external proficiency testing programs. These key management<br />

objectives can and should be assessed by external stakeholders surveys. Laboratories need to be<br />

continually improving their management and efficiency to provide the best possible advice to their clients or<br />

risk being 'out of touch' and underutilised.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1330 - 1500 Concurrent Session 1.5 - Diagnostic assays - ELISA & molecular<br />

COMPLETE SEQUENCE ANALYSES OF THE GENOME OF EPIZOOTIC HAEMORRHAGIC DISEASE<br />

VIRUS (EHDV): THE DESIGN OF DIAGNOSTIC ASSAYS.<br />

Anthony, SJ*; Maan, N; Maan, S; Attoui, H; Darpel, K; Mertens, P.P.C<br />

Introduction:<br />

EHDV is an infectious, non-contagious arbovirus. It is transmitted between domestic and wild ruminants via<br />

heamatophagus midges of the Culicoides spp and can cause clinical disease in both wild and domestic<br />

species. The objective of this study was to generate a sequence database for all ten genome segments of<br />

EHDV for molecular epidemiology studies, and for the design of improved methods of diagnosis.<br />

Methods:<br />

Full length sequences were generated for all 10 genome segments using the anchor-primer method<br />

developed by Maan, Rao et al (2007). Initial sequence was achieved using the anchor primer, and<br />

completed by gene-walking.<br />

Results:<br />

Full-length nucleotide sequence analyses and phylogenetic comparisons were completed for the entire<br />

genome of eleven EHDV reference strains. These included isolates from each known serotype, and different<br />

geographic origins (: www.iah.bbsrc.ac.uk/dsRNA_virus_proteins/ ReoID/EHDV-isolates.htm ). This<br />

represents the first complete sequence database for EHDV for molecular epidemiology studies. The<br />

sequences of genome-segments 2 and 6 (encoding outer-capsid protein VP2 and VP5) are discussed in<br />

another abstract. The level of variation and phylogeny in each genome segment was assessed. In many<br />

cases the results mirror groupings seen in the bluetongue viruses.<br />

Conserved sequences within specific genome segments were used to design EHDV species/serogroupspecific<br />

diagnostic assays (RT-PCR based). The assays targeting genome-segment 7 (encoding VP7, the<br />

outer core protein and major EHDV species/serogroup specific antigen) were used to identify EHDV from<br />

disease outbreaks in domestic cattle over the past 2-3 years, caused by EHDV-6 in Morocco, Algeria, and<br />

EHDV-7 in Israel. These diagnostic assays were evaluated for detection of diverse strains of EHDV, and for<br />

specificity using strains of other closely related Orbivirus species.<br />

Conclusion:<br />

RT-PCR assays targeting conserved genome segments were developed to detect and identify EHDV.<br />

Sequence analyses can also be used to indicate the geographic origins of an isolate.<br />

*Presenting author<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NEW APPROACHES IN MOLECULAR AND SEROLOGICAL DIAGNOSIS OF AFRICAN SWINE FEVER<br />

(ASF) ON NEW EMERGING VARIANTS.<br />

C. Gallardo 1, 2 , R. Bishop 2, D. M. Mwaengo 3 , E. Martin 1 , J. M. Macharia 4 , and M. Arias 1 .<br />

1 OIE Reference Laboratory for African Swine Fever, CISA-INIA, Valdeolmos, Madrid, Spain;<br />

2 International Livestock Research Institute (ILRI), Kabete, Nairobi (Kenya)<br />

3 Dep. Of Medical Microbiology, College of Health Sciences, University of Nairobi (Kenya).<br />

4 Central Veterinary Laboratory, Kangemi, Nairobi (Kenya)<br />

1. Introduction and Objectives<br />

African swine fever (ASF) is a highly contagious swine viral disease with very high mortality. ASF is prevalent in more than 20 sub-<br />

Saharan African countries, and since 1999, Italy was the only European country with reported outbreaks in Sardinia. However, on June<br />

2007, ASF outbreaks were described throughout Georgia and Armenia with a significant mortality. The source of infection seems to be<br />

from Eastern of Southern Africa. From this situation one of our main objectives has been focused on molecular characterization of<br />

emerging variants of ASFV, as well as to study the suitability of current serological diagnostic tools that enable the detection of<br />

antibodies to a wider range of ASF viruses. We report the molecular characterization on ASF isolates collected from last outbreaks in<br />

Kenya by a first genotyping sequencing of the gene encoding VP72 (Bastos et al., 2003) and VP54 antigenic proteins to place isolates<br />

into major subgroups, following by sub-typing analysing three variable regions of the ASFV genome marked by the presence of tandem<br />

repeats sequences (TRS). These dates were correlated with the serological results obtained from sera collected in the same outbreaks<br />

using the current serological ELISA tests.<br />

2. Material and methods<br />

ASF antibody detection; For the detection of antibodies the indirect ELISA and IB test described in the OIE Manual of diagnosis (OIE,<br />

2004) were used. The INGENASA ELISA commercial kit Ingezim PPA Compac (11.PPA k3) and the recombinant ELISAs previously<br />

described by Gallardo et al 2006, were also used for comparison.<br />

ASF virus detection; DNA was extracted from tissue, ticks samples homogenates and sera using a nucleic acid extraction kit<br />

(Nucleospin/ Machery-Nagel –Cultek) following the manufactures procedures. A PCR assay using the ASF diagnosis primers<br />

PPA1/PPA2 was used to confirm the presence of ASFV DNA (Agüero et al., 2003; Chapter 2.1.12 OIE 2004). 25 Kenyan ASFV isolates<br />

from domestic pigs and 10 Kenyan ASFV isolates from ticks were isolated, prior to characterization, after three passages in leukocytes<br />

as described previously (Malquimst W.A. et al., 1960).<br />

DNA sequencing and molecular characterization; p72 genotyping was achieved by PCR as described Bastos et al 2003. The complete<br />

gene encoding VP54 protein was amplifying by PCR using the primer set PPA89-PPA722 (Gallardo et al p.c). For amplifying the TRS<br />

by PCR, different set of primers were selected (Nix et al 2006). PCR products were excised and purified by Quiaex gel extraction<br />

(QUIAGEN) and sequenced using an automated sequencer 3730 DNA analyzer” (Applied Biosystems). Sequence alignment was<br />

performed with the CLUSTAL W package and the phylogenetic and molecular evolutionary analyses were conducted using MEGA 4.0.<br />

More than 100 ASF viruses available in our institute and in GENBANK of the XXII previously identified p72 genotypes were included for<br />

phylogenetic analysis purposes.<br />

3. Results.<br />

ASF antibody detection; a total of 105 sera collected in Kenya were analyzed, 86 obtained from healthy domestic pigs in a surveillance<br />

programme performed in Nairobi’s slaughterhouse (2005) and 19 from the last outbreaks occurred in 2006-2007. The results obtained<br />

analyzing 105 serum samples have been negative on antibody detection for all techniques.<br />

ASF molecular characterization; ASFV Kenyan isolates were first genotyped by partial p72 gene characterization. 13 ASFV Kenyan<br />

isolates collected in the last outbreaks occurred in 2006-2007 were classified into genotype IX which comprises exclusively domestic pig<br />

strains. However, ASFV Kenyan viruses from ticks (10) and from sera (12) collected in a surveillance programme performed in Nairobi’s<br />

slaughterhouse (2005) were placed into genotype X associated with a sylvatic cycle. The same result was obtained sequencing the<br />

complete gene that encodes the p54 protein. Although the p72 and p54 genes are useful for identifying the major genotypes, higher<br />

resolution of virus relationships is required to uncover epidemiological links. To this end, sequences of three variables regions<br />

characterized by the presence of TRS were generated from Kenyan isolates. The results obtained from ASFV Kenyan isolates<br />

collected in the outbreaks occurred in 2006-2007 showing a high degree of homology with the last viruses characterized in Uganda in<br />

2003 placing in the same group. However between the ASFV isolates characterized in this study associated to p72 genotype X, a high<br />

variability was found, revealing different groups between Kenyan isolates due to the absence of TRS.<br />

4. Discussion and Conclusions<br />

ASF serological surveillance performing in Kenya has shown low seroprevalence of seropositive pigs in parallel to the high incidence of<br />

ASF virus-positive animals. These findings seems to be related with the results obtained typing the Kenyan isolates reflecting the cocirculation<br />

of different variants of ASFV in Kenya with a significant variability in antigenic proteins used as antigens in ELISA tests like<br />

VP72 or VP54. The molecular characterization on ASFV Kenyan isolates may suggest the presence of two different epidemiological<br />

patterns. The first one is associated with the last outbreaks occurred in Kenya where pig-to-pig cycle is present exemplified by p72<br />

genotype IX with a high degree of homology with Ugandan isolates. This fact suggests that the disease may have been introduced<br />

recently in Kenya-Uganda border district either through swill from boat or plane, through pork products, or through live pigs. A genetic<br />

feature of a domestic pig cycle appears to be a pronounced lack of genetic variation analysing the TRS with most isolates being<br />

identical to each other. The other epidemiological pattern is related with the ASFV Kenyan isolates obtained from healthy pigs and ticks<br />

placed into a p72 genotype associated with a sylvatic cycle. In this genotype a high intragenetic variability has been demonstrated<br />

analysing TRS suggesting that the disease has been maintained in Kenya in an endemic form between argasid ticks, wild or domestic<br />

pigs. Both the sylvatic and domestic pig cycles appear to play an important role in the epidemiology of ASF in Kenya with multiple<br />

genotypes within country that must be considered to develop a robust ASF antibody detection method.<br />

5.References<br />

1. Bastos A.D.S. et al., (2003). Arch Virol 148 (4), 693-706.<br />

2. Agüero M. et al, (2003). J Clin Microbiol, Sept p. 4431-4434.<br />

3. Malmquist and Hay (1960). Am. J. Vet. Res. 21, 104-108.<br />

4. Nix et al., (2006). Arch Virology Dec; 151(12):2475-94.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

RAPID HIGH-THROUGHPUT REAL-TIME RT-PCR TESTING FOR PRRSV:<br />

PROBLEMS AND SOLUTIONS<br />

*K. Harmon, A. Chriswell, K. Behrens, R. Hansell, K-J. Yoon, Veterinary Diagnostic Laboratory,<br />

Iowa State University, Ames, IA<br />

Introduction<br />

Use of real-time PCR has escalated due its sensitivity, specificity and rapidity. At ISU-VDL, porcine<br />

reproductive and respiratory syndrome virus (PRRSV) RT-PCR increased from 1685 tests in 1999 to almost<br />

30,000 in 2006. Increasing sample throughput without sacrificing test quality has been a major goal.<br />

Another objective has been improving our PRRSV RT-PCR test, as we encounter strains that are not<br />

recognized by our primer/probe set, resulting in false negatives, due to a high viral mutation rate. To<br />

address these issues, we have assessed different extraction methods for higher test capacity, and have<br />

evaluated a commercially available kit for real-time RT-PCR (rRT-PCR) testing for PRRSV.<br />

Material & methods<br />

RNA extractions were performed using the QIAamp ® Viral RNA mini kit (Qiagen) or the Applied<br />

Biosystems/Ambion MagMAX ® Viral RNA kit, manually and in conjunction with the Kingfisher 96 ® Magnetic<br />

Particle Processor (Thermo Scientific). The rRT-PCR was performed with ISU’s in-house ORF7 assay,<br />

using TaqMan Fast PCR Universal Mastermix coupled with Multiscribe Reverse Transcriptase and RNase<br />

inhibitor (all from Applied Biosystems) or with the AgPath-ID ® NA/EU PRRSV Multiplex Reagent kit (AB<br />

Ambion). Reactions were run on the Cepheid ® SmartCycler or Applied Biosystems 7500 Fast ® instrument.<br />

Confirmatory gel-based RT-PCR was performed using the first-stage of the gel-based assay (ORF7 target)<br />

described by Christopher-Hennings et al (1995).<br />

Results<br />

Comparison between column and magnetic bead extraction on serum, lung and nasal swab for PRRSV virus<br />

indicated that the latter method consistently performed as well as, or better than, single column extraction,<br />

yielding comparable or, in most cases, lower Ct values. We also evaluated use of the Kingfisher ® Magnetic<br />

Particle Processor in conjunction with the magnetic bead kit (n=89). Ct values were, in the majority of cases,<br />

equivalent or lower with the extracts obtained with the use of the magnetic particle processor, and RNA<br />

extraction time for 96 samples decreased to less than 1.5 hours.<br />

Results from the AgPath-ID ® kit were compared to those from our in-house PRRSV rRT-PCR test and a<br />

previously published gel-based PRRSV RT-PCR test. Initially, all samples (n = 175) were tested with both<br />

real-time assays. Samples yielding discrepant results were subjected to gel-based RT-PCR. All samples<br />

that were positive with the ISU rRT-PCR were also positive by the AgPath-ID ® kit (n = 66). However, ISU<br />

rRT-PCR was negative on some samples which were positive by the AgPath-ID ® kit (n=38). In all clinical<br />

cases where the AgPath-ID ® kit detected PRRS viral RNA but the ISU real-time assay did not (n = 12),<br />

PRRSV RNA was confirmed with the gel-based RT-PCR assay.<br />

Discussions & conclusions<br />

A purported benefit of real-time PCR is its high degree of specificity, which may be an advantage but can be<br />

a drawback to rRT-PCR testing for certain agents. In the case of RNA viruses, which undergo a high<br />

mutation rate, targets that were chosen because of their highly conserved nature can also undergo changes<br />

and fail to be recognized by the primer/probe set. Several times since implementing rRT-PCR testing for<br />

PRRSV, our laboratory has needed to redesign the primer/probe set to compensate for the genetic changes<br />

we have observed in PRRSV field strains. In this study, we evaluated the AgPath-ID ® NA/EU PRRSV<br />

Multiplex Reagent kit (Applied Biosystems Ambion), which detects both North American and European<br />

PRRSV strains simultaneously. This kit contains 2 different primer/probe sets for each of the PRRSV<br />

genotypes, enhancing detection of viral field strains.<br />

Coupling this kit with a 96-well magnetic bead extraction and magnetic particle robot allows our laboratory to<br />

increase sample capacity, decrease sample preparation time (total time required for testing 96 samples is<br />

less than three hours) and maintain test integrity. Use of this technology has enabled our laboratory to meet<br />

the demands of our clients, generating rapid, high quality results from large sample numbers.<br />

References<br />

Christopher-Hennings, J. et al., 1995. Detection of Porcine Reproductive and Respiratory Syndrome Virus in<br />

Boar Semen by PCR. J Clin Microbiol. 33(7): 1730-1734.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT AND INITIAL EVALUATION OF A LATERAL FLOW DEVICE FOR THE RAPID<br />

DETECTION OF CONTAGIOUS BOVINE PLEUROPNEUMONIA.<br />

R. D. Ayling 1* , C. P. Churchward 1 , J. Flint 2 , C. Danks 2 , I. Humbert 2 , E. Hashem 2 , R. A. J. Nicholas 1 .<br />

1 Mycoplasma Group, Statutory and Exotic Bacterial Diseases Department, Veterinary Laboratories Agency, Woodham Lane,<br />

Addlestone, Surrey. KT15 3NB.<br />

2 Foresite diagnostics, Central Science Laboratory, Sand Hutton, York YO41 1LZ.<br />

Introduction<br />

Mycoplasma mycoides subspecies mycoides small colony type (MmmSC) is the causative organism of<br />

contagious bovine pleuropneumonia (CBPP), which is an economically important disease that is endemic to<br />

many sub-Saharan African countries. In Africa mortality rates of cattle affected by CBPP are typically<br />

between 10-70% and it is highest in newly affected regions (Egwu et al., 1996). Many factors affect the<br />

failure to control the disease. Nomadism and the trekking of trade cattle together with economic weakness<br />

and political and military disturbances have made movement restrictions and other control measures difficult.<br />

Vaccination campaigns have generally been unsuccessful (Rweyemamu and Benkirane, 1996). There are<br />

also a lack of diagnostic tools and an epidemiological surveillance network. Currently diagnosis is based on<br />

a complement fixation test (CFT) or a cELISA, both of which are known to have reasonable specificity but<br />

low sensitivity. These tests also need to be performed by skilled technicians in a laboratory that can often be<br />

hundreds of kilometres away from an outbreak. Serological diagnostic tests that can be performed near to<br />

the animal would help with the diagnosis of CBPP thus facilitating action that may help towards controlling<br />

the spread of CBPP. We report on the development of a lateral flow device (LFD) that will support the rapid<br />

diagnosis of CBPP in ‘the field’.<br />

Material & methods<br />

Earlier and ongoing work has focussed on the production of a rapid latex agglutination test (LAT) (Ayling et<br />

al., 1999). The LAT was developed using a carbohydrate extract from MmmSC and has been successfully<br />

used in ‘the field’ and in the laboratory on whole blood and serum. A further development of this test using a<br />

similar antigen is the LFD. The test utilises similar latex technology but includes the test and control within<br />

one slide. Serum or blood samples are diluted in a prepared buffer and 75 µl is added to the slide. Within a<br />

few minutes a single line develops if the sample is negative and two lines if the sample is positive. These<br />

lines can easily be seen but the LFD can also be read using a handheld battery operated device that will give<br />

an unambiguous reading. The test has been optimised using CBPP positive and negative samples as well<br />

as potentially cross-reacting Mycoplasma bovis positive serum. This test has been evaluated in the<br />

laboratory and is currently being tested in Africa.<br />

Results<br />

Using a bank of CBPP positive and negative control serum, the LFD has given comparable results to those<br />

obtained using other serological tests. In some cases detecting infection earlier and for longer periods in<br />

experimentally infected animals than the CFT and cELISA. Tests on whole blood have also been<br />

successful, giving the same results to those obtained from serum.<br />

Discussions & conclusions<br />

The LFD test is easy to perform, stable, relatively cheap and quick with results being obtained within a few<br />

minutes. Use of the electronic reader gives a % titre in comparison with the anti-species control line, thereby<br />

giving further opportunity to quantify the test to give defined cut-off points. This LFD rapid pen-side test has<br />

the potential to resolve the problems of diagnosing CBPP in remote areas of Africa.<br />

References<br />

Ayling, R. D., Regalla, J., Nicholas, R. (1999). A field test for detecting antibodies to Mycoplasma mycoides<br />

subsp. mycoides small colony type using the latex slide agglutination test. In: COST 826 Agriculture and<br />

biotechnology. Mycoplasmas of ruminants: pathogenicity, diagnostics, epidemiology and molecular genetics.<br />

(Stipkovits, L., Rosengarten, R., Frey, J. eds.) Vol III pp.155-158 European Commission, Brussels.<br />

Egwu, G. O., Nicholas, R. A. J., Ameh, J. A., Bashiruddin, J. B. (1996). Contagious Bovine<br />

Pleuropneumoniae: An Update. Veterinary Bulletin. 66: 9, 875-888.<br />

Rweyemamu, M. M., Benkirane, A. (1996). Global impact of infections with organisms of the “Mycoplasma<br />

mycoides cluster” in ruminants. In: COST 826 Agriculture and biotechnology. Mycoplasmas of ruminants:<br />

pathogenicity, diagnostics, epidemiology and molecular genetics. (Frey, J. Sarris, K. eds) pp. 1-11. European<br />

Commission, Brussels.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1330 - 1500 Concurrent Session 2.5 - Biosecurity, IT, & LIMS<br />

BENEFITS REALIZED FROM THE FIRST BIOSAFETY LEVEL 3 LARGE ANIMAL NECROPSY FACILITY<br />

IN A STATE DIAGNOSTIC LABORATORY IN NORTH AMERICA<br />

S. D. Fitzgerald 1 and W. M. Reed 2<br />

1 Diagnostic Center for Population & Animal Health, Department of Pathobiology & Diagnostic Investigation, Michigan State University,<br />

Lansing, MI 48910 USA<br />

2 School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907 USA<br />

Our objective is to illustrate the benefits realized from opening the first Biosafety Level 3 (BL3) large animal<br />

necropsy facility in any state diagnostic laboratory in North America. In the spring of 2004 the DCPAH was<br />

completed with $58 million funding from the state legislature of Michigan. The need for this facility grew out<br />

of the recognition of the increasing importance of zoonotic and emerging diseases in animals. Specifically<br />

the diseases of interest included bovine tuberculosis, chronic wasting disease, and neurologic viruses such<br />

as West Nile virus, eastern equine encephalomyelitis, and rabies infections. The DCPAH houses all current<br />

diagnostic disciplines such as necropsy, histopathology and immunohistochemistry, bacteriology, virology,<br />

parasitology, nutrition, endocrinology, toxicology and immunodiagnostics; but also includes space for our<br />

partners in disease surveillance, the MI Departments of Natural Resources and Agriculture.<br />

During the last three and one half years of operation this 152,000 gross square feet facility has utilized each<br />

of its 5 necropsy floors, 4 biosecure cattle holding facilities, BL3 microbiology laboratory, incinerator, and<br />

liquid waste treatment equipment, to deal with several existing and emergent infectious disease problems.<br />

Features specific to BL3 will be illustrated and highlighted. Some of these features include double door<br />

entries for personnel and animals, coded keypads to prevent non-approved personnel access, HEPA filtering<br />

of incoming and outgoing air, negative air pressure in the BL3 areas relative to the rest of the facilities,<br />

treatment of waste water under specific conditions of temperature and pressure, and high temperature<br />

incineration to inactivate bacteria, viruses and prions. To date we have processed over one thousand cattle<br />

and tens of thousands of deer for bovine tuberculosis, thousands of deer for chronic wasting disease,<br />

thousands of sheep for scrapie, hundreds of neurologic horses, and thousands of birds and wild mammals<br />

for West Nile virus. In addition to improved bio-security for protecting the health of our faculty, staff and<br />

veterinary students, this facility also has promoted a closer working relationship with our government<br />

partners resulting in rapid recognition and response to newly emerging disease conditions.<br />

In summary, these new BL3 level laboratory facilities have resulted in greater safety for workers, greater<br />

containment of infectious agents, as well as providing for a closer and better working relationship amongst<br />

the various animal disease control groups in the state of Michigan.<br />

Wed 14 November<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LINKING MULTIPLE VETERINARY LABORATORY INFORMATION MANAGEMENT SYSTEMS TO<br />

SUPPORT THE NATIONAL ANIMAL HEALTH LABORATORY NETWORK<br />

The National Animal Health Laboratory Network (NAHLN) currently involves more than fifty veterinary<br />

diagnostic laboratories throughout the United States of America and is dependent on real time reporting of<br />

diagnostic test results using standardized and secure techniques. This presentation will focus on the history,<br />

requirements analysis and current design of the information technology infrastructure which supports the<br />

NAHLN and the challenges we have worked through to implement a scalable network and database system<br />

which allows diverse laboratory information systems to provide timely and consistent data.<br />

Four years ago, a small group of veterinary diagnostic laboratories representing the American Association of<br />

Veterinary Diagnostic Laboratories (AAVLD) worked collaboratively with the USDA APHIS VS National<br />

Veterinary Service Laboratories (NVSL) and the USDA APHIS VS Centers for Epidemiology and Animal<br />

Health (CEAH) to conceptualize an approach to sharing data. The group envisioned an approach to<br />

aggregating information using processes independent of the type or version of Laboratory Information<br />

Systems in use by veterinary diagnostic labs, and establishing standards which would allow analysis of the<br />

data.<br />

The concept was proven successful in a pilot project and lessons learned from the pilot project and<br />

application of the approach to an on-going surveillance program were used as the basis for implementing the<br />

first version of the NAHLN Information Technology System.<br />

The design and lessons learned from the implementation of the information technology system which now<br />

supports the National Animal Health Laboratory Network are independent of factors specific to the United<br />

States and will be shared through this presentation with the anticipation that that they will be beneficial to<br />

other countries. Anticipated future functionality will also be identified.<br />

It is also hoped that this same approach may lead to the possibility of the real time sharing of Veterinary<br />

Diagnostic Laboratory Information internationally in the near future. With that as a potential goal, benefits of<br />

such sharing of real time international data will also be identified.


World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

IMPLEMENTATION, FUNCTION AND BENEFITS OF A LABORATORY INFORMATION MANAGEMENT<br />

SYSTEM IN A NATIONAL VETERINARY LABORATORY SERVICE.<br />

Author: *Paul M. Collery, Irish Department of Agriculture, Fisheries and Food, Veterinary Laboratory Service.<br />

Background<br />

The Irish Veterinary Laboratory Service (VLS) - a division of the Department of Agriculture, Fisheries and<br />

Food (DAFF) - comprises a Central Veterinary Research Laboratory (CVRL) at Backweston, Co. Kildare and<br />

six Regional Laboratories at strategic locations throughout the country. The VLS provides laboratory support<br />

for national animal disease control and surveillance schemes – as well as a veterinary diagnostic pathology<br />

service to the farming community.<br />

In May 2001, the VLS commenced a process of moving an almost entirely paper-based submission<br />

recording and reporting system to a proprietary electronic Laboratory Information Management System<br />

(LIMS). The LIMS now encompasses the entire laboratory network and provides electronic tracking of all<br />

submission information from sample receipt, through test assignment and result entry, to reporting and data<br />

retrieval and analysis. This presentation comprises a description of the functions, scope, management and<br />

benefits of the LIMS - with particular reference to the challenges and complexities of its implementation in a<br />

multi-discipline veterinary laboratory environment - as well as to the opportunities provided for enhanced<br />

support of national animal disease surveillance activities.<br />

Scope<br />

This is a large system supporting about 160 users – at six geographically distinct locations - and<br />

electronically mapping the over 800 tests available from the Laboratory Service repertoire. The system<br />

manages a wide range of submission types – from high volume surveillance samples to carcass and other<br />

animal materials for pathology and microbiology investigations. LIMS workflows map the movement, receipt<br />

and testing of submissions through the VLS. Result recording ranges from manual input text-intensive<br />

morphological pathology – to automated transfer from a range of instruments. Currently about 30<br />

instruments have direct connections to the LIMS - allowing electronic transfer of test assignment and result<br />

data in the areas of microbiology, serology, PCR, haematology, biochemistry and genotyping. The LIMS also<br />

has direct links with two other DAFF animal health databases. These provide automated submission login<br />

and result reporting for the TB eradication and scrapie genotyping programs. Client invoicing is facilitated<br />

through connection to the Department’s financial accounting system.<br />

Implementation<br />

The specific LIMS application was selected in 2001 by international tender. The most significant challenges<br />

during project implementation reflected the diverse nature of veterinary diagnostic pathology procedures -<br />

and the related networking issues consequent upon high data transmission requirements in a multi-site<br />

environment. Fitting the LIMS to meet requirements involved a combination of close user involvement in<br />

initial design workshops, as well as a substantial amount of work by the suppliers on workflow development<br />

and report design.<br />

The first phase of the implementation – covering the CVRL (then located at Abbotstown, Co. Dublin) and one<br />

Regional Laboratory at Athlone - identified line-speed issues which were ultimately addressed by distributing<br />

the application to users as a ‘thin-client’ terminal service – rather than by the initial approach of direct<br />

installation on users’ PCs. While ultimately successful – allowing roll-out of the LIMS to all Regional<br />

Laboratories in 2003 - the solution introduced some of its own problems which have been far from trivial in<br />

impact and resolution. The use of ‘thin clients’, however, greatly facilitated the transfer of CVRL activities<br />

from Abbotstown to a purpose-built laboratory campus at Backweston in 2005/2006. Despite the size and<br />

complexity of the LIMS, all internal support to date has been by the VLS’ own users. Underlying operating<br />

system, hardware and networking support is provided by DAFF IT Division.<br />

Benefits<br />

The overall experience of the implementation of a LIMS has been extremely positive – both for users and the<br />

VLS as a whole. Time savings in areas with repetitive data recording have been substantial. Any<br />

disadvantages of increased PC-usage in areas such as post-mortem rooms and ‘wet’ laboratories are<br />

regarded as being heavily outweighed by improved data access and reporting. The need to ensure data<br />

quality and consistency has also encouraged standardisation of laboratory methodology in several areas of<br />

diagnostic pathology.<br />

Probably the greatest benefits at VLS-level derive from enhanced disease surveillance opportunities. While<br />

these are only in the early stages of exploitation, the LIMS has already facilitated preparation of Regional<br />

Laboratory surveillance reports - as well as national input to EU-wide zoonotic and Class A animal disease<br />

reporting.<br />

Looking to the immediate future, the potential for linkages with other DAFF national animal databases and<br />

farm mapping systems should assist in the establishment of more accurate baseline data on the occurrence<br />

and distribution of indigenous diseases in farm animals in Ireland.<br />

Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

Wed 14 November


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

BEYOND THE LABORATORY GATE – ENHANCING A LABORATORY INFORMATION MANAGEMENT<br />

SYSTEM (LIMS) FOR EXOTIC ANIMAL DISEASE PREPAREDNESS<br />

P. A. Durr, I. Zupanovic, J. Watson, L. Wright and M. Johnson<br />

Australian Animal Health Laboratory (AAHL), 5 Portarlington Road, East Geelong, Victoria, Australia.<br />

INTRODUCTION<br />

The UK FMD epidemic in 2001 exposed the importance of data management for effective exotic animal<br />

disease (EAD) preparedness, and in response many laboratories have introduced or upgraded their<br />

laboratory information management systems (LIMS). However, many commercial LIMS are inward-looking<br />

applications, and issues and problems remain in establishing effective communication with other laboratories<br />

and EAD management systems (EADMS). This is the current situation in Australia, where the pending<br />

introduction of such an EADMS (BioSIRT) requires that the exotic disease laboratory, the Australian Animal<br />

Health Laboratory (AAHL), develop a solution to enable the electronic submission of high volumes of data in<br />

the event of an EAD incursion and to permit reporting of results direct to the BioSIRT database. Here we<br />

describe progress made in resolving this problem, with the design of a custom built interfacing application,<br />

“STARS”, which is an acronym for Submission, Tracking And Reporting System.<br />

DESIGN OF STARS<br />

The design phase of STARS followed a requirements phase, when the underlying “business” of sample<br />

processing was carefully analyzed. Initially a design which made use of AAHL’s current commercial LIMS<br />

application, SampleManager, was explored, but the complexity of this implementation plus security concerns<br />

argued for an interfacing application specialized upon the handling of submissions and reporting. According<br />

to this design, AAHL’s sample processing will be split between two applications, with SampleManager<br />

handling specimen processing (leading to a test result) and STARS handling submission processing (leading<br />

to a client report).<br />

To enable submission and reporting from any client, STARS will be in essence a web-enabled database (1) ,<br />

with an architecture following closely that recommended by Microsoft within their .NET framework. The user<br />

interface is designed to be both intuitive and work-flow orientated, aiming to guide users through tasks.<br />

Accordingly, it makes extensive use of Web 2.0 technologies, such as AJAX, to provide users with<br />

enhancements, such as auto-suggestion and federated access. For data transfer, STARS uses XML<br />

technology to ensure encrypted security and permit direct database to database delivery of data.<br />

DISCUSSION<br />

At first impression, it might seem that splitting AAHL’s sample processing across two applications might be<br />

adding unnecessary complexity, and a solution based upon SampleManager would be preferable. However,<br />

the split architecture has a number of advantages, including removing the need for the current complex<br />

customization of SampleManager required to disaggregate submissions (“accessions”) into samples and<br />

then re-aggregate the test results back for reports. Accordingly, SampleManager will be able to be treated as<br />

a COTS (“commercial off-the-shelf”) application, minimizing the cost when version upgrading occurs. Added<br />

to the fact that STARS will be using industry common standards, and thus readily understood by a large<br />

potential pool of IT professionals, then AAHL will be minimising the risk of a failure of its LIMS systems<br />

during an EAD. A further benefit of the interfacing solution is that it will be adaptable to other veterinary<br />

laboratories within Australia, including those not using SampleManager. Accordingly the possibility for<br />

creating a true animal health laboratory network for Australia, with direct sharing of results, will move a step<br />

closer to realization.<br />

REFERENCE<br />

Durr, P.A. and Eastland, S. (2004). Use of web-enabled databases for complex animal health investigations.<br />

Revue Scientifique et Technique de L Office International Des Epizooties 23, 873-84.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE RELAIS PROJECT: AN EPI-INFORMATICS CONSTELLATION<br />

P-M Agapow*, Institute for Animal Health<br />

J. Bashiruddin, Institute for Animal Health<br />

Introduction<br />

Global disease control presents an information management problem. Routine epidemiological work<br />

generates vast amounts of data in various forms: outbreak reports, clinical samples, the results of laboratory<br />

analyses, genetic sequences, phylogenetic trees. Much of this data must in turn be distributed in a timely<br />

and secure fashion to scientists, field personnel and decision makers. Unfortunately and unremarkably,<br />

disease data currently tends to be dispersed over a variety of information systems, using incompatible<br />

formats. Where data sharing takes, it is on an ad hoc basis.<br />

Material & methods<br />

To this end, we have developed ReLaIS: the Reference Laboratory Information System. This loosely coupled<br />

constellation of software systems and databases allows allied laboratories to gather, query, visualise and<br />

share epidemiological data in semi-automated manner. Rather than force workers to adopt their style and<br />

data to a common format, and workflow the ReLaIS system instead makes it easy for data to be imported<br />

and communicated between systems. Analyses are controlled and visualised via web browser (TTW:<br />

through the web), so that remote workers may manipulate data just as easily as those in the lab, in a<br />

standardised environment.<br />

ReLaIS is based upon a stack of free open source software, including the programming language Python,<br />

bioinformatics library BioPython, the MySQL database, the content management system Plone for web<br />

presentation and geospatial visualisation via OpenLayers. The choice is not ideological, but practical: By<br />

adopting widely used FOSS tools, we ensure that development time is lowered, individual components have<br />

been rigorously tested, any software developed can be redistributed without problems and there exists a<br />

large pre-existing body of expertise for development and deployment. Finally, low cost is an obvious<br />

attraction, especially where systems may need to deployed in impoverished areas.<br />

RelaIS also includes Amergin, a web-based platform for storing, editing and analysing molecular<br />

epidemiological data. By working through-the-web, operators gain a consistent analysis environment where<br />

expensive computations are carried out on a remote server and data is stored consistently and safely in a<br />

single location. Amergin engages with the ReLaIS workflow so that updated data flows to and from the<br />

system.<br />

ReLaIS has been designed to be flexible and extensible for easy adoption and customisation for different<br />

laboratories and pathogens (currently the FMDV world reference laboratory, and Bluetongue reference<br />

laboratory at IAH-Pirbright by the end of 2007). Further, it has been constructed from widely used, open<br />

source software for which there exists a wide pool of expertise for support and modification and limited<br />

developmental and set-up costs.<br />

Discussions & conclusions<br />

Future developments include expansion of data and analyses (e.g. molecular structural functions), and the<br />

introduction of a tracking facility for field workers and clients to follow the analysis of their samples in real<br />

time.<br />

References<br />

OPENLAYERS. 1994. Website. http://openlayers.org.<br />

BIOPYTHON. 1994. Website. http://biopython.org.<br />

PYTHON. 1994. Website. http://www.python.org.<br />

PLONE. Website http://www.plone.org.<br />

Wed 14 November


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1530 - 1700 Concurrent Session 1.6 - Molecular Diagnostic assys- II (Viral)<br />

HIGH THROUGHPUT DIAGNOSTIC PCR FOR BLUETONGUE VIRUS<br />

Katarzyna Bachanek-Bankowska*, Carrie A. Batten, Andrew Shaw, Peter P. C. Mertens and Chris A.L. Oura<br />

Institute for Animal Health, Ash Road, Pirbright, UK, GU24 0NF.<br />

Introduction:<br />

In August 2006, bluetongue disease (BT) was confirmed in the Netherlands and the outbreak rapidly spread<br />

to Belgium, Germany, Luxembourg and northern France. By late September 2007 over 26,000 premises had<br />

been confirmed to be infected in Northern Europe. On the 21 st of September 2007 samples from a Highland<br />

cow in the county of Suffolk in the UK were submitted to the lab and were found to be positive for BTV<br />

antibodies by C-ELISA and BTV RNA by real-time RT-PCR. Following this initial case further positive cases<br />

rapidly followed and surveillance sampling of animals in the control zone revealed BTV was present and<br />

circulating in South East England. In order to respond to the expected high throughput of samples we have<br />

developed a high throughput RNA extraction and real-time RT-PCR system for the detection of BTV.<br />

Material & methods and Results:<br />

Two RNA extraction robots (the 96 well Universal Qiagen robot and the 32 well Roche MagNA Pure LC<br />

robot), based on different chemistries, have been validated for high throughput RNA extraction from cattle<br />

and sheep blood and a real-time RT-PCR assay for the detection of BTV RNA has been developed and<br />

validated. Generic RNA extraction protocols are available from Qiagen and are widely used for the extraction<br />

of RNA from human blood however these protocols do not work for animal blood due to clogging of the<br />

filters. The most likely reason for this clogging is due to the higher protein content in animal blood compared<br />

to human blood. It has been especially difficult to develop a protocol for sheep blood which contains higher<br />

levels of protein than cattle blood. We have worked with Qiagen to develop and optimise protocols for the<br />

extraction of RNA from both cattle and sheep blood. For bovine blood it is necessary extract RNA from a<br />

reduced volume of 80µl of blood and a protocol is in the process of being optimised for RNA extraction from<br />

sheep blood and will be discussed. Due to the different chemistry used by the Roche MagNA Pure LC Robot<br />

we have found that this robot effectively extracts RNA from both cattle and sheep blood using the standard<br />

protocols. The Universal Qiagen robot is able to extract RNA from 96 samples taking approximately two<br />

hours whereas the Roche MagNA Pure LC Robot can extract RNA from 32 samples in 1.5 hours. Although<br />

the MagNA Pure robot has a reduced throughput compared to the Qiagen Universal robot we have found it<br />

very useful for extracting RNA from fewer samples as it can process up to 16 samples in 45 min. We have<br />

also found that throughput can be increased by pooling blood samples. Results show that pooling blood<br />

samples up to 1:10 does not significantly compromise the sensitivity of the assay.<br />

A real-time RT-PCR assay has been developed (Shaw et al., 2007) using primers and probes targeting<br />

genome segment 1. In the assay a combination of two sets of primers and two probes are used. Reports<br />

from other labs and our experience have revealed that the level of background fluorescence in the assay is<br />

very dependent on the quality/batch of primers used. In order to solve this background problem a single<br />

degenerate probe has been designed for use with the same primers. This modified assay has been fully<br />

validated and results in a reduced level of background.<br />

Discussions & conclusions:<br />

Both the Universal Qiagen and Roche MagNA Pure LC Robotic extraction systems have been used to<br />

increase diagnostic throughput. Protocols have been developed to enable both cattle and sheep blood to be<br />

extracted by the Qiagen robot and the advantages and disadvantages of both systems will be discussed. A<br />

new real-time RT-PCR assay has been developed for the detection of BTV RNA that reduces the level of<br />

background fluorescence. Both the Qiagen Universal and the MagNA Pure RNA extraction robots have been<br />

used successfully in conjunction with real-time RT-PCR to process a high throughput of samples during the<br />

outbreak of BT in Northern Europe in 2006/2007.<br />

References<br />

A.E. Shaw, P. Monaghan, H.O. Alpar, S. Anthony, K.E. Darpel, C.A. Batten, A. Guercio, G. Alimena, M.<br />

Vitale, K. Bankowska, S. Carpenter, H. Jone, C.A.L. Oura, D.P. King, H. Elliott, P. Mellor and P.P.C<br />

Mertens. (2007) Development and initial evaluation of a real-time RT-PCR assay to detect bluetongue virus<br />

genome segment 1. Journal of Virological methods, 145: 115-126.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NUCLEIC ACID DIAGNOSTIC TOOLS FOR EARLY DETECTION OF<br />

ARTHROPOD-BORNE ANIMAL VIRUSES<br />

W.C. Wilson, E.S. O’Hearn, R.J. Lenhoff, B. Hindson, C. Torres, D.E. Stallknecht, D.G. Mead, C.Y. Kato, R.T. Mayer, B.S. Drolet, and<br />

J.O. Mecham<br />

USDA, ARS, Arthropod-Borne Animal Diseases Research Laboratory, Laramie WY (Drolet, Harpster, Kato, O’Hearn, Mayer, Mecham,<br />

Wilson); Lawrence Livermore National Laboratories, University of California, Livermore CA (Hindson, Lenhoff, Torres); Southeastern<br />

Cooperative Wildlife Disease Study, College of Veterinary Medicine, The University of Georgia, Athens, GA (Stallknecht, Mead)<br />

Introduction: The objective of this paper is to provide a review of our strategy for developing nucleic acid<br />

diagnostic tools for existing and emerging arthropod-borne animal viral diseases. The outbreak of West Nile<br />

virus in the United Sates and the recent outbreak of Rift Valley fever (RVF) virus in East Africa have<br />

highlighted the importance of validated early detection tools for arthropod-borne animal viral diseases. The<br />

Arthropod-Borne Animal Diseases Research Laboratory has been involved with the development of rapid<br />

nucleic acid detection tests for bluetongue virus (BTV) and the related epizootic hemorrhagic disease virus<br />

(EHDV). Bluetongue virus causes disease in sheep and cattle and has significant economic impact due to<br />

trade barriers. Although U.S. EHDV strains have not been experimentally proven to cause clinical disease in<br />

cattle, there is serologic evidence of widespread infection. Vesicular stomatitis, caused by vesicular<br />

stomatitis virus (VSV) is also an important arthropod-borne livestock disease because of its clinical<br />

resemblance to foot and mouth disease. In addition, the Rift Valley fever (RVF) outbreak has prompted the<br />

development of new and the evaluation of existing diagnostic assays on livestock samples.<br />

Materials and methods: A common approach has been used for all of these arboviruses. A target gene is<br />

selected based on existing virological data and known degree of genetic conservation. Additional<br />

phylogenetic studies are conducted where limited sequence data is available. Bioinformatic software is<br />

utilized to predict the most likely polymerase chain reaction (PCR) primer/probe set of signatures to be<br />

successful. Laboratory evaluation is done using standardized protocols for real-time quantitative reverse<br />

transcriptase-PCR qRT-PCR. The assays are then evaluated by collaborating diagnostic laboratories.<br />

Results: We have developed (qRT-PCR) tests that detect RNA from indigenous and exotic strains of BTV<br />

and EHDV based on sequencing of multiple target genes. The EHDV qRT-PCR detected all 40 field strains<br />

available. Additionally, 105 archived sheep, cattle, deer and other antelope samples were tested. The<br />

assay detected 90% of previously virus isolation-positive clinical samples which was 12% better than our<br />

previously published EHDV PCR protocols. The samples missed by the assay are being re-evaluated to<br />

determine if the sample was degraded. The BTV qRT-PCR detected 43 indigenous and exotic field strains<br />

available using a two probe detection assay. A similar design strategy reported in another presentation was<br />

used to develop a qRT-PCR for VSV. This assay is proving useful in determining potential transovarial<br />

transmission of VSV in Culicoides sonorensis biting midges. The existing RVF qRT-PCR assays have been<br />

combined into a multiplex assay utilizing a previously described RNA template control.<br />

Discussion and conclusion: The standardized approach to use a Phylogenetic/bioinformatics analysis to<br />

design qRT-PCR assays for arboviruses has proven to be successful. A similar approach has been<br />

successful in developing qRT-PCR assays for these viruses by other researchers as well. The next<br />

generation of nucleic acid diagnostic tests will include multiplex or multi-target assays either to compensate<br />

for genetic variation and to differentiate pathogens that cause similar clinical signs, or differentiate species of<br />

a specific pathogen. Such assays are currently in development at Lawrence Livermore National<br />

Laboratories where they have incorporated qRT-PCR into single multiplex Luminex assays for multiple<br />

related pathogens. These assays have undergone a multiple laboratory field evaluation study that<br />

demonstrated the potential of this technology. ABADRL researchers are also developing non-enzymatic<br />

nucleic acid hybridization detection assay using fluorescence signal amplification and Raman spectroscopy<br />

technologies. In conclusion, a number of new nucleic acid tests have been and are being developed for early<br />

detection of arboviral animal pathogens, and are, or soon will be, available to veterinary diagnostic<br />

laboratories. The concept of simultaneous detection of multiple genomic targets for a single pathogen will<br />

help to ensure detection of RNA viruses which are known to be highly variable genetically.<br />

References:<br />

Drosten et al., J. Clin. Microbiol. 40, 2323-2330. 2002.<br />

Garcia et al., J. Clin. Microbiol. 39, 4456-4461. 2001.<br />

Moniwa et al., J. Vet. Diag. Investig. 19, 9-20. 2007.<br />

Shaw et al., 2007. J. Virol. Methods 145, 115-126.<br />

Wed 14 November


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE DEVELOPMENT OF METHODS FOR THE DETECTION OF NEWCASTLE DISEASE VIRUS<br />

Carmel Hancock, David Townsend, Debby Cousins<br />

AB-CRC, Curtin University of Technology, Department of Agriculture & Food, Western Australia<br />

Objectives<br />

There are many infectious agents that require fast and reliable detection methods so as to prevent<br />

epidemics. The aim of this project is the development of techniques for the detection of Newcastle Disease<br />

Virus (NDV).<br />

Key Messages<br />

While both methods employ real-time PCR, the first is aimed at the detection of viral RNA. The fusion<br />

cleavage site of the NDV genome is a molecular determinant of virulence of the virus and was targeted in the<br />

project. The use of LightUp probes to detect this sequence in both a real-time PCR and a field setting is<br />

examined.<br />

The second method developed is known as immuno-PCR and combines the concepts of antigen/antibody<br />

detection via ELISA with the targeting of specific nucleic acid sequences in PCR. This project examines<br />

some of the various methods of constructing a viable immuno-PCR system and the most efficient method of<br />

doing so for detecting NDV.<br />

Conclusion<br />

The use of LightUp probes for the detection of NDV encountered difficulties due to the nature of the fusion<br />

cleavage site but has also shown aspects that are very promising for a field based detection system.<br />

The combination of the two methods in the immuno-PCR allows the detection of the antibody at much lower<br />

levels than typically possible with a traditional ELISA.<br />

Stream: New Technologies and Platforms for Diseases Diagnosis<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MOLECULAR DETECTION OF BETANODAVIRUSES FROM SUBCLINICALLY INFECTED AQUARIUM<br />

FISHES AND INVERTEBRATES<br />

D. K. Gomez* 1 , S. C. Park 1,2,4 , G. J. Heo 3 , J. H. Kim 2,4 and C. H. Choresca Jr. 1,2,4<br />

1 KRF Zoonotic Disease Priority Research Institute, Seoul National University, Seoul 151-742, Korea<br />

2 Laboratory of Aquatic Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea<br />

3 College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju 361-763,<br />

Korea<br />

4 Brain Korea 21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea<br />

Introduction<br />

Viral nervous necrosis (VNN) or viral encephalopathy and retinopathy (VER) caused by betanodavirus<br />

(Nodaviridae) is a worldwide disease affecting several species of cultured marine fish including Korea. The<br />

present study was conducted to investigate asymptomatic infection of betanodavirus from aquarium fishes<br />

and invertebrates.<br />

Material & methods<br />

A total of 237 apparently healthy imported aquarium fish, marine (65 species) and freshwater (12 species)<br />

fishes and marine invertebrates (4 species), were collected from November 2005 to February 2006 in a<br />

commercial aquarium in Seoul, Korea. The brains of the fish and other tissues of the invertebrates were<br />

examined by reverse transcriptase polymerase chain reaction (RT-PCR) and nested PCR to detect the coat<br />

protein gene of betanodavirus and analyzed by phylogenetic tree.<br />

Results<br />

Positive nested PCR (420 bp) results were obtained from the brains of 8 marine and 2 freshwater fish<br />

species and 1 marine invertebrate species. Phylogenetic analyses based on the partial nucleotide sequence<br />

(177 bases) of the RNA2 coat protein gene of 11 strains were all identical to one another (93–100%) and<br />

highly homologous to the redspotted grouper nervous necrosis virus (RGNNV) genotype. The identity of the<br />

strains with the other 3 genotypes ranged only from 66.5-73.9%.<br />

Discussions & conclusions<br />

The results indicate that all detected samples imported from different countries regardless of host species<br />

and geographical areas were subclinically infected and genetically closely related, suggesting an importance<br />

of these asymptomatic carrier fish or invertebrate as natural hosts of betanodavirus.<br />

References<br />

1. DK Gomez, J Sato, K Mushiake, T. Isshiki, Y Okinaka and T Nakai (2004): PCR-based detection of<br />

betanodaviruses from cultured and wild marine fish with no clinical signs. Journal Fish Diseases 27:603-<br />

608.<br />

2. Dennis Kaw Gomez, Dong Joo Lim, Gun Wook Baeck, Hee Jeong Youn, Nam Shik Shin, Hwa Young<br />

Youn, Cheol Yong Hwang, Jun Hong Park, Se Chang Park (2006): Detection of betanodaviruses in<br />

apparently healthy aquarium fishes and invertebrates. Journal of Veterinary Science 7(4): 369-374.<br />

3.G.W. Baeck, D.K. Gomez, K. S. Oh, J.H. Kim, C.H. Choresca Jr. and S.C. Park (2007): Detection of<br />

piscine nodaviruses from apparently healthy wild marine fish in Korea. Bulletin of the European Association<br />

of Fish Pathologists 27(3): 116-122.<br />

Wed 14 November


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CHARACTERISATION OF THE HERPES-LIKE VIRUS INFECTING AUSTRALIAN POPULATIONS OF<br />

ABALONE HALIOTIS SPP. BY WHOLE GENOME PYROSEQUENCING<br />

*F. Wong 1 , M. Lancaster 1 , M. Crane 3 , S. Corbeil 3 , A. Hyatt 3 , J. Tan 1 , K. Savin 1 , N. Cogan 2 , T. Sawbridge 2 , S. Warner 1<br />

1 2<br />

Department of Primary Industries, Animal Health Sciences, 475 Mickleham Road, Attwood, VIC 3049 Australia; Department of Primary<br />

Industries, Victorian AgriBiosciences Centre, 1 Park Drive, Bundoora, VIC 3086; 3 CSIRO Livestock Industries, Australian Animal Health<br />

Laboratory, 5 Portarlington Road, Geelong, VIC 3220 Australia<br />

Introduction<br />

Mortalities of wild populations of abalone due to infectious ganglioneuritis continue to occur and spread<br />

along the Victorian coast, although the first reported outbreak of disease had affected farmed abalone<br />

Haliotis rubra, H. laevigata and hybrids in the state’s southwest (2). The likely cause is infection by a herpeslike<br />

virus resulting in high rates of morbidity and mortality in affected populations. Gross signs have included<br />

loss of adhesion due to relaxation of the pedal muscle, swelling of the buccal region and aversion of the<br />

radula. Histopathology revealed severe lesions centred in nerve tissues including oedema, cell necrosis and<br />

haemocytic infiltrations. Herpes-like virus particles were isolated from infected abalone tissues for DNA<br />

extraction and application to whole genome pyrosequencing.<br />

Material & methods<br />

Pooled tissues from infected Victorian wild abalone were homogenised and sonicated for virus isolation.<br />

Virus particles were isolated using discontinuous 10-60% sucrose density gradient ultracentrifugation.<br />

Herpes-like virus particles were found to be concentrated at the 40-50% sucrose interface. DNA extraction<br />

from virus containing interface fractions produced a single high MW DNA fragment. The DNA extract was<br />

used for multiple displacement amplification using QIAGEN REPLI-g, which generated sufficient template for<br />

direct ultrahigh-throughput parallel DNA sequencing using the 454 Life Sciences Genome Sequencer-FLX<br />

System. The abalone herpes-like virus was characterised via de novo assembly and annotation of the<br />

generated DNA sequences.<br />

Results<br />

A single sequencing run yielded in excess of 20.4 megabases of parallelised sequence information from the<br />

abalone virus DNA sample. Analysis of the sequence data has assembled multiple contigs with the largest<br />

up to 46 kb and totalling to greater than 150 kb of non-overlapping sequence segments, putatively<br />

representing a large portion of the expected virus genome. Assembled contigs were initially analysed by<br />

BlastX comparison to the genome of ostreid herpesvirus OsHV-1, a virus that causes similar disease<br />

pathology in oysters and other bivalves (1). The translated sequence contigs from abalone herpes-like virus<br />

showed 25-50% peptide sequence similarities with those of OsHV-1. PCR primers based on the derived<br />

abalone virus sequences were designed for application to specific conventional end-point and real-time PCR<br />

detection assays for virus infected abalone tissues. PCR primers tested so far have shown promise when<br />

tested against infected abalone and non-infected controls.<br />

Discussions & conclusions<br />

Analysis of the putative genome sequences derived from our study has facilitated the definitive identification<br />

and characterisation of the herpes-like virus causing abalone mortalities in Australia, and in gastropod<br />

molluscs. The virus affecting Victorian abalone populations appear to be only distantly related to the OsHV-1<br />

herpesvirus affecting bivalve molluscs in other countries. The two respective herpes-like virus pathogens do<br />

not appear to be the same virus or closely related strains. At present there is no rapid or specific detection<br />

method available for identification of the abalone herpes-like virus, making environmental surveillance of the<br />

disease and study of the pathogen extremely difficult. The development of specific PCR primers and in situ<br />

DNA probes for immunohistological detection tests in our study would greatly facilitate further investigation of<br />

the environmental spread and distribution of the virus.<br />

References<br />

(1) Davison, A. J., B. L. Trus, N. Cheng, A. C. Steven, M. S. Watson, C. Cunningham, R. -M. Le Deuff, and<br />

T. Renault. 2005. A novel class of herpesvirus with bivalve hosts. J. Gen. Virol. 86: 41-53.<br />

(2) Hooper, C., P. Hardy-Smith, and J. Handlinger. 2007. Ganglioneuritis causing high mortalities in farmed<br />

Australian abalone (Haliotis laevigata and Haliotis rubra). Aust. Vet. J. 85: 188-193.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF A REAL-TIME DUPLEX TAQMAN RT-PCR ASSAY FOR THE DETECTION OF<br />

EQUINE RHINITIS A AND B VIRUSES IN CLINICAL SPECIMENS<br />

Mori A., De Benedictis P., Zecchin B., Marciano S., Capua I., Cattoli G.<br />

Istituto Zooprofilattico Sperimentale delle Venezie, Research & Development Department<br />

OIE/FAO and National Reference Laboratory for Avian Influenza and Newcastle Disease<br />

OIE Collaborating Centre for Epidemiology, Training and Control of Emerging Avian Diseases<br />

Viale dell’Università 10, Legnaro, Padova, Italy<br />

Introduction<br />

Equine rhinitis A and B viruses (ERAV and ERBV) are respiratory viruses of horses belonging to the family<br />

Picornaviridae, genus Aphthovirus and Erbovirus respectively. Their laboratory confirmation is rare, even<br />

when there is serological evidence of their circulation. Virus isolation is often unsuccessful, due to inefficient<br />

growth in cell cultures, in many cases without a cytopathic effect (Li et al., 1997; Black et al, 2007). We<br />

describe here a real-time duplex TaqMan RT-PCR as a diagnostic tool for the detection of these viruses.<br />

Material & methods<br />

Strains of European and North American origin were used as positive controls. The titre in tissue culture<br />

infectious dose/ml (TCID50/ml) in RK13 cells was calculated for selected strains (Lorenz and Bogel,<br />

1973).Total RNA was extracted using the “High Pure TM RNA extraction kit” (Roche Diagnostics). The RNA<br />

was reverse-transcribed using “High capacity cDNA archive kit” (Applied Biosystems). Two sets of primers<br />

and probe were designed to detect viral RNA of ERAV and ERBV respectively. Both sets were located in the<br />

highly conserved 3D region (Huang J-A et al., 2001). The Real Time Duplex PCR reaction was carried out in<br />

25 µl with 1X “TaqMan Universal PCR Master Mix 2X” (Applied Biosystems), 240 nM of each primer, 250 nM<br />

of each probe and 5 µl cDNA template. The reaction was performed in a 7300 Real Time PCR System<br />

(Applied Biosystems) with the following protocol: 2 minutes at 50°C and 10 minutes at 95°C followed by 45<br />

cycles at 95°C for 15 sec and 60°C for 1 minute.<br />

The specificity of the method was assessed by testing each set of primers and probe for the ERAV or the<br />

ERBV template distinctly. Common equine RNA and DNA viruses, bacterial and protozoan pathogens were<br />

analysed to further ensure the specificity of the primer-probe sets. To assess the sensitivity of the method,<br />

ten-fold serial dilutions of titrated ERAV and ERBV were tested in different matrices, namely tissue culture<br />

supernatant, lung homogenates, urine and nasal swabs. Three different concentrations (high, medium, low)<br />

for both ERAV and ERBV were analyzed to assess the intra-assay repeatability and the inter-assay<br />

reproducibility.<br />

Results<br />

The designed sets were able to distinguish ERAV and ERBV viral genomes. No cross interaction was<br />

observed among the two sets of primers and probes. The analysis of other equine viruses, bacterial and<br />

protozoan pathogens gave negative results.The limit of detection (LoD) was 10 TCID50/ml for ERAV and 1<br />

