22.11.2013 Views

Abstract Book of EAVLD2012 - eavld congress 2012

Abstract Book of EAVLD2012 - eavld congress 2012

Abstract Book of EAVLD2012 - eavld congress 2012

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

2 nd CONGRESSOFTHEEUROPEAN<br />

ASSOCIATIONOFVETERINARY<br />

LABORATORYDIAGNOSTICIANS(EAVLD)<br />

<br />

EAVLD <strong>2012</strong><br />

<br />

<br />

14July,<strong>2012</strong>,KazimierzDolny,Poland<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

organizedbythe<br />

NationalVeterinaryResearchInstitute,Pulawy


TM marks are owned by IDEXX Laboratories, Inc.<br />

<br />

<br />

BVDV Test<br />

A new diagnostic solution to detect bovine viral<br />

diarrhea virus (BVDV) in just 20 minutes.<br />

EU veterinarians estimate that BVDV costs producers<br />

over €25 per head, even after vaccination. 1 Test all new<br />

introductions with the IDEXX SNAP BVDV Test to identify<br />

persistently infected cattle.<br />

The IDEXX SNAP BVDV Test:<br />

<br />

<br />

<br />

To learn more about the<br />

IDEXX SNAP BVDV Test,<br />

visit idexx.com/snapbvdv.<br />

1. Data on fi le at IDEXX Laboratories, Inc. Westbrook, Maine USA.


Welcome<br />

Dear colleagues,<br />

Welcome to the 2nd EAVLD <strong>congress</strong> in Kazimierz Dolny, hosted by the National Veterinary Research<br />

Institute, Puawy. I am very honoured and proud that our Institute had an opportunity to organize the event.<br />

Nowadays, a constant demand for a higher quality <strong>of</strong> diagnostic services, also a competition on the market<br />

and funding shortages make diagnostic services a hard work. Therefore, the possibility <strong>of</strong> meeting other<br />

specialists in the field <strong>of</strong> veterinary laboratory diagnostics and exchanging <strong>of</strong> eye-to-eye experience are <strong>of</strong><br />

prime importance, which was also marked by the success <strong>of</strong> the 1 st EAVLD <strong>congress</strong>. The sponsors’<br />

response to this <strong>congress</strong> was beyond our expectations and we are pleased that participants will have the<br />

possibility to get acquainted with the variety <strong>of</strong> the latest diagnostic developments during the <strong>congress</strong>.<br />

Despite the economical situation encountered, by the end <strong>of</strong> May, <strong>2012</strong> we had over 170 registered<br />

participants from 26 countries including: Africa, Australia, Europe, North and South America.<br />

Walking in the footsteps <strong>of</strong> the first EAVLD <strong>congress</strong> held in 2010 in Lelystad by the CVI, made our<br />

preparations easier since we had a tremendous and invaluable back-up and advice from Dr. Willie Loeffen,<br />

Secretary <strong>of</strong> the EAVLD and the President <strong>of</strong> the Organizing Committee <strong>of</strong> the EAVLD 2010. At the same<br />

time, we are aware <strong>of</strong> the responsibility and high expectations from the participants due to the positive<br />

feedback from 1 st such an event organized in 2010. Despite recent renovations <strong>of</strong> our Institute and its training<br />

centre both based in Puawy, we had to find another location due to the space limits. We have chosen<br />

Kazimierz Dolny, a town situated nearby Puawy, located at the bank <strong>of</strong> Vistula river. We hope you will<br />

admire town’s beauty and its flavour when visiting different attractions during the <strong>congress</strong> free time.<br />

Myself and the whole Organizing Committee <strong>of</strong> <strong>EAVLD<strong>2012</strong></strong> hope that you will find this <strong>congress</strong> interesting<br />

and valuable source <strong>of</strong> the up-to-date knowledge within the field <strong>of</strong> veterinary laboratory diagnostics.<br />

We wish you all the best and enjoy your stay in Kazimierz Dolny and in Puawy, both scientifically and<br />

socially.<br />

Miroslaw P. Polak<br />

The Organizing Committee<br />

<strong>of</strong> the 2 nd EAVLD Congress<br />

Welcome to the second EAVLD <strong>congress</strong> and to the beautiful town <strong>of</strong> Kazimierz Dolny. I hope that you have<br />

time to explore the area and are looking forward to the Polish hospitality!<br />

The <strong>congress</strong> is being held at a time <strong>of</strong> great economic uncertainty for many countries and individuals. As<br />

veterinary diagnosticians we have a duty to continue to make new scientific advances while also working<br />

more efficiently to help reduced costs to governments and animal owners. We will not be able to do this<br />

unless we share ideas and learn from each other. I want to thank you for coming and supporting this<br />

<strong>congress</strong> and also to all our sponsors for helping to make the <strong>congress</strong> possible. Please make the most <strong>of</strong><br />

the commercial exhibition – which must be the most concentrated gatherings <strong>of</strong> companies involved in<br />

veterinary diagnostics in Europe since our last <strong>congress</strong> in 2010!<br />

Please get involved and come to the General meeting on Tuesday at 16.30. If you have ideas for how the<br />

association may be able to better serve its members please speak up at the General Meeting or discuss with<br />

an EAVLD board member.<br />

I wish to especially thank Miroslaw Pawel Polak and the team at National Veterinary Research Institute for all<br />

their hard work in organising this <strong>congress</strong>.<br />

At this meeting I will hand on the presidency <strong>of</strong> the association to Willie Loeffen who was elected as vice<br />

president at the last <strong>congress</strong> in 2010. I want to thank Willie for all the work he has done as secretary over<br />

the last few years and to wish him all the best for his term <strong>of</strong> <strong>of</strong>fice. He will have my full support and I know<br />

that he will do an excellent job as your new EAVLD President.<br />

Andrew Soldan<br />

EAVLD President


KEYNOTE SPEAKERS<br />

General Session<br />

Pr<strong>of</strong>. Dr. Wim H.M. van der Poel<br />

Pr<strong>of</strong>. Dr. Wim H.M. van der Poel, DVM is senior scientist at the Central Veterinary Institute <strong>of</strong><br />

Wageningen University and Research Centre, The Netherlands. His research activities have<br />

focussed on veterinary virology, emerging and zoonotic viruses and food borne viruses. He is<br />

also coordinator <strong>of</strong> the EPIZONE European Research Group, the network on epizootic<br />

animal diseases research. Previously, from 1996 to 2004, pr<strong>of</strong> Van der Poel has been<br />

working on research <strong>of</strong> viral zoonoses and food borne viruses at the National Institute for<br />

Public Health and the Environment in Bilthoven, The Netherlands. At this institute, from<br />

2000-2004, he was also heading the National Reference Laboratory for Microbiological<br />

Contamination <strong>of</strong> Bivalve Molluscs. Pr<strong>of</strong>. Van der Poel is a member <strong>of</strong> a number <strong>of</strong> scientific<br />

committees, boards and pr<strong>of</strong>essional bodies in the field <strong>of</strong> veterinary virology, and he has<br />

been involved in many national and international research projects on viruses in animals and<br />

foodstuffs. Throughout his career he has published a large number scientific papers and<br />

reviews in this area and he has <strong>of</strong>ten served as an ad hoc reviewer for scientific journals and<br />

granting agencies. In February 2009 he has accepted an honorary visiting chair on Emerging<br />

and Zoonotic Viruses at the University <strong>of</strong> Liverpool, United Kingdom.<br />

Pr<strong>of</strong>. van der Poel graduated in Veterinary Medicine in 1988 and completed his PhD in<br />

veterinary virology at the Utrecht University in 1995. He is a registered specialist in veterinary<br />

microbiology within the Netherlands Royal Veterinary Association and a registered research<br />

worker in medical microbiology within the Netherlands Royal Microbiology Association.<br />

Pr<strong>of</strong>. Dr. med. vet. Stefan Schwarz<br />

works in the Institute <strong>of</strong> Farm Animal Genetics <strong>of</strong> the Friedrich-Loeffler-Institut (= Federal<br />

Research Institute for Animal Health) in Neustadt-Mariensee, Germany. He heads the<br />

research group ‘Molecular Microbiology and Antimicrobial Resistance’ and is involved in both<br />

surveillance <strong>of</strong> antimicrobial resistance and analysis <strong>of</strong> the molecular genetics <strong>of</strong><br />

antimicrobial resistance. He also teaches various academic courses at the University <strong>of</strong><br />

Veterinary Medicine Hannover. Pr<strong>of</strong>. Schwarz is a specialist veterinarian in (a) microbiology,<br />

(b) epidemiology, and (c) molecular genetics and gene technology. He acts as editor/<br />

editorial board member <strong>of</strong> six international journals.<br />

Diagnostics at the point <strong>of</strong> interest<br />

Andrew Soldan<br />

qualified as a veterinary surgeon in 1984 from the Royal Veterinary College, University <strong>of</strong><br />

London and then worked in general mixed practice in Yorkshire, UK for 2 years. Following<br />

the completion <strong>of</strong> an MSc in Tropical Veterinary Medicine at the University <strong>of</strong> Edinburgh he<br />

working in Malawi for 5 years and helped set up an epidemiology unit. During this time he<br />

specialised in laboratory diagnosis <strong>of</strong> disease and control methods for tick borne disease in<br />

local cattle. For the latter work he was awarded a Doctorate <strong>of</strong> Veterinary Medicine by the<br />

University <strong>of</strong> Edinburgh. On return from Africa he worked as a government veterinary<br />

investigation <strong>of</strong>ficer before another 3 year spell in mixed practice. In 1999 he started work<br />

for the Veterinary Laboratories Agency and was heavily involved in the laboratory response<br />

to the CSF outbreak in 2000 and the FMD outbreak in 2001. He was head <strong>of</strong> testing for the<br />

VLA for 5 years, was Veterinary Director for 1 year and is currently Commercial Director and<br />

Head <strong>of</strong> AHVLA Scientific for the Animal Health and Veterinary Laboratories Agency<br />

following its formation in early 2011. Andrew has been involved in the development <strong>of</strong> new<br />

tests and bringing them into routine use in an ISO17025 accredited laboratory. He has also


worked with many diagnostic companies to ensure that new tests developed at the VLA have<br />

been available to international laboratories and end users. He has taken a special interest in<br />

tests that can be used outside a specialist laboratory - possibly informed by his time working<br />

in Africa. Andrew was a founding member <strong>of</strong> EAVLA and its first president.<br />

Emerging, re-emerging and wildlife diseases - diagnostic<br />

possibilities<br />

Dolores Gavier-Widén<br />

is a veterinary pathologist, Head <strong>of</strong> the Research and Development Division, Department <strong>of</strong><br />

Pathology and Wildlife Diseases, National Veterinary Institute (SVA) and Associated<br />

pr<strong>of</strong>essor at the Swedish University <strong>of</strong> Agricultural Sciences, Uppsala, Sweden. She<br />

obtained her veterinary degree at Buenos Aires University, Argentina, and her MS and PhD<br />

degrees at the University <strong>of</strong> California, Davis. She has worked on diagnosis in combination<br />

with applied research, in particular on infectious diseases <strong>of</strong> wild animals, for more than 25<br />

years, most <strong>of</strong> the time in Sweden and including 40 months <strong>of</strong> work at the Veterinary<br />

Laboratory Agency, Weybridge, England. She is the president <strong>of</strong> the Wildlife Disease<br />

Association (WDA) (www.wildifedisease.org) and past president <strong>of</strong> its Section, the European<br />

WDA. She has participated <strong>of</strong> several EU FP6, FP7 and LIFE projects, such as the current<br />

WildTech project, and also <strong>of</strong> several working groups <strong>of</strong> the European Food Safety Authority<br />

(EFSA) and <strong>of</strong> the OIE ad hoc Group on the validation on diagnostic tests for wildlife, in<br />

2011. She has contributed to the organization <strong>of</strong> several WDA, EWDA and other<br />

conferences, such as Cutting Edge Pathology (European Society <strong>of</strong> Veterinary Pathology), in<br />

2011. Dolores believes that surveillance <strong>of</strong> infectious diseases <strong>of</strong> wildlife is essential to<br />

improve and maintain the health <strong>of</strong> humans, domestic and wild animals.<br />

New techniques in bacteriology, parasitology and pathology<br />

Dr. Markus Kostrzewa<br />

In 1990 Dr. Kostrzewa obtained the Diploma in Biology, University Gießen, Germany, then in<br />

1993 he obtained the title <strong>of</strong> Dr. rer. nat, University Gießen, Germany. He joined Bruker in<br />

1998, where he started as project manager for DNA analysis by MALDI-TOF mass<br />

spectrometry. Later he became the Head <strong>of</strong> development <strong>of</strong> Clincal Proteomics (“ClinProt”<br />

system, a first mass spectrometry protein pr<strong>of</strong>iling system).<br />

Since 1999 he works in the field <strong>of</strong> MALDI-TOF MS in microbiology. Dr. Kostrzewa initiated<br />

and directed the development <strong>of</strong> microorganism identification by MALDI-TOF mass<br />

spectrometry (“MALDI Biotyper” system). He is heading development <strong>of</strong> clinical applications<br />

for mass spectrometry. Since 2005 he is the Director <strong>of</strong> Molecular Biology R&D at Bruker<br />

Daltonics and since <strong>2012</strong> Dr. Kostrzewa is the Vice President <strong>of</strong> Clinical Mass Spectrometry,<br />

R&D at Bruker Daltonics.


Sensitive and reliable animal pathogen detection<br />

QIAGEN veterinary testing solutions<br />

Subheadline optional<br />

can run over 1, 2 or 3 lines.<br />

QuantiFast® Pathogen +IC Kits provide versatile and sensitive detection.<br />

<br />

<br />

<br />

Simultaneous detection <strong>of</strong> viral RNA and DNA<br />

Internal Control for certainty in result interpretation<br />

Universal protocol for standard and fast cyclers<br />

Learn more at www.qiagen.com/QuantiFast<br />

For up-to-date trademarks and disclaimers, see www.qiagen.com.<br />

Sample & Assay Technologies


Products and services<br />

for scientists around the world<br />

The Animal Health and Veterinary Laboratories Agency has developed products and services<br />

and makes them available to organisations around the world, through AHVLA Scientific.<br />

For veterinary laboratories these include:<br />

Specialist laboratory reagents and diagnostic kits<br />

Extensive range <strong>of</strong> pr<strong>of</strong>iciency testing schemes<br />

Comprehensive bio-imaging service<br />

Unique cell characterisation service<br />

Internationally recognised R & D capability<br />

Laboratory testing services<br />

If you require further information on any products and services,<br />

please contact our Customer Service Team at<br />

ahvlascientific@ahvla.gsi.gov.uk or visit www.ahvlascientific.com


ScientificProgram<br />

DAY1<br />

17:0020:00<br />

19:0021:00<br />

DAY2<br />

08:0010:00<br />

09:0009:30<br />

09:3010:15<br />

10:1510.45<br />

10:4512:15<br />

12:3014:00<br />

14:0014:45<br />

14:4516.15<br />

16:1516.45<br />

<br />

Sunday,1July<strong>2012</strong><br />

RegistrationdeskinKrolKazimierzHotel(KKH)<br />

Welcomedrink(patio<strong>of</strong>KKH)<br />

<br />

Monday,2July<strong>2012</strong><br />

Registrationdesk(KKH)<br />

OpeningCeremony(lecturehall<strong>of</strong>KKH)<br />

1 st session[S1.]:GeneralSession<br />

(allaspects<strong>of</strong>veterinarydiagnosis&drugresistanceissues)<br />

lecturehall<strong>of</strong>KKH(09:3018:15)<br />

[S1.]Keynotelecture:Theemergence<strong>of</strong>Schmallenbergvirus–Howtorespondto<br />

newepizooticsinEurope?WimVanderPoel,DVM,PhD,CentralVeterinary<br />

Institute,WageningenUniversityandResearchCentre,Department<strong>of</strong>Virology,P.O.Box65,8200<br />

ABLelystad,Edelhertweg15,8219PHLelystad,TheNetherlands<br />

C<strong>of</strong>feebreak+postersession+sponsors’exhibitions(patio<strong>of</strong>KKH)<br />

[S1.]Oralpresentations(6)<br />

1. Oralfluids–samplematrixforeffectiveherdhealthmonitoring.ChristinaBoss,Life<br />

Technologies,Germany.<br />

2. Diagnosticproperties<strong>of</strong>severalPRRSantibodyELISAsinthecontext<strong>of</strong>a<br />

longitudinalstudy<strong>of</strong>afarmusingdifferentvaccinationstrategies.KatrinStrutzberg<br />

Minder,IVDGmbH(IVDInnovativeVeterinaryDiagnostics),Germany.<br />

3. Introductionrate<strong>of</strong>alowpathogenicavianinfluenzavirusinfectionindifferent<br />

Dutchpoultrysectors.RuthBouwstra,CentralVeterinaryInstitute(CVILelystad),<br />

TheNetherlands.<br />

4. AduplexonesteprealtimeRTPCRfordiagnosis<strong>of</strong>footandmouthdisease.Kamila<br />

Górna,ANSES,France.<br />

5. Validation<strong>of</strong>acompetitiveELISAforthedetection<strong>of</strong>antibodiesantip26<strong>of</strong>equine<br />

infectiousanemiavirusinequinesera.RobertoNardini,IZSLAZIOETOSCANA,Italy.<br />

6. Diagnosticperformance<strong>of</strong>acommercialPRRSserumantibodyELISAadaptedtooral<br />

fluidspecimens:fieldsamples.AndreaBallagi,IDEXXLaboratories,Sweden.<br />

Lunch(restaurant<strong>of</strong>KKH)+postersession+sponsors’exhibitions(patio<strong>of</strong>KKH)<br />

[S1.]Keynotelecture:Assessingtheantimicrobialsusceptibility<strong>of</strong>bacteria<strong>of</strong><br />

animaloriginsomebasicconsiderations.Pr<strong>of</strong>.Dr.StefanSchwarz,Institutfür<br />

NutztiergenetikFriedrichLoefflerInstitut(FLI),Höltystr.10,D31535NeustadtMariensee,Germany<br />

[S1.]Oralpresentations(6)<br />

7. Subtyping<strong>of</strong>swineinfluenzavirusesbymultiplexrealtimePCR.KeesvanMaanen,<br />

GDAnimalHealthService,TheNetherlands.<br />

8. Importance<strong>of</strong>continuousvalidation<strong>of</strong>molecularmethodsforroutinediagnosis<strong>of</strong><br />

PRRSVRNAinclinicalsamples.SandraRevillaFernández,AustrianAgencyfor<br />

HealthandFoodSafety/InstituteforVeterinaryDiseaseControlMödling,Austria.<br />

9. Viraldiagnosisusingtransmissionelectronmicroscopy.WilliamCooley,AHVLA,<br />

UnitedKingdom.<br />

10. Abeadbasedmultipleximmun<strong>of</strong>luorometricassayforscreeningandconfirmation<strong>of</strong><br />

allmajorpriontypesinsheep.JanLangeveld,CentralVeterinayInstitute<strong>of</strong><br />

WageningenUR,TheNetherlands.<br />

11. Usingoralfluidfortheserologicalmonitorization<strong>of</strong>PRRSVcirculationinagroup<strong>of</strong><br />

infectedgilts.XavierRebordosaTrigueros,HIPRA,Spain.<br />

12. Enhanceddetection<strong>of</strong>bovinerespiratoryvirusesbyinclusion<strong>of</strong>cohortanimals.<br />

RonanO’Neill,DAFMLaboratories,CentralVeterinaryResearchLaboratory,Ireland<br />

C<strong>of</strong>feebreak+postersession+sponsors’exhibitions(patio<strong>of</strong>KKH)


16:4518.15<br />

20:0023:00<br />

[S1.]Oralpresentations(6)<br />

13. Thefirstyear<strong>of</strong>obligatoryBVDcontrolinGermany–diagnosticstrategies,results<br />

andexperiences.HorstSchirrmeier,FriedrichLoefflerInstitut,Institute<strong>of</strong><br />

DiagnosticVirology,Germany.<br />

14. EuropeanPRRSv–diagnosticsolutionsforarapidlymutatingvirus.ChristinaBoss,<br />

LifeTechnologies,Germany.<br />

15. Serologicalanalysisandmonitoring<strong>of</strong>IBR.IsitpossibletocontrolIBRgEantibodies<br />

inabulktankmilk?LourdesPorquetGaranto,HIPRA,Spain.<br />

16. Developmentandvalidation<strong>of</strong>arealtimePCRzengelmixforthediagnosisand<br />

quantification<strong>of</strong>CoxiellaBurnetii.AleidaVillaEspinosa,EXOPOL,SL.Autovacunasy<br />

Diagnóstico,Spain.<br />

17. Ringtestevaluationforthedetection<strong>of</strong>PRRSVantibodiesinoralfluidspecimens<br />

usingacommercialPRRSVserumantibodyELISA.AndreaBallagi,IDEXX<br />

Laboratories,Sweden.<br />

18. RealtimePCR,MycoplasmaGallisepticum,Mycoplasmasynoviae,Mycoplasma<br />

meleagridis.AndréFuchs,IDEXXLivestockandPoultryDiagnostics,U.S.A.<br />

Barbecuepartyinthegarden<strong>of</strong>theNationalVeterinaryResearchInstitutein<br />

Pulawy(transportbybusestoandfromKazimierzDolnywillbeprovidedbythe<br />

organizer–buseswillleavefromKKHat19:40)


Day3<br />

09:0009:45<br />

9:4510:45<br />

10:4511.15<br />

Tuesday,3July<strong>2012</strong><br />

2 nd session[S2.]:Diagnosticsatthepoint<strong>of</strong>interest<br />

(pensitetests,SNAPtestsandhomemadetestsusedbeyondthelabbencharea)<br />

lecturehall<strong>of</strong>KKH(09:0010:45)<br />

[S2.]Keynotelecture:Whendoyouwanttheresult?Howmuchdoyouwanttopay!<br />

AndrewSoldan,DVM,PhD,Head<strong>of</strong>AHVLAScientific,AHVLA–Weybridge,WoodhamLane,<br />

NewHaw,Addlestone,SurreyKT153NB,UnitedKingdom<br />

[S2.]Oralpresentations(4)<br />

1. Development<strong>of</strong>arapidisothermalassaytodetectTaylorellaequigenitalis,the<br />

causativeagent<strong>of</strong>contagiousequinemetritis.SarahNorth,AHVLAWeybridge,<br />

TechnologyTransferUnit,SpecialistScientificSupport,UnitedKingdom.<br />

2. Laboratoryvalidation<strong>of</strong>animmunochromatographictestfortherapiddetection<strong>of</strong>koi<br />

herpesvirus(cyhv3)ingillswabs.RobertVrancken,OkapiSciencesN.V.,Belgium.<br />

3. Evaluation<strong>of</strong>rapidHRSVstriptestsfordetection<strong>of</strong>bovinerespiratorysyncytialvirus.<br />

WojciechSocha,Department<strong>of</strong>Virology,NVRIPulawy,Poland.<br />

4. Evaluation<strong>of</strong>Alatexagglutinationtestfortheidentification<strong>of</strong>Clostridiumdifficile<strong>of</strong><br />

porcineorigin.LauraVallsVila,Hipra,DiagnosticCenter,Girona,Spain.<br />

C<strong>of</strong>feebreak+postersession+sponsors’exhibitions(patio<strong>of</strong>KKH)<br />

3 rd session[S3.]Emerging,reemergingandwildlifediseasesdiagnosticpossibilities<br />

(detectiontoolsbasedoncommercialandhomemadetests,theirperformanceandvalidationinthelab)<br />

lecturehall<strong>of</strong>KKH(11:1516:30)<br />

11:1512:00<br />

12:0013:00<br />

13:0014:30<br />

14:3016:15<br />

16:3018:00<br />

20:0022:00<br />

[S3.]Keynotelecture:Emergingandreemergingwildlifediseases:pathologyand<br />

relatedtechniquesfordiagnosticsandforgeneralandtargetedsurveillance.<br />

DoloresGavierWidén,DVM,MS,PhD,AssociatePr<strong>of</strong>.,Department<strong>of</strong>Pathologyand<br />

WildlifeDiseases,DeputyHead<strong>of</strong>Dept.,HeadR&DDivision,NationalVeterinaryInstitute(SVA),<br />

SE75189Uppsala,Sweden<br />

[S3.]Oralpresentations(4)<br />

1. SchmallenbergvirusoutbreakinTheNetherlands:Routinediagnosticsandtestresults.<br />

RuthBouwstra,CentralVeterinaryInstitute(CVILelystad),TheNetherlands.<br />

2. 25years<strong>of</strong>passivesurveillance<strong>of</strong>batsintheNetherlands.Molecularepidemiology<br />

andevolution<strong>of</strong>EBLV1.BartKooi,CentralVeterinaryInstitute<strong>of</strong>WageningenUR–<br />

Lelystad,TheNetherlands.<br />

3. Diagnosis<strong>of</strong>QfeverinDairyCattlebyPhasespecificMilkSerology.JensBöttcher,<br />

BavarianAnimalHealthService,Germany<br />

4. EQUINEnocardi<strong>of</strong>ormplacentitis&abortionoutbreakandfarmbasedriskfactor<br />

study,20102011.CraigN.Carter,University<strong>of</strong>Kentucky,U.S.A.<br />

Lunch(restaurant<strong>of</strong>KKH)+postersession+sponsors’exhibitions(patio<strong>of</strong>KKH)<br />

[S3.]Oralpresentations(7)<br />

5. AnewdiagnostictoolforbovinetuberculosisIDEXXM.bovisantibodytestkit.<br />

ChristophEgli,IDEXXLabs,Inc.,U.S.A.<br />

6. Detection<strong>of</strong>SchmallenbergvirusintheUK.AnnaLaRocca,AnimalHealthand<br />

VeterinaryLaboratories(AHVLA),UnitedKingdom.<br />

7. Detection<strong>of</strong>WNVenzooticcirculationinhorseswithneurologicalsignsandincaptive<br />

sentinelchickensintheprefecture<strong>of</strong>Thessaloniki,Greece.SerafeimC.Chaintoutis,<br />

Faculty<strong>of</strong>VeterinaryMedicine,AristotleUniversity<strong>of</strong>Thessaloniki,Greece.<br />

8. Schmallenbergvirus:serologicalstudiesinGermanholdings.HorstSchirrmeier,<br />

FriedrichLoefflerInstitut,Institute<strong>of</strong>DiagnosticVirology,Germany.<br />

9. Differentdiagnostictoolsforabroadrange<strong>of</strong>macavirusesandtheirreservoirand<br />

susceptiblehosts.ChristineFoerster.Institute<strong>of</strong>virology,Diagnosticlaboratory,<br />

Germany.<br />

10. Diagnosticaspects<strong>of</strong>Suidherpesvirus1infectioninwildboar.AdolfSteinrigl,AGES<br />

InstituteforVeterinaryDiseaseControl,Austria.<br />

11. Preliminaryvalidation<strong>of</strong>theIDScreen®SchmallenbergvirusIndirectELISA.Philippe<br />

Pourquier,IDVet,France.<br />

GeneralMeeting<strong>of</strong>EAVLD(EAVLDmembersonly)<br />

Galadinner(restaurant<strong>of</strong>KKH)


Day4<br />

Wednesday,4July<strong>2012</strong><br />

4 th session[S4.]Newtechniquesinbacteriology,parasitologyandpathology<br />

(newadvancesindiagnostictechniques)<br />

lecturehall<strong>of</strong>KKH(09:0011:15)<br />

09:0009:45 [S4.]Keynotelecture:MALDITOFandothernewdiagnostictechniques.Dr.Markus<br />

Kostrzewa,Director<strong>of</strong>MolecularBiology,R&D,BrukerDaltonikGmbH,Fahrenheitstr.4,D28359<br />

Bremen,Germany<br />

09:4511:15 [S4.]Oralpresentations(6)<br />

1. Suitability<strong>of</strong>recombinantproteinsforthediagnosis<strong>of</strong>leptospirosisinpigs.Ulrike<br />

Ripp,Synlab.vetLeipzig,Germany.<br />

2. BiologgenerationIII,matrixassistedlaserdesorption/ionisationtime<strong>of</strong>flight(MALDI<br />

TOF)massspectrometryand16srRNAgenesequencingfortheidentification<strong>of</strong><br />

bacteria<strong>of</strong>veterinaryinterest.PeterWragg,AnimalHealthandVeterinary<br />

LaboratoriesAgency(Weybridge),UnitedKingdom.<br />

3. Evaluation<strong>of</strong>thediagnosticperformance<strong>of</strong>peptidecocktailsintheinterferongamma<br />

assayfordiagnosis<strong>of</strong>tuberculosisincattle.AlexRaeber,PrionicsAG,Switzerland.<br />

4. Matrixassistedlaserdesorptionionizationtime<strong>of</strong>flightmassspectrometry(MALDI<br />

TOFMS)inaveterinarydiagnosticlaboratory.AnnetHeuvelink,GDAnimalHealth<br />

Service,TheNetherlands.<br />

5. Rapididentification<strong>of</strong>bovinemastitispathogensusingMALDITOF.GudrunOveresch,<br />

Institute<strong>of</strong>VeterinaryBacteriology,University<strong>of</strong>Bern,Switzerland.<br />

6. 15minuteELISAusingalowcostcommercialbiosensor.JasonSawyer,AnimalHealth<br />

andVeterinaryLaboratoriesAgency(Weybridge),UnitedKingdom.<br />

11:1511:45 Closingceremony,EAVLD2014<br />

11:4513:00 Lunch(restaurant<strong>of</strong>KKH)+postersession+sponsors’exhibitions(patio<strong>of</strong>KKH)<br />

13:00 End<strong>of</strong>theCongress<br />

(shuttlebusestoWarsawAirport)


GD Animal Health Service<br />

Custom made<br />

animal health solutions<br />

GD Pr<strong>of</strong>iciency testing schemes<br />

R&D<br />

Veterinary laboratory<br />

GD Diagnostics<br />

Consultancy<br />

www.gddeventer.com


Oral presentations<br />

“General Session”<br />

(1 st session)


S1 - K - 01<br />

THE EMERGENCE OF SCHMALLENBERG VIRUS – HOW TO RESPOND TO NEW EPIZOOTICS IN<br />

EUROPE<br />

Wim H. M. van der Poel<br />

Central Veterinary Institute <strong>of</strong> Wageningen University and Research Centre, Lelystad, The Netherlands, Tel. +31320238383, email:<br />

wim.vanderpoel@wur.nl<br />

Schmallenberg virus, epizootic, sheep, cattle, goat<br />

The emergence <strong>of</strong> Schmallenberg virus<br />

Schmallenberg virus was discovered in November 2011,<br />

and named after the village in Germany where it was<br />

first detected in blood samples from a dairy herd (1).<br />

The provisionally named “Schmallenberg virus” is an<br />

enveloped, negative-sense, segmented, single-stranded<br />

RNA virus. It belongs to the Bunyaviridae family, within<br />

the Orthobunyavirus genus. Schmallenberg virus is<br />

related to the Simbu serogroup viruses, which also<br />

includes ruminant viruses like Shamonda, Akabane,<br />

Sathuperi, Douglas and Aino virus. Based on what is<br />

already known about the genetically related Simbu<br />

serogroup viruses, Schmallenberg virus affects domestic<br />

ruminants. At its first occurrence in dairy cattle in both<br />

Germany and The Netherlands Schmallenberg virus<br />

infections presented with fever and reduced milk yield, in<br />

the Netherlands also severe diarrhoea (2). In early<br />

December 2011, congenital malformations were reported<br />

in new-born lambs in the Netherlands, and<br />

Schmallenberg virus was detected in and isolated from<br />

the brain tissue. Thereafter the virus was also detected<br />

in malformed calves and goat kids. Gross pathology in<br />

malformed animals and stillbirths (calves, lambs, kids)<br />

included arthrogryposis, hydrocephaly, brachygnathia<br />

inferior, ankylosis, torticollis, scoliosis, hydranencephaly,<br />

hypoplasia <strong>of</strong> the central nervous system, porencephaly<br />

and subcutaneous oedema (3). The symptoms can be<br />

summarised as arthrogryposis and hydranencephaly<br />

syndrome (AHS). The spatial and temporal distribution<br />

suggests that the disease is first transmitted by insect<br />

vectors, in particular culicoides spp and then vertically in<br />

utero. The detection <strong>of</strong> SBV in midges (culicoides spp) in<br />

several countries supports this assumption.<br />

The emergence <strong>of</strong> Schmallenberg virus in Europe<br />

resulted in a rapid increase <strong>of</strong> malformations in new<br />

borne lambs and later on calves. In spring <strong>2012</strong> the<br />

numbers <strong>of</strong> cases started to decrease, first in sheep and<br />

then in cattle. By May <strong>2012</strong> Schmallenberg virus had<br />

affected farms in at least 8 countries in Western Europe,<br />

and although the epidemic curve seemed to come to an<br />

end by May <strong>2012</strong> a further spread over Europe still is<br />

likely (4). Schmallenberg virus clearly causes severe<br />

disease in ruminants and as a result economic losses<br />

which may be enhanced by trade restrictions. As the<br />

family <strong>of</strong> Bunyaviridae contains several important<br />

zoonoses, studies were performed to elucidate its<br />

zoonotic potential. In a rapid risk assessment in Dec<br />

2011 it was concluded that human infections were<br />

unlikely but could not be excluded. Therefore both in the<br />

Netherlands and Germany serosurveys in the human<br />

population were performed. In the Netherlands 301<br />

persons exposed to SBV, farmers and veterinarians,<br />

were tested and in North Rhine-Westphalia 60 cattle and<br />

sheep farmers were tested. None <strong>of</strong> the tested<br />

individuals showed antibody to SBV and it was concluded<br />

that there is no evidence for zoonotic infection (5).<br />

How to respond to new epizootics in Europe<br />

The “Schmallenberg virus experience” again has shown<br />

that the introduction <strong>of</strong> a completely new virus on the<br />

continent can encompass an important threat to animal<br />

health and public health. Moreover, continued changes in<br />

human and animal demography, coupled with<br />

environmental changes and changes within a virus itself<br />

make it likely that the trend for increased viral disease<br />

emergence will continue.<br />

Strategies to improve veterinary and public health<br />

protection with regard to emerging pathogens have<br />

focused towards improved surveillance. Improved<br />

detection <strong>of</strong> viruses in reservoirs, early disease outbreak<br />

detection, or broadly based research to clarify important<br />

factors that favour (re-)emergence. In order to recognize<br />

and combat viral diseases, it is pivotal to understand the<br />

epidemiology <strong>of</strong> these infections. We need to know the<br />

pathogen, its vertebrate hosts and the methods <strong>of</strong><br />

transmission between these hosts. This should be<br />

coupled with knowledge <strong>of</strong> spatio-temporal disease<br />

patterns together with changes over time. Together,<br />

these can be used to build a picture <strong>of</strong> the dynamic<br />

processes involved in virus transmission that can be<br />

used to account for observed disease patterns and<br />

ultimately to forecast spread and establishment into new<br />

areas.<br />

Longitudinal veterinary surveillance should include food<br />

producing animals as well as wildlife and also insect<br />

vectors should be considered. A main goal <strong>of</strong> infectious<br />

disease surveillance is the early detection <strong>of</strong> new<br />

emerging pathogens. For this we will primarily be<br />

dependant <strong>of</strong> clinicians and laboratories testing field<br />

samples from potential reservoirs. Case reports will have<br />

to be generated and combined to early detect new<br />

emerging pathogens. Electronic systems, preferably<br />

web-based, could be very helpful to achieve this. In<br />

addition, improved detection may also be achieved<br />

through use <strong>of</strong> syndromic approaches. Syndromic<br />

surveillance, which collects non-specific syndromes<br />

before diagnosis, has great advantages in promoting the<br />

early detection <strong>of</strong> new emerging diseases before disease<br />

confirmation. By combining syndromic surveillance with<br />

case report surveillance in online reporting systems, a<br />

sensitive early detection system for new emerging<br />

diseases could be build.<br />

Novel molecular methods, for example DNA microarrays<br />

and whole genome approaches <strong>of</strong>fer unprecedented<br />

opportunities for rapid detection but these require<br />

significant optimisation and validation before they can be<br />

deployed broadly. Also due to costs limitations, the rapid<br />

detection <strong>of</strong> a new virus will only be feasible by<br />

employing the different molecular techniques, including<br />

microarray, (RT)-PCR and whole genome sequencing, in<br />

a sensible combination. By applying molecular<br />

approaches, positive detections <strong>of</strong> a lot <strong>of</strong> different<br />

pathogens in a lot <strong>of</strong> different samples have been<br />

performed. However, it is much more difficult to pro<strong>of</strong><br />

causation. The agent should be present at high<br />

concentrations and seroconversion should be<br />

demonstrated. Confidence in a causal relationship<br />

between a candidate pathogen and a disease is<br />

enhanced by fulfilment <strong>of</strong> Kochs’ Postulates (i.e.<br />

demonstration <strong>of</strong> the presence <strong>of</strong> an agent in all cases <strong>of</strong><br />

a disease and not in the absence <strong>of</strong> disease, replication<br />

<strong>of</strong> disease following ex vivo cultivation and introduction<br />

into a naïve host); however, this will not always be<br />

feasible. Apart from the fact that this can be extremely<br />

time-consuming, some viruses cannot be cultured and<br />

experimental infection can be extremely difficult.<br />

Prompt detection and instigation <strong>of</strong> control measures<br />

such as vaccination are crucial to prevent spread. Cloned


antigens or attenuated vaccines can be modified into the<br />

appropriate antigenic forms and in the case <strong>of</strong><br />

arboviruses, approaches targeting replication in the<br />

arthropod vector, or the vectors themselves, may <strong>of</strong>fer<br />

substantial benefit for control. However, the<br />

development <strong>of</strong> a safe and efficacious vaccine including<br />

its registration for use is very time-consuming. Moreover<br />

history has learned that it is extremely difficult to swiftly<br />

design a new reactive vaccination strategy, which means<br />

that in the first phase <strong>of</strong> every new epidemic we will<br />

have to rely on biocontainment and biosecurity<br />

measurements.<br />

National and International cooperation can be helpful to<br />

early identify new pathogens and will be needed to<br />

develop innovative and rapid control strategies to<br />

combat emerging diseases. Therefore this should be<br />

stimulated as much as possible. Knowledge exchange<br />

and research networking should become a commonplace.<br />

In addition the One Health approach, involving inclusive<br />

collaborations between physicians, veterinarians and<br />

other health and environmental pr<strong>of</strong>essionals, will be<br />

more and more important to combat emerging viral<br />

disease outbreaks. National authorities as well as<br />

international organisations like WHO, OIE, FAO and also<br />

ECDC therefore should actively support such cooperative<br />

initiatives to achieve an optimal response to new<br />

epizootics in Europe.<br />

References<br />

1. H<strong>of</strong>fmann B, Scheuch M, Höper D, Jungblut R, Holsteg M,<br />

Schirrmeier H, Eschbaumer M, Goller KV, Wernike K, Fischer M,<br />

Breithaupt A, Mettenleiter TC, Beer M (<strong>2012</strong>) Novel<br />

orthobunyavirus in Cattle, Europe, 2011. Emerg Infect Dis. 18:<br />

469-472.<br />

2. Muskens J, Smolenaars AJ, van der Poel WH, Mars MH, van<br />

Wuijckhuise L, Holzhauer M, van Weering H, Kock P (<strong>2012</strong>)<br />

Diarrhea and loss <strong>of</strong> production on Dutch dairy farms caused by<br />

the Schmallenberg virus. Tijdschr Diergeneeskd. 137:112-115.<br />

3. Van den Brom R, Luttikholt SJ, Lievaart-Peterson K,<br />

Peperkamp NH, Mars MH, van der Poel WH, Vellema P (<strong>2012</strong>)<br />

Epizootic <strong>of</strong> ovine congenital malformations associated with<br />

Schmallenberg virus infection. Tijdschr Diergeneeskd. 137:106-<br />

111.<br />

4. Armin R.W. Elbers , Willie L.A. Loeffen, Sjaak Quak, Els de<br />

Boer-Luijtze, Arco N. van der Spek, Ruth Bouwstra, Riks Maas,<br />

Marcel A.H. Spierenburg, Eric P. de Kluijver, Gerdien van Schaik,<br />

Wim H.M. van der Poel (<strong>2012</strong>) Seroprevalence <strong>of</strong> Schmallenberg<br />

Virus Antibodies among Dairy Cattle, the Netherlands, Winter<br />

2011–<strong>2012</strong>. Emerging Infectious Diseases 18:..<br />

5 .Chantal Reusken, Cees van den Wijngaard, Paul van Beek,<br />

Martin Beer, Gert-Jan Godeke, Leslie Isken, Hans van den<br />

Kerkh<strong>of</strong>, Wilfrid van Pelt , Wim van der Poel, Johan Reimerink,<br />

Peter Schielen, Jonas Schmidt-Chanasit, Piet Vellema, Ankje de<br />

Vries, Inge Wouters and Marion Koopmans (<strong>2012</strong>) Public health<br />

response to an emerging arbovirus: the case <strong>of</strong> Schmallenberg<br />

virus (submitted for publication).