TCID50/ml for ERBV in all tested matrixes. The Coefficient of Variation (CV) was calculated as below 7% and<br />

below 12%, for the intra-assay repeatability and the inter-assay reproducibility respectively.<br />

Discussions & conclusions<br />

The real-time duplex TaqMan-PCR developed was capable of detecting and differentiating both ERAV and<br />

ERBV. The method demonstrated excellent specificity and sensitivity, intra-assay repeatability and interassay<br />

reproducibility. The results of this investigation indicate that the molecular assay can be considered a<br />

valuable tool to aid the diagnosis of respiratory viral infections in horses. The high sensitivity and specificity<br />

of the test make it a valid instrument to provide further insights into the pathogenesis of these infections.<br />

References<br />

Black WD, Hartlley CA, Ficorilli NP, Studdert MJ. Arch Virol (2007) 152:137-149.<br />

Huang J-A, Ficorilli N, Hartley CA, Wilcox RS, Weiss M, Studdert MJ. J Gen Virol (2001) 82:2641-2645.<br />

Li F, Drummer HE, Ficorilli N, Studdert MJ, Crabb BS. J Clin Microbiol. (1997) 35:937-943.<br />

Lorenz RG., Bogel K. In: Kaplan MM, Koprowski H, eds. Laboratory techniques in rabies, 3rd ed. Geneva,<br />

Switzerland: World Health Organization (1973) 321:35-41. Dean AG, Dean JA, Burton AH, et al. Epi Info<br />

Version.<br />

Acknowledgements<br />

The authors wish to thank Udeni B. R. Balasuriya, University of Kentucky-USA, Janet Daly and Bob<br />

Geragthy, Animal Health Trust, Suffolk-UK, for kindly supplying ERAV and ERBV strains used in this work.<br />

Wed 14 November


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

1530 - 1700 Concurrent Session 2.6 - Epidemiology & Surveillance<br />

EFFECTS OF DYNAMIC ‘COMMUNAL’ INTERACTIONS ON DISEASE PROCESSES AND INDIVIDUAL<br />

AGENT DIAGNOSIS WHERE MULTIPLE INFECTIOUS AGENTS ARE PRESENT IN SHEEP<br />

K.L Tyrrell 1* , J.R White 2 , S.J McClure 1 & J. Lello 3<br />

1<br />

CSIRO Livestock Industries, Locked Bag 1, Armidale, NSW 2350<br />

2<br />

Australian Animal Health Laboratory, CSIRO Livestock Industries, Private Bag 24, Geelong, VIC 3220<br />

3<br />

School of Biosciences, Cardiff University, Cardiff, UK<br />

Introduction<br />

The concept of community ecology (an assemblage of living organisms that share and interact within a<br />

common environment) is explored in relation to multiple infections of parasitic worm species in sheep.<br />

Although single parasitic infections are not uncommon, mixed infection with various species or with several<br />

different types of parasites is generally considered to be normal. There are a number of examples in the<br />

literature which have shown that in mixed infections the presence of one pathogen can have either a<br />

detrimental, neutral or positive effect on the other (Kaufmann et.al 1992, Yabob et.al 2002, Dobson and<br />

Barnes 1995). It has also been shown that these interactions can affect the hosts response to one pathogen<br />

thereby giving an advantage to the other pathogen (Sharma et.al, 2000). In previous studies involving<br />

infection with a combination of up to three species, results showed a marked immunological difference<br />

between sheep with a single infection of Trichostrongylus colubriformis and dually infected sheep which<br />

were given a mixture of T. colubriformis and H. contortus. Total T. colubriformis worm counts were higher in<br />

dually infected sheep and there was a trend for lower T. colubriformis egg outputs in these sheep. The<br />

current study further examined this interaction to determine the mechanisms involved.<br />

Material & methods<br />

The study was conducted on 132 5 month old merino sheep. Animals were trickle infected with single or<br />

mixed nematode species over a period of 14 weeks. Faecal collections were made over the course of the<br />

study to measure nematode worm egg count and worm species composition. Blood collections were made<br />

to measure haematocrit, specific antibody levels and nutritional parameters. At the conclusion of the study<br />

animals were euthanased for total nematode worm counts and abomasal and jejunal tissue collected for use<br />

in histological assessment of immune response and antibody concentration.<br />

Results<br />

Integration of the various data collected, including worm egg count, worm count, histology and antibody<br />

titers, provided evidence to suggest that H. contortus suppresses those immune responses normally<br />

associated with a single T. colubriformis infection. Adult T. colubriformis worm counts were higher in dually<br />

infected sheep from day 70 through to the conclusion of the study at Day 120 post initial infection. Further, a<br />

suppressive effect was seen in antibody titers in mixed infection animals, whereby titers did not reach levels<br />

seen in animals infected with T. colubriformis only. This effect on the immune response was confirmed in the<br />

histology data, where smooth muscle and globular leucocyte responses to T.colubriformis were reduced<br />

when H. contortus was present. Glucose and protein levels taken through out the study did not suggest any<br />

role for nutrition in this suppression.<br />

Discussions & conclusions<br />

This study has shown that in a mixed infections of H. contortus and T. colubriformis, that H. contortus<br />

suppresses the immune response normally associated with a T. colubrifomis infection despite the spatially<br />

difference niches which each pathogen has within the host gut. The results suggest that a number of<br />

parameters may need to be carefully considered when attempting to accurately determine the aetiological<br />

agent/s responsible for a given disease syndrome. By understanding the possible ‘communal relations’<br />

(inter-specific interactions) between species, it is then possible to 1) improve parasite control programs 2)<br />

offer possibilities for alternative control 3) understand the treatment and immunological effect of targeting<br />

one species within a biological system 4) reduce chemical use in control programs and 5) improve diagnosis<br />

and reduce severity of disease outbreaks.<br />

References<br />

Kaufmann, J. et.al. (1992). Vet Para, 43:157-170.<br />

Yacob, H.T. et.al (2002). Vet Para. 104: 307-317.<br />

Dobson, R.J & Barnes, E.H (1995). Int J for Para 25 (4):495-501.<br />

Sharma, D.K. et.al. (2000). Vet Para 92:261-267.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

USE OF MOLECULAR AND EPIDEMIOLOGICAL INFORMATION FOR THE COST-EFFECTIVE<br />

DIAGNOSIS OF BOVINE VIRAL DIARRHOEA INFECTION IN NEW ZEALAND DAIRY CATTLE<br />

Introduction<br />

F I Hill*, M P Reichel, D J, Tisdall, Gribbles Veterinary, Palmerston North, New Zealand, *presenting author<br />

Veterinary interest in bovine viral diarrhoea (BVD) in New Zealand has been increasing since 2004.<br />

Representatives from the veterinary and farming communities formed groups to investigate the significance<br />

of the disease and the merits and method of diagnosis and control. Diagnostic tests allowing cost effective<br />

identification of BVD virus (BVDV) infected herds and individuals were a key requirement for farmers and<br />

veterinarians. BVDV persistently infected (PI) animals excrete vast amounts of virus in all body secretions<br />

including milk. Quantitative (or real-time) PCR technology has the ability to detect virus in ready-made pools,<br />

such as bulk tank milk in a vat. Epidemiological studies have shown that persistent BVDV infection in dairy<br />

cattle has a detrimental effect on lactation (Voges et al 2006) with most milking PI cattle found within the<br />

lowest producing 10% of the herd. This study reports on the use of PCR technology and that<br />

epidemiological information to identify PI animals in milking herds in a cost-effective manner.<br />

Materials & methods<br />

Milk samples were collected from the milk vat of dairy herds where veterinary practitioners suspected a<br />

problem with BVDV infection. Positive bulk tank milk was followed up by collection of individual serum<br />

samples. Testing of individual samples by PCR and antigen capture enzyme-linked immunosorbent assays<br />

(ACE) was used to identify PI animals. PCR and ACE are known to correlate well (Hill et al. 2007). After<br />

removal of the PI from the herd, repeat testing of another bulk milk sample was used to verify clearance of<br />

the infection.<br />

Results<br />

From 40 bulk milk samples initially tested by PCR, 6 were positive for BVDV virus. Four of these herds were<br />

investigated in depth. Serum samples were collected from every cow in the herd and 1/223, 1/130, 2/800<br />

and 1/275 PI’s identified. After removal of the PI from the herd another bulk milk sample was now negative.<br />

PIs were always found in the bottom 10% of producers.<br />

If BVDV infection was confirmed in the milking herd, investigation of the BVDV status of non-milking stock<br />

was undertaken using BVD antibody surveillance testing of 15 animals from each age group. Cohorts of<br />

seropositive groups of animals were tested in real time PCR in pools of 20 sera, followed by ACE testing to<br />

identify any viraemic animals. Consideration of cost-effectiveness and prevalence of PIs, rather than the<br />

technical ability of the assay determined pool size.<br />

Discussion & conclusions<br />

Targeted testing of dairy herds using PCR technology, in conjunction with epidemiological information, has<br />

markedly reduced the cost of diagnostic testing for BVDV in dairy herds in New Zealand over the past two<br />

years.<br />

References<br />

Hill, F.I., M.P.Reichel, D.J.Tisdall, and R.J.McCoy. 2007. Evaluation of two commercial enzyme-linked<br />

immunosorbent assays for detection of bovine viral diarrhoea virus in serum and skin biopsies of cattle. N. Z<br />

. Vet. J. 55:45-48.<br />

Voges, H., Young, S., Nash, M., 2006, Direct adverse effects of persistent BVDv infection in dairy heifers – a<br />

retrospective case control study. VetScript, 22-25.<br />

Wed 14 November


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ASSESSING THE WITHIN HERD PREVALENCE OF ANTIBODY POSITIVE COWS TO BOVINE<br />

HERPEVIRUS 1 USING AN INDIRECT ELISA ON BULK TANK MILK<br />

Background<br />

1.Aims<br />

The objective of the study was to evaluate the efficacy of a milk Indirect antibody ELISA to assess the within<br />

herd prevalence of cows seropositive to BHV 1.<br />

2.Methods<br />

Bulk milk samples from 799 dairy herds from Terceira Island in Azores/Portugal were initially tested for BHV<br />

1 antibodies using a commercial ELISA. The herds were then ranked in three cohorts according to their<br />

ELISA result sample to positve ratio (S/P) as follows: cohort 1 (C1) S/P


Wed 14 November<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ZOO ANIMALS AS A POTENTIAL RESERVOIR OF GRAM-NEGATIVE BACTERIA HARBORING<br />

INTEGRONS AND ANTIMICROBIAL RESISTANCE GENES<br />

Ashraf M. Ahmed 1,3* Yusuke Motoi 1 , Maiko Sato 1 , Akito Maruyama 1 , Hitoshi Watanabe 2 , Yukio Fukumoto 2 and Tadashi Shimamoto 1<br />

1 Laboratory of Food Microbiology and Hygiene, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-<br />

8528, Japan; 2 Hiroshima City Asa Zoological Park, Asa-cho Asakita-ku, Hiroshima 731-3355, Japan 3 Department of Microbiology,<br />

Faculty of Veterinary Medicine, Kafr El-Sheikh University, Kafr El-Sheikh 33516, Egypt<br />

Background:<br />

Problems associated with the development and spread of antimicrobial resistance in clinical practice have<br />

been increasing since the early 1960s and are currently viewed as a major threat to the public health on a<br />

global level. Zoo animals constitute a potential source of zoonotic infections and, thus, a public health risk.<br />

Of particular concern is the potential transmission of multidrug-resistant (MDR) zoonotic pathogens from<br />

animals to humans.<br />

Aims:<br />

As little is known about antimicrobial-resistant bacteria in zoo animals, this study was conducted to monitor<br />

the incidence and prevalence of antimicrobial resistance genes in gram-negative bacteria isolated from<br />

mammals, reptiles and birds housed at Asa Zoological Park, Hiroshima prefecture, Japan.<br />

Methods:<br />

A total of 103 swabs (68 fecal, 33 water and two nasal swabs) were randomly taken from different mammals,<br />

reptiles, birds and water sources between June and September 2006 at Asa Zoological Park, Hiroshima<br />

prefecture, Japan. Biochemical, antibiograms, PCR and DNA sequencing techniques were used for<br />

identification and molecular characterization of bacteria and antimicrobial resistance genes.<br />

Results:<br />

A total of 232 isolates of gram-negative bacteria were recovered, the most common being Escherichia coli<br />

122 (52.6%), Klebsiella pneumoniae 17 (7.3%), Proteus mirabilis 16 (6.9%), Enterobacter aerogenes 13<br />

(5.6%), Klebsiella oxytoca 13 (5.6 %), Pseudomonas aeruginosa 12 (5.2%) and Enterobacter cloacae 12<br />

(5.2%). A total of 49 isolates (21.1%) showed resistance phenotypes to two or more antimicrobial agent and<br />

harbored at least one antimicrobial-resistant determinant. PCR screening for integrons showed that 16<br />

(6.9%) and four (1.7%) isolates were positive for class 1 and class 2 integrons, respectively. The βlactamase-encoding<br />

genes blaTEM-1, blaOXY-2, blaSHV-36 and blaCTX-M-2 were identified in 19 (8.2 %), three<br />

(1.3%), two (0.9%) and one (0.43%) isolate, respectively, in addition to a novel ampC β-lactamase gene,<br />

blaCMY-26, identified in a single isolate. The plasmid-mediated quinolone resistance genes, qnr and aac(6’)-Ibcr,<br />

were identified in 10 (4.3%) and one (0.43% ) isolate, respectively.<br />

Conclusion:<br />

Although zoo animals do not naturally come into contact with antibiotics, the results of this study established<br />

zoo animals as a potential reservoir of antimicrobial-resistant bacteria and clinically important resistance<br />

genes. This study highlights the potential risk factor of zoo animals to public health.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PORCINE REPRODUCTIVE & RESPIRATORY SYNDROME (PRRS) - A NEW CHALLENGE<br />

Nguyen Van Long<br />

Not available at time of printing.<br />

Wed 14 November


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

POSTER PRESENTATIONS<br />

PROXIMITY LIGATION ASSAY – DETECTION OF INDIVIDUAL MICROBIAL PATHOGENS<br />

A Nordengrahn, SM Gustafsdottir, M Merza*<br />

Svanova Biotech AB, Uppsala Science Park, 751 83 Uppsala, Sweden<br />

Introduction<br />

The Proximity ligation assay (PLA) enables sensitive detection of proteins in samples by binding of affinity<br />

probes to the target proteins. These affinity probes are equipped with DNA strands that can be joined by<br />

ligation when two such reagents are brought into proximity by binding to the same target molecule. The DNA<br />

ligation products are subsequently detected by DNA amplification. Using real time PCR detection system,<br />

the product can be quantified i.e. the number of targets in the sample can be calculated.<br />

We have successfully used PLA for the detection of three pathogens, two viruses Foot-and mouth disease<br />

and Porcine parvovirus and the bacterium Lawsonia Intracellularis.<br />

Material & methods<br />

In the assays monoclonal antibodies specific to each of the pathogens were used as affinity probes. Nonsense<br />

oligonucleotides were attached to the antibodies by biotin-avidin coupling.<br />

By generating titration curves of the studied pathogens with known viral/bacterial titres the detection limit of<br />

the different systems were established.<br />

Real time PCR was used for visualisation of the reactions.<br />

The results were compared with Capture ELISA and Quantitative PCR.<br />

Results<br />

The results show very good sensitivity and specificity with detection limits down to less than 10 copies of the<br />

virus/bacteria. This was in line with the qPCR tests used for comparison. The Capture Elisa’s on the other<br />

hand where about 100 times less sensitive than the PLA.<br />

Discussions & conclusions<br />

Our data strongly support the use of PLA as a diagnostic tool enabling a very sensitive and specific detection<br />

of microbial proteins, far more sensitive than the Capture ELISA, and being alternative to nucleic detection<br />

showing a sensitivity and specificity in line with the PCR.<br />

References<br />

Gustafsdottir SM, Nordengrahn A, Fredriksson S, Wallgren P, Rivera E, Schallmeiner E, Merza M,<br />

Landegren U. 2006. Detection of Individual Microbial Pathogens by Proximity Ligation. Clin. Chem. 52:6,<br />

1152-1160.<br />

Nordengrahn A, Gustafsdottir SM, Ebert K, Reid SM, King DP, Ferris NP, Brocchi E, Grazioli S, Landegren<br />

U, Merza M. Evaluation of a novel proximity ligation assay for the sensitive and rapid detection of foot-andmouth<br />

disease virus. Vet. Microbiol. In press.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

IMPROVED DETECTION OF BOVINE REPRODUCTIVE DISEASE PATHOGENS<br />

A.E. Lew 1,3 , B. Venus 1 , B. Wlodek 1 , S-Y. Guo 2 , G. Fordyce 1 , P. Moolhuijzen 3 , M.I. Bellgard 3 , G. Coleman 2 , D. Trott 2 , P. Burrell 1 , J.T.<br />

Ellis 4 , B. Corney 1 , W.K. Jorgensen 1 *<br />

1 Department of Primary Industries & Fisheries, Yeerongpilly, Qld, Australia<br />

2 School of Veterinary Science, The University of Qld, Qld, Australia<br />

3 Centre for Comparative Genomics, Murdoch University, WA, Australia<br />

4 University of Technology Sydney, NSW, Australia<br />

Introduction<br />

Reproductive diseases are known to cause bovine embryonic loss and calf abortion but it is often difficult to<br />

determine the aetiological pathogen using gold standard methods such as serology, microscopy and culture<br />

isolation. Sensitive molecular probe based polymerase chain reaction (PCR) assays (real time assays) were<br />

developed and evaluated for the improved detection of four pathogens implicated in the following<br />

reproductive diseases: trichomoniasis (Tritrichomonas foetus), campylobacteriosis (Campylobacter fetus<br />

subsp. venerealis), ephemeral fever (bovine ephemeral fever virus), and neosporosis (Neospora caninum).<br />

Material & methods<br />

Probes based on 3’ minor groove binder probe (TaqMan®MGB) technology were developed for each<br />

pathogen targeting the internal transcribed spacer region for T. foetus and N. caninum respectively (2,5), the<br />

parA gene in C. fetus (4), and the BEFV glycoprotein RNA (RT-PCR) (1).<br />

Results<br />

All assays demonstrated sensitivity and specificity improvements over existing methods and conventional<br />

PCR protocols. For the bovine venereal diseases, a new animal sampling tool (Tricamper TM Sampling<br />

Device) proved more efficient and improved the sensitivity of pathogen identification over conventional<br />

sampling methods (by culture and PCR). Real time PCR demonstrated a higher field prevalence of<br />

campylobacteriosis pathogens than previously determined due to a two thousand fold increase in sensitivity<br />

compared with the gold standard culture method. Comparative genomic studies have identified new gene<br />

targets for C. fetus subsp. venerealis molecular diagnostic assay development to confirm this high<br />

prevalence (3). The BEFV real time PCR identified cattle previously not identified as infected using culture<br />

and the assay did not amplify closely related ephemeroviruses. The application of the N. caninum real time<br />

assay demonstrated the presence of N. caninum in Hammondia heydornii (CZ and NZ) preparations<br />

originating from dogs. The assay was useful in detecting N. caninum in serum, organs and milk from<br />

infected cattle and dogs, and in formalin fixed mouse brains (infection trial).<br />

Discussions & Conclusions<br />

The above assays will assist in identifying the contributing factors to substantial (>10%) unexplained<br />

reproductive losses (confirmed pregnancy to weaning) that occur in many northern Australian beef herds.<br />

These initial assays for each of the four pathogens have been adopted by Australian diagnostic laboratories<br />

and have provided useful disease research tools. The C. fetus subsp. venerealis parA gene assay has been<br />

submitted to the Sub-Committee on Animal Health Laboratory Standards (Australia/NZ) for evaluation.<br />

This research was funded by Meat & Livestock Australia.<br />

References<br />

1. Lew AE, Corney B, Minchin CM, Wright L. (2005a) Real time RT-PCR detection of bovine ephemeral<br />

fever virus infections. National Conference for the Australian Society for Microbiology, Canberra, Australia,<br />

25-29 th September, 2005.<br />

2. Lew A, Ellis J, Coleman G, McMillen L, Turner S, Venus B. (2005b) Real time PCR detection of<br />

Neospora caninum. World Association for the Advancement of Veterinary Parasitology WAAVP, New<br />

Zealand, 16-20th October 2005.<br />

3. Lew A, Guo SY, Venus B, Moolhuijzen P, Sanchez D, Trott D, Burrell P, Wlodek B, Bellgard M. (2007)<br />

Comparative genome analysis applied to develop novel PCR assays to characterise and identify<br />

Campylobacter fetus subsp. venerealis isolates. Zoonoses and Public Health, 2007, 54 (Supplement 1),<br />

19-155.<br />

4. McMillen, L., Fordyce, G., Doogan, V. J., Lew, A.E. (2005) Comparison of culture and a novel 5' Taq<br />

nuclease assay for the direct detection of Campylobacter fetus subspecies venerealis in clinical specimens<br />

from cattle. Journal of Clinical Microbiology 44(3): 938-945.<br />

5. McMillen, L. & Lew, A.E. (2006) Improved detection of Tritrichomonas foetus in bovine diagnostic<br />

specimens using a novel probe based real time PCR assay. Veterinary Parasitology 141: 204-215.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

IDENTIFICATION OF A NOVEL BABESIA SPECIES FROM SABLE, ROAN AND GIRAFFE BY MEANS<br />

OF THE REVERSE LINE BLOT (RLB) HYBRIDIZATION ASSAY<br />

MC Oosthuizen 1* , E Zweygarth 2 & BL Penzhorn 1<br />

1 Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort,<br />

0110, South Africa. (E-mail: marinda.oosthuizen@up.ac.za)<br />

2 Onderstepoort Veterinary Institute, Private Bag X05, Onderstepoort, 0110, South Africa<br />

Introduction<br />

Sable (Hippotragus niger) and roan (Hippotragus equinus) antelope populations have shown alarming<br />

decreases in numbers in the past decades. Sable antelope are listed as “conservation dependent” on the<br />

IUCN Red List and declining numbers could lead to a “threatened” listing in the near future. Roan antelope<br />

are classified as vulnerable. They are specialist grazers, rarely feeding on browse, making them endangered<br />

in some areas such as the Kruger National Park.<br />

Wild ruminants are known to harbour a variety of intra-erythrocytic parasites. Theileria sp., Babesia sp. and<br />

Anaplasma sp. have previously been reported from sable antelope, mostly from asymptomatic carriers;<br />

clinical signs only appear when the animals are stressed. Babesiosis has been reported in roan antelopes.<br />

Piroplasms have also previously been reported from giraffes in Kenya and Namibia and in one case<br />

theileriosis was suspected of causing mortality. Specimens from sable, roan and giraffe that presented a<br />

sudden onset of disease and subsequently died were submitted for molecular characterization. The Reverse<br />

Line Blot (RLB) assay, developed for the simultaneous detection and identification of tick-borne parasites<br />

infecting cattle and small ruminants, was successfully used to identify previously undescribed Theileria and<br />

Babesia species infecting wild ruminants.<br />

Material & methods<br />

Blood smears, blood and/or spleen samples collected from sable, roan and giraffe were received. DNA was<br />

extracted; the V4 variable region of the 18S rRNA gene amplified and analyzed using RLB. PCR products<br />

which did not hybridize with any of the Babesia or Theileria species-specific probes on the blot, but only with<br />

the Babesia / Theileria genus-specific probe were further investigated. Full-length 18S rDNA was amplified,<br />

cloned and the recombinants were analysed by sequencing analysis. Sequencing data were analysed using<br />

the Staden package, aligned with published sequences of related genera using ClustalX and phylogenetic<br />

trees were constructed using neighbor-joining in combination with the bootstrap method.<br />

Results<br />

Microscopic examination of thin blood smears revealed the presence of small piroplasms. RLB results<br />

showed that the PCR products amplified from the giraffe, sable and roan specimens hybridized only with the<br />

Babesia / Theileria genus-specific probe, but not with any of the Babesia or Theileria species-specific probes<br />

present on the blot. This probably indicated the presence of a novel Babesia or Theileria species or variant<br />

of a species.<br />

Discussions & conclusions<br />

Sequence analysis revealed the presence of novel Babesia and/or Theileria species in sable, roan and<br />

giraffe that presented a sudden onset of disease and subsequently died. Molecular characterization<br />

indicated that the parasite in sable antelope is a member of the Babesia sensu stricto clade. It is tempting to<br />

propose that the parasite described in this study is B. irvinesmithi, first described in 1930 and responsible for<br />

the fatal cases reported in sable antelope in the past. The 18S rRNA gene sequences of the parasites<br />

isolated from roan and giraffe were most similar to Babesia sp Xinjiang-2005, a sheep isolate, which was<br />

originally isolated in China and described as often leading to clinically inapparent infection in sheep. Also, a<br />

Theileria species infection was present in some of the giraffe samples. Piroplasmosis could therefore be<br />

implicated as a possible cause of mortality in sable, roan and giraffe. It is not known whether these were<br />

isolated incidents or whether these parasites cause severe disease in free-ranging populations.<br />

Acknowledgements<br />

<strong>Symposium</strong> attendance sponsored by: FAO-EMPRES<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DETECTION OF BLV IN FROZEN SEMEN SAMPLES BY PCR ASSAY<br />

L. Rossich 1 , J. Gutierrez 1 , S. Rodriguez 1 , E. Lefebvre 2 , K. Trono 1 and M. J. Dus Santos 1*<br />

(1) Instituto de Virología, CICVyA, INTA-Castelar. CC 77. Buenos Aires, Argentina.<br />

(2) Centro de Inseminación Artificial La Elisa. Argentina.<br />

* mdussantos@cnia.inta.gov.ar<br />

Introduction<br />

The major route of transmission of BLV is horizontal by direct exposure to biological fluids contaminated with<br />

infected lymphocytes, mainly blood. Although viral antigens and proviral DNA has been identified in semen,<br />

milk and colostrums, natural transmission through these secretions has not been demonstrated. The sanitary<br />

and economic impact of BLV infection is associated with the interference in the international movement of<br />

cattle and their germ plasm. Although experimental data support the improbability that semen from BLVpositive<br />

bulls could infect recipient cows, restriction for commercialization of semen from infected animals is<br />

still present 1-6 . Since there is no vaccine or treatment available, eradication and control of the disease is<br />

exclusively based on early diagnostic and segregation of infected animals. In this context the specificity and<br />

sensitivity of the diagnostic test used is a critical point.<br />

We have previously developed a PCR assay with high sensitivity and specificity to detect BLV genome in<br />

frozen semen samples.<br />

The objective of this work was to study the detection of BLV in semen.<br />

Material & methods<br />

Samples: Serum, whole blood and semen samples were obtained from CIALE (Artificial Insemination Centre<br />

La Elisa, Capitan Sarmiento, Buenos Aires, Argentina)<br />

Serology: The agar gel immunodifussion (AGID) test kit used to detect antibodies to BLV was produced by<br />

the Faculty of Veterinary, La Plata University, Argentina.<br />

An indirect ELISA using recombinant p24 as antigen was used to detect antibodies to BLV. This assay has<br />

been completely developed and standardized in the Institute of Virology, INTA-Castelar, Buenos Aires,<br />

Argentina.<br />

Detection of proviral DNA: DNA was extracted from PMBCs (purified from whole blood) and semen samples<br />

(fresh and straw). PCR assays that amplified a region of genes pol and gag and a nPCR that amplified a<br />

region of gene env were developed.<br />

Results<br />

Two PCR assays were standardized for detecting BLV genome in semen and PMBC. The limit of detection<br />

of viral particles was assessed by the addition of purified pBLV344 (a plasmid containing the complete BLV<br />

genome, kindly provided by Dr. Willems, Faculté Universitaire des Sciences Agronomiques, Gembloux,<br />

Belgium) to DNA from semen or PMBCs of a seronegative bull. By gag-PCR and pol-PCR, it was possible to<br />

detect 60 viral particles, using bromide staining after electrophoresis separation of DNA. In order to increase<br />

analytical sensitivity, a nested-PCR was developed which amplified a region of env gene. The n-PCR was<br />

able to detected 6 viral particles. Assessment of the limit of detection was highly repetitive under the<br />

standardized conditions.<br />

Frozen semen samples from 30 seropositive bulls were remitted to the laboratory and analyzed in the period<br />

2005-2007. It was possible to detect proviral DNA in 172 out of 862 samples. BLV genome detection<br />

occurred in several collections but in an alternated way with non detection periods.<br />

Fresh semen samples, straws and whole blood were also obtained from 5 seronegative and 5 seropositive<br />

bulls and tested together for the presence of BLV provirus. Results indicated that while detection of provirus<br />

was positive in PMBC from all seropositive bulls, detection of gag, pol and env genes in semen did not<br />

occurred in all the samples.<br />

Discussions & conclusions<br />

The results obtained suggest that BLV could present an intermittent pattern of excretion.<br />

Further studies should be done to confirmed the results obtained and to establish why the presence of BLV<br />

provirus in semen is not constant.<br />

References<br />

1. Choi, K, et al. 2002. Vet Diagn Invest 14: 403-406.<br />

2. Kaja, R. and Olson, C. 1982. Theriogenology 18: 107-112.<br />

3. Miller, J. and Van Der Maaten, M. 1979. J Natl Cancer Inst 62; 425-428.<br />

4. Monke, D. 1986. JAVMA 188 (8): 823-826.<br />

5. Romero, C., et al. 1983. Tropical Animal Health and production. 15: 215-218.<br />

6. Straub O. 1982. In: Fourth International <strong>Symposium</strong> of Bovine Leukosis. The Hague: Martinus Nijhoff<br />

Publishers: 299-308.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A SIMPLE INTERNAL PCR CONTROL FOR MYCOBACTERIUM AVIUM SUBSP. PARATUBERCULOSIS<br />

CONSTRUCTED BY PCR TECHNIQUES<br />

Introduction<br />

I. Marsh, M. McLoon, S. Austin, S. Fell, V. Saunders and L.Reddacliff<br />

New South Wales Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Australia<br />

Polymerase chain reaction (PCR) is routinely used to confirm the presence of Mycobacterium avium subsp.<br />

paratuberculosis (MAP) in the diagnosis of ovine Johne’s disease (OJD). A limitation of diagnostic PCR<br />

assays is false negative results, that is, samples that are positive but do not amplify due to inhibition of the<br />

assay. PCR assays that do not include an internal control for each individual reaction can at best produce<br />

an interpretable result from non-inhibited positive samples and this makes it difficult to interpret negative<br />

results. Under these conditions diagnosticians cannot determine if a negative result is truly negative or if the<br />

reaction failed as a result of inhibitory substances present in the sample. Even though the appropriate<br />

positive and negative controls may have been included within a PCR run, generally run as individual<br />

reactions, these are only useful for establishing the integrity of the PCR reagents and act as references for<br />

which the samples can be compared against. To overcome this situation PCR-based diagnosticians have<br />

begun to introduce internal controls such that each amplification reaction can be monitored. A number of<br />

these internal controls have been reported 1-3 but have yet to be adopted widely by the M. a. paratuberculosis<br />

research or diagnostic community. Generally, these internal controls are produced using DNA cloning<br />

techniques and this may be a limitation on their widespread use, as many laboratories may not have time,<br />

finances or the laboratory expertise to prepare these internal controls. To overcome this we have designed<br />

a PCR-based method for producing an internal amplification control (IAC), based on a method described and<br />

used in the detection of Bordetella pertussis by PCR 4 . Unlike other internal controls that require cloning and<br />

other molecular manipulations, this IAC only requires PCR capability for its production.<br />

Method<br />

The IAC was constructed in a two step PCR process that amplified a region of the Mycobacterium avium<br />

subsp. avium (MAA) genome that is not present in the MAP genome using composite primers made of an<br />

MAA region and an MAP region. The IAC was then incorporated in to a multiplex PCR that included a new<br />

MAP specific target to increase specificity. The analytical sensitivity of the IAC and multiplex PCR was<br />

established prior to evaluation on DNA samples that had been previously examined for OJD.<br />

Results<br />

The IAC had no adverse effects on the analytical sensitivity of the MAP specific multiplex PCR. The new<br />

PCR test was successfully used to determine the presence/absence of MAP in 25 faecal samples with<br />

known OJD status and simultaneously determine the integrity of each reaction. Future work includes<br />

modifying this PCR test to a real-time PCR format.<br />

Conclusion<br />

We present a new multiplex PCR test for MAP that incorporates an IAC. The procedure used to produce the<br />

IAC is simple and highly adaptable to other PCR-based diagnostic tests.<br />

References<br />

1. Englund, S., A. Ballagi-Pordany, G. Bolske, and K. E. Johansson. 1999. Single PCR and nested PCR with<br />

a mimic molecule for detection of Mycobacterium avium subsp. paratuberculosis. Diagn Microbiol<br />

Infect Dis 33:163-71.<br />

2. Englund, S., G. Bolske, A. Ballagi-Pordany, and K. E. Johansson. 2001. Detection of Mycobacterium<br />

avium subsp. paratuberculosis in tissue samples by single, fluorescent and nested PCR based on<br />

the IS900 gene. Vet Microbiol 81:257-71.<br />

3. Brey, B. J., R. P. Radcliff, D. L. Clark Jr, and J. L. Ellingson. 2006. Design and development of an internal<br />

control plasmid for the detection of Mycobacterium avium subsp. paratuberculosis using real-time<br />

PCR. Mol Cell Probes 20:51-9.<br />

4. Muller, F. M., N. Schnitzler, O. Cloot, P. Kockelkorn, G. Haase, and Z. Li. 1998. The rationale and method<br />

for constructing internal control DNA used in pertussis polymerase chain reaction. Diagn Microbiol<br />

Infect Dis 31:517-23.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INVESTIGATING SHEDDING RATES AND THE PROPORTION OF ANIMALS SHEDDING<br />

MYCOBACTERIUM AVIUM SUBSP PARATUBERCULOSIS IN CATTLE AND GOAT HERDS<br />

G.J. Eamens, NSW Department of Primary Industries<br />

Introduction<br />

Many diagnostic tests for Johne’s disease rely on, or correlate with, the level of faecal shedding of<br />

Mycobacterium avium subsp paratuberculosis (Map). Among faecal culture positive cattle, approximately<br />

70% are considered low faecal shedders of Map and 10% moderate shedders, 6 with low shedders the most<br />

difficult to detect in all diagnostic tests. Assessment of test performance must therefore be based on<br />

populations weighted more toward low to moderate shedder animals. However, such categories have only<br />

been defined from cultural results on solid media. 6 In recent studies of radiometric pooled faecal culture<br />

(PFC) for cattle and goats, a need to better understand shedding rates was highlighted.<br />

Material & methods<br />

During PFC studies in cattle and goats, 1-3 the growth of Map in dilutions in radiometric (Bactec) culture media<br />

with IS900 PCR/REA confirmation was used to identify a critical threshold for positive cultures. Map was<br />

isolated from faeces of 20 cattle and 16 goats in the studies. From several samples, the number of viable<br />

Map cells in the inoculum of each animal was calculated by endpoint titration (Most Probable Number, MPN)<br />

and then related to the rate of growth in radiometric culture. 4 This data was used to establish a regression<br />

equation to estimate the concentration of Map in all inocula, and then shedding rates in faeces of all animals<br />

were estimated, based on known processing losses during Bactec culture. 5 From the highest culture<br />

positive dilutions used in the PFC evaluations (between neat to 1:50), the minimal threshold number of Map<br />

was estimated. Estimates of growth from identical inocula on Herrold’s solid medium and Bactec media were<br />

compared to adjust known cutpoints for low to high shedders on solid media.<br />

Results<br />

Bactec culture was found to be 10 fold analytically more sensitive than solid media culture, so figures applied<br />

to delineate low to high shedding rates from solid culture growth were considered to contain 10 fold more<br />

bacteria when radiometric culture was applied. Based on adjusted cutpoints for Bactec culture of < 7.5 x 10 4<br />

/g for low shedders and > 7.5 x 10 5 /g for high shedders in either species, 8, 3 and 9 samples were classified<br />

as low, medium and high shedders respectively in cattle and 9, 5 and 2 were similarly classified in goats.<br />

This information was vital in establishing the applicability of the PFC study findings. There was also a<br />

correlation between the maximum dilution positive in PFC and the estimated shedding rate. These estimates<br />

included an allowance for processing losses estimated to be 250-300 fold/g faeces based on earlier studies. 5<br />

As all animals shedding > 5 x 10 5 Map/g were positive at the highest PFC dilution (1:50), estimates of the<br />

minimum number of viable Map in the Bactec inoculum required for positive culture were limited to 11 cattle<br />

and 14 goats excreting < 5 x 10 5 Map/g. From these (which included 5 cattle and 7 goats still positive at the<br />

1:50 dilution), Bactec culture was capable of detecting < 20 viable Map/g from an inoculum (Table 1).<br />

5<br />

Table 1. Detection limits of radiometric culture from 11 cattle and 14 goats excreting < 5 x 10 Map/g faeces<br />

Detection limit Detection limit<br />

Mean/g < 4.9 x 10 3<br />

Mean/g < 3.7 x 10 3<br />

Processing loss/g 250 x Processing loss/g 300 x<br />

Map in inoculum < 19.8 Map in inoculum < 12.3<br />

Discussion & conclusions<br />

From known processing losses, a minimum shedding rate of 2.5-3 × 10 2 Map/g of undiluted faeces would be<br />

required based on a single organism in the inoculum. The above results indicated that detection of < 3.7 x<br />

10 3 Map/g is readily achievable. Since these results allow populations to be categorised by growth rate in<br />

Bactec media, and more easily define populations as low, medium and high shedders, the same approach<br />

can be used to better evaluate the expected sensitivity of diagnostic tests that rely on or correlate with<br />

shedding rates. Further, these results indicate that to improve detection rates of low shedder animals using<br />

Bactec culture, an increase in the final number of Map cells in the Bactec inoculum is obligatory. Studies to<br />

capture larger numbers of Map cells or reduce losses during decontamination are therefore warranted.<br />

References<br />

1. Eamens GJ, et al (2007). Aust Vet J 85, 243-251<br />

2. Eamens GJ, et al (2007). Vet Microbiol 119, 184-193<br />

3. Eamens GJ (Unpublished). Final research report to Meat and Livestock Australia, 2006. Project PSHIP.184A<br />

4. Reddacliff LA, et al (2003). Appl Environ Microbiol 2003; 69, 3510-3516<br />

5. Reddacliff LA, Vadali A, Whittington RJ. (2003). Vet Microbiol 95, 271-282<br />

6. van Shaik G, et al (2003). J Vet Diag Invest 15, 233-241<br />

Supported in part by Meat and Livestock Australia<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A STREAMLINED WORKFLOW FOR RAPID AND SENSITIVE DETECTION OF Mycobacterium avium<br />

subsp. paratuberculosis (MAP) IN BOVINE FECAL SAMPLES BY REAL-TIME PCR<br />

D. A. Myers 1 , Q. Hoang 1 , R. Shah 1 , I. M. Moon 1 , R. C. Willis 1 , W. Xu 1 , A. M. Burrell 1 , W. Ge 1 , M. Bounpheng 1 , X. Fang 1 , J. El-Attrache 1* ,<br />

and L. Effinger 2<br />

1 Applied Biosystems, Austin, TX, 2 Animal Health Lab, Oregon Department of Agriculture, 635 Capitol St. NE, Salem, OR 97301, *John<br />

El-Attrache, Applied Biosystems., 2130 Woodward Street, Austin, Texas 78744, Ph: (512) 651-0200 ext.6605 , Fax: (512) 651-0201,<br />

email: john.el-attrache@appliedbiosystems.com<br />

Johne’s disease is a chronic wasting disease of ruminants, caused by an incurable infection of the intestinal<br />

tract by the bacterium Mycobacterium avium subsp. paratuberculosis (MAP). Currently, the laboratory<br />

standard for diagnosing Johne’s disease is fecal culture. Cultures of samples taken from a herd’s<br />

environment are also used to identify a herd’s overall risk for Johne’s disease. Unfortunately, due to the<br />

slow growth of the MAP organism, cultures typically take between 6 and 16 weeks to determine MAP<br />

presence. This allows more time for infected animals to shed MAP into the environment, increasing the risk<br />

of infection to healthy animals in the same herd.<br />

Quantitative real-time PCR (qPCR) provides a much faster solution for the detection of MAP from animal<br />

feces with results available within hours. Applied Biosystems has developed an integrated workflow,<br />

consisting of streamlined sample preparation, high throughput nucleic acid isolation and purification, and<br />

qPCR for detection of the bovine strain of MAP in bovine fecal samples as well as environmental samples<br />

from cow alleyways and manure pits. The MAP DNA isolation protocol combines the use of chemical and<br />

mechanical cell lysis with the Ambion ® MagMAX technology - a rapid, high-throughput magnetic beadbased<br />

nucleic acid isolation and purification method - to purify MAP DNA with minimal reaction inhibitors<br />

commonly found in animal feces. The purified nucleic acid is analyzed using our AgPath-ID MAP reagent<br />

kit on the Applied Biosystems 7500 Real-Time PCR system. To minimize false negatives, our MAP assay<br />

contains XenoDNA, a unique nucleotide sequence that serves as an internal process control to monitor the<br />

presence of qPCR inhibitors and reagent functionality.<br />

Using 80 field samples with known MAP culture status, our workflow method resulted in 100% sensitivity and<br />

specificity, out-performing the currently available commercial fecal DNA isolation methods and MAP qPCR.<br />

In addition, we identified all samples in the 2007 NVSL Johne’s Check Test with 100% accuracy using our<br />

MAP detection method. We also tested 87 bovine fecal samples that grew ≤1 colony of MAP per culture<br />

tube on Herrold’s egg yolk medium with mycobactin J and antibiotics (HEY) resulting in 78 samples detected<br />

as MAP positive samples by our method. With our method 17 out of 19 fecal samples with growth of ≥100<br />

MAP colonies on HEY culture were easily distinguishable from all fecal samples we tested that grew ≤30<br />

MAP colonies on HEY culture (n=145). This allows samples with high quantities of MAP to be distinguished<br />

consistently from medium to low MAP positive samples, providing reliable data for determining the risk an<br />

animal presents to the rest of a herd.<br />

Performance of our MAP detection protocol was evaluated using 409 field samples of unknown status that<br />

were later tested by HEY culture at an independent NVSL certified laboratory. For high and medium-low<br />

MAP shedders (≥3 colonies MAP/tube on HEY culture, n = 47), this evaluation resulted in 97.9% sensitivity<br />

and 100% specificity for our MAP detection method when using HEY culture as the “gold standard.” HEY<br />

culture results for low MAP shedders (


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AUTOMATED DNA ISOLATION: AN ADVANTAGEOUS TOOL IN MOLECULAR DIAGNOSTIC OF<br />

DERMATOPHYTOSIS<br />

Jose L. Blanco *, Sergio Alvarez, Patricia Alba, Marta E. García<br />

Dpto Sanidad Animal. Facultad de Veterinaria. UCM. 28040 Madrid. Spain.<br />

Introduction:<br />

The development and standardization of new techniques of DNA isolation from fungi is urgently needed. The<br />

use of automated procedures is an attractive alternative, and results in maximum efficiency when large<br />

numbers of specimens are handled. In this work, an automated system of DNA isolation from canine hairs<br />

experimentally infected by dermatophytes was compared with a manual protocol.<br />

Material & Methods:<br />

Hair samples from a healthy dog were experimentally infected by M. canis and T. mentagrophytes cultures.<br />

For the experimental infection, a small amount of hair was inserted into plate cultures of M. canis and T.<br />

mentagrophytes grown on Sabouraud agar containing 0.5 g l -1 of chloramphenicol (Sigma-Aldrich, St. Louis,<br />

USA) and 0.5 g l -1 of actidione (Sigma-Aldrich) that had been previously incubated for 12 d at 30ºC. After<br />

incubation at 30ºC for 72 h, approximately 5 mg of infected hairs were introduced into 1.5 ml Eppendof<br />

tubes. After the experimental infection, fungal DNA was isolated by two different procedures: a phenolchloroform<br />

method (manual protocol) and the automated system QuickGene-810 (Fujifilm, Japan). This<br />

automated system relies on pressured filtration technology through a porous membrane (50μm) that<br />

immobilizes nucleic acids due to their hydrophilic nature. The kit used (QuickGene DNA, Tissue Kit S) was<br />

initially designed for DNA extraction from different tissue samples, and has proven to be valid for isolation of<br />

fungal DNA. These procedures were compared at three levels: yield, purity of the DNA extract and time<br />

requirement of the protocol. To determine the quantity and quality of the DNA, the absorbance at 260 nm<br />

and the ratio of absorbances at 260 and 280 nm were measured.<br />

Results:<br />

The quantity and quality of DNA obtained in both procedures were suitable for the molecular techniques<br />

used in our laboratory to diagnose dermatophytosis (Table 1). Although a significatively lower yield was<br />

obtained with the automated procedure than with the manual protocol, purity of the DNA extracts was higher<br />

when DNA was isolated from M. canis infected hairs by the automated system. Moreover, the use of the<br />

automated procedure saved up to 22.5 minutes per sample with respect to the manual protocol. This<br />

procedure also reduces the time for which the direct intervention of a technician is required, which could<br />

compensate for the initial investment in the acquisition of the apparatus and the cost of the isolation kits. In<br />

addition, standardization of the extraction protocol is obtained, which is desirable in any laboratory.<br />

Discussion & Conclusions:<br />

The automated procedure of DNA extraction presents clear advantages: reduces considerably the handling<br />

time, prevents the use of nocive chemicals, minimizes the risk of cross-contaminations and improves the<br />

repeatability of the DNA isolation process. These multiple advantages of the automated DNA extraction<br />

procedure make it suitable for its application to the molecular techniques used in laboratorial diagnostic of<br />

dermatophytosis.<br />

Table 1: Comparison of procedures for DNA extraction from A. fumigatus mycelium and conidia<br />

Sample type<br />

Mycelium<br />

Conidia<br />

Extraction procedure<br />

*<br />

n<br />

Yield<br />

(µg DNA/mg sample)<br />

X ± SD<br />

Purity<br />

(DO260/DO280)<br />

X (range)<br />

M 4 3.46 ± 1.00 2.02 (1.98-2.06)<br />

A 4 0.91 ± 0.44 † 2.12 † (2.08-2.17)<br />

M 4 0.02 ± 0.01 1.48 (1.14-1.71)<br />

A 4 0.12 ± 0.06 † 1.38 (1.26-1.50)<br />

* Extraction procedure: M (manual), A (automated).<br />

† Significant difference (p < 0.05) between extraction procedures.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF REAL TIME PCRS TO DETECT IMPORTANT<br />

BACTERIAL PATHOGENS OF THE HORSE.<br />

S. North*, P. R. Wakeley, N. Mayo, J. Mayers, J. Sawyer.<br />

Technology Transfer Unit, Biotechnology Department, Veterinary Laboratories Agency, New Haw, Addlestone, Surrey, England.<br />

Introduction<br />

We have developed three real time TaqMan ® PCRs to detect the causative organisms of two important<br />

bacterial diseases of the horse. Both of these diseases, and their control, have a significant economic<br />

impact on the equine industry and the welfare of horses. Two PCRs were developed to detect the causative<br />

agents of equine metritis, Klebsiella pneumoniae and Pseudomonas aeruginosa. Another PCR was<br />

designed to detect the causative agent of horse strangles, Streptococcus equi equi. These PCRs were<br />

designed to detect their target organisms directly from swabs.<br />

Material & methods<br />

Two real time PCRs detected two of four causative organisms of CEM, Klebsiella pneumoniae and<br />

Pseudomonas aeruginosa, straight from swabs (we have previously validated a real time PCR to detect the<br />

other two, Taylorella equigenitalis and Taylorella asinigenitalis 1 ). The PCR to identify K. pneumoniae<br />

amplified a 95bp region of the 23S rDNA genes, the P. aeruginosa and S. equi equi PCRs amplified 131bp<br />

and 143bp fragments of the gyrase B gene. Another PCR detected Streptococcus equi equi, the causative<br />

organism of strangles, straight from swabs. For all three PCRs, swabs were firstly used to inoculate culture<br />

medium before a crude DNA extraction was performed, enabling a comparison with conventional culture<br />

methods. A control 16S rDNA PCR was run simultaneously with the diagnostic PCRs to determine whether<br />

the DNA extraction was successful. PCRs were tested against a panel of bacterial species, including horse<br />

commensals such as Rhodococcus equi and Oligella urethralis, to determine their analytical specificity. The<br />

sensitivity and efficiency of each PCR were also calculated.<br />

Results<br />

The PCRs did not cross-react with any of the 36 different bacterial organisms tested, including closely<br />

related species and known horse commensal bacteria. DNA was successfully extracted from nasal, abscess<br />

and genital swabs. All of the PCRs successfully identified a panel of positive and negative samples. The<br />

Pseudomonas aeruginosa PCR can detect as little as 20.3 gene copies and has an efficiency of ~100%.<br />

Discussions & conclusions<br />

We have successfully developed real time PCRs to identify two of the causative bacteria of equine metritis<br />

and horse strangles. These tests, alongside that previously published, provide a fast and accurate means of<br />

bacterial identification enabling rapid diagnosis and treatment.<br />

References<br />

1 Wakeley, P.R., et al. 2006. Development of a real time PCR for the detection of Taylorella equigenitalis<br />

directly from genital swabs and discrimination from Taylorella asinigenitalis. Vet. Microbiology. 118, 247-<br />

254.<br />

�<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ENHANCED CULTURAL SENSITIVITY OF STAPHYLOCOCCUS AUREUS FROM BOVINE MASTITIS<br />

MILK BY SAMPLE FREEZING AND CENTRIFUGATION<br />

1 Msoud Ghorbanpoor, 2 Seed Goraninejad and 3 Mansoreh Keshavarzi<br />

1 Department of Pathobiology, 2 Department of Clinical Science, 3 Graduate of, Faculty of Veterinary Medicine, Shahid Chamran<br />

University, Ahvaz, Iran.<br />

Bacterial culture of some milk samples may yields no bacterial growth due to presence of low colony forming<br />

unite of bacteria in the samples. These false negative results are more likely to occur with coliforms and<br />

S.aureus. Zecconi et al (1997) reported a 94% increased recovery rate of S. aureus from sediment after<br />

centrifugation of bovine mastitis milk. Villanueva et al (1991) reported a 1.5 times increased recovery rate of<br />

S. aureus by pre-culture freezing. Pre-culture freezing and incubation may also increased S. aureus recovery<br />

rate (sol et al, 2002). In attempts pre-culture freezing and centrifugation were examined to increase the<br />

recovery rate of S. aureus from milk of clinical bovine mastitis.<br />

Culture of milk from 60 affected quarters were done by standard techniques (0.01 ml of samples streaked on<br />

sheep blood agar). The result of this method was compared with that of pre-culture centrifugation (1500g for<br />

5 minutes), pre-culture freezing (12 hours at -56˚C) and pre-culture freezing and centrifugation.<br />

S. aureus were isolated from 22(36.6%) samples by standard method. Pre-culture freezing or freezing and<br />

centrifugation of these samples reveal 30(50%) positive S. aureus infection, in the other hand, freezing<br />

cause's detection of 8 additional S. aureus positive samples.<br />

Pre-culture centrifugation significantly increased the count of recovered S.aureus. Freezing enhanced<br />

(P


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MULTILOCUS SEQUENCE ANALYSIS AS A TOOL FOR IDENTIFICATION OF VIBRIO HARVEYI-<br />

RELATED ISOLATES FROM THE LARVAL REARING SYSTEM OF THE TROPICAL ROCK LOBSTER<br />

PANULIRUS ORNATUS<br />

A. Cano Gomez 1,2 , M.R. Hall 1 , L. Owens 2 , D.G. Bourne 1 , L. Høj 1 *<br />

1 Australian Institute of Marine Science, PMB 3, Townsville MC, QLD 4810; 2 Department of Microbiology and Immunology, James Cook<br />

University, Townsville, QLD 4811<br />

Introduction<br />

The development and sustainability of the crustacean aquaculture sector requires a fast and reliable<br />

technique for the detection of pathogenic vibrios in order to manage potential infections. At the Australian<br />

Institute of Marine Science (AIMS) we currently work towards closing the life cycle of the tropical rock lobster,<br />

Panulirus ornatus, which is a potential high value aquaculture candidate in Australia. The species has an<br />

extended larval phase (6 months), which makes controlling the microbial hatchery environment particularly<br />

challenging. Vibrio harveyi and related species are relevant pathogens of reared crustacean and fish causing<br />

severe economic losses due to high mortality rates (Haamed et al. 1996, Karunasagar et al.1994).<br />

Conventional biochemical tests and analysis of the 16S ribosomal RNA marker gene are unable to<br />

differentiate between these species as they share many phenotypic and genotypic characteristics (Gómez-<br />

Gil et al. 2004). To solve this problem we used a Multilocus Sequence Analysis (MLSA) which included<br />

screening of the housekeeping genes 16S rRNA, recA, rpoA, pyrH, dnaJ, and the V. harveyi toxR and luxN<br />

genes to obtain a precise identification of V. harveyi-related strains isolated from the larval rearing system of<br />

P. ornatus during mass mortality events.<br />

Materials & methods<br />

DNA was extracted from V. harveyi-related strains isolated from different compartments of the larval rearing<br />

tank of P. ornatus at AIMS. PCR amplifications of the rpoA, recA, and pyrH genes, the dnaJ gene, the toxR<br />

gene and the luxN gene were based on Thompson et al. (2005), Nhung et al. (2006), Conejero and<br />

Hedreyda (2003), and Bassler et al. (1993), respectively. Sequenced PCR products were aligned and<br />

phylogenetic trees were constructed using ContigExpress, AlignX, (Vector NTI Advance Software,<br />

Invitrogen) and Mega 3.1 Software. In addition, for each gene and strain the closest relatives were identified<br />

using the BLAST database algorithm available from NCBI.<br />

Results<br />

PCR products from the housekeeping genes were visualized in agarose gels as individual bands with the<br />

expected size for all tested strains. After BLAST search the isolates were identified as either V. harveyi, V.<br />

campbellii or V. rotiferianus with sequence identities of 99-100% for the 16S rRNA, rpoA, and pyrH genes,<br />

97-98% for the recA gene and 96-99% for the dnaJ gene. The identity of the two isolates identified as V.<br />

harveyi strains by the MLSA was confirmed by specific amplification of the V. harveyi toxR and luxN genes.<br />

The luxN was also amplified for two strains identified as V. campbellii but these sequences showed only 96%<br />

homology to the luxN gene of V. harveyi in the databases.<br />

Discussions and conclusions<br />

This study used a MLSA approach including the 16S rRNA, recA, rpoA, pyrH, dnaJ, toxR and luxN genes to<br />

specifically identify closely related Vibrio strains as either V. harveyi, V. campbellii or V. rotiferianus. The<br />

housekeeping genes included in the analysis have been reported as the most appropriate phylogeny<br />

markers for the identification of V. harveyi-related species (Thompson et al., 2005; Nhung et al., 2006).<br />

Identification based on the dnaJ gene was however not consistent with the other genes. This discrepancy<br />

with the literature is potentially due to the limited number of sequences included in previous studies. The V.<br />

harveyi luxN primers amplified the gene also for strains identified as V. campbellii. The luxN gene codes for<br />

one of the components of the quorum sensing system of V. harveyi but the results suggest that the gene<br />

may not be specific of this bacterium. This study suggests that a MLSA is required for a definitive<br />

identification of V. harveyi-related species and it will form the basis of a potential diagnostic assay for the<br />

detection and monitoring of Vibrio populations associated to the larval rearing system of P. ornatus and other<br />

aquaculture species.<br />

References<br />

Bassler BL, Wright M, Showalter RE, Silverman MR, 1993. Mol. Microbiol. 9: 773-786.<br />

Bourne DG, Høj L, Webster NS, Swan J, Hall MR, 2006. Aquaculture 260 (1-4): 27-38.<br />

Conejero MMU, Hedreyda CT, 2003. J. Appl.Microbiol.95: 602-611<br />

Gómez-Gil B, Soto-Rodríguez S, García-Gasca A, Roque A Vazquez-Juarez R, Thompson FL, Swings J 2004. Microbiology 150: 1769-<br />

1777.<br />

Haamed ASS, Rao PV, Farmer JJ, Brenner FWH, Fanning GR, 1996. Aquac. Res.27: 853-863.<br />

Karunasagar I, Pai R, Malathi GR, Karunasagar I, 1994. Aquaculture 128: 203-209.<br />

Nhung PH, Shah MM, Ohkusu K, Noda M, Hata H, Sun XS, Iihara H, Goto K, Masaki T, Miyasaka J, Ezaki T 2006. Syst. Appl.<br />

Microbiol. 30 (4): 309-315.<br />

Thompson FL, Gevers D, Thompson CC, Dawyndt P, Naser S, Hoste B, Munn CB, Swings J 2005 Appl Environ Microbiol 71: 5107-<br />

5115.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INDONESIA VETERINARY LABORATORY CAPACITY BUILDING PROJECT FOR HIGHLY<br />

PATHOGENIC AVIAN INFLUENZA<br />

S Gibbs*#, N Widowati†, A Axell*, R Lunt*, C Groocock‡, L Lauerman‡, A Foord*, I Pritchard*, T Taylor*, P Selleck*, M Johnson*, E<br />

Siregar†, B Poermadjaja†, and P Daniels*<br />

*Australian Animal Health Laboratory, LI, CSIRO, Geelong, Victoria, Australia; †Directorate of Animal Health, Department of Agriculture,<br />

Jakarta, Indonesia; ‡United States Department of Agriculture, APHIS, Jakarta, Indonesia<br />

# presenting author, poster submission<br />

Introduction<br />

In 2006, the Australian Animal Health Laboratory began a project in collaboration with the Disease<br />

Investigation Centers (DICs) of the Directorate General of Livestock Services (Indonesia), Balitvet<br />

(Indonesia), and the United States Department of Agriculture to ensure that highly pathogenic avian<br />

influenza diagnostic testing in Indonesia, and the surveillance programs which rely on that testing, achieve<br />

and sustain international best practice standards in managing the threat of pandemic influenza. Through this<br />

work, the Surveillance and Epidemiology and Laboratory Services elements of the Indonesian National<br />

Strategic Work Plan are supported.<br />

Objectives<br />

The objectives of the project are to support the use of laboratory testing capabilities for on-going and durable<br />

surveillance programs that investigate outbreaks, monitor the spread of highly pathogenic avian influenza,<br />

and monitor vaccination coverage, as well as promote the use of the OIE reference laboratory to analyze<br />

changes in the genetic composition of circulating avian influenza viruses.<br />

Methods<br />

Real-time RT-PCR units were purchased and installed in each of eight regional veterinary laboratories (DICs<br />

and Balitvet). Training, support, reference materials, and proficiency testing for real-time RT-PCR and<br />

serological testing (HI and ELISA) were provided to each laboratory. Formal training sessions in<br />

epidemiological techniques, on-site support visits, and field exercises for sample collection, biosafety and<br />

biosecurity training were also delivered.<br />

Results<br />

The first round of proficiency testing by the laboratory capacity building project has shown a good level of<br />

capability in the regional veterinary laboratory network with both HI serology and real-time RT-PCR. The<br />

real-time RT-PCR is ready for use in routine diagnosis of AI in Indonesia, with ongoing support from the<br />

project. Epidemiological training has helped to refine field procedures (blood, tissue, and swab collection),<br />

improve the use of personal protective equipment, increase understanding of data collection and analysis<br />

approaches, improve outbreak investigation techniques, and refine the sampling strategy at each regional<br />

laboratory.<br />

Conclusions<br />

Highly pathogenic avian influenza continues to be a major concern in Indonesia, both for human and poultry<br />

health. The transmission of subtype H5N1 directly from infected birds to humans has raised concerns that<br />

the potential for an influenza pandemic is increasing. There are also concerns about the potential for field<br />

strains of avian influenza viruses to mutate with consequences for diagnostic tests, appropriateness of<br />

vaccine strains and infectivity for people. Addressing these problems will require continuing surveillance<br />

throughout Indonesia. Isolation and characterization of AI viruses from the field will serve as important<br />

guides for adjusting laboratory tests to suit current strains, as well as providing information for vaccine<br />

development and application. Serological surveillance of poultry populations will provide needed information<br />

about vaccine coverage, exposure, and immunity. Continued public education campaigns and community<br />

involvement will be vital to the success of avian influenza control efforts in Indonesia.<br />

Poster Presentations


Poster Presentations<br />

Introduction<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MANAGEMENT OF A DIVA VACCINATION PROGRAMME FOR THE CONTROL OF LOW<br />

PATHOGENICITY AVIAN INFLUENZA (LPAI) IN ITALY<br />

M. Dalla Pozza 1 *, L. Busani 1 , C Ceolin 1 , C. Terregino 1 , G. Vicenzoni 1 , L. Bonfanti 1 , G. Ortali 2 , S. Marangon 1<br />

1 Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Padova, Italy<br />

2 Gruppo Veronesi<br />

From 2000 to 2005, Italy was affected by several Low Pathogenecity Avian Influenza (LPAI) epidemics<br />

caused by viruses of the H5 and H7 subtypes. During these epidemic waves, a set of control measures and<br />

restriction policies were enforced together with a “DIVA” – Differentiating Infected from Vaccinated Animals –<br />

vaccination strategy based on heterologous vaccination (1.2). Collection, storage, and processing of all the<br />

data arising from the vaccination programme and monitoring activity were of fundamental importance in<br />

ensuring the complete control of the epidemiological situation in the vaccination area. This paper describes<br />

the features of the vaccination strategy implemented and the Information System (IS) established for the<br />

continuous monitoring of vaccination campaign activities and results.<br />

Material & methods<br />

The vaccination program was applied in a well-defined area in the northern part of Italy (Lombardia and<br />

Veneto Regions). The criteria taken into account to define the area to be vaccinated were: risk factors for<br />

virus introduction and spread; occurrence of epidemics in previous years; poultry densities; presence of<br />

highly susceptible bird species (such as turkeys) and economic impact of the restriction measures in the<br />

vaccination zone. The vaccination strategy was targeted to specific types of poultry with long life-span and<br />

high LPAI infection risk (meat turkeys and layers). According to EU legislation (3) and guidelines a<br />

vaccination approach able to distinguish between vaccinated and infected birds through the use of a “DIVA”<br />

strategy was implemented. This strategy was based on the use of a vaccine containing a virus strain of the<br />

same H type as the field virus but with a heterologous neuraminidase. Heterologous monovalent and<br />

bivalent vaccines were selected depending on which virus was circulating. An intensive monitoring<br />

programme was implemented to ensure a prompt identification of field-exposed vaccinated flocks. To<br />

ensure the success of the control programme, detailed information on vaccinated farms and results of the<br />

monitoring programme were collected, stored, and processed in a central database (db). This permitted the<br />

continuous monitoring of the field situation including the results of the vaccination campaign.<br />

Results<br />

From 2003 to 2006 a total of 188,858,000 doses of vaccines were delivered, 697,712 serum samples and<br />

14,666 virological samples were tested in the framework of the vaccination monitoring programmes. A<br />

continuous flow of information to the central db was implemented from: farms (data on identification and<br />

location, species, vaccination schedule and production cycle); laboratory (data from the monitoring<br />

programme); vaccine delivery unit (doses of vaccine delivered to each farm). A centralised database to store<br />

and process all the data arising from the vaccination program was developed.<br />

Discussions & conclusions<br />

The success of a vaccination campaign is correlated with the implementation of restriction measures and the<br />

monitoring programmes associated with the DIVA vaccination strategy. The development and<br />

implementation of a dedicated I.S. allows the continuous monitoring of the epidemiological situation, linking<br />

laboratory information with field data, and eventually provides the evidence of lack of virus circulation in<br />

vaccinated flocks. In addition, a dedicated I.S. is a valuable tool to manage emergency situations during<br />

LPAI epidemics.<br />

References<br />

1. European Union (2000). Commission Decision 2000/721/EC of 7 November on introducing vaccination to<br />

supplement the measures to control avian influenza in Italy and on specific movement control measures.<br />

Off.J.Eur. Communities, L291, 33-36.<br />

2. CAPUA I, MARANGON S, 2005, The use of vaccination to combat multiple introductions of Notifiable<br />

Avian Influenza viruses of the H5 and H7 subtypes between 2000 and 2006 in Italy. Vaccine 2005; 25:<br />

4987-4995.<br />

3. CAPUA I, MARANGON S, 2006, Control of Avian Influenza in poultry. Emerg. Infect. Dis. 2006; 12: 1319-<br />

1324.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ADAPTATION OF REAL TIME PCR ASSAYS FOR DETECTION OF NEW AND EVOLVING AVIAN<br />

INFLUENZA VIRUS STRAINS<br />

Introduction<br />

A.J. Foord* and H.G. Heine<br />

CSIRO Livestock Industries, Australian Animal Health Laboratory, PO Bag 24, Geelong VIC 3220, Australia<br />

The design of TaqMan real-time PCR assays for rapid detection of AI H5N1 strains is based on available<br />

sequence information at the time of assay design. AI H5N1 continues to spread and cause disease in<br />

different geographic regions and hosts. Various genetic sublineages or clades of H5N1 strains have evolved<br />

and some recent isolates have acquired multiple genetic changes in primer or probe target regions. This has<br />

led to reported test failures for established TaqMan assays. Sequence analysis of GenBank data suggests<br />

that some of the recent AI strains will not be identified by the existing TaqMan assay. The project addresses<br />

the recent test failures of established TaqMan assays. We assessed the implications of sequence mismatches<br />

in detection probes and evaluated the use of generic detection dyes such as SYBR Green to<br />

overcome test failure due to nucleotide changes in the TaqMan probe binding region.<br />

Material & methods<br />

Sequences of AI isolates where TaqMan assays have failed have been determined and analysed. The<br />

potential use of generic dyes such as SYBR Green an alternative to TaqMan has been evaluated using<br />

recent AI strains from South East Asia and a panel of laboratory reference strains.<br />

Results & discussion<br />

Real-time PCR assays using SYBR Green were able to detect strains where TaqMan assay had failed due<br />

to nucleotide mismatches in the probe region. The same technology was able to differentiate highly<br />

pathogenic from low pathogenic AI strains based on sequence differences in the hemagglutinin cleavage<br />

sites and could potentially be used for subtyping and molecular pathotyping. Real time PCR using SYBR<br />

Green was less specific than TaqMan assays and resulted in cross-reactivity of some closely related<br />

subtypes. Results have to be analysed carefully by melting curve analysis of amplified product. Generic dyes<br />

such as SYBR Green offer a robust alternative to TaqMan assays for detection of AI, esp. newly evolving AI<br />

strains that may fail in TaqMan assays.<br />

References<br />

Spackman E., Senne D. A., Myers T. J., Bulaga L. L., Garber L. P., Perdue M. L., Lohman K., Daum L. T.,<br />

Suarez D. L.. (2002). Development of a real-time reverse transcriptase PCR assay for type A influenza virus<br />

and the avian H5 and H7 hemagglutinin subtypes. J. Clin. Microbiol. 40:3256–3260<br />

Heine H.G., Trinidad L., Selleck P., Lowther S. (2007). Rapid Detection of Highly Pathogenic Avian Influenza<br />

H5N1 Virus by TaqMan Reverse Transcriptase-Polymerase Chain Reaction. Avian Diseases 51: 370-372<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

IDENTIFICATION OF A NOVEL HEMAGGLUTININ CLEAVAGE SITE IN A HIGHLY PATHOGENIC AVIAN<br />

INFLUENZA H7 FROM NORTH KOREA<br />

Introduction<br />

H.G. Heine 1* , L. Gleeson 1, 2 , G. Fusheng 3 , L. Sims 4 , L. Trinidad 1 , S. Lowther 1 , J. Bingham 1 , P. Selleck 1<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory, PO Bag 24, Geelong VIC 3220, Australia<br />

2 current affiliations: United Nations Food and Agriculture Organisation (FAO), Bangkok.<br />

3 United Nations Food and Agriculture Organisation (FAO), Beijing<br />

4 Asia Pacific Veterinary Information Services, Manuda, Queensland<br />

An avian influenza (AI) H7N7strain was isolated from an outbreak associated with high mortality in chicken in<br />

the Democratic Peoples Republic of Korea (North Korea) amidst the H5N1 epidemic in surrounding countries<br />

in 2005. Virus was submitted to the OIE regional reference laboratory at the Australian Animal Health<br />

Laboratory in Geelong for further characterisation.<br />

Material & methods<br />

Virus was propagated in embryonated eggs and the intravenous pathology index (IVPI) determined.<br />

Pathology and histology were determined in 9 and 12 week old chickens. Molecular characterisation of the<br />

virus was by real-time PCR TaqMan assays (1) for type A, H5 and H7 and by conventional PCR and<br />

sequence analysis of the hemagglutinin gene.<br />

Results<br />

The original diagnostic specimen consisted of a lung sample from which virus was propagated in<br />

embryonated eggs. The virus was highly pathogenic avian influenza (HPAI) with an IVPI of 3.0. Oral/nasal<br />

challenge resulted in high mortality within 48 to 96 hours after inoculation confirming the virulence of the<br />

virus by natural routes of infection. TaqMan assays performed on the original submitted sample and the<br />

derived allantoic fluid of the first passage in embryonated eggs were strong positive for type A and H7, but<br />

negative for H5, ruling out potentially mixed populations with H5 virus. Brain, pancreas, kidney, lung and<br />

spleen from 4 chickens infected by natural route were AI TaqMan positive by day 1-2 post infection,<br />

indicating a high virus load in all tissues. Sequence and phylogenetic analysis of the hemagglutinin placed<br />

the virus into the Eurasian lineage of H7 strains. The molecular pathotype was determined as highly<br />

pathogenic by the presence of multiple basic amino acid residues at the H0 cleavage site and a unique<br />

amino acid sequence was identified at the haemagglutinin cleavage site that was longer than previously<br />

observed in HPAI strains of the Eurasian lineage. The cleavage region contained aromatic amino acids<br />

histidine and proline previously not observed in this context.<br />

Discussions & conclusions<br />

Molecular analyses indicate that a novel HPAI has been infecting poultry on the Korean Peninsula. The virus<br />

contained a unique haemagglutinin cleavage site that was longer than those observed in HPAI strains from<br />

previous outbreaks in Australia (1997), Italy (1999) and the Netherlands (2003). The outbreak of an HPAI H7<br />

amidst the H5N1 epidemic in surrounding countries emphasizes the necessity for broad and comprehensive<br />

detection methods in avian influenza surveillance and diagnosis. The occurrence of novel changes in the<br />

virus sequence highlights the unpredictable nature of influenza virus.<br />

References<br />

(1) Heine H.G., Trinidad L., Selleck P., Lowther S. (2007). Rapid Detection of Highly Pathogenic Avian<br />

Influenza H5N1 Virus by TaqMan Reverse Transcriptase-Polymerase Chain Reaction. Avian Diseases 51:<br />

370-372<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PRELIMINARY VALIDATION OF A COMMERCIAL AVIAN INFLUENZA N1 ANTIBODY COMPETITIVE<br />

ELISA KIT THAT CAN BE USED AS PART OF A DIVA STRATEGY<br />

WG Dundon 1 , C. Terregino 1 , V. Tuttoilmondo 1 , M. Pizzuto 1 , L. Busani 2 , M. Mancin 2 ,<br />

G. Cattoli 1 , I. Capua 1<br />

1 OIE\FAO and National Reference Laboratory for Avian Influenza and Newcastle Disease, Istituto Zooprofilattico Sperimentale delle<br />

Venezie, Legnaro (PD), ITALY ;<br />

2 Centro Regionale di Epidemiologia Veterinaria, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD, ITALY<br />

1.Introduction and Objectives<br />

Key words: DIVA, Avian influenza, kit validation<br />

The use of vaccination as part of a set of coordinated measures to combat AI is deemed to be successful in<br />

achieving the goal of eradication if it allows for the “DIVA” (Differentiation of Infected from Vaccinated<br />

Animals) principle (1). These systems enable the detection of field exposure in vaccinated flocks and through<br />

this, infected flocks may be properly managed. One of the systems that enables the detection of field virus in<br />

a vaccinated population is based on the use of a vaccine containing a seed virus of the same H subtype but<br />

of a different N subtype to the field virus (eg H5N9 vaccine against H5N1 field virus). Cross-protection is<br />

ensured by the same H group and antibodies to the N of the field virus are a result of field infection. In the<br />

framework of Workpackage 4.3 of EPIZONE (DIVA diagnostics) a commercially available competitive ELISA<br />

kit (ID Screen®, ID-VET, France) which is designed to specifically detect antibodies directed against the N1<br />

antigen was validated using poultry sera from the OIE\FAO and National reference laboratory for Avian<br />

influenza and Newcastle disease, Padova Italy.<br />

2.Material and methods<br />

The ID Screen® ELISA kits were used according to the manufacturer’s instructions. The sera tested were<br />

from (A) chickens vaccinated with an H7N5 subtype virus and challenged with an H7N1 subtype virus<br />

(n=33); (B) turkeys (n=13) and chickens (n=8) negative for type A influenza virus; (C) unvaccinated turkeys<br />

infected with H7N1 subtype virus (n=41); (D) unvaccinated turkeys infected with H7N3 subtype virus (n=40).<br />

Each sera was tested in duplicate. The results obtained from the ID Screen® ELISA were compared to the<br />

indirect immunofluorescence antibody assay (iIFA) which was taken as the gold standard (2). Cohen's<br />

Kappa statistic (K) value was calculated to assess the agreement between the tests.<br />

3.Results<br />

The overall sensitivity of the ID Screen® ELISA test as compared to the gold standard iIFA was 93.0% (CI<br />

95% 85.0-98.0), 91% for chickens (CI 95% 76.0-98.0) and 95% for turkeys (CI 95% 83.0-99.0). The overall<br />

specificity of the ID Screen® ELISA test as compared to the gold standard iIFA was 100% (CI 95% 94.0-<br />

100.0), 100% for chickens (CI 95% 63.0-100.0) and 100% for turkeys (CI 95% 93.0-100.0). The K value was<br />

calculated as 0.9264 indicating “excellent agreement” between the two tests according to Landis & Koch (3).<br />

4.Discussion and Conclusions<br />

The preliminary validation of the ID Screen® ELISA test has revealed an almost perfect agreement with the<br />

iIFAT, with excellent specificity and a sensitivity of 93%. This is most probably due to the competitive nature<br />

of the test. Optimisation and further validation studies will be carried out using a larger number of sera from a<br />

larger variety of birds (e.g. ducks, quail, ostriches etc).<br />

5.References<br />

(1) Capua I, Cattoli G, Marangon S. (2004) DIVA--a vaccination strategy enabling the detection of field<br />

exposure to avian influenza.Dev Biol 119:229-33<br />

(2) Cattoli, G., Terregino, C., Brasola, V., Rodriguez, JF, Capua, I (2003) Development and preliminary<br />

validation of an ad hoc N1-N3 discriminatory test for the control of avian influenza in Italy.<br />

Avian Dis. 47:1060-2<br />

(3) Landis J, Koch G.(1977) An application of hierarchical kappa-type statistics in the assessment of majority<br />

agreement among multiple observers. Biometrics. 33:363-74.<br />

Poster Presentations


Poster Presentations<br />

Introduction<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INACTIVATION OF AVIAN INFLUENZA VIRUSES (AIVs)<br />

BY NUCLEIC ACID EXTRACTION REAGENTS<br />

Beato M.S., Milani A., Mazzacan E., Cattoli G.<br />

Istituto Zooprofilattico Sperimentale delle Venezie, Research & Development Department<br />

OIE/FAO and National Reference Laboratory for Avian Influenza and Newcastle Disease<br />

OIE Collaborating Centre for Epidemiology, Training and Control of Emerging Avian Diseases<br />

Viale dell’Università 10, Legnaro, Padova, Italy<br />

Biosafety issues related to handling and shipment for diagnostic purposes of samples potentially<br />

contaminated by avian influenza viruses (AIVs) is a matter of concern for preventing the introduction of the<br />

agent in new areas or the infection of animals and human beings. In this study, the lysis buffer of two<br />

commonly used RNA extraction kits (High Pure RNA isolation kit, Roche; RNEasy kit, Qiagen) and of the<br />

FTA (Flinders Technology Associates) filter papers have been compared for their efficacy at inactivating<br />

AIVs.<br />

Materials and methods<br />

Two Low Pathogenicity AIVs (LPAIV): H5N2 A/turkey/Italy/80 (10 5.23 EID50/100µl) and H7N3<br />

A/chicken/Italy/8000/02 (10 6 EID50/100µl); and two Highly Pathogenic AIVs (HPAIV): H5N1 HPAI<br />

A/chicken/Yamaguchi/7/2004 (10 6.74 EID50/100µl) and H7N1 HPAI A/cicken/Italy/3665/99 (10 5.75 EID50/100µl);<br />

were incubated with the reagents at +4°C, 37°C and room temperature (25°) for 4 hours, 48 hours and seven<br />

days. Before using the lysis buffers or the filter cards, their toxicity to Specific Pathogen Free (SPF) chicken<br />

embryos were tested. Each lysis buffer was diluted in PBS with antibiotics from 10- 1 to 10- 3 . The active area<br />

of FTA filter card was removed and soaked in 2 ml of PBS with antibiotics. The supernatant was used to<br />

perform the toxicity test. The results of the toxicity test and virus inactivation were assessed by virus isolation<br />

performed in SPF eggs. The amplificability of the extracted RNA was tested by Real Time RT-PCR.<br />

Results<br />

Lysis buffers (Qiagen and Roche) were able to inactivate the four AI viruses at all the temperatures and<br />

contact times evaluated. The FTA filter papers inactivated both LPAIVs at all the temperatures and contact<br />

times, but the HPAI viruses were not inactivated under all conditions.<br />

The strain H5N1 (A/chicken/Yamaguchi/7/2004) was not inactivated after 4 hours at +°4C, but was<br />

inactivated at +37°C and at room temperature with this contact time. The same strain was inactivated after<br />

48 hours and seven days at all the temperatures tested.<br />

The strain H7N1 HPAI (A/chicken/Italy/3665/99) was not inactivated after 4 hours at any of the temperatures<br />

tested. Furthermore inactivation of this strain was not achieved after 48 hours at +4°C and +37°C but was<br />

achieved at room temperature. The H7N1 HPAI was inactivated after 7 days at room temperature and at<br />

+37°C but not at +4°C. Due to the discordance of results with LPAI and HPAI strains using the FTA<br />

technology, the test was repeated twice and the results reported above were confirmed. The viral RNA<br />

extracted with the three products was amplified at all temperatures and contact times.<br />

Discussion & Conclusions<br />

Data on the inactivation of AI viruses by chemical compounds are fragmentary and not in agreement (De<br />

Benedictis et al., 2007). Here we report results on the inactivation of AI viruses by different chemical<br />

compounds: lysis buffer of two different RNA extraction kits and FTA technology.<br />

Our data indicate that the inactivation of AI viruses by both lysis buffers was reliable under all the conditions<br />

evaluated. In contrast, the inactivation of AI viruses by FTA filter papers, is dependent on the subtype and<br />

pathogenicity of viruses as different results were obtained according to the strain tested. Moreover it seems<br />

that the HPAI viruses were not inactivated by FTA filter papers at +4°C even after 7 days with the H7N1<br />

virus. At the higher temperatures a contact time of 7 days was required to fully inactivate both HPAI viruses.<br />

References<br />

De Benedictis P, Beato MS, Capua I. Inactivation of avian influenza viruses by chemical agents and physical<br />

conditions: a review. Zoonoses Public Health. 2007;54(2):51-68.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A Rapid Laboratory Diagnosis of H5N1 Avian Influenza in Saudi Arabia<br />

Huaguang Lu 1 , Yousef Mohammed 2 , Owais Ahmed Khan 2 , Mansoar Hashem 2 , Mohamed Shuaib 2<br />

1 Animal Diagnostic Laboratory, Pennsylvania State University, University Park, PA 16802, USA<br />

2 Central Veterinary Diagnostic Laboratory, Riyadh 11454, KSA<br />

An avian influenza (AI) suspicious case submission with trachea, lung, brain, intestine, and other organs<br />

collected from one dead chicken and one sick chicken of a family owned “back yard poultry” was received at<br />

the Central Veterinary Diagnostic Laboratory (CVDL) in Riyadh of Saudi Arabia in May 9, 2007. The data<br />

submission sheet described that 41 chickens were dead out of 150 total, 2 turkeys dead (total unknown),<br />

sick birds had respiratory signs. This case was treated as a high priority case at the CVDL. Six tissue<br />

specimens including trachea/lung, brain and intestine from both live and dead birds were tested positive for<br />

Flu A virus by the BinaxNOW ® Influenza A & B test, and positive for H5 subtype by the Dot-ELISA test using<br />

H5-specific monoclonal antibody. Real-time RT-PCR test on the 6 tissues was positive for H5 subtype of AI<br />

virus (AIV). Meanwhile, the 6 tissues were processed and inoculated into embryonating chicken eggs for<br />

virus isolation. A 50% of embryo death occurred within 15 hours post inoculation in next morning.<br />

Chorioallantoic fluid (CAF) samples were harvested from the dead embryos and AIV confirmation tests<br />

including BinaxNOW ® Influenza A & B test, Dot-ELISA and HA/HI tests for H5 subtype, and real-time RT-<br />

PCR test for H5 and N1 subtype were conducted on the 6 CAF samples. These tests confirmed that the 6<br />

CAF samples were all positive for H5N1 subtype of AIV. Therefore, a final laboratory diagnosis of this H5N1<br />

positive case was made within 24 hours after case submission.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SEROPREVALENCE OF H9N2 ANTIBODY IN POULTRY FARM AND<br />

SLAUGHTER-HOUSE WORKERS OF IRAN USING HI TEST<br />

Masoud Hosseini 1,3,* , Effat Alizadeh 1 , Rouzbeh Bashar 2 , Vahideh Mazaheri 2 , Mansoureh Tabatabaeian 2 , Masoumeh Tavassoti Kheiri 2<br />

1) Department of Microbiology, Faculty of Biological Sciences, Shahid-Beheshti University, 2) Influenza Unit, Pasteur Institute of<br />

Iran 3) Central Veterinary Laboratory, Iran Veterinary Organization, Tehran, IRAN<br />

Introduction<br />

A number of different subtypes of influenza A viruses have emerged as agents of avian influenza in humans<br />

including H5N1, H7N2, H7N7 and H9N2. Most cases of avian influenza infection in humans have resulted<br />

from direct or closed contact with secretions and excretions from infected birds in poultry farm and abattoir.<br />

An outbreak of H9N2 infection in poultry farms is first reported in 1998 in Iran which is now endemic and<br />

vaccination against this subtype is practicing. The aim of this study is to investigate seropositivity among<br />

people with occupational risk of exposure to poultry subjects against H9N2 virus in Tehran province which is<br />

the most populated province with the highest number of poultry farms in Iran.<br />

Material & methods<br />

Sera were collected from poultry farms (no.65) and slaughter houses (no.62) workers from Tehran province.<br />

Only 42 of sera were from vaccinated workers with European trivalent commercial influenza vaccine. Also 25<br />

serums were collected from individuals who were not working with poultry, set as non-contact group. HI<br />

assay were performed as described in WHO recommendations against locally circulating H9N2 virus.<br />

Results<br />

The total seropositivity in groups who were in contact with poultry subjects were 37 %( 48/127), whereas it<br />

was 4% (1/25) in non-contact group, i.e., 9.25 times higher. Seropositive workers in slaughter houses (52%)<br />

were 2.2 times higher than in poultry-farms (23%). Seropositive workers in visceral removing line (83%) were<br />

2.64 times higher than feather removing line workers (31.5%) in slaughter-houses. Cross reaction was<br />

observed between AntiH3 and anti H9 antibodies. This cross reaction was eliminated by adsorption of<br />

vaccinated people sera by H3N2 virus.<br />

Discussions & conclusions<br />

Our study clearly showed that poultry-to-human transmission of avian influenza A (H9N2) viruses is feasible<br />

in contact people. However, the seropositivity rate could be different as other study (2) reported only 26% in<br />

compare with 37% in our work. Interestingly, the workers who were in contact with carcass visceral showed<br />

the highest seropositivity(83%), compare with other workers who were working in feather removing line<br />

(31.5%) or who were working with live poultry (23%). This implies again that visceral from infected birds in<br />

slaughter-house is the most contaminated material.<br />

References<br />

1) Manual on Animal Influenza Diagnosis and Surveillance (2002) WHO/CSR/NCS/2002.5<br />

2) Cheng X et al. Virological and Serological Surveys for H9N2 Subtype of influenza A Virus in Chickens and<br />

men in Shenzhen city. (2002) Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi (2002) December 16<br />

(4): 319-321<br />

3) Uyeki TM et al. Lack of Evidence for Human-to-Human Transmission of Avian Influenza A (H9N2) Viruses<br />

in Hong Kong, 1999 (2002) Emerging Infectious Diseases, Vol 8, No.2, February: 154-159<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ISOLATION OF PARAMYXO VIRUS-1 INFECTION IN PIGEON.<br />

M.D.Meshram and D.B.Sarode.<br />

Maharashtra Animal and Fishery Sciences University, Nagpur, (M.S.) India.<br />

The mortality in pigeons was observed in winter season. Mortality percentage in young ones was higher than<br />

adults. The mortality in male and female was not significant.<br />

The paramyxo virus-1 virus was isolated during the episode of mortality. The virus was measuring 220 nm in<br />

length. The virus was showing herrhingo pattern with envelop.<br />

The clinical signs were touticoulis, ataxia, greenish diarrhea, dehydration severe conjunctivitis with<br />

lacrymation and nasal discharge. The pigeons died suddenly with out any clinical signs were higher in young<br />

ones.<br />

The paramyxo virus-1 has got Zoonotic importance as the research workers suffered with infection during the<br />

period of study.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ADVANCING THE ROLE OF A REGIONAL REFERENCE LABORATORY FOR FOOT–AND-MOUTH<br />

DISEASE IN SOUTH EAST ASIA.<br />

RA Lunt 1 , W Linchongsubongkoch 2 ,LJ Gleeson 1 , R Abila 3 ,CJ Morrissy 1 ,C Leowijuk 4 ,SF Edwards 1 ,PM Le Blanc-Smith 1 and JM<br />

Hammond 1<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory, P.O. Bag 24, Geelong 3220, Australia<br />

2 Regional Reference Laboratory for FMD in South East Asia, National Institute of Animal Health, Department of Livestock<br />

Development, Pakchong, Nahkonratchasima, 30130, Thailand<br />

3 OIE SEAFMD Regional Coordination Unit, 69/1 Phaya Thai Road, Rajathevee, 10400, Bangkok, Thailand<br />

4 Department of Livestock Development, 69/1 Phaya Thai Road, Rajathevee, 10400 Bangkok, Thailand<br />

Objectives<br />

To assist development of a recently established BSL-3 containment laboratory for FMDV at Pakchong,<br />

Thailand to meet the scientific, biocontainment and quality standards necessary to operate as an<br />

international reference laboratory for FMD for ASEAN countries.<br />

Key Messages<br />

FMD is the major transboundary livestock disease affecting all sub-continental SEA countries and a<br />

significant threat to neighbouring countries including Australia. The Southeast Asian Foot and Mouth<br />

Disease Regional Reference Laboratory (SEAFMD RRL) has been the focus for the ASEAN-Australia<br />

Development Cooperation Program Regional Partnerships Scheme (AADCP RPS) project “Establishment of<br />

a reference laboratory for the Southeast Asian foot and mouth disease control program” which concluded a<br />

two-year operation in April 2007.<br />

Key outcomes from the project promoted regional and reference functions of SEAFMD RRL through:<br />

• development of a strategic plan for operation of SEAFMD RRL<br />

• a regional workshop/training course for representatives from ASEAN FMD testing laboratories<br />

• formation of LabNet, an ASEAN regional network of laboratory expertise<br />

• review and harmonisation of regional laboratory testing methods for FMD<br />

• assisting in accreditation to quality standard, engineering maintenance and the safe operation of SEAFMD<br />

RRL as a secure laboratory for work with FMD<br />

• training modules for laboratory staff at SEAFMD RRL<br />

• acting in complement with goals of the SEAFMD Campaign for regional FMD control, an initiative of World<br />

Organisation for Animal Health (the OIE or Office International des Epizooties)<br />

The project has also made successful the application to the OIE for additional support for a collaborative<br />

“twinning” process between AAHL and SEAFMD RRL. This process will strengthen the case for formal<br />

international recognition through the OIE of SEAFMD RRL as a reference centre of expertise for FMD.<br />

Conclusion<br />

Laboratory diagnosis is essential to correctly identify foot and mouth disease (FMD) in livestock, to trace the<br />

possible origin of the outbreak and to assist selection of an effective vaccine. The project assisted SEAFMD<br />

RRL meet the scientific, biocontainment and quality standards necessary for a regional reference laboratory.<br />

The project brought together as major partners the Australian Animal Health Laboratory (AAHL) and<br />

SEAFMD RRL to deliver outputs across laboratory management, technical operation and training.<br />

FMD severely impairs the potential for productivity and earnings from livestock such as buffalo, cattle, sheep,<br />

goats and pigs. The virus readily spreads by movement of infected livestock and livestock products. The<br />

effective operation of SEAFMD RRL as an ASEAN regional resource of FMD expertise is important to the<br />

success of control strategies within affected countries and for alleviation of FMD risk across the region.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

HETEROGENEITY IN THE OUTER CAPSID-CODING REGION OF FOOT-AND-MOUTH DISEASE VIRUS<br />

A AND O SEROTYPES IN AFRICA<br />

M Chitray 1,2* , B Blignaut 1,3 , FF Maree 1 , W Vosloo 1,2<br />

1 Onderstepoort Veterinary Institute, Exotic Diseases Division, Private Bag X05, Onderstepoort 0110, South Africa<br />

2 Department of Veterinary Tropical Diseases, Faculty of Veterinary Sciences, University of Pretoria, Onderstepoort 0110, South Africa<br />

3 Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0001, South Africa<br />

Introduction<br />

Foot-and-mouth disease (FMD), of which FMD virus is the causative agent, is a highly infectious disease of<br />

cloven-hoofed animals belonging to the Aphthovirus genus of the Picornaviridae family. To provide some<br />

insight into the complexity of disease control in Africa, the molecular epidemiology of FMDV in sub-Saharan<br />

Africa has focused on the SAT types in the past, despite the fact that A and O serotypes are also prevalent<br />

to the northern parts of the continent. Studies on evolutionary changes in A and O isolates in Africa are<br />

hampered due to insufficient nucleotide sequence data for the entire capsid-coding (P1) region. This led to<br />

the aim of this study i.e. to investigate the genetic variation in the P1 region of A and O serotypes from<br />

different countries and regions in Africa isolated from bovine over the last 32 years.<br />

Materials & Methods<br />

Nine O and eight A FMD viruses were chosen for P1 characterisation. Total RNA was extracted using a<br />

modified guanidium thiocyanate (GuSCN)/silica method, as described by Boom et al. (1990). The viral RNA<br />

was reverse-transcribed from the extracted total RNA using AMV reverse transcriptase enzyme (Promega)<br />

with the WDA (5’-GAAGGGCCCAGGGTTGGACTC-3’) antisense primer. The entire P1 region was<br />

amplified for all of the FMDV A and O type virus samples using two primers i.e. NCR1 (5’-<br />

TACCAAGCGACACTCGGGATC T-3’), which anneals in the 5’ non-coding region and WDA, which anneals<br />

at the 2A/2B junction. The PCR amplicons were separated on a 1.5% agarose gel, excised and purified.<br />

The P1 regions were determined using ‘genome-walking’ sequencing utilizing the ABI PRISM BigDye<br />

Terminator Cycling Ready Reaction Kit v3.0 (Applied Biosystems) as per the manufacturer’s instructions.<br />

The partial P1 sequences obtained for each virus were edited and aligned to form the complete P1 contigs<br />

using the SEQUENCHER TM 4.7 DNA sequence analysis software. Phylogenetic trees were constructed<br />

using MEGA 3 and hyper-variable regions identified using MEGA 1.<br />

Results<br />

The complete P1 sequences were successfully determined for all of the FMDV A and O viruses chosen in<br />

this study. Based on P1 phylogeny, most viruses clustered into the topotypes previously assigned using 1D<br />

data only, but differences were observed for serotype O viruses. Regions of variability were identified, which<br />

correlate with neutralisation sites that were previously determined using monoclonal antibodies and escape<br />

mutants of FMDV A and O viruses (Crowther et al. 1993, Samuel et al. 1990, Baxt et al. 1989). The RGD<br />

sequence, which is essential for receptor binding, is conserved in FMDV and this was true for the A and O<br />

types included in this study. The amino acid sequence alignment of the FMDV A types revealed a unique<br />

arginine residue neighbouring the highly conserved RGD sequence on the G-H loop of 1D.<br />

Discussions & Conclusions<br />

Phylogenetic analysis indicated that different clusters occur when utilising P1 regions compared to 1D<br />

sequences only. Alignments revealed hyper-variable regions in the outer capsid proteins, corresponding to<br />

or in close proximity to previously identified immuno-dominant sites on FMDV A and O types. Unique amino<br />

acid changes were observed in highly conserved residue positions neighboring the RGD sequence on the G-<br />

H loop of 1D. This study contributed to the expansion of the genetic database for FMDV and broadened the<br />

knowledge of the genetic variation within the capsid-coding regions of African field isolates.<br />

References<br />

Baxt, B., Vakharia, V., Moore, D.M., Franke, A.J., Morgan, D.O. 1989. Analysis of neutralizing antigenic<br />

sites on the surface of type A12 foot-and-mouth disease virus. J. Virol., 63: 2143-2151.<br />

Boom, R., Sol, C.J., Salimans, M.M.M., Jansen, C.L., Wertheim-Van Dillen, P.M.E. & Van Der Noordaa, J.<br />

1990. Rapid and simple method for purification of nucleic acids. J. Clin. Micro., 28: 495-503.<br />

Crowther, J.R., Rowe, C.A., Butcher, R. 1993. Characterisation of monoclonal antibodies against a type<br />

SAT 2 foot-and-mouth disease virus. Epidemiol. Infect., 111: 391-406.<br />

Samuel A.R., Ouldridge, E.J., Arrowsmith, A.E.M., Kitching, R.P., Knowles, N.J. 1990. Antigenic analysis of<br />

serotype O foot-and-mouth disease virus isolates from the Middle East, 1981 to 1988. Vaccine 8: 390-396.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

QUANTIFICATION OF WATER BUFFALO (Bubalus bubalis) CYTOKINE EXPRESSION IN RESPONSE<br />

TO INACTIVATED FOOT-AND-MOUTH DISEASE (FMD) VACCINE USING REAL-TIME PCR ASSAY<br />

C. N. Mingala ab *, S. Konnai a , F. A. Venturina b , M. Onuma a and K. Ohashi a<br />

a Laboratory of Infectious Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Japan<br />

b Philippine Carabao Center National Headquarters and Genepool, Science City of Munoz, Philippines<br />

Introduction<br />

Recently, there are a lot of assays introduced to measure cytokine expressions of different animal species,<br />

which are mostly protein-based tests such as ELISA, Elispot and Flowcytometry. Other tests were PCR<br />

based, which rely on the analysis of expression kinetics through electrophoresis reading. Most of these<br />

experiments engaged in in-vitro stimulation of cytokine-secreting cells. In water buffalo, cytokine expression<br />

reports were already published mainly using the latter procedure since protein based assays have its own<br />

limitations due to unavailability of specific antibodies. In this regard, real-time PCR has been developed as a<br />

simple and rapid quantification of several genes. The outcome of this study significantly confirmed the water<br />

buffalo cytokine expression in response to immunogen. This was the first time that water buffalo cytokine<br />

expression was quantified by the use of real-time PCR quantification assay.<br />

Material & methods<br />

Twelve riverine type water buffaloes were used as experimental and control animals. The treatment animals<br />

were vaccinated with Aftofor ® , an inactivated FMD strain “O” vaccine. Heparinized whole blood were<br />

collected from all of the animals at day 0 and consequently after 1, 2 and 3 weeks post-vaccination. Total<br />

cellular RNA was extracted and cDNA was synthesized. Real-time PCR was performed using a Light cycler,<br />

to quantify the cytokines (IL-2, IL12p40, IFNγ, IL-6, IL10, TNFα, IL-4) vis a vis the β-actin expression. The<br />

expression of the target cytokines was also compared with the humoral response of the animals by liquidphase<br />

blocking ELISA.<br />

Results<br />

Findings revealed that all of the cytokines were upregulated. IL-2 was immediately increased on the first<br />

week post-vaccination compared to the rest of the cytokines. Interestingly, IFNγ, IL-10 and TNFα had peaked<br />

in the last week of the observation period. TNFα significantly increased on the second week. The rest of the<br />

cytokines minimally started to increase on the first week and peaked on the following week. However, these<br />

cytokines (IL-2, IL-12p40, IL-4, IL-6) started to decline on the third week. To compare the antigen-antibody<br />

interaction with that of the cytokine expression of the experimental animals, a liquid-phase blocking ELISA<br />

was performed. The result showed that after the first week post-vaccination, all of the treatment animals<br />

reached the optimum antibody titer of more than 256.<br />

Discussions & conclusions<br />

The upregulation of IL-2 and IL-12p40 (both Th1), and drop of IL-4 (Th2) expression in a short span of time<br />

is a classic counter reaction between Th1 and Th2 cytokines. It was well noted that while IL-4 is upregulated,<br />

the IFNγ was not. IL-4 promotes the production of Th1 cytokines in company with suppressing the<br />

expression of the Th1 cytokines including IFNγ, and vice versa (Forshubor, 1996; Bembridge et al., 1998).<br />

Interestingly, TNFα and IL-6, which are also considered as Th2 cytokines, although could be suppressed by<br />

the Th1 cytokines, the synergy between TNFα to IL-6 could be a reason in maintaining its continuous<br />

expression (Frei et al., 1989; Walker et al., 2002).<br />

The early expression of IL-2 and IL-12p40 could also correspond to the action of the cell-mediated response<br />

where Th1 type cytokines are related. The subsequent development of specific immunity would be one<br />

parameter, with which the elevated IL-12p40 level after the challenge could be the reason. The continuous<br />

expression of Th2 cytokine (IL-6), which is mainly related to humoral immune responses, is connected with<br />

the progressive titer generation of the animals by the result of the LPB-ELISA. The result of this study would<br />

be an initial step towards an in-depth study of the water buffalo immune system using a real-time PCR<br />

assay.<br />

References<br />

Bembridge, G.P., Lopez, J.A., Cook, R., Melero, J.A., Taylor, G., 1998. Recombinant vaccinia virus coexpressing the F protein of<br />

respiratory syncytial virus (RSV) and interleukin-4 (IL-4) does not inhibit the development of RSV-specific memory cytotoxic T<br />

lymphocytes, whereas priming is diminished in the presence of high levels of IL-2 or gamma interferon. J. Virol. 72,4080-4087.<br />

Frei, K., Malipiero, U.V., Leist, T.P., Zinkernagel, R.M., Schwab M.E., Fontana, A., 1989. On the cellular source and function of<br />

interleukin 6 produced in the central nervous system in viral diseases. Eur. J. Immunol. 19,689-694.<br />

Forshubor, T., Yip, H.C., Lehmann, P.V., 1996. Induction of TH1 and TH2 immunity in neonatal mice. Science. 271,1728-1730.<br />

Walker, D., Jason, J., Wallace, K., Slaughter, J., Whatley, V., Han A., 2002. Spontaneous cytokine production and its effect on induced<br />

production. Clin. Diagn. Lab. Immunol. 9,1049-1056.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF AN IMPROVED CAPABILITY IN SUPPORT OF NATIONAL BIOSECURITY FOR THE<br />

SURVEILLANCE AND CONTROL OF FOOT & MOUTH DISEASE IN CATTLE AND PIGS IN VIETNAM.<br />

1 C. J. Morrissy, 2 Ngo Thanh Long, 1 J. Hammond, 1 L. Wright, 1 I. Pritchard, 1 D. Schafer, 1 W. Ha, 1 W. Goff, 2 Dong Manh Hoa, 1 M. Johnson<br />

and P, Daniels.<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory (AAHL),<br />

Geelong, Victoria, Australia.<br />

2 Regional Animal Health Centre, Ho Chi Minh City (RAHC-HCMC), South Vietnam.<br />

Objectives<br />

To determine why FMD vaccination of livestock does not give the expected protection against disease and to<br />

fully determine which serotypes of FMDV are circulating in Vietnam. This will enable better vaccination<br />

strategies to be employed. Regional diagnostic laboratories will be established with the capacity to carry out<br />

rapid and accurate FMDV serology, virus isolation and detection of antigen.<br />

Key Messages<br />

Development and implementation of disease control strategies depend upon a good understanding of FMD<br />

epidemiology which in turn requires accurate laboratory testing for both virus typing and serosurveillance.<br />

The establishment of an integrated National laboratory network is vital, with National Biosecurity being an<br />

important issue for the Vietnamese Government. Pilot zones have been established in provinces near<br />

Vietnam’s borders to gain insight into FMDV serotypes circulating in these areas. Molecular analysis of the<br />

FMDV isolates from these provinces will provide information on the effectiveness of border control and the<br />

origin of FMDV circulating in Vietnam each year. Such a diagnostic capacity for FMD will allow the early<br />

detection and identification of FMD enabling better control of disease and help reduce livestock losses and so<br />

improve productivity.<br />

Conclusion<br />

This project has established an improved diagnostic capacity for FMD in Vietnam through development of an<br />

integrated network of laboratories. Nucleotide sequence anlaysis has been carried out on 50 FMDV isolates<br />

providing vital new information on circulating viruses. The project is aligned with the National FMD plan and<br />

the information generated is aiding the control of FMD in Vietnam<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NUCLEOTIDE SEQUENCE ANALYSES OF EPIZOOTIC HAEMORRHAGIC DISEASE VIRUS (EHDV)<br />

GENOME SEGMENTS ENCODING THE OUTER CAPSID PROTEINS: A SEROLOGICAL AND GENETIC<br />

RE-EVALUATION OF SEROTYPES.<br />

Anthony, SJ*; Maan, N; Batten, C; Maan, S; Kgosana, L; Bachanek-Bankowska, K; Attoui, H; Darpel, K; Mertens, P.P.C<br />

Introduction:<br />

EHDV is an infectious, non-contagious arbovirus. It is transmitted between domestic and wild ruminants via<br />

heamatophagus midges of the Culicoides spp and can cause clinical disease in both wild and domestic<br />

species. The objectives of this study were to establish an initial sequence database for EHDV genome<br />

segments 2 and 6 for molecular epidemiology studies, to define the number and relationship of EHDV<br />

serotypes, and to develop molecular ‘typing’ assays.<br />

Methods:<br />

Full length sequences were generated for all 10 genome segments using the anchor-primer method<br />

developed by Maan, Rao et al (2007). Initial sequences were achieved using the anchor primer, and<br />

completed by gene-walking. Serological relationships between different serotypes were assessed by<br />

neutralisation assays, and compared with the molecular, phylogenetic data.<br />

Results:<br />

Full nucleotide sequence analysis and phylogenetic comparisons were completed for genome segments 2<br />

and 6 (encoding the type specific outer coat proteins VP2 and VP5 respectively) of eleven EHDV reference<br />

strains. These included isolates from each serotype, and from different geographic origins. This represents<br />

the first complete sequence database for genome segments 2 and 6 of different EHDV serotypes, providing<br />

a basis for molecular epidemiology studies. The sequence data were used to assess levels of variation in<br />

these genome segment and the proteins they encode, and were compared to results of serum-neutralisation<br />

assays for the strains analysed. This genetic and serological re-evaluation identified only 7 distinct EHDV<br />

serotypes (the isolates of existing serotypes 1 and 3 cross-neutralised and are closely related in both Seg-2<br />

and Seg-6). Strain ‘318’ was identified as a member of EHDV serotype 6.<br />

Conclusion:<br />

Serological and genetic analyses identified only seven serotypes of EHDV. The viruses previously identified<br />

as serotypes 1 and 3 appear to belong to the same Seg-2 nucleotype and serotype. The sequence data for<br />

genome segment 2 form a basis for molecular epidemiology studies and for molecular typing assays (typespecific<br />

RT-PCR).<br />

*Presenting author<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PHYLOGENETIC ANALYSIS OF AFRICAN SWINE FEVER VIRUSES FROM SOUTH AFRICA,<br />

MOZAMBIQUE AND TANZANIA FOR THE PERIOD 2001-2007<br />

RM Dwarka 1* , N Mtshali 1 , BA Lubisi 1 , OC Phiri 1,2 , ML Penrith 3,6 , A Nhamusso 3 , J Banze 3 , JIG Masambu 4 , W Vosloo 1,5<br />

1 ARC, Onderstepoort Veterinary Institute, Transboundary Animal Diseases Division, Private Bag X05, Onderstepoort, 0110,<br />

South Africa<br />

2 Novartis South Africa (Pty) Ltd, Animal Health Business Unit, P.O. Box 92 Isando, 1600, South Africa<br />

3 Instituto de Investigacao de Mozambique, Direccao de Cienca Animal, Maputo, Mozambique<br />

4 Animal Disease Research Institute, Dar Es Salaam, Tanzania<br />

5 Department of Veterinary Tropical Diseases, University of Pretoria, Private Bag X07, Onderstepoort, 0110, South Africa<br />

6 TAD Scientific, 40 Thomson Street, Colbyn, 0083, South Africa<br />

Introduction<br />

African swine fever (ASF), a highly contagious disease of domestic pigs, is caused by a virus classified into<br />

the Asfarviridae family, genus Asfivirus. It is endemic in many African countries and is characterized by high<br />

morbidity and mortality of up to 100%. When ASF outbreaks occur, it is imperative to determine the source<br />

of the infection in order to prevent future re-introductions since no vaccine or treatments are available. PCR<br />

amplification and characterisation of a 478 bp region at the C-terminal end of the p72 gene, coding for the<br />

major capsid protein, VP72, permits differentiation of ASF viruses into genotypes (Bastos et al., 2003;<br />

Boshoff et al., 2007; Lubisi et al., 2005).<br />

Material & methods<br />

DNA was extracted from tissue specimens, using a silica/guanidium-based nucleic acid extraction method<br />

(Boom et al., 1990). A PCR for molecular epidemiological analysis of ASF virus was performed, where the<br />

C-terminal region of the p72 gene was amplified using epidemiological primers P72-U<br />

(5'GGCACAAGTTCGGACATGT3') and P72-D (5' GTACTGTAACGCAGCACAG3'), as described previously<br />

(Bastos et al., 2003). Amplification products were separated on a 1.5 % agarose gel, excised and purified.<br />

The nucleotide sequence was determined by automated sequencing on an ABI Prism 310 Genetic Analyser<br />

(Applied Biosystems) using a Big Dye version 3.1 kit using the same primers as for PCR. Sequences were<br />

aligned using the DAPSA programme and neighbour joining trees constructed using MEGA 3.<br />

Results<br />

Previously, 4 genotypes were described in Mozambique and recent ASF viruses characterized from 2001-<br />

2005 clustered in 2 of these. ASF isolates obtained from Tanzania for the period 2001-2005 grouped in 4<br />

genotypes. Two consisted exclusively of Tanzanian isolates from 2001 and 2003 and are newly described<br />

genotypes while the other 2 genotypes clustered with viruses previously isolated from Kenya, Uganda and<br />

Burundi. In South Africa ASF is endemic only in the northern parts of the country where a sylvatic cycle<br />

exists, involving warthogs and argasid ticks. Part of the ASF surveillance strategy involves characterization<br />

of viruses obtained from ticks collected from warthog burrows within the control zone as well as from<br />

outbreaks in domestic pigs. All isolates characterized from pigs, ticks and warthog between 2001-2007<br />

clustered within 8 genotypes.<br />

Discussions & conclusions<br />

These studies demonstrate the way in which molecular epidemiological studies add value to diagnostic<br />

services and disease control as well as contributing to the risk assessment of ASF as information on<br />

geographic distribution of genotypes can be determined. In addition, phylogenetic trees can contribute to<br />

determining the genetic relatedness between strains. A molecular database can be useful when<br />

communicating the risk of infection to relevant stakeholders as it provides scientific proof of the presence of<br />

the risk and its possible origin which can be used to justify the management procedures.<br />

References<br />

Bastos, A.D., Penrith, M.L., Cruciere, C., Edrich, J.L., Hutchings, G., Roger, F., Couacy-Hymann, E. R.,<br />

Thomson, G. 2003. Genotyping field strains of African swine fever virus by partial p72 gene characterisation.<br />

Arch Virol, 148:(4): 693-706.<br />

Boom, R., Sol, C.J., Salimans, M.M.M., Jansen, C.L., Wertheim-Van Dillen, P.M.E. & Van Der Noordaa, J.<br />

1990. Rapid and simple method for purification of nucleic acids. J. Clin. Micro, 28: 495-503.<br />

Boshoff CI, Bastos AD, Gerber LJ, Vosloo W. 2007. Genetic characterisation of African swine fever viruses<br />

from outbreaks in southern Africa (1973-1999). Vet Microbiol, 121(1-2): 45-55.<br />

Lubisi BA, Bastos AD, Dwarka RM, Vosloo W. 2005. Molecular epidemiology of African swine fever in East<br />

Africa. Arch Virol, 150(12):2439-52.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EFFORTS TO CONTROL AND ERADICATE BOVINE TUBERCULOSIS IN A WILDLIFE RESERVIOR IN<br />

MICHIGAN, USA: A PRELIMINARY SUCCESS STORY<br />

S. D. Fitzgerald 1 , D. J. O’Brien 2 , S. M. Schmitt 2 , and J. B. Kaneene 3<br />

1 Diagnostic Center for Population & Animal Health, Department of Pathobiology and Diagnostic Investigation, College of Veterinary<br />

Medicine, Michigan State University, Lansing, MI 48910<br />

2 Wildlife Disease Laboratory, Michigan Department of Natural Resources, Lansing, MI 48910<br />

3 Center for Comparative Epidemiology, College of Veterinary Medicine, Michigan State University, Lansing, MI 48910 USA<br />

Bovine tuberculosis (TB) has been recognized in wild white-tailed deer (Odocoileus virginianus) as its<br />

principal reservoir host since 1994 in Michigan (MI), USA. Ongoing surveillance and control measures for<br />

the last 12 years have involved the cooperation of state, federal and university scientists in a multiinstitutional<br />

task force. We report here the methods and results of our efforts.<br />

To date, over 150,000 free-ranging deer have been tested for TB, with over 550 infected individuals<br />

identified. Approximately 98% of positive deer have originated within seven contiguous counties located in<br />

the northeastern lower peninsula of MI. The vast majority of deer tested have been voluntarily harvested by<br />

hunters. Hunters submit heads from their deer at road-side check stations, where data are entered on<br />

gender, age, and location of harvest. Heads are then forwarded to the Diagnostic Center where parotid,<br />

submandibular and medial retropharyngeal lymph nodes are examined. Those with suspicious lesions are<br />

harvested for routine histopathology, acid-fast staining, and mycobacterial isolation and identification. 1<br />

Control strategies have focused on reducing deer densities through increased hunter harvesting, and<br />

decreased human-caused congregation of deer by restriction or elimination of baiting and supplemental<br />

feeding. Significant decreases in disease prevalence have been achieved with these methods. 2 All-age<br />

apparent prevalence in the core outbreak area has decreased from 4.9% in 1995 to 1.2% in 2005. Apparent<br />

prevalence in yearling deer, a crude index of the rate of new infections, has dropped from 1.9% in 1995 to<br />

0.2% in 2005. Significant decreasing trends in prevalence have been observed in both yearlings (p =<br />

0.0007) and deer of all ages (p < 0.0001) over this period. Unfortunately, during the 2006 survey a<br />

significant increase in deer tuberculosis prevalence was found. Continued efforts include development of<br />

rapid ante-mortem testing to detect positive deer which have been live-trapped in high prevalence locations.<br />

While still decades away from disease eradication, MI has made encouraging progress in its initial efforts to<br />

control and eliminate TB in free-ranging deer.<br />

References<br />

1 Fitzgerald SD, Kaneene JB, Butler KL, Clarke KR, Fierke JS, Schmitt SM, Bruning-Fann CS, Berry DE,<br />

and JB Payeur: Comparison of post-mortem techniques for the detection of Mycobacterium bovis in whitetailed<br />

deer. J. Vet. Diagn. Invest. 12:322-327, 2000.<br />

2 O’Brien DJ, Schmitt SM, Fitzgerald SD, Berry DE, and GJ Hickling: Managing the wildlife reservoir of<br />

Mycobacterium bovis: The Michigan, USA, experience. Vet. Microbiol. 112:313-323, 2006.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DETECTION OF MAJOR BACTERIAL AND VIRAL PATHOGENS FROM<br />