S1 - K - 02<br />

ASSESSING THE ANTIMICROBIAL SUSCEPTIBILITY OF BACTERIA OBTAINED FROM ANIMALS<br />

Stefan Schwarz 1 , Peter Silley 2,3 , Shabbir Simjee 4 , Neil Woodford 5 , Engeline van Duijkeren 6 , Alan P. Johnson 7<br />

and Wim Gaastra 8<br />

1<br />

Institute <strong>of</strong> Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt-Mariensee, Germany<br />

2<br />

MB Consult Limited, Lymington, UK<br />

3<br />

Department <strong>of</strong> Biomedical Sciences, University <strong>of</strong> Bradford, Bradford, UK<br />

4 Elanco Animal Health, Basingstoke, UK<br />

5 Antibiotic Resistance Monitoring and Reference Laboratory, Centre for Infections, Health Protection Agency, London, UK<br />

6<br />

Centre for Infectious Disease Control Netherlands (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands<br />

7 Department <strong>of</strong> Healthcare-associated Infections and Antimicrobial Resistance, Centre for Infections, Health Protection Agency, London, UK<br />

8<br />

Department <strong>of</strong> Infectious Diseases and Immunology, Faculty <strong>of</strong> Veterinary Medicine, Utrecht University, Utrecht, The Netherlands<br />

Antimicrobial susceptibility testing, interpretive criteria, MIC 50 , MIC 90 , multi-resistance<br />

Introduction<br />

In recent years, antimicrobial resistance in bacteria <strong>of</strong> animal<br />

origin, including food-producing animals, pet and companion<br />

animals, fish and other aquatic animals as well as wild animals,<br />

has gained particular attention. Consequently, an increasing<br />

number <strong>of</strong> studies that include antimicrobial susceptibility testing<br />

have been published. However, an analysis <strong>of</strong> recently published<br />

articles revealed a number <strong>of</strong> frequently occurring shortcomings,<br />

which may have an impact either directly on the quality <strong>of</strong> the<br />

results obtained or on the conclusions drawn. An editorial has<br />

been published simultaneously in Veterinary Microbiology (1) and<br />

in the Journal <strong>of</strong> Antimicrobial Chemotherapy (2) and is intended<br />

to highlight the major pitfalls and provide guidance for authors,<br />

and reviewers on the correct performance <strong>of</strong> antimicrobial<br />

susceptibility testing as well as the presentation <strong>of</strong> the obtained<br />

results and the proper comparison <strong>of</strong> data from different studies.<br />

Methodology<br />

Several methods, like disc diffusion, E-test, agar dilution, broth<br />

microdilution and broth macrodilution are suitable for in-vitro<br />

antimicrobial susceptibility testing (AST). Whichever method is<br />

used, the tests have to be performed in accordance with an<br />

internationally accepted procedure, such as those published by<br />

the Clinical and Laboratory Standards Institute (CLSI) among<br />

others. The documents issued are regularly updated and, since<br />

the methodologies and interpretative criteria change over time, it<br />

is important to always follow the latest edition. In contrast to most<br />

other organizations, the CLSI is unique in that it produces<br />

separate documents for use in human and veterinary<br />

microbiology. The CLSI documents are not freely available, but<br />

must be purchased.<br />

The status <strong>of</strong> the various types <strong>of</strong> documents is clarified below.<br />

The CLSI for example differentiates between “standards” and<br />

“guidelines”. A “standard” is a document that clearly identifies<br />

specific and essential requirements for materials, methods, and<br />

practices to be used in an unmodified form. A standard may, in<br />

addition, contain discretionary elements, which are clearly<br />

identified. In contrast, a “guideline” is a document describing<br />

criteria for a general operating practice, procedure, or material for<br />

voluntary use. A guideline can be used as written or modified by<br />

the user to fit specific needs.<br />

The current CLSI document for testing antimicrobial<br />

susceptibilities <strong>of</strong> bacteria isolated from animals, M31-A3 (3), is<br />

an approved standard and cannot be used in a modified form.<br />

Clear and precise instructions on how to perform AST in vitro are<br />

given. They include, for example, the medium to be used<br />

(including any supplements required to support the growth <strong>of</strong><br />

specific bacteria), the inoculum density, the incubation time, the<br />

temperature and test conditions. These instructions are not<br />

optional, but are strict rules that must be adhered to for good<br />

laboratory practice. Thus, statements such as “Susceptibility<br />

testing mainly followed the recommendations given in the CLSI<br />

document M31-A3” are not acceptable. Any deviation from the<br />

approved test conditions, such as the use <strong>of</strong> a different medium<br />

or extended incubation times for slow-growing bacteria, have to<br />

be specified and justified by the authors.<br />

Most AST documents cover testing <strong>of</strong> numerous different<br />

bacterial species. However, for several bacterial pathogens<br />

relevant to the veterinary field, such as Haemophilus parasuis or<br />

Riemerella anatipestifer, no approved methodology exists. If<br />

authors adopt a method approved for a phylogenetically closelyrelated<br />

organism, it must be stated clearly that the method used<br />

has not been approved for the species tested, but for another<br />

member the same genus (e.g. if the method for testing<br />

Haemophilus influenzae is used to test H. parasuis). Whenever<br />

susceptibility testing is undertaken on bacteria for which there is<br />

no approved standard available, the methodology chosen has to<br />

be validated first, as detailed in CLSI document M37-A3 (4).<br />

Quality controls<br />

It is essential to test approved AST reference strains in parallel<br />

with the test strains for quality control (QC) purposes. Lists <strong>of</strong><br />

approved reference strains are included in the documents<br />

mentioned. They also contain acceptable MIC and zone diameter<br />

ranges for these reference strains and clearly state the<br />

methodology (e.g. broth microdilution) and the medium (e.g.<br />

Mueller-Hinton agar) that the values relate to. The reference<br />

strains must be relevant to the bacterial species tested, e.g.<br />

Escherichia coli ATCC 25922 may be used when testing<br />

Enterobacteriaceae. Furthermore, authors must ensure (a) that<br />

reference strains are suitable for quality control <strong>of</strong> the<br />

antimicrobial agents tested, (b) that the range <strong>of</strong> concentrations<br />

(in broth microdilution) tested spans the entire approved QC<br />

ranges, and (c) that discs (in disc diffusion tests) contain the<br />

quantity <strong>of</strong> antimicrobial for which the QC ranges are approved.<br />

Interpretation <strong>of</strong> the results<br />

AST studies seek to categorize bacterial isolates as susceptible,<br />

intermediate or resistant to each antimicrobial tested, on the<br />

basis <strong>of</strong> the MICs or the zone diameters obtained. Such<br />

classification requires approved interpretive criteria. Currently,<br />

two different types <strong>of</strong> interpretive criteria are available, clinical<br />

breakpoints and epidemiological cut-<strong>of</strong>f values (5,6). The precise<br />

emphasis <strong>of</strong> a particular study will dictate which criteria must be<br />

applied. If data are intended to guide a therapeutic approach (i.e.<br />

the aim <strong>of</strong> the study is to determine which antimicrobial agents<br />

are most likely to lead to therapeutic success), clinical<br />

breakpoints must be applied. Epidemiological cut-<strong>of</strong>f values<br />

should be used to describe MIC distributions <strong>of</strong> bacteria without<br />

clinical context. Clinical breakpoints and epidemiological cut-<strong>of</strong>f<br />

values may be very similar or even identical for some<br />

bacteria/drug combinations; however, authors need to<br />

understand that epidemiological cut-<strong>of</strong>f values are determined by<br />

a different approach than clinical breakpoints and do not<br />

necessarily take into account the results <strong>of</strong> clinical efficacy<br />

studies, dosing and route <strong>of</strong> administration <strong>of</strong> the antimicrobial<br />

agents, nor the drug’s pharmacokinetic and pharmacodynamic<br />

parameters in the respective animal species. The term<br />

“breakpoint” should be used exclusively for clinical breakpoints<br />

and “susceptible”, “intermediate” and “resistant” categories<br />

should also be reserved for classifications made in relation to the<br />

therapeutic application <strong>of</strong> antimicrobial agents. When reporting<br />

data using epidemiological cut-<strong>of</strong>f values, the term ‘resistant’ is<br />

inappropriate, rather bacteria should be reported as ‘wild type’ if<br />

the MIC or zone diameter falls within the wild type range, or ‘nonwild<br />

type’ if the MIC is higher or the zone diameter smaller than<br />

the wild-type range.<br />

The CLSI document M31-A3 (3) lists exclusively clinical<br />

breakpoints and includes the largest collection <strong>of</strong> approved<br />

clinical breakpoints for bacteria <strong>of</strong> animal origin currently<br />

available, a considerable number <strong>of</strong> which represent veterinaryspecific<br />

breakpoints. Many <strong>of</strong> the latter have been approved for<br />

specific disease conditions <strong>of</strong>ten caused by particular bacterial<br />

species in defined animal host species. For example, approved<br />

clinical breakpoints for enr<strong>of</strong>loxacin in cattle apply exclusively for<br />

bovine respiratory diseases due to Pasteurella multocida,<br />

Mannheimia haemolytica and Histophilus somni. The use <strong>of</strong><br />

these breakpoints for other bovine bacteria and different disease


conditions, e.g. Staphylococcus aureus from bovine mastitis, is<br />

unacceptable. Thus, the scope <strong>of</strong> application <strong>of</strong> the veterinaryspecific<br />

breakpoints is clearly defined and cannot be altered.<br />

All standards for performance <strong>of</strong> AST contain interpretive<br />

criteria which refer specifically to that particular methodology.<br />

Thus a certain methodology and its associated interpretive<br />

criteria are an entity, and as such belong together. It is not good<br />

practice to ‘mix and match’ testing methodologies and interpretive<br />

criteria issued by different organizations. Authors who perform E-<br />

tests must refer to the interpretive criteria given by the<br />

manufacturer <strong>of</strong> the E-test strips. Since these interpretive criteria<br />

are not veterinary-specific, but are adopted from human<br />

medicine, their true value for veterinary pathogens is unkown.<br />

The same holds for CLSI-approved breakpoints adopted from<br />

human medicine and listed in CLSI document M31-A3 (3).<br />

Authors who describe AST <strong>of</strong> animal isolates, <strong>of</strong>ten use incorrect<br />

or out-dated interpretive criteria derived from their own or others’<br />

previous publications. This is also bad practice and <strong>of</strong>ten results<br />

in cumulative errors. Authors must ensure that correct (at the<br />

time <strong>of</strong> submission) interpretive criteria are used. In addition,<br />

there is an onus on reviewers to verify whether the correct<br />

interpretive criteria were used.<br />

When comparing rates (percentages) <strong>of</strong> resistance between<br />

published studies, authors must make sure that the same<br />

methodologies and the same interpretive criteria have been used.<br />

Interpretive criteria <strong>of</strong>ten change over time and lowering the<br />

breakpoint(s) for a specific antimicrobial agent will result in a<br />

higher percentage <strong>of</strong> isolates being classified as resistant even if<br />

the MIC/zone size distribution <strong>of</strong> the population has not changed.<br />

As a consequence, an artefactual increase in the percentage <strong>of</strong><br />

resistant strains may be noted (Fig. 1). Publication <strong>of</strong> the full MIC<br />

distributions for each species/drug combination reduces the<br />

potential for this error since the data can be re-analysed by<br />

others if interpretive criteria change.<br />

(a)<br />

Number <strong>of</strong> Salmonella isolates<br />

(b)<br />

Number <strong>of</strong> Salmonella isolates<br />

2500<br />

2006<br />

2000<br />

1500<br />

1000<br />

500<br />

0<br />

2500<br />

2011<br />

2000<br />

1500<br />

1000<br />

500<br />

0<br />

susceptible: 2 mg/L<br />

susceptible: 0.06 mg/L<br />

MARAN 2004 (n = 2195)<br />

MARAN 2005 (n = 2238)<br />

resistant: 0.12 mg/L<br />

56 94 60 13 2 2<br />

0.06 0.12 0.25 0.5 1 2 4 8 16<br />

MIC values <strong>of</strong> cipr<strong>of</strong>loxacin in mg/L<br />

resistant: 4 mg/L<br />

63 79 33 8 5 1<br />

0.06 0.12 0.25 0.5 1 2 4 8 16<br />

MIC values <strong>of</strong> cipr<strong>of</strong>loxacin in mg/L<br />

0.27 %<br />

10.14 %<br />

Figure 1: Distribution <strong>of</strong> cipr<strong>of</strong>loxacin MIC values among<br />

Salmonella isolates collected in the MARAN studies 2004 and<br />

2005 and distinctly different percentages <strong>of</strong> resistance resulting<br />

from the use <strong>of</strong> different interpretive criteria in 2004 (a) and in<br />

2005 (b)<br />

Such tables or histograms are <strong>of</strong>ten large and due to limitations<br />

on journal space, may need to be provided as supplemental<br />

material.<br />

Before performing disc diffusion, authors need to make sure that<br />

the discs contain the correct quantity <strong>of</strong> antibiotic for which<br />

interpretive criteria are available. Unfortunately, for many<br />

antimicrobial agents a range <strong>of</strong> discs with varying amounts <strong>of</strong> the<br />

antimicrobial agent are commercially available. However, zone<br />

diameter interpretive criteria are commonly available only for a<br />

single specific disc strength. For example, discs charged with 10<br />

µg, 15 µg or 30 µg erythromycin are available, but CLSI<br />

interpretive criteria and QC ranges for reference strains (3) refer<br />

only to zone diameters around a 15 µg disc. Since it is not<br />

possible to adjust the values measured with a 10 µg or a 30 µg<br />

disc to the values for an approved 15 µg disc, zone sizes<br />

obtained with the 10 µg or a 30 µg disc can neither be interpreted<br />

nor validated.<br />

A standard dilution series for AST consists <strong>of</strong> doubling antibiotic<br />

concentrations and includes the reference concentration 1 mg/L<br />

(e.g. 0.125, 0.25, 0.5, 1, 2, 4, 8 mg/L etc.). E-test strips indicate<br />

half-log values and so MICs determined by E-test should be<br />

‘rounded up’ to the next highest value on the standard series. For<br />

example, if an E-test indicates that growth is inhibited at 0.38<br />

mg/L (which is not a concentration in the standard series), the<br />

MIC should be rounded up and reported as 0.5 mg/L.<br />

MIC 50 and MIC 90 values<br />

MIC 50 and MIC 90 values as well as the range <strong>of</strong> values obtained<br />

are important parameters for reporting results <strong>of</strong> susceptibility<br />

testing when multiple isolates <strong>of</strong> a given species are tested. The<br />

MIC 50 represents the MIC value at which at least 50% <strong>of</strong> the<br />

isolates in a test population are inhibited; it is equivalent to the<br />

median MIC value. Given n test strains and the values y 1 , y 2 , ...,<br />

y n representing a graded series <strong>of</strong> MICs starting with the lowest<br />

value, the MIC 50 is the value at position n x 0.5 as long as n is an<br />

even number <strong>of</strong> test strains. If n is an odd number <strong>of</strong> test strains,<br />

the value at position (n+1) x 0.5 represents the MIC 50 value. The<br />

MIC 90 represents the MIC value at which at least 90% <strong>of</strong> the<br />

strains within a test population are inhibited; the 90 th percentile.<br />

The MIC 90 is calculated accordingly, using n x 0.9. If the resulting<br />

number is an integer, this number represents the MIC 90 ; if the<br />

resulting number is not an integer, the next integer following the<br />

respective value represents the MIC 90 . MIC 50 and MIC 90 values<br />

should always be presented as concentrations on the standard<br />

AST dilution series. If using a statistical package to calculate the<br />

values, never use intermediate values. It should be noted that<br />

MIC 50 and MIC 90 values are not necessarily suitable parameters<br />

to describe bimodal or trimodal MIC distributions, although a<br />

discrepancy <strong>of</strong> several dilution steps between the MIC 50 and<br />

MIC 90 values, e.g. MIC 50 at 0.25 mg/L and the MIC 90 at 16 mg/L,<br />

might point towards the presence <strong>of</strong> at least two subpopulations<br />

which differ distinctly in their MICs to a given antimicrobial agent.<br />

As an example, in a test population <strong>of</strong> 70 strains, the MIC 50 is the<br />

value at position 35 and the MIC 90 the one at position 63 in a<br />

graded series <strong>of</strong> MICs starting with the lowest MIC value at<br />

position 1. In a test population <strong>of</strong> 71 strains, the MIC 50 is the<br />

value at position 36 and the MIC 90 the one at position 64 in the<br />

aforementioned graded series <strong>of</strong> the MICs.<br />

Although MIC 50 and MIC 90 values can also be calculated for<br />

small test populations <strong>of</strong> e.g. 10–30 strains, under such<br />

conditions few strains with high MICs will have a<br />

disproportionately high influence on the MIC 50 and MIC 90 values.<br />

Thus, researchers are encouraged not to overemphasize MIC 50<br />

and MIC 90 data obtained from small test populations. Since the<br />

significance <strong>of</strong> MIC 50 and MIC 90 increases with the numbers <strong>of</strong><br />

strains tested, sufficiently large test populations should be used<br />

for most meaningful statements on MIC 50 and MIC 90 values.<br />

Multi-resistance<br />

The term ‘multi-resistance’ exclusively refers to acquired<br />

resistance properties. Bacteria may occasionally exhibit intrinsic<br />

(primary) resistance to certain antimicrobial agents. Intrinsic<br />

resistance may be based on either the lack or the inaccessibility<br />

<strong>of</strong> the antimicrobial target site among the bacteria in question. In<br />

other cases, intrinsically resistant bacteria produce inactivating<br />

enzymes, such as species-specific β-lactamases, contain<br />

multidrug transporters, and/or exhibit permeability barriers. Such<br />

intrinsic resistances must be excluded when describing multiresistance<br />

patterns.<br />

There is no universally accepted definition <strong>of</strong> ‘multi-resistance’.<br />

As a consequence, this term is used inconsistently in the<br />

literature. The following suggestions are intended to provide<br />

guidance for the most accurate presentation <strong>of</strong> multi-resistance<br />

patterns:


(a) If only phenotypic susceptibility testing is performed,<br />

resistance to three or more classes <strong>of</strong> antimicrobial agents can<br />

be referred to as multi-resistance. For example, resistance to<br />

enr<strong>of</strong>loxacin, marb<strong>of</strong>loxacin, difloxacin and orbifloxacin<br />

represents resistance to one antimicrobial class, since all agents<br />

are fluoroquinolones and resistance is most likely mediated by<br />

the same mechanism(s). In the case <strong>of</strong> fluoroquinolones (and<br />

some other antimicrobial classes), resistance to a single<br />

representative <strong>of</strong> this class <strong>of</strong> antibiotic agent can reasonably be<br />

extrapolated to resistance (or reduced susceptibility) to other<br />

members <strong>of</strong> that class. However, single class representatives<br />

cannot always be validly defined, e.g. for -lactams and<br />

aminoglycosides. In these cases, resistance is not a class effect<br />

and multiple, diverse resistance mechanisms exist, each <strong>of</strong> which<br />

confers resistance to sub-groups <strong>of</strong> the respective antimicrobial<br />

class. Resistance to sub-groups should be counted separately<br />

e.g. resistance to streptomycin and spectinomycin is distinct from<br />

resistance to gentamicin, kanamycin and/or tobramycin.<br />

(b) If phenotypic susceptibility testing is supplemented with<br />

molecular analysis for the resistance genes present, multiresistance<br />

should be assessed at the molecular level. Bacterial<br />

isolates exhibiting the presence <strong>of</strong> three or more resistance<br />

genes or mutations, all <strong>of</strong> which are associated with a different<br />

resistance phenotype (i.e. affecting different antimicrobial classes<br />

or sub-groups), may be referred to as multi-resistant. Exceptions<br />

to this rule would include those cases where a single resistance<br />

gene or a gene complex is associated with resistance to<br />

structurally and/or functionally different antimicrobial agents, e.g.<br />

the gene cfr for resistance to phenicols, lincosamides,<br />

oxazolidinones, pleuromutilins, and streptogramin A antibiotics or<br />

the erm genes for combined resistance to macrolides,<br />

lincosamides and streptogramin B antibiotics.<br />

Bacteria <strong>of</strong> Animal Origin; A Report. (ISBN Number:1-56238-765-0). CLSI<br />

document X08-R. Wayne, PA: Clinical and Laboratory Standards Institute.<br />

Conclusions<br />

As indicated above, conducting AST and subsequent data<br />

interpretation is a complex matter. A number <strong>of</strong> competent<br />

authorities provide instructions for performing AST and data<br />

interpretation. Each should be followed precisely. Importantly,<br />

protocols for AST and data interpretation from different<br />

authorities cannot be interchanged. AST data intended for the<br />

recommendation <strong>of</strong> therapy should be interpreted and reported<br />

using clinical breakpoints, whereas AST data intended for<br />

surveillance purposes may be reported using epidemiological cut<strong>of</strong>f<br />

values. Moreover, the comparison <strong>of</strong> data generated in<br />

different studies requires not only a common methodology, but<br />

also the preferential presentation <strong>of</strong> the data as MIC distribution<br />

which allows for fast and easy re-evaluation <strong>of</strong> the original data<br />

even if the interpretive criteria change over time. In addition to the<br />

two editorials published in 2010 (1,2), CLSI has published in<br />

2011 a comprehensive report (CLSI document X08-R)<br />

“Generation, Presentation and Application <strong>of</strong> Antimicrobial<br />

Susceptibility Test Data for Bacteria <strong>of</strong> Animal Origin” (7). This<br />

report provides additional information on how to avoid mistakes in<br />

AST methodology, AST data reporting and AST data<br />

interpretation.<br />

References<br />

1. Schwarz, S, Silley, P, Simjee, S, Woodford, N, van Duijkeren, E,<br />

Johnson, AP, Gaastra, W (2010). Assessing the antimicrobial susceptibility<br />

<strong>of</strong> bacteria obtained from animals. Veterinary Microbiology, 141, 1-4.<br />

2. Schwarz, S, Silley, P, Simjee, S, Woodford, N, van Duijkeren, E,<br />

Johnson, AP, Gaastra, W (2010). Assessing the antimicrobial susceptibility<br />

<strong>of</strong> bacteria obtained from animals. Journal <strong>of</strong> Antimicrobial Chemotherapy,<br />

65, 601-604.<br />

3. Clinical and Laboratory Standards Institute (CLSI), 2008a. Performance<br />

standards for antimicrobial disk and dilution susceptibility tests for bacteria<br />

isolated from animals; approved standard – third edition (ISBN Number:1-<br />

56238-659-X). CLSI document M31-A3. Clinical and Laboratory Standards<br />

Institute, Wayne, PA, USA.<br />

4. Clinical and Laboratory Standards Institute (CLSI), 2008b. Development<br />

<strong>of</strong> in vitro susceptibility testing criteria and quality control parameters for<br />

veterinary antimicrobial agents; approved guideline - third edition (ISBN<br />

Number 1-56238-660-3). CLSI document M 37-A3. Clinical and Laboratory<br />

Standards Institute, Wayne, PA, USA.<br />

5. Bywater, R, Silley, P, Simjee, S (2006). Antimicrobial breakpoints –<br />

definitions and conflicting requirements. Veterinary Microbiology 118, 158-<br />

159.<br />

6. Simjee, S, Silley, P, Werling, HO, Bywater, R (2008). Potential confusion<br />

regarding the term ‘resistance’. Journal <strong>of</strong> Antimicrobial Chemotherapy 61,<br />

228-229.<br />

7. Clinical and Laboratory Standards Institute (CLSI) (2011). – Generation,<br />

Presentation and Application <strong>of</strong> Antimicrobial Susceptibility Test Data for


S1 - O - 1<br />

ORAL FLUIDS – SAMPLE MATRIX FOR EFFECTIVE HERD HEALTH MONITORING<br />

C.Boss 1 , R. Shah 2 , C. O’Connell 2<br />

1<br />

Life Technologies,Darmstadt, Germany<br />

2<br />

Life Technologies, Austin, USA<br />

Oral Fluids, PRRS, PCV2, SIV<br />

Introduction<br />

The use <strong>of</strong> oral fluid as a sample matrix in porcine reproductive<br />

and respiratory syndrome virus (PRRSV) surveillance has<br />

increased in many parts <strong>of</strong> the world over the last few years.<br />

Advantages <strong>of</strong> using this matrix are ease <strong>of</strong> collection, reduced<br />

stress to the pigs, the low cost <strong>of</strong> collection and minimum labor<br />

required. Successful implementation <strong>of</strong> sample testing through<br />

the use <strong>of</strong> this sample matrix has extended to the evaluation <strong>of</strong> its<br />

testing for other swine diseases, including porcine circovirus type<br />

2 (PCV2), swine influenza virus (SIV), and mycoplasma<br />

hyopneumoniae (M. Hyo).<br />

Materials & methods<br />

In a multi-center, collaborative study, cotton ropes were used to<br />

collect samples <strong>of</strong> oral fluid from pigs that were experimentally<br />

infected with PRRSV and PCV2. Additionally, oral fluid samples<br />

from SIV negative pens were spiked with SIV for evaluation.<br />

Nucleic acid was extracted from oral fluid samples using a<br />

MagMAX based magnetic bead extraction and then amplified<br />

using a TaqMan® based real-time PCR assay for each virus. The<br />

method is semi automated, involving the use <strong>of</strong> the MagMAX<br />

Express-96 Magnetic Particle Processor for the purification <strong>of</strong><br />

nucleic acid.<br />

For PRRSV testing, in collaboration with Kansas State University,<br />

oral fluids were collected from pens on the day that the pigs were<br />

experimentally infected through 40 days post-infection. Serum<br />

was collected from each pig within the pen on the same day that<br />

oral fluids were collected from the pen. Serum samples and oral<br />

fluids were both tested using real-time PCR.<br />

For PCV2 testing, in collaboration with Iowa State University,<br />

twenty-four 21-day-old pigs free <strong>of</strong> PRRSV and SIV were<br />

assigned to 1 <strong>of</strong> 4 groups and housed in pens in separate rooms.<br />

Pen number One was the control (None-Challenge) group. Each<br />

pig in the other 3 groups received PCV2a strain at 11 weeks <strong>of</strong><br />

age (dpi 0). Six pigs (Pen 3) were re-challenged with PCV2a<br />

strain at 35, 70, and 105 dpi. Each pig in Pen 4 group<br />

alternatively received PCV2a (dpi 0 and 70) and PCV2b (dpi 35<br />

and 105). The two PCV2a strains used were heterologous. Oral<br />

fluids samples were collected, dpi 0, 2, 4, 6, 8, 10, 12, 14, and<br />

weekly thereafter until dpi 140. The results are shown in Figure 2<br />

for the time periods tested using the PCV2 real-time PCR<br />

reagents (dpi 2-98<br />

For SIV testing, in collaboration with Iowa State University, a total<br />

<strong>of</strong> 180 oral fluid samples were spiked with high, medium and low<br />

copy numbers <strong>of</strong> SIV. The extracted nucleic acids were also<br />

tested with subtyping reagents for the identification <strong>of</strong><br />

hemagglutinin and neuraminidase subtypes.<br />

<strong>of</strong> ~20 swine, the proposed method <strong>of</strong> sample preparation and<br />

nucleic acid purification efficiently processes multiple samples,<br />

thereby decreasing screening time.<br />

A major advantage <strong>of</strong> this high throughput method, combined<br />

with the ease <strong>of</strong> collection <strong>of</strong> oral fluid, is that large numbers <strong>of</strong><br />

pigs can be tested without increased cost or labor. This provides<br />

a more efficient and cost effective testing environment and the<br />

ability to better curb incidences <strong>of</strong> infection.<br />

Acknowledgements<br />

Dr. Bob Rowland, Kansas State University<br />

Dr. Tanja Opriessnig, Iowa State University<br />

Dr. Christa Irwin, Iowa State University<br />

Dr. Jeff Zimmerman, Iowa State University<br />

Results<br />

PRRSV nucleic acid was amplified from all oral fluid samples<br />

derived from the experimentally infected pigs. Lower Cts,<br />

indicating a higher copy number <strong>of</strong> viral RNA, were present in the<br />

serum samples early post-infection; but similar levels were<br />

detected in serum and oral fluid at later collection points. These<br />

results indicate a high level <strong>of</strong> correlation between PRRSV<br />

detection from serum collected from individual animals and from<br />

the pen or herd based oral fluid samples.<br />

Using the SIV screening real-time PCR reagents, SIV nucleic<br />

acid was detectable in all <strong>of</strong> the spiked oral fluid samples and at<br />

each concentration level. Additionally, the results <strong>of</strong> the subtyping<br />

study indicate that all positive samples could be sub-typed.<br />

High titers <strong>of</strong> PCV2 were detected from day 12 through 28 post<br />

infection, and virus was detectable throughout the entire testing<br />

period (dpi 2 – 98).<br />

Discussion & conclusion<br />

In summary, oral fluid samples were demonstrated to provide<br />

sensitive detection <strong>of</strong> PRRSV, SIV, and PCV2. As these<br />

samples could be used for the detection <strong>of</strong> virus in a population


S1 - O - 2<br />

DIAGNOSTIC PROPERTIES OF SEVERAL PRRS ANTIBODY ELISAS IN THE CONTEXT OF A<br />

LONGITUDINALSTUDY OF A FARM USING DIFFERENT VACCINATION STRATEGIES<br />

Strutzberg-Minder K 1 , Seidenspinner M 2 , Schuh H 2 , Böttcher J 3 , Wendt M 4 , Leibold W 5<br />

1 IVD GmbH, Hannover, Germany,<br />

2 Dr. Hermann Schuh, Veterinary Practice, Ipsheim, Germany<br />

3 Bavarian Animal Health Service, Poing, Germany,<br />

University <strong>of</strong> Veterinary Medicine, Foundation, 4 Klinik für kleine Klauentiere, 5 Institute <strong>of</strong> Immunology, Hannover, Germany<br />

PRRS virus, serology<br />

Introduction<br />

Porcine reproductive and respiratory syndrome (PRRS) is one <strong>of</strong><br />

the most important diseases in pig farms almost worldwide (1).<br />

The PRRS virus (PRRSv) causes severe economic losses due to<br />

reproductive failures in sows and gilts and respiratory distress in<br />

piglets and fattening pigs (1). Among the objectives <strong>of</strong> the study<br />

was a longitudinal analysis <strong>of</strong> the antibody response following<br />

different vaccination strategies by routine diagnostic ELISA.<br />

Materials & methods<br />

The study was performed on an intensive pig farm in Germany<br />

with about 120 sows, 450 raising sites and 500 fattened pigs in a<br />

closed system. 8 to 12 piglets <strong>of</strong> 6 sows examined preliminarily<br />

were reassembled in 4 different groups with a total <strong>of</strong> 15 piglets<br />

in each. Blood samples were taken at 10 weeks (w), 13 w, 16 w,<br />

21 w, 26 w and 27 w <strong>of</strong> age. Pigs were vaccinated with Porcilis ®<br />

PRRS (live European PRRSV strain DV, Intervet International,<br />

now: MSD Animal Health, USA) and Progressis ®<br />

(inactivated<br />

PRRSv European strain P120, Merial, UK) as described in the<br />

instruction manuals and in the following table.<br />

Table 1: Vaccination schedule<br />

Group 1 Group 2 Group 3 Group 4<br />

Age <strong>of</strong> pigs<br />

(red)<br />

(blue)<br />

(green) (yellow)<br />

8 Progressis ®<br />

11<br />

Porcilis ® Porcilis ® Porcilis ®<br />

PRRS<br />

PRRS<br />

PRRS<br />

20 Progressis ® Progressis ®<br />

25<br />

Porcilis ® Porcilis ®<br />

PRRS<br />

PRRS<br />

Progressis ®<br />

Porcilis ®<br />

PRRS<br />

Porcilis®<br />

PRRS<br />

Serum samples were analysed with the following PRRS antibody<br />

ELISAs:<br />

A) HerdChek* PRRS 2XR, IDEXX Laboratories, USA. A<br />

sample/positive control (S/P) ratio (blanked by normal host<br />

cells antigen for control) ≥ 0.400 is positive.*<br />

B) HerdChek* PRRS X3 (now: IDEXX PRRS X3), IDEXX<br />

Laboratories, USA. An S/P ratio (blanked by negative<br />

control, NC) ≥ 0.400 is positive.*<br />

C) INGEZIM PRRS UNIVERSAL, INGENASA, Spain. S/P ratio<br />

transformed in OD%; an OD% > 15 is positive.<br />

D) INGEZIM PRRS EUROPA, INGENASA, Spain. S/P ratio<br />

transformed in OD%; an OD% > 15 is positive.<br />

E) INGEZIM PRRS AMERICA, INGENASA, Spain. S/P ratio<br />

transformed in OD%; an OD% > 15 is positive.<br />

F) CIVTEST SUIS PRRS E/S (European strains), Laboratories<br />

HIPRA, Spain. An IRPC (Index Relative to Positive Control)<br />

value ((sample - NC) / (PC - NC) x 100) > 20 is positive.<br />

G) CIVTEST SUIS PRRS A/S (American strains), Laboratories<br />

HIPRA, Spain. IRPC value >20 is positive<br />

H) INGEZIM DR, INGENASA, Spain. S/P ratio transformed in<br />

OD%; an OD% > 17.5 is positive.<br />

I) PRRSv, BioChek, The Netherlands. An OD > 0.500 is<br />

positive.*<br />

*Results <strong>of</strong> ELISA with real numbers were multiplied by 100 to<br />

yield a positive integer for better comparison <strong>of</strong> the different<br />

ELISA results in one diagram (Fig. 1). ELISA H and I were used<br />

only for group 2.<br />

Results<br />

The individual course <strong>of</strong> antibody development detected by all<br />

ELISA differed most in group 4. However, the arithmetic means<br />

smoothed the individual differences (Fig. 1). Results for group 4<br />

were significantly higher (p ≤ 0.0606) than for group 1 and 2 at 13<br />

w. Maximum antibody levels were observed between 21 w and<br />

26 w (10 w to 15 w after priming with Porcilis ® PRRS) for all<br />

ELISA (Fig. 1). These peaks were more or less independent <strong>of</strong><br />

subsequent boosting with Progressis ® or with Porcilis ® PRRS (s.<br />

Tab. 1).<br />

Mean <strong>of</strong> ELISA results<br />

3.500<br />

3.000<br />

2.500<br />

2.000<br />

1.500<br />

1.000<br />

0.500<br />

ELISA B<br />

0.000<br />

7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28<br />

Age <strong>of</strong> pigs (weeks)<br />

Group 1 Group 2 Group 3 Group 4<br />

Figure 1: Pigs' developing antibody response (mean ELISA<br />

results) with 4 different PRRSv vaccination strategies detectable<br />

by different ELISAs (here: ELISA B) for PRRSv antibodies<br />

The earliest positive results after exposition with live PRRSv<br />

vaccine at 11 w were detected by ELISAs A and B in at least 6<br />

(group 3) <strong>of</strong> 15 samples and all means <strong>of</strong> all groups (Fig. 2) at 13<br />

w (2 w after priming with Porcilis ®<br />

PRRS). ELISA H tested<br />

positive in all but 2 samples at all dates before and after<br />

vaccination without significant changes in results (Fig. 2). The<br />

earliest positive results <strong>of</strong> the other ELISAs C, D, F and I in<br />

almost all samples and all means <strong>of</strong> groups 1-3 were at 16 w (5 w<br />

after priming with Porcilis ® PRRS) (Fig. 2). Only group 4 tested<br />

positive also by ELISAs C, D and F at 13 w.<br />

Mean <strong>of</strong> ELISA results<br />

450<br />

Group 2<br />

400<br />

350<br />

300<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28<br />

Age <strong>of</strong> pigs (weeks)<br />

ELISA A ELISA B ELISA C ELISA D ELISA F ELISA H ELISA I<br />

Figure 2: Pigs' developing antibody response with different<br />

PRRSv vaccination strategies (here: group 2 mean ELISA<br />

results) detectable by different ELISAs for PRRSv antibodies<br />

Discussion & conclusion<br />

As ELISA H results did not reflect any PRRS vaccination they<br />

were not useful for understanding antibody development as an<br />

immunological response to PRRSv. The earliest detection <strong>of</strong><br />

exposing pigs to live PRRSv was possible with ELISA A and B at<br />

the first sampling (2 w after priming). Pre-immunization with<br />

Progressis ® (group 4) also made it possible to detect antibodies<br />

against PRRSv at 13 w by the other positive ELISAs C, D and F,<br />

but individual serological pr<strong>of</strong>iles were very heterogeneous, while<br />

all other individual serological pr<strong>of</strong>iles (groups 1-3) were more<br />

homogenous and represented appropriately by the group means.<br />

ELISAs A and B can be used at the latest 2 w after priming with<br />

PRRSv for early detection, and other ELISAs except E, G and H,<br />

for monitoring the antibody response.<br />

References<br />

1. Zimmermann et al. (2006) in: Straw et al. (eds.), Diseases <strong>of</strong> Swine,<br />

9thed., 387-417<br />

Note: These data are in part results <strong>of</strong> the thesis <strong>of</strong> Marc Seidenspinner


S1 - O - 03<br />

INTRODUCTION RATE OF A LOW PATHOGENIC AVIAN INFLUENZA VIRUS INFECTION IN<br />

DIFFERENT DUTCH POULTRY SECTORS<br />

Jose Gonzales 1 , Guus Koch 1 , Ruth Bouwstra 1 , Armin Elbers 1 , J.J. de Wit 2 , Arjan Stegeman 3<br />

1 Central Veterinary Institute, <strong>of</strong> Wageningen University and Research Centre (CVI-Lelystad), Lelystad, The Netherlands<br />

2 Animal Health Service, Deventer, The Netherlands<br />

3 Utrecht University, Faculty <strong>of</strong> Veterinary Medicine, Department <strong>of</strong> Farm Animal Health, Utrecht, The Netherlands<br />

Avian Influenza, low pathogenic, introduction rate<br />

Introduction<br />

Low Pathogenic Avian Influenza (LPAI) viruses <strong>of</strong> the H5 and H7<br />

subtype have the potential to evolute to Highly Pathogenic Avian<br />

Influenza (HPAI) viruses in poultry and therefore infections with<br />

these subtypes are notifiable . Consequently, member states <strong>of</strong><br />

the European Union have implemented surveillance programmes<br />

(1). In the Netherlands a syndromic surveillance and serological<br />

monitoring programme is in place. In the monitoring programme,<br />

all poultry farms are tested 1-4 times a year. Frequency differs<br />

between the different poultry types and housing systems (indoor<br />

and outdoor layer chickens, broilers, ducks, turkeys, etc) based<br />

on the supposed differences in the risk <strong>of</strong> introduction <strong>of</strong> LPAI<br />

infections. However, quantitative information regarding the<br />

possible differences in risk between these poultry types is sparse.<br />

In this study the rate <strong>of</strong> introduction <strong>of</strong> LPAI in different poultry<br />

types was quantified. .<br />

Materials & methods<br />

Data from the Dutch LPAI surveillance programme(2007–2010)<br />

were analysed using a generalised linear mixed and spatial<br />

model. All poultry farms should be tested at least once a year. In<br />

addition, outdoor-layer farms are tested 3 to 4 times per year.<br />

Farms were identified by their unique farm number (UBN) and<br />

poultry sector (duck-breeders, duck-meat (meat production),<br />

turkeys, broilers, indoor-layers, outdoor- layers, pullets and<br />

breeders). Based on the sampling frequency (time interval<br />

between samplings), the time at risk <strong>of</strong> exposure to a LPAI<br />

infection (“time at risk”) was calculated per poultry sector. For<br />

poultry sectors sampled once a year or once per production<br />

cycle, the age <strong>of</strong> the birds, at the moment <strong>of</strong> sampling, was taken<br />

as the time at risk. For poultry sectors sampled more than once<br />

per production cycle, the average sampling interval was taken as<br />

the time at risk. Positive cases were defined as: 1) farms with at<br />

least one seropositive animal – to any LPAI strain – in both, the<br />

screening test (IDEXX FLockCheck AI MultiS-Screen or agar gel<br />

precipitation, which is only used for broilers) and the confirmatory<br />

test (Hemagglutination Inhibition test) or 2) three or more<br />

positives in the screening test. Furthermore, only primary cases<br />

were included.<br />

Results<br />

The results are summarised in Table 1. Almost all seropositive<br />

results appeared to be single introductions. Results showed that<br />

outdoor-layer farms had a 11, turkey 8, duck-breeder 23 and<br />

meat-duck 13 times higher rate <strong>of</strong> introduction <strong>of</strong> LPAI than<br />

indoor-layer farms.<br />

Table 2. Relative risk (RR), with accompanying lower (LCI) and<br />

upper (UCI) 95% confidence intervals, <strong>of</strong> introduction <strong>of</strong> a LPAIv<br />

infection on poultry farms. Indoor - layers were considered as the<br />

reference category.<br />

Poultry Type<br />

RR<br />

Mean LCI UCL<br />

breeders 0.3 0.0 2.4<br />

pullets 0.7 0.1 5.7<br />

layers indoor 1.0<br />

layers outdoor 11.0 4.9 24.8<br />

turkeys 7.6 2.0 29.0<br />

duck meat 12.8 1.6 102.7<br />

duck breeders 23.0 6.2 85.7<br />

Discussion & conclusion<br />

Our analysis shows that outdoor-layer farms, duck (breeders and<br />

meat) farms and turkey farms have a significantly higher risk <strong>of</strong><br />

introduction <strong>of</strong> a LPAI infection compared to indoor-layer farms.<br />

Breeder ducks have the highest risk. This could be related to 1)<br />

their higher susceptibility to infection with LPAI <strong>of</strong> wild bird origin<br />

(ducks, geese, swans) than chickens, 2) their long production<br />

cycle (time <strong>of</strong> exposure), and 3) their higher exposure to LPAI<br />

from a contaminated environment and/or contact with wild<br />

waterfowl. The latter could also be the reason for the higher risk<br />

observed in outdoor-layer than indoor-layer farms.<br />

In the Netherlands, turkeys are raised indoors and despite the<br />

small population <strong>of</strong> turkey farms, we observed a higher risk <strong>of</strong><br />

introduction <strong>of</strong> a LPAI infection on turkeys than indoor-layers.<br />

This higher risk might be partly associated with the apparent<br />

higher susceptibility <strong>of</strong> turkeys to LPAI infections than chickens<br />

(2). We also observed a significant higher risk <strong>of</strong> introduction in<br />

meat-duck farms. This was surprising because this poultry type is<br />

kept indoors and has a short production cycle (6 weeks). The<br />

above mentioned higher susceptibility <strong>of</strong> ducks than chickens<br />

could be one reason for the observed higher risk. Differences in<br />

the rate <strong>of</strong> introduction <strong>of</strong> LPAI could be used to (re)design a<br />

targeted risk-based surveillance programme.<br />

References<br />

1.European Commission, 2007. Commission Decision 2007/268/EC <strong>of</strong> 13<br />

April 2007 on the implementation <strong>of</strong> surveillance programmes for avian<br />

influenza in poultry and wild birds to be carried out in the Member States<br />

and amending Decision 2004/450/EC. OJEU ;L 115:3.5.2007, p.2003.<br />

2. Tumpey, T, M, Kapczynski, D, R, Swayne, D,E 2004. Comparative<br />

susceptibility <strong>of</strong> chickens and turkeys to avian influenza A H7N2 virus<br />

infection and protective efficacy <strong>of</strong> a commercial avian influenza H7N2<br />

virus vaccine. Avian Diseases 48:167-176.


S1 - O - 04<br />

A DUPLEX ONE-STEP REAL TIME RT-PCR FOR DIAGNOSIS OF FOOT AND MOUTH DISEASE<br />

Kamila Górna, Romey Aurore, Anthony Relmy, Aude Allmandou, Sandra-Blaise Boisseau, Stephan Zientara,<br />

Labib Bakkali Kassimi<br />

ANSES, Laboratoire de Santé Animale, UMR1161 ANSES INRA ENVA, Maisons-Alfort 94706 France<br />

Foot- and –Mouth Disease; Diagnosis; Real Time RT-PCR<br />

Introduction<br />

Foot-and-mouth disease (FMD) is a highly contagious transboundary<br />

disease <strong>of</strong> wild and domesticated cloven-hooved<br />

ruminants. It is one <strong>of</strong> the most economically devastating<br />

diseases in livestock, due to measures employed to control the<br />

outbreak and trade loss resulting from embargos on the country.<br />

The causative agent, the FMD virus (FMDV), is a single stranded<br />

RNA virus belonging to the Picornaviridae family, genus<br />

Aphtovirus. There are seven immunologically distinct serotypes<br />

<strong>of</strong> the virus, namely types O, A, C, Asia 1, SAT 1, SAT 2 and<br />

SAT 3.<br />

Quick diagnosis <strong>of</strong> FMD is critical for the rapid implementation <strong>of</strong><br />

control measures to limit the spread <strong>of</strong> the virus and eradicate the<br />

disease. The recommended real-time RT-PCR method (rtRT-<br />

PCR) for detection <strong>of</strong> FMDV is a two-step simplex method<br />

targeting conserved regions in the internal ribosomal entry site<br />

within the 5’ untranslated region (IRES) and in the RNA<br />

polymerase gene (3D)(1). In parallel, a rtRT-PCR targeting a<br />

cellular gene (endogenous control) is performed to allow<br />

normalization <strong>of</strong> results. This method has the disadvantage <strong>of</strong><br />

being performed in two separate steps (RT and PCR) and<br />

detects only one target at a time. It is therefore time-consuming,<br />

and since several pipetting steps are required, this method can<br />

be subject to possible errors and contamination. Multiplex rtRT-<br />

PCR is a modification <strong>of</strong> the basic rtRT-PCR method in which<br />

pairs <strong>of</strong> primers targeting different targets are used in the same<br />

reaction. In one-step multiplex rtRT-PCR, the reverse<br />

transcription and subsequent amplification take place in a single<br />

tube, thereby reducing the reaction time and number <strong>of</strong> reaction<br />

mixtures, and also diminishing the risk <strong>of</strong> contamination. In this<br />

study, duplex one-step rtRT-PCRs were established to detect<br />

and type FMDV. These methods are simple, specific, and<br />

sensitive for early diagnosis <strong>of</strong> FMDV infection.<br />

Material & methods<br />

Samples: Artificially FMDV spiked samples were prepared by<br />

making 10 fold serial dilutions <strong>of</strong> virus stock (strains O1 Manisa,<br />

A22 Iraq or Asia 1) in FMDV-free bovine tongue epithelial tissue<br />

suspension. Field bovine epithelium samples (n:19) were<br />

collected from suspected cases in West Africa. Each sample was<br />

subjected to virus isolation, Ag-ELISA, Chromatographic test<br />

(Svanodip) and rtRT-PCR. Virus Isolation: Samples were prediluted<br />

5 and 10 times in cell culture medium and inoculated into<br />

goat tongue cell cultures (ZZ-R). Chromatographic test (lateral<br />

flow device): Svanodip (SVANOVA Biotech AB) test was<br />

performed according to the manufacturer’s instructions. Antigen<br />

Capture Elisa was performed as described in the OIE manual<br />

(1). rtRT-PCR: RNA was extracted from samples by using the<br />

QIAamp Viral RNA mini-kit (QIAGEN). Two step simplex rtRT-<br />

PCRs for IRES, 3D and -actin targets were performed using<br />

TaqMan Reverse Transcription Reagents kit and TaqMan PCR<br />

Core Reagents kit (Life technologies) following protocols<br />

described previously (2, 3, 4). One Step duplex rtRT- PCRs were<br />

performed using the AgPath-IDOne-Step RT-PCR Reagents<br />

(Life Technologies). The thermal conditions were as follows: 10<br />

min at 45°C (RT), 10 min at 95°C (denaturation) and 45 cycles <strong>of</strong><br />

15s at 95°C and 1 min at 60°C. The final concentration <strong>of</strong> primers<br />

and probes in duplex assay was optimized for IRES, 3D and -<br />

actin targets and was as previously described for the typing<br />

protocol (5). Each probe for FMDV specific targets, marked by<br />

FAM dye, was coupled in the same test tube with β-actin probe<br />

labelled with VIC dye.<br />

with two other vesicular disease viruses, swine vesicular disease<br />

and vesicular stomatitis viruses. Artificially FMDV spiked samples<br />

containing different amounts <strong>of</strong> virus were used to compare the<br />

sensitivity <strong>of</strong> FMDV diagnostic methods. Both one-step and twostep<br />

rtRT-PCR methods were 1000 to 10,000 fold more sensitive<br />

than the antigen capture Elisa and Svanodip test. VI and all rtRT-<br />

PCRs displayed quite similar sensitivity. The 3D/-actin duplex<br />

real-time one-step method gave better results than the two-step<br />

method. The serotype-specific one-step duplex rtRT-PCR was<br />

slightly less sensitive than the pan-FMDV one-step duplex rtRT-<br />

PCR method. A better detection <strong>of</strong> the internal β-actin control<br />

was observed with the one-step protocol. All rtRT-PCR methods<br />

were evaluated with 19 field samples. FMDV was isolated from<br />

all samples. The one-step duplex rtRT-PCR for both IRES and<br />

3D targets was more sensitive than the two-step simplex rtRT-<br />

PCR. Moreover, four samples found negative for IRES by a twostep<br />

method were positive using the one-step method. It was not<br />

possible to type the 19 field samples by rtRT-PCR, despite the<br />

presence <strong>of</strong> FMDV type O and A in these samples, as revealed<br />

by VP1 sequencing.<br />

Discussion & conclusion<br />

This study reports the development <strong>of</strong> a one-step duplex rtRT-<br />

PCR method for detection <strong>of</strong> FMDV and characterization <strong>of</strong> the<br />

three most commonly occurring serotypes worldwide. This<br />

method is more sensitive than other recommended FMDV<br />

detection methods tested. Simultaneous detection <strong>of</strong> FMDV and<br />

-actin within the same reaction permits the exclusion <strong>of</strong> false<br />

negatives that may result from improper extraction or degradation<br />

<strong>of</strong> the RNA, and permits normalization <strong>of</strong> the results. Thus, this<br />

one-step duplex rtRT-PCR method could be <strong>of</strong> interest for FMD<br />

diagnostic laboratories. We were unable to type FMDV from the<br />

field samples by rtRT-PCR. Analysis <strong>of</strong> the FMDV VP1 sequence<br />

from these samples revealed multiple mismatches in the primer<br />

and probe sequences for type O and type A. This is not<br />

surprising since the primers and probes were designed from<br />

sequences <strong>of</strong> FMDV strains belonging to lineages not present in<br />

West Africa (5). New primers and probes specific to WA lineages<br />

will be designed and evaluated. The development <strong>of</strong> a multiplex<br />

one-step rtRT-PCR could provide an improved method for rapid<br />

detection <strong>of</strong> FMDV, as it should allow detection <strong>of</strong> more targets in<br />

the same test and in less time.<br />

Acknowledgements<br />

The author thanks Jennifer Richardson for editing the English<br />

version <strong>of</strong> the manuscript.<br />

References<br />

1-Manuel <strong>of</strong> Diagnostic Test and Vaccines for Terrestrial Animals, chapter<br />

2.1.5.1C, 2009, OIE.<br />

2- M. Reid et al. Detection <strong>of</strong> all seven serotypes <strong>of</strong> foot-and-mouth<br />

disease virus by real-time, fluorogenic reverse transcription polymerase<br />

chain reaction assay. J. Virol. Methods, 2002, 105 : 67-80<br />

3- JD Callaham et al. Use <strong>of</strong> a portable real-time reverse transcriptasepolymerase<br />

chain reaction assay for rapid detection <strong>of</strong> foot-and-mouth<br />

disease virus. YAVMA, 2002, 220, 11:1636-42.<br />

4- J.F. Toussaint et al. Bluetongue virus detection by two real-time RTqPCR<br />

targeting two different genomic segments. J. Virol. Methods, 2007,<br />

140 : 115-128<br />

5- Reid et al. Detection <strong>of</strong> FMDV serotypes O, A and Asia1 by real-time<br />

RT-PCR. EuFMD 2008 Meeting. The global control <strong>of</strong> FMD – Tools, ideas<br />

and ideals – Erice, Italy 14-17 October 2008.<br />

Results<br />

One step duplex rtRT-PCR methods for pan-FMDV (IRES and<br />

3D) successfully detected all 7 serotypes <strong>of</strong> FMDV. The typing<br />

rtRT-PCRs detected exclusively the specific serotype and did not<br />

cross react with other serotypes. All tests gave negative results


S1 - O -05<br />

VALIDATION OF A COMPETITIVE ELISA FOR THE DETECTION OF ANTIBODIES ANTI-p26 OF<br />

EQUINE INFECTIOUS ANEMIA VIRUS IN EQUINE SERA<br />

R. Nardini 1 , G.L. Autorino 1 , R.Lorenzetti 1 , P. Cordioli 2 , 1 A. Caprioli, M.T. Scicluna 1<br />

1<br />

National Reference Centre for equine infectious diseases, Istituto Zoopr<strong>of</strong>ilattico Sperimentale Lazio e Toscana, 00178 Roma Italia. 2 Istituto<br />

Zoopr<strong>of</strong>ilattico Sperimentale Emilia Romagna e Lombardia, Via A. Bianchi 9, 25124<br />

Keywords: Equine infectious anemia, competitive ELISA, validation<br />

Introduction<br />

Equine infection anemia (EIA) is a viral infection <strong>of</strong> equidae,<br />

caused by a virus (EIAV) <strong>of</strong> the Lentivirus genus, Retroviridae<br />

family. Hematophagous insects belonging to the Tabanidae,<br />

Hippoboscidae and Muscinae families are involved as passive<br />

vectors in viral transmission. The infection is usually persistent,<br />

and can occur as an asymptomatic form or with recurrent febrile<br />

episodes. Since 2006, Italy has adopted a National Surveillance<br />

Plan (NSP) (1) consisting in a serological screening <strong>of</strong> all animals<br />

except for meat horses. According to the Italian Regulations, the<br />

<strong>of</strong>ficial confirmatory test is the Agar Gel Immunodiffusion (AGID)<br />

test which detects antibodies anti-p26, a highly conserved<br />

capsidic protein <strong>of</strong> the virus. Since this technique has a low<br />

detection limit, development <strong>of</strong> new and more sensitive tests has<br />

become necessary. WOAH Manual (2) indicates ELISA,<br />

immunoblotting and PCR as confirmatory tests for the serological<br />

diagnosis <strong>of</strong> EIA. The ELISA, for its characteristics, is the most<br />

suitable for screening purposes. The aim <strong>of</strong> this paper is to<br />

present the results <strong>of</strong> validation <strong>of</strong> a competitive ELISA (c-ELISA)<br />

for the detection <strong>of</strong> antibodies anti-p26 <strong>of</strong> EIAV in equine sera.<br />

Materials and methods<br />

The procedure <strong>of</strong> the c-ELISA, object <strong>of</strong> this validation, is briefly<br />

described as following. A 96-well microplate is pre-adsorbed<br />

overnight at 4°C with a monoclonal antibody (Mab) anti-p26.<br />

Serum samples and a recombinant antigen p26, produced in E.<br />

coli, are mixed on a separate plate and incubated at 37°C for 75<br />

minutes. At the end <strong>of</strong> the incubation, the serum-antigen mix is<br />

transferred onto the pre-absorbed plate and a second Mab<br />

conjugated with horseradish peroxidase is added. After another<br />

incubation at 37°C for 75 minutes, ortophenyl-diamine substrate<br />

is added and the plate is incubated at room temperature for 15<br />

minutes in the dark. The reaction is stopped by the addition <strong>of</strong><br />

sulphuric acid and the optical density (OD) is read with a 492nm<br />

filter. Sera are categorized as positive, negative or equivocal<br />

according to the percentage inhibition (PI), calculated as the ratio<br />

between the sample and the internal negative control.<br />

Validation <strong>of</strong> the c-Elisa was performed according to WOAH<br />

Manual guidelines (3) and the aim <strong>of</strong> this test was for screening<br />

purposes.<br />

For its validation the following parameters were evaluated.<br />

Analytical specificity was estimated at three different levels:<br />

1. Selectivity, defined as the capability to detect the target<br />

analyte in the presence <strong>of</strong> other interferences, was evaluated by<br />

changing the composition <strong>of</strong> the wash solution and processing<br />

positive and negative International Reference Serum (IRS).<br />

2. Exclusivity, considered as the capacity to discriminate<br />

target analyte from other crossreactive analytes, was evaluated<br />

processing ten sera positive for each <strong>of</strong> the following virus: Feline<br />

Immunodeficiency Virus, Feline Leukaemia Virus and Visna<br />

Maedi Virus. Each sample was repeated ten times.<br />

3. Inclusivity which is how a test can differentiate between<br />

different serovars. p26 protein is highly conserved in this virus<br />

and (4) thus evaluation was not necessary.<br />

Analytical sensitivity is represented by limit <strong>of</strong> detectability (LOD)<br />

and in this validation, the LOD <strong>of</strong> ELISA was compared with the<br />

AGID LOD, analysing progressive dilutions <strong>of</strong> a positive IRS in<br />

both methods.<br />

Repeatability was evaluated estimating the standard deviation<br />

(S r ) <strong>of</strong> the OD values <strong>of</strong> 30 repetitions <strong>of</strong> a negative IRS; this<br />

value was compared with another set <strong>of</strong> 30 values, processed by<br />

the same person during the same session.<br />

Reproducibility was estimated taking into account both qualitative<br />

and quantitative characteristics <strong>of</strong> ELISA test.<br />

a) To analyse qualitative reproducibility a Ring Test was<br />

performed by eleven laboratories and the statistic K <strong>of</strong> Cohen<br />

was calculated.<br />

b)Quantitative reproducibility was evaluated in seven sessions<br />

examining 30 replica <strong>of</strong> negative IRS in each, and calculating S R<br />

<strong>of</strong> OD values.<br />

Diagnostic performances were evaluated from sensitivity (Dse)<br />

and specificity (Dsp). Minimum number <strong>of</strong> samples to evaluate a<br />

sensitivity <strong>of</strong> 90% and a specificity <strong>of</strong> 80% with confidence<br />

interval <strong>of</strong> 99% and a precision <strong>of</strong> 5% was calculated. 1095 sera<br />

from field samples and from European Union Reference<br />

Laboratory, consisting in 857 positive and 238 negative samples<br />

were analysed with both ELISA and AGID, using the latter as<br />

Gold standard (GS). Dse, Dsp, positive and negative predictive<br />

values, efficacy and test Bias were calculated (5).<br />

For statistical analysis XL-Stat 2011 ® was used.<br />

Results<br />

1.ELISAs performed changing the washing solution did not<br />

correctly recognise positive and negative sera.<br />

2.All sera positive for other Lentivirus were classified as negative<br />

by the ELISA.<br />

3.LOD <strong>of</strong> ELISA test is at least 1 Log 10 more sensitive than AGID.<br />

4.Comparison between two data sets for evaluation <strong>of</strong><br />

repeatability did not show significant differences with test F<br />

(p=0.42) and S r resulted to be equal to 0.184.<br />

5.Statistic K value was equal to 0.967.<br />

6.Comparison between seven data sets for evaluation <strong>of</strong><br />

reproducibility did not show significant differences with test F<br />

(p=0,092) and S r was equal to 0.129.<br />

7.Dse and Dsp resulted respectively 100 and 80,3%.<br />

8.Positive and negative predictive values resulted respectively<br />

94.8% and 100%<br />

9.Efficacy <strong>of</strong> ELISA which is the percentage <strong>of</strong> samples correctly<br />

identified (positive and negative together) compared with the GS<br />

resulted equal to 95.7%<br />

10.Test Bias which is the ratio between apparent and real<br />

prevalence resulted 1.05.<br />

Discussion and conclusions<br />

The results <strong>of</strong> the parameters evaluated for the validation <strong>of</strong> the<br />

c-ELISA are all satisfactory. In particular, the lower Elisa LOD<br />

would allow not only the detection <strong>of</strong> “poor responders” then<br />

when using the AGID only, but also the possibility <strong>of</strong> detecting<br />

earlier, new cases. Although the Dsp is 80%, this value is still<br />

acceptable for a sreening test compared to the advantages that it<br />

presents to the AGID, i.e the possibility <strong>of</strong> its standardization, the<br />

objectivity <strong>of</strong> the results reading, the lower amount <strong>of</strong> reagents<br />

used and the higher processing <strong>of</strong> samples per unit time. The use<br />

<strong>of</strong> this diagnostic test is an added value, together with other<br />

measures, in a control programme for AIE.<br />

ReferenceS<br />

1.Ordinanza 8 agosto 2010 Piano di sorveglianza nazionale per l'anemia<br />

infettiva degli equidi. G.U. Serie Generale n. 219 del 18 settembre 2010<br />

2.Manual <strong>of</strong> Diagnostic Tests and Vaccines for Terrestrial Animals 2010,<br />

Chapter 2 . 5 . 6 .Equine infectious anaemia<br />

3.Manual <strong>of</strong> Diagnostic Tests and Vaccines for Terrestrial Animals<br />

2010,Chapter 1.1.4/5. Principles <strong>of</strong> validation <strong>of</strong> diagnostic assays for<br />

infectious diseases.<br />

4.Charles J. Issel, R. Frank Cook (1993)REVIEW ARTICLE A review <strong>of</strong><br />

techniques for the serologic diagnosis <strong>of</strong> equine infectious anemia J Vet<br />

Diagn Invest 5: 137-141<br />

5.Signorelli C. Elementi di metodologia epidemiologica SEU, 2005


S1 - O - 06<br />

DIAGNOSTIC PERFORMANCE OF A COMMERCIAL PRRS SERUM ANTIBODY ELISA ADAPTED TO<br />

ORAL FLUID SPECIMENS: FIELD SAMPLES<br />

Apisit Kittawornrat 1 , John Prickett 1 , Chong Wang 1, 2 , Chris Olsen 1 , Yaowalak Panyasing 1 , Andrea Ballagi 3 ,<br />

Anna Rice 3 , Sergio Lizano 3 , John Johnson 1 , Rodger Main 1 , Chris Rademacher 4 , Marlin Hoogland 4 ,<br />

Jeffrey Zimmerman 1<br />

1 Department <strong>of</strong> Veterinary Diagnostic and Production Animal Medicine, 2 Department <strong>of</strong> Statistics, Iowa State University, Ames, Iowa, United States, 3 IDEXX<br />

Laboratories, Westbrook, Maine, United States, 4 Murphy-Brown LLC, Ames, Iowa, United States<br />

Oral fluid, ELISA, Field samples, PRRSV<br />

Introduction<br />

Oral fluid samples are <strong>of</strong> interest because <strong>of</strong> their ease <strong>of</strong><br />

collection and documented use in surveillance <strong>of</strong> PRRSV and<br />

other pathogens (1,2). Previous work showed that a commercial<br />

PRRS ELISA (HerdChek® PRRS X3 ELISA) could be adapted to<br />

detect anti-PRRSV IgG in oral fluid specimens (IgG ELISA). The<br />

objective <strong>of</strong> the current study was to evaluate the ability <strong>of</strong> the<br />

assay to detect anti-PRRSV IgG antibody in pen-based oral fluid<br />

field samples.<br />

References<br />

1. Kittawornrat A, et al. 2010. PRRSV in serum and oral fluid samples from<br />

individual boars. Virus Res 154:170-176.<br />

2. Prickett, J et al.: 2011. Prolonged detection <strong>of</strong> PCV2 and anti-PCV2<br />

antibody in oral fluids following experimental inoculation. Transbound<br />

Emerg Dis 58:121-127<br />

3. Ramirez et al. <strong>2012</strong>. Efficient surveillance <strong>of</strong> pig populations using oral<br />

fluids. Prev Vet Med (Epub ahead <strong>of</strong> print).<br />

Materials & methods<br />

Positive samples were derived from a longitudinal field study in<br />

10 wean-to-finish barns (3). At each site, oral fluid samples were<br />

collected from the same 6 pens at 2-week intervals (total <strong>of</strong> 10<br />

sampling points per barn). Positive oral fluid samples were<br />

defined as all samples collected from a pen after the first PRRSV<br />

PCR positive oral fluid sample from that pen (n = 250). Negative<br />

oral fluid (n = 284) field samples were diagnostic samples<br />

submitted to the ISU VDL for PRRSV qRT-PCR testing from<br />

expected-negative herds.<br />

Results<br />

The cumulative S/P results for defined positive and negative oral<br />

fluid samples are given in Figure 1. Of 284 expected-negative<br />

field samples, all were negative on the IgG ELISA (S/P


S1 - O - 07<br />

SUBTYPING OF SWINE INFLUENZA VIRUSES BY MULTIPLEX REAL-TIME PCR<br />

Kees van Maanen 1 , Ingrid Wiggers 1 , Chris Schouten 3 , Tom Duinh<strong>of</strong> 1 , Paolo Cordioli 2 , Remco Dijkman 1<br />

1<br />

GD-Animal Health Service, Diagnostics,Research and Epidemiology,Deventer , the Netherlands<br />

2 Istituto Zoopr<strong>of</strong>ilattico Esperimentale Brescia, the Netherlands<br />

3 DAP Aadal, Erp, the Netherlands<br />

Swine Influenza Virus; real-time PCR; subtyping; lung lavage samples; diagnosis<br />

Introduction<br />

Swine influenza is an acute respiratory disease <strong>of</strong> swine caused<br />

by type A influenza viruses. In Europe, swine influenza is<br />

considered one <strong>of</strong> the most important primary pathogens <strong>of</strong> swine<br />

respiratory disease and infection is primarily with H1N1, H1N2<br />

and H3N2 influenza A viruses. Avian-like H1N1 and human-like<br />

H3N2 swine influenza viruses (SIV) have been considered<br />

widespread among pigs in Western Europe since the 1980s, and<br />

a novel H1N2 reassortant with a human-like H1 emerged in the<br />

mid 1990s. These viruses have remained endemic in European<br />

pig populations but significant differences in the circulation <strong>of</strong><br />

these strains occur at a regional level across Europe (1,2).<br />

For diagnosis <strong>of</strong> swine influenza infections serological and<br />

virological tests can be used. For detection <strong>of</strong> subtype-specific<br />

antibodies HI-tests are commonly used. However, crossreactions<br />

can occur, and the interpretation can be difficult<br />

depending on the vaccination and infection history <strong>of</strong> the herd.<br />

For diagnosis <strong>of</strong> recent infections paired serum samples are<br />

required collected with a 2-3 week interval.<br />

For rapid detection and discrimination <strong>of</strong> the different swine<br />

influenza subtypes several PCRs have been described. However,<br />

these methods were either conventional PCRs, targetted only the<br />

Haemagglutinin gene, or the coverage for European SIV strains<br />

was too low according to our BLAST analysis results.<br />

Therefore we decided to develop an in-house subtype-specific<br />

SIV real-time PCR. In the course <strong>of</strong> the project a prototype realtime<br />

PCR for detection and differentiation <strong>of</strong> SIV (Life<br />

Technologies) became available and was also evaluated.<br />

Materials & methods<br />

In this study two RNA extraction methods (High Pure nucleic acid<br />

kit, Roche Applied Science, and MagMax AM1836, Life<br />

Technologies) were compared in combination with either the inhouse<br />

designed primer/probe sets + AgPath-ID multiplex onestep<br />

RT-PCR kit (Life Technologies) or a prototype SIV<br />

Screening qRT-PCR kit (Life Technologies). All tests were run on<br />

an ABI7500 fast platform.<br />

Primers and probes were designed in silico using GenBank SIV<br />

sequences <strong>of</strong> the last 15 years. Primers and probes should have<br />

a coverage <strong>of</strong> at least 95%, and were used in working dilutions <strong>of</strong><br />

10 and 5 µM, respectively. Primers and probes were designed to<br />

enable all sorts <strong>of</strong> combinations (singleplex, multiplex H+N,<br />

multiplex N and multiplex H in parallel). Test results were<br />

compared with the results <strong>of</strong> an influenza A real-time PCR<br />

targetting the M gene that had been validated and implemented<br />

several years ago.<br />

Detection limits and efficiency were determined for decimal<br />

dilution series <strong>of</strong> H1N1, H3N2, and H1N2 reference viruses, as<br />

compared with the influenza A matrix real-time PCR. Subtypespecificity<br />

was evaluated by testing 22 different SIV strains<br />

provided by the IZSLER institute, Brescia, Italy. Diagnostic<br />

performance for lung lavage samples was evaluated by testing<br />

influenza A PCR positive pooled lung lavage samples (n=17) <strong>of</strong><br />

4, 7 and 10 week old piglets. For some pools the lung lavage<br />

samples that contributed to the pool were also tested individually<br />

(n=25) in order to compare the reproducibility <strong>of</strong> subtyping results<br />

and the presence <strong>of</strong> dual infections.<br />

Results<br />

For the haemagglutinin genes <strong>of</strong> H1N1, H1N2, and H3N2 viruses<br />

81, 36, and 61 entries were aligned for the development <strong>of</strong><br />

primer/probe sets. For the neuraminidase genes <strong>of</strong> the same<br />

subtypes 67, 28, and 25 entries were aligned for the development<br />

<strong>of</strong> primer/probe sets.<br />

Both for the in-house PCRs and for the prototype SIV Screening<br />

qRT-PCR kit RNA extraction with the MagMax AM1836 kit<br />

yielded 10-100 times lower detection limits than the High Pure<br />

nucleic acid kit. Using the MagMax AM1836 kit for RNA<br />

extraction, the detection limits <strong>of</strong> the in-house PCRs and the<br />

prototype commercial kit were comparable for the neuraminidase<br />

genes, whereas detection limits showed more variation for the<br />

haemagglutin genes. Overall, the detection limits <strong>of</strong> the subtypespecific<br />

primer/probe sets were equivalent to 10 times higher<br />

than the detection limit <strong>of</strong> the influenza A real-time PCR targeting<br />

the matrix gene.<br />

Subtype-specificity <strong>of</strong> the in-house PCR and the commercial kit<br />

were investigated by blind testing <strong>of</strong> a panel <strong>of</strong> Swine influenza<br />

virus isolates (H1N1 n=10; H1N2 n=4; H3N8 n=8) generously<br />

provided by the IZSLER institute, Brescia, Italy. The in-house<br />

PCRs failed to subtype several strains and also one probe<br />

appeared to be labile. In contrast the prototype commercial kit<br />

subtyped 21/22 strains correctly (for one sample a dual infection<br />

was reported, whereas this sample only contained H3N2). All<br />

strains were detected with the influenza A matrix PCR with Ctvalues<br />

ranging from 20-33.<br />

A first diagnostic evaluation <strong>of</strong> the commercial kit was performed<br />

by testing influenza A positive (results from another laboratory)<br />

pooled lung lavage samples (n=17) from piglets <strong>of</strong> 4-10 weeks <strong>of</strong><br />

age. All pools were also tested in our in-house influenza A PCR<br />

and results were confirmed. In the subtype-specific PCR 15/17<br />

pools yielded a single subtype (H1N2: n=9; H1N1: n=5; H3N2:<br />

n=1), whereas 2 pools yielded 2 or 3 subtypes. Retesting <strong>of</strong> the<br />

individual lung lavage samples that contributed to the latter pools<br />

confirmed the presence <strong>of</strong> dual infections in the majority <strong>of</strong> these<br />

piglets. Retesting <strong>of</strong> the individual lung lavage samples <strong>of</strong> three<br />

pools that were subtyped unequivocally yielded exactly the same<br />

subtype in all individual samples.<br />

Discussion & conclusion<br />

Influenza viruses are highly variable RNA viruses that show rapid<br />

evolution <strong>of</strong> especially the haemagglutinin and neuraminidase<br />

genes. Therefore, development <strong>of</strong> reliable PCR protocols for the<br />

subtyping <strong>of</strong> these viruses is not easy. Despite the effort we<br />

spent on selection <strong>of</strong> primers and probes, the results <strong>of</strong> our inhouse<br />

PCR were disappointing and inferior when compared to a<br />

prototype commercial real-time PCR kit.<br />

The latter kit showed a good performance when combined with<br />

RNA extraction on a MagMax AM1836 platform. Detection limits<br />

were comparable to tenfold higher than the influenza A matrix<br />

PCR, 95% <strong>of</strong> well-defined SIV strains were subtyped correctly,<br />

and the test also demonstrated a good diagnostic performance<br />

for subtyping <strong>of</strong> influenza A positive lung lavage samples.<br />

Although some practices in the Netherlands are very experienced<br />

in collecting lung lavage samples, the method is cumbersome;<br />

collecting other types <strong>of</strong> samples like nasal swabs or chewing<br />

ropes is much less invasive laborious. However, nasal and oral<br />

virus excretion is <strong>of</strong>ten short-lived. In the next study we will<br />

evaluate the diagnostic performance <strong>of</strong> real-time influenza A PCR<br />

and subtype-specific PCRs for nasal swabs and chewing ropes in<br />

fattening pigs with acute respiratory disease.<br />

Acknowledgements<br />

We thank Life Technologies for generously providing the<br />

prototype SIV Screening qRT-PCR kit for this study.<br />

References<br />

1. Van Reeth, K, Brown, IH, Dürrwald, R, Foni, E, Labarque, G, Lenihan,<br />

P, Maldonado, J,,Markowska-Daniel, I, Pensaert, M, Pospisil, Z, Koch, G<br />

(2008). Seroprevalence <strong>of</strong> H1N1, H3N2 and H1N2 influenza viruses in pigs<br />

in seven European countries in 2002-2003. Influenza Other Respi Viruses,<br />

2(3), 99-105.<br />

2: Brown, IH (<strong>2012</strong>). History and Epidemiology <strong>of</strong> Swine Influenza in<br />

Europe. Curr Top Microbiol Immunol Jan 11. [Epub ahead <strong>of</strong> print].