TRASH FISH FOR CULTUREDFLOUNDER<br />

Ji Hyung Kim a,c, *, Se Chang Park a,b,c , G.J. Heo d , Dennis K. Gomez a,b and Casiano H. Choresca Jr. a,b,c<br />

a Laboratory of Aquatic Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea<br />

b KRF Zoonotic Disease Priority Research Institute, Seoul National University, Seoul 151-742, Korea<br />

c Brain Korea 21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea<br />

d College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Korea<br />

Introduction<br />

Several bacterial and viral diseases usually take place during feeding of trash fish to cultured flounder<br />

especially in Korea. The aim of this study is to know the possibility of trash fish-direct infection to cultured<br />

flounder. Target pathogens are Streptococcus iniae and S. parauberis (streptococcosis); iridovirus (iridovirus<br />

infection) and betanodavirus (viral nervous necrosis).<br />

Material & methods<br />

Twenty-six different species of trash fish samples were collected from 11 different hatcheries in Jeju Island in<br />

November 2006. Pooled organs (liver, kidney for Streptococcus sp.; eye, brain for betanodavirus; gills,<br />

spleen for iridovirus) were collected aseptically, homogenized and processed for detection of pathogens<br />

using polymerase chain reaction (PCR) based method. Positive iridovirus sample was analyzed by<br />

phylogenetic tree.<br />

Results<br />

Streptococcus iniae (300 bp) PCR positive results were obtained from Pacific cutlass fish Trichiurus lepturus,<br />

mysidacea Neomysis awatschensis and mixed fish samples. S. parauberis (718 bp) and Iridovirus (1299 bp)<br />

PCR positive results were obtained from 2 and 1 mixed fish samples, respectively. Phylogenetic analysis of 1<br />

iridovirus strain based on the partial nucleotide sequence (355 bases) portion of the MCP gene was identical<br />

and homologous with other known fish iridovirus: seabass and red seabream (98.3%); rock bream (97.4%);<br />

turbot and Korean flounder (95.7%). Betanodavirus was negative for PCR result.<br />

Discussions & conclusions<br />

These results indicate that trash fish poses a serious danger for the spread of diseases to the mariculture,<br />

and can be one of the sources or carrier of pathogenic microbial agents (bacterial and viral) for cultured<br />

flounder infection in Korea.<br />

References<br />

1. Ghittino C, Latini M, Agnetti F, Panzieri C, Lauro L, Ciappelloni R, Petracca G. Emerging pathologies in<br />

aquaculture: effects on production and food safety. Vet. Res. Comm. 2003, 27, 471-479.<br />

2. Kim WS, Oh MJ, Jung SJ, Kim YJ, Kitamura SI. Characterization of an iridovirus detected from cultured<br />

turbot Scophthalmus maximus in Korea. Dis. Aquat. Org. 2005, 64, 175-180.<br />

3. Baeck GW, Kim JH, Dennis KG, Park SC. Isolation and characterization of Streptococcus sp. from<br />

diseased flounder (Paralichthys olivaceus) in Jeju Island. J. Vet. Sci. 2006, 7(1), 53-58.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SURVEY ON SALMONELLA SPP INFECTION IN SLAUGHTER PIGS IN SASKATCHEWAN BY<br />

CULTURE AND PCR<br />

N. Atashparvar 1 ,R.C. Mainar-Jaime 2 , M. Chirino-Trejo 2<br />

1)-Dept. of Microbiology, School of Veterinary Medicine, Lorestan University, Khorram-Abad, Iran<br />

2)-Department of Veterinary Microbiology, WCVM, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4<br />

In Canada salmonellosis ranks as the second most common bacterial foodborne illness in humans, and pork<br />

and its products a possible source of infection. We carried out a survey in Saskatchewan to collect baseline<br />

information on the prevalence of Salmonella infection from slaughter pigs through bacteriological culture and<br />

PCR. Salmonella spp was isolated from 13.9% (95%CI=9.2%-18.6%) and 5.4% (95%CI=2.3%-8.5%) of the<br />

caecal content and lymph nodes samples, respectively. When PCR was used the prevalence of Salmonella<br />

infection from caecal content was significantly higher (32.6%; 95%CI=26.2%-39%). The large differences<br />

observed suggested that the accuracy of both techniques should be evaluated on field samples before<br />

deciding which one is more advantageous for surveillance purposes. While among all samples analysed<br />

main Salmonella serotypes detected (Derby –23.1%-, Typhimurium var. Copenhagen -15.4%-, and<br />

California -7.7%-) were also commonly found in pigs from other areas of Canada, it was also detected a high<br />

prevalence of the serotype Enteritidis (20.5%) which is commonly associated with infection in humans. A<br />

larger survey would be advisable to determine the extent of this serotype in the province and its potential<br />

implication in human infection. Antimicrobial resistance, although detected to some common antibiotics<br />

(ampicilline, chloramphenicol, tetracyclin, and trimethoprim/sulfamethoxazole), appeared to be a lesser<br />

problem compared to overall AR results in Canada.<br />

Key Words: Salmonella, PCR, Pig, Slaughter house.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EXPERIMENTAL TRANSMISSION OF TRANSMISSIBLE SPONGIFORM ENCEPHALOPATHIES<br />

(SCRAPIE, CHRONIC WASTING DISEASE, TRANSMISSIBLE MINK ENCEPHALOPATHY) TO CATTLE<br />

AND THEIR DIFFERENTIATION FROM BOVINE SPONGIFORM ENCEPHALOPATHY<br />

A.N. Hamir, R.C. Cutlip, J.M. Miller, R.A. Kunkle, J.A. Richt, J.J. Greenlee, E.M. Nicholson, M.E. Kehrli.<br />

Virus and Prion Diseases of Livestock Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA<br />

50010, USA.<br />

Introduction: Experimental cross-species transmission of TSE agents provides valuable information for<br />

identification of potential host ranges of known TSEs. This report provides a synopsis of TSE (scrapie, CWD,<br />

TME) transmission studies that have been conducted in cattle and compares these findings to those seen in<br />

animals with BSE.<br />

Materials & Methods: Generally 6-month-old bull calves were obtained and assigned to inoculated and<br />

control groups. Inoculated calves were housed in a Biosafety Level 2 isolation barn at the National Animal<br />

Disease Center (NADC), Ames, Iowa. Calves were inoculated intracerebrally with 1 ml of a 10% TSE brain<br />

inoculum.<br />

Results: Results of various TSE cattle experiments with intracerebral inoculation of scrapie, CWD and TME<br />

are shown in tabular form (Table 1).<br />

Table 1. Comparison of experimental scrapie, chronic wasting disease (CWD) and transmissible spongiform<br />

encephalopathy (TME) in cattle inoculated by the intracerebral route during first passage of the inocula.<br />

Scrapie CWD TME<br />

Abnormal CNS signs Anorexia, weight loss, leg Anorexia, weight loss, occasional Variable hyperexcitability with<br />

and back stiffness. Some showed aimless circling, listlessness and occasional falling to the ground.<br />

incoordination and posterior excited by loud noises. Some showing circling and<br />

weakness. Eventual severe lethargy. aggressive behavior.<br />

Incubation (survival) time 14 – 18 months. 23 – 63 months. 13 – 16 months.<br />

Attack rate 100% CWD from mule deer: 38%. 100%<br />

CWD from elk: 86%.<br />

Histopatholgic lesions Some vacuolation and central of Isolated vacuolated neurons, a Extensive vacuolation of neuronal<br />

chromatolysis of neurons*. few degenerate axons, and a mild perikarya and neuropil. Presence of<br />

astrocytosis. mild multifocal gliosis.<br />

Western blot (brainstem) All three isoforms of PrP res present. All three isoforms of PrP res seen. All three isoforms of PrP res seen.<br />

Immunohistochemistry<br />

PrP res in lymphoreticular<br />

tissues. Not present. Not present. Not present.<br />

PrP res in CNS Present within perikaryon and Multifocal distribution with Diffusely present and usually evenly<br />

processes of neurons. labeling primarily in glial cells distributed in neuropil.<br />

(astrocytes).<br />

Conclusions: 1. All three TSEs agents (scrapie, CWD and TME) are capable of propagating in<br />

cattle tissues when administered intracerebrally.<br />

2. All three TSEs can be distinguished from each other and from BSE when inoculated<br />

intracerebrally by histopathology, immunohistochemistry and Western blot techniques.<br />

Poster Presentations


Poster Presentations<br />

Objectives<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

TSE SURVEILLANCE IN AUSTRALIA: A REGIONAL LABORATORY PERSPECTIVE<br />

E. Houston<br />

Biosecurity, Department of Primary industries & Fisheries, Queensland<br />

The aim of this project was to evaluate Australia’s ability to implement enhanced TSE surveillance in a<br />

regional laboratory. The Animal Disease Surveillance Laboratory (ADSL) in Toowoomba was subcontracted<br />

by Animal Health Australia to conduct this work. This followed validation by the Australian Animal Health<br />

Laboratory (AAHL) at Geelong of the BioRad TeSeE ELISA kit method, for the purification and detection of<br />

resistant prion proteins. This method was validated for use under Australian conditions.<br />

Key messages<br />

• ADSL is the only regional facility in Australia conducting TSE surveillance using this method<br />

• The facility was upgraded from PC2 to derogated PC3 to perform this work<br />

• Equipment and reagents were purchased by ADSL<br />

• Procedures and methods were evaluated on site<br />

• The validation phase of the project involved the testing of 200 samples<br />

• The working phase of the project involved the testing of 2000 samples from throughout Australia<br />

• Accreditation with NATA was achieved<br />

• Requirements for future work were highlighted<br />

Conclusions<br />

ADSL was able to renovate an existing PC2 laboratory to derogated PC3. Equipment was purchased and<br />

installed and procedures and methods implemented to test bovine and ovine brain stem material, sourced<br />

from around the country, using the TeSeE Bio-Rad ELISA. The laboratory’s scope of accreditation by NATA<br />

to ISO/IEC 17025 in the field of Veterinary Testing was extended in 2006 to include prion detection by<br />

immunological methods in production animals. Test validation was completed on 200 samples provided by<br />

AAHL, followed by the testing of 2000 surveillance samples. The information gained from this project was<br />

reported to Animal Health Australia.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

BOVINE SPONGIFORM ENCEPHALOPATHY PREVENTION PROGRAM IN ARGENTINA.<br />

SURVEILLANCE OF BRAIN SAMPLES<br />

F.J.Blanco Viera*; E.L.Weber; F.Delgado; G.Pinto; F.Capellino; L. Jiménez; M.C.Tagle; Gl. Francinelli ; Cl. Moreno; B.J.Carrillo<br />

Laboratorio Nacional de Referencia. CICVyA. INTA Castelar Buenos Aires Argentina C.C 77 (1708) Morón<br />

Introduction<br />

The surveillance program, together with other activities such as the risk analysis are the best tools for a<br />

country to make a preventive study and show its sanitary status. One important issue of the surveillance<br />

program is the continuous monitoring of brain samples. In Argentina this activity has been carried out since<br />

1992, and during this time it was modified to qualitatively and quantitatively improve the results, according to<br />

the international requirements. The objective of this presentation is to show the surveillance activities of the<br />

TSE National Reference Laboratory of Argentina, with special emphasis in the last seven years<br />

Material & methods<br />

First, a surveillance network was designed, with the main purpose of collecting brain samples for analysis,<br />

but also to make contacts with the actors of the cattle production system. The network was formed by official<br />

institutions (Secretary of Agriculture, Fishery and Food, National Agrifood Quality Service, National Institute<br />

of Agricultural Technology, County authorities, rabies control centers), Universities, official and private<br />

diagnostic laboratories, veterinary practitioners, federal and provincial slaughterhouses and abattoirs,<br />

professionals related with the agricultural production. At the beginning of the Program, the whole encephalic<br />

mass was collected, preserving its integrity and separating it from the craneal bone structure following preestablished<br />

methods; sampling through the foramen magnus was also implemented (3), and the technique<br />

was transferred to the members of the surveillance network. The encephalic samples were analyzed by. a)<br />

histopathology, following the procedures established at the Spongiform Encephalopathies Diagnosis<br />

Workshop III, Veterinary Laboratory Agency (VLA), Weybridge, United Kingdom (UK) (3). In some cases<br />

immunohistochemistry was applied, following procedures accepted by the International Animal Health<br />

Organization (OIE) (1); b) Immunochemical analysis for detection of the infectious prion protein (PrP Sc ),<br />

using the “traditional” Western Blot method following the protocol of the VLA Weybridge, UK (1,3), and the<br />

rapid tests Prionics Check Western and Enfer (Abbott).<br />

Results<br />

Between 1992 and 2006 a total of 20,995 brain samples from bovines of different provinces and different<br />

subpopulations were collected. During the last seven years 16,797 bovine brain samples were analyzed. The<br />

distribution of samples according to age, sampling categories and points asigned to each samples, following<br />

the suggestions of OIE (2) were showed. The summing up of the points these past seven years equals<br />

521,342 points, figure that exceeds the suggestions of OIE for a type A surveillance (2). No lesions<br />

compatible with BSE were observed. The biochemical analysis for PrP Sc detection were all negative.<br />

Discussions & conclusions<br />

These negative results in 100% of microscopical examination and biochemical analysis for PrP Sc detection<br />

allow the conclusion that BSE is not present in the country. The continuity of the surveillance program, plus<br />

the other measures taken by the organisms in charge of surveillance for animal diseases will secure this<br />

condition and it will allow the international recognition of Argentina as a BSE free country.<br />

References<br />

1-0ffice International Des epizooties (OIE). Manual of Standards for Diagnostic Test and Vaccines. 5th<br />

Edition 2004 ISBN 92-9044-632-3<br />

2-0ffice International Des epizooties OIE - Bovine Spongiform Encephalopathy – Terrestrial Animal Health<br />

Code – Chapter 2.3.13 y 3.8.4 16 th Edition. 2007. ISBN 978-92-9044-696-5 www.oie.int<br />

3)-Wood J.L.N.; Done S.A.; Bradley R. “Diagnosis of BSE and Scrapie”. Spongiform Encephalopathy<br />

Diagnosis Workshop III. Central Veterinary Laboratory Weybridge England oct. 1997.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

COMPLETE AUTOMATION OF A BSE POST MORTEM DIAGNOSTIC TEST<br />

P. Marguerat 1 , J. Rodriguez 2 , S.Koller 3 , V. Leathers 3 , M. Zalunardo 4<br />

IDEXX Laboratories<br />

1 Eragny- sur Oise, France, 2 Barcelona, Spain,<br />

3 Westbrook, Maine, USA, 4 Rydalmere, New South Wales, Australia<br />

The IDEXX Herdchek* BSE Antigen and BSE-Scrapie Antigen Test kits have recently been validated for full<br />

automation from the starting tissue homogenate tube. These are the only commercial BSE kits on the market<br />

capable of automating the entire protocol of sample preparation and EIA after tissue homogenization. This is<br />

possible because the assay only involves pipetting tissue homogenate onto a microtiter plate with no sample<br />

processing requirements (no proteinase K). This unique feature has allowed us to propose a “front-end<br />

automated system” with high throughput which guarantees full traceability of samples that is not possible<br />

with other systems on the market. Since sample preparation is the rate-limiting step in all TSE assays and<br />

due to the fact that samples arrive continuously during the day, our automation system allows for continuous<br />

loading mode as compared to batch mode. The equipment reads the barcode of the tubes just before the<br />

transfer of the homogenate to the dilution plate, guaranteeing full sample traceability. The robot automatically<br />

changes tips during sample transfer or if there is a clogged tip. The robot controls all pipetting and incubation<br />

steps. In addition the data is recorded and can be transferred directly to the LIMS. The fully automated<br />

IDEXX Herdchek* BSE Antigen test was validated with 100% sensitivity and 100% specificity.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EMERGENCE OF CANINE PARVOVIRUS (CPV-2C) IN NORTH AMERICA: 2006<br />

S. Kapil*, E. Cooper, B. Murray, L. Johnston III, C. Lamm, G. Rezabek, G. Campbell, B. Johnson<br />

Oklahoma Animal Disease Diagnostic Laboratory, Farm and Ridge Road, Stillwater, OK 74076, USA<br />

Introduction<br />

Canine parvovirus (CPV) is the most significant cause of viral enteritis in puppies after 2 weeks of age. There<br />

are several variants of CPV: 2, 2a, 2b, and 2c that have evolved over time. In 2001, a new variant (CPV-2c)<br />

was first reported in Italy in clinically-affected puppies (Buanavoglia et al., 2001). CPV-2c has been detected<br />

in Europe, Asia, and South America. To our knowledge, there are no published reports of CPV-2c from<br />

Australia, and Africa. There are recent reports of of detection of CPV-2c in the USA (Hong et al., 2007 and<br />

Kapil et al., 2007).<br />

Materials and Methods<br />

Many cases were submitted with the history of CPV vaccination and/or lack of reactivity in commercial CPV<br />

diagnostic test but the clinical condition and lesions were compatible with CPV. Total DNA was extracted<br />

from fecal, intestinal, and tongue samples from cases suspected of CPV. The PCR protocol has been<br />

described (Desario et al., 2005). PCR was followed by sequencing to detect the CPV genotypes circulating<br />

in the USA during Feb. 2006-August, 2007. A 583 base pair PCR product was amplified, and sequenced.<br />

Codon positions 426, 494, and 572 were recorded for the CPV isolates. Fifty of the CPV samples (tongue<br />

and intestines) were inoculated in Crandell feline kidney cell line (CRFK). The presence of the CPV antigen<br />

in cell culture supernatant was detected with hemagglutination test with swine erythrocytes (1%) in PBS<br />

buffer with 1% fetal calf serum.<br />

Results<br />

A wide range of breeds of dogs were involved in these CPV cases. The distribution of CPV genotypes in<br />

OADDL cases was CPV-2 (n=4); CPV-2b (n=26) and CPV-2c (n=27). OADDL received the first case of<br />

CPV-2c from Texas, USA in February, 2006. To our knowledge, this was the first detection of CPV-2c in the<br />

USA (Kapil et al., 2007a).We found that the identification of CPV-2c in canines has increased over the last<br />

20 months to about 47% and CPV-2c is wide spread in the USA. We observed that the CPV isolates (CPV-<br />

2b) in USA have pin-point mutations that are unique to the North American CPV viruses. On codon 426, AAT<br />

(CPV-2 or CPV-2a), GAT (CPV-2b), and GAA (CPV-2c) were observed. Position 426 was used to assign the<br />

CPV genotype. In CPV-2b isolates, the codons at positions 494 and 572 were TGC and GTC, except three<br />

isolates that had TGT and GTA, respectively. In CPV-2c, the codons at positions 494 and 572 were TGT<br />

and GTA. We also observed few changes at other codon positions (431, 440, 449, 469, 562 and 573) in a<br />

few CPV isolates. We were able to propagate approximately 45% of the CPV isolates in CRFK cell line.<br />

Discussion and Conclusions<br />

In the American CPV-2c cases, many dogs had either yellow mucous diarrhea or hemorrhagic feces based<br />

on gross necropsy. Anti-CPV antibody (clone A3B10, VMRD, WA, USA) reacts well with the CPV variants in<br />

FA and IHC procedures for CPV diagnosis, including new CPV-2c variants. It is critical to monitor the CPV<br />

genotypes because the virus can infect in presence of low levels of antibodies. Unique mutations in VP 2<br />

gene of CPV can serve as “epidemiological markers” in tracing the origin of CPV. CPV-2c has spread around<br />

the globe.<br />

References<br />

Buanavoglia, C., V. Martella, A. Pratelli, M. Tempesta, A. Cavalli, D. Buonavoglia, G. Bozzo, G. Elia, N.<br />

Dacrdo, and L. Carmichael. 2001. Evidence for evolution of canine parvovirus type 2 in Italy. J. Gen. Virol.<br />

82:3021-3025.<br />

Desario, C., N. Decaro, M. Campolo, A. Cavalli, F. Cirone, G. Elia, V. Martella, E. Lorusso, M. Camero, and<br />

C. Buonavoglia. 2005. Canine parvovirus infection: Which diagnostic test for virus? J Virol. Meth. 126:179-<br />

185.<br />

Hong, C., N. Decaro, C. Desario, P. Tanner, M.C. Pardo, S. Sanchez, C. Buonovglia, and J. Saliki. 2007.<br />

Occurrence of canine parvovirus type 2c in the United States. J. Vet. Diagn. Invest. 19:535-539.<br />

Kapil, S., E. Cooper, G. Campbell, C. Lamm, G. Rezebek, and B. Johnson. February, 2007a. Canine<br />

parvovirus variants circulating in South-Central USA, Western Veterinary Conference, Las Vegas, NV.<br />

Kapil, S., E. Cooper, C. Lamm, B. Murray, G. Rezebek, L. Johnston, G. Campbell, and B. Johnson. 2007b.<br />

Canine parvovirus types 2b and 2c circulating in North American dogs: 2006-2007. J. Clin. Microbiol. (In<br />

Press).<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PREVALENT STUDY OF PARASITIC INFESTATION IN MYANMAR TIMBER ELEPHANT<br />

Tin Tin Myaing, Soe Soe Wai, Latt Latt Tun, Kyaw San Linn and<br />

Tay Zar Aye Cho<br />

Almost 12,000 elephants are populated in Myanmar and most of them are utilizing as drought power in<br />

timber production. Even recommended dose of anthelmintics were administered routinely to them, some<br />

baby elephants were infested heavy parasitic infestation in those areas. The aim of the study is to investigate<br />

the prevalence of parasites in Myanmar timber elephants and to promote the health and care of baby<br />

elephants as there was a very limited record relevant to parasitic infestation in elephants. A total of (811)<br />

fresh faecal sample was collected per rectum on every month from both adult and baby elephants from the<br />

year 2004 to 2006 from middle part of Myanmar. Sample collection and faecal examination was done every<br />

month during the three years. All faecal samples were examined for intestinal nematodes eggs and larvae<br />

by using faecal sedimentation- floatation method and identified under microscope. The sedimentationfloatation<br />

technique was followed to get a minimum estimate of the number of eggs per gram of dung, which<br />

was used as an index of parasite load.<br />

Intestinal parasites are best identified by examining adult worms Strongyloides eggs 111/811(13.7%),<br />

Strongyloides larvae 114/811(14.1%), Amphistome eggs 32/811(3.9%), Coccidia eggs 2/811 (0.2%) and<br />

Toxocara eggs 2/811 (0.2%) were identified from the year 2004 to 2006. Strongyloides eggs 11/87(12.6%),<br />

26/81(32.0%), 16/120(13.3%) and Strongyloides larva 15/87(17.2%), 25/81(30.7%) and 8/120 (6.7%) was<br />

identified in summer, rainy and winter season, respectively in the year 2004. Amphistome eggs 7/87(8.0%) ,<br />

9/81(11.1%) and 2/120(1.7%) had been observed in summer, rainy and winter season, respectively in<br />

2004.Strongyloides eggs 13/68(13.26%), 10/68(14.7%), 4/28(14.2%) and Strongyloides larva 25/98(25.5%),<br />

26/68(38.23%) and 5/28 (17.85%) was identified in summer, rainy and winter season, respectively in the<br />

year 2004 while Amphistome eggs 9/98(9.1%) , 7/68(10.29%) and 0/28( 0%) had been observed in<br />

summer, rainy and winter season, respectively in 2005. Strongyloides eggs 5/74(6.8%), 15/144(10.4%),<br />

11/110(10.0%) and Strongyloides larva 1/74(1.4%), 6/144(4.1%) and 3/110 (2.7%) was identified in summer,<br />

rainy and winter season, respectively in the year 2006 while Amphistome eggs 0/74(0%) , 7/144(4.9%) and<br />

1/110(0.9%)had been observed in summer, rainy and winter season, respectively in 2006. Ectoparasites<br />

(mites) and Hypoderma species (warble fly) were also observed. Only one Metastrongylus larvae has been<br />

identified where earthworms persisted in the pasture.<br />

The higher percentage of parasites eggs and larvae were investigated in rainy and winter season than<br />

summer season. Potential factors determining the parasitic infestations were influenced by environmental<br />

factors such as climate, humidity and temperature that affect the viability and behavior of parasite<br />

propagates and feeding. Transmission through contaminated forage may be one of the sources due to<br />

survivorship of eggs and juvenile stages of many nematodes is higher under moist conditions and because<br />

more food is consumed during the wet than in the dry season. Transmission through water may play as an<br />

important role since elephants often defecate near or in water where stream or river available at their place.<br />

Diet may influences the host’s nutritional status, and hence probably resistance to parasitic attack A<br />

moderate percentage of intestinal parasites eggs and larva identified in timber elephants may be due to the<br />

presence of large population of suitable host and favorable climate in these locations even anthelmintic had<br />

been given. Furthermore emerging parasitic zoonotic diseases transmitted from wild life should be<br />

investigated as mahout and their families are still living closed together with elephants.<br />

Key words: elephants, nematodes, Strongyloides, favourable climate, zoonoses<br />

Corresponding author: tintinmyaing@mail4u.com.mm<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE CONCEPT OF EARLY DETECTION AND RAPID RESPONSE TO TAD'S<br />

By APH, IAEA.<br />

TAD’s, i.e. highly infectious diseases with great socio-economic impact, present a major challenge to<br />

national veterinary services. Proper measures have to be put in place to monitor the absence and to alert the<br />

service in case of potential outbreaks. This implies the formation of field staff in clinical and epidemiological<br />

features of such diseases and the procurement of pen side tests for an early detection, demands direct<br />

information of the responsible veterinary laboratory for further investigations and entails a response plan<br />

involving further investigations and control measures at the veterinary and political level.<br />

To allow such a straight forward approach diagnostic tools fit for the purpose ( field or laboratory test, known<br />

diagnostic sensitivity and specificity, robustness) have to be available in assured quality and sufficient<br />

quantity. Results from the field, i.e. alert messages, have to be communicated to a “centre” where decisions<br />

on the next appropriate steps can be made without delay (Send field teams, collect samples). At the same<br />

time the responsible diagnostic laboratory needs this information to prepare for the analysis and a differential<br />

diagnosis.<br />

Molecular techniques offer the chance to identify the potential pathogens already at the field level.<br />

Communicating these results to the “centre” will allow a much easier risk assessment and a better focused<br />

response strategy. Only positive confirmation tests will enact the full response plan, but delays as with CBPP<br />

of several weeks after the alert should become history in this way.<br />

The concept of transporting mainly DNA back to the laboratory has the charm of a much lower risk of<br />

spreading disease or contaminating laboratory workers, a more stable substrate to be analysed and a much<br />

quicker test procedure for most diseases in the responsible laboratory.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

BRUCELLOSIS IN MARINE MAMMALS: VETERINARY LABORATORIES AGENCY INVOLVEMENT.<br />

E. STUBBERFIELD*, C. DAWSON, L. PERRETT, S. BREW, and J. STACK<br />

Veterinary Laboratories Agency, Woodham Lane, Addlestone, Surrey, KT15 3NB, UK<br />

Brucellosis is an important zoonotic disease usually associated with cattle, sheep goats and pigs. In 1994<br />

Brucella was isolated from four common seals (Phoca vitulina), two harbour porpoises (Phocoena phocoena)<br />

and one common dolphin (Delphinus delphis) in Scotland 1 and a captive bottlenose dolphin (Tursiops<br />

truncatus) in the USA 2 . These isolates were found to be morphologically and molecularly different to the<br />

terrestrial strains and have been proposed the names Brucella maris or B. cetacea and B. pinnipedia on host<br />

specificity 3 .<br />

There is serological evidence of brucellosis in marine mammal sp. around the world however isolation of the<br />

organism has only occurred in the northern hemisphere 4 . Brucella maris has been shown to be zoonotic by<br />

four reported cases of infection in humans. The first human case was reported in 1999, a laboratory acquired<br />

infection in the UK 5 , a further three cases have occurred as natural causes of infection 6,7 – believed to be<br />

linked with the consumption of raw seafood.<br />

The Veterinary Laboratories Agency (VLA) has been involved in the examination of marine mammals both<br />

serologically and with the culture and molecular typing of Brucella isolated from various tissues. Since the<br />

first isolations in 1994 VLA has tested 150 isolations from a variety of marine mammals, primarily from British<br />

waters, but also isolates from USA, Canada and Northern Europe. We have tested over 3900 serological<br />

samples from around the world, both captive and wild animals, by three ELISAs.<br />

This poster summarises brucellosis in marine mammals and shows the results obtained from work done at<br />

the VLA.<br />

Acknowledgements:<br />

The authors would like to thank G. Foster (SAC, Inverness, Scotland), N. Davison (VLA Truro, Cornwall) and<br />

other laboratories that have kindly supplied material.<br />

References:<br />

1. Ross, H.M., Foster, G., Reid., Jahans, K.L. and MacMillan, A.P., Brucella species infection in seamammals.<br />

(1994) Vet. Rec., 134(14):359<br />

2. Ewalt, D.R., Payeur, J.B., Martin, B.M., Cummins, D.R. and Miller, W.G. Characteristics of a Brucella<br />

species from a bottlenose dolphin (Tursiops truncatus). (1994) J. Vet. Diagn. Invest. 6:448-452.<br />

3. Cloeckaert, A., Verger, J-M. Grayon, M., Paquet, J-Y., Garin-Bastuji, B., Foster, G. and Godfroid, J.<br />

Classification of Brucella spp. isolated from marine mammals by DNA polymorphism at the omp2 locus.<br />

(2001) Microbes and Infection, 3:729-738.<br />

4. Foster G, MacMillan AP, Godfroid J, Howie F, Ross HM, Cloeckaert A, Reid RJ, Brew S and Patterson<br />

IAP. A review of Brucella sp. Infection of sea mammals with particular emphasis on isolates from Scotland.<br />

Dec(2002) Veterinary microbiology 90(1-4):563-80<br />

5. Brew, S.D., Perrett, L.L., Stack, J.A., MacMillan, A.P. and Staunton, N.J. Human exposure to Brucella<br />

recovered from a sea mammal. (1999) Vet. Rec., 144:(17)483.<br />

6. Sohn AH, Probert WS, Glasser CA, Gupta N, Bollen AW, Wong JD, Grace EM and McDonald WC. Human<br />

neurobrucellosis with intracerebral granuloma caused by a marine mammal Brucella spp. Apr(2003)<br />

Emerging infectious diseases (USA) 9(4):485-8<br />

7. Mc Donald WL, Jamaludin R, Mackereth G, Hansen M, Humphrey S, Short P, Taylor T, Swingler J,<br />

Dawson CE, Whatmore AM, Stubberfield E, Perrett LL and Simmons G Characterization of a Brucella sp.<br />

strain as a Marine Mammal Type Despite Isolation from a Patient with Spinal Osteomyelitis in New Zealand.<br />

(2006) Journal of Clinical Microbiology 44(12): 4363-70<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PREVALENCE OF METHICILLIN RESISTANT STAPHYLOCOCCUS AUREUS (MRSA) IN A<br />

VETERINARY TEACHING HOSPITAL.<br />

Magda Dunowska 1 *, David C. VanMetre 1 , Gage Patterson 1 , Scott Weese 2 , Doreene R. Hyatt 1 , Paul S. Morley 1<br />

1<br />

Animal Population Health Institute, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins,<br />

CO, 80523,<br />

2<br />

Dept of Clinical Studies, University of Guelph, Guelph, Ontario , Canada<br />

Magda Dunowska*, David C. VanMetre, Gage Patterson, Scott Weese 2 , Doreene R. Hyatt, Paul S. Morley<br />

*Current address: Department of Primary Industries, PIRVic, Victoria, Australia<br />

Introduction<br />

In general, Methicillin Resistant Staphylococcus aureus (MRSA) is regarded as a human pathogen and there<br />

are numerous publications describing problems associated with high prevalence of MRSA infections in<br />

human hospital settings. 1 However, several recent reports described MRSA isolation from animals. 2-5<br />

Although humans are a likely source for limited numbers of MRSA infections in animals, such reports raise<br />

concerns that MRSA would establish itself in the animal population. The purpose of this study was to<br />

determine the prevalence of MRSA carriage among animal patients and the extent of MRSA environmental<br />

contamination at the Veterinary Medical Center at Colorado State University (VMC-CSU).<br />

Material & methods<br />

This was a prospective study conducted over a period of 9 months between February and October 2004.<br />

Patients were chosen by convenience from both in-patients and out-patients at the VMC-CSU. A total of 473<br />

nasal swabs were collected from 450 animals, including dogs, horses and cattle. In addition, 70 udder swabs<br />

were collected from cattle. Environmental samples (n = 218) were collected using electrostatic wipes.<br />

Bacteriological swabs and electrostatic wipes were cultured for the presence of MRSA. Selected MRSA<br />

isolates were typed by pulsed field gel electrophoresis (PFGE) of bacterial DNA digested with the restriction<br />

enzyme SmaI.<br />

Results<br />

Overall, 3.5 % of all animals and 3.2 % of the environmental samples tested positive for MRSA. Hospitalized<br />

horses showed highest prevalence of MRSA shedding (5.8 %), followed by canine patients (2.6 %). None of<br />

the bovine patients were positive for MRSA. The majority of samples tested by PFGE belonged to one of the<br />

two PFGE types, classified as USA100 and USA500. Positive environmental samples were collected from<br />

the small animal areas (4 samples), large animal areas (2 samples) or mixed areas (1 sample).<br />

Discussions & conclusions<br />

Although, at present, the prevalence of MRSA carriage among veterinary patients seems to be low, the<br />

potential for emergence of MRSA as animal pathogen is concerning. 3 While humans are presumably the<br />

initial source of MRSA infection in animals, it is likely that a carrier animal may then serve as a vector for<br />

disseminating MRSA among other animals or people. The fact that identical MRSA strains have been<br />

isolated from veterinary patients and veterinary clinic personnel in several studies seems to support this<br />

hypothesis. 2,5 This may be of particular concern for pet owners who are immunocompromised or those in<br />

contact with immunocompromised people.<br />

MRSA was isolated from the environmental samples collected from VMC-CSU. Although the percentage of<br />

positive samples was low, these samples were collected from the high traffic areas. Similarly to the situation<br />

in human hospitals, appropriate infection control measures are recommended.<br />

References<br />

1. Richards MJ, Russo PL. Surveillance of hospital-acquired infections in Australia--One Nation, Many<br />

States. J Hosp Infect. 2007;65 Suppl 2:174-81.<br />

2. O'Mahony R, Abbott Y, Leonard FC, et al. Methicillin-resistant Staphylococcus aureus (MRSA) isolated<br />

from animals and veterinary personnel in Ireland. Vet Microbiol 2005;109:285-96.<br />

3. Waller A. The creation of a new monster: MRSA and MRSI--important emerging veterinary and zoonotic<br />

diseases. Vet J 2005;169:315-6.<br />

4. Weese JS. Methicillin-Resistant Staphylococcus aureus: An Emerging Pathogen in Small Animals. J Am<br />

Anim Hosp Assoc 2005;41:150-7.<br />

5. Weese JS. Methicillin-resistant Staphylococcus aureus in horses and horse personnel. Vet Clin North Am<br />

Equine Pract 2004;20:601-13.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A VIRUS NEUTRALIZATION TEST FOR THE DETECTION OF ANTIBODY TO AUSTRALIAN BAT<br />

LYSSAVIRUS<br />

KM Newberry, RA Lunt, KA McColl and T Chamberlain<br />

CSIRO Livestock Industries, Australian Animal Health Laboratory, P.O. Bag 24, Geelong 3220, Australia<br />

Introduction<br />

Australian bat lyssaviruses (ABLV) form a monophyletic group (genotype 7) with two clades that correlate<br />

with the identified biological reservoirs in flying foxes (Megachiroptera spp.) and one species of<br />

insectivorous bat (Microchiroptera Saccolaimus flaviventris). The ABLV genotype is more closely related to<br />

the terrestrial rabies virus (RABV, genotype 1) than any of the other six genotypes currently recognised, and<br />

consequently shares a common serotype (serotype 1).<br />

In published studies to date 1, 2 , the efficacy of RABV vaccination for protection against ABLV has been based<br />

on relatively limited data and has not accounted for possible differences due to ABLV variants. We have<br />

developed neutralization tests based on the FAVN for measuring antibody to pteropid and insectivorous bat<br />

variants of ABLV (ABLV-pt and ABLV-ins). These tests have been used to assess various sets of sera for<br />

antibody titres to RABV and ABLV.<br />

Materials & methods<br />

Sera: Variously obtained as sequential bleeds from animals (feline, canine, pteropid) used in ABLV<br />

experimental transmission studies (39 animals, 130 sera), pteropid field surveillance sera, various and<br />

unspecified collections (164 sera), human post RABV vaccination (118 sera) and human RABV/ABLV clinical<br />

investigations (3 individuals, 11 sera).<br />

Viruses: 96-0648 (ABLV-pt) and 96-1256 (ABLV-ins), Challenge Virus Standard RABV (CVS-11 genotype 1)<br />

were used in comparative serology tests.<br />

Serology tests: Two virus neutralization serology techniques were used, the Rapid Fluorescent Focus<br />

Inhibition Test (RFFIT) 3 and the Fluorescent Antibody Neutralization Test (FAVN) 4 . Only RABV was used in<br />

the RFFIT, while rabies, ABLV-Pt and ABLV-In all used in the FAVN.<br />

Results<br />

Summarised results from this work are:<br />

- FAVN format assays were set up and optimised for both ABLV variants<br />

- Qualitative agreement between tests (RABV RFFIT, RABV FAVN, ABLV-ins FAVN, ABLV-pt FAVN)<br />

assays ranged between 94 to 97.6%<br />

- 4/95 human sera were RABV antibody test-positive, but test-negative for one or both ABLV variants<br />

- Regression plots of ABLV against rabies titres for 95 post vaccination sera demonstrate equivalence<br />

at around 0.5 IU/ml, but with slope and intercept parameters favouring a differential for RABV at<br />

higher antibody titres<br />

Discussions & conclusions<br />

ABLV is closely related to RABV and has been associated with a rabies-like disease in both bats and<br />

humans. Vaccination for rabies is commonly used in Australia to protect bat handlers and others against<br />

potential infection with ABLV. In the absence of a specific ABLV antibody test, rabies serology tests have<br />

been used as a guide for effective response to vaccination. Furthermore, the lack of a specific assay for anti-<br />

ABLV antibody has impaired evaluation of results from assays employing heterologous virus (RABV) to<br />

measure antibody responses in animals that may have been exposed to ABLV. Our results show that the<br />

modified FAVN can be effectively applied for comparative measurement of antibody to RABV and ABLV<br />

variants. The results are significant both for appropriate test selection (e.g. in ABLV surveillance serology)<br />

and for interpretation of protective antibody levels against ABLV in rabies vaccine recipients.<br />

References<br />

1. Fraser GC, Hooper PT, Lunt RA, Gould AR, Gleeson LJ, Hyatt AD, Russell GM, Kattenbelt JA. 1996.<br />

Encephalitis caused by a lyssavirus in fruit bats in Australia. Emerg. Infect. Dis.; 2: 327-331.<br />

2. Brookes SM, Parsons G, Johnson N, McElhinney LM, Fooks AR. 2005. Rabies human diploid cell vaccine<br />

elicits cross-neutralising and cross-protecting immune responses against European and Australian bat<br />

lyssaviruses. Vaccine; 23: 4101-4109.<br />

3. Smith JS, Yager PA, Baer GM 1973. A rapid reproducible test for determining rabies neutralizing<br />

antibody. Bull. WHO, 48: 535-541.<br />

4. Cliquet F, Aubert M, Sagné L (1998). Development of a fluorescent antibody virus neutralisation test<br />

(FAVN test) for the quantification of rabies-neutralising antibody. J. immunol. Meth., 212:79-87.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF PORCINE CIRCOVIRUS (PCV) DIAGNOSTIC CAPABILITY AT AAHL ALLOWING<br />

THE DETECTION AND NUCLEOTIDE SEQUENCE ANALYSIS OF PCV FROM DISEASE OUTBREAKS.<br />

1 C. J. Morrissy, 1 D. Schafer, 1 L. Wright, 1 J. Hammond, 1 J. Bingham, 3 Nguyen Thi Thu Hong, 2 E. Neumann, 1 D. Middleton, 1 W. Goff, 1 W,<br />

Ha, 2 P, Jaros,<br />

2 L McIntyre and 1 M. Johnson<br />

1 CSIRO Livestock Industries, Australian Animal Health Laboratory (AAHL),<br />

Geelong, Victoria, Australia.<br />

2 EpiCentre, Massey University, Palmerston North, New Zealand.<br />

3 National Veterinary Company (NAVETCO), Ho Chi Minh City, Vietnam.<br />

Objectives<br />

1. To establish a diagnostic capability at AAHL for PCV.<br />

2. To collate nucleotide sequence data from Australian, New Zealander (NZ) and Vietnamese viruses<br />

isolated from pigs showing a variety of clinical signs of disease.<br />

Key Messages<br />

Postweaning multisystemic wasting syndrome (PMWS) causes severe loss of weight and mortality in weaner<br />

pigs. There is an increasing body of evidence that PCV type 2 (PCV2) is associated with PMWS. PMWS and<br />

PCV associated disease have been described in commercial pig populations in many countries worldwide<br />

including the USA, the UK, Asia and most of Europe; it has not been reported from Australia. We have<br />

isolated PCV2 from a number of samples submitted from Australia and NZ. We have also assisted<br />

NAVETCO, in Vietnam, to isolate PCV from pigs as part of a technology transfer program. Nucleotide<br />

sequence analysis has enabled comparisons with historical Australian PCV isolates and with other<br />

sequences from PMWS endemic countries.<br />

Conclusions<br />

AAHL has developed a diagnostic capability for PCV which includes virus isolation, immunodetection<br />

techniques including immunohistochemistry, PCR and nucleotide sequencing. The sequence data<br />

demonstrate that the most recent Australian PCV isolates differ from historical Australian PCV isolates and<br />

that these isolates are different from those viruses isolated in NZ.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

STUDY OF BRUCELLOSIS AMONG ABATTOIR WORKERS IN ISFAHAN, IRAN<br />

Babak Asghari*, Jamshid Faghri, Foad Abdollahpour<br />

Brucellosis is a serious zoonotic problem in most developing countries, caused by Brucella spp.Brucella can<br />

spread from animals to people. Brucellosis is an occupational hazard particularly for abattoir workers,<br />

veterinarians and farm workers. Brucellosis was determined through a sampling of 150 abattoir workers in<br />

Isfahan. Diagnosis was obtained from Rose Bengal plate test, standard tube agglutination test (STA) and<br />

blood culture. The prevalence of brucellosis was 2.1% among abattoir workers. It would also be beneficial to<br />

create awareness about brucellosis in such professional so that essential precautions and repeated<br />

screening of such occupationally exposed individual can be done. Vaccination of animals for Elimination of<br />

the infection to produce Brucella free animals can prevent the infection in humans.<br />

*Babak Asghari– Dept. of Microbiology, School of Medicine, Isfahan Medical Sciences University<br />

Post Box: 81745-359<br />

ISFAHAN-IRAN<br />

E-mail : bab.asghari@gmail.com<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

GENETIC VARIABILITY ANALYSIS OF RABIES VIRUS -SEROTYPE 1- ON ROMANIAN TERITORY<br />

M. Turcitu*, Handan Coste, St Nicolae, Gh. Barboi<br />

Institute for Diagnostic and Animal Health, Bucharest, Romania<br />

Introduction<br />

Nucleotides sequence analysis for virus isolates belonging to different geographical regions of Romania<br />

allows phylogenetic diversities screening for a particular region, as well as similarity degree especially with<br />

isolates from different european countries.<br />

Material and methods<br />

Viral genome sequencing was performed at nucleoprotein gene region, due to the relatively high genetic<br />

stability, with the pathologic material consisting of brain homogenates from different species of domestic and<br />

wild animals (2 dogs, 3 cats, 1 jackal, 4 foxes, 2 cows, 1 horse, and 1 sheep) find with rabies through direct<br />

immunofluorescence and mice inoculation.<br />

For preparation of virus RNA, commercially available kits were used (HighPure RNA Isolation Kit- Roche<br />

Applied Science, RNEasy Mini Kit- Qiagen), followed by nested PCR using primers previously described<br />

(Heaton et al). Revers Transcription and the first PCR were carried out in a single tube RT-PCR (OneStep<br />

RT-PCR kit- Qiagen), followed by second PCR. The amplicons obtained (606bp) were purified from gel<br />

agarosis using "MinElute Gel Extraction Kit- Qiagen" and subjected to direct sequencing using "BigDye<br />

Terminator Cycle Sequencing Kit- Applied Biosystems ".<br />

Results<br />

Sequences obtained were aligned using Clustal W Alignment software and a phylogenetic tree was build<br />

using Mega 3.1 software. Data analysis showed three major characteristics regarding virus circulation: virus<br />

isolates belonging to central and southern Romania (Danube natural barrier with Bulgaria) tends to form a<br />

separate cluster, without many foreign infusion of genetic material; for western and south-western isolates,<br />

due to the high phylogenetic similarities with hungarian and former Yugoslavia strains, showed the<br />

permanent exchange of genetic material through bidirectional movement of virus. Finally, eastern and northeastern<br />

isolates falls into clusters with sequences from north-eastern Europe (Finland, Estonia), probably via<br />

Ukraine.<br />

Discussions and conclusions<br />

Data obtained show two majors route of virus circulation in Romania that extends into neighboring countries<br />

to witch Romania has terrestrial borders: west and south-west with Hungary and former republic of<br />

Yugoslavia, east and north-east with Ukraine. However, Danube river (witch is the natural border with<br />

Bulgaria to the south) tends to act as an efficient barrier for virus movement, whiteout any genetic material<br />

exchange detected so far.<br />

The results obtained needs to be completed with new data from all romanian territory to have a clear view<br />

upon virus circulation; moreover, sequencing of larger fragments could be relevant for phylogenetic analysis,<br />

although with the genetic diversity detected in this study do not appear to have a decisive role.<br />

References<br />

1 PAUL R. HEATON, LORRAINE M. McELHINNEY, J. PAUL LOWINGS (1999). Detection and identification<br />

of rabies and rabies-related viruses using rapid-cycle PCR, Journal of Virological Methods, 81, 63-69.<br />

2. HERVE BOURHY, BACHIR KISSI, LAURENT AUDRY, MARCIN SMRECZAK, MALGORZATA<br />

SADKOWSKA-TODYS, KATARIINA KULONEN, NOEL TORDO, JAN F. ZMUDZINSKI, EDWARD C.<br />

HOLMES(1999). Ecology and evolution of rabies virus in Europe, Journal of General Virology, 80, 2545-<br />

25573.<br />

3. S KUMAR, K TAMURA, and M NEI (2004) MEGA3: Integrated software for Molecular Evolutionary<br />

Genetics Analysis and sequence alignment. Briefings in Bioinformatics 5:150-163.<br />

*corresponding author: e-mail Turcitu.Mihai@idah.ro<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ANALYSIS OF CYTOKINE EXPRESSION IN CULTURE OF MONONUCLEAR CELLS FROM<br />

TUBERCULOUS BOVINES<br />

1 *Díaz-Otero F., Lugo-Arriaga TM., Jaramillo-Meza L., González Salazar D., Espitia-Pinzón CI, Arriaga-Díaz<br />

1 CENID-Microbiología-INIFAP 2 INIB-UNAM<br />

The aim of this work was to evaluate expression of IFN-γ, IL-2, IL-4 and IL-10 by RT-PCR in bovines<br />

showing different reactivity in the diagnostic tests of bovine tuberculosis. Group 1 was formed by animals<br />

negative to the tuberculin skin test, ELISA and IFN-γ test, group 2 by animals positive to all tests and group 3<br />

by animals positive only to the ELISA test. Peripheral blood mononuclear cells (PBMC) were obtained from<br />

all animals and were stimulated in vitro with PPD bovis, PPD avium and Concanavalin A and assayed for<br />

cytokine expression. Expression of IFN-γ and IL-2 was evident in cells of group 1 stimulated with Con A while<br />

no significant expression of these cytokines was observed when stimulated with PPD bovis or PPD avium. In<br />

cells from group 2 stimulated with PPD bovis high expression of IFN-γ, IL-2 and IL-4 was observed; when<br />

stimulated with PPD avium expression of IL-2 was lower than with IFN-γ. In contrast, cells of group 3 showed<br />

higher expression of IL-4 and IL-10 in the presence of PPD bovis. These results indicate that as the infection<br />

progresses, the pattern of cytokine expression changes from Th1 type to Th2, which was confirmed by the<br />

presence of high levels of antibodies and no reactivity to the skin tuberculin test that was observed in group<br />

3; this immune response is usually observed in advanced stages of the disease.<br />

Supported in part by CONACYT Nº.- D43244-Z<br />

1 Corresponding author: Fernando Díaz Otero, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias. Centro Nacional<br />

de Investigación Disciplinaria en Microbiología. Departamento de Biotecnología Aplicada. Carretera México-Toluca, Km. 15.5 Col. Palo<br />

Alto, 05110, México, DF. Teléfono: 55703100 EXT.-34, Fax: 55704073, E-mail: diof0009@servidor.unam.mx<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INTER-LABORATORY EVALUATION OF A REAL-TIME PCR ASSAY FOR DETECTION OF BOVINE<br />

HERPESVIRUS 1 IN BOVINE SEMEN<br />

J. Wang 1* , J. O’Keefe 1 , D. Orr 1 , L. Loth 1 , S. Cork 1 , M. Banks 2 , P. Wakeley 2 , D. West 2 , R. Card 2 , G. Ibata 2 , C. Van Maanen 3 , P. Thoren 4 ,<br />

M. Isaksson 4 , P. Kerkhofs 5<br />

1 Investigation and Diagnostic Centre, Biosecurity New Zealand, Upper Hutt, New Zealand<br />

2 Veterinary Laboratories Agency, Weybridge, United Kingdom<br />

3 Animal Health Service, Deventer, The Netherlands<br />

4 National Veterinary Institute, Uppsala, Sweden<br />

5 Veterinary and Agrochemical Research Centre, Brussels, Belgium<br />

*Presenting author<br />

Introduction<br />

Bovine herpesvirus 1 (BoHV-1) is an important pathogen economically in cattle causing different syndromes,<br />

including infectious bovine rhinotracheitis (IBR), and infectious pustular vulvovaginitis (IPV) and<br />

balanoposthitis (IBP). BoHV-1 infected bulls are regarded as life-long carriers of the virus and may<br />

potentially shed virus intermittently in their semen. BoHV-1 is of significance to international bovine semen<br />

trade. To guarantee the safety of semen for artificial insemination, rapid, sensitive and reliable tests for virus<br />

detection are crucial. Virus isolation is routinely used for the detection of BoHV-1 in bovine semen, which is<br />

also the prescribed test for international trade by the Office International des Epizooties (OIE). However, this<br />

method is time and labour-consuming, and lacks sensitivity 1 . A more sensitive and reliable method would be<br />

valuable. We have previously developed and validated a real-time PCR assay for the detection of BoHV-1 in<br />

extended semen 2 . The PCR assay was shown to be more sensitive than virus isolation, highly specific and<br />

repeatable. The specific objectives of this study were: (1) to evaluate the test reproducibility when the same<br />

real-time PCR assay was used in different laboratories employing different personnel and instrument; (2.) to<br />

further assess the sensitivity and specificity of the real-time PCR assay.<br />

Material & methods<br />

An international inter-laboratory ring trial was performed with the participation of six laboratories from five<br />

countries. Sets of coded samples were prepared in one laboratory and distributed to each of the participating<br />

laboratories. The sample panel consisted of semen from naturally and artificially infected bulls, serial<br />

dilutions of positive semen, semen from uninfected bulls, spiked negative semen with virus, as well as serial<br />

dilutions of reference virus. The samples were tested using a previously validated real-time PCR assay for<br />

the detection of BoHV-1 in each participating laboratory 2 . The PCR tests were conducted with four different<br />

real-time PCR amplification/detection platforms. Virus isolation using one set of samples was performed in<br />

one laboratory.<br />

Results<br />

The PCR results from the participating laboratories were compared with one another, and with those of virus<br />

isolation. Analysis using к statistic showed that there was a substantial level of agreement on PCR testing<br />

results between the laboratories (к = 0.59-0.95). The sensitivity of the PCR ranged from 78.6% to 89.3%,<br />

with an overall result at 82.7%, in comparison to 53.6% for virus isolation. The overall specificity for the PCR<br />

across all six participating laboratories was 93.6%, while the specificity for virus isolation was only 84.6%.<br />

Discussion & conclusions<br />

The real-time PCR assay applied in the ring trial provided a satisfactory reproducibility when conducted by<br />

different personnel in different laboratories. The sensitivity and specificity of the PCR assay was greater than<br />

those of virus isolation. The high specificity and sensitivity of the real-time PCR assay, in combination with<br />

significant reduction of time for detecting amplified products, make it a valuable alternative to the slow and<br />

laborious virus isolation for the detection of BoHV-1 in extended semen. The real-time PCR assay has been<br />

ratified by the OIE as a prescribed test for international trade.<br />

References<br />

1. Weiblen, R., Kreutz, L., Canaboroo, T. F., Schuch, L. C., Rebelatto, M. C., 1992. Isolation of bovine<br />

herpesvirus 1 from perputial swabs and semen of bulls with balanoposthitis. J. Vet. Diag. Invest. 4, 341-343.<br />

2. Wang, J., O’Keefe, J., Orr, D., Loth, L., Banks, M., Wakeley, P., West, D., Card, R., Ibata, G., Van<br />

Maanen, C., Thoren, P., Isaksson, M., Kerkhofs, P. 2007. Validation of a real-time PCR assay for the<br />

detection of bovine herpesvirus 1 in bovine semen. J. Virolo. Methods. 144, 103-108.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

THE AUSTRALIAN NATIONAL QUALITY ASSURANCE PROGRAM (ANQAP)<br />

*PL Young, ME Beggs, LME McCauley, Department of Primary Industries, Victoria<br />

The Australian National Quality Assurance Program (ANQAP) is an international proficiency testing program<br />

for veterinary laboratories. The major focus of the program is to evaluate tests associated with disease<br />

control programs, quarantine and export health certification. Participation in proficiency testing provides<br />

laboratories with an objective means of assessing and demonstrating the reliability of the results produced.<br />

ANQAP currently offers proficiency testing for 36 different tests used for the diagnosis of 21 diseases in the<br />

areas of veterinary virology, serology and bacteriology. The tests available include agar gel immunodiffusion<br />

(AGID), enzyme linked immunosorbent assay (ELISA), virus neutralisation, complement fixation, virus<br />

isolation, microscopic agglutination (MAT) and polymerase chain reaction (PCR) tests. In the current 2007<br />

testing cycle there are 33 participant laboratories from Australia, New Zealand, Asia, Africa, Europe and<br />

Northern America.<br />

For each proficiency test, a panel of six samples is sent to each laboratory for testing. The samples are<br />

either freeze dried serum or whole blood which are reconstituted by the participant laboratory prior to testing.<br />

The panel of samples will generally be comprised of at least one negative, one low positive and one high<br />

positive sample to reflect a range of results that are likely to be found in routine diagnostic testing.<br />

Laboratories are encouraged to treat the panel as diagnostic specimens and include them in their routine<br />

testing schedule. For each assay evaluated, ANQAP collates the results reported by the individual<br />

laboratories and prepares a report that summarises all of the results reported by the participating<br />

laboratories. The ANQAP report includes a statistical analysis of the results where appropriate, and a<br />

classification of the results. Results that fall within an acceptable variation range will be classified as<br />

satisfactory. If the results demonstrate variation, the laboratory is given the opportunity to retest with a fresh<br />

vial of the same sample. Following retesting the result will be classified as either retest satisfactory, retest<br />

demonstrating minor variation or retest unsatisfactory. Laboratories receiving an unsatisfactory classification<br />

are encouraged to take corrective action.<br />

ANQAP has been providing proficiency testing since 1991. The program began with only four tests and has<br />

undergone continuous growth to offer 36 tests in 2007. The number of tests monitored by ANQAP will<br />

continue to increase as additional tests are considered. In 2006 ANQAP included the first molecular assay in<br />

the program: Bovine viral diarrhoea PCR. Other molecular tests will be included in the program over the next<br />

few years beginning with the Influenza A and Newcastle disease virus PCR tests.<br />

Objectives<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LESTER: THE USE OF A SIMULATION TOOL TO ENABLE DETAILED EVALUATION OF<br />