S1 - O - 08<br />

IMPORTANCE OF CONTINUOUS VALIDATION OF MOLECULAR METHODS FOR ROUTINE<br />

DIAGNOSIS OF PRRSV RNA IN CLINICAL SAMPLES<br />

Sandra Revilla-Fernández 1 , Adolf Steinrigl 1 , Tatjana Sattler 2 , Friedrich Schmoll 1<br />

1<br />

AGES Institute for Veterinary Disease Control, Department for Molecular Biology, Robert Koch Gasse 17, 2340 Mödling, Austria<br />

2<br />

Large Animal Clinic for Internal Medicine, University Leipzig, An den Tierkliniken 11, 04103 Leipzig, Germany<br />

PRRSV, real-time PCR, sensitivity, specificity, validation<br />

Introduction<br />

PPRSV is a small, enveloped, positive single-stranded RNA virus<br />

belonging to the Arteriviridae family, recognised world-wide as an<br />

important cause <strong>of</strong> reproductive failure and pneumonia in pigs.<br />

Since PRRSV was first described at the beginning <strong>of</strong> the 1990´s,<br />

wide genomic, antigenic and clinical differences have been<br />

described in isolates belonging to the European (EU) and North<br />

American (NA) genotypes. Several molecular-based methods like<br />

conventional and real-time RT-PCR (RT-qPCR) were<br />

demonstrated to be fit for detection <strong>of</strong> PRRSV in clinical samples.<br />

However, the high mutation rate <strong>of</strong> PRRSV poses a diagnostic<br />

challenge (4, 5). Therefore, the purpose <strong>of</strong> this study was to<br />

perform an in-house evaluation <strong>of</strong> recently established PRRSV<br />

molecular diagnostic methods and to compare them. Six different<br />

assays were tested with a series <strong>of</strong> PRRSV EU and NA field<br />

strains from different subtypes and origin. Diagnostic and<br />

analytical sensitivity and specificity were extensively studied. This<br />

is essential to control the disease spread and to guarantee the<br />

negative status <strong>of</strong> the herds (1).<br />

Materials & methods<br />

Six different one-step RT-PCR-based assays (Table 1), including<br />

one published RT-qPCR (3), four commercially available kits and<br />

one conventional ORF7 duplex RT-nested PCR (2) were tested<br />

for assay sensitivity and specificity with a series <strong>of</strong> PRRSV EU<br />

and NA field strains from Central Europe and Russia, samples<br />

from two European Ring trials and dilution series <strong>of</strong> in vitro EU<br />

and NA transcripts. The target sequences for the commercial RTqPCR<br />

kits were unknown. All but one test distinguish between<br />

EU and NA genotypes (Table 1). Runs were performed in two<br />

different real-time PCR cyclers. The ORF5 region <strong>of</strong> field<br />

samples was sequenced and phylogenetically analyzed with<br />

Bionumerics S<strong>of</strong>tware (Applied Maths), using reference strains <strong>of</strong><br />

all PRRSV genotypes and subgroups (4, 5). Quantitative RTqPCR<br />

data (Cq values) was analysed statistically by 2-sided<br />

paired t-test.<br />

Table 1: Summary <strong>of</strong> PRRSV PCR methods applied for<br />

validation, references and system descriptions<br />

conven<br />

-tional<br />

RTqPCR<br />

Assay<br />

Ref.<br />

comm. = commercially available<br />

Differentiation<br />

NA/EU<br />

RNA Input<br />

(µl)<br />

ORF7 2 yes, one-tube 3.5<br />

ORF6 3 yes, two-tube 5<br />

Kit A comm. yes, one-tube 8<br />

Kit B comm. yes, one-tube 5<br />

Kit C comm. no 5<br />

Kit D comm. yes, one-tube 5<br />

Results<br />

In general, the commercially available RT-qPCR kits showed<br />

higher sensitivity than the conventional method. RT-qPCR<br />

efficiency and sensitivity calculated from serial dilutions <strong>of</strong><br />

standard EU and NA PRRSV strains were optimal. However,<br />

specificity problems were seen with the published ORF6 based<br />

assay to detect some regional EU strains and the highly<br />

pathogenic (HP) NA strains. EU-1 field strains from Austria,<br />

Slovenia and Russia were equally well detected by all assays<br />

tested, whereas German strains showed the highest inter-assay<br />

differences.<br />

Taking into account different RNA sample inputs, statistical<br />

analysis <strong>of</strong> Cq values confirmed relevant differences between<br />

assays: lowest Cq values, indicating highest analytical sensitivity,<br />

were consistently observed with kits A, B and C. However, kit A<br />

did not efficiently detect some EU-2 and EU-unassigned strains,<br />

whereas kit B showed problems resulting from crosstalk between<br />

fluorescence channels in one <strong>of</strong> the real-time PCR cyclers used<br />

for validation, leading to false positive results. Kit C had<br />

specificity problems with NA HP strains included in one <strong>of</strong> the<br />

ring trials. The conventional RT-PCR method was shown to be<br />

less sensitive than the commercial real-time PCR Kits. Moreover,<br />

differentiation <strong>of</strong> double infection with both genotypes was not<br />

possible by real-time PCR with kit C and difficult when using the<br />

RT-nested PCR.<br />

Discussion & conclusion<br />

As shown, in-house validation may be helpful to identify the bestsuited<br />

methods for PRRSV diagnosis. To overcome these key<br />

aspects in PRRSV diagnosis, continuous re-validation <strong>of</strong> applied<br />

methods is necessary. If this is not possible, diagnostic<br />

laboratories should closely collaborate with the manufacturers <strong>of</strong><br />

commercial kits, supporting them with new field strains and with<br />

field observations.<br />

In many European countries, as in Austria or Germany, both EU<br />

and NA strains can be detected within the same herd or even<br />

within the same animal (1, and personal observations). In this<br />

situation, differentiation <strong>of</strong> both PRRSV genotypes is directly<br />

demanded by the clinical veterinarians since it is useful<br />

information for animal trade and epidemiological studies.<br />

In conclusion, this validation shows that there is currently no<br />

single assay or commercial kit with optimal analytic and<br />

diagnostic sensitivity. This indicates that various laboratory<br />

technical specifications, including the PCR machines, should be<br />

also considered for evaluation to avoid false results. The major<br />

problem to underline is the low specificity <strong>of</strong> some assays for<br />

detecting the newly evolved PRRSV strains that are<br />

underestimated if only PRRSV reference strains are considered<br />

for validation. Consequently, continuous evaluation and<br />

adaptation <strong>of</strong> routinely applied methods to currently circulating<br />

strains is necessary in each laboratory.<br />

References<br />

1. Große Beilage E, Bätza HJ. (2007): PRRSV-Eradikation: Eine Option für<br />

Schweinebestände in Deutschland? PRRSV-eradication: An option for pig<br />

herds in Germany? Berl Münch Tierärztl Wochenschr. 120, 11/12,470-479.<br />

2. Reiner G, Fresen C, Bronnert S, Willems H. (2009): Porcine<br />

Reproductive and Respiratory Syndrome Virus (PRRSV) infection in wild<br />

boars. Vet Microbiol. 12;136(3-4):250-8.<br />

3. Revilla-Fernández S, Wallner B, Truschner K, Benczak A, Brem G,<br />

Schmoll F, Mueller M, Steinborn R. (2005): The use <strong>of</strong> endogenous and<br />

exogenous reference RNAs for qualitative and quantitative detection <strong>of</strong><br />

PRRSV in porcine semen. J Virol Methods. 126:21-30.<br />

4. Shi M, Lam TT, Hon CC, Hui RK, Faaberg KS, Wennblom T, Murtaugh<br />

MP, Stadejek T, Leung FC. (2010): Molecular epidemiology <strong>of</strong> PRRSV: a<br />

phylogenetic perspective. Virus Res. 154(1-2):7-17.<br />

5. Stadejek T, Oleksiewicz MB, Scherbakov AV, Timina AM, Krabbe JS,<br />

Chabros K, Potapchuk D. (2008): Definition <strong>of</strong> subtypes in the European<br />

genotype <strong>of</strong> porcine reproductive and respiratory syndrome virus:<br />

nucleocapsid characteristics and geographical distribution in Europe. Arch<br />

Virol. 153(8):1479-88.


S1 - O - 09<br />

VIRAL DIAGNOSIS USING TRANSMISSION ELECTRON MICROSCOPY<br />

W.A. Cooley and D. J. Everest<br />

Animal Health and Veterinary Laboratories Agency-Weybridge, Bio-Imaging Unit, New Haw, Addlestone, Surrey KT15 3NB, UK<br />

Diagnosis, Electron Microscopy, Virus detection<br />

Introduction<br />

The size <strong>of</strong> most viruses places them well below the resolving<br />

power <strong>of</strong> the standard light microscope. However they are quite<br />

readily viewed and identified by transmission electron microscopy<br />

(TEM) and therefore TEM has provided contributions to virology<br />

and in particular to the discovery, detection and diagnosis <strong>of</strong><br />

various viral infections. Virus diagnosis by electron microscopy is<br />

based on the visualization and morphological identification <strong>of</strong><br />

virus particles. Molecular or immunological methods, such as<br />

Polymerase Chain Reaction (PCR) and Enzyme Linked Immuno<br />

Sorbent Assay (ELISA), are very effective in detecting known<br />

viruses and can be used to scrutinize a large amount <strong>of</strong> samples.<br />

However, one <strong>of</strong> the main advantages <strong>of</strong> using TEM for viral<br />

diagnosis is that it does not require organism-specific reagents<br />

for recognizing the pathogenic agent. Therefore, for new or<br />

unknown pathogens that may occur in the context <strong>of</strong> bio-terrorism<br />

attacks, or as a result <strong>of</strong> the manifestation <strong>of</strong> new pathogens,<br />

electron microscopy remains the only method that can provide a<br />

quick assessment <strong>of</strong> all pathogens present in a sample, providing<br />

an “open view”, which is why electron microscopy is considered a<br />

“catch all method”. Electron microscopy can also be used to<br />

investigate viruses isolated in cell cultures, which is useful in<br />

controlling the results <strong>of</strong> the virus isolation process, among other<br />

applications. The AHVLA at Weybridge, UK, provides a rapid<br />

(diagnosis within 15 minutes <strong>of</strong> arrival) viral diagnostic service for<br />

various diseases affecting farmed livestock and wildlife species<br />

through veterinary surveillance as part <strong>of</strong> the Great Britain<br />

Wildlife Disease Surveillance Partnership and also for captive<br />

and zoological animals. The samples most frequently received for<br />

viral examination in our diagnostic TEM laboratory are fresh<br />

unfixed samples such as scabs, warts, gut and faecal samples.<br />

Materials & methods<br />

For viral diagnosis we perform the negative staining procedure.<br />

This is a simple, rapid and well suited technique for the<br />

examination <strong>of</strong> small particulate suspensions. For viewing the<br />

particulate specimens for organisms such as viruses, a support<br />

membrane must first be prepared onto our TEM grids. This will<br />

provide a support platform to hold the small particles. Examples<br />

<strong>of</strong> support films we may use include Formvar, Collodion, Butvar,<br />

and Piol<strong>of</strong>orm. Once the grids are coated with the films, they are<br />

then stabilized by evaporating a thin carbon coat over them to<br />

render them conductive and keep them from melting in the<br />

electron beam followed by plasma glowing, to ensure the grids<br />

are highly hydrophilic, to obtain maximum sample adsorption The<br />

received samples are now ground in the presence <strong>of</strong> a phosphate<br />

buffer to break up the tissue and release any viral particles which<br />

may be present. A drop <strong>of</strong> the ground suspension is then<br />

adsorbed onto the coated grid for 30 seconds. After wicking dry<br />

with a piece <strong>of</strong> filter paper the grid with sample now adsorbed to<br />

the surface is then floated on a drop <strong>of</strong> negative stain for about<br />

one minute. The most common negative stains are 1% (60 mM)<br />

aqueous uranyl acetate, pH 2-4.5, and 2% (5 mM)<br />

phosphotungstic acid, pH adjusted to 6.6 with NaOH. After<br />

approximately 15 seconds on the negative stain any excess stain<br />

is wicked away and the grid air dried after which it is ready for<br />

examination by TEM. Structures on the grid are surrounded and<br />

stabilized by the drying stain, thus they appear as transparent,<br />

highly detailed negative images within a dark halo <strong>of</strong> stain.<br />

Results<br />

Amongst the most common viruses we diagnose are the<br />

poxviruses (Fig 1-3). These viruses infect both vertebrate and<br />

invertebrate animals and are <strong>of</strong>ten detected from cattle, sheep<br />

and goats causing external scabby lesions. The virus group is<br />

well known as it includes smallpox (variola) an orthopoxvirus.<br />

Poxviruses are regarded as the major contributor responsible for<br />

the decline in the UK <strong>of</strong> the indigenous Red Squirrel (Sciurus<br />

vulgaris). This is due to its susceptibility to Squirrel pox virus<br />

(SQPV). This invariable fatal disease was first reported in 1981<br />

(1) using TEM at the AHVLA. SQPV is approximately the same<br />

size and shape (ovoid) as the parapoxvirus and is differentiated<br />

using TEM due to differences in the spiral coat (longitudinal as<br />

opposed to transverse). TEM is also used to detect ‘enteric<br />

viruses’ which are an important, but diverse, group <strong>of</strong> viruses<br />

found in the intestinal tract <strong>of</strong> animals (and humans). Examples<br />

which we commonly detect are rotavirus and adenovirus.<br />

Adenovirus (Fig.4) is also an emerging problem contributing to<br />

the decline <strong>of</strong> the Red Squirrel and was first reported in 1997 (2)<br />

using TEM at the AHVLA. As mentioned above, the “open view”<br />

occasionally detects mixed viral infections. A published example<br />

(3) being both parapox and calicivirus detected in a scab from a<br />

Grey Seal (Halichoerus grypus). Calicivirus is also the cause <strong>of</strong><br />

Rabbit haemorrhagic disease or viral haemorrhagic disease<br />

(VHD). This is a highly infectious and <strong>of</strong>ten fatal disease that<br />

affects wild and domestic rabbits and was a notifiable disease in<br />

the UK for several years during the 1990s.<br />

Fig. 1. Squirrel poxvirus, 2. Parapoxvirus, 3. Orthopoxvirus, 4.<br />

Adenovirus.<br />

Discussion & conclusions<br />

TEM remains essential for certain diagnostic aspects <strong>of</strong> Virology<br />

(and bacteriology). It was and still is necessary for new virus<br />

characterization and for the initial identification <strong>of</strong> unknown viral<br />

agents in particular outbreaks. This especially applies to<br />

veterinary virology, in which several agents are continuously<br />

being identified with this technique over the past few years. It is<br />

also recommended by regulatory agencies for investigations <strong>of</strong><br />

the viral safety <strong>of</strong> biological products and/or cells. The nature <strong>of</strong><br />

the samples to be analyzed can be tremendously diverse, from<br />

body fluids or biopsies performed or scabs, warts, gut and faecal<br />

samples. An additional advantage <strong>of</strong> electron microscopy is<br />

through its reassurance in having the picture <strong>of</strong> the screened<br />

virus and “a picture is worth a thousand words”. The results by<br />

TEM are therefore <strong>of</strong>ten regarded by many as the 'Gold<br />

Standard', as viral particles are actually observed. The “open<br />

view” approach <strong>of</strong> electron microscopy permits rapid and “catchall”<br />

detection <strong>of</strong> viruses, if these are present in a detectable<br />

concentration, and makes it especially useful as demonstrated<br />

here for the identification and diagnosis <strong>of</strong> various animal viruses,<br />

as well as being used in the initial identification <strong>of</strong> unknown viral<br />

agents in particular disease emergencies and outbreaks and/or in<br />

suspected bioterrorism.<br />

References<br />

1. Scott, A. C., Keymer, I. F. and Labram, J. (1981). Parapoxvirus infection<br />

<strong>of</strong> the red squirrel (Sciurus vulgaris). Vet. Rec. 109, 202.<br />

2. Sainsbury, A.W., Adair, B., Graham, D., Gurnell, J., Cunningham, A.A.,<br />

Benko, M. and Papp, T. (2001). Isolation <strong>of</strong> a novel adenovirus associated<br />

with splenitis, diarrhoea, and mortality in trans-located red squirrels,<br />

Sciurus vulgaris. Verhandlungs Bericht über die Erkrankung der Zootiere<br />

40, 265-270.<br />

3. Stack M.J., Simpson V.R. and Scott AC (1993). “Mixed poxvirus and<br />

calicivirus infections <strong>of</strong> grey seals (Halichoerus grypus) in Cornwall.” Vet.<br />

Rec. 132: 163-165.


S1 - O - 10<br />

A BEAD BASED MULTIPLEX IMMUNOFLUOROMETRIC ASSAY FOR SCREENING AND<br />

CONFIRMATION OF ALL MAJOR PRION TYPES IN SHEEP<br />

Yue Tang 1 , Jan PM Langeveld 2 , Adriana Gielbert 1 , Jorg G Jacobs 2 , Thierry Baron 3 , Olivier Andreoletti 4 , Takashi<br />

Yokoyama 5 , Alex Bossers 1 , Maurice J Sauer 2<br />

1<br />

Animal Health and Veterinary Laboratories Agency, New Haw, Addlestone, Surrey KT15 3NB, UK<br />

Central Veterinary Institute <strong>of</strong> Wageningen UR, Lelystad, The Netherlands<br />

Agence Francaise de Sécurité Sanitaire des Aliments Lyon, Lyon, France<br />

UMR INRA ENVT 1225, 31076 Toulouse, France<br />

National Institue <strong>of</strong> Animal Health,Tsukuba, Ibaraki 305-0856, Japan<br />

Scrapie, discrimination, TSE, BSE, prion, multiplex, assay<br />

Introduction<br />

Prion diseases or transmissible spongiform encephalopathies<br />

(TSEs) in small ruminants are presented in many forms: classical<br />

scrapie, Nor98/atypical scrapie, CH1641 scrapie and bovine<br />

spongiform encephalopathy (BSE). The discrimination between<br />

BSE and CH1641 is especially <strong>of</strong> interest for food safety reasons,<br />

but also methods that can directly co recognize atypical<br />

Nor98/scrapie cases are lacking.<br />

Materials & methods<br />

We previously described a multiplex immun<strong>of</strong>luorometric assay<br />

(mIFMA), based on a bead array flow cytometry technology,<br />

which provided, in a single assay, discrimination between BSE (in<br />

cattle and sheep) and classical scrapie (Tang et al., 2010). In this<br />

study, we extended the mlFMA assay to differentiate classical<br />

scrapie, atypical scrapie, BSE (experimentally infected sheep and<br />

naturally infected cattle) and CH1641 (both experimental and<br />

natural CH1641-like infections in sheep). Three prion protein<br />

(PrP) specific capture antibodies were used, two distinct<br />

N-terminus specific antibodies 12B2 and 9A2, and core specific<br />

antibody 94B4 (Fig. 1). Western blotting procedures were as<br />

described for discriminating bovine BSE types (Langeveld et al.,<br />

2011).<br />

Figure 1. The principle <strong>of</strong> discrimination <strong>of</strong> BSE/CH1641, scrapie<br />

and atypical scrapie using multiplex IFMA.After PK digestion,<br />

9A2, 12B2 and 94B4 antibodies can all bind to classical scrapie<br />

PrP res ; only 9A2 and 94B4 bind substantially to BSE and CH1641<br />

PrP res ; only 12B2 and 9A2 bind substantially to atypical scrapie<br />

PrP res<br />

.The capture antibodies 12B2 for amino acid residues<br />

93 - 97, 9A2 for ovine PrP N-terminal amino acid residues 102104,<br />

and 94B4 for amino acid residues 190-197, are coupled to<br />

distinct bead types. The reporter antibody Sha31 (epitope;<br />

residues 148-155) is biotinylated. Theoretical idealised pr<strong>of</strong>iles <strong>of</strong><br />

MFI readings, associated with 12B2 (black filled bar), 9A2<br />

(unfilled bar) and 94B4 (grey filled bar) respectively, are indicated<br />

on the right side <strong>of</strong> the simplified representation <strong>of</strong> the PrP res<br />

protein primary structure. CH1641 may be differentiated from<br />

BSE by analysis <strong>of</strong> a minor 14 kDa C-terminal fragment and also,<br />

the 9A2 region might be different from BSE which would result in<br />

lower 9A2 values than in BSE.<br />

suggesting that major PrP Sc cleavage <strong>of</strong> the N-terminus occurs<br />

further towards the C terminus in CH1641 than in BSE. The ratios<br />

<strong>of</strong> 12B2/94B4 and 9A2/94B4 were similar between experimental<br />

CH1641 and CH1641-like cases, although two CH1641 like<br />

subjects displayed slightly elevated ratios <strong>of</strong> both 12B2/94B4 and<br />

9A2/94B4. To verify this finding for PrP res , mass spectrometry<br />

quantification to determine the absolute abundance <strong>of</strong> the<br />

peptides associated with all three antibody binding regions<br />

confirmed that there was a 2.2 fold reduction <strong>of</strong> peptides<br />

containing the 9A2 epitope for experimental CH1641 PrP res<br />

in<br />

comparison to BSE PrP res . Observation <strong>of</strong> reduced PrP res<br />

may<br />

serve as a new marker for CH1641. Finally, Western blotting<br />

further confirmed that the CH1641-like cases have to be<br />

considered as a TSE class different from the experimental<br />

CH1641 cases.<br />

Discussion & conclusion<br />

This mIFMA may thus provide the basis for simplified TSE<br />

diagnosis with capability for simultaneous screening and<br />

differential diagnosis.<br />

Acknowledgements<br />

Financial support for this work was provided by the Department<br />

for Environment, Food and Rural Affairs (Defra, UK; project<br />

SE1700/ CSA 7335). JPML is partly supported by the Dutch<br />

Ministry <strong>of</strong> Agriculture, Nature and Food Quality project number<br />

WOT-01-002-14 001.01. JPML and OA are partly supported by<br />

the EU STREP project GoatBSE FOOD15 CT-2006-36353. We<br />

thank Linda Davis and Sharon Everitt for Western blotting<br />

analysis. The help <strong>of</strong> Dr Marion Simmons and the VLA TSE<br />

Archive team for provision <strong>of</strong> samples is gratefully acknowledged.<br />

References<br />

1. Tang, Y, Thorne, J, Whatling,K, Jacobs, J, Langeveld, J and Sauer, M.<br />

2010. A single step multiplex immun<strong>of</strong>luorometric assay for differential<br />

diagnosis <strong>of</strong> BSE and scrapie. J. Immunol Meth. 356:29–38.<br />

2. Langeveld, JPM, Erkens, JHF, Rammel, I, Jacobs, JG, Davidse, A, van<br />

Zijderveld, F, Bossers, A, and Schildorfer, H. 2011. Four independent<br />

molecular prion protein parameters for discriminating new cases <strong>of</strong> C, L,<br />

and H BSE in cattle. J Clin Microbiol., 49:3026–3028.<br />

Results<br />

All three antibodies were shown to bind classical scrapie PrP res<br />

strongly, whereas in Nor98/atypical scrapie PrP res only 12B2 and<br />

9A2 binding was observed. PrP res binding <strong>of</strong> 12B2 was low for<br />

both BSE and CH1641, as expected. Furthermore, analysis <strong>of</strong><br />

serially diluted samples indicated that the assay provided a<br />

similar level <strong>of</strong> sensitivity for atypical scrapie as that found using<br />

a well established commercial test. Unexpectedly, 9A2 binding to<br />

CH1641 PrPres was reduced by 2.1 fold both for experimental<br />

CH1641 and CH1641 like scrapie when compared with BSE,


S1 - O - 11<br />

USING ORAL FLUID FOR THE SEROLOGICAL MONITORIZATION OF PRRSV CIRCULATION IN A<br />

GROUP OF INFECTED GILTS<br />

Martos-Raich, Alba 1 , Coma-Oliva, Ester 1 ; Serra-Martínez, Jordi 2 ; Barrera-Toro, Xavier 3 ; Planasdemunt-Regàs,<br />

Llorenç 3 ; Maldonado-García, Jaime 1 ; Porquet-Garanto, Lourdes 1 ; Rebordosa-Trigueros, Xavier 1 .<br />

1 HIPRA 17170 Amer, Girona, Spain.<br />

2 Bi<strong>of</strong>ar Laboratoris, S.L 08261 Cardona, Barcelona, Spain.<br />

3 AVP Planasdemunt i Associats 17400 Breda, Girona, Spain<br />

Introduction<br />

The use <strong>of</strong> oral fluid-based assays is proving every day that is<br />

well suited for monitoring <strong>of</strong> different pathogens in farm animals 1 .<br />

In the case <strong>of</strong> PRRSV different studies have shown that pig oral<br />

fluid (OF) is a suitable sample for monitoring both viremia (PCR)<br />

and sero-conversion (ELISA) after infection 2 . In this study a new<br />

methodology to adapt the existing CIVTEST SUIS PRRS E/S and<br />

A/S ELISA kits (Hipra) to be used with OF matrix instead <strong>of</strong><br />

serum was evaluated under field conditions. The ultimate goal <strong>of</strong><br />

this study was to compare the performance <strong>of</strong> the ELISA kits<br />

using individual or pooled serum samples, as well as OF from<br />

groups <strong>of</strong> animals.<br />

PPRS virus, oral fluid, ELISA, alternative for monitoring<br />

Materials & methods<br />

The study included a single group <strong>of</strong> 10 gilts coming from a<br />

PRRSV-free farm (as confirmed by PCR and ELISA at the<br />

beginning <strong>of</strong> the study). The animals were infected in isolation<br />

using a field strain <strong>of</strong> PRRSV Type 1 on day 0. The study lasted<br />

for 9 weeks (wk) with weekly sampling <strong>of</strong> blood from each<br />

individual and OF samples from the group. The presence <strong>of</strong><br />

antibodies to PRRSV Type 1 and 2 was evaluated by using the<br />

CIVTEST SUIS PRRSV E/S and CIVTEST SUIS PRRS A/S<br />

ELISA kits respectively (Hipra) and also by Western blot based in<br />

a European strain. Individual serum samples were tested for the<br />

presence <strong>of</strong> viral RNA by Real Time RT-PCR 3 . Serums samples<br />

were analyzed individually and as a pool <strong>of</strong> 10 (pool/10). For OF<br />

serology different conjugates (anti-IgG1,-IgG2, -IgA) for the<br />

ELISA were evaluated.<br />

Results<br />

The incubation conditions for OF were fixed at 4 ºC <strong>of</strong> a ½<br />

dilution <strong>of</strong> the sample. In all cases an IRPC <strong>of</strong> 20.0 was used as<br />

a cut-<strong>of</strong>f (as defined in CIVTEST for individual serum). As seen in<br />

Table I, results from individual serum showed 80% <strong>of</strong> positive<br />

animals at wk3 (100% at wk4). The IRPC values for the pool/10<br />

and OF samples showed a value above 20 at wk4. Both, the<br />

mean IRPC value <strong>of</strong> the group and the IRPC <strong>of</strong> the pool/10<br />

increased until the end <strong>of</strong> the study. By contrast, IRPC values<br />

obtained for the OF sample showed a maximum at wk4, and then<br />

dropped down till the end <strong>of</strong> the trial. No reactivity was observed<br />

with the A/S kit (data not shown). The use <strong>of</strong> different conjugates<br />

for the analysis <strong>of</strong> OF samples indicated that the most abundant<br />

type <strong>of</strong> Ig after infection was IgA (already detectable at wk1 postinfection;<br />

Figure 1). The kinetics <strong>of</strong> IgG1 showed sero-conversion<br />

around 3-4 weeks after infection. IgG2 levels were nondetectable<br />

(data not shown). The maximum sensitivity (IRPC<br />

above 20 at wk1) was achieved by using a combination <strong>of</strong> IgG1<br />

plus IgA conjugates.<br />

Figure1. OD450 values obtained with the oral fluid weekly<br />

samplings using conjugates with different antibody-type<br />

specificity<br />

Discussion & conclusions<br />

CIVTEST SUIS PRRS E/S has shown a good adaptation to the<br />

OF matrix. This sample evidenced antibody dynamics similar to<br />

those observed with the pool/10 serum sample, representing a<br />

suitable alternative for monitoring PRRSV circulation in pig herds.<br />

The use <strong>of</strong> different channels (IgG1 or IgA) for the analysis <strong>of</strong> the<br />

OF sample would allow to asses maximum earliness in the<br />

detection <strong>of</strong> infection or detection <strong>of</strong> the fall in post-infection<br />

immunity.<br />

References<br />

1. Prickett, J.R. et al: 2010, Anim Hlth Res. Rev. 11(2): 207-216.<br />

2. Kittawornrat, A. et al: 2010, Virus Res. Dec;154(1-2):170-176.<br />

3. Martínez, E. et al: 2008, Res. Vet. Sci. 85: 184-193<br />

Table I. Serology results obtained with serum and oral fluid using<br />

the CIVTEST SUIS PRRS E/S at weekly intervals after infection<br />

(IRPC values above 20.0 appear in shaded boxes)<br />

PRRS E/S<br />

% Pos<br />

(ELISA)<br />

wk<br />

0<br />

wk1 wk2 wk3 wk4 wk5 wk6 wk7 wk8 wk9<br />

0% 0% 0% 80% 100% 100% 100% 80% 100% 100%<br />

Mean (IRPC) -1,49 -1,38 8,64 24,32 41,75 46,56 70,47 50,96 72,67 72,00<br />

Pool/10<br />

(IRPC)<br />

-1,80 -2,09 4,19 11,52 23,95 54,09 57,88 48,48 68,97 87,19<br />

OF (IRPC) -5,20 -4,22 0,13 14,08 46,65 31,48 35,19 29,74 20,52 32,66<br />

OF/IgG1+IgA<br />

(IRPC)<br />

4,33 44,02 60,07 71,66 99,72 96,37 97,72 86,04 80,60 99,95


S1 - O - 12<br />

ENHANCED DETECTION OF BOVINE RESPIRATORY VIRUSES BY INCLUSION OF COHORT<br />

ANIMALS<br />

Ronan O’Neill, Emily Connaghan, Jean Mooney<br />

CVRL, Virology Division, Backweston, Celbridge, Ireland<br />

Respiratory virus, bovine, multiple, PCR, diagnostics<br />

Introduction<br />

Bovine respiratory disease (BRD) in calves remains a hugely<br />

significant problem to Irish cattle breeders. Until recently BRD<br />

diagnostics were quite limited but this has improved due to the<br />

adoption <strong>of</strong> molecular techniques, the introduction <strong>of</strong> a pooled<br />

swab matrix and a parallel programme <strong>of</strong> veterinary education.<br />

These advances have allowed a much more accurate<br />

representation <strong>of</strong> the dynamics <strong>of</strong> BRD in terms <strong>of</strong> time <strong>of</strong> year<br />

and pathogen involved. This study provides support to the<br />

clinical judgement <strong>of</strong> vets in making an initial pathogen<br />

identification and initiating remedial actions at the earliest<br />

stage.<br />

Materials & methods<br />

The analysis is based on data generated over the three year<br />

period <strong>of</strong> 2009 to 2011. The calf age–group dataset consisted<br />

<strong>of</strong> 3822 PCR tests from 753 submissions, representing 30%<br />

(n=12642) and 31% (n=2405) <strong>of</strong> total live animal respiratory<br />

submissions over the period. The sample matrix used<br />

throughout was cotton swabs, without transport media.<br />

Veterinarians were encouraged to collect up to six swabs from<br />

the index and cohort animals which were then pooled in the<br />

lab for testing (same cost) so the analysis includes results<br />

based on individual swabs and results based for pooled<br />

swabs.<br />

The normal respiratory panel consisted <strong>of</strong> five real-time PCR<br />

tests for Bovine Herpesvirus 1 (BHV1) (1), Bovine Coronavirus<br />

(BoCo)(2), Bovine Respiratory Syncytial virus (BRSV)(3),<br />

Bovine Viral Diarrhoea virus (BVDv)(4) and Parainfluenza 3<br />

virus (PI3). All swabs were also assessed for nucleic acid yield<br />

/polymerase inhibition using a B-actin internal control.<br />

Results<br />

In calves overall, BHV1 was detected in 8% <strong>of</strong> swabs tested,<br />

BoCo in 37%, BRSV in 11%, BVDv in 4% and PI3 in 9%.<br />

However respiratory samples in this age group are extremely<br />

seasonal with 46% <strong>of</strong> samples in months November,<br />

December and January (Figure 1). In the calf age group, the<br />

months with the highest rates <strong>of</strong> detection were December for<br />

BRSV (44%) and PI3 (26%), November for BoCo (18%) and<br />

January for BHV1 (15%) and BVDv (15%).<br />

In calves there were 2622 PCR tests done on individual swabs<br />

and 1199 PCR tests done on pooled swabs – sufficient<br />

numbers to allow statistical comparison. On average using<br />

pooled swabs rather than individual swabs increased the virus<br />

detection rate for BHV1 by a factor <strong>of</strong> 2.4 (p


S1 - O - 13<br />

THE FIRST YEAR OF OBLIGATORY BVD CONTROL IN GERMANY – DIAGNOSTIC STRATEGIES,<br />

RESULTS AND EXPERIENCES<br />

Horst Schirrmeier, Günter Strebelow, Martin Beer<br />

Institute <strong>of</strong> Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald-Insel Riems, Germany<br />

Pestivirus, disease controlj<br />

Introduction<br />

Since November 2004, Bovine viral diarrhea/Mucosal disease<br />

(BVD/MD) has been a notifiable disease in Germany. An<br />

obligatory BVD-legislation and control strategy <strong>of</strong> the Federal<br />

Government is active since the 1st <strong>of</strong> January 2011. The<br />

implemented eradication strategy is focused on the direct<br />

detection <strong>of</strong> persistently infected (PI) animals by using antigen<br />

ELISAs or real-time RT-PCR.<br />

References<br />

1. Anonymus (2008). „Verordnung zum Schutz der Rinder vor einer<br />

Infektion mit dem Bovinen Virusdiarrhoe-Virus (BVDV-Verordnung) v.<br />

11.12.2008 (BGBl. I S. 2461), neugefasst durch Bek. v. 4.10.2010 I 1320,<br />

1498,geändert durch Art. 1 V v. 17.12.2010 I 2131, diese geändert durch Art. 1 V<br />

v. 31.5.2011 I 1002<br />

2. Schirrmeier, H. Strebelow, G. (<strong>2012</strong>). BVD-Pflichtbekämpfung – Wie ist<br />

die epidemiologische Situation in Deutschland?, LBH, 6. Leipziger<br />

Tierärztekongress – Tagungsband 3: 95-98<br />

Materials & methods<br />

The major goal <strong>of</strong> the obligatory control program is a fast and<br />

efficient reduction <strong>of</strong> the prevalence <strong>of</strong> PI animals and the<br />

establishment <strong>of</strong> so-called “unsuspicious (virus free) animals” and<br />

farms with a certified status. Therefore, four basic rules were<br />

defined:<br />

1. Obligatory investigation <strong>of</strong> all newborn calves for BVDV<br />

(antigen/genome) before the age <strong>of</strong> 6 months<br />

2. Efficient elimination <strong>of</strong> all PI animals.<br />

3. Trade with certified “unsuspicious animals” only<br />

4. Prevention <strong>of</strong> reinfections by qualified measures (e.g.<br />

biosafety and voluntary vaccination)<br />

Approved methods taking the so-called “diagnostic gap”<br />

(method-dependent limitation <strong>of</strong> virus/genome detection due to<br />

BVDV-specific colostral antibodies) into account, and a system <strong>of</strong><br />

measures for quality control and assurance, like pr<strong>of</strong>iciency tests<br />

or the licensing <strong>of</strong> diagnostic assays ensure a high standard <strong>of</strong><br />

BVDV diagnostics within the German program. Ear notches are<br />

the main sample material allowing a reliable and very early<br />

detection and subsequent elimination <strong>of</strong> PI animals. All diagnostic<br />

results are <strong>of</strong>ficially documented in a central data base (HIT).<br />

Additionally, in order to get more information about the molecular<br />

epidemiology <strong>of</strong> BVDV in Germany, more than 400 BVDVpositive<br />

samples or virus isolates from PI animals were collected<br />

and further analysed by sequencing their 5’ untranslated genome<br />

region (5´UTR)<br />

Results<br />

In the last year, more than 6 million tests for the detection <strong>of</strong><br />

BVDV were conducted in German laboratories. Real-time RT-<br />

PCR assays and antigen-capture-ELISAs were used in an<br />

approximately equal amount. 17 410 PI animals had been<br />

detected and were removed which is a percentage <strong>of</strong> 0.14% in<br />

relation to the German cattle population, and <strong>of</strong> 0.36% in relation<br />

to all calves born in 2011. The prevalence <strong>of</strong> PI animals differed<br />

between the German federal states from 0.02% to 0.58%. In<br />

6016 cattle holdings at least one PI animal was detected (4.1%).<br />

Phylogenetic analysis <strong>of</strong> 445 German BVDV isolates clearly<br />

showed that only two BVDV subgroups (BVDV-1b and BVDV–1d)<br />

represented more than 75% <strong>of</strong> all strains investigated. Apart from<br />

this dominating subgroups, BVDV-1f (8.8%), BVDV-1e (3.5%),<br />

BVDV-1h (2.9%), BVDV-1g (0.7%), and BVDV-1k (0.2%) could<br />

be observed. Regional differences were also noticeable. The<br />

percentage <strong>of</strong> BVDV-2 was 5.1%, and the subtype “Germany 2a”<br />

was the dominating subgroup within this genotype.<br />

Discussion & conclusions<br />

During the first year <strong>of</strong> the German compulsory BVDV control<br />

program, more than 17 000 PI animals were detected and<br />

removed. Furthermore, a reduction <strong>of</strong> the number <strong>of</strong> notified PI<br />

cases was obvious during the last three month due to an efficient<br />

elimination at the beginning <strong>of</strong> the year. We therefore conclude,<br />

that the diagnostic concept, focussed on an as early as possible<br />

detection and elimination <strong>of</strong> PI calves is successful. However,<br />

with the progress in BVDV-eradication, the risk and the economic<br />

outcome <strong>of</strong> reinfections have to be taken into consideration, and<br />

accompanying measures like serological monitoring and<br />

vaccination have to be implemented into the complex disease<br />

control system..