LABORATORY CAPACITY TO RESPOND TO AN OUTBREAK.<br />

DR JAMES WATSON<br />

To design and implement a simulation tool that would enable detailed assessment of laboratory capabilities<br />

as part of an emergency animal disease exercise.<br />

Key Messages<br />

As part of Exercise Hippolytus, a program of emergency animal disease (EAD) exercises for animal health<br />

laboratories, a tool (LESTER) was developed to allow detailed simulation of laboratory systems.<br />

This tool, based on manipulation of magnetic tokens and labels on whiteboards laid out to represent<br />

functional areas of the laboratory, allowed laboratory staff to participate directly as players in the unfolding<br />

scenario. Tokens represented actual staff members and real resources, including key reagents; availability<br />

was based on an unannounced laboratory audit. Strict time constraints were enforced to mimic realistic<br />

workflow.<br />

A spreadsheet model of key functional areas was also developed to explore the effects of management<br />

decisions and tune the simulation for maximum benefit.<br />

The simulation identified key constraints on laboratory surge capacity as well as generating feedback on<br />

ways to improve AAHL’s EAD response plan. The outcomes provided a realistic assessment of the<br />

laboratory’s surge capacity in an outbreak situation. Direct involvement of staff, not usually the case in such<br />

exercises, was a useful training exercise.<br />

Conclusion<br />

LESTER enabled detailed simulation of laboratory response to an outbreak, providing useful data on<br />

laboratory capacity and constraints as well as providing considerable input towards process improvements.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

REPRODUCIBILITY OF RESULTS IN BATCHES OF 3 ELISA KITS FOR JOHNES’ DISEASE:<br />

RECOMMENDATIONS FOR KIT EVALUATION CRITERIA<br />

J.M. Gwozdz*, M. Carajias<br />

OIE and National Reference Laboratory for Johne’s Disease<br />

Department of Primary Industries<br />

475 Mickleham Road, Attwood, Victoria 3049, Australia, phone: 61 3 9217 4277<br />

e-mail: Jacek.Gwozdz@dpi.vic.gov.au<br />

Introduction<br />

Johne's disease (JD) is a chronic, wasting disease of ruminants caused by infection with Mycobacterium<br />

paratuberculosis.<br />

Currently, there are 3 commercially available ELISA kits that are approved for testing cattle for JD in<br />

Australia. It is crucial that they show high reproducibility of results as they are used in the National Johne's<br />

Disease Market Assurance Program for Cattle. Therefore, new batches of the ELISA kits are subjected to<br />

independent evaluation to assess reproducibility of the assay performance prior to release of a kit.<br />

The objective of this study was to validate criteria for the evaluation of batches of the 3 ELISA kits.<br />

Materials & methods<br />

Three new batches of 3 ELISA kits supplied by Prionics (Switzerland), Institut Pourquier (France) and IDEXX<br />

(USA) were evaluated over a period of 3 years. A batch was considered new when there was a change in<br />

the source, production or processing of any biological component of a kit. The kits were coded to maintain<br />

confidentiality of suppliers.<br />

A panel consisting of about 180 sera from cattle from a region considered as free of JD and about 50 sera<br />

from cattle with JD was used for the evaluation of the within-plate, between-plates and between-batches<br />

variations, and specificity and sensitivity of each batch. Testing and interpretation of results were carried out<br />

following the kit manufacturer’s recommendations. Coefficient of variation (CV) of optical density (OD) values<br />

of replicates of each serum was calculated to assess the within-plate, between-plates and between-batches<br />

variations. In addition, the percentage of diagnostic agreement was used to further assess the betweenbatches<br />

reproducibility.<br />

Results<br />

The total average CVs of OD values within a plate (among wells), between plates and between batches of<br />

the three kits were 6.8% (3.73 to 9.12%), 1 9.3% (5.35 to 14.9%) and 13% (8.8 to 16.98%), respectively.<br />

The overall average agreement of diagnostic classification for all kits and batches was 99% (98 to 100%).<br />

The overall average specificity and sensitivity for all kits and batches were 99.75% (98.53 to 100%) and<br />

78.6% (70.6 to 90.9%), respectively.<br />

Data derived from this study was used to formulate acceptance criteria for the evaluation of new batches of<br />

the ELISA kits. For the within-plate, between-plates and between-batches variations, the sum of the average<br />

CV of OD values and 2 standard deviations (SD), representing the upper scale of 95% of the population, was<br />

adopted as an acceptance limit. For the percentage of diagnostic agreement, the minimum acceptance limit<br />

was established by deducting 2 SD from the overall average.<br />

In addition, data derived from this study was used to formulate the measurement of uncertainty for assay<br />

results.<br />

Discussions & conclusions<br />

The high reproducibility of results warrants the use of the 3 ELISA kits in market assurance programs to<br />

consistently assess levels of M. paratuberculosis infection in cattle herds.<br />

The recommended acceptance criteria for the evaluation of new batches of the 3 ELISA kits are as follows:<br />

• Within-plate variation < 10% CV of OD values of serum replicates<br />

• Between-plates variation < 15% CV of OD values of serum replicates<br />

• Between-batches variation < 20% CV of OD values of serum replicates<br />

• % diagnostic agreement < 98%<br />

Acknowledgment<br />

This study was supported by the Department of Primary Industries Victoria and suppliers of the ELISA kits:<br />

IDEXX, Institut Pourquier and Prionics.<br />

1 , figures in brackets indicate a range of results.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LATERAL FLOW TECHNOLOGY-PENSIDE TEST<br />

FOR THE DETECTION OF FOOT-AND-MOUTH DISEASE<br />

T Kristersson, A Nordengrahn, M Merza*<br />

Svanova Biotech AB, Uppsala Science Park, 751 83 Uppsala, Sweden<br />

Introduction<br />

Consistent with the guidelines of new policy oriented research is the development of rapid, simple tests that<br />

could be used as first line diagnostics. Pen-side tests represent tools that satisfy these requirements: their<br />

application could be useful as a first line diagnosis for Veterinarians in slaughter house, in farms and in<br />

simply equipped regional laboratories, mainly to control the spreading of viral infections when the primary<br />

outbreak has been confirmed by a Reference laboratory. The on site assays are mainly based on antigen or<br />

antibody recognition, consequently they use antibody-based detection systems.<br />

Material & methods<br />

An FMDV pan-reactive mAb was bound to colloid gold as well as immobilised on a nitrocellulose membrane.<br />

If virus is present in the sample it will bind to the gold-antibody and form an immune complex. The complex<br />

then migrates by capillary action along the membrane until it reaches the immobilised antibody on the<br />

membrane. The complex will bind, resulting in a read line.<br />

In order to evaluate the assay, inactivated viral cellcultures representing serotype A, O, C, Asia1, Sat1, Sat2<br />

and Sat3 as well as samples from Vesicular stomatitis virus (VSV), Swine disease virus (SVDV) and normal<br />

non infected cell cultures were analysed.<br />

Results<br />

Results show that all seven serotypes were detected by the assay, but with a weaker reaction with Sat1,<br />

Sat2 and Sat3 samples. No reaction was seen with VSV, SVDV or with normal cells.<br />

Discussions & conclusions<br />

We have successfully used the lateral flow technology in order to develop a test system for FMDV. By the<br />

use of a panreacting monoclonal antibody a good sensitivity and specificity was established as all seven<br />

serotypes of FMDV were recognised, while samples from other vesicular diseases such as VSV and SVDV<br />

as well as other cellculture samples did not react.<br />

The Pen-side test is considered to be a quick and easy to perform assay and our results shows that it could<br />

be of great value as the first line of diagnosis. By using the Pen-side test a yes/no answer could be obtained<br />

within 10 minutes giving the responsible Veterinarian the ability to quickly take the right precautionary<br />

measures in the matter.<br />

Further evaluations are ongoing in order to develop a new line of diagnostics for FMDV market.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF A HIGH THROUGHPUT ROBOTICS-BASED ELISA FOR THE DETECTION OF<br />

ANTIBODIES SPECIFIC FOR PARASITE WORM SPECIES IN SHEEP<br />

Introduction<br />

K. L. Tyrrell 1* & J. R. White 2<br />

1 CSIRO Livestock Industries, Locked Bag 1, Armidale, NSW 2350<br />

2 Australian Animal Health Laboratory, CSIRO Livestock Industries, Private Bag 24, Geelong, VIC 3220<br />

Traditionally ELISA have been performed manually in the laboratory with little, if any input from automated<br />

machinery due to either the sample type or the complexity of the plate formats required. Manually operated<br />

ELISA assays require scrupulous adherence to test parameters to maintain acceptable intra- and inter-test<br />

reproducibility. Assay consistency can suffer due to such factors as operator error, the throughput of large<br />

numbers of samples within short given timelines and the involvement of different operators. An existing<br />

ELISA protocol for the detection of separate immunoglobulin isotype responses in sheep plasma to the<br />

parasitic worms Haemonchus contortus and Trichostrongylus colubriformis, was adapted to perform on a<br />

Tecan Genesis 200 robotic deck. The primary aim of this activity was to enable the rapid processing of a<br />

very large number of samples in a highly consistent and reproducible manner and simultaneously investigate<br />

the robustness of a ‘roboticised’ ELISA format.<br />

Material & methods<br />

In adapting the assay to an automated format, careful consideration needed to be given to the mechanical<br />

capabilities / limitations of the robot in relation to the degree of permitted modifications within individual steps<br />

of the assay. A program for the Tecan Genesis 200 robotic deck was written to accomplish the major steps<br />

involved in the ELISA with little or no interference required by the operator. These steps included sample,<br />

antibody and conjugate addition to individual plates together with the associated washing steps involved for<br />

each step. 18 standard ELISA plates were pre-coated with antigen two days prior to testing and<br />

subsequently blocked with a !% casein solution one day prior to testing, using a PerkinElmer MultiProbe<br />

robotic deck. At the start of the ELISA assay proper, all plates were placed on the deck of the Tecan Genesis<br />

200 in the correct order for sample addition. Individual animal plasma samples were prepared in 96 deep<br />

well plates which acted as the stock supply to be aliquotted by the robotic arm to specific plates. Each<br />

sample was fully titrated across the plate. Plasma standards were also included in the deep well format and<br />

needed to be individually placed in the appropriate plates by predefined programming of the robotic arm. At<br />

the completion of the robotic steps on the deck, substrate was added manually and the reaction stopped for<br />

plates to be read on a standard plate reader. The assay was completed in approximately 7.0 hr.<br />

Results<br />

The ELISA was successfully translated to a robotics format (RELISA) with equivalent results obtained when<br />

the assay was conducted manually. Comparisons of standard curves obtained from both the robot and<br />

manual assays showed an improvement in the consistency in the robotic data between plates and between<br />

days that the assay was performed. It was determined that leaving the plates soaking in assay wash buffer<br />

(NaCl 3.85M, Na2HPO4 281mM, KCL 67mM, KH2PO4 plus 0.05% Tween 20) on the deck, for prolonged<br />

periods at the end of the assay run, did not affect the expected OD levels. This finding meant the RELISA<br />

was able to routinely process 36 plates per day and up to 54 plates per 24 hour period where a third<br />

overnight run was employed. A total of 730 plates were run in a 25 day period, providing data on 610<br />

individual animal samples.<br />

Discussions & conclusions<br />

The improved consistency of standards both within and between ‘runs’ suggests that there will be an<br />

increase in the quality of the data produced when the robotic deck is employed to process samples. The<br />

successful translation of the RELISA to a high-throughput, automated protocol has also provided invaluable<br />

insights into the various factors and parameters that need to be considered for the adaptation of other<br />

ELISA-based assays to a robotics format. At the Australian Animal Health Laboratory (AAHL) we wish to now<br />

extend this process to other existing ELISA formats in particular, those assays specific for exotic agents with<br />

the potential to cause major, widespread disease outbreaks.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LATE-PCR DETECTION OF FOOT AND MOUTH DISEASE VIRUS<br />

Kenneth E. Pierce 1 , *Rohit Mistry 2 , Scott M. Reid 3 , Juliet P. Dukes 3 , Katja Ebert 3 , Donald P. King 3 and Lawrence J. Wangh 1 .<br />

1 Brandeis University, Waltham, MA, USA<br />

2 Smiths Detection, Watford, WD23 2BW, UK<br />

3 Institute for Animal Health, Pirbright, GU24 0NF, UK.<br />

*presenting author<br />

Introduction: Foot and mouth disease (FMDV) is caused by a positive-strand RNA virus and outbreaks are<br />

highly contagious and costly. Virus detection is complicated by the presence of 7 serotypes and variation<br />

within serotypes. We aim to construct a pan-FMDV assay using a novel technology, LATE-PCR suitable for a<br />

sophisticated field instrument.<br />

Materials & Methods: The most highly conserved sequences in the 3D (RNA polymerase) gene were used<br />

to design a Limiting and an Excess Primer according to the criteria of Linear-After-The-Exponential (LATE)-<br />

PCR 1,2,3 . A one-step RT-PCR was performed using the Excess Primer to initiate cDNA synthesis. The two<br />

primers then generate an abundance of single-stranded DNA that is freely available to hybridize with a<br />

fluorescent probe at low temperature. The probe is mis-match tolerant and therefore hybridizes to sequence<br />

variants within its target sequence. Fluorescence detection during a post-PCR melt identifies and quantifies<br />

the amplification products. This assay also includes an internal control target that is amplified by the same<br />

primers and hybridizes to a probe with a different fluorophore. In a parallel experiment, a one-step real-time<br />

RT-PCR assay (designed to 3D) in routine use by national reference laboratories 4, 5, 6 was used to test<br />

identical viral RNA samples.<br />

Results: Amplification of synthetic DNA templates demonstrated that the signal at end-point was<br />

proportional to number of initial target molecules over the range 10 to 1 million copies. Serial dilutions of<br />

viral RNA also showed quantitative end-point signals, although variation was higher. A single pair of LATE-<br />

PCR primers amplified RNA for multiple strains from all 7 FMDV serotypes. All samples which contained<br />

culturable FMDV, as well as several samples with titers below culturable limits were positive. There were no<br />

false positive signals with RNA from other viruses that cause vesicular disease in livestock. Each FMDVnegative<br />

sample generated a signal from an internal control sequence, insuring against amplification failures.<br />

Comparison of the LATE and reference laboratory RT-PCR assay data showed the results were concordant.<br />

Conclusions: Our LATE-PCR pan-FMDV assay uses a single pair of primers and a single probe to detect a<br />

wide spectrum of FMDV isolates in a convenient quantitative-end-point assay suitable for a field instrument.<br />

Supported by: Smiths Detection, Inc 1, 2 and Defra UK 3 (project SE 1121).<br />

References:<br />

1. Sanchez, J.A., Pierce, K.E., Rice, J.E., and Wangh, L.J. (2004). Linear-After The Exponential (LATE)-<br />

PCR: An advanced method of asymmetric PCR and its uses in quantitative real-time analysis. Proc. Natl.<br />

Acad. Sci (USA) 101, 1933-1938.<br />

2. Pierce, K.E., Sanchez, J.A., Rice, J.E., and Wangh, L.J. (2005). Linear-After-The-Exponential (LATE)-<br />

PCR: optimizing primer design for high yields of specific single-stranded DNA and improved real-time<br />

detection. Proc. Natl. Acad. Sci (USA) 102, 8609-14.<br />

3. Pierce, K.E, and Wangh, L.J. (2007). LATE-PCR and allied technologies: Real-time detection strategies<br />

for rapid, reliable diagnosis from single cells. In: Single Cell Diagnostics, A.R. Thornhill (ed.), Humana<br />

Press, pages 65-85.<br />

4. Callahan JD, Brown F, Osorio FA, Sur JH, Kramer E, Long GW, Lubroth J, Ellis SJ, Shoulars KS, Gaffney<br />

KL, Rock DL, Nelson WM.(2202). Use of a portable real-time reverse transcriptase-polymerase chain<br />

reaction assay for rapid detection of foot-and-mouth disease virus. J Am Vet Med Assoc. 220 (11), 1636-42.<br />

5. Reid SM, Grierson SS, Ferris NP, Hutchings GH, Alexandersen S. (2003). Evaluation of automated RT-<br />

PCR to accelerate the laboratory diagnosis of foot-and-mouth disease virus. J Virol Methods 107 (2),129-39.<br />

6. King DP, Ferris NP, Shaw AE, Reid SM, Hutchings GH, Giuffre AC, Robida JM, Callahan JD, Nelson WM,<br />

Beckham TR. 2006. Detection of foot-and-mouth disease virus: comparative diagnostic sensitivity of two<br />

independent real-time reverse transcription-polymerase chain reaction assays. J Vet Diagn Invest 18, 93-7<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A HIGHLY MULTIPLEXED RT-LATE-PCR ASSAY FOR DETECTION AND ANALYSIS OF SUBTYPES OF<br />

AVIAN INFLUENZA VIRUS<br />

C. Hartshorn, A.H. Reis, Jr., J.E. Rice, L.J. Wangh<br />

Brandeis University, Waltham, MA, USA 02254-9110<br />

(email: Wangh@brandeis.edu)<br />

Introduction: Pathogenic strains of avian influenza, particularly subtypes H5N1 and H7N1 continue to<br />

spread and evolve through out Eurasia and Africa, causing death and requiring culling of millions of birds.<br />

Several hundred people have died of avian influenza primarily following direct contact with infected livestock,<br />

but there is reason to fear that relatively few genetic changes could lead to efficient human-to-human<br />

transmission raising the specter of a devastating global pandemic. Rapid accurate detection of the presence<br />

and type of influenza is needed urgently, particularly in remote rural areas where people live in close<br />

proximity with their livestock. Using novel technologies developed in our laboratory at Brandeis University,<br />

we are constructing a highly multiplexed assay that can detect and distinguish between several subtypes of<br />

avian influenza and their strains in a single tube. This assay is designed to work on an advanced “pen side”<br />

sample preparation and PCR system which is being designed and built by Smiths Detection (see C.Volpe et<br />

al in this volume).<br />

Materials & Methods: Our multiplexed Avian Influenza assay utilizes Linear-<br />

After-the-Exponential (LATE)-PCR and PrimeSafe (For more information about these technologies see<br />

Wangh et al. in this volume.) In the proof-of-principle experiments described here we utilize viral RNA<br />

sequences transcribed in vitro from plasmid construct (GenScript Corporation ) using T7 RNA polymerase.<br />

Each of these RNAs, either alone or in a multiplex, is hybridized first to one of the LATE-PCR primers (the<br />

RT primer) and is then reverse transcribed at 55°C. The second LATE-PCR primer, together with one or<br />

more fluorescently tagged probes, is then added to the cDNA, along with a thermally stabile polymerase. In<br />

accord with the logic of LATE-PCR, exponential amplification of double-stranded DNA is carried out until<br />

each limiting primer is exhausted (phase I) and then is followed by ten or more cycles of linear amplification<br />

(phase II), during which the single-stranded amplicon for each target accumulates. It is possible to follow<br />

LATE-PCR amplification in real-time by measuring probe-target hybridization after the extension step of<br />

each thermal cycle (see Wangh et al. this volume). However, in the LATE-PCR Avian Influenza assay<br />

discussed here, no real-time product detection is performed. Instead, each single-stranded amplicon is<br />

detected at end-point by dropping to temperatures at which probe-target hybridization takes place and is<br />

measured. Each of the four fluorescent colors available on the instrument is used to detect two different<br />

targets using two different probes having different hybridization temperatures.<br />

Results: Multiplexed (RT)-LATE-PCR assays are highly versatile and inclusion of PrimeSafe suppresses<br />

mis-priming and primer-dimer formation among the pairs of primers. The four-color, double-temperature,<br />

Avian Influenza assay, that we are currently constructing, is comprised of 9 pairs of primers that reverse<br />

transcribe 9 RNA sequences and then generate 10 single-stranded amplicons which are detected using 10<br />

different fluorescent probes, all in a single closed-tube. (One pair of primers is used to amplify both RNA<br />

and an internal DNA control.) Depending on which virus is present this assay will generate independent<br />

positive signals for: 1) Avian Influenza – Subtypes: H5-LPA, H5-HPA, H7-LPA, H7-HPA, N1; 2) Newcastle<br />

Disease Virus (if Avian Influenza is absent). In addition, each sample will contain an internal control, to verify<br />

that PCR amplification has occurred and will be spiked with an additional control to verify that sample<br />

preparation has been successfully achieved.<br />

Discussion & Conclusions: Once this assay is fully implemented on the “pen side” instrument,, sample<br />

preparation, addition of all reagents and PCR will all be performed by the instrument and will require no<br />

manual actions by the operator. We estimate that the total elapsed time from sample-to-read out will be less<br />

than one hour. Finally, because of the nature of the LATE-PCR amplification process, each of the singlestranded<br />

amplicons generated in a multiplex reaction of the type described here can be used for Dilute-‘N’-<br />

Go dideoxy-sequencing which is both convenient and cost-effective (see Wangh et al. this volume). This is<br />

particularly important in the case of RNA viruses like Avian Influenza in which sequence changes occur<br />

readily and can significantly alter the infectivity and evolution of the virus.<br />

References:<br />

All of our publications on LATE-PCR are available at: http://www.brandeis.edu/projects/wanghlab/<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ONE-STEP PRIMER-PROBE ENERGY TRANSFER (PRIPROET) REAL-TIME RT-PCR DETECTION<br />

OF SVDV USING ROTOR-GENE 6000<br />

M. Hakhverdyan 1 , T.B. Rasmussen 2 , Å. Uttenthal 2 , S. Belák 1<br />

1 Joint Research and Development Division, Departments of Virology, The National Veterinary Institute and the Swedish University of<br />

Agricultural Sciences, Uppsala, Sweden;<br />

2 National Veterinary Institute, Technical University of Denmark, Lindholm, Kalvehave, Denmark<br />

Introduction<br />

The aim of the present work was to develop a one-step real-time RT-PCR based on primer-probe energy<br />

transfer (PriProET) technology for sensitive and specific detection of swine vesicular disease virus (SVDV).<br />

The main importance of this exotic pig disease is that the clinical signs produced by the SVDV are<br />

indistinguishable from those caused by foot-and-mouth disease virus (FMDV) and vesicular stomatitis virus<br />

(VSV). Earlier, PriProET was successfully applied for the detection of FMDV, SVDV and VSV in two-step<br />

PCR protocol (1-4).<br />

Material and Methods<br />

Fifteen SVDV strains, FMDV O and C, VSV New Jersey and Indiana, human CV-B5 strain Faulkner used in<br />

this study were obtained from IAH (Pirbright, UK) and CISA-INIA (Valdeolmos, Madrid, Spain). The basic<br />

principle of the assay relies on fluorescence energy transfer between primer and probe. In the PriProET<br />

system, one of the two primers is labelled with a 5’-donor fluorophore (FAM) and the probe is labelled with a<br />

3’-reporter fluorophore (Texas Red or Cy5). During the PCR cycles, when the probe anneals to the extended<br />

fluorescent primer, energy transfers from the donor to the reporter due to their close proximity. The reporter<br />

emits fluorescence that is monitored and quantified. Directly following the amplification, a melting profile is<br />

generated for determination of specific probe melting peaks.<br />

Results<br />

Fifteen SVDV strains, eight of which had one or two mutations in the probe region, were successfully<br />

amplified resulting in positive amplification plots and specific melting peaks. The mutations in the probe<br />

region were reflected in the melting profile by a shift in observed melting peaks. In average, one mutation<br />

decreases probe Tm on 6 o C. Heterologous viruses like FMDV, VSV or human CV-B5 remained negative.<br />

Discussions and Conclusions<br />

The ability of PriProET to detect SVDV strains with mutations in the probe region makes it superior for<br />

analysis of unknown diagnostic specimens. For the PriProET system even a low efficient hybridising probe<br />

will bring the reporter fluorophore in proximity of the donor enabling release of reporter fluorescence. There<br />

is no competition with the stem-loop structure (as in molecular beacons) and there is no need for probe<br />

degradation to release fluorescence (as in TaqMan), which impairs detection of strains with mutations in the<br />

probe target region. In addition, the PriProET system gives a specific Tm for each SVDV strain, which can<br />

reveal mutations in the target. There is a chance to identify phylogenetically divergent strains of SVDV,<br />

which may appear negative in other probe-based real-time PCR assays. At the same time any difference in<br />

melting points may provide an indication of divergence in the probe region. The one-step SVDV PriProET<br />

assay may improve the early and rapid detection of viral nucleic acids of a wide range of SVDV strains,<br />

allowing reduced turnaround time and use of high-throughput, automated technology.<br />

Acknowledgements<br />

We thank Dr. Donald King, Dr. Scott Reid (IAH, Pirbright, UK), Dr. Montserrat Agüero, Dr. Jovita Fernandez<br />

(CISA-INIA, Valdeolmos, Madrid, Spain) for providing us material. This study was supported by the EU<br />

projects QLK2-CT-2000-00486 (Multiplex PCR) and SSPE-CT-2004-513 645 (LAB-ON-SITE).<br />

References<br />

1. Hakhverdyan M, Rasmussen TB, Thorén P, Uttenthal Å, Belák S (2006). Development of a real-time<br />

PCR assay based on Primer-Probe Energy Transfer for the detection of swine vesicular disease virus.<br />

Arch Virol 151: 2365-2376.<br />

2. Rasmussen TB, Uttenthal A, de Stricker K, Belak S, Storgaard T (2003). Development of a novel<br />

quantitative real-time RT-PCR assay for the simultaneous detection of all serotypes of foot-and-mouth<br />

disease virus. Arch Virol 148: 2005-2021.<br />

3. Rasmussen TB, Uttenthal A, Fernandez J, Storgaard T (2005). Quantitative multiplex assay for<br />

simultaneous detection and identification of Indiana and New Jersey serotypes of vesicular stomatitis<br />

virus. J Clin Microbiol 43: 356-362.<br />

4. Rasmussen TB, Uttenthal A, Agüero M (2006). Detection of three porcine vesicular viruses using<br />

multiplex real-time primer-probe energy transfer. J Virol Methods 134, 176-182.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT AND VALIDATION OF A REAL TIME RT-PCR ASSAY FOR THE SIMULTANEOUS<br />

DETECTION OF AVIAN INFLUENZA VIRUSES BELONGING TO THE H5, H7 AND H9 SUBTYPES<br />

Monne I., Salviato A., Ormelli S., De Battisti C., Salomoni A., Drago A., Zecchin B., Bertoli E.,<br />

Capua I, Cattoli G.<br />

Istituto Zooprofilattico Sperimentale delle Venezie, Research & Development Department<br />

OIE/FAO and National Reference Laboratory for Avian Influenza and Newcastle Disease<br />

OIE Collaborating Centre for Epidemiology, Training and Control of Emerging Avian Diseases<br />

Viale dell’Università 10, Legnaro, Padova, Italy<br />

Introduction<br />

Influenza A viruses are responsible for avian influenza, a disease of great importance for animal health and<br />

sometimes also for human health. Several subtypes of AI have been identified basing on antigenic<br />

differences between the two surface glycoproteins HA and NA (1). Among the different HA subtypes, the H5,<br />

H7 and H9 strains are of great interest for the scientific community because of serious consequences for the<br />

poultry industry and the increasing frequency of direct transmission of these viruses to humans. In certain<br />

areas of the world, these subtypes are co-circulating in domestic birds and in order to manage this situation<br />

appropriately an adequate and rapid diagnostic tool is necessary. Here we report the development and<br />

validation of a one step reverse transcription real time PCR (RRT-PCR) assay to detect simultaneously the<br />

H5, H7 and H9 subtypes of AIVs.<br />

Material and Methods<br />

A one step real time PCR was developed using fluorogenic hydrolysis-probes to detect the H5, H7 and H9<br />

subtypes of AI from clinical samples. Multiple alignments of previous and recent H5, H7 and H9 HA<br />

sequences were performed to design primers and probes minimizing mismatches.<br />

The specificity of the primer-probe sets was examined with RNA and DNA extracted from a diverse array of<br />

viral and bacterial avian pathogens. The sensitivity were tested using in vitro-transcribed RNA and ten-fold<br />

serial dilutions of titrated avian influenza viruses. Intra and inter-assay variability of the method were<br />

assessed. The test was also validated on clinical samples of avian origin.<br />

Results<br />

The protocol allows a rapid (less than 2 hrs) determination of the three AI subtypes. The H5, H7 and H9<br />

primer and probe sets were able to detect the nucleic acids only of isolates of their respective subtypes. High<br />

sensitivity levels were obtained, with limits of detection ranging from 10 2 to 10 4 RNA copies. Excellent results<br />

were achieved in the intra- and inter-assay variability tests. The coefficient of variation intra-assay and<br />

interassay resulted < to 2,64% and < to 7,22% respectively. Results derived from the test validation on<br />

clinical samples were in agreement with the gold standard applied (virus isolation).<br />

Discussion and Conclusion<br />

Conventional avian influenza diagnostic tools are time consuming and require facilities (e.g. BSL3) not easily<br />

available in many affected areas. The molecular tests for diagnosis of AI are commonly applied for type A<br />

influenza viruses. However, the only Real Time PCR detection assay published for H7 subtype has been<br />

validated only on strains belonging to the AI American lineage, and probes are currently not available for the<br />

identification of H9 subtypes.<br />

The high sensitivity, specificity and reproducibility of the H5, H7 and H9 RRT-PCR assay together with its<br />

rapidity indicate that this method is suitable as a routine laboratory test for rapid detection and differentiation<br />

of three prevalent avian influenza virus subtypes in samples of avian origin.<br />

To date, this is the first real time PCR protocol available for the simultaneous detection of AI viruses<br />

belonging to H5, H7 and H9 subtypes.<br />

References<br />

1. Fouchier RA, Munster V, Wallensten A, et al. (2005). Characterization of a novel influenza A virus<br />

hemaglutinin subtype (H16) obtained from blackheaded gulls. J Virol; 79: 2814-2822.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AN EVALUATION OF COMMERCIALLY AVAILABLE NUCLEIC ACID EXTRACTION METHODS FOR<br />

THE PROCESSING AND SUBSEQUENT qPCR ANALYSIS OF SAMPLES INFECTED WITH AVIAN<br />

INFLUENZA VIRUS.<br />

John R. White 1* , Glen R. Hewitson 2 , Tyrone S. McDonald 3 , Eric Hansson 1 , Hans-G Heine 1 , Adam J. Foord 1 and Ibrahim S. Diallo 2 .<br />

Introduction<br />

1 Australian Animal Health Laboratory, CSIRO, Division of Livestock Industries, P.O. Bag 24, Geelong, VIC.<br />

2 Animal Research Institute, Queensland Dept. Primary Industry and Fisheries, Locked Bag 4, Moorooka,<br />

QLD.<br />

3 School of Life and Enviromental Sciences , Faculty of Science and Technology, Deakin University, Pigdon’s<br />

Rd., Waurn Ponds, Geelong, VIC.<br />

The Australian Animal Health Laboratory (AAHL) is responsible for the ongoing monitoring of the exotic<br />

disease-free status of endemic animal populations via regular submission of field samples and in cooperation<br />

with relevant State Departments, specifically targeted field surveillance programs. At AAHL, the<br />

quantitative (real-time) polymerase chain reaction (qPCR) is usually one of the first methods of choice for<br />

rapid infectious agent identification. To accommodate the large daily sample numbers expected during an<br />

outbreak and an ongoing surveillance phase, we need to have proven sample processing protocols in place<br />

that will minimise ‘bottlenecks’ prior to final qPCR analysis. In collaboration with the Animal Research<br />

Institute (Queensland Department of Primary Industry and Fisheries) we have therefore undertaken a study<br />

of a number of commercially available methods for the extraction of viral RNA from field and laboratoryderived<br />

samples known, or suspected to be, infected with strains of avian influenza virus. The efficiency and<br />

sensitivity of each method was assessed by the ultimate detection of viral genomic material in an appropriate<br />

qPCR reaction 1 . In addition to overall extraction efficiency, each method was also assessed for ease of use,<br />

extent of operator exposure to material, suitability for adaptation to a high throughput requirement and<br />

applicability for adoption in regional laboratory settings.<br />

Material & methods<br />

Original animal swab material was suspended in collection medium (0.01M Phosphate-buffered saline, 1%<br />

foetal calf serum, 0.1 % glucose and 0.1 % (v/v) phenol red and/or inoculated into embryonated eggs.<br />

Between 10 -100 ul of subsequently inactivated allantoic fluid or swab suspension was then used for<br />

influenza virus RNA extraction. A small number of various tissue types from experimentally infected ducks<br />

were also used for extraction. The column matrix based RNeasy Kit (Qiagen) and the magnetic bead based<br />

MagMax Kit (Ambion - Applied Biosystems) were used to extract viral RNA according to the manufacturer’s<br />

instructions. The thermostable protease based nucleic acid extraction system from Zygem (New Zealand)<br />

(PrepGEM) was used basically according to manufacturer’s instructions but with minor modifications.<br />

Extracted RNA derived from each kit was then used as the template in a previously described Taqman©<br />

assay 1 for the detection of influenza virus type A matrix protein-specific RNA. This assay was analysed on an<br />

Applied Biosystems 7500 System using a threshold setting of 0.05 units.<br />

Results<br />

All three nucleic acid extraction methods produced acceptable and generally similar Ct values (within 1-2 Ct<br />

units) for specific samples. However, after considering all the data obtained, the MagMax Kit appeared<br />

potentially the most sensitive method and the PrepGEM Kit was marginally the least sensitive.<br />

Discussions & conclusions<br />

All three extraction kits are reliable for the isolation of influenza virus RNA. The MagMax and PrepGEM Kits<br />

are the most appropriate for application to automated high throughput systems. The PrepGEM Kit is the<br />

simplest and most rapid procedure and involves least exposure of reaction material during the extraction<br />

process. There is potential for the sensitivity of the PrepGEM Kit to be further optimised.<br />

References<br />

1 Heine HG, Trinidad L, Selleck PW, Lowther S. (2007). Avian Diseases. 51: 370-372.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

HIGH THROUGHPUT ISOLATION AND DETECTION OF NORTH AMERICAN AND EUROPEAN<br />

PORCINE REPRODUCTIVE AND RESPIRATORY VIRUS BY QRT-PCR<br />

A. M. Burrell 1* , W. Xu 1 , Q. Hoang 1 , R. C. Willis 1 , R. Shah, M. Bounpheng 1 , and X. Fang 1<br />

PRRSV, an RNA virus, order Nidovirales of the Arteriviridae family, is one of the most costly infectious<br />

diseases for swine producers. Two main antigenic subtypes of PRRSV exist: North American strain (ATCC<br />

VR-2332) and European strain (Lelystad strain). Infected animals present with reproductive difficulties, and<br />

respiratory disease often associated with secondary infections.<br />

We have developed an integrated workflow consisting of high throughput nucleic acid purification, and qRT-<br />

PCR for concurrent detection of North American and European PRRSV. Using an Ambion ® MagMAX TM high<br />

throughput magnetic bead-based viral RNA isolation method, RNA can be isolated from diverse sample<br />

matrices. Purified nucleic acid is analyzed with the Ambion AgPath-ID NA/EU PRRSV Reagent Kit, which<br />

can detect North American and European strains of PRRSV, allowing subtyping. As few as 50 copies of<br />

PRRSV RNA are consistently detected using this assay. An internal control RNA, XenoRNA, is provided to<br />

monitor nucleic acid purification efficiency, and to detect PCR inhibitors. It ensures that false negative results<br />

are minimized, if not eliminated.<br />

Assay performance was evaluated using >100 field samples of known PRRSV status. RNA was isolated<br />

from swine serum, tonsil, and lung tissues and the purified RNA underwent qRT-PCR using the AgPath-ID<br />

qRT-PCR Kit. Results showed 98% concordance with secondary laboratory PCR results. Furthermore,<br />

subtyping of North American and European strains were 100% concordant demonstrating that this<br />

method provides an economical and rapid solution for PRRSV detection and identification.<br />

For research use only, not for use in diagnostic procedures. Not licensed by the USDA.<br />

1 Applied Biosystems, Austin, TX<br />

* Angela M. Burrell, Applied Biosystems,<br />

2130 Woodward Street, Austin, Texas 78744<br />

Ph: (512) 651-0200, Fax: (512) 651-0201, E-mail: angela.burrell@appliedbiosystems.com<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CSFV DIVA ASSAY: POTENTIAL APPLICATION OF A ROBUST MULTIPLEX REAL-TIME RT-PCR IN<br />

WILD BOAR VACCINATION AND CONTROL<br />

L. Liu a , B. Hoffmann b , F. Widén a , C. Baule a , M. Beer b , S. Belák a<br />

a Joint R&D Division in Virology, The National Veterinary Institute & The Swedish University of Agricultural Sciences, SE-751 89<br />

Uppsala, Sweden. b Friedrich-Loeffler-Institute Federal Research Institute for Animal Health, Boddenblick 5a, D-17493 Greifswald - Insel<br />

Riems, Germany<br />

Introduction:<br />

CSF is a highly contagious and devastating viral disease affecting both domestic pigs and wild boars. In<br />

Europe, the virus is largely maintained in wild boar reservoir and is reintroduced to domestic pigs by contacts<br />

between domestic and free-living species. About 80% of the first outbreaks of CSF have occurred in regions<br />

where CSF in wild boar is endemic. Within the EU project “CSFV Vaccine & Wild Boar”, a chimeric marker<br />

vaccine CP7_E2alf has been developed and evaluated for application in wild boar 1 . The aims of this study is<br />

development of a multiplex real-time RT-PCR assay for detection and differentiation of wild type CSFV from<br />

the marker vaccine CP7_E2alf, and to highlight its potential application in CSFV vaccination and control in<br />

wild boar.<br />

Methods:<br />

Multiplex DIVA assay includes two sets of primers and probes, one for the marker vaccine CP7_E2alf (CP7<br />

assay) with TexasRed as reporter dye and another for wild type CSFV (CSF assay) with 6’-FAM as reporter<br />

dye. The reaction was carried out with QuantiTect Probe RT-PCR kit.<br />

Results:<br />

The performance of DIVA assay was highly specific, with either CP7- or CSFV-specific amplification out of a<br />

total of 35 related pestiviruses (including vaccine virus). The DIVA assay had same sensitivity as single<br />

assays, with excellent reaction efficiencies (close to 100%) and good linearity.<br />

Conclusions:<br />

The robust multiplex DIVA assay was developed with excellent performance. With this high throughput<br />

diagnostic tool to differentiate infected from vaccinated animals, its potential application will be effect control<br />

of CSF in wild boar population in European countries.<br />

References:<br />

1. Koenig P, Lange E, Reimann I, Beer M. CP7_E2alf: a safe and efficient marker vaccine strain for oral<br />

immunisation of wild boar against Classical swine fever virus (CSFV). Vaccine. 2007 Apr 30;25(17):3391-9.<br />

2: Hoffmann B, Beer M, Schelp C, Schirrmeier H, Depner K. Validation of a real-time RT-PCR assay for<br />

sensitive and specific detection of classical swine fever. J Virol Methods. 2005 Dec;130(1-2):36-44.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A REAL-TIME RT-PCR (RRT-PCR) WITH INTERNAL CONTROL TO DETECT PERSISTENT BVD VIRUS<br />

(BVDV) INFECTION IN POOLED EAR NOTCH SAMPLES<br />

A. G. Wise, P. Wu, J. Harmala, C. Benson, J. Heissler, L. Stolle and R. K. Maes x<br />

Persistently infected (PI) cattle are the main source of spread of BVDV. In the US, ear notches are the most<br />

common diagnostic sample. Assay formats include IHC, antigen capture ELISA (ACE), and RT-PCR on<br />

pooled PBS extracts. Our aim was to develop and validate a Taqman-based rRT-PCR with internal control to<br />

detect PI animals.<br />

Primers and probe were designed from conserved sequences within the 5’ UTR. The detection limit was 0.8<br />

TCID50. Dynamic range was determined with a panel provided by Dr. Ridpath. Initial testing of RNA from<br />

individual PBS extracts of 42 known positive samples yielded only 38 positives. In contrast, all RNA samples<br />

from tissue pools made by mixing each of the same 42 positive samples with 9 known negative samples,<br />

tested positive by rRT-PCR. Subsequently, 2,570 ear notch samples were analyzed by rRT-PCR (257 pools)<br />

and individually by ACE. Twenty-one pools were positive and were subsequently found to contain at least<br />

one positive sample by ACE. All individual samples in the 236 negative pools tested negative by ACE.<br />

Our current preference is for testing of pooled ear notch tissue pieces rather than pooled PBS extracts. An<br />

internal control in the rRT-PCR assay is essential to detect assay inhibition, resulting in false negatives.<br />

DCPAH- MSU, 4125 Beaumont Road, Lansing, MI 48910, USA.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

RAPID DETECTION OF CLASSICAL SWINE FEVER VIRUS IN PORK PRODUCTS BY A REAL-TIME<br />

RT-PCR ASSAY<br />

Songhua Shan 1 *,Yi Xia 2 , Zhongrong Huang 1 , Yi Zhou 2 , Chungyang Li 1<br />

1 Shanghai Entry-Exit Inspection and Quarantine Bureau, Shanghai, China; * Murdoch University, Australia; 2 Shanghai Fosun Med-Tech<br />

Development Co., Ltd, Shanghai,China<br />

Introduction<br />

Although some countries have successfully eradicated classical swine fever virus (CSFV), outbreaks still<br />

continue to occur throughout the world. CSFV-contaminated swine products have become the main risk<br />

factor for the introduction of CSFV into CSF-free countries (1, 2). Real-time PCR has been increasingly used<br />

for the diagnosis, quantitative and qualitative measurement of a broad range of animal pathogens, which has<br />

attractive advantages over RT-PCR (3, 4, 5). However, there are limited reports on CSFV detection in<br />

musculature or meat products by RT-PCR, although there are some promising reports about detection of<br />

other viruses in meat (2). The aim of the present study was to develop a CSFV-specific real-time RT-PCR<br />

and evaluate its suitability for the detection of small amounts of CSFV in swine products.<br />

Material & methods<br />

CSFV Thiveral strain (T strain), bovine diarrhoea virus (BVDV) Oregon C24V strain and PK-15 cells were<br />

provided by National Control Institute of Veterinary Bioproducts and Pharmaceuticals, China. CSFV lapinized<br />

Chinese virus (C strain), BVDV Singler, Drapper, NADL and NY-1 strains were donated by Nanjing<br />

Agricultural University. Other reference isolates including PPV, PRCV, TGEV, PRV, and PRRSV were from<br />

the Shanghai Entry-Exit Inspection and Quarantine Bureau, China. Primers and FAM-labeled Taqmanprobes<br />

specific for CSFV were designed using Primer Express 1.0 software based on the consensus<br />

sequence of the 5’ non-translated region of available CSFV nucleotide sequences. Parameters (the ratio of<br />

primer/probe, reverse transcriptase/Taq polymerase, the concentration of Mg 2+ , dNTPs, annealing<br />

temperature and PCR cycle) of real-time RT-PCR were optimized using CSFV T strain. In total, 211 samples<br />

from clinically suspected cases, imported pigs and porcine products were simultaneously tested by real-time<br />

RT-PCR and CSFV-specific antigen-capture ELISA (IDEXX kit).<br />

Results<br />

The one-step real-time RT-PCR showed 100% specificity against CSFV, other pestiviruses, porcine viruses<br />

and cell cultures. The PCR product of T strain was sequenced, and showed the expected sequence. The<br />

detection limit of the real-time RT-PCR is 1 TCID50/mL. Serial 10-fold dilutions of virus stock tested by realtime<br />

RT-PCR on LightCycler, SLAN, PE5700, iCycler, PE7000 and PE7700 present similar results. A<br />

comparison of three RNA extraction kits from Roche, Invitrogen and Shanghai Fosun Med-Tech<br />

Development Co., Ltd. showed similar results. 75 visceral organs from CSFV suspect infected pigs and 132<br />

imported swine products were tested by real-time RT-PCR and ELISA. The results demonstrated that realtime<br />

RT-PCR is more sensitive than ELISA, showing significant difference (P


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DETECTION AND DIFFERENTIATION OF AVIAN INFLUENZA AND NEWCASTLE DISEASE VIRUS<br />

DIRECTLY FROM POULTRY SPECIMENS BY MULTIPLEX RT-PCR<br />

Songhua Shan A* , Weirong Jing B , Min Wang B , Caozhe Xu A , Hongyou Qin B , Qi Tao A and Jinping Liu A<br />

A Shanghai Entry-Exit Inspection and Quarantine Bureau, Shanghai, China;<br />

*Murdoch University, Australia; B Shanghai Huaguan Biochip Company, Shanghai, China<br />

Introduction<br />

Avian Influenza (AI) and Newcastle Disease (ND) are the most devastating infectious diseases in poultry<br />

industry. Confirmatory diagnosis of AI and ND depends on the isolation of the virus (1). RT-PCR has<br />

currently been applied in rapid detection and identification of AI and ND (1). Because AIV and NDV share<br />

many similar characteristics, it was considered appropriate to develop an multiplex RT-PCR (mRT-PCR) for<br />

both AI and ND. However, reports on mRT-PCR for detection of AIV and NDV are limited (2, 3). In this study,<br />

an mRT-PCR capable of simultaneously detection of AIV and NDV is described, focusing on the detection of<br />

AIV and NDV from tissue and organ samples, swabs and poultry products.<br />

Material & methods<br />

NDV (F48E9, Mukteswar, N79 isolates), A/chicken/HK/1000/97(H5N1), A/Chicken/HK/216.3/01 (H6N?),<br />

A/chicken/HK/230.2/01 (H9N?), A/Shanghai/29/78(H1N1), A/Shanghai/49/78(H3N2), Duck hepatitis virus<br />

(DHV) and Duck plague virus (DPV) used were from Shanghai Entry-Exit Inspection and Quarantine Bureau,<br />

China. Eighty isolates with haemagglutinating activity were collected and identified as AIV or NDV by HI.<br />

Other avian pathogens including IBV, ARV, IBDV, MDV, EDSV, chicken coccidiosis and Infectious Coryza<br />

were from different commercial vaccines.<br />

The primers were designed from the nucleocapsid protein (NP) gene of AIV and the fusion protein (F) gene<br />

of NDV to amplify products of 502bp and 421bp, respectively. Parameters (concentration of primer, Mg2+,<br />

dNTPs and Taq DNA Polymerase, PCR cycle, annealing temperature) of the single-virus RT-PCR ( sRT-<br />

PCR) were optimized by checkerboard titration. mRT-PCR procedures were determined by balancing<br />

different parameters based on the result of sRT-PCR.<br />

Results<br />

The ratio of 1 to 1.25 of NDV vesus AIV primers and the annealing temperature (55�) suitable for bigger<br />

fragment amplification(AIV) was chosen for the mRT-PCR. In addition to testing different avian pathogens,<br />

the PCR products of nine AIV and ten NDV isolates were sequenced to further confirm the specificity of<br />

mRT-PCR. The detection limit of AIV and NDV by mRT-PCR were respectively 10 3.5 EID50 and 10 4.75 EID50,<br />

being the same sensitivity as sRT-PCR for AI and 100 fold decrease for ND compared to sRT-PCR. Thirtynine<br />

AIV consisting of H1, H5, H9 subtype and 41 NDV of different origins (chicken, duck, goose, ostrich,<br />

pigeon and peacock) were compared by multiplex RT-PCR and typing by HI, showing 100%(80/80)<br />

agreement. Tissue and visceral organ samples from the chickens experimentally infected with AIV or NDV<br />

were tested by mRT-PCR and virus isolation(VI), showing 93.8%(30/32 ) agreement for AIV and 95.2%<br />

(20/21 ) for NDV. Additionally, 35 samples from imported poultry products and 32 cloacal swabs from<br />

Chinese domestic markets were tested by mRT-PCR and VI , and showed 100% (67/67) agreement for AIV<br />

and 97.1%(65/67) agreement for NDV. These data showed that VI is more sensitive than mRT-PCR.<br />

Discussions & conclusions<br />

Due to the primer design based on the conserved sequence of AIV NP gene and NDV F gene, different<br />

subtype AIVs and NDVs tested could all be detected. mRT-PCR was able to simultaneously detect and<br />

differentiate AIV and NDV in one day and showed the good correlation (97.1%) with VI. Although real-time<br />

PCR(4) and NASBA(5) have recently been developed to reduce the time and increase specificity and<br />

sensitivity, these instruments are very expensive. Thus, the mRT-PCR described here provides a rapid, costeffective<br />

way for simultaneous detection of AI and ND.<br />

A single-tube mRT-PCR for simultaneous detection of APV, AIV, and NDV was reported to show similar<br />

detection limits to sRT-PCR(2). In our study, although mRT-PCR for AI has the same sensitivity as sRT-<br />

PCR, the detection limit decrease for NDV was 100 fold less than that by sRT-PCR. The reasons may be<br />

formed primer-dimers, competition for dNTP, Taq DNA polymerase in a PCR tube.<br />

References<br />

1. Office International des Epizooties: Highly pathogenic avian influenza, Newcastle Disease. In: Manual of standard for diagnostic tests<br />

and vaccines, 4th ed. Office International des Epizooties, Paris, France. http://www.oie.int.20005.<br />

2. Malik Y.S., D.P. Patnayak, and S.M. Goyal. Detection of three avain respiratory viruses by single-tube multiplex reverse<br />

transcription-polymerase chain reaction assay. J Vet Diagn Invest. 16(3): 244-248. 2004.<br />

3. Farkas T, M. Antal, L.Sami, P. German, S. Kecskemeti, G, Kardos, S. Belak, I. Kiss. Rapid and simultaneous detection of avian<br />

influenza and newcastle disease viruses by duplex polymerase chain reaction assay. Zoonoses Public Health. 54(1):38-43. 2007.<br />

4. Spackman E., D.A. Senne, L.L. Bulaga, S. Trock, and D.L. Suarez. Development of multiplex real-time RT-PCR as a diagnostic tool<br />

for avian influenza. Avian Dis. 47(3 Suppl):1087-1090. 2003.<br />

5. Shan S.H., L.S. Ko, R.A. Collins, Z.L.Wu , J.H. Chen, K.Y. Chan, J. Xing, L.T. Lau, and A.C.H.Yu. Comparison of different methods<br />

for avian influenza subtype H5N1 detection. Biochem.Biophy. Res. Commun. 302: 377�383. 2003.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SAMPLE PREPARATION METHODS FOR HIGH QUALITY NUCLEIC ACID ISOLATION FROM A<br />

VARIETY OF VETERINARY SAMPLES<br />

S. Ullmann 1 , D. Herold 1 , R. Hodgson 2 , R. Soeller, 3 S. Cramer 3<br />

1 QIAGEN GmbH, Hilden, Germany; 2 QIAGEN Pty Ltd, Doncaster, Vic, Australia;<br />

3 QIAGEN Hamburg GmbH, Hamburg, Germany<br />

Introduction<br />

Effective and reproducible isolation and purification of nucleic acids is one of the key success factors for<br />

amplification and detection of pathogenic nucleic acids using molecular methods. Diagnostic tests using<br />

PCR (e.g., for Johne’s disease in cattle) are replacing traditional ELISA or culture based methods due to<br />

higher sensitivity and speed.<br />

An easy and standardized method for nucleic acid isolation that gives high-quality nucleic acids is required.<br />

The major challenge when working with such diverse material is to develop optimized pretreatments for all<br />

sample types. Dependent on sample type, content of inhibitors, and content of nucleic acids, the samples<br />

demand different pretreatment conditions such as mechanical disruption, enzymatic digestion, and<br />

incubation times.<br />

Material & methods<br />

Sample sources include various animal organ tissues, fresh and dried pig ears, horse hair, ruminant feces<br />

and ticks. Nucleic acid purification used either manual or automated sample preparation protocols. Manual<br />

preparation comprises different methods depending on the starting material (e.g., DNeasy ® Blood & Tissue<br />

Kit, QIAamp ® DNA Stool Mini Kit). For automated sample preparation, the low-throughput QIAGEN ®<br />

BioSprint 15 and BioRobot ® EZ1 systems were used.<br />

Results<br />

DNA isolation from a variety of different samples and animals was successful, and a range of typical DNA<br />

yield depending on the starting material will be presented. Quality of the isolated DNA is shown by gel<br />

electrophoresis, enzymatic digestion, and standard as well as real-time PCR as downstream applications.<br />

All types of pig ear samples, including frozen, lyophilized, and dried tissue, gave good yields and were<br />

successfully used in PCR analysis. Real-time PCR data for detection of M. paratuberculosis DNA isolated<br />

from ruminant feces have been performed with no observed PCR inhibition. Detection of Borrelia DNA with<br />

the artus ® Borrelia LC PCR Kit subsequent to a DNA isolation from ticks has shown a high sensitivity: the<br />

lowest positive curve in the real-time PCR corresponded to 5 copies.<br />

DNeasy Blood & Tissue Kit, BioSprint 15, BioRobot EZ1, artus Borrelia LC PCR Kit: For Research Use Only.<br />

Not for use in diagnostics procedures. No claim or representation is intended to provide information for the<br />

diagnosis, prevention, or treatment of a disease. The QIAamp DNA Stool Mini Kit is intended for general<br />

laboratory use. No claim or representation is intended to provide information for the diagnosis, prevention, or<br />

treatment of a disease.<br />

Discussions & Conclusions<br />

Purification, downstream analysis and archiving of high quality nucleic acid from various animal samples are<br />

now a standard requirement for many diagnostic tests in Veterinary pathology. Various sample types,<br />

matched with an appropriate extraction protocol, can be processed using manual or automated procedures<br />

to fit workflow throughput needs. For optimum performance and comparison of test results the key<br />

parameters to maximise in purified nucleic acid preparations are; quality, integrity, yield, reproducibility and<br />

standardization.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EVALUATION OF REAL-TIME PCR BASED ASSAYS FOR THE DETECTION AND QUANTITATION OF<br />

DIFFERENT DISEASES IN ANIMALS<br />

Introduction<br />

T. Laue 1 , H. Marquardt 1 , S. Gauch 2 , S. Schlemm 2 , R. Hodgson, 3* P. Wakeley 4 and S. Ullmann 2<br />

1 QIAGEN Hamburg GmbH, Hamburg, Germany; 2 QIAGEN GmbH, Hilden, Germany;<br />

3 QIAGEN Pty Ltd, Doncaster, Vic, Australia; 4 Veterinary Laboratories Agency, Weybridge, United Kingdom<br />

In 2006 QIAGEN acquired a license to commercialize a portfolio of selected PCR-based, veterinary<br />

molecular test (assays) developed by the Veterinary Laboratories Agency (UK). The initial portfolio consists<br />

of different PCR-based assays for infectious veterinary diseases affecting wildlife and farmed livestock.<br />

These assays were designed at the VLA and will be available from QIAGEN as a “complete solution”<br />

including recommendations for nucleic acid preparation plus optimized kits and protocols for multiplex and<br />

real-time PCR.<br />

Material & methods<br />

The assays will be validated by the VLA using manual and/or automated technologies for DNA and RNA<br />

extraction from different samples.<br />

Results<br />

This poster presents data for analytical sensitivity and specificity and diagnostic specificity and sensitivity.<br />

Discussions & Conclusions<br />

This presentation describes the cooperation between the two partners and shows the procedure of<br />

development and validation of the veterinary assays using the example of the specific assay for detection of<br />

Bovine Viral Diarrhoea Virus, an infectious disease causing infertility, abortion, and congenital defects in<br />

calves.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

Introduction:<br />

ADVANCES IN FLUORESCENCE POLARIZATION FOR DIAGNOSIS<br />

OF BOVINE BRUCELLOSIS IN MILK<br />

Samartino L., Conde, S., Schust, M., Piazza, E., Salustio, E.<br />

Instituto de Patobiología, CICVyA INTA<br />

Las Cabañas y Los Reseros, 1712, Villa Udaondo, Castelar, Buenos Aires, Argentina.<br />

luersa00yahoo.com.ar<br />

Bovine brucellosis is still an important health problem in Argentina and other countries of Latin America. The<br />

milk ring test is the mandatory screening test used for surveillance 4 times a year in most of these countries.<br />

Argentina recently also adopted the indirect enzyme-immunosorbent assay (IELISA) which is more precise<br />

but far more expensive. Fluorescence polarization immunoassays (FPA) were introduced in the 90s for<br />

serological diagnosis of brucellosis in cattle. This test is based on measuring the polarization of light caused<br />

by changes in molecular size as a result of antigen-antibody reactions. The test is simple to perform, gives<br />

rapid results, and reduces the errors that can occur when reading the conventional agglutination tests. In<br />

addition, FPA is adaptable to field implementation. We understand that this test could be also applied for milk<br />

studies.<br />

Objective:<br />

To evaluate the Fluorescence Polarization Assay (FPA) for diagnosis of bovine brucellosis in milk.<br />