S1 - O - 14<br />

EUROPEAN PRRSV – DIAGNOSTIC SOLUTIONS FOR A RAPIDLY MUTATING VIRUS<br />

R. Shah 2 , C. Boss 1 , C. O’Connell 2<br />

1<br />

Life Technologies,Darmstadt, Germany<br />

2<br />

Life Technologies, Austin, USA<br />

Introduction<br />

Porcine Reproductive and Respiratory Syndrome virus (PRRSV)<br />

is a highly infectious disease that is endemic in pigs throughout<br />

Europe. Estimates on its economic impact vary from as little as<br />

€60 per infected animal to as much as €75 for every animal in<br />

production on an annualized basis. Detection and removal <strong>of</strong><br />

infected animals followed by surveillance to avoid re-infection is<br />

the preferred strategy to manage the impact <strong>of</strong> the disease in<br />

European Pig Populations.<br />

The virus is continuously and rapidly evolving, generating “new<br />

variants” and expanding its diversity. Therefore there is a need to<br />

update diagnostic solutions at a pace that provides efficacious,<br />

appropriate and cost competitive solutions to the marketplace.<br />

In 2010, diagnostic studies showed that a mutation in one <strong>of</strong> the<br />

9 open reading frames <strong>of</strong> the North American PRRS viral<br />

genome had rendered our Taqman NA/EU PRRSV reagents<br />

ineffective for detection <strong>of</strong> certain newly mutated variants.<br />

Materials & methods<br />

Laboratories using the Life Technologies Taqman PRRSV assay<br />

sent samples to our lab for sequencing analysis. Several <strong>of</strong> the<br />

questionable samples were sequenced and the resulting<br />

sequence information was used to design a set <strong>of</strong> primers and<br />

probes that were perfect matches to the new sequence. This new<br />

set <strong>of</strong> oligos is refered to as a “patch”.<br />

The new patch was added to the existing assay and optimized so<br />

as to increase sensitivity <strong>of</strong> the North American PRRSV assay<br />

while not affecting sensitivity <strong>of</strong> the European PRRSV assay that<br />

was multiplexed with it.<br />

Results<br />

Over 130 samples from different labs that contained the new<br />

NAPRRSV strain were tested with the patched assay as well as<br />

the old assay to show that the patch really did pick up the new<br />

strain that the old assay was missing. No non specific<br />

interactions were obsereved when the patch was added to the<br />

old assay reagents.<br />

The new patched assay enables sensitive and specific detection<br />

<strong>of</strong> current and old NAPRRSV strains as well as the ability to<br />

subtype EUPRRSV strains from NAPRRSV strains. All<br />

parameters <strong>of</strong> the old assay such as thermal pr<strong>of</strong>ile and PCR<br />

mastermix preparation remain the same.<br />

Discussion & conclusions<br />

Given the rapid mutation rate <strong>of</strong> PRRSV, the creation and<br />

maintenance <strong>of</strong> a diagnostic solution is a moving target.<br />

Therefore collaborations across veterinary laboratories and other<br />

key stakeholders is essential for monitoring the degree <strong>of</strong><br />

divergence that is occurring and to aid in building robust<br />

diagnostic solutions.<br />

This North American example provides a template, combined<br />

with newly available technologies such as whole genome<br />

sequencing, for creating a regionally appropriate assay and<br />

monitoring program for the detection <strong>of</strong> rapidly mutating viruses<br />

like PRRSV.<br />

References<br />

Iowa State University, Iowa Select Farms, University <strong>of</strong> Minnesota<br />

PRRSV


S1 - O - 15<br />

SEROLOGICAL ANALYSIS AND MONITORING OF IBR<br />

IS IT POSSIBLE TO CONTROL IBRgE ANTIBODIES IN A BULK TANK MILK?<br />

Xavier Rebordosa-Trigueros, Ester Coma Oliva, Santiago Casademunt Garre, Lourdes Porquet-Garanto*<br />

HIPRA, 17170 Amer (Girona), SPAIN<br />

IBRgE, Bulk Tank Milk (BTM), lower prevalence detection.<br />

Introduction<br />

The detection <strong>of</strong> antibodies in Bulk Tank Milk (BTM) provides<br />

an easy and cheap methodology for monitoring the health<br />

status <strong>of</strong> the herd 1 . The application <strong>of</strong> this methodology to the<br />

detection <strong>of</strong> IBR-gE antibodies presents some limitations.<br />

Blocking IBRgE-ELISAs have low sensitivity 2 . So, IBRgE-<br />

ELISAs are only capable <strong>of</strong> detecting positive tanks when the<br />

prevalence in the animals in production is greater than 15-<br />

20% 3 . Under these conditions, gE detection in the BTM is not<br />

adequate for monitoring the tank and even less so for the<br />

classification <strong>of</strong> the farms. The objective <strong>of</strong> this work is to<br />

develop and validate an IgG concentration method to increase<br />

the sensitivity <strong>of</strong> the IBR-gE detection systems in BTM<br />

sample.<br />

Materials & methods<br />

We have developed a simple methodology that in less than 60<br />

minutes can concentrate up to 30 times the IgGs from a<br />

sample <strong>of</strong> 5.0 ml <strong>of</strong> BTM. To validate this methodology 17<br />

dairy-farms from the NW <strong>of</strong> Spain with a known IBR status<br />

were selected (8 vaccinated and 9 non-vaccinated). From<br />

each farm we have obtained Individual serum <strong>of</strong> all the<br />

animals in production and a sample <strong>of</strong> each tank. Taking into<br />

account the IBRgE results in sera the real prevalence <strong>of</strong> each<br />

tank was calculated. BTM samples were also analysed for gE<br />

pure and concentrated.<br />

Results<br />

The actual prevalence against gE, calculated from individual<br />

serum samples in the 17 selected farms was: 0% (5 farms),<br />

4%, 15%, 17%, 18% 19%, 21%, 24%, 26%, 33%, 43% and<br />

44% (2 farms). By using pure milk (non-concentrated) only 5<br />

tanks were consistently detected as positives to gE. These<br />

tanks corresponded to the farms with the higher prevalence<br />

values (from 26% to 44%). By contrast, applying to the same<br />

samples the methodology <strong>of</strong> concentration <strong>of</strong> IgGs, all tanks<br />

were detected positive, even the lowest prevalence one. This<br />

methodology did not affect the 5 negative tanks that remained<br />

negative. Fig. 1.<br />

Figure 1. BTM ELISA IBRgE titres from 17 dairy-farms.<br />

Prevalence detection from 0 to 44%. Column □ nonconcentrated<br />

BTM (pure milk) and Column ■ salting-out<br />

concentrated BTM (IgGs concentration). CIVTEST ®<br />

BOVIS<br />

IBRgE interpretation: Higher than 20.0 (%INH) = positive<br />

sample, Lower than or equal to 20.0 = negative sample.<br />

Discussion & conclusions<br />

These results indicate that the proposed IgG concentration<br />

system does not affect the specificity <strong>of</strong> the ELISA but<br />

increases the sensitivity, having allowed in this study to detect<br />

tanks with the lower prevalence (4%).<br />

References<br />

1.Nylin, B., Strøger, U., Rønsholt, L. “A retrospective<br />

evaluation <strong>of</strong> a Bovine Herpesvirus-1 (BHV-1) antibody ELISA<br />

on bulk-tank milk samples for classification <strong>of</strong> the BHV-1<br />

status <strong>of</strong> Danish dairy herds”. (2000). Preventive Veterinary<br />

Medicine. 47, 91-105.<br />

2. Kramps, J.A., Banks, M., Beer, M., Kerkh<strong>of</strong>s, P., Perrin, M.,<br />

Wellenberg, G.J. and Van Oirschot, J.T. (2004) “Evaluation <strong>of</strong><br />

tests for antibodies against bovine herpesvirus 1 performed in<br />

national reference laboratories in Europe”. Vet Microbiol.,<br />

8;102(3-4):169-81.<br />

3. Wellenberg, G.J., Verstraten, E.R.A.M., Mars, M.H., Van<br />

Oirschot, J.T. (1998) “ELISA detection <strong>of</strong> antibodies to<br />

glycoprotein E <strong>of</strong> bovine herpesvirus 1 in bulk milk samples”.<br />

Vet Rec. 142(9), 219 – 20.


S1 - O - 16<br />

DEVELOPMENT AND VALIDATION OF A REAL-TIME PCR ZEN GEL MIX FOR THE DIAGNOSIS AND<br />

QUANTIFICATION OF COXIELLA BURNETII<br />

Aleida Villa 1 , José L Arnal 1 , Daniel Serrano 1 , Alfredo A Benito 1 , Rafael Baselga 1 .<br />

EXOPOL Autovacunas y Diagnóstico. Pol. Río Gállego, 50840 San Mateo de Gállego Zaragoza, España. Tel 976 69 45 25, exopol@exopol.com<br />

Coxiella burnetii, qPCR, validation, diagnostics<br />

Introduction<br />

Coxiella burnetii is the ethological agent <strong>of</strong> Q Fever a zoonotic<br />

and emergent disease with implications for public and animal<br />

health. Reliable diagnosis <strong>of</strong> C. burnetii is the vertebral point for<br />

the control <strong>of</strong> the spread <strong>of</strong> these bacteria highly infectious.<br />

Here we describe a novel PCR (qPCR) assay simple, rapid,<br />

sensitive, specific, robust and well validated for routine diagnosis<br />

<strong>of</strong> Coxiella burnetii based in a new type <strong>of</strong> "ZEN" double<br />

quencher probe (1) that increases the accuracy, sensitivity and<br />

significantly decreases the background fluorescence, reducing<br />

Cq values consistently with a greater precision and signal. In this<br />

qPCR all the reaction components (master mix, enzymes,<br />

specific primers, probes, buffer and internal controls) have been<br />

stabilized within a plate using a technology called gelation (2)<br />

allowing their use at room temperature and in a "ready to use”<br />

format.<br />

Materials & methods<br />

To ensure high performance and bio-safety on the DNA process<br />

extraction, the procedure was performed in an automated Lab<br />

Turbo robot (Taigen, Bioscience, Taiwan) and compared to an<br />

established manual DNA extraction method (Mo Bio). The<br />

samples were run .through the kits according to the<br />

manufactures` instructions. The qPCR set up protocol were<br />

single consisting <strong>of</strong> RNAse/ DNAse free water and 5µl <strong>of</strong> DNA<br />

sample Amplification reactions were carried out on a MiniOpticon<br />

RT System CFB3120 thermal cycler (Bio Rad, s<strong>of</strong>tware CFX<br />

manager 2.0). The assay targets a C. burnetii specie specific<br />

region <strong>of</strong> the Trans gene was selected, the internal probe trans-p<br />

was labeled with (FAM/ZEN/Iowa Black). The capacity <strong>of</strong> the<br />

ZEN Coxiella burnetii Gel Mix to discriminate between target and<br />

non target microorganisms was assessed using 1ng/µl <strong>of</strong><br />

genomic DNA from various sources: including 4 reference strain<br />

and 73 field strain and 1407 clinical cases; from sheep, goat and<br />

bovine origin: (milk, feces, vaginal exudates, tissue from<br />

abortions and parturient cows, water and other environmental<br />

samples) and 40 strains <strong>of</strong> entities prevalent in ruminants. The<br />

positives samples obtained by qPCR were used for C. burnetii<br />

isolation in VERO cell line. The obtained strains were identified<br />

by immun<strong>of</strong>luorescence and quantified by qPCR, and then<br />

confirmed by inoculation in chick embryo yolk sac and genotyping<br />

in the reference center <strong>of</strong> Coxiella <strong>of</strong> Institute Carlos III in Madrid.<br />

The parameters <strong>of</strong> analytical and diagnostic sensitivity; analytical<br />

and diagnostic specificity; precision intra-assay and inter-assay<br />

were evaluated.<br />

Results<br />

ZEN qPCR Coxiella Gel mix demonstrated an excellent ability to<br />

quantify, defined by its wide dynamic range (at least 7 log units),<br />

with a minimum detection limit equivalents to 2 genome copies <strong>of</strong><br />

C. burnetii and a maximum value <strong>of</strong> > 10 10 . The slopes <strong>of</strong> the<br />

linear regression curves were calculated within an interval <strong>of</strong> 7-<br />

log dilutions and were similar to the theoretical optimum <strong>of</strong> -3.32<br />

(-3.475), showing an effective amplification efficiency (E = 0.90),<br />

and R 2 values close to 1 (0.9988) indicating that the assay was<br />

extremely linear. Confidence intervals based on the standard<br />

deviations <strong>of</strong> the Cq values not overlapped, up to 10 target<br />

molecules, indicating that it is possible a reliable quantification<br />

above this limit. The intra-assay CVs (5 replicates and 5 assays)<br />

ranged between 9.8% and 31%. The inter-assay CVs obtained<br />

was from 12.6% to 24.5% after 5 independent trials. The<br />

sensitivity <strong>of</strong> the real-time quantitative PCR was 2, 6 times higher<br />

than that isolation in VERO cell line. All isolates (73/194) were<br />

phase I and highly pathogenic in chick embryos<br />

Table 1. qPCR positive tissue from abortions Vs Isolation <strong>of</strong> C.<br />

burnetii<br />

Species Positive<br />

qPCR cases<br />

C. burnetii<br />

isolates cases<br />

% <strong>of</strong> isolation Vs<br />

qPCR positive cases<br />

Bovine 70 41 58,6<br />

Caprine 31 10 32,3<br />

Ovine 93 22 23,7<br />

The results <strong>of</strong> specificity detection <strong>of</strong> 40 species <strong>of</strong> other common<br />

pathogens agents proved to be negative. The positive rates <strong>of</strong><br />

Coxiella burnetii shedding in abortions were 27.21% (123/452),<br />

while in post partum cases was very low 5.99% (22/345). The<br />

excretions <strong>of</strong> C. burnetii in ruminants herds in productions<br />

resulted: vaginal swabs 26.81% (74/276); milk tanks 31.58%<br />

(12/38); pooled milks 22.22% (40/180); bovine feces 2.17(2/90).<br />

C. burnetii was also detected in ground samples 33.3% (1/3), but<br />

not on in water (0/10) and cereal (0/3) samples.<br />

Discussion & conclusion<br />

Abortions and/or birth <strong>of</strong> weak <strong>of</strong>fspring are the most serious<br />

consequence by C. burnetii infection in animals. Abortions<br />

usually occur in the second half <strong>of</strong> gestation (3) . Infected animals<br />

are usually asymptomatic and is not possible establish any<br />

relationship between antibody titers and individuals excretion.<br />

Studies by Rousset et al. (4)<br />

showed that a non negligible<br />

proportion <strong>of</strong> seronegative animals in delivered normally could<br />

excrete C. burnetii to remain seronegative and spread massive<br />

amounts <strong>of</strong> microorganisms during calving. C. burnetii was<br />

detected in 26.42% <strong>of</strong> cases <strong>of</strong> abortions and 61.98% <strong>of</strong> these<br />

viable bacteria were recovered in phase I with high pathogenicity<br />

and 11.36% <strong>of</strong> them were characterized to belong to genotype III,<br />

where are included strain from animals, ticks and those that<br />

cause acute Q fever in humans. These data shows that Coxiella<br />

infection in rumiants animals have a strong impact on<br />

environmental contamination and its a potential dangerous<br />

source for humans infection, even if people have or not contact<br />

with infected animals. The high number <strong>of</strong> Coxiella positive cases<br />

found in this study claim the need for stablish strict health and<br />

vaccination programs.The qPCR developed and validated for us<br />

and used in this studies has proved a wonderful diagnostic tool<br />

and is available commercially.<br />

References<br />

1.- CORALVILLE, IA. ZEN double-quenched probes can now be<br />

combined with a range <strong>of</strong> fluorescent dyes in: www.idtdna.com<br />

2.- Biotools. Método de estabilización de Biomoléculas y mezclas<br />

de reacción complejas mediante la gelificación Patentes<br />

Internacionales PCT ES2002/000109 PCT ES2004/000024.<br />

3. Sitio Argentino de Producción Animal. Enfermedad infecciosa<br />

de los ovinos. S.S.Diab and Uzal F.A. 2007. Disponible en:<br />

www.produccion-animal.com.ar<br />

4. Rousset E, Berri M, Durand B, Dufour P, Prigent M, Delcroix T,<br />

et al. A. Coxiella burnetii shedding routes and antibody response<br />

after Q fever abortion outbreaks in dairy goat herds.Appl Environ<br />

Microbiol. 2008, Nov 14.<br />

5. Guatteo R, Beaudeau F, Joly A and Seegers H. Coxiella<br />

burnetii shedding by dairy cows. Vet Res 2007 Nov-Dec;<br />

38(6):849-60.


S1 - O - 17<br />

RING TEST EVALUATION FOR THE DETECTION OF PRRSV ANTIBODIES IN ORAL FLUID<br />

SPECIMENS USING A COMMERCIAL PRRSV SERUM ANTIBODY ELISA<br />

Apisit Kittawornrat 1 , Chong Wang 1 , Gary Anderson 2 , Andrea Ballagi 3 , Andre Broes 4 , Susy Carman 5 , Kent<br />

Doolittle 6 , Judith Galeota 7 , John Johnson 1 , Sergio Lizano 3 , Eric Nelson 8 , Devi Patnayak 9 , Roman Pogranichniy 10 ,<br />

Anna Rice 3 , Gail Scherba 11 , Jeffrey Zimmerman 1<br />

1 Iowa State University, 2 Kansas State University, 3 IDEXX Laboratories,Inc., 4 Biovet Inc., 5 University <strong>of</strong> Guelph, 6 Boehringer Ingelheim Vetmedica Inc.,<br />

7 University <strong>of</strong> Nebraska, 8 South Dakota State University, 9 University <strong>of</strong> Minnesota, 10 Purdue University, 11 University <strong>of</strong> Illinois<br />

Oral fluid, ELISA, Ring test, PRRSV<br />

Introduction<br />

A commercial PRRS serum antibody ELISA (IDEXX<br />

Laboratories, Inc., Westbrook ME USA) was recently adapted to<br />

detect anti-PRRSV antibody in oral fluid specimens (1). Based on<br />

testing <strong>of</strong> field and experimental samples, diagnostic sensitivity<br />

and specificity was estimated at 94.7% (95% CI: 92.4, 96.5) and<br />

100% (95% CI: 99.0, 100.0), respectively, at a sample-to-positive<br />

(S/P) cut<strong>of</strong>f <strong>of</strong> ≥ 0.40. 1 The purpose <strong>of</strong> this study was to evaluate<br />

the reproducibility and repeatability <strong>of</strong> the PRRS oral fluid ELISA<br />

in a ring test (check test) format.<br />

Materials & methods<br />

A total <strong>of</strong> 276 oral fluid samples were collected, completely<br />

randomized, and sent for testing in 12 collaborating diagnostic<br />

laboratories. In addition to the set <strong>of</strong> oral fluid samples, each<br />

laboratory received the materials required for conducting the test:<br />

ELISA plates (HerdChek® PRRS X3 ELISA, lot #40959-W721),<br />

reagents, positive and negative controls, pre-diluted conjugate<br />

antibody, and a copy <strong>of</strong> the standard operating procedure for the<br />

PRRS oral fluid IgG ELISA. The laboratories tested the samples<br />

and returned the results for analysis. Assay results were<br />

analyzed as S/P ratios, with S/P ratios ≥ 0.40 considered<br />

positive.<br />

Results<br />

Distribution <strong>of</strong> the results from the 12 laboratories is summarized<br />

in Figure 1. As is shown in Figure 1, variation in S/P results<br />

increased as the concentration <strong>of</strong> antibody in the sample<br />

increased. Overall, this had little impact on categorical results.<br />

That is, among the 276 samples tested by the 12 laboratories,<br />

133 samples tested positive in all laboratories; 136 samples<br />

tested negative in all laboratories, and 7 samples had discordant<br />

results (Table 1). With the exception <strong>of</strong> sample #7, a discordant<br />

result was reported in each case by only one <strong>of</strong> the 12<br />

laboratories. Discordant results for sample #7 were reported at 3<br />

laboratories, but this may be explained by the fact that all results<br />

for sample #7 clustered close to the 0.40 cut<strong>of</strong>f.<br />

Table 1: Summary <strong>of</strong> discordant results<br />

Laboratory<br />

Sample 1 2 3 4 5 6 7 8 9 10 11 12<br />

1 0.03 0.00 0.02 0.05 0.00 0.08 0.03 0.51 0.01 0.00 0.06 0.02<br />

2 0.05 0.01 0.00 0.01 0.01 0.04 0.04 0.03 0.00 0.72 0.00 0.01<br />

3 0.03 0.01 0.04 0.05 0.00 0.04 0.02 0.01 3.03 0.02 0.02 0.00<br />

4 1.19 0.03 0.04 0.06 0.00 0.04 0.01 0.08 0.03 0.02 0.04 0.04<br />

5 0.07 0.04 0.01 0.00 0.02 0.02 0.04 0.00 0.07 0.96 0.00 0.02<br />

6 0.05 0.03 0.00 0.02 0.02 0.00 0.02 0.03 0.02 0.01 0.04 1.21<br />

7 0.27 0.26 0.26 0.39 0.45 0.27 0.33 0.41 0.40 0.26 0.26 0.25<br />

Discussion & conclusions<br />

The ring test results showed that the PRRS oral fluid IgG ELISA<br />

was highly reproducible across laboratories. These results<br />

support the routine use <strong>of</strong> this test in laboratories providing<br />

diagnostic service to pig producers. Thus, herd monitoring based<br />

on oral fluid sampling could be one part <strong>of</strong> a PRRSV control<br />

and/or elimination program. Further, the successful adaptation <strong>of</strong><br />

one assay to the oral fluid matrix suggests that this approach<br />

could provide the basis for monitoring specific health and welfare<br />

indicators in commercial swine herds using a "pig friendly"<br />

approach.<br />

References<br />

1. Kittawornrat, A et al.: <strong>2012</strong>. Detection <strong>of</strong> PRRSV antibodies in oral fluid<br />

specimens using a commercial PRRSV serum antibody ELISA. J Vet<br />

Diagn Invest 24 (2):262-263<br />

Figure 1: PRRS oral fluid antibody ELISA results from 237<br />

samples tested in 12 laboratories


S1 - O - 18<br />

REAL TIME PCR, MYCOPLASMA GALLISEPTICUM, MYCOPLASMA SYNOVIAE, MYCOPLASMA<br />

MELEAGRIDIS<br />

Pablo Lopez 1 , Phyllls I. Tyrrell 2<br />

1<br />

IDEXX Laboratories, Livestock and Poultry Diagnostics, Westbrook, Maine, USA<br />

2<br />

IDEXX Laboratories, Research and Development, Westbrook, Maine, USA<br />

Introduction<br />

Pathogenic Mycoplasma species are widely prevalent, and<br />

detrimental to poultry health and production. The pathogenic<br />

Mycoplasma species for poultry include M. gallisepticum (Mg), M.<br />

synoviae (Ms) and M. meleagridis (Mm). Infection with these<br />

pathogens can lead to airsaccultis, synovitis, respiratory disease,<br />

decreased growth and decreased egg production (3). Detection<br />

has historically been via Hemagglutination Inhibition (HI), Serum<br />

Plate Agglutination (SPA) and Enzyme Linked Immunosorbent<br />

Assay (ELISA), with culture isolation as the gold standard <strong>of</strong><br />

testing. The use <strong>of</strong> Real-time PCR for the detection <strong>of</strong> pathogenic<br />

mycoplasmas has emerged as a rapid and highly accurate<br />

technique for the recognition or confirmation <strong>of</strong> common<br />

Mycoplasma species in a flock. The reagent sets use<br />

hybridization probes to allow for the creation <strong>of</strong> melting curves<br />

that provide another level <strong>of</strong> diagnostic sensitivity and confidence<br />

in test results.<br />

Materials & methods<br />

All samples were run according to reagent set instructions, using<br />

IDEXX Mg Detection Reagents (lot #1058), Ms Detection<br />

Reagents (lot #1060) or Mm Detection Reagents (lot #1062).<br />

Briefly, reactions consisted <strong>of</strong> 4 μL Roche Genotyping Master<br />

Mix, 9 μL nuclease-free PCR grade water, 2 uL <strong>of</strong> species -<br />

specific detection reagent and 5 μL DNA extraction sample.<br />

Cycling conditions were as follows:<br />

Activation: 95°C, 10 minutes, 1 cycle<br />

Amplification (45 cycles): 95°C, 20 seconds<br />

60°C, 20 seconds, single acquisitions<br />

73°C, 15 seconds<br />

Melting Curve: 95°C, 1 minute<br />

45°C, 1 minute<br />

80°C, 0.14°C/second ramp rate, continuous acquisitions<br />

All samples were tested on a Roche LC 480 LightCycler at either<br />

IDEXX Laboratories or field sites, using 96-well reaction plates<br />

with optical film covers.<br />

Results<br />

M. gallisepticum M. synoviae M. meleagridis<br />

Sample DNA Crossing T m Crossing T m Crossing T m<br />

Point Point Point<br />

M. iowae negative negative negative negative negative negative<br />

M. lip<strong>of</strong>aciens negative negative negative negative negative negative<br />

M. cloacale negative negative negative negative negative negative<br />

M. imitans negative negative negative negative negative negative<br />

M. gallisnarum negative negative negative negative negative negative<br />

M. gallinaceum negative negative negative negative negative negative<br />

M. glycophilum negative negative negative negative negative negative<br />

M. pullorum negative negative negative negative negative negative<br />

M. gallopavonis negative negative negative negative negative negative<br />

10 fg 28.58 63.99 28.92 68.48 28.71 71.48<br />

1 fg 31.70 63.64 32.22 67.92 32.84 71.42<br />

nc negative negative negative negative negative negative<br />

nc negative negative negative negative negative negative<br />

Discussion & conclusions<br />

The use <strong>of</strong> hybridization probe technology allows for the<br />

simultaneous generation <strong>of</strong> crossing point data as well as melting<br />

curve analysis to confirm positive results. While ELISA<br />

technology remains the current standard in screening assays for<br />

avian mycoplasmas, we hope that these standardized reagent<br />

sets will improve testing confidence and be incorporated into<br />

laboratory protocols.


Oral presentations<br />

“Diagnostics<br />

at the point <strong>of</strong> interest”<br />

(2 nd session)


S2 - K - 01<br />

WHEN DO YOU WANT THE RESULT? HOW MUCH DO YOU WANT TO PAY!<br />

Andrew Soldan<br />

Animal Health and Veterinary Laboratories Agency, AHVLA Weybridge, New Haw, Addlestone, Surrey, KT15 3NB, UK<br />

The results <strong>of</strong> some tests are not time critical. For<br />

surveillance testing <strong>of</strong> healthy populations to confirm the absence<br />

<strong>of</strong> disease the cost <strong>of</strong> testing and the ability to handle large<br />

numbers <strong>of</strong> tests is likely to be more important than the speed to<br />

result. In many other circumstances a result the day after<br />

sampling is perfectly adequate. However there are situations<br />

when a same day result or even a next to patient result would be<br />

advantageous. In general these tests can justify a higher unit kit<br />

cost. This paper explores the relationship between cost,<br />

convenience and speed to result <strong>of</strong> tests for infectious disease in<br />

animals. It considers what an ideal rapid patient side test would<br />

look like.<br />

In human medicine very large sums are being invested to<br />

develop point to care testing, especially for molecular agent<br />

detection. However point <strong>of</strong> care testing in humans is not the<br />

equivalent <strong>of</strong> pen-side testing for animals. It is likely that a spin<br />

<strong>of</strong>f from the investment in human diagnostics will be significant<br />

improvements in near patient animal testing. This will be done in<br />

practice labs, labs set up close to a disease outbreak and in<br />

regional rather than central labs. The new tests will be rapid,<br />

accurate, and need little skill or training to run. For veterinary<br />

applications it is likely that platforms with lower instrument costs<br />

will find greater acceptance. An issue that both veterinary<br />

practitioners and regulators will need to address is that owners<br />

will be able to buy and run many <strong>of</strong> these tests themselves.<br />

This paper discusses the major categories <strong>of</strong> rapid tests for<br />

serology and agent detection including consideration <strong>of</strong> some <strong>of</strong><br />

the commercial technologies that are just coming to market.<br />

Rapid tests, point <strong>of</strong> care testing, pen-side testing, cost, speed


S2 - O - 01<br />

DEVELOPMENT OF A RAPID ISOTHERMAL ASSAY TO DETECT TAYLORELLA EQUIGENITALIS, THE<br />

CAUSATIVE AGENT OF CONTAGIOUS EQUINE METRITIS.<br />

S. E. North, P. Das, P.R. Wakeley, J. Sawyer<br />

Technology Transfer Unit, Specialist Scientific Support Department, Animal Health Veterinary Laboratories Agency, Weybridge, United Kingdom.<br />

Introduction<br />

Contagious Equine Metritis (CEM) is an infection <strong>of</strong> the equine<br />

genital tract, caused by the bacterium Taylorella equigenitalis.<br />

Identifying T. equigenitalis from equine genital swabs, and<br />

differentiating it from the closely related Taylorella asinigenitalis,<br />

using a standard bacteriological culture methodology is laborious,<br />

slow and relies on the presence <strong>of</strong> live bacteria. We have<br />

developed a rapid isothermal assay for the detection <strong>of</strong> T.<br />

equigenitalis, the causative agent <strong>of</strong> CEM, using Recombinase<br />

Polymerase Amplification (RPA). RPA is a method <strong>of</strong> amplifying<br />

DNA or RNA. Like the conventional PCR method <strong>of</strong> amplifying<br />

nucleic acid, RPA employs the use <strong>of</strong> two primers and a probe,<br />

however, the reaction proceeds at a single temperature (37C)<br />

and does not cycle through various temperatures as PCR does.<br />

The reactions can be monitored in real time, based on<br />

fluorescence readings, or the products <strong>of</strong> amplification can be<br />

detected at the endpoint <strong>of</strong> the reaction on a lateral flow device<br />

(LFD).<br />

Materials & methods<br />

Equine genital swabs were used to inoculate culture medium<br />

before a crude DNA extraction was performed, enabling<br />

comparisons to be made with conventional culture methods. The<br />

assay was tested, in real time, against 60 equine genital swabs<br />

(30 positive and 30 negative, as determined by culture). To<br />

determine the analytical specificity <strong>of</strong> the assay it was tested<br />

against a panel <strong>of</strong> 38 bacterial species, including closely related<br />

species and equine commensals. The analytical sensitivity <strong>of</strong> the<br />

assay was calculated using a dilution series <strong>of</strong> target DNA which<br />

was tested in triplicate. A second probe was introduced, specific<br />

for T. asinigenitalis, which enabled detection and differentiation <strong>of</strong><br />

both T. equigenitalis and T. asinigenitalis. The T. equigenitalis<br />

assay was adapted, by use <strong>of</strong> a labelled probe, to enable<br />

detection <strong>of</strong> amplification products using a LFD.<br />

Results<br />

The assay to identify T. equigenitalis amplified a 121bp fragment<br />

<strong>of</strong> the 23S rDNA genes. A crude DNA preparation <strong>of</strong> a control<br />

strain <strong>of</strong> T. equigenitalis could be consistently detected in five<br />

minutes and the assay will detect as few as 46 gene copies in 12<br />

minutes. The assay did not cross-hybridise with any <strong>of</strong> the 38<br />

different bacterial organisms tested to determine specificity,<br />

including closely related species and known equine commensal<br />

bacteria. Fifty-seven <strong>of</strong> the 60 DNA extracts from genital swabs<br />

were correctly identified. The introduction <strong>of</strong> a second probe into<br />

the assay enabled the simultaneous detection and differentiation<br />

between T. equigenitalis and T. asinigenitalis. The T.<br />

equigenitalis assay was successfully adapted to enable detection<br />

<strong>of</strong> amplification products using a LFD.<br />

Discussion & conclusions<br />

We have successfully developed a rapid and sensitive isothermal<br />

assay to identify T. equigenitalis. Additionally, the inclusion <strong>of</strong> a<br />

second probe enables the detection <strong>of</strong> T. asinigenitalis. The<br />

amplification products can be detected in real time or via LFD.<br />

This test provides a fast (under 15 minutes) and accurate means<br />

<strong>of</strong> identification. The low energy requirements, lack <strong>of</strong> expensive<br />

equipment, freeze dried reagents and LFD readout makes this<br />

test a suitable candidate for rapid diagnosis and treatment in low<br />

technology laboratories or near to the point <strong>of</strong> interest.<br />

Isothermal, amplification, equine, metritis, RPA


S2 - O - 02<br />

LABORATORY VALIDATION OF AN IMMUNOCHROMATOGRAPHIC TEST FOR THE RAPID<br />

DETECTION OF KOI HERPESVIRUS (CyHV-3) IN GILL SWABS<br />

R. Vrancken 1 , N. Goris 1 , T. Leclipteux 2 , F. Lieffrig 3 , S. Wera 1 , J. Neyts 1 and A. Vanderplasschen 4<br />

1 Okapi Sciences NV, B-3001 Heverlee, Belgium<br />

2 Coris BioConcept, R&D Department, B-5032 Gembloux, Belgium<br />

3 CER Groupe, B-6900 Marloie, Belgium<br />

4 University <strong>of</strong> Liège, Department <strong>of</strong> Infectious and Parasitic Diseases (B43b), B-4000 Liège, Belgium<br />

Immunochromatographic test, Koi herpesvirus, Cyprinid herpesvirus-3<br />

Introduction<br />

Carp are one <strong>of</strong> the most economically valuable fresh-water fish<br />

in aquaculture. Although the majority <strong>of</strong> this species is cultivated<br />

for human consumption (Cypinus carpio carpio), the more<br />

colourful variant <strong>of</strong> this species, the koi carp (Cyprinus carpio<br />

koi), is a popular ornamental fish and may be highly valuable.<br />

In the late 1990’s, both common carp and koi carp were struck<br />

with mass mortality, caused by Cyprinid herpesvirus-3 (CyHV-3)<br />

(1), commonly known as Koi herpesvirus (KHV). Koi herpesvirus<br />

disease (KHVD) is generally seasonal and may develop rapidly at<br />

water temperatures between 23 and 25°C inducing mortality up<br />

to 100% (2). Although clinical signs can be obvious, they are not<br />

pathognomonic, therefore diagnosis <strong>of</strong> CyHV-3 relies exclusively<br />

on laboratory techniques like (q)PCR, virus isolation,… Since<br />

early detection <strong>of</strong> KHVD is crucial for disease control, we<br />

developed an immunochromatographic test for the specific and<br />

rapid (pond-side) detection <strong>of</strong> CyHV-3 antigen.<br />

In this study we evaluated the analytical and diagnostic specificity<br />

and sensitivity under laboratory conditions.<br />

Materials & methods<br />

Rapid test. Briefly, samples were collected by taking a swab from<br />

the gills <strong>of</strong> the animal, which was eluted in 200µl <strong>of</strong> elution buffer.<br />

One hundred µl <strong>of</strong> eluate was subsequently applied to the test<br />

well <strong>of</strong> the test cassette and results were read after 15 minutes.<br />

Analytical specificity. Possible cross-reaction with other<br />

pathogens affecting carp was evaluated on diseased fish for:<br />

Ichthyobodo sp., Dactylogyrus sp. and Gyrodactylus sp.,<br />

Ichthyophthirius sp., Trichodina sp., Aeromonas sp., Saprolegnia<br />

sp., Branchiomyces sp., For Carp Pox virus (CyHV-1) and Spring<br />

Viremia <strong>of</strong> Carp virus (SVCV) in vitro cultured virus was used.<br />

Analytical sensitivity. Serial dilutions <strong>of</strong> in vitro cultured CyHV-3<br />

from different geographical regions were used. These included a<br />

German, an Israeli and two Belgian CyHV-3 field isolates.<br />

Diagnostic specificity. Animals (n=60) known to be free <strong>of</strong> CyHV-<br />

3 were tested using the rapid test as described.<br />

Diagnostic sensitivity. Two identical animal experiments were set<br />

up using either the Belgian field strain M1 or M3. Hereto, 120<br />

common carp (Cyprinus carpio carpio) were distributed over 4<br />

tanks. Three tanks containing 30 animals were infected by bath<br />

infection for 1 h at 40 pfu/ml <strong>of</strong> CyHV-3. One tank <strong>of</strong> 30 animals<br />

served as a negative control. Each day, 2 animals per tank were<br />

sacrificed and tested using the rapid test. All animals found dead<br />

were analysed as well. DNA <strong>of</strong> gill tissue was extracted and the<br />

presence <strong>of</strong> CyHV-3 was evaluated by PCR (Bercovier TK<br />

primers) (2).<br />

Results<br />

Analytical specificity. No cross-reactivity was observed. Hence,<br />

the analytical specificity was 100%.<br />

Analytical sensitivity. All field strains were detected with the rapid<br />

test with a limit <strong>of</strong> detection from 1,25 x 10² to 2,4 x 10 4 pfu/ml.<br />

Diagnostic specificity. No false positive results were observed.<br />

Hence, the diagnostic specificity was 100%.<br />

Diagnostic sensitivity.<br />

Experiment 1: CyHV-3 M1<br />

The rapid test detected the viral antigen from day 5 post infection<br />

(p.i.) at a detection rate <strong>of</strong> 33.3% increasing to 100% on day 9 p.i.<br />

The detection rate subsequently decreased until the end <strong>of</strong> the<br />

observation period (16 days p.i.) (see Figure 1). All animals found<br />

dead due to KHVD tested positive on the rapid test. Mortality<br />

started at 7 days p.i. and a relative diagnostic sensitivity <strong>of</strong> 52.6%<br />

was estimated when using PCR as golden standard.<br />

Figure 1. Detection rate in fish infected with CyHV-3-M1.<br />

Experiment 2: CyHV-3 M3<br />

The rapid test detected the viral antigen from day 4 p.i. at a<br />

detection rate <strong>of</strong> 33.3%, rapidly increasing to 100% on day 7 p.i.<br />

(see Figure 2). All animals found dead due to KHVD tested<br />

positive on the rapid test. Mortality was observed from 5 days p.i.<br />

and a relative diagnostic sensitivity <strong>of</strong> 71.7% was estimated when<br />

using PCR as golden standard.<br />

Figure 2. Detection rate in fish infected with CyHV-3 M3.<br />

Discussion & conclusions<br />

We here present the first rapid immunochromatographic test for<br />

the specific detection <strong>of</strong> CyHV-3-antigen. This non-invasive test<br />

was able to detect the CyHV-3 antigen in a population (aquarium<br />

or pond) on day 4 or 5 post infection and confirmed the presence<br />

<strong>of</strong> CyHV-3 in fish that succumbed to the disease (100% detection<br />

rate). Although the diagnostic sensitivity <strong>of</strong> this technique is<br />

inferior to PCR, this method does have some major advantages<br />

over PCR and other laboratory techniques.<br />

The rapid detection <strong>of</strong> CyHV-3 is crucial in preventing the further<br />

spread <strong>of</strong> the disease. With this pond-side test, results are<br />

available within 15 minutes and carp breeders/koi owners would<br />

have the opportunity to take the necessary control measures.<br />

This test does not require trained personnel or expensive<br />

equipment and samples do not have to be shipped to<br />

laboratories, thereby saving time. Furthermore, a suspicion <strong>of</strong><br />

KHVD is generally raised in case <strong>of</strong> mortality and given the 100%<br />

sensitivity in fish succumbed to KHVD, the detection <strong>of</strong> an<br />

emerging infection in the field is highly likely, which makes this<br />

test fit for purpose.<br />

In conclusion, we developed the first pond-side test based on<br />

immunochromatography for the specific detection <strong>of</strong> CyHV-3 as a<br />

first line diagnostic tool in the field.<br />

Acknowledgments<br />

This study was financed by a project <strong>of</strong> the Walloon Region (APE<br />

grant MA-13387-00).<br />

References<br />

1. Michel, B., Fournier, G., Lieffrig, F., Costes, B., Vanderplasschen, A.<br />

(2010). Emerging infectious diseases, 16, 1835-43.<br />

2. Manual <strong>of</strong> Diagnostic Tests for Aquatic Animals (OIE, 2009)


S2 - O - 03<br />

EVALUATION OF RAPID HRSV STRIP TESTS FOR DETECTION OF BOVINE RESPIRATORY<br />

SYNCYTIAL VIRUS<br />

Wojciech Socha, Jerzy Rola<br />

National Veterinary Research Institute, Department <strong>of</strong> Virology, Pulawy, Poland<br />

Introduction<br />

Bovine respiratory syncytial virus (BRSV) is one <strong>of</strong> the major viral<br />

pathogens responsible for respiratory tract diseases in cattle<br />

worldwide. Currently most widely used diagnostic methods for<br />

direct detection <strong>of</strong> BRSV are virus isolation test, antigen ELISA<br />

and RT-PCR (1). Each <strong>of</strong> them is relatively complicated and<br />

requires properly equipped laboratory and trained personnel.<br />

Therefore there is a need for a simple and rapid diagnostic test<br />

for detection <strong>of</strong> BRSV in the field conditions. Rapid<br />

immunochromatographic strip tests detecting viral antigens have<br />

been developed for closely related to BRSV, Human Respiratory<br />

Syncytial Virus (HRSV) (2). Due to the high antigenic similarities<br />

between both viruses it could be theoretically possible to adapt<br />

this tests for the diagnosis <strong>of</strong> BRSV. The aim <strong>of</strong> this study was to<br />

evaluate this possibility on animals inoculated with BRSV vaccine<br />

strain.<br />

Materials & methods<br />

Five clinically healthy and serologically negative for BRSV calves<br />

approximately 6-8 weeks old were selected for experiment. Three<br />

calves were inoculated intranasally with 2 ml <strong>of</strong> live vaccine<br />

Rispoval RS-PI3 (Pfizer). Control calves received 2 ml <strong>of</strong> sterile<br />

water. Both groups <strong>of</strong> calves were housed separately in isolation<br />

to prevent the spread <strong>of</strong> the vaccine virus. Nasal swabs were<br />

taken both from the inoculated and control calves from day -1 to<br />

28 post inoculation (dpi) and placed in liquid transport medium. In<br />

the laboratory swabs were shaken, centrifuged and the<br />

supernatant was stored at -80 o C until examination.<br />

Three different rapid HRSV strip tests: RSV Respi-strip (Coris),<br />

TRU RSV (Meridian Bioscience) and BinaxNOW RSV (Inverness<br />

Medical) were evaluated in the study. All the tests were<br />

performed according to manufacturer’s instructions.<br />

The detection limit <strong>of</strong> all tests was determined by analysing<br />

a 2-fold dilution series <strong>of</strong> BT cell cultures infected with BRSV<br />

strain 375, with each <strong>of</strong> the strip tests and RT-PCR. Specificity <strong>of</strong><br />

the tests was investigated using two positive reference strains<br />

(BRSV 375, BRSV A51908) and two strains <strong>of</strong> other<br />

paramyxoviruses as a negative control (HRSV A2, BPIV3 SB).<br />

Another negative control consisting <strong>of</strong> transport medium from<br />

UTM-RT Copan system was included.<br />

Total RNA was extracted from the supernatant <strong>of</strong> nasal swabs<br />

using TRI reagent (Sigma) according to the producer’s<br />

instructions. RT-PCR was performed using Titan One-Tube RT-<br />

PCR System (Roche) with primers B7 and B8 specific to gene<br />

encoding glycoprotein G (4). The RT-PCR assay was used as the<br />

gold standard for this study and samples positive by this method<br />

were considered true positives.<br />

Results<br />

The RT-PCR with primers for glycoprotein G gene was specific<br />

for BRSV. None <strong>of</strong> the negative control strains was detected. All<br />

rapid strip tests reacted positively both with HRSV strain A2 and<br />

BRSV reference strains and negatively with BPIV-3. The<br />

detection limit for RT-PCR was 39.1 TCID 50 whereas for each <strong>of</strong> the<br />

rapid tests it was around 156 TCID 50 <strong>of</strong> the virus .<br />

All three calves vaccinated with the live vaccine Rispoval RS-PI3<br />

were PCR positive on nasal swabs. In total the vaccine virus was<br />

found in 12 swabs out <strong>of</strong> 30 swabs collected from calves<br />

experimentally vaccinated. Two control calves remained negative<br />

in PCR throughout the study. Using RSV Respi-strip test virus was<br />

detected in 7 swabs, four nasal swabs were positive in TRU RSV<br />

test and 9 in BinaxNOW RSV. All <strong>of</strong> tests showed negative results for<br />

control calves. Diagnostic sensitivity, specificity, positive predictive<br />

value (PPV) and negative predictive value (NPV) were calculated<br />

for each <strong>of</strong> the tests (Table 1)<br />

Table 1. Performance <strong>of</strong> rapid strip tests for BRSV detection.<br />

Test<br />

RSV Respistrip<br />

Diagnostic<br />

sensitivity Specificity PPV NPV<br />

33% 87% 57% 71%<br />

TRU RSV Test 33% 100% 100% 74%<br />

BinaxNOW<br />

RSV<br />

50% 87% 67% 77%<br />

Discussion & conclusions<br />

Results <strong>of</strong> our studies confirmed that rapid<br />

immunochromatographic HRSV tests are able to detected BRSV.<br />

They also showed high specificity reacting only with HRSV and<br />

BRSV strains. Positive reaction with HRSV, should not be a<br />

problem because it is known that this virus has highly restricted<br />

host range and does not have the ability to infect cattle (3).<br />

All <strong>of</strong> the strip tests used in this study were previously evaluated<br />

in HRSV diagnostics, showing high specificity (97- 98%) with<br />

sensitivity ranging from 58 to 91%. In our study we made similar<br />

calculations for the same three strip tests for their performance in<br />

BRSV detection. Compared with RT-PCR, specificity <strong>of</strong> strip tests<br />

was relatively high (87-100%) however their diagnostic sensitivity<br />

was low (33-50%). This showed that although strip tests reacted<br />

with similarly high specificity with both BRSV and HRSV, their<br />

sensitivity was clearly affected when used in detection <strong>of</strong> bovine<br />

pathogen. This could be explained by existing differences in<br />

amino acid sequence <strong>of</strong> F and N proteins between BRSV and<br />

HRSV, which can be reason for overall weaker reaction <strong>of</strong> the<br />

assays.<br />

It is also possible that lower diagnostic sensitivity detected in our<br />

study stemed from different type <strong>of</strong> samples used for testing.<br />

Nasopharygneal washes are described as a recommended<br />

specimen for immunchromatographic tests and previous<br />

evaluations were done basing on this type <strong>of</strong> samples. However,<br />

due to the fact that collection <strong>of</strong> nasal washes from animals could<br />

be complicated, we have decided to use nasal swabs as they are<br />

much easier to get in the field conditions.<br />

Basing on the evaluation <strong>of</strong> sensitivity, specificity, PPV and NPV<br />

<strong>of</strong> the strip tests, it can be concluded that most reliable was TRU<br />

RSV. Although its sensitivity was lower compared to BinaxNOW<br />

RSV it was characterized by very high specificity as well as PPV<br />

and NPV.That meant that samples positive by TRU RSV were<br />

true positives. TRU RSV could therefore be used as a supportive<br />

rapid screen test for BRSV detection at the farm. However, due<br />

to the limited sensitivity, negative results would have to be<br />

confirmed by more sensitive tests like RT-PCR.<br />

References<br />

1.Larsen, L.E., 2000, Bovine respiratory syncytial virus (BRSV):<br />

a review. Acta Ve. Scand 41: 1-24.<br />

2.Selvarangan, R., Abe, D., Hamilton, M., 2008. Comparison <strong>of</strong> BD<br />

Directigen EZ RSV and Binax NOW RSV tests for rapid detection <strong>of</strong><br />

respiratory syncytial virus from nasopharyngeal aspirates in a pediatric<br />

population. Diagn Microbiol Infect Dis 62: 157-161.<br />

3.Valarcher, J.F., Taylor, G., 2007. Bovine respiratory syncytial virus<br />

infection. Ve. Res 38: 153-180.<br />

4.Vilcek, S., Elvander, M., Ballagi-Pordány, A., Belák S., 1994.<br />

Development <strong>of</strong> nested PCR assays for detection <strong>of</strong> bovine respiratory<br />

syncytial virus in clinical samples. J Clin Microbiol 32: 2225-2231.


S2 - O - 04<br />

EVALUATION OF A LATEX AGGLUTINATION TEST FOR THE IDENTIFICATION OF CLOSTRIDIUM<br />

DIFFICILE OF PORCINE ORIGIN<br />

Introduction<br />

Clostridium difficile-associated disease (CDAD) in piglets less<br />

than 1 week old is considered emerging and <strong>of</strong> great importance<br />

for its direct effects on piglet performance. Although no direct<br />

evidence <strong>of</strong> animal-to-human transmission, isolation and<br />

accurate identification <strong>of</strong> CD strains are crucial for both<br />

diagnostics and epidemiology <strong>of</strong> CDAD, due to the significant<br />

overlap between strains implicated in CD infection in humans and<br />

animals. A definitive diagnostic <strong>of</strong> CD infection is usually<br />

achieved by combining toxin testing <strong>of</strong> stool specimens, along<br />

with isolation and further toxin production test by CD isolates.<br />

Jaime Maldonado 1 , Laura Valls 1<br />

1<br />

Hipra, Diagnostic Center, Girona, Spain<br />

Clostridium difficile, latex, agglutination<br />

In addition to the colony morphology and growth features in<br />

selective culture media, definitive identification <strong>of</strong> CD in pure<br />

cultures is <strong>of</strong>ten accomplished by biochemical pr<strong>of</strong>iling, and by<br />

PCR amplification <strong>of</strong> CD-specific or toxin genes. Commercial<br />

latex agglutination test (LAT) for presumptive identification <strong>of</strong> CD<br />

in stools, broth cultures and solid media are available as an<br />

alternative to costly and labour-intensive assay. They are rapid<br />

and simple assays for the early identification <strong>of</strong> CD. In this study<br />

we aimed to evaluate the usefulness <strong>of</strong> a LAT kit for the<br />

presumptive identification <strong>of</strong> CD strains <strong>of</strong> porcine origin.<br />

Materials & methods<br />

The C. difficile Test Kit (Oxoid, Hampshire, UK) was evaluated<br />

using three groups <strong>of</strong> bacteria: 1) The test was validated with five<br />

well identified CD strains <strong>of</strong> porcine origin from previous studies<br />

(1), with known toxin and ribotype pr<strong>of</strong>iles; 2) Further evaluation<br />

was carried out with 41 CD field isolates coming from suckling<br />

piglets suffering from diarrhoea in the 1 st week <strong>of</strong> age. They came<br />

from the same number <strong>of</strong> epidemiologically unrelated pig<br />

breeding units, and were collected in the period 2008-2011.<br />

Bacterial culture and identification were performed as previously<br />

described (1). Definitive identification to species level was carried<br />

out using the API Rapid ID 32 A kit (BioMerieux, Marcy l'Etoile,<br />

France); 3) Cross-reactivity in the LAT kit was determined using<br />

reference and collection strains <strong>of</strong> swine Escherichia coli (n=4),<br />

Clostridium perfringens types A, B, C, D and E (n=5), Salmonella<br />

typhimurium (n=1), Proteus mirabilis (n=1) and Clostridium novyi<br />

(n=1). The LAT kit was performed as per manufacturer’s<br />

instructions, with all three groups <strong>of</strong> bacteria involved in the<br />

study. Agglutination reactions were scored as negative (0), mild<br />

(1+), moderate (2+), or strong (3+).<br />

Results<br />

When LAT was carried out with well characterized CD antigens <strong>of</strong><br />

porcine origin, it correctly identified all five bacteria with a strong<br />

(3+) agglutination reaction. Similarly, all 41 field CD strains<br />

identified by the API system with excellent (24,4%) or good<br />

(75.6%) identification scores, also tested positive with 1+<br />

(14.6%), 2+ (29.3%), or 3+ (56.1%) agglutination reactions. No<br />

correlation between the two scores was observed.<br />

Nevertheless, with respect to cross-reactivity, two strains <strong>of</strong> C.<br />

perfringens (Types A [3+] and E [1+]), one strain <strong>of</strong> E. coli (3+),<br />

and the swine pathogen C. novyi (2+) tested positive in the LAT,<br />

and hence were incorrectly identified as being CD.<br />

Figure 1: Latex agglutination test: 1. Swine C. difficile strong<br />

reaction (3+); 2-4. Cross-reactivity <strong>of</strong> swine C. novyi (2+), C.<br />

perfringens type A (3+) and E. coli (3+), respectively; 5.<br />

Negative control <strong>of</strong> the test. 6. Positive control <strong>of</strong> the test.<br />

Discussion & conclusion<br />

Taking together, the results obtained in this study indicates that<br />

the LAT kit has a good ability to identify CD, since none <strong>of</strong> the<br />

collection or field CD strains tested negative. However, the fact<br />

that three bacterial species that are part <strong>of</strong> the pig intestinal flora<br />

have tested positive, indicates that positive results using this kit<br />

should be interpreted with caution. In fact, the kit manufacturer<br />

warns <strong>of</strong> the need to confirm all positive results, which is<br />

evidenced in this study.<br />

Considering that a wide variety <strong>of</strong> CD strains were tested by LAT<br />

in the present study, it is reasonable to assume that negative<br />

agglutination test results obtained with the evaluated kit, under<br />

the described conditions (pure cultures on solid media), may<br />

correspond to true negative. Consequently, the LAT kit described<br />

could eventually be used to test porcine bacterial cultures<br />

suspicious <strong>of</strong> being CD; positive results must be confirmed by a<br />

more specific assay, and negative results should be coupled with<br />

growth features and case history.<br />

Although CDAD is considered a major threat for the swine<br />

industry, and despite the obvious similarities between the CD<br />

strains affecting humans and domestic animals, existing<br />

diagnostic tools have not been evaluated in depth using clinical<br />

samples <strong>of</strong> porcine origin. It would be necessary to do so, with<br />

those test intended for toxin detection in stools and molecular<br />

detection <strong>of</strong> toxin genes, in order to improve the current<br />

diagnostic approach to CD-related neonatal diarrhoea in swine.<br />

Acknowledgements<br />

We thank Carmen Sánchez and Lidia López who collected and<br />

identified bacterial strains during the study.<br />

References<br />

1. Alvarez-Perez S, Blanco JL, Bouza E, Alba P, Gibert X, Maldonado J,<br />

Garcia. (2009). Prevalence <strong>of</strong> Clostridium difficile in diarrhoeic and nondiarrhoeic<br />

piglets. Vet. Microbiol, 137, 302-5.


Oral presentations<br />

“Emerging, re-emerging<br />

and wildlife diseases –<br />

diagnostic possibilities”<br />

(3 rd session)


S3 - K - 01<br />

EMERGING AND RE-EMERGING WILDLIFE DISEASES: PATHOLOGY AND RELATED TECHNIQUES<br />

FOR DIAGNOSTICS AND FOR GENERAL AND TARGETED SURVEILLANCE<br />

Dolores-Gavier Widén<br />

Department <strong>of</strong> Pathology and Wildlife Diseases, Deputy Head <strong>of</strong> Department, National Veterinary Institute (SVA), SE‐75189 Uppsala, Sweden<br />

Wildlife surveillance, emerging diseases, pathology<br />

The importance <strong>of</strong> wildlife and wildlife diseases<br />

Wild animals have high economic, social, ecological and cultural<br />

value and play fundamental roles in the maintenance <strong>of</strong> stable<br />

ecosystems. Wildlife contributes significantly to recreation and<br />

tourism. Hunting is an important socio-economic activity. Within<br />

the EU hunting is estimated to be worth €16 billion 1 and there are<br />

more than 7 million hunters in Europe. 2<br />

Wildlife trade is a fast growing market and contributes to the<br />

worldwide spreading <strong>of</strong> new pathogens and emerging diseases,<br />

for example giant liver fluke (Fascioloides magna) was introduced<br />

into Europe in elk from North America. Natural migration <strong>of</strong> wild<br />

animals has similar consequences, as exemplified by the global<br />

spread <strong>of</strong> highly pathogenic H5N1 avian influenza. Wildlife<br />

reservoirs maintain infections which may spillover to domestic<br />

animals, making the control and eradication difficult, for example<br />

tuberculosis and classical swine fever. Wild animals host<br />

numerous zoonotic infections. Humans can acquire infections<br />

through direct contact with wild animals, for instance rabies and<br />

tularemia, through food consumption, as trichinosis and hepatitis<br />

E, or from the environment contaminated by wildlife, such as<br />

hantavirus infections or alveolar echinococcosis.<br />

Mass mortality in wild animals results in public concern, as seen<br />

during botulism outbreaks in waterfowl, trichomoniasis epidemics<br />

in wild finches and morbillivirus infections in seals. Furthermore,<br />

diseases may have a devastating impact on wildlife populations<br />

and threaten their conservation, like White Nose Syndrome<br />

(Geomyces destructans infection) in bats.<br />

Therefore, the value <strong>of</strong> maintaining diverse and healthy wildlife<br />

populations cannot be overestimated. The importance <strong>of</strong> wildlife<br />

surveillance and the interest on diagnosing infections in wild<br />

animals has increased rapidly in Europe and globally. This is<br />

clearly supported by the Director General <strong>of</strong> the OIE, who stated<br />

that “Surveillance <strong>of</strong> wildlife diseases must be considered equally<br />

as important as surveillance and control <strong>of</strong> diseases in domestic<br />

animals”. 3<br />

Emerging and re-emerging infectious diseases<br />

Emerging infectious diseases (EID) include broad categories <strong>of</strong><br />

infections characterized by changes in their epidemiological<br />

presentation. EID are diseases whose incidence has increased in<br />

a defined time period and location or threatens to increase. If the<br />

infection was unknown in the place before, the disease is<br />

regarded as emerging, but if it had been present at the location in<br />

the past and was considered controlled or eradicated, the<br />

infection is considered to be re-emerging. The following situations<br />

are all examples and mechanisms <strong>of</strong> EID: new pathogens or<br />

newly evolved strains <strong>of</strong> existing pathogens, spread to new<br />

geographic area (for example through wildlife migration or<br />

translocation), spread to new populations, increase in<br />

prevalence, detection <strong>of</strong> previously unrecognized pathogens and<br />

old infections reemerging as a result <strong>of</strong> antimicrobial resistance.<br />

Emerging infectious diseases (EID) in humans are dominated by<br />

zoonoses, and constitute 60% <strong>of</strong> EID events. 4<br />

A high rate <strong>of</strong><br />

these zoonoses (72%) are caused by pathogens with a wildlife<br />

origin, and this rate has increased significantly with time<br />

(controlling for reporting effort). 4<br />

As a whole, zoonoses from<br />

wildlife are considered the most significant, growing threat to<br />

global health <strong>of</strong> all EID. Vector-borne diseases are frequent<br />

among EID events (22.8% <strong>of</strong> events) and these infections have<br />

also increased over time. 4 Climate change is probably implicated,<br />

however, it is difficult to demonstrate causal relationships<br />

between EID events and climate change. Emerging pathogens<br />

are more likely to be zoonotic or vector-borne with a broad host<br />

range. 4 Drivers such as human population density, antibiotic drug<br />

use and agricultural practices are major determinants <strong>of</strong> the<br />

distribution <strong>of</strong> EID events. 4<br />

Good knowledge and rapid identification <strong>of</strong> emerging infectious<br />

diseases occurring in wildlife are essential for the design and<br />

implementation <strong>of</strong> preventing, control and mitigating measures to<br />

protect the health <strong>of</strong> humans and animals.<br />

Wildlife health surveillance<br />

Surveillance, is defined by the OIE) as “the systematic ongoing<br />

collection, collation, and analysis <strong>of</strong> information related to animal<br />

health and the timely dissemination <strong>of</strong> information to those who<br />

need to know so that action can be taken”. 5<br />

General wildlife<br />

disease surveillance, also named passive surveillance, identifies<br />

ill or dead wild animals and conducts diagnostic investigations to<br />

determine the cause <strong>of</strong> disease or death. The investigations<br />

include <strong>of</strong> a broad range <strong>of</strong> wildlife hosts and pathogens or<br />

diseases. Targeted surveillance, also named specific or active<br />

surveillance, identifies a specific infection or disease,<br />

serologically or demonstrating the pathogen. Surveillance<br />

informs about occurrence <strong>of</strong> pathogens, wildlife hosts and<br />

locations <strong>of</strong> endemic infections and can also result in the<br />

detection <strong>of</strong> new pathogens or diseases. Long term surveillance<br />

detects changes in disease patterns, which contribute to<br />

epidemiological analyses and risk assessment. All together,<br />

surveillance is essential in the control and eradication <strong>of</strong><br />

infectious diseases.<br />

There is a great variation in the level <strong>of</strong> wildlife surveillance<br />

conducted in different countries in Europe. As a whole, more<br />

than 18,000 wild animals are investigated by general surveillance<br />

and more than 50,000 by targeted surveillance annually in<br />

Europe. 6<br />

The choice <strong>of</strong> diagnostic tests to be used for surveillance <strong>of</strong><br />

wildlife diseases needs to take into consideration several factors,<br />

for example the purpose <strong>of</strong> the testing (such as certifying<br />

freedom from infection before translocation, estimating<br />

prevalence, or others), the practicalities and applicability to field<br />

investigations, the cost <strong>of</strong> the tests and the known performance<br />

<strong>of</strong> the method in the host species to be tested.<br />

Test validation<br />

The performance <strong>of</strong> tests designed and validated for use in<br />

domestic animals is most <strong>of</strong>ten not known when applied to<br />

wildlife species. Diagnostic testing <strong>of</strong> wild animals is challenging<br />

in several ways, for example the samples may be <strong>of</strong> poor quality<br />

or difficult to obtain. Likewise, validation <strong>of</strong> assays for wildlife also<br />

presents difficulties. In 2011, the OIE established an ad hoc<br />

group which drafted guidelines on “Principles and methods for<br />

the validation <strong>of</strong> diagnostic tests for infectious diseases<br />

applicable to wildlife”, 7 which take into consideration the intrinsic<br />

difficulties <strong>of</strong> validating tests for wildlife. In the guidelines,<br />

pathways and stages are proposed for validation <strong>of</strong> tests with two<br />

different starting points, when a previously validated test in<br />

domestic animals exists and when it does not. It is proposed that<br />

when full validation is not possible, the best alternative may be to<br />

evaluate the fitness <strong>of</strong> the test in a small number <strong>of</strong> reference<br />

samples. These preliminary estimates may give sufficient<br />

information to obtain provisional acceptance <strong>of</strong> the test by the<br />

authorities; this is a new step /category within stage 2 <strong>of</strong> the<br />

validation process.<br />

Pathology<br />

Pathology is a primary component in general wildlife disease<br />

surveillance. Rudolph Virchow (1821-1902), a founding father <strong>of</strong><br />

pathology, made seminal advancements in the understanding <strong>of</strong><br />

infectious diseases based on observations at abbatoirs and <strong>of</strong><br />

tissue changes at the cellular level; he was active in the<br />

implementation <strong>of</strong> meat inspection. Today, despite the technical


advances, the fundamental tools <strong>of</strong> pathology are still post<br />

mortem examination and histopathology, and these are the<br />

starting point for many diagnostic investigations. Additionally,<br />

pathology is essential in the early and rapid identification <strong>of</strong><br />

emerging infections and/or changes in disease patterns.<br />

Pathology techniques<br />

Histopathology is conducted as a standard on haematoxylin-eosin<br />

stained sections. Specific stains are used to identify structures<br />

and substances and to help visualize pathogens, for example<br />

Congo red for amyloid and Ziehl-Neelsen for acid fast bacteria.<br />

Enzyme histochemistry can be used to test enzyme activities in<br />

muscle diseases and in other conditions. Cytology, the study <strong>of</strong><br />

cells spread on slides, is applied on clinical samples, such as<br />

liquid punctures, or smears <strong>of</strong> organs. Electron microscopy is<br />

used to characterize structural changes in cells and tissues and to<br />

support the morphological changes observed by microscopy.<br />

Immunohistochemistry is used to reveal specific antigens or<br />

proteins in tissue sections applying labelled antibodies against the<br />

particular protein and visualizing the antigen-antibody reaction by<br />

a marker such as fluorescent dye or enzyme. IHC is a very useful<br />

technique used to classify tumors and detect bacteria, viruses and<br />

parasites. Importantly, IHC allows associating the presence <strong>of</strong><br />

pathogens with lesions. IHC can be automated and is widely<br />

applied to diagnosis. Double-labelling techniques are used to<br />

identify two or more different proteins, giving information on for<br />

example cell type and presence <strong>of</strong> a particular virus in the cell.<br />

Immuno-electron microscopy uses immunolabelling <strong>of</strong> ultrathin<br />

sections, and is visualized with markers as colloidal gold particles.<br />

Quantifying pathology gives detailed information on lesions<br />

numbers and size. This results in accurate descriptions, for<br />

example <strong>of</strong> degree <strong>of</strong> advancement and dissemination <strong>of</strong> lesions,<br />

and allows comparison <strong>of</strong> lesion severity between animals, for<br />

instance, in assessment <strong>of</strong> disease protection by vaccine<br />

candidates. Morphometry, the macroscopic or microscopic<br />

measurement <strong>of</strong> lesions, or parts <strong>of</strong> lesions (for example necrosis)<br />

gives quantitative data. Computerized image analysis allows<br />

collection <strong>of</strong> large amounts <strong>of</strong> data in a short time. Scoring <strong>of</strong><br />

lesions is a semi-quantitative assessment <strong>of</strong> lesions size and/or<br />

number, the scoring has to be standardized for each application.<br />

Imaging techniques developed for clinical application in humans<br />

allow tridimensional measurements. They can also be used in<br />

veterinary pathology for measuring lesions in dead animals,<br />

mostly in experimental settings. Some examples are magnetic<br />

resonance imaging (MRI) and multidetector computed<br />

tomography (MDCT), used for example to measure the<br />

magnitude <strong>of</strong> lesions in pulmonary tuberculosis. X-rays are used<br />

frequently in wildlife pathology for forensic investigations to detect<br />

bullet fragments in carcasses.<br />

Molecular diagnostics are increasingly used to identify pathogens<br />

in combination with the diagnostic histopathology. In situ<br />

hybridization is based on the hybridization <strong>of</strong> a labelled probe (a<br />

small specific DNA or RNA sequence); with a complementary<br />

sequence <strong>of</strong> for example virus, on a tissue section, the reaction<br />

can be visualized by chromogens or fluorescein (FISH). PCR and<br />

the numerous rapidly developing PCR-based techniques are<br />

widely used to establish an etiological diagnosis. PCR is usually<br />

performed in unfixed (fresh or frozen) samples, but in many cases<br />

can be applied to formalin fixed paraffin embedded tissue. The<br />

target lesion, for example a granuloma, can be micro-dissected<br />

and tested individually. Microarray technology detects large<br />

numbers <strong>of</strong> genomes simultaneously allowing identification <strong>of</strong><br />

multiple infectious agents at the same time in one sample. The<br />

EU project WildTech, 8 is developing tests for screening wildlife<br />

populations for infections, including nucleic acid microarrays and<br />

ELISA technology adapted to microarray format for serology.<br />

Obtaining the correct sample <strong>of</strong> target tissues and lesions for<br />

laboratory testing is <strong>of</strong> utmost importance, in particular when<br />

small amounts <strong>of</strong> tissues are tested. A very sensitive method<br />

applied to “the wrong sample” can have a small chance <strong>of</strong><br />

detecting the pathogen.<br />

The importance <strong>of</strong> networking<br />

Wildlife disease surveillance is a very extensive field involving<br />

different diagnostic disciplines and a broad range <strong>of</strong> wildlife hosts<br />

and pathogens. It also includes epidemiology and communication<br />

activities. Networking among disciplines and across national and<br />

organization boundaries is <strong>of</strong> great benefit in surveillance<br />

programs. Organizations such as the European Association <strong>of</strong><br />

Veterinary Laboratory Diagnosticians (EAVLD) 9 and the Wildlife<br />

Disease Association (WDA) 10 , with its European Section (EWDA)<br />

are excellent forums.<br />

Conclusions<br />

In conclusion, pathology plays a key role in wildlife surveillance<br />

and in discovering new diseases and is the starting point <strong>of</strong><br />

diagnostic investigations. Importantly, pathology informs about<br />

the significance, in terms <strong>of</strong> disease, <strong>of</strong> the presence <strong>of</strong> infectious<br />

agents or particular strains <strong>of</strong> agents demonstrated by laboratory<br />

tests. Classical descriptive pathology is today more refined by<br />

new techniques but cannot be replaced by other disciplines and<br />

will continue to be a cornerstone in the detection and<br />

characterization <strong>of</strong> emerging diseases. Wild animals play an<br />

increasingly important role in emerging infectious diseases, in<br />

particular zoonoses. This trend will possibly continue in the<br />

future.<br />

Acknowledgement<br />

WildTech, a project <strong>of</strong> the European Commission under the Food,<br />

Agriculture and Fisheries, and Biotechnology Theme <strong>of</strong> the 7th<br />

Framework Programme for Research and Technological<br />

Development, grant agreement no. 222633.<br />

References<br />

1. Kenward, R. & Sharp, R. (2008) Use Nationally <strong>of</strong> Wildlife Resources<br />

Across Europe, 117-132.: in Manos, P. & Papathanasiou, J. [eds.] (2008)<br />

GEM-CON-BIO: Governance & Ecosystems Management for the<br />

Conservation <strong>of</strong> Biodiversity. Thessaloniki<br />

2. Federation <strong>of</strong> Associations for Hunting and Conservation <strong>of</strong> the EU<br />

(FACE), at: www.face.eu/aboutus-en.htm<br />

3. Vallat B. 2008, OIE editorial. Improving wildlife surveillance for its<br />

protection while protecting us from the diseases it transmits. At :<br />

www.oie.int/en/for-the -media/editorials/detail/article/improving -wildlifesurveillance-for-its-protection-while-protecting-us-from-the-diseases-ittransmit/<br />

4. Global trends in emerging infectious diseases. Kate E. Jones, Nikkita<br />

G. Patel, Marc A. Levy, Adam Storeygard, Deborah Balk, John L.<br />

Gittleman, & Peter Daszak. Nature. Vol 451| 21 February 2008|<br />

doi:10.1038/nature06536<br />

5. World Organization for Animal Health (OIE) (2009) Terrestrial Animal<br />

Health Code , 18 th Ed. Glossary. OIE. Paris. Available at:<br />

www.oie.int/index.php?id=169&L=0&htmfile=glossaire.htm#terme_surveill<br />

ance).<br />

6. Establishing a European network for wildlife health surveillance. Kuiken,<br />

T., Ryser-Degiorgis M.-P., Gavier-Widén, D., Gortázar, C. Rev. Sci. Tech.<br />

Off. Int. Epiz, 2011, 30 (3), 755-761.<br />

7. REPORT OF THE MEETING OF THE OIE BIOLOGICAL STANDARDS<br />

COMMISSION Paris, 14–16 September 2011. At: www.oiw.int/<br />

8.WildTech at: www.wildtechproject.com/wildtech/<br />

9. European Association <strong>of</strong> Veterinary Laboratory Diagnosticians<br />

(EAVLD), at www.<strong>eavld</strong>.org/<br />

10. Wildlife Disease Association (WDA), at : www.wildlifedisease.org


S3 - O - 01<br />

SCHMALLENBERG VIRUS OUTBREAK IN THE NETHERLANDS: ROUTINE DIAGNOSTICS AND TEST<br />

RESULTS<br />

Ruth Bouwstra 1 , Wim van der Poel 1 , Eric de Kluijver 1 , Betty Verstraten 1 , Johan Bongers 1<br />