Methods: Test samples: Milk from 252 pools ( Holstein cows) was obtained from different farms from<br />

samples taken for routine surveillance testing.<br />

Tests performed:<br />

The milk ring test (MRT), the routine test for surveillance in dairy cattle in Argentina, was performed<br />

according to the current international proceedings. The IELISA was also used. Cut off for the IELISA has<br />

been established previously as 23% positive (pool). FPA was done following the instruction of the<br />

commercial kit (Prionics) and adapted for milk. FPA was applied to stored whole milk and to milk sera using<br />

a portable Diachemix Sentry 100 polarization analyzer instrument (single tube reader).<br />

Results:<br />

The following results were observed: 71.8% were positive for MRT, 82.1% were positive for IELISA and<br />

15.1% and 71.0% of whole milk and milk sera, respectively, were positive for FPA.<br />

Conclusions:<br />

FPA results, using milk sera, agreed with MRT results, while the results obtained when whole milk was used<br />

were very poor. IELISA seems to have more sensitivity that the other tests. However, based in this<br />

preliminary study, we conclude that FPA gave results comparable to MRT and is a promising technique for<br />

the diagnosis of bovine brucellosis in milk. Current studies are targeted toward improving the sensibility of<br />

the test.<br />

References<br />

Samartino, L., Gregoret, R., Gall, D., Nielsen, K. Fluorescence polarization assay: Application to the diagnosis of<br />

bovina brucellosis in Argentina. J. Of Immunassay, 20 (3), 115-126. 1999.<br />

Nielsen, K., Smith, D., Gall, D., Perez, B., Samartino, L., Nicoletti, P., Dajer, A., Rojas, X., Nelly, W. Validation of<br />

the fluorescence polarization assay for detection of milk antibody to Brucella abortus. J. Immunoassay 22: 203-<br />

211. 2001.<br />

Gall, D, Nielsen, K., Bermudez, R., Moreno, F., Smith, P. Fluorescence Polarization Assay for Detection of<br />

Brucella abortus. Clin. Diag. Lab. Immuno. 1356-1360. 2002.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EFFECTS OF DIFFERENT POPULATIONS ON AN ASSESSMENT OF THE<br />

SENSITIVITY AND SPECIFICITY OF AN ANTIGEN ELISA FOR BVDV.<br />

J Zhang* 1 , L Brown 1 , A Elliott 1 , S Fell 1 and PD Kirkland 1<br />

1 Virology Laboratory, Elizabeth Macarthur Agricultural Institute, NSW DPI, Menangle, NSW Australia<br />

Introduction<br />

Enzyme linked immunosorbent assays (ELISAs) to detect antigens of bovine viral diarrhoea virus (BVDV),<br />

the so called pestivirus antigen capture ELISA (PACE), have made a major contribution to improved<br />

diagnosis of BVDV infection over the last decade. Assays are typically used to detect BVDV antigens in<br />

lymphoid or epithelial cells in blood and tissues such as spleen, lymph nodes and lung. In recent years, a<br />

modified assay with high sensitivity has been used to test skin samples and is in widespread use in many<br />

countries but has not been fully validated for use in Australia, where there is a different population of viruses<br />

to those found in the USA and Europe. During the evaluation of a new assay, it is important to have a good<br />

understanding of the epidemiology and pathogenesis of the disease under investigation, as well as to have<br />

samples from animals of known disease status. The status of the test samples can be established by<br />

selecting animals from herds known to be free of the test agent (for the determination of assay specificity)<br />

and to have animals or samples that have tested positive by a “gold standard” (assessment of assay<br />

sensitivity). This study involves the evaluation of a commercial ELISA kit that is used for the detection of<br />

BVDV antigens in skin samples in 2 different animal populations. The results illustrate the importance of<br />

knowledge of the disease and performance of the assays being used to ensure the correct assessment of<br />

assay sensitivity and specificity.<br />

Material & methods<br />

Whole, heparin treated blood and skin biopsy samples were collected from more than 1200 cattle. Plasma<br />

and white blood cell (‘buffy coat’) fractions were separated for testing. Skin samples were transported to the<br />

laboratory in a liquid sample buffer. The buffer from the skin samples and detergent extracts of buffy coat<br />

samples were tested in separate antigen ELISAs designed to detect BVDV antigens in white cell<br />

preparations or skin extracts. Plasma samples were used for virus isolation in cell culture, PCR and serology<br />

by VNT and AGID. Any animal that gave a positive result in an antigen ELISA was resampled after 4-8<br />

weeks. Virus isolation was conducted by inoculating pre-confluent monolayers of secondary bovine testis<br />

cells while the real time PCR was based on a previously published method 1 .<br />

Results<br />

Compared to virus isolation, both ELISAs had a sensitivity of >99%. However, depending on the cattle<br />

population, the diagnostic specificity of the ELISA varied from 81% to >99% for skin samples. In a large dairy<br />

herd, where the youngest BVDV infected animal was 2 years old, there was complete agreement between<br />

virus isolation and both of the antigen ELISAs. Each of the BVDV infected animals was seronegative,<br />

remained positive in both tests when resampled and did not seroconvert. In this population, with only 9<br />

persistently infected (PI) animals, the relative sensitivity and specificity of the PACE for testing of skin<br />

samples were both 100%. However, in a large population of approximately 1500 beef cattle, 247 calves were<br />

tested and 172 reactors were detected when extracts of skin samples were tested. These positive samples<br />

were all collected from calves that were approximately 5-6 months old. In contrast only 139 of these samples<br />

were positive by virus isolation and by the PACE with buffy coat samples. Of the 172 animals that gave<br />

positive or inconclusive reactions with skin samples, 32 were seropositive at the time of first sampling. When<br />

these animals were re-sampled, only 139 gave positive ELISA results and on both skin and buffy coat<br />

samples. All of the animals that had previously reacted in the ELISA with skin samples were now<br />

seropositive. In this herd, if the assay performance characteristics were calculated from the results of the first<br />

test, the diagnostic specificity relative to virus isolation was only 81%. However, the specificity for the test on<br />

the later extracts of skin samples was 100%. Throughout the study, the sensitivity and specificity of the<br />

ELISA remained high (approaching 100%) at all times when buffy coat extracts were tested.<br />

Discussion & conclusions<br />

When testing for the detection of BVDV infected animals, the main purpose is to detect PI animals as these<br />

are the reservoirs of the virus in a population. While testing of skin samples has the advantage of high<br />

analytical sensitivity, care must be exercised to ensure that reactors are not acute, transiently infected<br />

animals. To overcome this problem and avoid unnecessary culling, all reactors must be retested. The results<br />

obtained in these herds amply demonstrate the need to have a thorough understanding of the disease and to<br />

be able to apply appropriate supplementary tests when determining assay performance characteristics.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

COMPARISON OF SEROLOGIC TECHNIQUES: MAT AND ELISA, WITH BACTERIOLOGIC METHODS:<br />

DFM EXAMINATION, IFAT, MICROBIOLOGIC CULTURAL ISOLATION AND PCR FOR DIAGNOSIS OF<br />

BOVINE LEPTOSPIROSIS IN IRAN<br />

Ehsanollah Sakhaee* Gholamreza Abdollahpour, Mahmoud Blourchi, Abdolmohammade Hasani Tabatabayi, Mohammad reza Mokhber<br />

Dezfouli, Rahim Haji Hajikolayi, Afshin Raoofi, Saeed Sattari<br />

Leptospirosis is a worldwide zoonosis caused by Leptospira interrogans. This study was conducted to<br />

evaluate serologic and bacteriologic findings of leptospirosis in clinically suspected cows. Three hundred and<br />

eighty sera and thirty three urine samples were collected from 6 industrial dairy farms in Tehran suburb, from<br />

December (2004) to June (2005). The prevalence of disease was determined by microscopic agglutination<br />

test (MAT), enzyme linked immunosorbent assay (ELISA), direct dark-field microscopic (DFM) examination,<br />

indirect fluorescent antibody test (IFAT), microbiologic cultural isolation technique and polymerase chain<br />

reaction (PCR). Antibodies were detected by MAT at least against one serovar of Leptospira interrogans in<br />

55 sera (14.47%) among 380 samples at a dilution 1:100 or greater, and L. icterohaemorrhagiae was the<br />

most prevalent serovar. Leptospiral antibodies were detected by ELISA in 85 sera (22.37%) among 380<br />

samples. Four samples (12.12%) among 33 urine samples were suspected by DFM examination and no<br />

positive sample by IFAT was observed. Leptospires could be isolated from none of the 33 samples taken<br />

from industrial farms, but leptospires were isolated from urine samples taken from two clinically affected<br />

calves in a traditional farm. In this study, positive controls were detected only at a dilution equal to or greater<br />

than 2000 leptospires per each ml of urine sample by PCR, therefore, no DNA from serum and urine<br />

samples were collected from 6 industrial dairy farms, could be detected by this method. It seems that, to<br />

increase the precision in the diagnosis of the disease, using a range of reliable techniques and comparing<br />

the results is important in reaching final conclusion.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF A NEW AVIAN INFLUENZA ANTIBODY BLOCKING ELISA<br />

M. Zalunardo, K. Velek, R. Muñoz, J. Taxter, S. Michaud, V. Leathers<br />

IDEXX Laboratories<br />

One IDEXX Drive, Westbrook, Maine 04092, USA<br />

A new blocking enzyme-linked immunosorbent assay (ELISA) was developed based on nucleoprotein (NP)<br />

antigen coated on the solid phase which binds avian serum antibodies against avian influenza virus. A<br />

conjugate, prepared with monoclonal antibody directed against the conserved Influenza A nucleoprotein, is<br />

then used as the detection reagent. The presence of AI antibodies in a test sample is determined by the<br />

ability of the test sample to inhibit binding of the anti-NP conjugate to the plate. Because the basis of the test<br />

is the ability of sample antibody to block a conserved immunodominant epitope on the avian influenza<br />

nucleoprotein (NP) coated plate, the assay format does not require use of any species specific reagents and<br />

is therefore broadly applicable for any avian species that generates an antibody response to the NP protein<br />

of avian influenza.<br />

Well characterized serum samples from different avian species (commercial and non-commercial chickens,<br />

turkeys, ducks, geese, quail and ostriches) were analyzed with this new ELISA. The results showed an<br />

overall specificity of 99.7% and excellent correlation with other traditional test methods, such as agar gel<br />

immunodiffusion (AGID) and hemagglutination inhibition (HI) tests, and improved specificity when compared<br />

to indirect ELISAs.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIAGNOSTICS OF PARASITIC DISEASES OF<br />

AGRICULTURAL ANIMALS IN UZBEKISTAN<br />

M.Aminjonov, professor<br />

Sh. Temirov, a post-graduated student<br />

(Uzbek veterinary scientific research institute)<br />

One of the wide spread and dangerous to human health and animals parasitic diseases in Uzbekistan is<br />

echinococcosis. According to Ministry of public health of the republic of Uzbekistan annually more then 6<br />

thousand people of difference age (18-20% children up to 15 years old, more then 50% women) are<br />

operated. Operation fee in echinococcosis of one man is more then 10 thousand dollars (USD).<br />

There is observed a trend of echinococcosis growing so among the population as among the animals.<br />

According to our data sheep are infected with echinococcosis up to 75-80%, cattle is infected – 45% and<br />

more, donkeys – 35,0% and so on.<br />

Economic damage from echinococcosis among the animals a year was equal to 5 mln. dollars (USD).<br />

Success of struggle against echinococcosis in many times depend on in-time its diagnostics.<br />

There is widely used reaction indirect hemaglutination in Stepanovskiy’s modification for recognizing of<br />

echinococcosis in Uzbekistan.<br />

With aim of discovery of antibodies’ titres in above mentioned reaction 10 heads lambs experimentally<br />

infected with echinococcosis were used. As control was 5 heads lambs not infected. In experimentally<br />

infected animals positive titres appeared from 40 th day after infection (1:320) and maximally reached on 180 th<br />

day. On that day antibodies’ titres of few animals were 1:5260 and than there was observed decreasing of<br />

titres and in one year was equal to 1:320 and 1:160.<br />

Titres reaction of some animals did not exceed 1:160.<br />

Slaughter of animals in one year showed high infection. Number of cycts echinococcus formed in average<br />

52-712 copies a head, control animals were free from invasion.<br />

So, sensitivity and specificity of used reaction was 80,0% and higher, and it is acceptable in condition of<br />

sheep farmers of Uzbekistan and neighboring republics of Central Asia.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

TARGETED SELECTIVE TREATMENT OF OSTERTAGIA OSTERTAGI<br />

- THE USE OF AN ELISA AS A DIAGNOSTIC TOOL FOR THE CONTROL OF THE PARASITE IN<br />

CATTLE<br />

J Troeng, J Vercruysse, J Charlier, A Nordengrahn, M Merza*<br />

Svanova Biotech AB, Uppsala Science Park, 751 83 Uppsala, Sweden<br />

Introduction<br />

A prerequisite to identify herds affected by parasites and then to treat only those animals that suffer from<br />

clinically significant parasitism (according to TST) is access to diagnostic tool that can be included in<br />

surveillance programs. The nematode infestations in adult cows are predominanty subclinical, but may lead<br />

to a decrease in milk production. One of the most important factors is to find the infestation level<br />

corresponding to the necessity of anthelmintic treatment. The screening for O. Ostertagi antibodies in cattle<br />

milk samples have been demonstrated to be a promising parameter to determine the infestation level, thus<br />

becoming an instrument to determine the need for antheliminic control.<br />

Material & methods<br />

The kit procedure is based on a solid-phase indirect ELISA where milk samples are exposed to O. Ostertagi<br />

antigen. A HRP conjugated a-bovine Ig is used as detection antibody.<br />

In order to establish the relation between OD value read in a spectrophotometer and the change in milk<br />

yield, bulk milk samples from more than 800 European herds were studied.<br />

Results<br />

The study established a good correlation between OD values and change in milk yield. The data were further<br />

analysed and put into a diagram which can be used for the interpretation of single bulk milk sample tested in<br />

the ELISA in order to assess the importance of the infestation in a specific herd.<br />

Discussions & conclusions<br />

We have developed an O. Ostertagi ELISA for antibody detection in serum and milk. This ELISA used on<br />

bulk tank milk, is a promising diagnostic tool that can be used on a large-scale to identify dairy herds where<br />

the infestation level with gastrointestinal nematodes is likely to reduce productivity. By using this ELISA the<br />

correlation between a certain level of antibodies to O. Ostertagi in a bulk milk sample and the herd average<br />

milk yield reduction can be established and thereby being a tool for determination of the need for<br />

anthelmintic control. The ELISA has been commersialised as Svanovir® Ostertagia Ostertagi ab ELISA.<br />

References<br />

Charlier J., Duchateau, L., Claerebout E., Vercruysse J., 2007. Predicting milk-production responses after an<br />

autumn treatmend of pastured dairy herds with eprinomectin. Vet. Parasit. 143, 197-390.<br />

Charlier J., Claerebout E., Duchateau L., Vercruysse J., 2005. A survey to determine relationships between<br />

bulk tank milk antibodies against Ostertagia ostertagi and milk production parameters. Vet. Parasit.<br />

129, 67-75.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

EVALUATION OF POSTVACCINAL ANTIBODIES LEVEL AFTER VACCINATION WITH MARKER GE<br />

BHV1 VACCINE IN BOVINES, USING ELISA METHOD<br />

C. Radulescu, I. Olvedi<br />

Institute for Diagnosis and Animal Health, Virology Dep., Bucharest, Romania<br />

Key words: vaccination, marker, BHV1, ELISA<br />

Aims:<br />

Bovine Herpesvirus 1 (BHV1), a member of the Herpesviridae family (subfamily Alphaherpesvirinae), is the causative agent of infectious<br />

bovine rhinotracheitis (IBR), infectious pustular vulvovaginitis (IPV), infectious balanoposthitis (IBP) and several other clinical<br />

syndromes. BHV1 infection usually affects the respiratory or genital tracts.<br />

Infectious bovine rhinotracheitis is highly contagious and can be spread through airborne and sexual transmission, although infection<br />

can also occur through other routes. The disease is widespread throughout the world. In Romania, IBR was first diagnosed in 1964, but<br />

serological surveillance started in 2000.<br />

The purpose of this work is the evaluation of postvaccinal antibodies level after vaccination with marker gE BHV1 vaccine in bovines,<br />

using ELISA method.<br />

Materials and methods:<br />

Vaccine and vaccination scheme: the bovines were vaccinated with IBR live marker vaccine; this is an attenuated vaccine containing at<br />

least 10 5.7 TCID50 of gE¯ BHV-1, strain GK/D.<br />

Challenge study: was made on 50 bovine which were grouped in 7 age categories:<br />

Batch no. I: 7 dairy cattle;<br />

Batch no. II: 7 heifers;<br />

Batch no. III: 8 heifers at 12-18 month old;<br />

Batch no. IV: 10 heifers at 6-12 month old;<br />

Batch no. V: 10 calves at 3-6 month old;<br />

Batch no. VI: 4 calves at 1-2 month old;<br />

Batch no. VII: 4 control animals for uninfected adults.<br />

These bovines were vaccinated with 2 ml/animals following the program:<br />

Primary vaccination: - intranasal for the calves a 1-2 month old;<br />

- intramuscular for other batches.<br />

Secondary vaccination: was applied only the calves at 1-2 month old, intramuscular, after 3-4 month from the first vaccination.<br />

Samples: Blood samples were harvest from all 50 animals before vaccination.<br />

After the vaccination, the samples were taken thus:<br />

- after 28 and 90 days from primary vaccination for all bovines;<br />

-after 21 days, from recall vaccination for calves a 1-2 month old.<br />

Methods: - Chekit Trachitest Serum screening, ELISA kit, used for titrations the antibodies against bovine herpes virus 1 (BHV) before<br />

and after vaccinations;<br />

- Chekit BHV1 gE, ELISA kit used for detection vaccinal antibodies<br />

Results:<br />

During this experiment, all animals were clinically examined and the body temperature was measured. There were no clinical alterations<br />

or mortality in inoculated animals.<br />

Before challenge:<br />

- bovines of the batch no. I showed a high, stable and homogenous BHV1 titer of antibodies (around 1:64 128);<br />

- animals of the batch no. II present a low titer of antibodies (approximately 1:4);<br />

- only 2 heifers a 12-18 month old have detectable antibodies titer (1:4);<br />

- the batches no. IV and V showed no antibodies, being negative at ELISA;<br />

- controls (batch no. VII) remained seronegative, indicating the absence of the wild BHV1 contamination.<br />

After challenge:<br />

- the seroconversion observed at 28 days was significant for batch<br />

no. I, II, III, IV, V vaccinated i.m., and less seroconversion in batch nr. VI that received the vaccine on the nasal route;<br />

- at 90 days after vaccination, titers of detected antibodies were<br />

lower than previous determination, but higher than initially observed;<br />

- at the animals with secondary i.m. vaccination the titers of anti-<br />

bodies were higher then titers after nasal vaccination, but<br />

lower when compared with titers registered in the rest of batches;<br />

- controls remain negative, showing no detectable antibodies.<br />

Conclusion:<br />

It was observed after 28 days post inoculation significant seroconversion consequently the inoculation of the IBR live marker vaccine by<br />

intramuscularly route and low seroconversion following nasal route administration. After 90 days post inoculation antibodies titer<br />

decreased, but it was higher than initially observed (before inoculation). Seronegative animals presented at all serological exams for<br />

specific antibodies detection, performed with Chekit BHV1 gE ELISA kit, negative results, although all bovines permanently stayed<br />

together (infected animals – seropositive- and seronegative).<br />

Reference:<br />

1. Flint.S.J., Enquist, L.W., Skalka,A.M., 2003- Priciple of virology, vol1.<br />

2. Monika Fuchs, Peter Hübert, Jan Detterer, and Hanns-Joachim<br />

Rziha - Detection of Bovine Herpesvirus Type 1 in Blood from<br />

Naturally Infected Cattle by Using a Sensitive PCR That<br />

Discriminated between Wild-Type Virus and Virus Lacking Glycoprotein E.<br />

3. Jones C. Alphaherpesvirus latency: its role in disease and survival of the virus in nature.<br />

4. Engels M, Ackermann M. Pathogenesis of ruminant herpesvirus infections.<br />

5. Mylène Lemaire, 1 Vincent Weynants, 2 Jacques Godfroid, 3 Frédéric Schynts, 1 Gilles Meyer, 1 Jean-Jacques Letesson, 2 and Etienne<br />

Thiry 1* - Effects of Bovine Herpesvirus Type 1 Infection in Calves with Maternal Antibodies on Immune Response and Virus Latency .<br />

6. Kupferschmied HU, Kihm U, Bachmann P, Muller KH, Ackermann M. Transmission of IBR/IPV virus in bovine semen: a case report.<br />

7. OIE, (2004b) Office International des Epizooties, Manual of Diagnostic Test and Vaccines for Terrestrial Animals, Fifth Edition, 2004,<br />

Chapter 2.3.5 – Infectious bovine rhinotracheitis/Infectious pustularã vulvovaginitis.<br />

8. Kramps JA, Banks M, Beer M, Kerkhofs P, Perrin M, Wellenberg GJ, Oirschot JT. - Evaluation of tests for antibodies against bovine<br />

herpesvirus 1 performed in national reference laboratories in Europe.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

VALIDATION OF AN AGAR GEL IMMUNODIFFUSION AND WESTERN BLOT ASSAYS FOR<br />

DIAGNOSIS OF EQUINE INFECTIOUS ANEMIA USING A RECOMBINANT p26 PROTEIN<br />

Alvarez, I. 1* , Gutierrez, G. 1 , Vissani, A. 1 , Ostlund, E 2 ., Barrandeguy, M. 1 and Trono, K. 1<br />

1 Instituto de Virología. Centro de Investigaciones en Ciencias Veterinarias y Agronómicas. INTA. C.C. 1712. Castelar. Argentina.<br />

2 NVSL: National Veterinary Services Laboratoryies. P. O. Box 844, Ames, IA 50010. USA * ialvarez@cnia.inta.gov.ar<br />

Introduction<br />

In Argentina, laboratory diagnosis of equine infectious anemia (EIA) is currently made by agar gel<br />

immunodiffusion test (AGID) designed by Coggins in 1970 (1). It is an inexpensive, simple and highly<br />

specific assay to identify EIAV-infected animals and it is still internationally recognized as the “gold standard”<br />

test by the World Organization for Animal Health (OIE) (2). The detection of EIA antibodies by ELISA has<br />

also been described and test kits have been approved in some countries. Several factors can contribute to<br />

yield conflicting results that need to be resolved using an alternative reliable and highly specific diagnostic<br />

tool. Western blot (WB) is currently used as confirmatory test and as an important tool for managing human<br />

and animal retroviral infections.<br />

The objective of this study was to validate an AGID assay and a confirmatory WB test using a recombinant<br />

p26 protein (rp26) produced in Escherichia coli as antigen.<br />

Materials and methods<br />

We employed international guidelines (3-5) to analyze the performance characteristics of both tests and we<br />

compared the results of a large number of field serum samples against those obtained with an imported<br />

commercially available AGID test kit.<br />

Results<br />

For the rp26-AGID test, a panel of 1855 field serum samples was analyzed, showing total concordance<br />

(100%) with the results obtained on the same samples using a commercial test kit.<br />

The agreement between results from 567 sera analyzed with the rp26-WB and commercial AGID was very<br />

good, with 98.9% of concordance. Six samples yielded discordant results between rp26-WB and a<br />

commercial AGID. These samples were sent to the NVSL (EIA reference laboratory) and fully examined<br />

using multiple EIA test formats The positive results obtained by using rp26-WB were considered to be truepositive<br />

since they were coincident with those obtained with the NVSL set of serologic tests. The rp26-AGID<br />

and rp26-WB assays demonstrated excellent performance characteristics. Excellent repeatability and<br />

reproducibility and total agreement with blind previous results from 5 proficiency test panels indicated that<br />

both test were precise assays. No cross-reactivity was observed when serum samples from horses with<br />

other infectious diseases or with poorly preserved samples were analyzed by rp26-AGID or rp26-WB assays.<br />

In the analytical sensitivity trial, positive sera showed nearly the same endpoint dilutions for rp26-AGID and<br />

commercial tests, and for the rp26-WB, demonstrated to be higher than AGID.<br />

Discussion and conclusions<br />

This is the first time that a recombinant AGID assay able to identify EIAV infections has been standardized<br />

and validated in Argentina according to international guidelines. Based on the results obtained, the p26-<br />

AGID could be adopted in the Argentinean current context as an official test method. Additionally, the rp26-<br />

WB was shown to be an accurate confirmatory diagnostic tool to be used as a complementary test after a<br />

doubtful ELISA or AGID test, available at national level for confirmation of conflicting results that contribute to<br />

resolve legal and/or sanitary situations that can be propitious to the dissemination of AIE in Argentina.<br />

References<br />

(1) Coggins, L., and N. L. Norcross (1970). Immunodiffusion reaction in equine infectious anemia. Cornell<br />

Vet. 60(2):330-335.<br />

(2) World Organization for Animal Health (OIE) (2004). Manual of Diagnostic Test and Vaccines for<br />

Terrestrial Animals. Volume II. Chapter 2.5.4. Equine infectious anemia.<br />

(3) International Committee of Harmonisation. Tripartite Guideline Q2A: Text on validation of analytical<br />

procedures, (1994); and Tripartite Guideline Q2B: Validation of analytical procedures: methodology, (1996).<br />

(4) World Organization for Animal Health, (OIE). 2004. Manual of Diagnostic Test and Vaccines for<br />

Terrestrial Animals, Volume I. Chapter 1.1.3. Principles of validation of diagnostic assays for infectious<br />

diseases.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NEW DIAGNOSTIC ASSAYS FOR THE DETECTION AND MONITORING OF COCCIDIOSIS IN POULTRY<br />

J. A. T. Morgan 1 , C. Constantinoiu 1,2 , J. B. Molloy 1 , G. T. Coleman 2 and W. K. Jorgensen 1*<br />

1 Emerging Technologies, Department of Primary Industries and Fisheries, Yeerongpilly, Qld.<br />

2 School of Veterinary Science, University of Queensland, Qld.<br />

Introduction<br />

Poultry farms suffer significant economic loss from infection with the parasitic disease coccidiosis (Shirley,<br />

Smith and Tomley, 2005). Transmission is via ingestion of infective oocysts which are shed in the faeces.<br />

Seven species have been identified from chickens based on varying biology and pathogenicity (Sutton,<br />

Shirley and Wisher, 1989). Control methods include chemical coccidiostats and live species-specific<br />

vaccines. To maximise the effectiveness of coccidiosis control programs effective diagnostic assays are<br />

essential. Species diagnosis has traditionally been based on morphological characters and developmental<br />

biology but these methods have proved problematic, particularly with mixed species infections. Diagnostic<br />

tests must be suitable for epidemiological investigations and investigating coccidiosis outbreaks.<br />

Conventional PCR and sequence based DNA markers have improved species discrimination but lack the<br />

ability to quantify Eimeria load, particularly in animals with mixed species infections. Quantitative detection<br />

and serological assays are both essential to meet these industry requirements.<br />

Material & methods<br />

Seven real-time PCR assays were developed for the detection and quantification of Australian Eimeria<br />

species. The assays use species-specific minor groove binder (MGB) TaqMan probes designed with FAM or<br />

VIC flourophores so that the seven species can be screened by multiplexing in 4 reaction tubes. Assays can<br />

be applied to DNA extracted directly from faecal samples are non-invasive and permit accurate identification<br />

to species, and quantification of oocyst load. Profiles of mixed species infections in commercial flocks were<br />

monitored over time. An indirect ELISA has also been developed from merozoite antigens of E. tenella.<br />

Since they detect antibodies rather than the parasites themselves, ELISAs can detect exposure to Eimeria<br />

species even when the parasites are no longer present. The indirect ELISA was used to assess sera from<br />

vaccinated and unvaccinated birds from commercial farms.<br />

Results<br />

The seven newly developed real-time PCR assays show no cross reactivity to non-target species. Profiling of<br />

poultry broiler farms found that mixed species infections were common, there was large bird to bird<br />

variability, and that peak oocyst shedding occurred after 18 days. The newly developed indirect ELISA can<br />

detect exposure of chickens to low numbers of oocysts of E. tenella. Antibodies were detected by 14 days<br />

post-infection and, depending on the rearing system, persisted in most birds for greater than 10 weeks. In<br />

field screens 99% of birds in a flock vaccinated against coccidiosis and 97% of free-range unvaccinated<br />

layers, were positive in the ELISA. In contrast, less than 1% of unvaccinated broilers fed a diet containing a<br />

chemical coccidiostat were positive.<br />

Discussions & conclusions<br />

Current Australian vaccines only protect against 4 species, therefore accurate disease diagnosis is important<br />

to the poultry industry where coccidiosis is a costly and virtually ubiquitous problem. The developed DNA<br />

based assays are species-specific and quantitative. They are sensitive enough to detect low level infections<br />

allowing infections at economic thresholds to be identified. The indirect ELISA provides a quick and<br />

inexpensive test that has high throughput capacity. The new assays can be used to investigate outbreaks,<br />

monitor and quality control vaccination programmes, check the effectiveness of biosecurity measures and<br />

identify the development of chemical resistance.<br />

References<br />

Shirley, M. W., Smith, A. L. and Tomley, F. M. (2005) The biology of avian Eimeria with an emphasis on their<br />

control by vaccination. Advances in Parasitology 60: 285–330.<br />

Sutton, C. A., Shirley, M. W. and Wisher, M. H. (1989) Characterisation of coccidial proteins by twodimensional<br />

sodium dodecyl sulphate-polyacrylamide gel electrophoresis. Parasitology 99: 175-187.<br />

Acknowledgement<br />

This research was partly funded by RIRDC project DAQ-316A<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ACCURATE DETECTION OF BOVINE LEUKEMIA VIRUS USING RECOMBINANT P24-ELISA AND PCR<br />

G. Gutiérrez*, M.J. Dus Santos, I. Alvarez, M. Lomónaco, R. Politzki, S. Rodriguez and K. Trono. Instituto de Virología, Centro de<br />

Investigaciones en Ciencias Veterinarias y Agronómicas, INTA, CC 1712 Castelar, Argentina<br />

Introduction<br />

Diagnosis of Bovine leukemia virus (BLV) infection is currently carried out by agar gel immunodiffusion<br />

(AGID) and ELISA (1). Even when both tests are approved by the OIE for trade purposes, several factors<br />

can contribute to yield conflicting results that need to be confirmed. In this work, ELISA and PCR were used<br />

as complementary tools to confirm the real sanitary status for specific situations as breeding or<br />

commercialization. The performance characteristics of an in-house developed recombinant ELISA (rp24-<br />

ELISA) were evaluated according to international guidelines (2,3).<br />

Material & methods<br />

The BLV p24 major capsid protein was expressed as a recombinant thioredoxin-6xHis fusion protein in<br />

Escherichia coli in our laboratory. The purified rp24 was used as antigen for the ELISA test (rp24-ELISA) to<br />

detect antibodies to BLV. To assess the performance characteristics of this test 710 field serum samples<br />

were analyzed by rp24-ELISA and a commercial AGID test (UNLP AGID). Amplification of the envelope gene<br />

by nested PCR (nPCR) (4) and an in-house Western Blot assay were used to confirm discordant results on a<br />

well-characterized herd.<br />

Results<br />

rp24-ELISA and AGID showed a 90.5% of concordance when the 710 field serum samples were analyzed<br />

on the accuracy trial. rp24-ELISA could detect 269 positives samples while AGID only detected 210. When<br />

nPCR was carried out 18 out of 22 samples that were declared as positive by rp24-ELISA were positive by<br />

nPCR. In addition, the rp24-ELISA was able to detect 5 out of 21 infected animals that were not detected by<br />

AGID. Five out of 8 samples that were not declared as positives by AGID were detected by WB and rp24-<br />

ELISA. Complementary, the rp24-ELISA demonstrated to be a precise assay with excellent repeatability,<br />

reproducibility and better analytical sensitivity than AGID<br />

Discussions & conclusions<br />

Taking these results into account, the rp24-ELISA could be adopted as an official test method for diagnosis<br />

and control of BLV in Argentina, and together with the nested-PCR assay could be considered as<br />

complementary tools for confirmatory situations.<br />

References<br />

1- World Organization for Animal Health (OIE). 2004. Manual of Diagnostic Test and Vaccines for Terrestrial<br />

Animals (mammals, birds and bees). Chapter 2.3.4. Enzootic bovine leukosis<br />

2- World Organization for Animal Health (OIE). 2004. Manual of Diagnostic Test and Vaccines for Terrestrial<br />

Animals (mammals, birds and bees). Chapter 1.1.3. Principles of validation and of diagnostic assays for<br />

infectious diseases.<br />

3- International Committee of Harmonisation. Tripartite Guidelina Q2A: Text on Validation of Analytical<br />

Procedures, 1994, and Tripartite Guideline Q2B: Validation of Analytical Procedures: Methodology, 1996.<br />

4-Lew, AE; Bock, RE; Molloy, JB; Minchin, CM; Robinson, SJ; Steer, P. Sensitive and specific detection of<br />

proviral bovine leukemia virus by 5´Taq nuclease PCR using a 3´ minor groove binder fluorogenic probe. J<br />

Virol Methods, 2004 Feb;115(2):167-75<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

IS THE COXIELLA BURNETII NINE MILE STRAIN A SUITABLE ANTIGEN FOR DETECTION OF<br />

Q FEVER ANTIBODIES OF RUMINANTS BY ELISA?<br />

*A Rodolakis 1 , M Bouzid 1,2 , A Souriau 1 , P Camuset 3 , M Berri 1 1 IASP INRA, Nouzilly France, 2 IRVT Tunis Tunisie 3 SNGTV Paris France<br />

Introduction<br />

Coxiella burnetii, the causative agent of Q fever, a worldwide zoonosis, infects various animal species.<br />

However ruminants are considered as the most common reservoir for human infection as infected ruminants<br />

shed high concentration of C burnetii in placenta and parturition products but also in milk, in faeces and in<br />

urine, which are the source of environmental contaminations causing human disease. In ruminants C burnetii<br />

may cause abortion, pneumonitis and retained placentitis leading to metritis and infertility namely in cows.<br />

But C burnetii infections are frequently asymptomatic and numerous ruminants without any clinical symptom<br />

shed the bacteria. The diagnosis of Q fever in ruminants is generally performed by PCR combined with<br />

serological analysis of serum samples by ELISA. However, several female animals that shed C burnetii in<br />

vaginal mucus or milk even for several months remained ELISA negative. As human chronic Q fever could<br />

be related to immunodeficiency, the aim of this work was to investigate if these seronegative animals were<br />

unable to produce antibodies against C burnetii or if the antigen used in the ELISA kit failed to detect them.<br />

Material & methods<br />

Seven C burnetii strains were used: the Nine Mile (NM) strain isolated from ticks which is the antigen used in<br />

ELISA kits and 6 strains isolated from ruminants. Four of them were isolated from placenta of aborted<br />

females: CbB1 from a cow, CbO1 and CbO184 from a ewe and CbC1 from a goat. The CbC2 and CbB2<br />

strains were isolated from the milk of an asymptomatic goat and an infertile cow respectively 1 . The antigenic<br />

variability of C burnetii strains were analyzed by immunoblotting with the serum of a goat naturally infected<br />

and by immunofluorescence (IF) 2 with serum of mice immunized with CbB1, CBC1, CbC2, CbO1 and NM C<br />

burnetii strains. The presence of antibodies against the 4 C burnetii strains, NN, CbO1, CbC1, CbC2 in the<br />

ELISA-negative serum samples of ruminants shedding C burnetii in vaginal mucus, milk or feces 3 was<br />

investigated by IF<br />

Results<br />

Immunoblotting and IF using immune sera obtained from either mice or naturally infected ruminants<br />

exhibited antigenic variability between NM strain and strains isolated from domestic animals. These<br />

techniques also revealed minor antigenic variability inter strains isolated from ruminants. The serum of the<br />

animals that shed C burnetii and which were ELISA negative, were also negative in IF with the NM antigen<br />

but they were positive against at least one of the 3 strains isolated from ruminants: 29 sera were positive<br />

against the CbO1 strain and 13 gave of doubtful reaction. CbC1 and CbC2 strains detected 19 and 17<br />

positive sera respectively. For both of them, 3 sera gave a doubtful reaction.<br />

Discussions & conclusions<br />

The ELISA seronegative cows, ewes or goats that shed C burnetii in vaginal mucus, feces or milk were able<br />

to produce antibodies but the antigenic variability between NM strain and the strains isolated that infect<br />

ruminants did not allow their detection. So the C burnetii N M strain, widely used in ELISA kits, is not suitable<br />

for the diagnosis of Q fever in ruminants, as there were lots of false negative responses with such kits. IF<br />

tests are not widely used in veterinary diagnosis as they cannot be automated; and they require skilled<br />

technicians and expensive equipment. Nevertheless ELISA is supposed to be slightly more sensible than IF 4 .<br />

So these results have lead us to develop an ELISA kit using a C burnetii strain isolated from ovine as antigen<br />

that improves the sensitivity of the serological diagnosis of Q fever in ruminants. To valid a test, this work<br />

underlines how, in absence of “Gold Standard”, it is crucial to dispose of a huge collection of serum samples<br />

from flocks of well known sanitary status.<br />

References<br />

1 Arricau-Bouvery, N., Hauck Y., Bejaoui A., Frangoulidis D., Bodier C.C., Souriau A., Meyer H., Neubauer<br />

H., Rodolakis A., Vergnaud G. BMC Microbiology, 2006 Apr 26; 6(1):38<br />

2 Souriau, A., Le Rouzic, E., Bernard, F., Rodolakis, A., 1993. Vet. Rec. 132, 217-219.<br />

3 Rodolakis A Berri M, Héchard C, Caudron C, Souriau A, Bodier CC, Blanchard B, Camuset P,<br />

Devillechaise P, Natorp JC, Vadet JP, Arricau-Bouvery N. 2007 JDS In press<br />

4 Rousset E, Durand B, Berri M, Dufour P, Prigent M, Russo P, Delcroix T, Touratier A., Rodolakis A, Aubert<br />

M. 2007 Vet Microbiol, 124: 286-297<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIFFERENTIATION OF FOOT-AND-MOUTH DISEASE VIRUS (FMDV) INFECTED PIGS FROM<br />

VACCINATED PIGS USING A WESTERN BLOTTING (WB) ASSAY BASED ON BACULOVIRUS<br />

EXPRESSED FMDV 2C AND 3D ANTIGENS<br />

K. Fukai*, K. Morioka, S. Ohashi, R. Yamazoe, K. Yoshida, K. Sakamoto<br />

Exotic Diseases Research Station, National Institute of Animal Health, Japan<br />

Introduction<br />

FMD is probably the most contagious live stock disease known. FMDV is a member of the genus Aphtovirus<br />

within the family Picornaviridae. Seven serotypes have been identified, and multiple antigenic variants occur<br />

within each serotype. Outbreaks of FMD in a disease-free country or region are detrimental to the agriculture<br />

industry due to trade and movement restrictions and the severe impact on animal health and welfare, as well<br />

as society as a whole. Until now, the outbreaks within a FMD-free country have been controlled by a<br />

combination of stamping out and movement control. However, the massive destruction of animals during the<br />

FMD epidemic in the United Kingdom in 2001 emphasized the need alternative control procedures. Indeed,<br />

the European Union has been changed to allow the use of emergency vaccination for the control of FMD in<br />

Europe. In concordance, the time period given in the Terrestrial Animal Health Code of the Office<br />

International des Epizooties, before a country which has resorted to emergency vaccination can regain<br />

status as ‘disease free without vaccination’, has been reduced to six months, provided serological surveys<br />

for antibodies to FMDV non-structural proteins are carried out to ensure correct classification of infected<br />

animals in the presence of vaccination. Therefore, the availability of tests with high sensitivity and specificity<br />

as well as high capacity, is essential. In this study, to differentiate FMDV infected pigs from vaccinated pigs,<br />

a WB assay was performed with using baculovirus expressed FMDV 2C and 3D non-structural proteins as<br />

antigen.<br />

Material & methods<br />

Sera by infection were collected in constant intervals from three pigs inoculated 10 6 TCID50 FMDV Asia1<br />

Shamir. Ones by vaccination were prepared with a single administration of 6 PD50 FMDV Asia1 Shamir and<br />

O1 Manisa inactivated vaccine to four pigs. FMDV 2C and 3D proteins were expressed by using a<br />

baculovirus expression system. The serum samples were examined by a WB assay based on the<br />

baculovirus expressed 2C and 3D non-structural proteins of FMDV O/JPN/2000. And same samples were<br />

also tested by liquid phase blocking ELISA (LPBE) and virus neutralization (VN) assay.<br />

Results<br />

In the WB assay, positive reactions were obtained for sera from pigs infected with FMDV Asia1 Shamir. The<br />

assay detected initially antibodies from 6 days following experimental infection of non-vaccinated pigs, and<br />

the positive reactions persisted for up to 41 days after infection. In contrast, no positive reaction was<br />

obtained from vaccinated pigs for up to 210 days after vaccination. In the LPBE and VN assay, positive<br />

reactions were obtained for sera from both infected and vaccinated pigs. And the positive reactions persisted<br />

for up to 41 and 210 days after infection and vaccination, respectively.<br />

Discussions & conclusions<br />

In this study, the WB assay based on baculovirus expressed FMDV 2C and 3D antigens could clearly<br />

differentiate FMDV infected pigs from vaccinated pigs. In the future, the applicability of this WB assay needs<br />

to be verify using the field samples in FMDV endemic countries and the serum samples of pigs administrated<br />

an inactivated vaccine many times. And the assay which can simultaneously examine a lot of samples, i.e. a<br />

ELISA system, using these baculovirus expressed FMDV 2C and 3D non-structural proteins also needs to<br />

be established.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF A COMMERCIAL ELISA FOR THE DETECTION OF SERUM ANTIBODY TO<br />

AKABANE VIRUS<br />

Lunt RA * , White JR * , Newberry KM * , Pourquier P † , Comtet, L † , Whittle, S ‡ .<br />

* CSIRO Livestock Industries, Australian Animal Health Laboratory, P.O. Bag 24, Geelong 3220, Australia<br />

† ID VET, Cap Gamma - 167 rue Mehdi Ben Barka - 34070 Montpellier, France<br />

‡ Laboratory Diagnostics, Serenity Cove Corporate Park Kurnell, Australia<br />

Introduction<br />

Akabane virus (AKAV; family: Bunyaviridae, Genus: Orthobunyavirus) is an insect-borne virus of ruminants<br />

associated with epizootics of abortion, stillbirth and congenital abnormalities in cattle, sheep and goats. The<br />

virus has wide distribution across Africa, the Middle East, Cyprus, Asia and Australia 1 .<br />

We have developed a monoclonal antibody competition ELISA for the detection of antibody in ovine and<br />

bovine sera. Sensitivity and specificity of the assay has been investigated using panels of experimentallyderived<br />

and field collection sera. The assay has been transferred to a commercial company (ID-VET,<br />

France) for development as a commercial kit product. This paper describes the work undertaken to<br />

characterize assay performance.<br />

Material & methods<br />

Sera: 187 AKAV VNT positive sera and 1041 AKAV VNT negative sera were variously sourced as: 41 sera<br />

from animals experimentally infected with AKAV, 146 VNT positive field sera, 424 bovine and 593 ovine sera<br />

from non-endemic areas, 24 VNT-negative sera from AKAV endemic areas.<br />

Monoclonal antibody: A monoclonal antibody (17A12) was identified in preliminary work as an AKAV<br />

neutralizing antibody, with binding specificity for G1 protein identified by radio-immunoprecipitation, but not<br />

detected in western blots.<br />

Virus: AKAV isolate B8935 was used in VNT serology and antigen production for use in ELISA kits.<br />

VNT: sera were characterized by antibody neutralization for AKAV and Aino virus (AINOV) in a standard<br />

VNT format using VERO cells and 100TCID50 virus input. Sera with titres of greater than 4 were considered<br />

VNT antibody positive.<br />

ELISA: Following initial characterization, antigen and monoclonal antibody were incorporated into an ID-VET<br />

ELISA. Kit components included microplates coated with AKAV antigen (inactivated virus), control sera,<br />

horseradish peroxidise conjugated 17A12 monoclonal antibody, buffers and substrate solutions. Sera were<br />

tested according to the product insert method and in single wells. In brief, sera diluted 1/10 were added to<br />

plates for 45 minutes at 37°C. Plates were washed and conjugate added for 30 minutes. Substrate was<br />

reacted in washed plates for 15 minutes. After addition of stop solution, plates were read at OD 450nm. The<br />

sample to negative control ratio (S/N %) was used to identify reactive sera: S/N < 50% was regarded as<br />

positive.<br />

Results<br />

At the cut-off level (S/N = 50%), assay sensitivity and specificity were estimated at 0.94 and 0.98<br />

respectively, relative to VNT positive and negative division of the entire 1228 panel of sera. 19 of 20 VNT-<br />

NEG, ELISA-POS sera were derived from areas endemic for AKAV. Possible influence in the ELISA of<br />

antibody against other Simbu serogroup viruses such as AINOV was investigated. A panel of 9 AKAV VNT-<br />

NEG, AINOV VNT-POS field sera included 2 ELISA positives. However tests of sera from sheep<br />

experimentally exposed to various Simbu viruses (Peaton, Douglas, Simbu and Tinaroo) did not confirm any<br />

cross-reactivity. 9 of 11 AKAV VNT-POS, ELISA-NEG sera had VNT tires of ≤ 1/32. Of these sera, 6 of 11<br />

had S/N values between 50 to 70%, indicating that some below threshold inhibition of monoclonal binding<br />

was occurring.<br />

Discussions & conclusions<br />

The development of ELISA-based assays as alternatives to VNT assays has advantages in sample<br />

throughput capability and the removal of dependencies on live virus and cell culture requirements. An AKAV<br />

neutralizing monoclonal antibody (17A12) was identified as potentially useful in a competition format ELISA<br />

for serum antibody. Through collaboration with ID VET, an ELISA kit was developed and assessed. A range<br />

of sheep and cattle sera from field and experimentally infected animals were used in determinations of<br />

operational levels of sensitivity and specificity. Performance characteristics of the ID-VET AKAV competition<br />

ELISA suggest suitable applications could include diagnostic investigations and livestock movement<br />

certifications in testing that might otherwise require VNT serology.<br />

Reference<br />

1. Saeed MF,Li L, Wang H,Weaver SC. Barrett ADT (2001). Phylogeny of the Simbu serogroup of the genus<br />

Bunyavirus. J Gen. Virol. 82: 2173-2181.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PRELIMINARY VALIDATION OF A NEW MULTI-SPECIES COMPETITIVE ELISA KIT FOR THE<br />

DETECTION OF ANTIBODIES DIRECTED AGAINST THE WEST NILE VIRUS: ID SCREEN® WEST NILE<br />

COMPETITION<br />

P. Pourquier, S. Lesceu, ID VET, Montpellier, France<br />

Introduction and objectives:<br />

The West Nile virus (WNV), which causes encephalitis in infected humans and horses, is maintained in<br />

nature by a mosquito vector and bird reservoir host. Given the need for a rapid test able to diagnose the<br />

West Nile virus in horses and birds, ID VET (www.id-vet.com) has developed a multi-species competitive<br />

ELISA.<br />

The aim of this study was to validate a commercial ELISA developed by ID VET for the detection of anti-<br />

WNV antibodies in serum from different species (equine, avian).<br />

Test principle:<br />

The test is a competitive ELISA. The antigen is an immunocaptured E antigen. The conjugate is an anti-E<br />

peroxidase (Po).<br />

Methods :<br />

Sensitivity was evaluated by testing 20 positive horse sera from the South of France where WNV outbreaks<br />

were observed in 2003 and 2006.<br />

Specificity was studied by testing 320 negative horse sera from Calvados, France and 500 disease-free<br />

avian sera (ducks and chickens) from the Loire-Atlantique region in France.<br />

Results :<br />

The ID Screen® kit detected all 20 WNV-positive horse sera as positive and all WNV-negative horse and<br />

avian sera as negative.<br />

Conclusion:<br />

The ID Screen® test gives satisfactory specificity results on both bird and horse sera, and satisfactory<br />

sensitivity results on horse sera. The test is rapid, standardized and easy-to-use.<br />

The next step in the kit’s validation is to test positive avian sera. These samples are difficult to find given the<br />

low volume of sera obtained from wild birds.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

VALIDATION OF A NEW COMMERCIAL KIT FOR THE DETECTION OF ANTIBODIES DIRECTED<br />

AGAINST LEISHMANIA INFANTUM IN CANINE SERA: ID SCREEN® LEISHMANIASIS INDIRECT<br />

P. Pourquier 1 , S. Lesceu 1 , J. Dereure 2 , N. Keck 3<br />

1 ID VET, Montpellier France<br />

2 Parasitology-Mycology Laboratory, Centre Hospitalier Universitaire Montpellier, Leishmaniasis National Reference Centre,<br />

Montpellier, France<br />

3 Laboratoire Départemental Vétérinaire, Montpellier, France.<br />

Introduction:<br />

The visceral form of leishmaniasis is caused by Leishmania infantum in the Mediterranean area. The<br />

domestic dog, sensitive to infection by L. infantum, is an important reservoir host for transmission of the<br />

human disease.<br />

The aim of this study was to validate a commercial ELISA developed by ID VET (www.id-vet.com) for the<br />

detection of anti-L. infantum antibodies in canine sera.<br />

Test principle:<br />

The test is an indirect ELISA. The antigen used is a purified extract of the promastigote form of L. infantum.<br />

The conjugate is an anti-dog peroxidase.<br />

Methods:<br />

Canine sera from Hungary (disease-free zone, n=192), from Gers, Ariège, and Aude, France (moderatelyinfected<br />

zones, n=515), and from Hérault, France (highly-infected zone, n=144) were studied. Samples were<br />

tested using the ID Screen® ELISA and results compared to those obtained by IFAT (Indirect Fluorescent<br />

Antibody Test) and counter immuno-electrophoresis (Hérault sera only).<br />

Results:<br />

Specificity of the ID Screen® ELISA was estimated to be superior to 99% and sensitivity superior to 98 %.<br />

Excellent correlation with IFAT for titers greater than or equal to 1/80 was observed.<br />

Conclusion:<br />

The ID VET test is an effective and rapid tool for the detection of anti-L. infantum antibodies in canine sera.<br />

The test demonstrates high specificity and sensitivity, and excellent correlation with IFAT.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF A ELISA KIT TO DETECT AND EVALUATE BOVINE VIRAL DIARRHEA VIRUS<br />

ANTIGEN IN THE CRITICAL STAGES OF VACCINE PRODUCTION<br />

A. Pecora 1 , A. Ostachuk 1 , S. Levy 2 , A. Espinoza 2 , M. J. Dus Santos 1* and A. Wigdorovitz 1<br />

(1) Instituto de Virología, CICVyA, INTA-Castelar, Buenos Aires, Argentina<br />

(2) Biogénesis-Bago, Argentina<br />

* mdussantos@cnia.inta.gov.ar<br />

Introduction<br />

Bovine viral diarrhea virus (BVDV) is the causal agent of a worldwide spread disease. It infects bovines of all<br />

ages, causing reproduction problems and altering biological products of high commercial value, resulting in<br />

considerable economic losses. Persistently infected cattle disseminate large amounts of virus into the<br />

environment representing a mechanism by which the virus persist.<br />

Due to worldwide distribution of BVDV, disease control is based on the segregation of persistently infected<br />

cattle and efficient vaccination of the herd. According to that, the aim of vaccination against BVDV is to<br />

prevent transplacental transmission of the virus.<br />

Production of a BVDV vaccine usually presents the difficulty of obtaining enough antigen in order to induce<br />

high levels of neutralizing antibodies.<br />

The objective of this work was to standardize an enzyme-linked immunosorbent assay (ELISA) to detect and<br />

evaluate BVDV antigen in the critical stages of vaccine production.<br />

Material & methods<br />

ELISA procedure<br />

U-bottom polystyrene microplates (Maxisorp, NUNC) were coated with an anti-E2 mouse monoclonal<br />

antibody (1:100 dilution). Samples were added at the corresponding dilution and incubated 1h at 37°C. As<br />

primary antibody, sera from rabbit immunized with a truncated version of E2 (1:2000) were used.<br />

Peroxidase-conjugated goat anti-rabbit IgG (1:2000) (KPL) were used as secondary antibody. The reaction<br />

was visualized with ABTS (Sigma) and absorbance was measured at 405 nm in a Multiskan EX<br />

(Labsystyem) reader.<br />

Daily calibration curves using four concentrations (104 ng/ml, 26 ng/ml, 6.5 ng/ml and 1.6 ng/ml) (each level<br />

in duplicate) were prepared with purified tE2, and used to compute E2 levels.<br />

Standard curves<br />

A stable CHO-K1 cell line expressing a truncated version of BVDV E2 glycoprotein (tE2) was used for<br />

construction of standard curves. E2t was purified by Immobilized Metal Affinity Chromatography. Purity and<br />

protein concentrations were determined by SDS-PAGE and Coomasie blue staining using a BSA standard<br />

curve followed by densitometry analysis using Image J software and bicinconic method<br />

Results<br />

A sandwich ELISA kit for the detection of BVDV was developed. In order to quantify E2 in the sample tested,<br />

a standard curve was performed using purified tE2 as antigen. 8 concentrations of purified E2t were<br />

prepared for using as standards. An aliquot of each standard was tested 12 times. A linear dose-response<br />

curve was obtained (from 417 to 0.4 ng/ml of purified E2t) with a coefficient of correlation R≥0.93.<br />

Four concentrations of purified tE2 (104 ng/ml, 26 ng/ml, 6.5 ng/ml and 1.6 ng/ml) were prepared for use as<br />

QC samples for evaluation of intra and inter-assay precision and accuracy. Excellent levels of repeatability<br />

and reproducibility were obtained.<br />

The limit of detection of E2 in the supernatant of the stable cell line was 0.15 ng E2/ul. The test also allows<br />

detecting E2 glycoprotein in BVDV cell culture production. For that purpose BVDV samples were treated with<br />

0.25% Triton X114 (Sigma-Aldrich). The assay could detect up to 10 4,5 TCID50/ml BVDV, Singer strain.<br />

Different BVDV productions and experimental and commercial vaccines were assayed. The ELISA was able<br />

to detect and quantify E2 in live BVDV, inactive BVDV and in the formulated vaccine.<br />

Discussions & conclusions<br />

Results obtained indicate that the assay developed is a reliable tool to monitor the most relevant BVDV<br />

antigen in the process of vaccine formulation.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

FINE-TUNING A BULK MILK EBL ELISA AS PRIMARY SCREENING TOOL FOR ALL EBL-FREE<br />

HERDS IN NEW ZEALAND<br />

H Voges*, M Nash and T Trotter (LIC)<br />

Introduction<br />

Over 99.9% of New Zealand dairy herds have for several years achieved freedom from enzootic bovine<br />

leukosis (EBL) in accordance with OIE guidelines.<br />

An efficient tool to monitor these herds regularly as proscribed by the OIE Code is needed.<br />

The LACTELISA TM BLV Ab Bi Indirect Tank 250 test is designed for EBL antibody detection in vat milk<br />

samples from herds ranging of 150 to 250 cows (Ridge 2005). In the early stages of the NZ Dairy EBL<br />

Control Scheme, the Tank250 was employed to prioritise herds. Based on this dataset, Hayes & Burton<br />

(1998) reported test sensitivity and specificity equal to 81.9% and 98.7% respectively for herds of all sizes.<br />

Milk E4 dilution series<br />

Methods<br />

Using commercial EBL reference serum, a dilution trial was carried out with E4in EBL antibody-negative milk<br />

at 1: 200000 to 1: 1250000.<br />

Results<br />

At the recommended cut-off (0.2 S/P), test sensitivities fell to ~60% at high dilutions. In contrast, sensitivity<br />

was maintained at ~95% amongst high dilutions when the cut-off was reduced to 0.1 S/P. Clear separation<br />

between all E4 dilutions and EBL-free milk (+2σ) was maintained, suggesting a reduced cut-off may be<br />

feasible.<br />

Historical field data analysis<br />

Methods<br />

In 2000/01 & 2001/02 all NZ dairy herds were scheduled for a Tank250 test on a bulk milk vat sample. Case<br />

definition was difficult and imperfect, but only herds with 3 season test data were included:<br />

• Case + herds: 144 EBL-positive herds (at beginning and end of the season)<br />

• Case – herds: 13133 EBL-free herds that tested EBL negative on pool milk<br />

Results<br />

Overall Tank250 SE for the two seasons was 75.7% and the SP equal to 99.7% (95% CI: 99.5% - 99.7%). A<br />