1<br />

Central Veterinary Institute, <strong>of</strong> Wageningen University and Research Centre (CVI-Lelystad), Department <strong>of</strong> Virology, Lelystad, The Netherlands.<br />

Introduction<br />

At the end <strong>of</strong> 2011, a new Orthobunya virus named<br />

Schmallenberg virus (SBV), was discovered in Germany. Soon<br />

thereafter in the Netherlands, the virus was associated with<br />

decreased milk production, watery diarrhoea and fever in dairy<br />

cows, and subsequently also with congenital malformations in<br />

calves, lambs and goat kids (1,2). By the 20th <strong>of</strong> December 2011<br />

in the Netherlands malformations in new-borns <strong>of</strong> ruminants were<br />

made notifiable. After a notification by a farmer or veterinarian, a<br />

maximum <strong>of</strong> five malformed new-borns per farm were<br />

necropsied. The diagnosis <strong>of</strong> Schmallenberg virus disease was<br />

based on the pathologic findings and RT-PCR test results <strong>of</strong><br />

brain tissue <strong>of</strong> the malformed new-borns. In addition blood<br />

samples from mothers <strong>of</strong> affected new-borns were collected and<br />

tested for antibodies against SBV using a virus neutralization<br />

test.<br />

Schmallenberg virus, routine diagnostics, test results<br />

Materials & methods<br />

Brain samples were tested with the RT-PCR for Schmallenberg<br />

virus that has been developed by the Friedrich-Loeffler Institute in<br />

Germany. Serum samples were tested in a virus neutralisation<br />

test (VNT) against SBV developed by the Central Veterinary<br />

Institute. A virus isolate from brain tissue <strong>of</strong> a lamb, 4th passage<br />

on VERO cells, was used in the test (3).<br />

Results<br />

Between 20th <strong>of</strong> December and March the 8th, in total 1165 brain<br />

tissue samples were tested in the RT-PCR: 577 originated from<br />

lambs, 444 from calves and 42 from goat kids. In the VNT 681<br />

blood samples were tested: 329 originated from ewes, 329 from<br />

cows and 23 from goats. Results showed that 8% <strong>of</strong> the tested<br />

calf brains, 31% <strong>of</strong> the tested lamb brains and 12% <strong>of</strong> the tested<br />

goat kid brains were RT-PCR positive. The number <strong>of</strong> malformed<br />

lambs and RT-PCR positive lamb brains decreases over time<br />

while the number <strong>of</strong> malformed calves and RT-PCR positive calf<br />

brains increases (fig 1,2). In the VNT 95% <strong>of</strong> the ewes, 93% <strong>of</strong><br />

the cows and 23% <strong>of</strong> the goats tested positive. Combining the<br />

results <strong>of</strong> the RT-PCR and the VNT, 20% <strong>of</strong> all farms tested<br />

positive in both the RT-PCR (genetic material <strong>of</strong> SBV in brain<br />

tissue in malformed new-borns) and the VNT (antibodies against<br />

SBV in blood from mothers <strong>of</strong> affected new-borns ). The results<br />

reported here are based on testing up to the first week <strong>of</strong> March<br />

<strong>2012</strong>. Additional test results which will be available in June will<br />

be presented also.<br />

Figure 2: Number <strong>of</strong> RT-PCR positive (dark grey bars) and RT-<br />

PCR negative (light grey bars) calf brain tissue samples per<br />

week.<br />

Discussion & conclusions<br />

In goats the number <strong>of</strong> seropositives is far lower than in sheep<br />

and cattle. Less samples from goats were tested and the<br />

estimated seroprevalence is therefore less precise. Furthermore<br />

number <strong>of</strong> RT-PCR positive calves and goat kids is far lower than<br />

in sheep. Given that goats, in contrast to cattle and sheep, are<br />

<strong>of</strong>ten housed indoors, and the SBV is supposedly transmitted by<br />

Culicoides vectors, this lower number <strong>of</strong> test positive results<br />

would not be surprising. In addition, the difference in pregnancy<br />

length between cows and sheep might explain why it is more<br />

difficult to detect genetic material <strong>of</strong> SBV in brain tissue <strong>of</strong> calves<br />

and, assuming that infection <strong>of</strong> cows and sheep was around the<br />

same period, it might also explain the difference in number <strong>of</strong><br />

malformations and RT-positives over time. Supposing that all<br />

malformations notificated are truly caused by the Schmallenberg<br />

virus, on farm level, diagnostic sensitivity <strong>of</strong> the RT-PCR is much<br />

lower in comparison with the VNT.<br />

Acknowledgements<br />

We thank our colleagues from the Dispatching Service Unit and<br />

the Virological Diagnostic Laboratory for execution <strong>of</strong> the<br />

diagnostic testing.<br />

References<br />

1. Muskens J, Smolenaars AJG, van der Poel WHM, Mars MH, van<br />

Wuijckhuise L, Holzhauer M, van Weering H, Kock P. Diarrhea and loss <strong>of</strong><br />

production on Dutch dairy farms caused by the Schmallenburg virus.<br />

Tijdschr Diergeneeskd. <strong>2012</strong>; 137: 112-5.<br />

2. Van den Brom R, Luttikholt SJM, Lievaart-Peteron K, Pperkamp NHMT,<br />

Mars MH, van der Poel WHM, Vellema P. Epizootic <strong>of</strong> ovine congenital<br />

malformations associated with Schmallenberg virus infection. Tijdschr<br />

Diergeneeskd <strong>2012</strong>; 137: 106-11.<br />

3. Loeffen WLA et al. J Virol Methods <strong>2012</strong> (in preparation)<br />

Figure 1: Number <strong>of</strong> RT-PCR positive (dark grey bars) and RT-<br />

PCR negative (light grey bars) lamb brain tissue samples per<br />

week.


S3 - O - 02<br />

25 YEARS OF PASSIVE SURVEILLANCE OF BATS IN THE NETHERLANDS. MOLECULAR<br />

EPIDEMIOLOGY AND EVOLUTION OF EBLV-1.<br />

Introduction<br />

Since 1986 a reactive surveillance program is running in the<br />

Netherlands to investigate the presence <strong>of</strong> European Bat<br />

Lyssavirusses (EBLV-1 and EBLV-2) in bats. In this program all<br />

contact cases, with human or pets, are send to the laboratory in<br />

Lelystad and tested for EBLV with a EU prescribed immune<br />

fluorescence test (IFT). Up to date almost 5000 samples have<br />

been tested. The main reservoir for EBLV is Eptesicus serotinus,<br />

334 positive cases so far which is about 22% <strong>of</strong> the samples<br />

tested, second species is Myotis dasycneme, with almost 4%<br />

positive thus far (Table 1). EBLV-2 has not been detected in the<br />

Netherlands since 1993.<br />

Materials & methods<br />

Bat brains were tested using the prescribed fluorescent antibody<br />

test (FAT). For conformation a real time PCR test on the N-gene<br />

was used. On the same nucleic acid isolation used for the realtime<br />

PCR N and G-gene sequencing was performed on an ABI<br />

sequencer according to the manufacturers protocol. The species<br />

<strong>of</strong> all submissions were determined by a bat expert.<br />

The MCC phylogenetic tree, estimates <strong>of</strong> the rate <strong>of</strong> molecular<br />

evolution (substitutions per site per year) and the TMRCA for the<br />

alignments were inferred using a Bayesian MCMC method in the<br />

BEAST package.<br />

B. Kooi, A. Vogel and E. Van Weezep<br />

Central Veterinary Institute <strong>of</strong> Wageningen UR, Lelystad, The Netherlands.<br />

Lyssavirus, EBLV, bats, surveillance, molecular epidemiology<br />

Results & Discussion<br />

We performed a molecular epidemiology study on a collection <strong>of</strong><br />

more then 40 EBLV-1 viruses that currently circulate in the<br />

Netherlands. Based on the sequences <strong>of</strong> the coding regions <strong>of</strong><br />

the N and G genes a phylogenetic analysis was performed in<br />

which the two distinct lineages or subgroups EBLV-1a and EBLV-<br />

1b were clearly visible (Fig. 1). Surprisingly the larger EBLV-1a<br />

group could be further subdivided into three phylogenetic groups<br />

that were consistent with topology. Two groups were<br />

predominantly found in the northern provinces whereas the third<br />

group was spread from east to west in the middle <strong>of</strong> the country.<br />

A most recent common ancestor analysis was performed which<br />

confirmed a distinct difference in origin <strong>of</strong> the two EBLV-1<br />

subgroups. Whereas the EBLV-1a subgroup migrated into<br />

Europe via the east-west route, EBLV-1b apparently entered the<br />

continent from the south, most likely through the Iberian<br />

Peninsula.<br />

.<br />

Table 1: Passive lyssavirus surveillance <strong>of</strong> bats (1986-2011)<br />

Species tested postitive<br />

no determination 10 -<br />

Myotis mystacinus 22 -<br />

Myotis nattereri 10 -<br />

Myotis daubentonii 124 -<br />

Myotis dasycneme 138 5<br />

Pipistrellus pipistrellus 2216 -<br />

Pipistrellus nathusii 358 -<br />

Nyctalus noctula 77 -<br />

Nyctalus leisleri 4 -<br />

Eptesicus serotinus 1491 334<br />

Vespertilio murinus 13 -<br />

Plecotus auritus 257 -<br />

total 4720 339<br />

Figure 1: Phylogenetic analysis <strong>of</strong> EBLV-1 based on G-protein<br />

coding regions <strong>of</strong> EBLV-1 from bats from the Netherlands (2004-<br />

2011)<br />

References<br />

1. Drummond AJ, Rambaut A. BEAST: Bayesian evolutionary analysis by<br />

sampling trees. BMC Evol Biol. 2007;7(1):214.<br />

2. Cliquet, F and Barrat J, 2008. Rabies (chapter 2.1.13). Manual <strong>of</strong><br />

diagnostic tests and vaccines for terrestrial animals, OIE (World<br />

Organisation for Animal Health), Paris, 1: 304-322


S3 - O - 03<br />

DIAGNOSIS OF Q FEVER IN DAIRY CATTLE BY PHASE-SPECIFIC MILK-SEROLOGY<br />

Böttcher J 1 , Frangoulidis D 2 , Schumacher M 1 , Janowetz B 1 , Gangl A 1 , Alex M 1<br />

1 Bavarian Animal Health Service, Poing, Germany,<br />

2<br />

Bundeswehr Institute <strong>of</strong> Microbiology, Munich, Germany<br />

Q fever, serology<br />

Introduction<br />

Dairy cattle herds are frequently endemically infected by C.<br />

burnetii the causative pathogen <strong>of</strong> Q fever. Shedding <strong>of</strong> C.<br />

burnetii at calving and in milk is <strong>of</strong> special concern. Chronic<br />

infection frequently results in an increased level <strong>of</strong> milk-shedding.<br />

The diagnostic value <strong>of</strong> phase-specific serology was assessed by<br />

a longitudinal study on milk samples and puerperal swabs in an<br />

endemically infected dairy cattle herd with about 100 dairy cows.<br />

PhII titer<br />

10000<br />

1000<br />

100<br />

10<br />

PhI-serology<br />

negative<br />

positive<br />

Materials & methods<br />

Cows were kept in two groups (MS and RB) with close contact<br />

within one barn. The same calving boxes are used for both<br />

groups. Individual milk samples were collected from March 2010<br />

until December 2011; since August 2010 puerperal swab were<br />

collected from a proportion <strong>of</strong> cows within 16 h after calving.<br />

Samples: 405 milk samples, 59 puerperal, 9 samplings (MS) and<br />

465 milk samples, 64 puerperal swabs, 10 samplings (RB).<br />

Phase-specific ELISAs were performed as described (1) with<br />

minor modifications: Milk samples were diluted 1/5 log 10 and the<br />

titer was calculated at 20% (OD%) <strong>of</strong> the positive control (serum<br />

control diluted 1/400).<br />

Frequency <strong>of</strong> positive samples<br />

1,4<br />

1,2<br />

1<br />

0,8<br />

0,6<br />

0,4<br />

0,2<br />

0<br />

swabs<br />

milk samples<br />

1003 1004 1007 1008 1009 1010 1011 1012 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112<br />

52 46 98 0 46 57 57 52 0 54 58 51 52 0 56 45 47 0 52 46<br />

0 0 0 5 11 2 10 9 5 9 8 5 0 7 5 3 6 8 11 6<br />

Sampling (YYMM) and number <strong>of</strong> milk-samples and swabs, respectively<br />

Fig. 1: Frequency <strong>of</strong> positive qPCR (CI95%) was plotted over<br />

year/month <strong>of</strong> sample collection (YYMM, top), the number <strong>of</strong> milk<br />

samples (middle) and the number <strong>of</strong> analysed puerperal swabs (bottom).<br />

Cut-<strong>of</strong>fs for milk&swabs were ≥10 C.b./ml milk and ≥1 C.b./swab.<br />

qPCR (C.b. log10/swab and ml milk, resp.)<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

1003<br />

1004<br />

puerperal swabs<br />

milk<br />

1007<br />

1008<br />

1009<br />

1010<br />

1011<br />

1012<br />

1101<br />

1102<br />

1103<br />

1104<br />

Sampling (YYMM)<br />

Fig. 2: Amount <strong>of</strong> C. burnetii-target in qPCR-positive swabs and milk<br />

samples. qPCR scored positive if the titer exceeded 1 C.b./swab and ml<br />

milk, respectively.<br />

Milk samples and puerperal swabs were analysed by quantitative<br />

(q) PCR (2). Target was quantified by a standard curve derived<br />

from diluted C. burnetii reference isolate Nine Mile RSA493<br />

(kindly provided by C. Heydel, Giessen).<br />

The status <strong>of</strong> milk-shedding was assessed by two procedures: (1)<br />

Mean shedding per cow was calculated (C.b./ml) and classified<br />

as 1000 C.b./ml. (2) Shedding<br />

pattern was defined as chronic shedding (CS; >3 times ≥10<br />

C.b./ml) irrespective <strong>of</strong> negative results in between. Intermittent<br />

shedding (IS3, IS2, IS1) with 3, 2 and 1 positive result, and<br />

negative, if all samples gave less than 10 C.b./ml. Classification<br />

relied on a minimum <strong>of</strong> three samples except for IS3, this status<br />

required four samplings.<br />

1105<br />

1106<br />

1107<br />

1108<br />

1109<br />

1110<br />

1111<br />

1112<br />

1<br />

1003<br />

1004<br />

1007<br />

1009<br />

1010<br />

1011<br />

1012<br />

1102<br />

1103<br />

1104<br />

Milk sampling (YYMM)<br />

Fig. 3: PhII-titers in cows (500. Only one multiparous CScow<br />

remained just below 500. Positive predictive value for PhI 500<br />

was 40%, whereas it was 25% for PhI 100 and PhII 100 , respectively.<br />

In contrast to CS-cows, IS1/IS2-cows were characterized by a<br />

striking instability <strong>of</strong> PhI- and PhII-results. Even after positive<br />

puerperal swabs seroconversion in milk was not a regular finding.<br />

PhI - /PhII + -pattern in young cows indicates the episode <strong>of</strong><br />

puerperal shedding at herd level and PhI 500 is a suitable<br />

screening for heavy milk shedders.<br />

Acknowledgements<br />

The present study was supported financially by the Free State Bavaria and the<br />

Bavarian Joint Founding Scheme for the Control and Eradication <strong>of</strong> Contagious<br />

Livestock Diseases (Bayerische Tierseuchenkasse).<br />

References<br />

1. Böttcher, J., Vossen, A., Janowetz, B., Alex, M., Gangl, A., Randt A., and Meier,<br />

N. (2011): Insights into the dynamics <strong>of</strong> endemic Coxiella burnetii infection in<br />

cattle by application <strong>of</strong> phase-specific ELISAs in an infected dairy herd.<br />

Veterinrary Microbiology, 151, 291-300.<br />

2. Böttcher, J., Frangoulidis, D., Schumacher, M., Janowetz, B., Gangl, A., Alex,<br />

M. (<strong>2012</strong>): Diagnostic value <strong>of</strong> Coxiella burnetii phase I and II antibody titers in<br />

individual milk samples <strong>of</strong> cows. In preparation.<br />

1108<br />

1109<br />

1111<br />

1112


S3 – O - 04<br />

EQUINE NOCARDIOFORM PLACENTITIS & ABORTION OUTBREAK<br />

AND FARM-BASED RISK FACTOR STUDY, 2010-2011<br />

Craig N. Carter 1 , Jacki C Cassady 1 , Noah Cohen 2 , Laura A Kennedy 1 , Erdal Erol 1 , Tamara Malm 1 ,<br />

Mike Donahue 1 , Steve Sells 1 , Neil Williams 1 Jacqueline Smith 1 , Roberta Dwyer 3<br />

1<br />

Veterinary Diagnostic Laboratory, Department <strong>of</strong> Veterinary Science, University <strong>of</strong> Kentucky, Lexington, KY USA<br />

2 Large Animal Clinic, College <strong>of</strong> Veterinary Medicine, Texas A&M University, College Station, TX USA<br />

3 Gluck Equine Research Center, Department <strong>of</strong> Veterinary Science, University <strong>of</strong> Kentucky, Lexington, KY USA<br />

Equine, horse, placentitis, abortion, epidemiology<br />

Introduction<br />

The syndrome now known as equine nocardi<strong>of</strong>orm placentitis &<br />

abortion (NPA) that was first seen in Kentucky in the 1980’s, is<br />

caused by nocardi<strong>of</strong>orm actinomycete bacteria. Crossiella equi,<br />

characterized and named in 2002 (1), and various species <strong>of</strong><br />

Amycolatopsis, 3 species characterized and named in 2003 (2),<br />

have been isolated from NPA cases since 1988. Two species <strong>of</strong><br />

Streptomyces (3), both isolated in 1999, have also been<br />

associated with NPA cases. The infection results in late<br />

abortions, stillbirths and premature foaling, sometimes leading to<br />

early neonatal mortality. The epidemiology <strong>of</strong> transmission is not<br />

known. Interestingly, nocardi<strong>of</strong>orm actinomycetes have rarely<br />

been isolated from any tissue other than placenta. Most cases<br />

have occurred in the Bluegrass region <strong>of</strong> Kentucky (Figure 1) but<br />

cases have also been diagnosed in Florida (4), Italy (5), and<br />

South Africa. During the 2010-2011 equine reproductive season,<br />

the Lexington laboratory identified gram positive branching bacilli<br />

in 118 cases by culture and PCR. The high incidence seen in<br />

this season precipitated this farm-level study to identify possible<br />

risk factors for NPA.<br />

Materials & methods<br />

A 34 question survey instrument was designed to gather<br />

information on farm size, number <strong>of</strong> mares, mare density, water<br />

sources, feeding/grazing practices, pasture management, pre<br />

and post breeding practices, drugs/supplements used and<br />

diseases/syndromes seen on the farms. A total <strong>of</strong> 497 farms<br />

were contacted with 153 responding. Five <strong>of</strong> the farms were<br />

disqualified, leaving 148 for analysis. Data was analysed using<br />

chi-squared, Wilcoxon rank-sum tests and logistic regression<br />

analysis using S-PLUS, v8.2.<br />

Results<br />

Of the farms surveyed, 98 were affected by NPA during the 2010-<br />

2011 with 50 farms unaffected. The average number <strong>of</strong> mares<br />

affected per farm was 4 (range 1-64). The total number <strong>of</strong> mares<br />

at risk on all farms was 8075 with 429 mares diagnosed with NPA<br />

(5.3% all farms, 6.5% affected farms). The outbreak involved 15<br />

cities and 27 zip codes. Affected farms tended to have greater<br />

acreage (p < 0.0001), had more mares bred (p < 0.0002) and<br />

greater mare density (p = 0.0095). The distribution <strong>of</strong> the number<br />

<strong>of</strong> hours horses were grazed from January-March differed<br />

significantly for affected farms (median 8 hours) and unaffected<br />

farms (median 12 hours). The proportion <strong>of</strong> affected farms using<br />

progesterone pre-breeding was significantly lower than that <strong>of</strong><br />

unaffected farms (OR = 2.9). The proportion <strong>of</strong> affected farms<br />

that administered HCG post-breeding was significantly lower than<br />

that <strong>of</strong> unaffected farms (OR = 2.3) Finally, the proportion <strong>of</strong><br />

affected farms that administered NSAIDs pre-breeding was<br />

significantly lower than that for unaffected farms (OR = 2.8).<br />

Discussion & conclusion<br />

The significant findings <strong>of</strong> farm size, mare numbers and density<br />

related to affected farms may be an indicator that simple<br />

crowding and intensive management <strong>of</strong> mares is a risk factor for<br />

NPA. It might also indicate greater opportunities for diagnosis <strong>of</strong><br />

NPA. The finding <strong>of</strong> longer grazing times January-March on<br />

unaffected farms may indicate a protective effect, i.e. the barn<br />

environment may be more risky. Use <strong>of</strong> progesterone prebreeding,<br />

HCG post-breeding and NSAIDs pre-breeding might<br />

indicate practices that reduce risk but further studies are needed<br />

as the chance for false discovery is high. These factors could be<br />

better assessed in a case-control study in which the unit <strong>of</strong><br />

analysis is the mare, not the farm. A spatial analysis <strong>of</strong> the data<br />

was not performed.<br />

Acknowledgements<br />

We owe much gratitude to the farms that participated in the<br />

survey, the Kentucky Thoroughbred Farm Managers Club, the<br />

Kentucky Thoroughbred Association (KTA), the Kentucky<br />

Thoroughbred Owners and Breeders (KTOB), the Kentucky<br />

Association <strong>of</strong> Equine Practitioners (KAEP), David Switzer and<br />

Vickie Garcia <strong>of</strong> KTA and KTOB, Drs. Ernie Martinez and Stuart<br />

Brown <strong>of</strong> the KAEP, and Barbara Dunaway and Susan Neal,<br />

UKVDL.<br />

References<br />

1.Donahue JM., Williams NM., Sells SF and Labeda DP: 2002, Crossiella<br />

equi sp nov., isolated from equine placentas. International Journal <strong>of</strong><br />

Systemic and Evalutionary Microbiology. 52, 2169-2173.<br />

2. Labeda DP, Donahue JM, Williams NM, et al.: 2003, Amycolatopsis<br />

kentuckyensis sp. nov., Amycolatopsis lexingtonensis sp. nov. and<br />

Amycolatopsis pretoriensis sp. nov., isolated from equine placentas.Int J<br />

Syst Evol Microbiol. 53(Pt 5):1601-5.<br />

3. Labeda DP, Price NP, Kroppenstedt RM, et al.: 2009, Streptomyces<br />

atriruber sp. nov. and Streptomyces silaceus sp. nov., two novel species <strong>of</strong><br />

equine origin. Int J Syst Evol Microbiol. 59(Pt 11):2899-903.<br />

4. Christiensen BW, Roberts JF, Pozor MA, et al.: 2006, Nocardi<strong>of</strong>orm<br />

placentitis with isolation <strong>of</strong> Amycolatopsis spp in a Florida-bred mare.J Am<br />

Vet Med Assoc. 228(8):1234-9.<br />

5. Cattoli G, Vascellari M, Corrò M, et al.: 2004, First case <strong>of</strong> equine<br />

nocardi<strong>of</strong>orm placentitis caused by Crossiella equi in Europe. Vet Rec.<br />

154(23):730-1.<br />

180<br />

160<br />

140<br />

120<br />

# <strong>of</strong> Cases<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

90 91 92 93 94 95 96 97 98 99 00 01 02 03 04 05 06 07 08 09 10 11<br />

Year<br />

Figure 1: History <strong>of</strong> NPA cases diagnosed in Kentucky


S3 - O - 05<br />

A NEW DIAGNOSTIC TOOL FOR BOVINE TUBERCULOSIS-IDEXX M.BOVIS ANTIBODY TEST KIT<br />

J. Lawrence 1 , N. Djuranovic 1 , C. Egli 2<br />

1 - IDEXX Labs, Inc., Livestock and Poultry Diagnostics, Westbrook, Maine, USA<br />

2 - IDEXX Switzerland,Livestock and Poultry Diagnostics, Bern, Switzerland, USA<br />

Tuberculosis, ELISA, IFN-g, SICCT, IDEXX<br />

Introduction<br />

The IDEXX M. bovis antibody test kit is an ELISA designed to<br />

detect the presence <strong>of</strong> antibody to M. bovis in bovine serum and<br />

plasma samples. This test could improve bTB detection <strong>of</strong> TB -<br />

infected animals in TB-infected herds or could be an easy, cost<br />

effective surveillance tool in TB-negative regions. The M. bovis<br />

ELISA test is currently in the approval process for the OIE<br />

registry <strong>of</strong> tests. Also, all documentation has been submitted to<br />

USDA for product licensure.<br />

2. Schiller, I., et al. 2010. Bovine tuberculosis: a review <strong>of</strong> current<br />

and emerging diagnostic techniques in view <strong>of</strong> their relevance for<br />

disease control and eradication. Transbound. Emerg. Dis.<br />

57:205–220.<br />

Material & methods<br />

Characterized serum and plasma samples were obtained from<br />

worldwide sources and were used to validate the M. bovis ELISA.<br />

Two temporal series were produced from animals exposed to M.<br />

bovis and followed over time. Positive samples from 3 different<br />

countries were obtained from either culture positive animals (n =<br />

307). Sample sets (n = 100) with varying skin or gamma<br />

interferon results were evaluated to demonstrate subsets <strong>of</strong><br />

positive animals within TB-infected herds (n = 45) and the power<br />

<strong>of</strong> combining tests to increase overall sensitivity. Negative<br />

samples (n = 1473) were obtained from 4 different countries with<br />

samples originating from negative herds, states or regions. In<br />

addition, to understand potential cross-reactivity with other<br />

Mycobacteria, samples were obtained from animals exposed to<br />

large doses <strong>of</strong> M. paratuberculosis, M. avium and M. kansasii or<br />

from a herd with a high Johne’s antibody levels.<br />

All samples were evaluated on an M. bovis antibody kit<br />

manufactured at production scale, according to the standard kit<br />

protocol. Briefly, samples and kit controls were diluted 1:50 in a<br />

sample diluent and applied to microtiter plates. After a 60-minute<br />

incubation, the plates were washed, followed by the addition <strong>of</strong><br />

an anti-bovine HRP conjugate (30-minute incubation). After<br />

another plate wash, TMB substrate was added. After color<br />

development <strong>of</strong> 15 minutes, plates were read on a<br />

spectrophotometer at 450nm. Sample optical densities were<br />

compared to those <strong>of</strong> the kit positive control to derive Sample-to<br />

Positive (S/P) ratios. Samples with S/P ratios <strong>of</strong> >/= 0.30 were<br />

considered positive for M. bovis antibodies.<br />

Results<br />

Data from the M. bovis temporal series revealed that animals can<br />

develop antibody titers within weeks <strong>of</strong> exposure and that an<br />

antibody response can be boosted after the application <strong>of</strong> a skin<br />

test. The M. bovis ELISA detected 197/307 samples from culture<br />

positive animals resulting in a sensitivity <strong>of</strong> 64.2% . Using a single<br />

test method, between 71.1% and 75.6% <strong>of</strong> herds would have<br />

been detected. Combining tests resulted in an increase in herd<br />

sensitivity to between 86.7% (GIFN and ELISA) and 97.8%<br />

(GIFN, SICCT and ELISA). On negative sample sets, the M.<br />

bovis ELISA demonstrated a specificity <strong>of</strong> 98% with no cross<br />

reactivity observed on M. paratuberculosis (both experimental<br />

and field infected) or M. avium samples. Transient, low level<br />

reactivity was observed with animals inoculated with large doses<br />

<strong>of</strong> M. kansasii.<br />

Discussion & conclusions<br />

The strategic supplemental use <strong>of</strong> the IDEXX M. bovis antibody<br />

test represents a fast, easy, objective and cost effective option for<br />

use in bTB programs and can increase overall diagnostic power<br />

by detectiing subsets <strong>of</strong> infected animals missed by current<br />

methods. This test's high specificity allows for an application <strong>of</strong><br />

this test in bTB- negative regions.<br />

References<br />

1. de la Rua-Domenech, R., A. T. Goodchild, H. M. Vordermeier,<br />

R. G. Hewinson, K. H. Christiansen, and R. S. Clifton-Hadley.<br />

2006. Ante mortem diagnosis <strong>of</strong> tuberculosis in cattle: a review <strong>of</strong><br />

the tuberculin tests, γ-interferon assay and other ancillary<br />

diagnostic techniques. Res. Vet. Sci. 81:190-210.


S3 – O - 06<br />

DETECTION OF SCHMALLENBERG VIRUS IN THE UK<br />

Julie Peake, Christopher Finnegan, Falko Steinbach, Akbar Dastjerdi and Anna La Rocca<br />

AHVLA (Animal Health and Veterinary Laboratories), MVIU (Mammalian Virus Investigation Unit), Dept <strong>of</strong> Virology, Addlestone, UK<br />

Introduction<br />

In November 2011, colleagues at the FLI using samples taken in<br />

Schmallenberg (Germany) detected a new virus. The virus<br />

identified has subsequently, but tentatively, been named<br />

Schmallenberg Virus (SBV)(1). This virus belongs to the genus<br />

Orthobunyavirus within the Bunyaviridae family. Since the end <strong>of</strong><br />

2011, traces <strong>of</strong> the virus were detected in several European<br />

countries. In the second half <strong>of</strong> January <strong>2012</strong>, the first PCR<br />

positive cases were detected in the UK, following reports <strong>of</strong><br />

clinical signs <strong>of</strong> the disease in aborted lambs from the south-east<br />

part <strong>of</strong> the country.<br />

Materials and Methods<br />

RNA was extracted using Trizol® (Invitrogen) followed by<br />

extraction <strong>of</strong> the acquoeus phase with the QIAamp Viral RNA<br />

mini Kit (Qiagen) and qPCR carried out with the primers<br />

described below using the QuantiTect® Virus RT-PCR kit<br />

(Qiagen). ELISA tests were carried out using the ID Screen®<br />

Schmallenberg virus Indirect ELISA (ID.VET)<br />

Results<br />

The first generation <strong>of</strong> qPCR assays based on the L segment<br />

were developed and kindly made available by FLI. In parallel, we<br />

used the virus sequence information to generate a set <strong>of</strong> primers<br />

for an AHVLA-qPCR. Shortly after, a second generation <strong>of</strong><br />

primers and probe for qPCR was available through FLI.<br />

Subsequently, LSI also marketed a kit. All these qPCRs are<br />

based on the S genome segment. We are, at present, comparing<br />

the use <strong>of</strong> Texas Red as fluorescent dye compared to FAM, in<br />

the current assays, as this could increase the sensitivity <strong>of</strong> the<br />

assay, especially the resolution <strong>of</strong> results close to the cut-<strong>of</strong>f<br />

value.<br />

Using the FLI second generation qPCR we have tested brains <strong>of</strong><br />

sheep and cattle to closely monitor the outbreak across the<br />

country. We, very interestingly, have observed that the spread <strong>of</strong><br />

the infection has been very quick across south but quite slow<br />

towards north <strong>of</strong> the UK. These observations lead us to the<br />

hypothesis that there could have been several incursions <strong>of</strong> the<br />

virus into the UK.<br />

Discussion and Conclusions<br />

Since mid-March, the number <strong>of</strong> positive brains from sheep has<br />

been declining and this correlates with the advancement <strong>of</strong> the<br />

lambing season. However, we have also detected five positive<br />

cases in April/mid-May in England. This challenges parts <strong>of</strong> our<br />

understanding about the transmission <strong>of</strong> SBV via midges, the<br />

observation <strong>of</strong> further cases until mid-May on Channel Islands is<br />

less astonishing.<br />

A particular challenging aspect <strong>of</strong> the SBV diagnosis has been<br />

the finding that the brains <strong>of</strong> many submitted suspects with the<br />

disease case definition were negative in our PCR assay. It is<br />

considered possible that in some instances the virus has been<br />

cleared by the developing immune system <strong>of</strong> the lambs (which<br />

would be traceable via antibodies to SBV). However, a recent<br />

study suggested looking also at further tissues besides brain to<br />

detect SBV (2).<br />

Ongoing studies in our lab are accordingly looking into the<br />

comparison <strong>of</strong> meconium, foetal fluid, and the dissemination <strong>of</strong><br />

SBV in brains. We are also using the recently established<br />

commercial ELISA to complement PCR results.<br />

References<br />

1. H<strong>of</strong>fmann B, Scheuch M, Höper D, Jungblut R, Holsteg M, Schirrmeier<br />

H, et al. Novel orthobunyavirus in cattle, Europe, 2011. Emerg Infect Dis<br />

<strong>2012</strong> Mar;18(3):469-72.<br />

2. Bilk S, Schulze C, Fischer M, Beer M, Hilnak A, H<strong>of</strong>fmann B. Organ<br />

distribution <strong>of</strong> Schmallenberg virus RNA in malformed newborns. Vet<br />

Microbiol <strong>2012</strong> Mar 30 Epub ahead <strong>of</strong> print.<br />

Tuberculosis, ELISA, IFN-g, SICCT, IDEXX


S3 - O - 07<br />

DETECTION OF WNV ENZOOTIC CIRCULATION IN HORSES WITH NEUROLOGICAL SIGNS AND IN<br />

CAPTIVE SENTINEL CHICKENS IN THE PREFECTURE OF THESSALONIKI, GREECE<br />

Chrysostomos I. Dovas 1 , Serafeim C. Chaintoutis 1 , Alexandra Chaskopoulou 2 , Nikolaos Diakakis 3 ,<br />

Ilias Bouzalas 1 , Maria Papanastassopoulou 1 , Kostas Danis 4 , Anna Papa 5 , Orestis Papadopoulos 1<br />

1<br />

Aristotle University <strong>of</strong> Thessaloniki, Faculty <strong>of</strong> Veterinary Medicine, Laboratory <strong>of</strong> Microbiology and Infectious Diseases, Thessaloniki, Greece<br />

2<br />

United States Department <strong>of</strong> Agriculture-Agricultural Research Service, European Biological Control Laboratory, Thessaloniki, Greece<br />

3<br />

Aristotle University <strong>of</strong> Thessaloniki, Faculty <strong>of</strong> Veterinary Medicine, Clinic <strong>of</strong> Companion Animals, Thessaloniki, Greece<br />

4<br />

Hellenic Centre for Disease Control and Prevention, Athens, Greece<br />

5<br />

Aristotle University <strong>of</strong> Thessaloniki, Faculty <strong>of</strong> Medicine, A’ Laboratory <strong>of</strong> Microbiology, Thessaloniki, Greece<br />

WNV, detection, characterization, horses, sentinel chickens<br />

Introduction<br />

During 2010, a large outbreak <strong>of</strong> West Nile virus (WNV)<br />

infections occurred in Greece, with 262 reported human cases.<br />

Among them, 197 were diagnosed with West Nile neuroinvasive<br />

disease (WNND) and 35 patients died. The epidemic reoccurred<br />

in 2011. Laboratory methods, including RT-PCR, ELISA,<br />

microtiter plaque reduction neutralization test (micro-PRNT),<br />

indirect immun<strong>of</strong>luorescence antibody test (IFAT) and virus<br />

isolation, were used to detect WNV enzootic circulation in horses<br />

with neurological signs (passive surveillance), as well as in<br />

captive sentinel chickens (active surveillance), prior to the onset<br />

<strong>of</strong> human WNND cases in the prefecture <strong>of</strong> Thessaloniki, Greece.<br />

Materials & methods<br />

Blood samples were obtained from 17 horses with neurological<br />

signs, during the epidemic <strong>of</strong> 2010 (with the first case being<br />

reported on August 1). Furthermore, blood was collected from<br />

one horse which showed ataxia during the epidemic <strong>of</strong> 2011, on<br />

August 13. Also, in May 2011, 7 coops <strong>of</strong> 6 chickens each were<br />

placed at the edges <strong>of</strong> Thessaloniki City and blood samples were<br />

collected weekly until October. In conjunction to blood sampling<br />

from chicken, mosquitoes were sampled using 28 CO 2 -baited<br />

CDC light traps.<br />

The sera obtained from the 18 horses were assayed with two IgM<br />

capture ELISA (MAC-ELISA) kits (IDEXX IgM WNV Ab Test and<br />

IDVet ID Screen West Nile IgM Capture) for the detection <strong>of</strong><br />

WNV specific IgM antibodies, sign <strong>of</strong> recent infection, as well for<br />

specific IgG antibodies, using IFAT. Plasma samples from these<br />

horses were tested with real-time RT-PCR for WNV (LSI TaqVet<br />

West Nile Virus – Dual IPC). Chicken sera were assayed by<br />

competitive ELISA (cELISA) (IDVet ID Screen West Nile<br />

Competition) for the detection <strong>of</strong> specific antibodies, and positive<br />

results were confirmed by micro-PRNT. Plasma samples from the<br />

seropositive chickens were screened using a WNV specific RT-<br />

PCR and confirmed using nested RT-PCR, employing specific<br />

primers for the WNV “Nea Santa-Greece-2010” strain, followed<br />

by sequencing <strong>of</strong> the NS3 gene for further molecular<br />

characterization. Vero cell cultures were inoculated with plasma<br />

samples from the RT-PCR positive chickens, for virus isolation.<br />

The collected mosquitoes were being counted and<br />

morphologically identified. 146 pooled samples, each one<br />

consisting <strong>of</strong> 50 mosquitoes <strong>of</strong> the same species (123 Culex<br />

pipiens and 23 Culex modestus pools) were examined by three<br />

different WNV specific real-time RT-PCR protocols, as well as<br />

with the nested RT-PCR protocol,<br />

Results<br />

WNV specific antibodies were detected in all 18 horse cases <strong>of</strong><br />

2010 and 2011, while the IgG IFAT titer was high (>250) in all <strong>of</strong><br />

them. Three horses were found positive with real-time RT-PCR<br />

(Ct ~39). Regarding chickens, seroconversion was detected in 11<br />

birds (10 in Western Thessaloniki). The first seroconversion<br />

occurred on June 29, a month before the onset <strong>of</strong> human cases<br />

in the area. The seroconversion rate peaked on August 17 with 4<br />

seropositive birds and the last seroconversion occurred on<br />

September 28. WNV was detected in 2/11 seropositive chickens<br />

with RT-PCR, from samples taken one week prior to<br />

seroconversion. According to BLAST algorithm, the partial NS3<br />

sequence presented highest nucleotide sequence identity<br />

(99,73%) to the strain “Nea Santa-Greece-2010”, responsible for<br />

the 2010 Greek epidemic. The inferred partial NS3 amino acid<br />

sequence was 100% identical to that <strong>of</strong> the “Nea Santa-Greece-<br />

2010”. The strains maintained the amino acid substitution H 249 P,<br />

which may be associated with increased virulence. WNV was<br />

successfully isolated in cell cultures. Chicken serum neutralizing<br />

antibody titers to WNV ranged between 20 and 40. Culex<br />

mosquito populations peaked in mid-June, ~10 days prior to the<br />

first chicken seroconversion, remained high throughout the<br />

summer and showed a significant drop in September. The most<br />

prevalent mosquito species in both residential and agricultural<br />

areas were Culex pipiens, followed by Culex modestus Ficalbi.<br />

Only one out <strong>of</strong> the 123 Culex pipiens pools tested was found<br />

positive for the WNV “Nea Santa-Greece-2010” strain using the<br />

nested RT-PCR, while the application <strong>of</strong> all different real-time RT-<br />

PCR protocols resulted in detection <strong>of</strong> additional mosquito<br />

flaviviruses, indicating problems <strong>of</strong> detection specificity.<br />

Figure 1. Location <strong>of</strong> mosquito traps (n=28) and sentinel chicken<br />

coops for WNV surveillance in Thessaloniki, 2011. Blue bullets<br />

represent the mosquito traps, while yellow dots represent the<br />

location <strong>of</strong> the 7 chicken coops. The 10 seroconverted chickens<br />

belonged to the coops No. 1, 2 and 3 (Western Thessaloniki),<br />

while one more chicken belonged to the coop No. 5, in Eastern<br />

Thessaloniki.<br />

Discussion & conclusion<br />

With passive surveillance, 18 clinical cases <strong>of</strong> horses were<br />

detected mainly in suburban areas <strong>of</strong> Thessaloniki. Unfortunately<br />

there could not be successfully used as an early warning system,<br />

given that the first human case in the prefecture <strong>of</strong> Thessaloniki<br />

was recorded 15 days prior to the onset <strong>of</strong> clinical cases in<br />

horses, both in 2010 and 2011. On the contrary, with the second<br />

surveillance system, it was shown that the “Nea Santa-Greece-<br />

2010” strain became endemic in Greece, as it was detected,<br />

molecularly identified and isolated early in 2011. Most<br />

importantly, enzootic circulation <strong>of</strong> WNV was successfully<br />

detected one month prior to human cases. As a result, health<br />

authorities were informed in a timely manner and were facilitated<br />

the successful implementation <strong>of</strong> preparedness plans to protect<br />

public health and minimize the impact <strong>of</strong> the epidemic <strong>of</strong> 2011. In<br />

regards to the use <strong>of</strong> real-time RT-PCR for vector surveillance,<br />

the methods need further optimization and validation, concerning<br />

the specificity <strong>of</strong> WNV detection and results should be further<br />

confirmed by sequencing.<br />

References<br />

Danis, K, Papa, A, Papanikolaou, E, Dougas, G, Terzaki, I, Baka, A, Vrioni,<br />

G, Kapsimali, V, Tsakris, A, Kansouzidou, A, Tsiodras, S, Vakalis, N,<br />

Bonovas, S, Kremastinou, J (2011). Ongoing outbreak <strong>of</strong> West Nile virus<br />

infection in humans, Greece, July to August 2011. Eurosurveillance,<br />

16(34), pii=19951.