ROC analysis shows modest SE gains were possible at the expense of small SP losses. Thus a cut-off at 0.1<br />

S/P resulted in an overall SE of 84.0% (SP 98.7%). In herds >500 cows SE rose from 64.3 to 76.2%.<br />

Operational performance and validation<br />

Methods<br />

During 2003/04 all high-risk and EBL-positive herds, as well as two-thirds of all EBL-free herds with up to<br />

300 cows, were screened by Tank250 using a reduced cut-off (S/P = 0.1).<br />

Results<br />

Changing the cut-off from 0.2 to 0.1 S/P resulted in big SE gains with a relatively small reduction in SP:<br />

� Tank250 SE amongst herds up to 600 cows was 100%<br />

� SE in herds >600 cows was 81.8% @ 0.1 S/P cut-off.<br />

� 4/5 herds >1000 milking cows (1-2 reactors) consistently tested positive on the repeated vat samples<br />

Repeatability of true positive results was extremely high for successive vat samples. Employing a serial 2test<br />

logic had no effect on test sensitivity but increased the SP substantially from 98.4% to 99.9% (p


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ADAPTING A DNA TISSUE SAMPLER AS AN AID FOR EFFICIENT BVDV ANTIGEN SCREENING<br />

H Voges, T Trotter, M Nash and M Walker(LIC, NZ)<br />

Introduction<br />

The use of ear biopsy samples to screen cattle for BVD virus by antigen-capture ELISA is common practice<br />

and may be used to avoid maternal antibody interference.<br />

The Genemark Punch has been designed to take 1mm samples using self-contained punch-pouches for<br />

DNA profiling of livestock. Principal advantages of the Punch for BVDv testing include prevention of crosscontamination<br />

while the small biopsy size minimizes pain and distress.<br />

Material & methods<br />

The trial objectives were to determine sample stability under adverse storage conditions and to ensure<br />

maximum test sensitivity for PI detection using the IDEXX HerdChek BVDv Antigen (Serum Plus) ELISA.<br />

Antigen extraction from tissues was carried out overnight and tested in accordance with the recommended<br />

protocol.<br />

Initial evaluation was carried out using Punch biopsies from two PI calves after storage for more than two<br />

weeks under a range of conditions as well as sample dilutions to 1:50.<br />

Thirty-five BVDv PCR-positive PIs – including 21 young calves (14 with maternal antibodies) – and 284<br />

PCR-negative in-contact cattle were sampled. All biopsies were tested after overnight extraction. A second<br />

(and third) extract from PI samples was also tested. Finally all PI extracts were diluted 1:13 in negative<br />

cohort extracts.<br />

Results<br />

The antigen ELISA results remained very stable over more than 2 weeks storage at a range of temperatures<br />

(4, 25, 37°C as well as fluctuating) and humidity.<br />

All raw PI sample extracts tested antigen-positive. All negative control biopsies were antigen-negative. A<br />

single pooled extract optical density reading fell below the test cut-off, although clearly higher than the<br />

negative controls. Punch ELISA optical densities of sero-positive calves were significantly lower than seronegative<br />

cattle (p < 0.01), although serum antigen ELISA results appeared unaffected by maternal<br />

antibodies.<br />

An additional 20 PIs were confirmed using the Punch ELISA.<br />

Discussions & conclusions<br />

The Punch was primarily designed to provide samples for parentage confirmation and PCR testing.<br />

However, BVDv is an RNA virus and hence sample integrity may be compromised by cellular RNAses<br />

(bovine of bacterial), especially in the presence of cell death as found in skin biopsies (Oakey 2007). The<br />

Punch was therefore not considered suitable for BVDv PCR testing as a PI screening tool. In contrast, the<br />

antigens detected by the IDEXX antigen ELISA are largely unaffected during storage.<br />

The Punch biopsy is a highly stable sample delivering excellent sensitivity for PI detection using the<br />

HerdChek Antigen Serum Plus ELISA. Sample pooling however resulted in unacceptable variability of<br />

optical densities.<br />

References<br />

Fux RG (2007) Entwicklung und Prüfung von Verfahren zum Nachweis des Virus der Bovinen Virusdiarrhoe<br />

in getrockneten Ohrgewebeproben mittels Antigen-ELISA und real time RT-PCR (Thesis). Ludwig-<br />

Maximilians-Universität, Munich<br />

Hill FI, Reichel MP, McCoy RJ and Tisdall DJ (2007) Evaluation of two commercial enzyme-linked<br />

immunosorbent assays for detection of bovine viral diarrhoea virus in serum and skin biopsies of cattle. New<br />

Zealand Veterinary Journal 55(1): 45-48<br />

Oakey HJ (2007) A universal test to determine the integrity of RNA, and its suitability for reverse<br />

transcription, in animal tissue laboratory specimens. Journal of Veterinary Diagnostic investigation 19: 459–<br />

464<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

VALIDATION OF AN INDIRECT BULK MILK BVD ANTIBODY ELISA FOR SERO-PREVALENCE<br />

INVESTIGATIONS AMONGST NEW ZEALAND DAIRY HERDS<br />

H Voges, T Trotter and M Nash (LIC, NZ)<br />

Introduction<br />

Given that contact with a persistently infected (PI) animal results in rapid sero-conversion of the majority of<br />

herd mates, the rationale behind a herd antibody, sero-prevalence test to gauge herd exposure to a BVDv PI<br />

animal is sound with careful interpretation, and has been used in BVDv control schemes overseas (Houe et<br />

al 2006; Niskanen 1993).<br />

We tested bulk milk vat samples using the indirect BVD antibody ELISA from IDEXX to assess the<br />

prevalence of BVDv infection amongst New Zealand dairy herds. To interpret the bulk milk test, we<br />

compared the result with individual sero-prevalence estimates in some herds.<br />

Material & methods<br />

Bulk milk vat samples from 350 herds across the country were obtained from the dairy companies and tested<br />

by BVD antibody ELISA in accordance with the manufacturer recommendations. Individual herd test milk<br />

samples (minimum 15) from 34 herds were subsequently tested by IDEXX BVD antibody ELISA for withinherd<br />

sero-prevalence estimation.<br />

Results<br />

Correlation between bulk milk ELISA and sero-prevalence estimates was remarkably high with R 2 equal to<br />

0.90.<br />

1.4<br />

1.2<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

BTM BVD indirect Ab ELISA (S/P)<br />

R 2 = 0.90<br />

– +<br />

proportion of BVD antibody-positive cows<br />

0.0<br />

0% 20% 40% 60% 80% 100%<br />

Vat milk samples from thirteen South Island herds were also tested by PCR. Three herds with low bulk milk<br />

antibody ELISA results were PCR negative, while four of ten high antibody herds (top right corner: S/P > 1.1,<br />

equivalent to ~90% sero-prevalence) were virus positive on PCR.<br />

Discussions & conclusions<br />

The national vat sampling and validation suggest that about one third of NZ dairy herds either harbour active<br />

BVD virus infection or have been infected recently (~90% sero-prevalence). Both Westland and Taranaki<br />

appear to have fewer BVD infected herds than other regions.<br />

References<br />

Houe, H., Lindberg, A., Moennig V. (2006) Test strategies in bovine viral diarrhea virus control and<br />

eradication campaigns in Europe. Journal of Veterinary Diagnostic Investigation 18 (5): 427-436.<br />

Niskanen, R. (1993) Relationship between the levels of antibodies to bovine viral diarrhoea virus in bulk tank<br />

milk and the prevalence of cows exposed to the virus. Veterinary Record. 133 (14): 341-344.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PRELIMINARY EVALUATION OF NEOSPORA CANINUM ELISA KITS FOR USE WITH NEW ZEALAND<br />

MILK SAMPLES<br />

H. Voges.<br />

A preliminary investigation was undertaken to assess the application of an indirect ELISA to detect<br />

antibodies against Neospora caninum in milk samples from New Zealand dairy herds.<br />

To determine the optimal rate of milk sample dilution for the IDEXX HerdChek Neospora caninum Antibody<br />

ELISA, individual milk samples were tested raw vs 1:2 vs 1:4 and compared to serum ELISA results. While<br />

the area under the ROC curve did not differ between treatments (0.998 vs 1.000 vs 1.000 respectively),<br />

greatest separation between positive and negative sample results was achieved at the 1:2 dilution with<br />

excellent correlation (R 2 = 0.91) between serum and milk results.<br />

Subsequently 308 bulk milk samples were selected randomly from across New Zealand. After 1:2 dilution the<br />

samples were tested by HerdChek Neospora ELISA. Using a cut-off at S/P >0.6 – indicating 15%+ seroprevalence<br />

according to Bartels et al (2005) – 7% of herds were found to be positive.<br />

The milk samples were also tested with the LSI Bovine Milk Neospora Caninum ELISA kit in accordance with<br />

manufacturer’s recommendations. The results were strongly correlated with serum test results (R 2 = 0.89) as<br />

well as the HerdChek (individual and bulk) milk ELISA. However the negative milk samples gave very high<br />

optical density readings and the kit will require fine-tuning for New Zealand milk.<br />

This limited trial data shows that milk samples may be useful to identify Neospora infected cattle or high<br />

prevalence herds in New Zealand, although the tests need further validation to refine the test cut-off values.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

ELISA BASED ON RECOMBINANT E RNS PROTEIN MIXTURES FOR SERELOGICAL DIAGNOSIS OF<br />

CLASSICAL SWINE FEVER<br />

*Seong-In Lim, Jae-Young Song, Eun-Jin Choi, Byoung-han Kim, Jaejo Kim, In-Soo Cho,<br />

Hoo-Don Joo 1 , Byoung-Sik Chang 1 , Hyun-Jung Lee 1<br />

Virology Division, National Veterianry Research & Quarantine Service, Anyang, Korea.<br />

1 Jenobiotech, Co., Ltd., Chuncheon, Gangwon-do, Korea.<br />

Introduction<br />

The principle of marker vaccine strategy is that the antibodies of vaccinated animal can be distinguished<br />

from those of infected animal against classical swine fever (CSF) virus. Serologically the detection of a CSF<br />

infection after vaccination with a marker vaccine depends entirely on the discriminatory ELISA. Recently,<br />

although two conventional anti-E rns ELISAs were developed and commercialized, the two test kits showed<br />

considerable discrepancies in CSF antibody titer between different CSFV genotypes (Vet Microbiol., 2001,<br />

83:121-136). In this study, a discriminatory ELISA was developed to detect antibody against the second viral<br />

envelope glycoprotein E rns of CSFV.<br />

Material & Methods<br />

The respective recombinant E rns proteins of 3 different genotypes of CSFV strains were expressed by<br />

baculovirus expression system. The E rns blocking ELISA was performed with the three recombinant E rns<br />

proteins mixtures as coating antigens and HRP conjugated-monoclonal antibody (Mab) against E rns protein of<br />

CSFV.<br />

Results<br />

Our data showed that the ELISA detected CSFV-specific antibodies as early as 14 days after infections of all<br />

genotypes (1, 2, and 3) of CSFVs, respectively. The sensitivity and specificity of the developed ELISA were<br />

validated using 266 CSF antibody positive reference sera and 1,871 CSF antibody positive field sera. The<br />

discriminatory E rns blocking ELISA scored a sensitivity of 93.2 % and a specificity of 94.2 %.<br />

Discussions & Conclusions<br />

Although this discriminatory ELISA kit was less sensitive than conventional E2 ELISA, this ELISA kit could be<br />

an effective method to detect antibodies of pigs infected with 3 genotypes CSFV after vaccination with E2<br />

marker vaccine, respectively.<br />

References:<br />

1. Floegel-Niesmann G. Classical swine fever (CSF) marker vaccine Trial �. Evaluation of discriminatory<br />

ELISAs. Vet Microbiol 2001, 83:121-136.<br />

2. Konig M, Lengsfeld T, Pauly T, Stark R and Thiel HJ. Classical swine fever virus: Independent induction of<br />

protective immunity by two structural glycoproteins. J Virol 1995, 69:6479-6486<br />

3. Moormann RJ, Bouma A, Kramps JA, Terpstra C and De Smit HJ. Development of a classical swine fever<br />

subunit marker vaccine and companion diagnostic test. Vet Microbiol 2000, 73:209-219.<br />

4. van Rijn PA, van Gennip HG and Moormann RJ. An experimental marker vaccine and accompanying<br />

serological diagnostic test both based on envelope glycoprotein E2 of classical swine fever virus (CSFV).<br />

Vaccine 1999, 17:433-440.<br />

�<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT AND EVALUATION OF ELISA FOR DETECTING ANTIBODIES AGAINST CAEV<br />

Introduction<br />

M. KONISHI 1, *, T. YAMAMOTO 1 , T. SHIMADA 1 , H. SHIRAFUJI 1 , H. SENTSUI 2 , K. MURAKAMI 1<br />

1 National Institute of Animal Health, 2 Nihon university<br />

Caprine arthritis-encephalitis (CAE) is a persistent virus infection caused by CAE virus (CAEV), which<br />

belongs to retrovirus. Infected animals shed the virus all their lives, but most of them are symptomless,<br />

therefore early detection of antibody for CAEV is very important to control this disease. In Japan, the first<br />

case of CAE was recognized in 2002. Since then, we have developed agar gel immunodiffusion test (AGID)<br />

using maedi virus, which is closely related to CAEV, as antigen for diagnosis of the disease. Although AGID<br />

is simple to perform, it requires experience for reading weak positive reactors. Furthermore, preparing<br />

antigen for the AGID is laborious, and it is difficult to keep uniform quality among different lots. In order to<br />

overcome the disadvantage of the AGID, two ELISAs were newly established. One used whole virus particle<br />

of CAEV (wELISA), and the other used recombinant core protein precursor, p55 (rELISA) as coating antigen.<br />

Sera of 745 goats in Japan were tested by the three assays. To evaluate the performance of the ELISAs,<br />

sensitivities and specificities were calculated using the result of AGID as a reference.<br />

Material & methods<br />

Preparation of antigen:<br />

1) AGID: Maedi virus was propagated in fetal lamb lung (FLL) cells. The virus was recovered from the<br />

supernatant of infected cells by ammonium sulfate precipitation followed by dialysis and dehydration. One<br />

percent of Triton-X 100 was added to the concentrated viral fluid.<br />

2) wELISA: CAEV was propagated in FLL cells, and was purified by density gradient centrifugation. Purified<br />

virus was resuspended in PBS, and was treated with NP-40.<br />

3) rELISA: P55, a precursor of core protein of CAEV, was expressed as His-Tag fusion protein. A gene<br />

encoding p55 was amplified from proviral DNA, which was extracted from infected FLL cells. The DNA<br />

fragment was cloned into an expression vector, pCold TF (TAKARA). The recombinant protein was<br />

recovered from the soluble fraction of E. coli and purified using a Ni-column.<br />

Sera: Sera of 745 goats were collected from 16 farms in 11 prefectures of Japan.<br />

Procedure of ELISA: Each ELISA was performed according to the conventional method of indirect ELISA.<br />

The optimal conditions of ELISAs were determined by checkerboard titration. Cut-off values were determined<br />

with the difference of OD values between positive and negative samples determined by AGID.<br />

Evaluation of ELISAs: Sensitivities and specificities were calculated using the result of AGID as a reference.<br />

The agreement between AGID and each ELISA was evaluated from the observed percentage of<br />

concordance and by Kappa statistic.<br />

Results<br />

AGID, wELISA and rELISA detected 38.7%, 45.4% and 48.9% as seropositives, respectively. Two hundred<br />

and thirty two sera were positive and 290 were negative by all the tests. AGID+/ELISAs- were 10, and AGID-<br />

/ELISAs+ were 24. Fifty-nine sera were positive only by wELISA, and rest of the 46 were negative only by<br />

one of the ELISAs. Sensitivities and specificities calculated for each ELISA were 0.88 and 0.82 for wELISA,<br />

and 0.89 and 0.76 for rELISA. Concordance percentages between AGID and ELISAs were 84.2% (wELISA)<br />

and 81.2% (rELISA). Kappa statistics of ELISAs were 0.68 (wELISA) and 0.62 (rELISA).<br />

Discussions & conclusions<br />

The relative sensitivity and specificity of each ELISA for AGID were almost similar. The observed percentage<br />

of concordance showed good agreement value between AGID and each ELISA. It is still necessary to verify<br />

the reference by some other serological tests like Western Blot, however, the results presented here indicate<br />

that these ELISAs can be used for initial screening test to detect anti-CAEV antibodies and may be suitable<br />

for analyzing large numbers of samples. Since ELISA is suitable for analyzing large numbers of samples, the<br />

ELISAs, especially rELISA, which the antigen is easily prepared, could be useful for CAE surveillance<br />

combined with AGID.<br />

References<br />

Konishi, M. et.al. 2004, An Epidemic of Caprine Arthritis Encephalitis in Japan: Isolation of the virus. J Vet<br />

Med Sci. Aug; 66 (8):911-7.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

A DIAGNOSTICALLY USEFUL PEPTIDE TO DETECT WEST NILE VIRUS INFECTION IN HORSES<br />

J.M. Hobson-Peters a,b,c , P.G. Toye b , M.D. Sanchez d , K.N. Bossart c,e , L.F. Wang c,e , D.C. Clark a ,<br />

W.Y. Cheah a , R.A. Hall a .<br />

a School of Molecular and Microbial Sciences, University of Queensland, St Lucia, Qld, Australia, 4072<br />

b AGEN Biomedical Ltd, Acacia Ridge, Qld, Australia, 4110<br />

c Australian Biosecurity CRC for Emerging Infectious Diseases, St Lucia, Qld, Australia, 4072.<br />

d Department of Microbiology, University of Pennsylvania, Philadelphia, PA U.S.A. 19104<br />

e CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Vic, Australia, 3220<br />

Introduction<br />

West Nile virus (WNV) is a mosquito-borne flavivirus responsible for recent outbreaks of fever and fatal<br />

encephalitis in humans and horses 1 . While highly virulent strains have been reported in Europe 2 and North<br />

America 3 , only a benign subtype of WNV, (Kunjin virus - KUNV), occurs in Australia. However, the presence<br />

of suitable vectors and reservoir hosts (birds) 4 , and the ability of WNV to spread between continents 5 , flag<br />

virulent WNV strains as a serious biosecurity threat to Australia. To compound this threat, the antigenic<br />

similarity between KUNV and exotic WNV strains will require highly specific serological assays to<br />

differentiate infections caused by the two viruses. The aim of this study was to identify diagnostically useful<br />

peptides in WNV proteins for incorporation into a rapid diagnostic assay for surveillance of exotic WNV<br />

strains in Australia.<br />

Materials and Methods<br />

6<br />

Panels of monoclonal antibodies were generated to the structural (prM/M and E) proteins of WNVNY99 and<br />

screened for their reaction to various WNV strains in ELISA. The conformation of the epitopes recognised by<br />

these antibodies was assessed by their reaction in Western blot after resolution of boiled, reduced and<br />

carboxymethylated viral antigens on SDS PAGE. The immunogenicity of the epitopes was also assessed by<br />

competition binding between Mabs and WNV-immune sera in a blocking ELISA. Site directed mutagenesis,<br />

amino acid sequence alignments, and synthetic peptides were then used to map the contact residues of<br />

immunogenic epitopes. Epitopes were then expressed as recombinant fusion proteins in mammalian cells<br />

and assessed for recognition by WNV- and KUNV-immune horse sera using Western blot.<br />

Results<br />

We identified a continuous (linear) epitope (WN19) that was highly immunogenic during equine WNV<br />

infection that mapped to a 19 amino acid sequence encompassing the WNV envelope (E) protein<br />

glycosylation site at position 154. Of six sera collected from horses experimentally infected with WNVNY99,<br />

four specifically bound peptide WN19. Four of six sera from horses with natural, subclinical infection with<br />

WNV also bound the peptide. Testing of four KUNV-immune horse sera revealed that three recognised<br />

peptide WN19. These three samples also had neutralizing antibodies to another Australian flavivirus called<br />

Murray Valley Encephalitis virus (MVEV). The sole sample that had KUNV-neutralizing antibodies, but not<br />

MVEV-neutralizing antibodies, did not react with the peptide. Failure of most (83%) of the immune sera to<br />

recognise the epitope as a synthetic peptide or as a degylcosylated fusion protein, also demonstrated that<br />

the N-linked glycan was critical for antibody recognition of the peptide.<br />

Conclusions<br />

Although peptide WN19 did not enable the clear differentiation between WNV and Australian flavivirus<br />

infections in horses, it shows potential as an antigen for incorporation into rapid WNV diagnostic assays.<br />

Further testing will be performed to determine whether the peptide is recognised by serum antibodies from<br />

other species infected with WNV, and can differentiate between WNV and KUNV infections.<br />

References<br />

1. van der Meulen, K.M., Pensaert, M.B., Nauwynck, H.J. (2005). Arch Virol 150, 637-57<br />

2. Tsai, T.F., Popovici, F., Cernescu, C., Campbell, G.L., Nedelcu, N.I. (1998). Lancett 352, 767-71.<br />

3. Hayes, E.B., and Gubler, D.J. (2006). Annu Rev Med. 57, 181-94<br />

4. Hall, R.A., Broom, A.K., Smith, D.W., Mackenzie, J.S. (2002). Curr Top Microbiol Immunol 267, 253-69.<br />

5. Lanciotti, R.S., Roehrig, J.T., Deubel, V., Smith, J., Parker, M., Steele, K., Crise, B., Volpe, K.E., Crabtree,<br />

M.B., Scherret, J.H., Hall, R.A., MacKenzie, J.S., Cropp, C.B., Panigrahy, B., Ostlund, E., Schmitt, B.,<br />

Malkinson, M., Banet, C., Weissman, J., Komar, N., Savage, H.M., Stone, W., McNamara, T., Gubler, D.J.<br />

(1999). Science 286, 2333-7.<br />

6. Sanchez, M.D., Pierson, T.C., McAllister, D., Hanna, S.L., Puffer, B.A., Valentine, L.E., Murtadha, M.M.,<br />

Hoxie, J.A., and Doms, R.W. (2005). Virology 336, 70-82<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

NOVEL ANTIGENS FOR THE DIAGNOSIS OF BOVINE TUBERCULOSIS<br />

V. Meikle, I. Durante, A. Cataldi, A. Alito<br />

Biotecnología, CICV y A INTA Castelar<br />

e-mail: vmeikle@cnia.inta.gov.ar.<br />

Aims<br />

The aim of the present work was to assess the cellular immune response against recombinant antigens of M.<br />

bovis in experimentally and naturally infected animals through the measurement of gamma interferon (γIFN)<br />

release.<br />

Methods<br />

Fourteen surgically-castrated Fresian calves of 6 months of age from a tuberculosis-free herd were used.<br />

Two animals used as negative controls.<br />

Eighty-five Fresian diary cows of two herds from Provincia de Buenos Aires with positive bovine PPD and 28<br />

diary cows with negative PPD were used.<br />

The selected recombinant proteins were: CFP10, ESAT6, Rv1636, Rv0138, Rv2524c, EsxV, Rv3740, HspX,<br />

TPX, Rv2624c, L7/L12, Rv3747, TrbB. γIFN release was measured and Specificity and Sensibility was<br />

assessed.<br />

Results<br />

All infected animals showed high levels of γIFN production when stimulated with DPP.<br />

Both CFP10 and ESAT6 showed high levels of γIFN throughout the infection (98 days). The stimulation rates<br />

were similar to those of DPP and in some cases even higher. No cross reaction with any M. avium protein for<br />

CFP10 was detected. TPX, Rv2624c and TrbB did not stimulate at the beginning of the infection; when they<br />

stimulated, the stimulation was not continuous but with high levels. TrbB stimulated from week 6 until the end<br />

of the assay. Rv2624c showed lower stimulation. It is induced by hypoxia as it is the case of HspX (Sherman<br />

D. et al., 2001). In naturally infected animals, EsxV could differentiate positive from negative animals. The<br />

combination of Rv1636/HspX and ESAT6/CFP10 reached 100% specificity and sensibility, and<br />

EsxV/Rv1636/Rv2524 showed 100% specificity and 67% sensibility. TPX, TrbB, Rv2624c, Rv3747 and<br />

L7/L12 reached specificities between 67 and 100% and sensibilities between 76 and 85%.<br />

Discusion<br />

The response of ESAT6 and CFP10 could show a persistence response throughout time, so it would be<br />

advisable to use these antigens for the diagnosis of TB. TPX, Rv2624c and TrbB seem to be involved at<br />

more advanced stages of infection.<br />

The combination of different recombinant antigens increased the sensibility of the assay, which proves that<br />

the combination of diverse epitopes influence the stimulation of more T-clones.<br />

Taking into account these results, we think it necessary for the diagnosis of the bovine tuberculosis through<br />

an γIFN test to evaluate different combinations of antigens, combining antigens that sense diverse stages of<br />

the infection.<br />

Bibliography<br />

-Young DB (1990) Stress proteins and the immune response. Antonie Van Leeuwenhoek. 58(3):203-8.<br />

Sherman DR, Voskuil M, Schnappinger D, Liao R, Harrel.<br />

-MI, Schoolnik GK. (2001) Regulation of the Mycobacterium tuberculosis hypoxic response gene encoding<br />

alpha-cristallin. Proc Natl Acad Sci USA. 2001 Jun 19;98(13):7534-9.<br />

-Daniel J, Deb C, Dubey VS, Sirakova T, Abomoelak B, Morbidoni H. Induction of a novel class of<br />

diacylglycerol acyltranferases and triacyl glycerol accumulation in Mycobacterium tuberculosis as it goes into<br />

a dormancy-like state in culture. J Bacteriol. 2004 Aug;1986(15):5017-30.<br />

-Lin MY Lack of immune responses to Mycobacterium tuberculosis DosR regulon proteins following<br />

Mycobacterium bovis BCG vaccination. Infect Immun. 2007 Jul;75(7):3523-30.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PRELIMINARY VALIDATION OF A NEW COMMERCIAL ELISA KIT FOR THE DETECTION OF<br />

ANTIBODIES DIRECTED AGAINST CHLAMYDIA ABORTUS<br />

P. Pourquier 1 , S. Lesceu 1 , A. Rodolakis 2 , K Yousef Mohamad 2<br />

1 ID VET, Montpellier France<br />

2 National Agronomie Research Institute (INRA), Nouzilly, France<br />

Introduction and Objectives:<br />

Serological diagnosis of Chlamydia abortus may be acheived by the complement fixation test (CFT) or by<br />

ELISA. These tests, which use LPS or whole bacteria as antigens, generally present low specificity and<br />

sensitivity levels, and cross reactions are often observed with Chlamydia pecorum.<br />

The aim of this study was to perform a preliminary validation of a commercial peptide-based ELISA<br />

developed by ID VET (www.id-vet.com) for the detection of anti-Chlamydia abortus antibodies in ovine and<br />

caprine sera.<br />

Test principle:<br />

The test is an indirect ELISA. The antigen is a synthetic peptide from a major outer-membrane protein<br />

specific to Chlamydia abortus. The conjugate is an anti-ruminant peroxidase.<br />

Methods:<br />

Serum samples from sheep herds with and without a history of abortions (n=10 and 26 respectively) were<br />

tested for C percorum by microimmunofluorescence, and for C. abortus by the ID Screen® ELISA.<br />

Sheep herds, naturally or experimentally-infected with C. pecorum (n=17 and 20 respectively), were also<br />

tested using the two techniques.<br />

Sensitivity was tested through the analysis of 8, C. abortus-infected animals by CFT and by the ID Screen®<br />

ELISA (n=8).<br />

Results:<br />

With the ID Screen® ELISA:<br />

- None of the animals from the abortion-free herd were found positive.<br />

- None of the 17 naturally-infected C. pecorum animals were found positive (10 of these animals were<br />

found doubtful when tested for C. pecorum, and 7 were found positive).<br />

- Only 3 out of 20 animals, experimentally-infected by C. pecorum, were found positive.<br />

- All sera with a CFT titre superior or equal to 1/20 were found positive.<br />

- An excellent correlation was obtained between CFT titres and S/P values.<br />

Conclusion:<br />

Through the use of a synthetic peptide antigen, the ID Screen® Chlamydia abortus Indirect ELISA reduces<br />

the number of cross-reactions with C. percorum while maintaining high sensitivity.<br />

It is an excellent tool for the specific detection of anti-C. abortus antibodies in ruminant sera.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PRELIMINARY VALIDATION OF A NEW COMMERCIAL DIAGNOSTIC TEST FOR THE DETECTION OF<br />

ANTI-EQUINE INFECTIOUS ANEMIA VIRUS (EIAV) ANTIBODIES IN HORSE SERA<br />

P. Pourquier 1 , L. Comtet 1 , S. Rosati 2<br />

1 ID VET, Montpellier, France<br />

2 Facoltà di Medicina Veterinaria,Torino, Italy<br />

Introduction and Objectives:<br />

Equine infectious anemia (EIA) is an infectious and potentially fatal viral disease caused by the equine<br />

infectious anemia virus (EIAV). Serology is often used to control and monitor this disease.<br />

The aim of this study was to perform a preliminary validation of a commercial ELISA developed by ID VET<br />

(www.id-vet.com), ID Screen ® Equine Infectious Anemia Indirect, for the detection of anti-EIAV antibodies in<br />

horse sera.<br />

Test principle:<br />

The test is an indirect ELISA using a p26/Tm recombinant antigen (1) and an anti-horse-peroxidase conjugate.<br />

Wells are coated with GAG and Transmembrane TM recombinant antigens.<br />

Methods and Results:<br />

Specificity was evaluated by testing 594 disease-free horse populations from France and Italy by AGID (Agar<br />

Gel ImmunoDiffusion) and by the ID Screen ® ELISA. Both techniques gave negative results for all samples,<br />

giving a measured specificity of the ID Screen ® test of 100% (IC95% 99.8 - 99.99 %).<br />

Sensitivity was evaluated by testing 24 AGID positive sera from France, Italy, the Czech Republic and the<br />

USA. All sera were found positive with the ID Screen ® test, giving a measured sensitivity of 100%.<br />

Detectability was evaluated by testing 10 repetitions of the OIE international standard (2) . All were<br />

found positive.<br />

Conclusion:<br />

The ID Screen ® ELISA is an effective, reliable and rapid tool for the specific detection of anti-EIAV antibodies<br />

in horse sera.<br />

References:<br />

(1) ROSATI S., et al.2004. Development of recombinant capsid antigen / transmembrane epitope fusion<br />

proteins for serological diagnosis of animal lentivirus infections. Journ. Vir. Methods, 121, 73-78.<br />

(2) OIE International Reference Equine Infectious Anemia (Ecole Nationale Vétérinaire, 7 avenue du Général<br />

de Gaulle, 94704 Maisons-Alfort Cedex, France).<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

APPLICATION OF IMMUNOMAGNETIC BEAD TECHNOLOGY FOR IMPROVED DIAGNOSIS OF<br />

CLASSICAL SWINE FEVER IN A LOW TECHNOLOGY SETTING<br />

J.V. Conlan* 1 , A. Colling 2 , C.J. Morrissy 2 , L.J. Gleeson 2 , C.R. Wilks 3 and S. Khounsy 1<br />

1 National Animal Health Centre, Department of Livestock and Fisheries, Vientiane Capital, Laos; 2 CSIRO Livestock Industries,<br />

Australian Animal Health Laboratory, Geelong, VIC Australia; 3 Faculty of Veterinary Science, University of Melbourne, Parkville, VIC<br />

Australia<br />

Introduction<br />

Classical Swine Fever (CSF) is a highly contagious viral disease that causes major losses to pig production<br />

in many parts of the world. CSF virus is a member of the genus Pestivirus of the family Flaviviridae and is<br />

closely related antigenically to other Pestiviruses; Bovine Viral Diarrhoea (BVD) virus and Border Disease<br />

(BD) virus. Laboratory testing is required to make a definitive diagnosis of CSF due of the difficulty of making<br />

a diagnosis based solely on clinical signs. Immunomagnetic bead (IMB) technology offers an alternative solid<br />

phase for the reliable and rapid detection of analyte in a diagnostic specimen. The research presented<br />

outlines the development and assessment of two tests using this technology: an enzyme immunoassay for<br />

the near-to-field detection of classical swine fever (CSF) virus antigen and a blocking ELISA for the detection<br />

of antibodies to CSF.<br />

Materials & methods<br />

Anti-pestivirus polyclonal capture antibody was covalently bound to the carboxyl modified surface of<br />

immunomagnetic beads (IMBs) (Spherotech, USA) to capture solubilised antigen from tissue samples.<br />

Monoclonal antibody (Mab) specific for the E2 protein of CSF virus was used to detect captured antigen and<br />

visualisation was achieved by adding horse-radish-peroxidase conjugate followed by TMB substrate (Sigma<br />

Aldrich, USA). The immunoassay for antigen detection was developed in a dropper bottle kit format for use<br />

near-to-field. The antigen capture immunoassay was further developed into a laboratory based blocking<br />

ELISA for the detection of antibodies using a Lao CSF virus strain as the standard antigen.<br />

Results<br />

The near-to-field antigen capture test was shown to have performance characteristics similar to an antigen<br />

capture ELISA, 100% diagnostic sensitivity and greater than 90% diagnostic specificity. The advantage that<br />

this test has over other more conventional tests is the simplicity, speed and cost. No sophisticated laboratory<br />

equipment is required and a result can be read by eye with a high level of inter-operator agreement. The<br />

blocking ELISA for the detection of antibodies to CSF virus is also rapid to perform and has excellent<br />

performance characteristics in comparison to a commercial ELISA (Cedi Diagnostics, The Netherlands) and<br />

the “gold standard” neutralising peroxidase linked assay (NPLA).<br />

Conclusions<br />

In Laos, where these tests were developed, CSF is endemic and outbreaks have adverse effects on the<br />

predominantly smallholder farming sector. Immunomagnetic bead technology is adaptable and versatile and<br />

can provide a platform for appropriate diagnostic test development in a low technology setting. The near-tofield<br />

test is inexpensive, portable and a reliable test that requires minimal training to implement and will<br />

improve diagnostic services for pig farmers. The blocking ELISA for antibody detection shows strong<br />

agreement with the ‘gold-standard’ NPLA and could be a valuable tool for monitoring vaccine uptake.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

LABORATORY EVALUATION OF THE APPROPRIATE VACCINATION PROGRAMS AGAINST<br />

NEWCASTLE DISEASE IN BROILER CHICKENS UNDER FIELD CONDITIONS IN MOSUL PROVINCE<br />

(IRAQ).<br />

A. Abdul -A had Shamaun * O.S. Beyon * A.M.SHAREEF**<br />

*Department of Pathology, College of Veterinary Medicine, Mosul, Iraq<br />

** Department of Veterinary Public Health, College of Veterinary Medicine, Mosul, Iraq<br />

Four experiments were conducted to examine the efficiency of 4 different Newcastle disease (ND) field<br />

vaccination programs. For all experiments young broiler chickens were obtained from breeder flocks<br />

regularly vaccinated against ND.<br />

The following vaccination programs were applied to 128000 broilers (four broiler flocks programs ,with of<br />

about 8000 broilers flock):<br />

1-Vaccination at 1 day of age with lentogenic Hitchner B1/spray with oil emulsion vaccine /SC; 7,14,25,35<br />

and 45 day old with Lasota/drinking water; 2- Vaccination at 1 day of age with oil emulsion vaccine /SC;<br />

7,14,25,35 and 45 day old with Lasota/drinking water; 3- Vaccination at 1 day of age with hitchner B1/spray<br />

7,14,25,35 and 45 day old with Lasota/drinking water;4- No vaccination at 1 day of age ; 7,14,25,35 and 45<br />

day old with Lasota/drinking water.<br />

In all experiments, 20 randomly chosen birds from each of 16 broiler flocks were bled just prior to vaccination<br />

and tested for Newcastle disease antibody by hemagglutination inhibition test(HI).Serological data indicated<br />

that the greatest response was found by appling the first program, when 1 day old chicks were vaccinated<br />

with combined Hitchner B1/spray and oil emulsion<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DEVELOPMENT OF THE RESAZURIN MICROTITER ASSAY<br />

FOR ASSESSMENT OF ANTI-SURRA DRUG<br />

Raadan Odbileg<br />

Animal trypanosomiasis (Surra) caused by Trypanosoma evansi is endemic through out Asia, Middle East,<br />

Africa and South America. Currently, Surra is on the list of notifiable diseases of OIE. With scant economic<br />

resources available for antiparasitic drug discovery and development, inexpensive, and high-throughput<br />

assays to screen potential new drugs are essential. A quantitative colorimetric assay using the resazurin<br />

was developed to measure the cytotoxicity of compounds against T. evansi. To validate the assay, the<br />

experimental conditions were adjusted, such as number of parasites, dye concentration, and time of<br />

incubation, with respect to linearity and lower limit of detection. We found that absorbances increased<br />

linearly, with the plating density of parasites as low as 2x10 5 /ml ~ 9x10 6 /ml per well when they were<br />

incubated for 5 to 48h at 37°C in the presence of 10% resazurin solution (2 mM). When the cytotoxicity of<br />

the reference drugs Samorin, Antrycide and Suramin were measured with this assay and compared to the<br />

microscopic counting method, the same range is obtained, demonstrating that the resazurin microtiter assay<br />

is valid for the screening of new trypanocidal compounds. Furthermore the emergence of resistance against<br />

major drug for the control of trypanosomiasis is now serious and increasing problem. The potential of this<br />

assay in detecting drug resistant trypanosome to some trypanocidal drug is also promising by subjecting<br />

these parasites to series of drug dilutions. Therefore, to evaluate a potential anti-trypanosomal drug<br />

candidate, resazurin microtiter assay was found reliable, simple, fast, sensitive and cheap, and could be<br />

easily automated for high-throughput screening.<br />

Poster Presentations


Poster Presentations<br />

Aims<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

WILD BIRDS – NATURAL RESEVOIR OF NEWCASTLE DISEASE VIRUS, IN ROMANIA<br />

Graziela NEAGOE¹, Iuliana ONIŢĂ¹, Adriana NEICUŢ¹, OLARU E. ¹,<br />

Aurelia IONESCU¹, DIACONU Cl. ¹ DONESCU D. ¹, Gina DRAGOMIR¹<br />

1 Institute for Diagnosis and Animal Health<br />

In Romania, during 2005 –2006, within the framework of Reference National Laboratory for Avian Influenza<br />

and Newcastle Disease from Institute for Diagnosis and Animal Health were tested samples collected from<br />

449 wild birds from 34 different species.<br />

The purpose of this workpaper is to distinguish the role of wild birds like important factor for spreading of<br />

Newcastle disease virus.<br />

Methods<br />

Samples were represented by tracheal and cloacal swabs, tissues. Brain and intestine samples were<br />

processed separately from other organs.<br />

Have been used virus isolation on allantoic cavity of 9-11- day – old embryonating fowl eggs and virus<br />

identification by haemagglutination inhibition test with specific antiserum from OIE and National Reference<br />

Laboratory for Avian Influenza and Newcastle disease, Istituto Zooprofilattico delle Venezie, Italy. The<br />

assessment of virus virulence was done by intracerebral pathogenicity index (ICPI) using 0.05 ml of diluted<br />

virus injected intracerebrally into each of ten chicks hatched from eggs an SPF flock.<br />

Results<br />

From 449 wild birds, 338 were negative, 37 positive for avian influenza virus (1 wild duck, 21 swan, 2 pigeon,<br />

1 Fulica atra, 2 crow, 7 wild goose, 1 dabchick, 1 heron) and 14 for Newcastle disease virus (2 swan, 6<br />

pigeon, 1 Fulica atra, 1 seagull, 1 dabchick, 1 stork, 2 partridge)<br />

ICPI values are comprised between 1.60 and 1.86 and belong to velogenic pathotype. The high values have<br />

been observed at partridge – 1.86, swan – 1.77, Fulica atra and pigeon – 1.75. There are not ICPI values<br />

less 1.6 in samples collected from wild birds.<br />

Those 14 wild birds positive at Newcastle disease virus are originated from 11 villages of 7 counties (Arges,<br />

Bucureşti, Constanta, Ilfov, Mehedinţi, Tulcea, Vrancea).<br />

Conclusions<br />

ICPI high values recorded at those 14 wild birds which are diagnosed with Newcastle disease framed the<br />

strains in velogenic pathotype.<br />

ICPI demonstrated the pathogenicity of isolates which circulated in Romania in wild birds, correlated with<br />

Newcastle disease outbreaks in fowls, in same time and area.<br />

The isolation and identification of Newcastle disease virus in wild birds from Romania suggested that these<br />

represent a natural reservoir of Newcastle disease virus.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AUSTRALIAN STRAINS OF BOVINE HERPESVIRUS 1 ARE GENETICALLY HOMOGENOUS.<br />

* X.Gu 1 P.D. Kirkland 1 , R.J. Davis 1 , C.L.Hornitzky 1 .<br />

1 Virology Laboratory, Elizabeth Macarthur Agricultural Institute, Camden, NSW, Australia<br />

Introduction<br />

Viruses belonging the bovine herpesvirus 1 (BHV-1) group cause respiratory and reproductive disease. In<br />

Australia neither severe respiratory disease nor abortion has been observed. It has been claimed that the<br />

restricted and less severe clinical manifestations observed in Australian cattle are due to an absence of the<br />

BHV1.1 and 1.2a subgroups 1 . The purpose of this study was to examine the characteristics of a large<br />

collection of Australian BHV-1 isolates.<br />

Material & methods<br />

Bovine herpesviruses from a diversity of geographical locations in Australia and isolated over a 30 period<br />

from different clinical presentations were examined. Genetic analyses employed digestion of semi-purified<br />

viral DNA by restriction enzymes (RE) followed by agarose gel electrophoresis and antigenic characterisation<br />

involved immunoperoxidase (IPX) staining with monoclonal antibodies that were either pan-reactive or<br />

specific to a sub-group 1,2 . RE profiles were developed for 121 Australian bovine strains and 9 overseas<br />

reference strains - 5 of BHV-1.1, 2 of BHV-1.2a and 2 of BHV-1.2b. One buffalo isolate (BHV-2), 2 BHV-5<br />

strains, 1 caprine isolate (GHV) and 1 equine isolate (EHV-1) were included for comparison. Antigenic<br />

analyses were conducted on 164 isolates by IPX staining of antigens in virus-infected, formalin fixed cell<br />

culture monolayers with a panel of 3 IBR specific monoclonal antibodies 2,3 .<br />

Results<br />

None of the Australian viruses examined had RE profiles that were indicative of the BHV-1.1 subgroup. The<br />

RE profiles conclusively showed that all of the Australian isolates belong to the BHV-1.2b subgroup (IPVlike)<br />

and were also readily distinguishable from viruses in the BHV1-2a subgroup. All of the viruses reacted<br />

in the IPX assays with the BHV-1 group-reactive monoclonal antibodies. Although some of the monoclonal<br />

antibodies were claimed to be subtype specific, a presumptively BHV-1.1 reactive mAb sometimes reacted<br />

with Australian viruses from the BHV-1.2b subgroup. It is of some interest that these viruses came from<br />

clusters in 3 discrete geographical areas.<br />

Discussion & conclusions<br />

Although Australian viruses were examined over a 30 year time period and from a range of clinical<br />

presentations, no isolates of BHV1.1 were identified. The results support the claim that all Australian strains<br />

of BHV1 belong to the BHV1.2b subgroup. The clinical presentations observed in Australia, notably the<br />

absence of abortigenic strains and those associated overseas with severe outbreaks of respiratory disease,<br />

are consistent with these findings. While a previous study 4 had suggested that variant strains of BVH-1 had<br />

recently been found in Australia, indistinguishable strains, isolated almost 20 years earlier, were identified in<br />

the present study.<br />

References<br />

1. Brake F, Studdert MJ (1985): Molecular epidemiology and pathogenesis of ruminant herpesviruses<br />

including bovine, buffalo and caprine herpesviruses l and bovine encephalitis herpesvirus. Aust Vet J,<br />

62:331-334.<br />

2. Metzler AE, Matile H, Gassmann U, Engels M, Wyle R (1985): European isolates of bovine herpesvirus 1:<br />

a comparison of restriction endonuclease sites, polypeptides, and reactivity with monoclonal antibodies. Arch<br />

Virol 85: 57-69<br />

3. Rijsewijk FA, Kaashoek MJ, Langeveld JP, Meloen R, Judek J, Bienkowska-Szewczyk K, Maris-Veldhuis<br />

MA, van Oirschot JT (1999): Epitopes on glycoprotein C of bovine herpesvirus-1 (BHV-1) that allow<br />

differentiation between BHV-1.1 and BHV-1.2 strains. J Gen Virol, 80:1477-1483.<br />

4. Smith GA, Young PL, Reed KC: Emergence of a new bovine herpesvirus 1 strain in Australian feedlots.<br />

Arch Virol 1995, 140:599-603.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INTERFERENCE IN DISEASE DIAGNOSIS<br />

IN PIGS BY PESTIVIRUS CROSS-REACTIONS<br />

R. R. Clough*, A. McFadden, J. Wang, K. Tham, W. Garrod, T. Haydon, D. Orr,<br />

R. Kittelberger, M. Hannah, K. Garnett, J. Jenner, W. Stanislawek,<br />

S. Cork, P. Bingham, C. Pigott<br />

Investigation and Diagnostic Centre-Wallaceville, MAF Biosecurity New Zealand, Upper Hutt, New Zealand<br />

*Presenting author<br />

Heterologous pestiviruses can cross-react in various serological assays 1,2 and therefore may interfere with<br />

determination of exotic disease status. By using a case study in pigs, we aimed to show that antibodies to<br />

pestiviruses can cross-react in CSF ELISAs that are claimed by manufacturers to be highly specific.<br />

Methods used in this investigation included three commercial classical swine fever (CSF) virus enzymelinked<br />

immunosorbent assays (ELISA) (IDEXX Herdcheck CSFV blocking, Pourquier CSF-Ab blocking, Cedi<br />

Ceditest CSFV 2.0 blocking) and a porcine reproductive and respiratory syndrome (PRRS) virus antibody<br />

detecting ELISA (IDEXX HerdChek PRRS 2XR), plus in-house bovine virus diarrhoea (BVD), CSF and<br />

Border disease/hairy shaker disease (BD/HSD) virus neutralization tests (VNT), CSF, BVD and porcine<br />

circovirus type 2 (PCV-2) polymerase chain reactions (PCR), necropsy, virus isolation and histopathology<br />

(subcontracted to the Institute of Veterinary, Animal and Biomedical Sciences, Massey University, NZ).<br />

During an exotic disease investigation with differentials of CSF and PRRS, serological tests indicated in 2 of<br />

19 pigs the presence of antibodies to CSF virus by a commercial competitive ELISA, and to BVD and CSF<br />

viruses by VNTs. Virus isolation from tissues of three affected pigs resulted in only PCV-2 being isolated<br />

(confirmed by PCV-2 PCR), and histology demonstrated results most consistent with post-weaning<br />

multisystemic wasting syndrome (PMWS). To Investigate the CSF serology results an additional 109 pigs<br />

were sampled, which was sufficient to estimate a prevalence


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AN UNUSUAL CASE OF INFECTIOUS LARYNGOTRACHEITIS VIRUS DETECTION<br />

IN A BACKYARD POULTRY FLOCK<br />

B.G. Corney*, S.M. Ossedryver, I.S. Diallo, G.R. Hewitson, A.J. De Jong, M.X. Tolosa, M.A. Kelly, B.J. Rodwell.<br />

Biosecurity Sciences Laboratory, Department of Primary Industries and Fisheries, Yeerongpilly, Queensland 4105, Australia.<br />

Introduction<br />

Infectious laryngotracheitis is a notifiable (in Queensland) respiratory disease of poultry. It is caused by an<br />

Alphaherpesvirus, Gallid herpesvirus 1 (Guy & Bagust, 2003; Roizman, 1982) commonly known as infectious<br />

laryngotracheitis virus (ILTV) (Roizman, 1982). Current dogma is that ILTV replication occurs exclusively in<br />

the respiratory tract and conjunctiva (Guy & Bagust, 2003). No viremic phase has been documented, and<br />

attempts at virus detection generally utilise samples collected from the respiratory tract and conjunctiva (Guy<br />

& Bagust, 2003). We report here an unusual case where ILTV was detected in cloacal swabs, but not<br />

trachea, collected from a hen with respiratory disease.<br />

Material & methods<br />

A sick layer hen from a backyard flock presented with poor body condition, head swelling, congestion of the<br />

comb, severe bilateral conjunctivitis, swelling over the right orbital fossa and greenish watery diarrhoea. The<br />

bird was necropsied and tissues and swabs were collected for histology, bacteriology, virus isolation and<br />

molecular tests for a range of bacterial and viral pathogens.<br />

Results<br />

Histology revealed moderate congestion and subcutaneous oedema of the comb and congestion of the<br />

kidney. Mycoplasma pullorum was isolated from a tracheal swab. Avibacterium (Pasteurella) gallinarum was<br />

isolated from several sites. Av. (Haemophilus) paragallinarum was also detected in several sites by real-time<br />

PCR. ILTV was detected in a cloacal swab but not in fresh trachea by real-time PCR. ILTV was also isolated<br />

from the cloacal swab only. The isolates were identified as herpesvirus by electron microscopy and ILTV by<br />

real-time PCR. The bird was diagnosed as having clinical conjunctivitis involving Av. paragallinarum, Av.<br />

gallinarum and ILTV.<br />

Discussions & conclusions<br />

The occurrence of ILTV in cloacal and faecal samples is not an entirely new finding (Creelan et al., 2006). In<br />

the case reported herein, ILTV was detected in cloacal but not tracheal samples. The presence of ILTV was<br />

initially demonstrated by real-time PCR and confirmed by virus isolation and sequencing.<br />

Kirkpatrick et al. (2006) describes ILTV genotypes exhibiting considerable variation in their tropism for<br />

trachea or conjunctival tissue. The bird examined in this case probably had a strain of ILTV with preferential<br />

tropism for the conjunctiva, causing clinical conjunctivitis in association with or complicated by the other<br />

bacterial organisms identified here, without overt tracheal pathology. Av. paragallinarum was detected by<br />

real-time PCR but not by culture probably due to overgrowth of the culture plates by other bacteria. The<br />

presence of Av. paragallinarum was confirmed by sequencing.<br />

Detection of ILTV in nonrespiratory sites is a rarity. This may be a reflection of current sampling strategies for<br />

ILTV detection (Guy & Bagust, 2003) rather than the actual occurrence of ILTV at these sites. Reassessment<br />

of current sampling strategies for ILTV may be warranted based on these findings, especially as birds<br />

shedding ILTV in faeces could act as a source of infection for other birds and possibly as a reservoir of<br />

infection in otherwise healthy flocks.<br />

References<br />

Creelan, J.L., Calvert, V.M., Graham, D.A. and McCullough, S.J. (2006). Avian Pathology 35, 173-179.<br />

Guy, J.S. and Bagust, T.J. (2003). Laryngotracheitis. In: Y.M. Saif, H.J. Barnes, J.R. Glisson, A.M. Fadly,<br />

L.R. McDougald and D.E. Swayne (Eds), Diseases of Poultry, 11 ed, Iowa State University Press, Ames,<br />

Iowa, USA, pp. 121-134.<br />

Kirkpatrick, N.C., Mahmoudian, A., Colson, C.A., Devlin, J.M. and Noormohammadi, A.M. (2006). Avian<br />

Pathology 35, 449-453.<br />

Roizman, B. (1982). The family Herpesviridae: general description, taxonomy, and classification. In: B.<br />

Roizmann (Ed), The Herpesviruses, Vol. 1, Plenum Press, New York, pp. 1-23.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CLINICO- PATHOLOGICAL STUDIES ON CLASSICAL SWINE FEVER VIRUS IN NATURALLY<br />

OCCURRING OUTBREAKS IN UNVACCINATED SWINE HERDS OF PUNJAB (INDIA).<br />

H. Kumar, V. Mahajan. S. Sharma and K.S. Sandhu.<br />

Department of Epidemiology and Preventive Veterinary Medicine, Guru Angad Dev Veterinary and Animal Sciences University,<br />

Ludhiana.<br />

Study was conducted to investigate and to confirm the outbreaks of classical swine fever and to formulate<br />

control strategies to check the spread of disease. Three outbreaks of classical swine fever (CSF) were<br />

investigated in different swine herds of Punjab (India). All herds were located in the district of Ludhiana but in<br />

different villages (Lopo, Haibowal and Changran) approximately at a distance of 20 km apart. A team of<br />

expert visited the places of outbreaks and conducted detailed investigations. About 2 ml blood samples were<br />

collected in heparinized vials for haematological examination and 0.5ml in nutrient broth for bacteriological<br />

isolation. Rectal temperature of infected and healthy animal was recorded in each herd. Dead animals were<br />

subjected to detailed post mortem examination. Tissues from kidney, lymph nodes, lungs, liver, intestine and<br />

brain were collected in 10% formalin for histopathology and in 50% glycerol saline (kidney, lymph nodes,<br />

intestine and tonsil) for virological studies. Confirmation of CSF virus in the tissues was made by RT-PCR.<br />

There were 125 pigs in all the three herds and 13 pigs died out of 32 pigs infected with classical swine fever<br />

virus. Over all morbidity, mortality and Case fatality rate observed were 25.6 % (32/125), 10.4 % (13/125)<br />

and 40.62% (13/32). Clinically, most of the affected animal were showing high fever; erythema of the skin of<br />

the ears, abdomen, medial thighs; and greenish watery diarrhea. Some of the animals were exhibiting<br />

staggering gait. Haematologically, all the infected animals were showing leucopenia and relative<br />

lymphocytosis. Postmortem lesions observed were intestinal ulcers (button ulcers); congestion, multifocal<br />

hemorrhages and infarcts of the spleen; enlarged, edematous, and hemorrhagic lymph nodes; and petechial<br />

hemorrhages on the kidneys. Sub capsular hemorrhages in kidneys and chronic necrotic enteritis were the<br />

significant histological lesions observed histopathologically. Perivascular cuffing and leptomeningitis was<br />

observed in brain. Lungs were showing haemorhages and bronco pneumonia. Samples subjected to RT-<br />

PCR revealed positive reaction confirming presence of classical swine fever virus.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

BOVINE LYMPHOTROPIC HERPESVIRUS (BLHV) IN THE UK<br />

*G Ibata, B Nash, J Peake, J P Frossard, F Steinbach and M Banks<br />

Veterinary Laboratories Agency, UK<br />

Introduction<br />

BLHV is a gamma herpesvirus, which had until recently only been described in the USA, and showed a<br />

tentative link with enzootic bovine leukosis (EBL) (3). It was first detected in the UK in December 2005 in<br />

dairy cattle presenting non-responsive post-partum metritis (NPPM), indicating the possible involvement of<br />

BLHV (2). A small scale investigation on 13 farms (2) provided evidence to support this hypothesis.<br />

Bovine leukaemia virus (BLV) is the causative agent of EBL and it has been suggested that BLHV may be a<br />

co-factor in the pathogenesis of EBL (3). To investigate this, BLHV positive samples were screened to<br />

ensure the samples were negative for BLV.<br />

Tissues from UK slaughterhouse carcasses from enlarged, caseous or tumorous lymph nodes are regularly<br />

received at the VLA for BLV detection from suspect EBL cases. These cattle had neither BLV involvement,<br />

nor any other indication of disease and were therefore considered to be a good source of material for BLHV<br />

screening.<br />

Also available for BLHV testing was a series of 158 EDTA bloods from a non UK herd that were BLV<br />

positive.<br />

Material & methods<br />

A nested PCR was employed using pan-herpesvirus primers DFA, ILK, TGV, IYG and KG1 (4) on DNA<br />

extracted from samples of vaginal mucus and swabs from NPPM cattle and from the slaughterhouse<br />

samples and foreign cattle samples indicated above. All BLHV positive samples were tested for BLV proviral<br />

DNA by PCR. Selected positive BLV and pan-herpes PCR amplicons were sequenced and assessed<br />

phylogenetically. The remaining pan-herpes positives were confirmed as BLHV by HindIII digestion; noncutting<br />

amplicons were sequenced as above.<br />

Results<br />

In 9/13 herds suffering from NPPM at least one animal tested positive for BLHV and one BLHV negative<br />

animal from a BLHV positive herd, tested positive for BoHV4.<br />

27% of the vaginal exudates and swabs were positive for BLHV. There was close DNA sequence (474 nt.)<br />

homology between the UK isolates (96.6-100%) and to the USA isolates (97.7-100%).<br />

All 95 UK EBL submissions were negative by BLV PCR, 53 submissions were positive to BLHV (56%) and<br />

all were confirmed as BLHV by sequence analysis or HindIII digestion.<br />

EBL submissions were received from17/25 Animal Health Divisional Offices. BLHV virus was detected in<br />

samples from 13 of these regions. BLHV positives submissions were from animals aged 2 to 16 years.<br />

All 60 UK BLHV positive cases were negative for BLV.<br />

All 158 non UK EBL submissions were positive for BLV; 81 (51%) of these were positive for BLHV.<br />

Discussions & conclusions<br />

• BLHV virus is present in cattle of all ages from across the UK and was detected in samples from<br />

abroad.<br />

• BLHV can be detected in blood and lymph nodes, and in vaginal exudates from cases of NPPM.<br />

• BLHV has been detected in BLV-positive tissues.<br />

• The UK BLHV prevalence cannot be determined from this study, but was high in the samples<br />

examined.<br />

• BLHV infection does not appear to predispose to the progression to BLV-induced lymphosarcoma as<br />

suggested earlier (3), but could play a role in non-BLV lymphomas.<br />

• To investigate a possible association of BLHV with lymphoid tumours, lymphoid tissue from tumour<br />

negative animals will be screened.<br />

References<br />

1. M. Banks, G. Ibata, A.M. Murphy, J.P. Frossard, T.R. Crawshaw and D.F. Twomey, (2007) The<br />

Veterinary Journal, in press.<br />

2. S.P. Cobb, M. Banks, C. Russell, and M. Thorne, Veterinary Record 158 (2006), pp807-808.<br />

3. J. Rovnak, S.L Quackenbush, R.A. Reyes, et al. (1998) Journal of Virology 72 pp. 4237-4242.<br />

4. D.R. Van Devanter, P. Warrener, L. Bennett, et al. (1996) Journal of Clinical Microbiology. 34 pp.1666-<br />

1671<br />

Introduction<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

REPRODUCTION OF PMWS OF HIGH MORTALITY WITH A PORCINE CIRCOVIRUS<br />

TYPE 2-GROUP 1 ISOLATE<br />

A. Cheung 1*<br />

, K. Lager 1<br />

, P. Gauger 1<br />

, A. Vincent 1<br />

, T. Opriessnig 2<br />

, M. Kehrli, Jr. 1<br />

1<br />

National Animal Disease Center, Ames, Iowa, USA, 50010;<br />

2<br />

Iowa State University, Ames, Iowa, USA, 50011<br />

In late 2005, sporadic cases of an acute onset of high mortality disease were observed in growing pigs<br />

among USA swine herds. PCV2-group 1 (Gp1) virus was consistently detected among the affected animals.<br />

Phylogenetic analysis showed that the PCV2 isolates from the United States until late 2005 belonged to<br />

PCV2-group 2 (Gp2) (1,2). The objective of this study was to examine the pathogenic capability of Gp1 and<br />

Gp2 viruses in germ-free pigs.<br />

Material & methods<br />

Virus. Infectious viruses were generated from bacterial plasmids containing Gp1 and Gp2 nucleotide<br />

sequences. The viral genomes were excised from the bacterial plasmids after delivered into PK15 cells via<br />

lipofectamine transfection. Infectious viruses were recovered from the transfection cell cultures after 1 week.<br />

Experimental design. Pigs were derived under sterile conditions and maintained germ-free in isolators. The<br />

pigs were fed a sterile milk diet and at 12 days of age (day 0 of the experiment) were given an intranasal<br />

inoculation of either Gp1 virus, Gp2 virus or cell culture medium. The pigs were monitored for clinical signs<br />

with a plan of euthanizing all pigs 41 days-post-inoculation.<br />

Results<br />

Mortality of the infected pigs in 2 independent experiments with Gp1 was 50% (6/12 pigs) and 100% (4/4<br />

pigs), respectively. In one experiment, Gp1 (4/4 pigs) was found to be more virulent than Gp2 (1/4 pigs). The<br />

most significant lesions observed in the diseased pigs were depletion and replacement of lymphoid follicles<br />

by macrophages, and severe vacuolar degeneration and necrosis of hepatocytes with multifocal areas of<br />

hemorrhagex. Abundant PCV2 antigen was detected in tissues of affected pigs. In our analysis, no other<br />

microorganisms were detected in the animals throughout the experiment. Since the viruses used in this<br />

study were derived from cloned DNA, we presume the clinical effects were attributed to the respective virus<br />

isolate.<br />

Discussions & conclusions<br />

Here, we report reproduction of PMWS of high mortality in germ-free pigs by a Gp1 virus alone and without<br />

immune stimulation or co-infection with another infectious agent (3,4). In this study, the Gp1 isolate was<br />

more pathogenic when compared to the Gp2 isolate identified in the same 2005 United States outbreak. In<br />

our analysis, no other microorganisms were detected in the affected pigs.<br />

References<br />

1. Olvera A, Cortey M, Segales J. (2007) Molecular evolution of porcine circovirus type 2 genomes:<br />

phylogeny and clonality. Virology 357: 175-185.<br />

2. Cheung A, Lager K, Kohutyuk O, Vincent A, Henry S, Baker R, Rowland R, and Dunham A. (2007)<br />

Detection of two porcine circovirus type 2 genotypic groups in United States swine herds. Arch Virol<br />

152:1035-1044<br />

3. Ellis J, Krakowka S, Lairmore M, Haines D, Bratanich A, Clark E, Allan G, Konoby C, Hassard L, Meehan<br />

B, Martin K, Harding J, Kennedy S, McNeilly F. (1999) Reproduction of lesions of postweaning multisystemic<br />

wasting syndrome in gnotobiotic piglets. J Vet Diagn Invest 11:3-14.<br />

4. Krakowka S, Eillis J, Meehan B, Kennedy S, McNeilly F, Allan G. (2000) Viral wasting syndrome of swine:<br />

experimental reproduction of postweaning multisystemic wasting syndrome in gnotobiotic swine by<br />

coinfection with porcine circovirus 2 and porcine parvovirus. Vet Pathol 37:254-263<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

AN UPDATE ON SEROPREVALENCE OF PORCINE REPRODUCTIVE AND RESPIRATORY<br />

SYNDROME IN MALAYSIA<br />

*JASBIR S. 1 , KAMARUDDIN M.I. 2 AND HASSAN L.. 3<br />

1<br />

Veterinary Research Institute, 59, Jalan Sultan Azlan Shah, 31400 Ipoh, Perak MALAYSIA<br />

2<br />

Department of Veterinary Services Head Quarters, 62630 Putrajaya MALAYSIA<br />

3<br />

Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor MALAYSIA<br />

*jasbir@jphvri.gov.my<br />

Introduction<br />

Porcine reproductive and respiratory syndrome (PRRS) is one of the most prevalent pig diseases in the<br />

world. It has swept across most of countries during the past two decades and brought enormous economic<br />

loss to the pig industry. The first laboratory confirmed case of PRRS in Malaysia was reported in 1997<br />

(Jasbir et al. 1997). The PRRS virus is suggested to be the primary cause of porcine respiratory disease<br />

complex (PRDC) in pigs (Thacker, 2001). This paper reports an update in the PRRS serological status of pig<br />

herds in Malaysia.<br />

Material & methods<br />

Out of the 670 pig farms in Peninsular Malaysia, 50 farms were selected randomly from major production<br />

areas in the country using stratified sampling methods to represent all the major production areas. A total of<br />

15 blood samples were collected from each farm, five samples from sows and 10 samples from porkers of<br />

age >4 months to avoid interference with maternal antibodies. A questionnaire on herd health and<br />

management practices was administered at the farms. The blood samples were tested by a commercial<br />

ELISA antibody test kit (Idexx, Inc.) at the Veterinary Research Institute, Ipoh and the results were analysed<br />

using the xChek software programme (Idexx, Inc.).<br />

Results<br />

None of the farms selected in this study vaccinate their animals against PRRS. From the total of 735 blood<br />

samples tested from the 50 farms, 613 (83.4%) of the samples and 47 (94%) of the farms were positive for<br />

PRRS antibodies. The results demonstrate widespread occurrence of the PRRS viral agent among the pig<br />

population in the country. Pig farms with good biosecurity were also not spared from PRRS. However, there<br />

was a lack of clear clinical signs of the syndrome observed in most of the seropositive pigs, indicating a<br />

probable subclinical infection.<br />

Discussions & Conclusions<br />

PRRS appears to be endemic in Malaysia, however very few farms vaccinate their animals against PRRS.<br />

The high seroprevalence of PRRS in the country clearly mimics the occurrence of the syndrome in many<br />

other parts of the world. A previous serological survey of PRRS conducted in 1996 indicated that out of 1990<br />

blood samples tested from 100 pig herds in the country, 1195 (60.1%) of the samples and 93 (93%) of the<br />

herds were tested positive for PRRS antibodies (Jasbir et al., 1996). The comparison between the years<br />

shows that the prevalence of PRRS in the country has remained almost the same even after a decade.<br />

PRRS has been suggested as a major co-factor of porcine multisystemic wasting syndrome (PMWS) [Kim et<br />

al., 2007]. A concurrent serological study in the state of Perak on porcine circovirus type 2 (PCV2), the<br />

probable causative agent of PMWS has suggested a high prevalence of the syndrome. Using a commercial<br />

ELISA test kit (Synbiotic SERELISA PCV2 antibody test, France), 9 of 15 pig herds (60%) tested were<br />

seropositive for PCV2 antibodies. Many of the pig farms in the country have poor hygiene, farm<br />

management and biosecurity and these factors may contribute to the high prevalence of the two syndromes<br />

in the country.<br />

Acknowledgement<br />

The author thanks the FAO for support to attend the <strong>WAVLD</strong> symposium and the Department of Veterinary<br />

Services (DVS), Malaysia for permission to publish this abstract. Appreciation is also extended to all DVS<br />

staff who contributed in the collection of organ and blood samples and to Komala T., Nisha T.G., Kong S.L.<br />

for help in the bench work.<br />

References<br />

1. DH Kim, S Kim, SW Wang, et al. (2007) Serological Prevalence of Porcine Reproductive and Respiratory<br />

Syndrome virus by ELISA in 2005/2006 in South Korea. In: Proc. 3 rd Congr Asian Pig Vet Soc. p. 93.<br />

2. Jasbir S, Hussin A.A., Murdan Z.A. and Too H.L. (1996). Porcine reproductive and respiratory syndrome in<br />

Malaysia. - In: Proc 8 th Vet Assoc Malaysia Sc Congr, p. 72-75.<br />

3. Jasbir S., Chen K.S. and Kono Y. (1997). Isolation and characterisation of Porcine reproductive and<br />

respiratory syndrome virus in Malaysia. - In: Proc 9 th Vet Assoc Malaysia Sc Congr, p. 27-29.<br />

4. Thacker EL. (2001). Immunology of the porcine respiratory disease complex. Vet Clin North Am Food<br />

Anim Pract. 17(3):551-65.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

INVESTIGATIONS OF NODAVIRUS INFECTIONS OF AUSTRALIAN BASS<br />

KE Arzey* 1 , Y Lele 1 , S Vrankovic 1 , DS Fielder 2 , KR King 1 , X Gu 1 and PD Kirkland 1<br />

1 Virology Laboratory, Elizabeth Macarthur Agricultural Institute, NSW DPI, Menangle, NSW Australia<br />

2 Port Stephens Fisheries Centre, NSW DPI, Taylor’s Beach NSW Australia<br />

Introduction<br />

In 2004, nodavirus infection was diagnosed for the first time as the cause of high mortality in a batch of<br />

Australian bass larvae hatched and reared in captivity for ultimate release into rivers and dams for<br />

recreational fishing. To protect wild populations, a program of testing of hatchery reared larvae/fingerlings for<br />

piscine nodavirus was initiated prior to release into the wild. However, little is known about the status of wild<br />

populations or the epidemiology of nodavirus in Australian bass. In 2006, a batch of Australian bass larvae<br />

were tested and found to be infected with nodavirus. This prevented the release of the larvae into the wild<br />

but provided an opportunity to assess the persistence of nodavirus infection in the batch and to compare<br />

virus detection methods. This paper presents the results of this investigation.<br />

Material & methods<br />

Samples from a batch of approximately 50,000 larvae were initially collected at 22 days post spawning and<br />

tested for evidence of nodavirus infection. Sections of formalin fixed eyes and brains (150 larvae) were<br />

examined by histopathology and RNA extracts from homogenates of 150 whole larvae (in pools of 10) were<br />

tested in a nested reverse transcriptase polymerase chain reaction (nRTPCR) 1 . When it was found that the<br />

broodstock pool had inadvertently included fish that had tested positive for nodavirus 2 years before, release<br />

of the larvae was suspended and further testing undertaken. At 3 months of age 500 fingerlings were<br />

sampled for intensive testing (in pools of 5) by nested PCR, virus isolation in cell culture and later in a real<br />

time RTPCR (qPCR) assay that was developed during the course of this study. Further samples of 1-200<br />

fingerlings were collected at 6, 11, 12 and 13 months and examined by qPCR, virus isolation and, at times,<br />

histopathology. Virus isolation was conducted by inoculating near confluent monolayers of SSN-1 cells with<br />

clarified homogenates of eyes and brains (usually from individual fish). Cultures were passaged up to 3<br />

times at intervals of 7 days. These homogenates were also tested in the qPCR. This assay was based on<br />

Taqman technology with primer sequences that were conserved across all nodavirus sequences lodged in<br />

Genbank and from the nodaviruses isolated from barramundi and bass in Australia and used an appropriate<br />

fluorogenic probe to match a region within the amplified sequence. From time to time histopathology was<br />

conducted on sections of formalin fixed paraffin embedded sections using conventional methods.<br />

Results<br />

There was no evidence of nodavirus infection in the samples of larvae that were examined by histopathology<br />

and nested RTPCR at 22 days post spawning. However, at 3 months, nodavirus was isolated in cell culture<br />

from each of the pools of fingerlings tested. At 6 months, virus isolation was conducted on 100 individual<br />

fingerlings and each was found to be infected with nodavirus. Each of the 3 & 6 month samples were also<br />

found to be positive by qPCR. About one third of the 3 month samples gave a positive result in the nRTPCR.<br />

At 11 - 13 months, nodavirus was detected in a smaller proportion of fingerlings by both virus isolation and<br />

qPCR. The results indicate that the qPCR has higher sensitivity than virus isolation. Despite the initial very<br />

high incidence of infection, there was no clinical evidence of disease or histological lesions in the<br />

larvae/fingerlings at any time. Sampling and testing are continuing.<br />

Discussion & conclusions<br />

Subclinical nodavirus infection has been described 2 in commercially farmed halibut in Norway that were<br />

survivors of a natural outbreak of viral encephalopathy and retinopathy (VER) and the virus was shown to be<br />

still viable in these fish 9 -12 months after hatching. This longitudinal study of Australian bass naturally<br />

infected with nodavirus has shown that the virus can persist in clinically normal fish for at least 12 months.<br />

Virus isolation was a more sensitive method than the conventional nested RTPCR for nodavirus detection.<br />

However, the real time RTPCR used in this study has very high sensitivity, perhaps more sensitive than virus<br />

isolation, and will be a valuable tool for the detection of nodaviruses in fish.<br />

References<br />

1. Moody NJ, Horwood PF and S McHardy (2004) FRDC Project No 2001/626<br />

2. Johansen R, Grove S, Svendsen AK, Modahl I and B Dannevig (2004) J Fish Dis. 24: 327-341<br />

Acknowledgements<br />

Funding for this project was provided by the NSW Recreational Fishing Trust and NSW DPI. We are<br />

indebted to Drs R. Reece and S. Hum for assistance with histopathology.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

DIAGNOSIS AND COUNTERMEASURE OF ALVEOLAR ECHINOCOCCOSIS IN RED FOXES UTILIZING<br />

LOCAL RESOURCES IN JAPAN<br />

M. Kamiya 1 and J.T. Lagapa 1,2<br />

(1) OIE Reference Laboratory for Echinococcosis and Laboratory of Environmental Zoology, Department of Biosphere and<br />

Environmental Sciences, Faculty of Environmental Systems, Rakuno Gakuen University, Bunkyodai-Midorimachi 582, Ebetsu, Hokkaido<br />

069-8501, Japan<br />

(2) Animal Disease Diagnostic Laboratory, Surgery and Zootechnics, College of Veterinary Medicine, Central Mindanao University,<br />

Musuan, Bukidnon, 8710, Philippines<br />

The paper describes successful field diagnosis and countermeasures against Echinococcus multilocularis in<br />

red foxes utilizing intravital coproantigen examination and anthelmintic laced bait made from local resources.<br />

Local residents through their Nonprofit Organizations (NPO) initiated the baiting campaigns with the<br />

assistance of Forum on Environment and Animals, a Limited Liability Company founded by our laboratory.<br />

This novel concept of using local resources such as manpower, fish-waste products and finances by NPO’s<br />

for diagnosis and control led to what is known as “endogenous development”. This is seen as a key to a<br />

wider and sustainable control campaigns against echinococcosis in Hokkaido, Japan.<br />

Initial prevalence rate of Echinococcus infection in foxes was determined in Koshimizu, Hokkaido (200 km 2 )<br />

in 1997. Annual fecal collection for diagnostic purposes and bait distribution were assumed by a local NPO<br />

(Okhotsk Sanctuary) from 2002 to 2006. Baits were made using fish-waste products and fortified with<br />

praziquantel (50 mg/bait). Prevalence rates in fox feces were assessed annually using coproantigen ELISA.<br />

In Kutchan, Hokkaido (100 km 2 ), a local residents’ NPO (WAO) initiated fecal collection to assess prevalence<br />

rates in 2005. From May to November, 2006, about 1,500 praziquantel-fortified baits were distributed<br />

monthly by the NPO at specific locations guided by a GIS-based map.<br />

Initial prevalence rates in Koshimizu in 1997 were 51.6-66.7%. Fecal examinations conducted in 2006<br />

revealed a zero coproantigen prevalence rate after annual bait distribution by local residents. In Kutchan, a<br />

coproantigen prevalence rate of 21% was registered during baseline surveys. Seven months after<br />

continuous baiting, the� prevalence rates of taeniid egg and coproantigen positive feces dropped to 0% and<br />

2%, respectively. The findings suggests a control initiative against alveolar echinococcosis by targeting the<br />

sources of infection which are the red foxes, the major definitive hosts. Baiting campaigns initiated by local<br />

residents using anthelmintic-laced baits and intravital diagnostic techniques by coproantigen examination<br />

conducted by FEA indicated a successful and sustainable approach in the elimination of alveolar<br />

echinococcosis in Japan.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SMART FIELD SAMPLING FOR DIAGNOSIS OF ECHINOCOCCOSIS IN WILDLIFE DEFINITIVE HOST<br />

USING GIS-BASED MAPS<br />

J.T. Lagapa 1,2 , Y. Oku 3 , M. Kaneko 4 and M. Kamiya 1<br />

(1) OIE Reference Laboratory for Echinococcosis and Laboratory of Environmental Zoology, Department of Biosphere and<br />

Environmental Sciences, Faculty of Environmental Systems, Rakuno Gakuen University, Bunkyodai-Midorimachi 582, Ebetsu, Hokkaido<br />

069-8501, Japan<br />

(2) Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Central Mindanao University, Musuan, Bukidnon, 8710,<br />

Philippines<br />

(3) Laboratory of Parasitology, Graduate School of Veterinary Medicine, Hokkaido University, Kitaku, Kita 18-jo Nishi 9 chome, Sapporo,<br />

Hokkaido 060-0818, Japan<br />

(4) Laboratory of Environmental Geographic Information System (GIS), Department of Biosphere and Environmental Sciences, Faculty<br />

of Environmental Systems, Rakuno Gakuen University, Bunkyodai-Midorimachi 582, Ebetsu, Hokkaido 069-8501, Japan<br />

Field sampling of fecal samples from wild red foxes for the diagnosis of echinococcosis requires an intensive,<br />

costly and laborious field works. To deplete these factors, Geographic Information System (GIS)-based maps<br />

on potential distribution of environmental contamination by eggs of Echinococcus multilocularis from feces of<br />

infected foxes were made in Nopporo Forest Park, Hokkaido, Japan. It has been known that red foxes used<br />

their feces as landmarks at their foraging habitats. Present study aimed to determine exact locations of fecal<br />

contamination using Global Positioning System (GPS) and to create seasonal distribution maps using GIS<br />

software. Exact location of fox feces was recorded (+50 cm) by a handheld GPS (Pathfinder Pro XR;<br />

Trimble) using ArcPad 6 software (ESRI). Data were fed into a computer using Microsoft Active Sync<br />

program. Seasonal distribution maps of fox feces were created using ArcView 8 software package (ESRI).<br />

Data indicated changes in foraging habitats of foxes as evidenced by fecal landmarks and were tested for<br />

fast and easy field sampling of wild fox feces. Digital map was downloaded to a hand-held GPS and enabled<br />

the researcher to reach the site with ease and accuracy and a higher probability of finding fox feces. This<br />

novel method reduced the required time to collect fecal samples for diagnosis by 80%.These GIS-based<br />

maps could also be utilized in optimizing bait distribution for cost-effective intervention and in monitoring<br />

control programs or re-emergence of the disease over time. Our study indicates a novel system that provides<br />

a smart field sampling technique for the diagnosis of E. multilocularis in wild fox population.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

Mycoplasma mycoides subsp. capri ASSOCIATE WITH GOAT RESPIRATORY DISEASE AND HIGH<br />

FLOCK MORTALITY<br />

Hernandez A. L 1 , Lopez J 2 , St-Jacques M 2 , Ontiveros CL1, Acosta J 1 , Handel K 2 .<br />

1 CENID-Microbiología INIFAP-SAGARPA, Bacteriology Laboratory, Carretera Libre México-Toluca, Km 15.5 Palo Alto, Cuajimalpa D.F.<br />

ZC. 05110, México. 2 National Centre for foreign animal Disease, Canadian Food Inspection Agency, 1015 Arlington Street Winnipeg,<br />

Manitoba R3E 3M4.<br />

Introduction<br />

A number of Mycoplasma species has been associated with caprine respiratory disease, but some of the<br />

most virulent ones are Mycoplasma capricolum subsp. capripneumoniae (Mccp), M. mycoides subsp.<br />

mycoides Large Colony Type (MmmLC) and M. mycoides subsp. capri (Mmc). All these mycoplasmas share<br />

genomic and antigenic features, which result in very closely similar biochemical and serologic properties,<br />

making precise identification of individual isolates a difficult task and contributing to confusion among<br />

diagnosticians. The prevalence and relative importance of Mycoplasma infection in Mexico is not known. By<br />

this reason the objectives were isolate and identify the mycoplasmal organisms causing pneumonia in goats.<br />

Material & methods<br />

An outbreak of respiratory disease occurred in a herd of 2000 Sannen dairy goats in the State of Durango,<br />

Mexico. This was a closed, intensive milk production flock. Due to increased demand for goat milk, animals<br />

of different breeds were imported from Canada and the USA, approximately 6 months before the start of the<br />

outbreak. Initially 200 animals died over a period of 15 d and after 6 months, the flock mortality had reached<br />

40%. Animals of all ages were affected, although they were most adults. The clinical picture was<br />

characterized by fever, abundant nasal secretion, difficult breathing, prostration, ear drop, and low milk<br />

production. Fourteen animals were necropsied.<br />

Results<br />

Grey, red, or both, areas of consolidation were seen in the lungs, affecting 1 or more lobes. All animals<br />

showed marked pleuritis and pleural effusion. The cut surface of some affected lungs revealed a fine<br />

granular texture with hepatisation. The histopathologic findings were consistent with proliferative interstitial<br />

pneumonia. Mycoplasma growth from pleural and fluid and lung samples was evident after 48 and 72 h<br />

incubation in Friis medium. To rule out the possibility that the outbreak might have been caused by the exotic<br />

Mycoplasma, the PCR-PstI digestion was followed, using DNA templates obtained from all the Mycoplasma<br />

isolates and directly from the pleural fluid and lung tissue. The isolates were identified as Mycoplasma<br />

mycoides subsp. capri.<br />

Discussions & Conclusions<br />

A high mortality outbreak of respiratory mycoplasmosis occurred in goats in Mexico. The clinicopathologic<br />

presentation resembled contagious caprine pleuropneumonia caused by Mycoplasma capricolum<br />

subspecies capripneumoniae. By using a battery of polymerase chain reaction assays, the Mycoplasma<br />

associated with this outbreak was identified as Mycoplasma mycoides subsp. capri. The prevalence and<br />

relative importance of Mmc infection in Mexico is not known. There is only 1 report of this condition in the 60s<br />

and, at the time, it was thought to be the first report of CCPP in the Americas.<br />

References<br />

Dedieu L, Mady V, Lefevre PC. Development of a selective polymerase chain reaction assay for the<br />

detection of Mycoplasma mycoides subsp. mycoides S.C. (contagious bovine pleuropneumonia agent). Vet.<br />

Microbiol 1994;42:327-339.<br />

Friis NF. Mycoplasmas cultivated from the respiratory tract of Danish pigs. Acta Vet Scand 1971;12:69-79<br />

Hotzel H, Sachse K, Pfützner H. A PCR scheme for differentiation of organisms belonging to the<br />

Mycoplasma mycoides cluster. Vet. Microbiol 1996;49:31-43.<br />

Solana P, Rivera E. Infection of goats in Mexico by Mycoplasma mycoides var capri. Ann NY Acad Sci<br />

1967;143:357-363.<br />

Thiaucourt F, Bölske G, Leneguersh B, Smith D, Wesonga H. Diagnosis and control of contagious caprine<br />

pleuropneumonia. Rev Sci Tech 1996; 15:1415-1429.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

RESERVOIR OF WEST NILE VIRUS AND ANOTHER ENCEPHALITIS VIRUS IN MEXICAN BATS<br />

(Desmodus rotundus).<br />

S. Cuevas*, A. Alvarado, P. Mejia, G. Colmenares, M. Camara (INIFAP-CENIDM-Mexico), N. Nemeth. (CDC-Fort Collins<br />

Colorado,USA) R. Navarro, Y. Barrera (CPA-SAGARPA-México), A. L. Salgado, G. Estrada, (UTMB-USA), E. Cruz (ZOOMAT-INHE).<br />

Introduction<br />

West Nile virus (WNV) is a flavivirus endemic in Africa, the Middle East and in South-western Asia. In North<br />

America the initial outbreak of WNV was reported in New York City in August 1999 with deaths reported in<br />

humans, horses and numerous species of birds. In the spring of 2003, the first WNV isolate found in Mexico<br />

was obtained from a raven in the southeastern state of Tabasco. The current study focused in determining<br />

the potential role of the hematofagous bat (Desmodus rotundus) might play as potential reservoir in the<br />

transmission cycles of WNV in Mexico. Insert text<br />

Material & methods<br />

Eighty bats captured from southeastern Mexican state of Chiapas were tested for reservoir potential of WNV.<br />

Previous to the experimental procedures all serum samples were screened for antibodies to WNV by plaque<br />

reduction neutralization tests (PRNT) and other arbovirus and assured free flavivirus antibody response.<br />

Those results plus experimental data are presented here. We also tested some brain tissues to WNV by<br />

Real time-PCR (TIB-Molbiol, Berlin, Germany) and Rabies virus by fluorescent antibodies test.<br />

Results<br />

All serum samples were negative to presence of WNV, St. Louis encephalitis virus (SLEV) and EEV<br />

antibodies. Twelve brain samples of the 80 Desmodus rotundus bats were tested negatives to presence of<br />

WNV by a commercial kit to Rapid Cycle Real Time PCR (TIB-Molbiol, Berlin, Germany) and also these<br />

samples were negatives to fluorescents antibodies to Rabies virus.<br />

Discussions & conclusions<br />

These finding suggests that Desmodus bats collected from an endemic WNV zone in Mexico, probably<br />

suggests a limited interaction between WNV and bats in the transmission cycle.<br />

References<br />

J. Wildl Dis, 2006, 42(2) 455-8. A survey for West Nile virus in bats from Illinois. Bunde JM, Heske EJ, et al.<br />

J. Wildl Dis, 2004, 40(2) 335-7. West nile virus antibodies in bats from New Jersey and New York. Pilipski<br />

JD, Pilipski LM, et al.<br />

Am J Trop Med Hyg. 2005,73(2):467-9.Experimental and natural infection of North American bats with West<br />

Nile virus. Davis A, Bunning M, et al.<br />

Stream: Diseases of Wildlife PresentationType: Poster<br />

Presenting Author: Sandra Cuevas Romero cuevas.julieta@inifap.gob.mx<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SEROEPIDEMIOLOGY OF THE ARBOVIRUS OF CRIMEAN- CONGO HEMORRHAGIC FEVER IN<br />

RURAL COMMUNITY OF BASRA<br />

Adel Alyabis* ,Hassan .J.Hasony<br />

*Laboratory Department ,Microbiology and Serology Sub department ,General Hospital ,Basra, Iraq.<br />

Key words; Seroepidemiology<br />

Introduction and objectives<br />

In 1979 cases of Crimean-Congo Hemorrhagic Fever (CCHF)were recognized in Iraq ,following years<br />

several cases of CCHF were diagnosed in Basra, southern Iraq. Aseroepidemiological survey was carried<br />

out in rural community of Basra to estimate the size of the enzootic focus in which the CCHF virus is<br />

circulating . A total of 682 serum samples were collected from apparently healthy individuals, their ages<br />

range from 5 to76 years , 20% of the collected sera were obtained from occupational risk groups<br />

(veterinarians , abattoirs workers , farmers).Sera collected from 74 sheep and 48 cattle , 42 tick pools<br />

were also gathered parallel to the human and animals sera in the same areas.<br />

Material and methods<br />

Enzyme-linked immunosorbent assay (Elisa), was used to detect the prevalence of circulating IgG antibodies<br />

in the collected sera.<br />

Results<br />

In general IgG antibodies against CCHFV were found in 4.3% of the resident in rural areas of Basra,<br />

Seropositive sera were detected in 9.7% of northern residents , while 20% of the sheep sera and 37% of<br />

cattle sera were seropositive which indicate that the virus is circulating in the area within the endemic level .<br />

The tick pools were identified , the predominant tick species was diagnosed as Hyaloma marginatum.<br />

Discussion and Conclusions<br />

The existence of enzootic focus for the CCHFV is maintained by ecological , socioeconomic variables , the<br />

possibility of emerging infections should always be considered.<br />

References<br />

1.AL-Tikriti Sk. Crimean -Congo Hemorrhagic Fever in Iraq. Bull WHO 1981;59:58-90.<br />

2.Tantawi HH. Moslih IM. Hassan Fk .Crimean –Congo Hemorrhagic Fever .Al-Mthana House Baghdad ,<br />

Iraq.1980;1-60.<br />

3.WHO Viral Hemorrhagic Fever. Report of WHO scientific Committee , series 1985 ;No.716.<br />

4.Hoogstral H.Tick Borne Crimean-Congo Hemorrhagic Fever; <strong>Handbook</strong> series of Zoo noses (ed) In chief<br />

Steel JH. 1' ed., section B., Viral Zoo noses , volume 1.Baron Gw.Florida Boka,Raton 1981;267-402.<br />

5.Gonzalis JB,Legunoo B,Gulland M,A fatal case of Crimean-Congo Hemorrhagic fever virus in Muritania,<br />

Virological &Serological Suggestion, Epidemic transmission Trans Roy Soc.Trop Med Hyg1990:84(4);573-<br />

576.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

SURVEILLANCE PROGRAM FOR NEWCASTLE DISEASE CONDUCTED SINCE 1998 IN ARGENTINA<br />

C. ESPINOSA 3 , M. V. TERRERA 3 , R. De BENEDETTI 3 , P. BORGNA 3 , E. PARRADO 3 1, 2<br />

and C. BUSCAGLIA<br />

1<br />

Cátedra de Zootecnia Especial III (Aves y Piliferos), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata. CC 296<br />

(B190AVW) La Plata. Argentina. E-mail: cb235@yahoo.com<br />

2 Comisión de Investigaciones Científicas de la Prov. Buenos Aires. Argentina.<br />

3 SENASA, Buenos Aires, Argentina<br />

Newcastle Disease (ND) was diagnosed for the first time in Argentina in 1961. The last outbreak of the<br />

disease produced by a velogenic strain was reported in the province of Entre Rios in 1987.The aim of the<br />

epidemiological surveillance is to establish the presence or absence of antibodies or virus of ND in all the<br />

avian population There are two regions in the country where the samples are obtained: a) region A that<br />

include the avian population in the border with Brazil, Bolivia, and Paraguay, and b) region B that include the<br />

rest of the provinces of the country.<br />

This study is part of the prevention program of AI and velogenic ND in Argentina that includes the avian<br />

populations such as backyard chickens, commercial poultry, all the controls for importation and exportation<br />

of live birds and the samples obtained since 1998/. More than 30.000 sera samples and tracheal or cloacal<br />

swabs were tested by ELISA and/ or HI, and for virus isolation in embryonated SPF eggs respectively. The<br />

results were negative for velogenic NDV.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

PURIFICATION OF A HERPES-LIKE VIRUS IN ABALONE<br />

(HALIOTIS SPP) AND DETECTION BY TRANSMISSION ELECTRON MICROSCOPY<br />

Jianming Tan 1 , Malcolm Lancaster 1 , Alex Hyatt 2 ,<br />

Rosemary van Driel 2 , Frank Wong 1 , Simone Warner 1<br />

1 Department of Primary Industries,<br />

475 Mickleham Rd, Attwood,VIC 3049, Australia<br />

2 CSIRO, Livestock Industries, Australian Animal Health Laboratory,<br />

5 Portarlington Rd, Geelong, VIC 3220, Australia<br />

A herpes-like virus was successfully purified from abalone diagnosed with ganglioneuritis on the south coast<br />

of Victoria, Australia. Pleuropedal ganglia, pedal nerve cords, head and epipodial tissue were collected and<br />

homogenized from abalone populations exhibiting high mortality and clinical signs consistent with herpes-like<br />

virus ganglioneuritis. After ultracentrifugation using a discontinuous 10-60% sucrose gradient, a total of five<br />

visual bands appeared at each of the five interfaces of the sucrose gradient. Herpes-like viral particles were<br />

mainly found at the 40-50% interface, and to a lesser extent at the 50-60% sucrose interface, as determined<br />

by examination of negatively stained preps on a transmission electron microscope (TEM). The virus particles<br />

were observed to have icosadeltahedral capsids, and many of the capsids appeared to be surrounded by a<br />

single envelope with numerous spikes on the external surface. The naked nucleocapsid ranged from 92 to<br />

109 nm, and the enveloped particle was approximately 150 nm in diameter. In order to determine the<br />

buoyant density of the virus, isopycnic gradient centrifugation was performed using both potassium tartrate<br />

and caesium chloride gradients. Using this technique, the buoyant density of the herpes-like virus was<br />

determined to be 1.17 and 1.18 g/mL, respectively.<br />

This is the first report to successfully purify herpes-like virus from infected abalone using ultracentrifugation.<br />

A clear result was obtained using the sucrose gradient, which reinforced its effectiveness in purification of<br />

the abalone herpes-like virus, where further purification with other gradients appeared unnecessary. The use<br />

of sea-water as the gradient buffer was critical in the preliminary purification of the virus.<br />

This approach to successfully purify herpes-like virus from infectious abalone establishes a firm ground for its<br />

further investigation in research, including the classification, identification and pathogenic studies of the<br />

virus. The appearance of the virus was observed clearly in negative staining by TEM, despite contamination<br />

by cellular debris. The quality and purity of the resultant virus was amenable to follow-on work including<br />

DNA extraction and sequencing.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

GENETIC HETEROGENEITY OF BOVINE VIRAL DIARRHOEA VIRUS (BVDV) ISOLATES FROM ITALY:<br />

IDENTIFICATION OF NEW BVDV-1 GENOTYPES<br />

M. Giammarioli, C. Pellegrini, E. Rossi, G.M. De Mia *<br />

Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche, Perugia (Italy)<br />

Introduction<br />

Bovine viral diarrhea virus (BVDV) belongs to the genus Pestivirus with classical swine fever virus and<br />

border disease virus in the Family Flaviviridae. Presently two species of BVDV are recognised. BVDV-1<br />

infection occurs worldwide involving mainly respiratory, reproductive and enteric organs. BVDV-2 causes<br />

similar clinical signs as BVDV-1, except that infection with highly virulent isolates may lead to fatal<br />

haemorrhagic syndrome. Genetic typing of BVDV has usually been performed using sequences from the 5’-<br />

UTR, N pro and E2 regions. Sequence analysis of 5’-UTR can distinguish BVDV-1 and BVDV-2 and can<br />

subdivide BVDV-1 into at least eleven genetic groups (3). Genetic typing of BVDV-2 isolates has not been as<br />

extensive and at present, only two to four genetic groups of BVDV-2 have been described.<br />

Studies on the prevalence of BVDV in Italy have been conducted providing evidence of circulation of five<br />

BVDV-1 genotypes (1). BVDV-2 circulation has been reported in cattle as well (2). To better define the<br />

genetic pattern within BVDV Italian isolates we studied a broad range of BVDV isolates. Our genetic study<br />

has been based on the 5’-UTR.<br />

Materials & Methods<br />

Eighty-eight BVDV strains have been collected mainly during a period of 8 years (2000-2007) from 12 Italian<br />

regions. The viruses were mostly from cattle (n=81), buffalo (n=3) and sheep (n=4). The samples have been<br />

originated from individual dead animals as well as persistently infected animals. The genomic region<br />

encoding the highly conserved 5'-UTR of the pestivirus genome was amplified using primers 324 and 326<br />

flanking a 288 bp DNA fragment (4). Nucleotide sequences were determined by cycle sequencing three<br />

independent cDNA clones. Sequences were aligned using the Clustal X (version 1.83) analysis program and<br />

were proof read using the BioEdit version 7.0.0. Phylogenetic trees was calculated using the Phylip (version<br />

3,66) program package based on the neighbor joining algorithm. Visualization of the phylogenetic trees was<br />

performed using TREEVIEW, version 1.6.6.<br />

Results<br />

All the 88 viruses were sequenced in the 5’-UTR. Genetic typing revealed that 83 isolates were BVDV-1 and<br />

the topology of the tree indicates that they belonged to 7 distinct genotypes namely respectively BVDV-1a<br />

(n=8), BVDV-1b (n=37), BVDV-1d (n=3), BVDV-1e (n=22), BVDV-1f (n=4), BVDV-1g (n=4) and BVDV-1h<br />

(n=5). Non-bovine isolates were all typed as BVDV-1.<br />

Five viruses that originated from Lombardia and Basilicata were typed as BVDV-2 and may be grouped into<br />

two distinct phylogenetic groups BVDV-2a (n=4) and BVDV-2b (n=1).<br />

Discussion & conclusions<br />

The aim of this work was to study the genetic variability of a broad range of BVD viruses circulating in Italy.<br />

For this purpose an extensive collection of 88 BVDV isolates collected all over the country, has been<br />

investigated by genetic typing, providing further evidence for the heterogeneity of BVDV.<br />

At the subgroup level, pair wise similarity and cluster analysis provided a clear-cut assignation to 7 distinct<br />

genotypes of 83 isolates typed as BVDV-1. Most cattle farms were infected by the predominant BVDV-1b<br />

and BVDV-1e isolates,the others genotypes occurred only sporadically. The results also provided evidence<br />

for circulation of BVDV-1a and BVDV-1g additional genotypes, which have been never shown before in Italy.<br />

Five field viruses were typed as BVDV-2 and were clustered into the genotypes BVDV-2a and BVDV-2b. It<br />

should be mentioned that in Italy little is done to prevent the spread of BVDV, nevertheless the emergence of<br />

BVDV-2 in the field is very uncommon supporting the hypothesis of a iatrogenic infection in relation with<br />

contaminated vaccines. In summary, the results presented in this work revealed a high BVDV genetic<br />

heterogeneity in Italy. This is the result of the absence of any BVDV systematic control measures. Indeed, in<br />

Italy there is no BVDV national control program and the management practices such as cattle trade and<br />

movement, expose cattle herds to a high risk of introduction of BVDV infection as well as of new genetic<br />

variant of BVDV as a consequence of a high diversity of BVDV.<br />

References<br />

1. Falcone, E., Cordioli, P., Tarantino, M., Muscillo, M., La Rosa, G., Tollis, M., 2003. Genetic heterogeneity<br />

of bovine viral diarrhea virus in Italy. Vet. Res. Commun. 27, 485-494.<br />

2. Luzzago, C., Bandi, C., Bronzo, V., Ruffo, G., Zecconi, A., 2001. Distribution pattern of bovine viral<br />

diarrea virus strains in intensive cattle herds in Italy. Vet. Microbiol. 83, 265-274.<br />

3. Vilček, Š., Ďurkovič, B., Strojni, L., Ibata, G., Moussa, A., Loitsch, A., Rossmanith, W., Vega, S.,<br />

Scicluna, M.T., Palfi, V., 2001. Bovine viral diarrhea virus genotype 1 can be separated into at least<br />

eleven genetic groups. Arch. Virol. 146, 99-115.<br />

4. Vilček, Š., Ďurkovič, B., Kolesàrovà, M., Greiser-Wilke, I., Paton, D., 2004. Genetic diversity of<br />

international bovine viral diarrhea virus (BVDV) isolates: identification of a new BVDV-1 genetic group.<br />

Vet. Res. 35, 609-615.<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

GENETIC CHARACTERISATION OF BORDER DISEASE VIRUS ISOLATED FROM CHAMOIS<br />

S.Vilcek 1* , K. Willoughby 2 , P.F. Nettleton 2<br />

1 University of Veterinary Medicine, Kosice, Slovakia; 2 Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian EH26<br />

0PZ, UK<br />

Introduction<br />

Border disease virus (BDV) belongs to four well recognised Pestivirus species, family Flaviviridae. A recent<br />

antigenic and genetic analysis of new pestiviruses isolated from sheep of continental Europe have led to the<br />

proposal that BDV strains can by phylogenetically allocated into three BDV genotypes, namely BDV-1, BDV-<br />

2 and BDV-3 (Becher et al., 2003). Recently we have typed a pestivirus isolated from chamois in Pyrenees<br />

(Andorra) as additional BDV-4 genotype (Arnal et al., 2004).<br />

The aim of this work was further characterization of the chamois pestivirus strain by sequencing of structural<br />

and non-structural genes.<br />

Material & methods<br />

Total RNA of a virus stock (Chamois-1) was extracted using the Viral RNA minikit (Quigen). The cDNA was<br />

prepared using random hexamers and Moloney Leukaemia Virus reverse transcriptase. Different PCR<br />

products of the pestivirus genome were prepared using single or nested PCR employing panpestivirus or the<br />

chamois virus specific primers and proofreading DNA polymerase. Longer fragments were prepared by the<br />

Expand Long Template system (Roche). Nucleotide sequences were proofread using the SeqMan II<br />

program from DNASTAR (Lasergene).The percentage of nucleotide and deduced amino acid similarities<br />

were calculated using the MegAlign program. Phylogenetic analysis was carried out using Neighbor-joining<br />

method (program NEIGBOR from PHYLIP inference package programs) including selected pestivirus<br />

nucleotide sequences from GenBank.<br />

Results<br />

The alignment in Npro region for all pestivirus genotypes revealed that its size is 168 amino acids and 66 of<br />

them are totally conserved in all pestiviruses. The Npro/C cleavage site is also conserved in all genotypes.<br />

Of six cysteine residues, five are conserved in all pestivirus genomes. The hydrophobicity profile is similar for<br />

all pestiviruses. No significant insertion/deletion was observed in any part of the pestivirus genome, including<br />

the NS2-3 region. The nucleotide sequence similarity between Chamois-1 strain and two other chamois<br />

isolates is in the entire E2 region around 86%.<br />

The nucleotide sequence similarity between Chamois-1 and other pestiviruses varied depending on the<br />

pestivirus genotypes. The most similar values were found for BDV-1 – BDV-5 viruses (62-67% in Npro, C,<br />

Erns and E1 regions, 53% in E2). The similarity values for CSFV varied in the range 68-74% and 59% in E2<br />

region. Corresponding values for BVDV-1 and BVDV-2 varied between 61-69% and 49-53% in E2 region.<br />

Phylogenetic analysis in all genomic region confirmed that Chamois-1 strain belongs to BDV species, BDV-4<br />

genotype.<br />

Discussions & conclusions<br />

The genetic characterization of Chamois pestivirus strain contributes to better understanding of genetic<br />

diversity of pestiviruses as a result of virus evolution at the genetic level. The relationship between<br />

pestiviruses identified in chamois and similar BDV isolates detected in sheep on Iberian Peninsula remains<br />

to be elucidated.<br />

References<br />

Arnal, M. et al.: J. Gen. Virol. 85, 3653, 2004; Becher, P. et al.: Virology 311, 96, 2003.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MOLECULAR CHARACTERIZATION OF NEWCASTLE DISEASE VIRUSES ISOLATED FROM RECENT<br />

OUTBREAKS IN ROMANIA<br />

Handan Coste, M. Turcitu*, St Nicolae, I. Sandu, Gh. Barboi<br />

Institute for Diagnostic and Animal Health, Bucharest, Romania<br />

Introduction<br />

Newcastle disease plays an important role in poultry losses in Romania due to the high morbidity and<br />

mortality registered in commercial and backyards farms, restrictions following the outbreaks and all the<br />

measures required for limiting and eventually eradicating the disease. Diagnostic approach of the disease<br />

needs to be fast, reliable and accurate in order to have a quick response to the emerging situations. Thus,<br />

sequencing and pathotyping of the isolates complete the virological methods for characterization of the<br />

isolates.<br />

Material and methods<br />

All samples subjected to the study - a total of 47 field isolates from 2000 to 2007 - were initially determined to<br />

be velogenic by plaque formation, the mean death time (MDT) of embryonated eggs and the intracerebral<br />

pathogenicity index (ICPI). Apart from those isolates, challenge strain "Craiova" (local velogenic strain) was<br />

analyzed.<br />

For preparation of virus RNA, commercially available kits were used (HighPure RNA Isolation Kit- Roche<br />

Applied Science, RNEasy Mini Kit- Qiagen), followed by single tube RT-PCR (OneStep RT-PCR kit- Qiagen)<br />

using primers from "F" gene region that spans the cleavage site. The amplicons obtained were purified from<br />

gel agarosis using "MinElute Gel Extraction Kit- Qiagen" and subjected to direct sequencing using "BigDye<br />

Terminator Cycle Sequencing Kit- Applied Biosystems ".<br />

Results<br />

By translation of the nucleotide sequence from the cleavage site, all samples showed an amino acid motifs<br />

109 119<br />

SGGRRQKR/FIG thus belonging to mesogenic/velogenic pathotype. Subsequently, the sequences<br />

were aligned using Clustal W Alignment software and a phylogenetic tree was build using Mega 3.1<br />

software. Phylogenetic analysis demonstrated that all field strains belong to genotype VII (fourth panzootic of<br />

ND), with close phylogenetic relationship. As expected, challenge strain "Craiova", isolated from outbreaks in<br />

1985, belong to genotype IV (responsible for first panzootic of ND).<br />

Discussions and conclusions<br />

Close phylogenetic relation between field isolates suggest the endemic evolution of ND in Romania, with one<br />

virus strain being responsible for the outbreaks. Moreover, molecular pathotyping proved to be a reliable tool<br />

providing important informations especially were no virus isolates can be achieved (eg low virus<br />

concentration samples, long time stored samples etc - data not shown).<br />

References<br />

1. Characterization of Newly Emerging Newcastle Disease Virus Isolates from the People's Republic of<br />

China and Taiwan: Li Yu, Zhilian Wang, Yihai Jiang, Leo Chang, and Jimmy Kwang - Journal of Clinical<br />

Microbiology, Oct.2001, p. 3512-3519<br />

2. Characterization of Newcastle Disease Virus Isolates by Reverse Transcription PCR Coupled to Direct<br />

Nucleotide Sequencing and Development of Sequence Database for Pathotype Prediction and Molecular<br />

Epidemiological Analysis: Bruce S. Seal, Daniel J. King, and Joyce d. Bennett - Journal of Clinical<br />

Microbiology, Oct. 1995, p. 2624-2630<br />

3. S Kumar, K Tamura, and M Nei (2004) MEGA3: Integrated software for Molecular Evolutionary Genetics<br />

Analysis and sequence alignment. Briefings in Bioinformatics 5:150-163.<br />

*corresponding author: e-mail Turcitu.Mihai@idah.ro<br />

Poster Presentations


Poster Presentations<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

MOLECULAR EPIDEMIOLOGY OF RABIES IN BAT-EARED FOXES (OTOCYON MEGALOTIS) IN<br />

SOUTH AFRICA.<br />

C.T. Sabeta a,b* , K.L. Mansfield c , L. M. McElhinney c , A. R. Fooks c , L H. Nel d<br />

a Rabies Unit, Onderstepoort Veterinary Institute, Private Bag X05, Onderstepoort, 0110, Pretoria, South Africa; b University of Pretoria,<br />

Department of Veterinary Tropical Diseases, P Bag XO4, Onderstepoort 0110, South Africa; c Rabies and Wildlife Zoonoses Group,<br />

WHO Collaborating Centre for the Characterisation of Rabies and Rabies-related Viruses, Veterinary Laboratories Agency, Weybridge,<br />

Woodham Lane, Surrey KT15 3NB, U.K and d University of Pretoria, Department of Microbiology and Plant Pathology, 0002 Pretoria,<br />

South Africa.<br />

Introduction<br />

Canid rabies was recently introduced into the southern Africa in the late 1940s from Angola (Swanepoel et<br />

al., 1993). It first entered the dog population in the northern Limpopo province of South Africa (Alexander,<br />

1952) and a few years later, the disease started to appear in black-backed jackal species C. mesomelas and<br />

the bat-eared fox O. megalotis (Swanepoel et al., 1993), most likely via a spill-over from the domestic dog.<br />

Rabies cases were later on reported in O. megalotis in the Central Cape Province, with another isolated<br />

case in the Etosha National Park (Namibia) in the 1950s. Rabies was identified once again in this species<br />

many years later in the western and central Cape Provinces, the western Free State and the rest of Namibia<br />

(Thomson and Meredith, 1993). In this study, we have addressed some questions pertaining to the<br />

transmission dynamics of RABV in this wildlife carnivore species. The primary objective of the study was to<br />

establish the genetic relationships of O. megalotis rabies virus strains with those obtained from other wildlife<br />

and domestic host species. In this way, we attempted to clarify whether rabies in O. megalotis is indeed a<br />

new and independent cycle and further to assess the public and veterinary health threat of the maintenance<br />

and expansion of rabies cycles in O. megalotis.<br />

Material & methods<br />

A cohort of rabies viruses (n=124) from wildlife host species (principally the bat-eared fox, Otocyon<br />

megalotis) and domestic carnivore species were collected between 1980 and 2005 from a region of South<br />

Africa associated with endemic bat-eared fox rabies. The highly variable G-L intergenic region and the<br />

conserved nucleoprotein gene of each of the rabies viruses in this South African panel were amplified,<br />

sequenced and analysed phylogenetically.<br />

Results<br />

Although it was demonstrated that all these viruses were very closely related (indicating a recent and<br />

common origin), they could be segregated into two major phylogenetic groups. The topologies of the<br />

phylogenetic trees obtained with analyses of the G-L intergenic and the N gene sequences were similar,<br />

although some isolates were better resolved in the former.<br />

Discussions & conclusions<br />

The data obtained in this investigation complement antigenic and surveillance data on rabies in this host<br />

species in South Africa. Most importantly these data support a hypothesis that the bat-eared fox<br />

independently maintains rabies cycles in specific geographical loci in south-western South Africa. This is the<br />

first molecular epidemiological investigation describing rabies transmission dynamics in this wildlife carnivore<br />

host species in South Africa and highlights the ever increasing radiation of rabies into new geographical<br />

areas and wildlife host species including the bat-eared fox.<br />

References<br />

1. Alexander, R.A. 1952. Rabies in South Africa. J. S. Afr. Vet. Med. Assoc. 23.<br />

2. Swanepoel, R., Barnard, B.J.H., Meredith, C.D., Bishop, G.C., Bruchner, G.K., Foggin, C.M. and<br />

Hubschle, O.J.B. 1993. Rabies in southern Africa. Onderstepoort J. Vet. Res. 60, 323-346.<br />

3. Thomson, G.R.and Meredith, C.D. 1993. Rabies in bat-eared foxes in South Africa. Onderstepoort J. Vet.<br />

Res. 60, 399-403.<br />

World Association of Veterinary Laboratory Diagnosticians – 13 th International <strong>Symposium</strong>, Melbourne, Australia, 11-14 November 2007<br />

CANINE RABIES IN SOUTH AFRICA: IDENTIFICATION OF A NEW LINEAGE IN LIMPOPO AND A<br />

RECENT SPREAD INTO THE FREE STATE PROVINCE<br />

Introduction<br />

Chuene Ernest Ngoepe a, * , Gugulethu Zulu a , Claude Sabeta a , Louis Nel b<br />

a Rabies Unit, Onderstepoort Veterinary Institute, Private Bag X05, Onderstepoort, 0110<br />

b University of Pretoria, Microbiology and Plant Pathology, 0002 Pretoria, South Africa<br />

In 2005, there was a drastic and unexpected increase of dog rabies cases in the Limpopo province.<br />

Laboratory confirmed cases increased from 5 in 2004 to 35 in 2005 and to 100 in 2006. The outbreak of<br />

rabies in domestic dogs was followed by a human rabies outbreak in which at least 30 human deaths were<br />

confirmed between 2005 and 2006. In contrast, the Free State province has been historically associated with<br />

endemic rabies in the yellow mongoose Cynictis penicillata (Snyman, 1940; Swanepoel et al. 1993). Due to<br />

spillover events, rabies viruses of the mongoose biotype were recovered from domestic dogs. More recently,<br />

there has been increased number of rabies cases of the canid biotype reported in domestic dogs in this<br />

province. The objectives of this study were; 1. to establish the exact source of infection in the human rabies<br />

cases and the origin of the rabies virus lineage responsible for the recent rabies outbreak in Limpopo, and 2.<br />

to trace the origin of canid rabies and assess the public health threat of mongoose rabies (in dogs) in the<br />

Free State province.<br />

Material & methods<br />

A molecular epidemiological study was therefore performed on a cohort of 98 rabies viruses recovered from<br />

domestic dogs between 1995 and 2007 from the Free State province and 52 rabies viruses recovered from<br />

domestic dogs, jackals and humans from Limpopo and southern Zimbabwe. The cytoplasmic domain of the<br />

glycoprotein and the G-L intergenic region of the rabies viruses in this study sample were amplified and<br />

sequenced. Phylogenetic trees were reconstructed from an alignment of a 592-bp region of the genome<br />

under investigation.<br />

Results<br />

Case 1: Phylogenetic analysis revealed that human rabies viruses were closely related to those obtained<br />

from domestic dogs in the same locality and the rabies viruses from Limpopo were closely related to viruses<br />

obtained from southern Zimbabwe.<br />

Case 2: The phylogenetic analyses segregated the rabies viruses in this study sample into two main<br />

clusters; the genetically compact canid rabies biotype and a second group of heterogeneous viruses of the<br />

mongoose rabies biotype. From the data, it could be demonstrated that viruses of the canid rabies group<br />

were recently introduced into this part of South Africa.<br />

Discussions & conclusions<br />

The reasons for the emergence and rapid dissemination of the new dog rabies strain in Limpopo are not very<br />

clear. It appears that common rabies infection cycles persist between domestic dogs and jackals in southern<br />

Zimbabwe and northern South Africa. It is evident that the new canid group belongs to the same<br />

epidemiological cycle circulating in dogs in both the Free State province and across the international border<br />

with Lesotho. Our results confirm that spillover of the mongoose biotype into domestic dogs lead to dead-end<br />

infections. In comparison to mongoose rabies that is endemic here, canid rabies has emerged to become of<br />

much greater importance to the public and veterinary health sectors of this region.<br />

References<br />

1. Swanepoel, R., Barnard, B. J. H., Meredith, C. D., Bishop, G. C., Bruckner, G. K., Fogging, C. M.,<br />

Hubschle, O. J. B., 1993. Rabies in Southern Africa. Onderstepoort Journal of Veterinary Research, 60: pp.<br />

325-346.<br />

2. Snyman, P.S., 1940. The study and control of vectors of rabies in South Africa. Onderstepoort Journal of<br />

Veterinary Science Animal Husbandry, 66: pp. 296-307<br />

Poster Presentations


www.wavld2007.com<br />

HOSTS GOLD SPONSOR<br />

SILVER SPONSORS<br />

<strong>WAVLD</strong> 2007 SYMPOSIUM MANAGERS<br />

The Meeting Planners 91–97 Islington Street Collingwood Victoria 3066 tel: +61 3 9417 0888 fax: +61 3 9417 0899 email: wavld2007@meetingplanners.com.au

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!