S3 - O - 08<br />

SCHMALLENBERGVIRUS: SEROLOGICAL STUDIES IN GERMAN HOLDINGS<br />

Horst Schirrmeier, Bernd H<strong>of</strong>fmann, Martin Beer<br />

Institute <strong>of</strong> Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald-Insel Riems, Germany<br />

Introduction<br />

In August 2011 first indications <strong>of</strong> a previously unknown disease<br />

in dairy cattle were observed in North Rhine-Westphalia,<br />

Germany, and in the Netherlands. Fever, decreased milk<br />

production and in some cases also diarrhea were the major<br />

symptoms. In November 2011, the detection <strong>of</strong> a novel<br />

Orthobunyavirus in blood samples <strong>of</strong> acutely diseased dairy cows<br />

was reported by the Friedrich-Loeffler-Institut, and the the virus<br />

was named as Schmallenberg virus (SBV). Since December<br />

2011, congenital malformations were observed, initially in sheep<br />

and goats, and subsequently also in calves. A real-time RT-PCR<br />

was developed very early after the first detection, and first<br />

serological methods were established for serological studies. The<br />

first findings including investigations about the SBV-prevalence in<br />

Germany are reported.<br />

Materials & methods<br />

The following methods for the detection <strong>of</strong> SBV-specific<br />

antibodies were established and used:<br />

1. serumneutralization test (SNT) in BHK-21 cells, clone<br />

CT (L164, CCLV, Insel Riems) using the cell culture<br />

isolate “Schmallenberg BH80”.<br />

2. indirect immun<strong>of</strong>luorescence in a multi-plate format<br />

with SBV-infected BHK-21 cells, using the SBVinfected<br />

cell clone BRS5 (L194, CCLV Insel Riems) as<br />

antigen matrix (Vero cells were used as an alternative<br />

cell culture).<br />

3. Indirect ELISA (in house development as well as a<br />

prototype batch <strong>of</strong> a commercial ELISA assay).<br />

Within a nationwide first monitoring study, cattle, sheep and goat<br />

sera were tested for SBV antibodies. Furthermore, sera from<br />

malformed calves and pre-colostral sera <strong>of</strong> newborn ruminants<br />

were investigated to improve diagnostic options in clinically and<br />

pathologically suspected, but PCR negative cases. A few sera<br />

from wild ungulates were also included, and in vitro crossreaction<br />

studies with other members <strong>of</strong> the Orthobunyavirus<br />

family were performed.<br />

Results & discussion<br />

SBV-specific antibodies could be detected in serum samples<br />

from cattle and sheep from all included federal states. The<br />

observed incidence was within a range <strong>of</strong> 10% to 60 %, with a<br />

single outlier <strong>of</strong> more than 90 %. Positive results were obtained<br />

most frequently in the epidemiological “core regions” in the<br />

Northwestern parts <strong>of</strong> Germany. The number <strong>of</strong> sero-reactors<br />

corresponded very well with the frequency <strong>of</strong> the notified clinical<br />

cases. However, the in-herd prevalence differed substantially and<br />

varied between single reactors and up to 80 % positive animals,<br />

and was generally higher in cattle herds than in sheep flocks.<br />

Apart from domestic ruminants, antibodies could be also detected<br />

in samples from bisons, roe deer, red deer and from one camel<br />

and lama, respectively. Furthermore, SBV-specific antibodies<br />

could be also detected in samples from PCR-negative animals as<br />

well as in sera <strong>of</strong> malformed calves without colostrum uptake.<br />

Our results will allow better insights into the epidemiolocial<br />

background <strong>of</strong> the SBV-infection, will expand the knowledge<br />

about pathogenetic mechanisms, and are able to substantiate the<br />

SBV case definition in PCR-negative animals.<br />

References<br />

H<strong>of</strong>fmann B, Scheuch M, Höper D, Jungblut R, Holsteg M, Schirrmeier H,<br />

Eschbaumer M, Goller KV, Wernike K, Fischer M, Breithaupt A,<br />

Mettenleiter TC, Beer M. (<strong>2012</strong>). Novel orthobunyavirus in cattle, Europe,<br />

2011, Emerg Infect Dis. <strong>2012</strong> Mar;18(3):469-72<br />

Orthobunyavirus, serology,


S3 - O - 09<br />

DIFFERENT DIAGNOSTIC TOOLS FOR A BROAD RANGE OF MACAVIRUSES AND THEIR<br />

RESERVOIR AND SUSCEPTIBLE HOSTS<br />

Christine Foerster 1 , Matthias Koenig 1 , Heinz-Juergen Thiel 1 , Jens-Ove Heckel 2<br />

1<br />

Institute <strong>of</strong> Virology, diagnostic laboratory, Justus-Liebig-University Giessen, Germany<br />

2<br />

Zoo Landau, Germany<br />

Macavirus, malignant catarrhal fever, reservoir and susceptible host<br />

Introduction<br />

Malignant catarrhal fever (MCF) is a fatal disease <strong>of</strong> Artiodactyla,<br />

which is caused by different macaviruses within the subfamily<br />

Gammaherpesvirinae (GHV) <strong>of</strong> the family Herpesviridae. Until<br />

now there are at least 6 distinct macaviruses known to cause<br />

MCF: Alcelaphine herpesvirus 1 (AlHV-1), ovine herpesvirus 2<br />

(OvHV-2), caprine herpesvirus 2 (CpHV-2), white tailed deer-<br />

MCF-virus (MCF-WTD), an unclassified macavirus from ibex and<br />

a virus resembling alcelaphine herpesvirus 2 (AlHV-2). Other<br />

closely related viruses are the hippotragine herpesvirus 1 (HiHV-<br />

1 in roan antelope) and GHV in springbok, impala, oryx, muskox<br />

and aoudad.<br />

MCF usually occurs only sporadically. Within the framework <strong>of</strong><br />

differential diagnostics the detection <strong>of</strong> macaviruses was<br />

stimulated since the first cases <strong>of</strong> bluetongue disease were<br />

noticed in northern Europe.<br />

Typical reservoir hosts which do not develop clinical disease are<br />

sheep, goat and wildebeest. Several species <strong>of</strong> Artiodactyla are<br />

susceptible to MCF, for example bison, cattle, deer and moose.<br />

In addition pigs, goats and experimentally infected rabbits show<br />

typical clinical symptoms like fever, mucosal erosions, nasal and<br />

lacrimal discharge, corneal opacity and skin lesions.<br />

Undirected virological methods like virus propagation in cell<br />

culture and electron microscopy are not promising with regard to<br />

macaviruses. PCR is the method <strong>of</strong> choice for laboratory<br />

diagnosis. Until now most laboratories use the OvHV-2 specific<br />

PCR (Baxter et al., 1993) for samples <strong>of</strong> cattle.<br />

Objective <strong>of</strong> this work was to compare and improve the<br />

diagnostic tools for the detection <strong>of</strong> a broad spectrum <strong>of</strong> potential<br />

MCF-viruses.<br />

Discussion & conclusions<br />

The competitive ELISA was clearly superior to SNA with regard<br />

to time, effort and quality <strong>of</strong> result. ELISA is recommended for<br />

routine diagnostic use, especially for reservoir hosts.<br />

The importance <strong>of</strong> broadly reactive PCR methods was<br />

demonstrated by 18 positive results <strong>of</strong> wildlife samples. All 18<br />

cases led to negative results in the OvHV-2-specific PCR, but<br />

reacted positive in the PAN-Macavirus-PCR. Cloning, sequencing<br />

and phylogenetic analyses <strong>of</strong> the amplificates led to the detection<br />

<strong>of</strong> CpHV-2, CpHV-2-like and BoHV-6-like macaviruses.<br />

For the first time CpHV-2-induced MCF was detected in two<br />

Banteng-cattle. Until now, CpHV-2 has been considered as a<br />

cause <strong>of</strong> MCF only in deer and moose. This implies that routine<br />

OvHV-2-specific-diagnostic in cattle is insufficient.<br />

The establishment <strong>of</strong> an OvHV-2-CpHV-2-discriminating realtime<br />

PCR allowed rapid differentiation between the two most common<br />

causative agents <strong>of</strong> MCF in Europe. This method is suitable for<br />

application in routine diagnostics <strong>of</strong> small ruminants and cattle.<br />

For the application in wildlife samples the conventional PCR is<br />

more suited, because primer pairs were selected for a broad<br />

range <strong>of</strong> macaviruses and the conventional PCR appeared to be<br />

more sensitive.<br />

References<br />

1. Baxter, S. I., Pow, I., Bridgen, A. and Reid, H. W. (1993). PCR detection<br />

<strong>of</strong> the sheep-associated agent <strong>of</strong> malignant catarrhal fever. Arch Virol 132<br />

(1-2), 145-59.<br />

2. Russell, G. C., Stewart, J. P. and Haig, D. M. (2009). Malignant<br />

catarrhal fever: A review. Vet J 179 (3), 324-35.<br />

Materials & methods<br />

Two new conventional PCR-methods and an OvHV-2-CpHV-2-<br />

discriminating realtime PCR were established, which allowed to<br />

detect a broad spectrum <strong>of</strong> macaviruses. They were compared to<br />

the commonly used OvHV-2-specific PCR. Amplified fragments<br />

were cloned, sequenced and used for phylogenetic analyses.<br />

Furthermore two antibody-test-systems, ELISA and serum<br />

neutralisation assay (SNA), were compared to each other.<br />

Samples <strong>of</strong> 276 wild and zoo animals, comprising 54 different<br />

genera <strong>of</strong> the family Bovidae, Cervidae, Camelidae and<br />

Giraffidae, 230 goats and 58 sheep were examined.<br />

Results<br />

39 cases out <strong>of</strong> 276 wild ruminants (14 %) led to a positive result<br />

in at least one procedure. In approximately half <strong>of</strong> the samples<br />

(19) OvHV-2 was identified, in 18 CpHV-2 or CpHV-2 like virus.<br />

Samples <strong>of</strong> one waterbuck contained sequences related to<br />

BoHV-6. 19 <strong>of</strong> 39 macavirus-positive animals showed MCF like<br />

disease.<br />

CpHV-2 was identified in conjunction with two cases <strong>of</strong> MCF in<br />

Banteng cattle.<br />

5 out <strong>of</strong> 230 goats (2 %) showed MCF like symptoms and 4 <strong>of</strong><br />

them also pathological and histological signs <strong>of</strong> MCF. In all<br />

samples <strong>of</strong> these animals OvHV-2 could be demonstrated.<br />

Samples <strong>of</strong> 92 (42 %) goats obtained positive results in at least<br />

one PCR method. Interestingly single and also double infection <strong>of</strong><br />

OvHV-2 and CpHV-2 were found.<br />

In contrast to the goats only OvHV-2 was found in 27 sheep<br />

samples (47 %).<br />

Realtime PCR was able to detect OvHV-2 and CpHV-2 in all<br />

cases <strong>of</strong> MCF-diseased animals. In latently infected animals high<br />

cycle treshold values were found.<br />

In some reservoir hosts antibodies were found but no viral DNA.<br />

33 <strong>of</strong> 59 animals (56 %) were found positive via ELISA, but<br />

negative by SNA, while only one animal (1,6 %) was estimated<br />

positive by SNA and negative via ELISA.


S3 - O - 10<br />

DIAGNOSTIC ASPECTS OF SUID HERPESVIRUS 1 INFECTION IN WILD BOAR<br />

Adolf Steinrigl, Sandra Revilla-Fernández, Eveline Wodak, Zoltán Bagó, Friedrich Schmoll<br />

AGES Institute for Veterinary Disease Control, Mödling, Austria<br />

SuHV-1, wild boar, prevalence, molecular characterization<br />

Introduction<br />

Many infectious diseases that are strictly controlled in livestock<br />

may remain unnoticed in wild animals, posing the risk <strong>of</strong> entering<br />

domestic animal populations with potentially devastating<br />

consequences. Suid herpesvirus 1 (SuHV-1), the causative agent<br />

<strong>of</strong> Aujeszky’s disease (AD), was eradicated from domestic pig<br />

populations in many European countries, including Austria. At the<br />

same time, SuHV-1 infection is common in European wild boar,<br />

as shown by a number <strong>of</strong> studies, reporting detection <strong>of</strong> SuHV-1<br />

DNA or antibodies in wild boar (1). An additional indicator <strong>of</strong><br />

SuHV-1 presence in wild boar are occasional cases <strong>of</strong> AD in<br />

hunting dogs, usually in close connection to hunting events (2).<br />

Recently, we reported about the first detection <strong>of</strong> SuHV-1 in<br />

Austrian wild boar and the molecular characterization <strong>of</strong> SuHV-1<br />

field isolates from wild boar and hunting dogs (3).<br />

In the course <strong>of</strong> the Wild Animal Survey 2011, a nationwide study<br />

that was partly financed by the Austrian Federal Ministry <strong>of</strong><br />

Health, additional sera and tissue samples from Austrian wild<br />

boar were collected and analysed, with the aim to provide a<br />

representative estimate <strong>of</strong> the prevalence <strong>of</strong> both, SuHV-1 DNA<br />

and antibodies, in Austrian wild boar. This work shows the results<br />

<strong>of</strong> this survey and reflects our experiences with detection and<br />

molecular characterization <strong>of</strong> SuHV-1 in wild boar.<br />

Materials & methods<br />

Wild boar tissues and sera were sampled according to a survey<br />

design that was based on recent wild boar hunting bag statistics.<br />

Anti-SuHV-1 gB ELISA, real-time PCR (qPCR), amplification and<br />

sequencing <strong>of</strong> the SuHV-1 glycoprotein C coding region,<br />

phylogenetic analysis and virus isolation were performed as<br />

described previously (3).<br />

Results<br />

From April 2011 to January <strong>2012</strong>, tissues from 298 wild boars<br />

were sampled, corresponding to 78% <strong>of</strong> the scheduled sample<br />

size for the whole country. Wild boar sera <strong>of</strong> sufficient amount<br />

and quality were available from 259 animals. Results <strong>of</strong><br />

serological and virological screening are shown in Table 1. All<br />

qPCR positive wild boars were adult females at 2-4 years <strong>of</strong> age.<br />

One <strong>of</strong> them was also serologically positive, while the other two<br />

animals were seronegative.<br />

Table 1: Results <strong>of</strong> serological (ELISA) and virological (qPCR)<br />

screening <strong>of</strong> Austrian wild boars<br />

Samples Samples Prevalence<br />

tested positive<br />

ELISA 259 59 22.8%<br />

qPCR 298 3 1.0%<br />

Virus was successfully isolated from one <strong>of</strong> the qPCR-positive,<br />

yet seronegative, wild boars: tissue homogenate from a qPCRpositive<br />

tonsil was inoculated onto PK15 cells, resulting in<br />

massive cytopathic effect within 24 hours post inoculation. An<br />

attempt to isolate virus from the other two qPCR positive animals<br />

was unsuccessful. Phylogenetic analysis <strong>of</strong> SuHV-1 gC<br />

sequences amplified from the three qPCR positive wild boars<br />

sampled in 2011 showed that all three animals hosted virus that<br />

was identical in gC sequence and belonged to one <strong>of</strong> the<br />

previously described lineages <strong>of</strong> SuHV-1 (3), which are currently<br />

present in Austria (Figure 1).<br />

Figure 1: Neighbour Joining tree, based on a 639 bp alignment <strong>of</strong><br />

the SuHV-1 partial gC coding region. Sequences were amplified<br />

from wild boars (WB), hunting dogs (HD) and from a domestic pig<br />

(DP). Year <strong>of</strong> isolation is in brackets. Numbers along the<br />

branches indicate the percentage <strong>of</strong> 1000 bootstrap replicates.<br />

Discussion & conclusions<br />

The presented study confirms the presence <strong>of</strong> SuHV-1 in<br />

Austrian wild boar. SuHV-1 seroprevalence detected during the<br />

Wild Animal Survey 2011 was lower than that previously<br />

described (3), which is probably due to the more representative<br />

sampling strategy applied for the current study. In contrast to the<br />

high numbers <strong>of</strong> wild boars with detectable SuHV-1 antibodies,<br />

SuHV-1 DNA was detected by qPCR in only a few animals.<br />

Furthermore, Cq-values <strong>of</strong> qPCR-positive tissue pools or<br />

individual organs were relatively high (30 - 38 Cq), indicating a<br />

low viral load in these tissues. This is also supported by our<br />

previous observation <strong>of</strong> weak immunohistochemistry signals in<br />

qPCR positive tissues (3) and by the fact that virus isolation was<br />

positive in a single case only. In summary, our results indicate<br />

that most SuHV-1 infected wild boars harbour virus in a latent<br />

form, whereas only a small proportion <strong>of</strong> infected wild boars is<br />

actually shedding virus and may infect other susceptible animals.<br />

This is also the most likely explanation for the relatively rare<br />

cases <strong>of</strong> AD in hunting dogs, despite rising numbers <strong>of</strong> wild boars<br />

in many European countries and the frequent involvement <strong>of</strong><br />

hunting dogs in wild boar hunting. Despite the fact that only few<br />

potentially virus shedding wild boars were found in this study,<br />

direct contact between wild boars and domestic pigs could result<br />

in SuHV-1 transmission. Feeding <strong>of</strong> wild boar intestines to<br />

domestic animals, especially pigs, dogs and cats, is another<br />

reasonable possibility <strong>of</strong> transmission and must be strictly<br />

discouraged.<br />

Acknowledgements<br />

This study was financially supported in part by the Austrian<br />

Federal Ministry <strong>of</strong> Health. The excellent technical assistance <strong>of</strong><br />

the laboratory staff <strong>of</strong> the Departments for Molecular Biology,<br />

Virology and Electron Microscopy, and Pathology is<br />

acknowledged.<br />

References<br />

1. Müller T, Hahn EC, Tottewitz F, Kramer M, Klupp BG, Mettenleiter TC,<br />

Freuling C (2011). Pseudorabies virus in wild swine: a global perspective.<br />

Arch. Virol., 156, 1691-705.<br />

2. Cay AB, Letellier C (2009). Isolation <strong>of</strong> Aujeszky’s disease virus from<br />

two hunting dogs in Belgium after hunting wild boar. Vlaams<br />

Diergeneeskundig Tijdschrift, 78, 194-195.<br />

3. Steinrigl A, Revilla-Fernández S, Kolodziejek J, Wodak E, Bagó Z,<br />

Nowotny N, Schmoll F, Köfer J (2011). Detection and molecular<br />

characterization <strong>of</strong> Suid herpesvirus type 1 in Austrian wild boar and<br />

hunting dogs. Vet Microbiol., Dec 30. [Epub ahead <strong>of</strong> print]


S3 - O - 11<br />

PRELIMINARY VALIDATION OF THE ID SCREEN ® SCHMALLENBERG VIRUS INDIRECT ELISA<br />

P. Pourquier, 1 , C. Loic, 1 , S. Zientara, 2 , E. Breard, 2 , C. Sailleau, 2 , C. Viarouge, 2 , A.-B.Cay, 3 , N. De Regge, 3<br />

1<br />

IDvet, Montpellier, France<br />

2 ANSES Maisons-Alfort, France<br />

Introduction<br />

Schmallenberg virus (SBV) is the name given to a vectortransmitted<br />

orthobunyavirus related to Shamonda virus, initially<br />

reported in November 2011 to cause foetal congenital<br />

malformations and stillbirths in cattle, sheep, and goats. The<br />

virus has since been detected in Germany, the Netherlands,<br />

Belgium, France, Luxembourg, Italy, Spain and the United<br />

Kingdom.<br />

Serological testing is essential in order to study and control this<br />

new emerging disease. While SBV specific antibodies can be<br />

detected by virus neutralization tests and indirect<br />

immun<strong>of</strong>luoresence, these techniques are time-consuming, not<br />

suitable for high-throughput, and do not <strong>of</strong>fer standardized result<br />

interpretation.<br />

Available as <strong>of</strong> March <strong>2012</strong>, the ID Screen ®<br />

Schmallenberg<br />

virus Indirect ELISA is the first ELISA developed for SBV<br />

diagnosis. The test allows for the detection <strong>of</strong> SBV antibodies in<br />

ruminant serum and plasma. It is a rapid, standardized assay<br />

which is automatable and therefore suited to high throughput<br />

testing.<br />

This study presents validation data for this test. Data will be<br />

added for any second generation tests developed by IDvet<br />

between March and June <strong>2012</strong>.<br />

Material & methods<br />

- The ID Screen ® Schmallenberg virus Indirect ELISA was<br />

performed as per manufacturer’s specifications.<br />

- Specificity was studied using panels <strong>of</strong> sera collected prior to<br />

2010.<br />

- Sensitivity was evaluated on sera tested positive with the virus<br />

neutralization test (VNT).<br />

- An experimental infection was performed in April <strong>2012</strong> in order<br />

to evaluate the seroconversion response in cattle, sheep and<br />

goats.<br />

Results & Discussion<br />

Preliminary validation studies indicate excellent test specificity<br />

and high correlation with other serological techniques, making the<br />

the ID Screen ® Schmallenberg virus Indirect ELISA an efficient<br />

tool for disease surveillance and epidemiological studies.<br />

3 CODA-CERVA, Department <strong>of</strong> Viral Diseases, Brussels, Belgium<br />

Schmallenberg virus, serology, ELISA


Oral presentations<br />

“New techniques in bacteriology,<br />

parasitology and pathology”<br />

(4 th session)


S4 - K - 01<br />

MALDI-TOF AND OTHER NEW DIAGNOSTIC<br />

Markus Kostrzewa<br />

Bruker Daltonik GmbH, Bioanalytical development, Bremen, Germany<br />

Microorganism identification, FT-IR spectroscopy, Raman spectroscopy, mass spectrometry, MALDI-TOF MS<br />

Introduction – the “status quo”<br />

Since more than one hundred years, identification <strong>of</strong><br />

microorganisms mainly is performed by methods investigating<br />

their phenotypic characteristics. Gram stain together with<br />

microscopy in combination gives a first quick hint about identity.<br />

The screening <strong>of</strong> series <strong>of</strong> tests for biochemical capabilities is<br />

used to generate a pattern which is more or less species specific.<br />

Although these tests now have been automated for the detection<br />

<strong>of</strong> the most prevalent microorganisms, the accuracy <strong>of</strong><br />

biochemical tests is limited, in particular for organisms with few<br />

characteristic biochemical features like Gram negative nonfermenting<br />

bacteria <strong>of</strong> anaerobes. Even worse is the situation for<br />

rare bacteria, mycobacteria or fungi where mainly labourintensive<br />

manual methods like API are applied for identification.<br />

Therefore, there is an urgent need for modern automated<br />

methods for identification which can be introduced in routine<br />

laboratories to shorten time-to-result and decrease the overall<br />

costs <strong>of</strong> the laboratories. Promising approaches appearing in the<br />

areas <strong>of</strong> molecular biology, optical technologies and mass<br />

spectrometry have appeared through the recent years and are on<br />

the way to revolutionize microbial analysis.<br />

Methods based on molecular biology have already been<br />

introduced in many laboratories, mainly PCR based tests to<br />

screen for dedicated microorganisms, e.g. food pathogens. While<br />

having the advantage <strong>of</strong> high sensitivity and short analysis time<br />

one drawback <strong>of</strong> PCR based tests is that they generally need a<br />

pre-assumption which microorganism is searched for, i.e. a<br />

dedicated primer design. To apply DNA analysis to broad species<br />

identification, PCR typically is coupled with subsequent sequence<br />

determination <strong>of</strong> the amplicon. Sequencing <strong>of</strong> the 16S rDNA<br />

region nowadays is regarded the gold standard for species<br />

identification. Nevertheless, 16S rDNA sequencing has never<br />

made it to the routine but is mainly a second- or third-line method<br />

for cases where other methods have failed. Reasons are the high<br />

costs, elaborate sample handling, and PCR-related<br />

contamination risk. Further, in quite a number <strong>of</strong> cases,<br />

determination <strong>of</strong> the 16S ribosomal RNA gene sequence is not<br />

sufficient for the discrimination <strong>of</strong> closely related species and<br />

further gene sequences have to be analysed, increasing time to<br />

result, costs and labour, significantly. Novel next generation<br />

sequencing technologies might change the position <strong>of</strong><br />

sequencing in future by their enormous throughput capability, but<br />

today they are still expensive and labour-intensive research tools.<br />

The upcoming alternatives<br />

Vibrational spectroscopy<br />

Optical technologies have been shown to be powerful for<br />

microorganism identification as well as for analyses on the<br />

subspecies level. These techniques are based on a molecular<br />

fingerprint <strong>of</strong> the entire biomass, proteins, carbohydrates, lipids,<br />

nucleic acids. Analyses are not depending on reagent kits, labels<br />

or drugs, and these technologies are applicable to a broad<br />

spectrum <strong>of</strong> organisms.<br />

Raman spectroscopy is a non-destructive optical technique,<br />

based on scattering <strong>of</strong> light by molecules, the so called “Raman<br />

effect”. The atoms in a molecule move in so called vibrational<br />

modes. When light, i.e. a photon, and a molecule interact, some<br />

<strong>of</strong> the energy <strong>of</strong> the photon can be transferred to the molecule.<br />

Thereby, one <strong>of</strong> the vibrational modes in the molecule is excited.<br />

The energy <strong>of</strong> the scattered photon is reduced by the exact<br />

amount <strong>of</strong> energy received by the molecule. The energy required<br />

to excite a molecular vibration is exactly defined and depends on<br />

the masses <strong>of</strong> the atoms involved in the vibration, on the type <strong>of</strong><br />

chemical bonds between these atoms, on the structure <strong>of</strong> the<br />

molecule and on interactions with its environment and other<br />

molecules. The spectrum <strong>of</strong> the scattered light is used as a<br />

pattern to identify microbes.<br />

In contrast, Fourier-transformation infrared spectroscopy uses<br />

absorption <strong>of</strong> an infrared light spectrum (not a single wavelength)<br />

to create a fingerprint <strong>of</strong> the whole cell composition. Mainly<br />

transmission <strong>of</strong> the light is used to create the characteristic<br />

pr<strong>of</strong>ile. The first deviation <strong>of</strong> the transmission pr<strong>of</strong>ile spectrum is<br />

generated and sophisticated mathematical algorithms are used to<br />

identify strains by matching against a database or to build<br />

relational dendrograms for classification.<br />

Restrictions <strong>of</strong> the optical technologies is mainly that they require<br />

a strict standardization <strong>of</strong> cultivation conditions (time, media,<br />

temperature) as these are influencing the cell content and<br />

thereby the optical fingerprint. The speed <strong>of</strong> the analyses is also<br />

counteracted by the fact that a pure culture is required (i.e. <strong>of</strong>ten<br />

no analysis from primary culture can be performed).<br />

Mass spectrometry<br />

Some technological approaches for identification and further<br />

analysis <strong>of</strong> microorganisms based on mass spectrometry may<br />

have the most impact on microbial characterization in the near<br />

future. One <strong>of</strong> these approaches is based on a combination <strong>of</strong><br />

molecular biology and electrospray time-<strong>of</strong>-flight mass<br />

spectrometry (ESI-TOF MS). In a first step, several target regions<br />

<strong>of</strong> the DNA from the unknown microorganism are amplified with<br />

generic primer pairs. This is creating a set <strong>of</strong> amplification<br />

products with specific molecular masses. Subsequently, the PCR<br />

products are purified with a magnetic bead based system and<br />

then injected in liquid phase into the ESI-TOF mass<br />

spectrometer. During the ionization process, the two DNA strands<br />

<strong>of</strong> an amplicon are separated. The mass <strong>of</strong> both DNA strands is<br />

measured in the ESI-TOF very accurately. From the combination<br />

<strong>of</strong> the accurate masses <strong>of</strong> the both pairing strands the<br />

composition <strong>of</strong> the polynucleotides (i.e. the number <strong>of</strong> A, C, G,<br />

and T, respectively) can be determined. The projection <strong>of</strong> the<br />

base compositions <strong>of</strong> the selected PCR products into a database<br />

is used for the identification <strong>of</strong> an organism. The more PCR<br />

products are analysed, the more accurate/discriminative the<br />

identification can be. One advantage <strong>of</strong> this technology is that in<br />

parallel to species identification also other characteristics, e.g.<br />

resistance genes or virulence factors, can be detected. On the<br />

other hand, this approach still relies on a relatively complex<br />

(although in parts automated) workflow, and both the instrument<br />

as well as consumable costs are significant.<br />

The second, currently most successful and promising mass<br />

spectrometry technology is matrix-assisted laserdesorption/ionization<br />

time-<strong>of</strong>-flight mass spectrometry (MALDI-<br />

TOF MS). In contrast to electrospray ionization the MALDI<br />

process (generating the ions measured in the TOF analyser) is<br />

performed from a solid sample. For this, the sample is deposited<br />

on a target plate, overlaid with the so called matrix, generally an<br />

organic acid related to cinnamic acid, which co-crystalizes with<br />

the sample. This matrix supports the evaporation <strong>of</strong> the sample<br />

and its ionization after absorption <strong>of</strong> the energy <strong>of</strong> a pulsed laser<br />

beam. One advantage <strong>of</strong> this technology is that the sample can<br />

be easily prepared <strong>of</strong>fline before measurement and then<br />

transported or stored until measurement.<br />

For one molecular biology based approach, MALDI-TOF MS is<br />

used to detect specific RNA fragments generated by PCR<br />

followed by transcription <strong>of</strong> both DNA strands and the cleavage <strong>of</strong><br />

the transcripts by RNAses. The exact masses <strong>of</strong> the fragments<br />

can be used as pattern for identification and typing <strong>of</strong><br />

microorganisms. Alternatively, minisequencing/primer extension<br />

<strong>of</strong> specific regions after PCR amplification, followed by mass<br />

determination in a MALDI-TOF MS, has been shown to have<br />

some potential for typing or resistance detection.<br />

While none <strong>of</strong> these DNA based mass spectrometry approaches<br />

made it to routine yet, MALDI-TOF MS whole cell protein pr<strong>of</strong>iling<br />

has entered many clinical and veterinary microbiology<br />

laboratories, already. This MALDI-TOF fingerprinting is based on<br />

the specific pattern <strong>of</strong> high abundant proteins <strong>of</strong> microbial cells<br />

which can be measured quickly and accurately by MALDI-TOF<br />

mass spectrometry. Sample preparation is very simple, fast and<br />

cheap: A small amount <strong>of</strong> a bacterial colony is transferred to a<br />

position on a MALDI sample target and smeared on it as a thin<br />

film. More robust cells can be broken by a short extraction with


acid and organic solvent. Then a droplet <strong>of</strong> matrix solution is<br />

added. Thereby, the cells are destructed, the protein content gets<br />

released and is embedded into the matrix crystals which are<br />

forming after solvent evaporation. During the subsequent MALDI-<br />

TOF analysis the most abundant proteins, in particular ribosomal<br />

proteins, are detected as a characteristic molecular pr<strong>of</strong>ile<br />

(fingerprint). This pattern then is compared with the pr<strong>of</strong>iles<br />

stored in a database. Identification is done based on highest<br />

similarity and a minimum identity score. The whole process, from<br />

sample preparation to identification, can be as fast as ten<br />

minutes for a single sample/colony, less than one hour for about<br />

one hundred samples. For difficult to prepare microorganisms like<br />

actinomycetes, fungi, or mycobacteria special protocols are<br />

available which allow also their analysis with high success rate.<br />

Dependance on cultivation conditions is generally low. Thereby,<br />

this is one <strong>of</strong> the broadest applicable technologies for microbial<br />

identification. MALDI-TOF fingerprinting is already used in many<br />

clinical and veterinary laboratories, frequently as the first-line<br />

routine identification method. Its accuracy has been reported in<br />

various publications. Extensive commercial databases are<br />

available, but also alternative or supplementary laboratoryspecific<br />

libraries can be established. Recent developments in the<br />

direction <strong>of</strong> direct specimen analysis, epidemiology, virulence<br />

typing, and resistance determination will make the technology<br />

even more valuable for the clinical microbiology.<br />

Conclusions<br />

New technologies for microorganism identification and<br />

characterization are no more on the horizon, they are on the<br />

playground. In the near future it can be expected that a<br />

combination <strong>of</strong> molecular and spectroscopic/spectrometric<br />

technologies will substitute most <strong>of</strong> phenotypic/biochemical<br />

assays, increasing accuracy, efficiency and speed <strong>of</strong> the clinical<br />

microbiology laboratory.


S4 - O - 01<br />

SUITABILITY OF RECOMBINANT PROTEINS FOR THE DIAGNOSIS OF LEPTOSPIROSIS IN PIGS<br />

Ripp, Ulrike 1,2 , Truyen, Uwe 2 , Homeier, Timo 2<br />

1<br />

Synlab.vet Leipzig, Leipzig, Germany<br />

2<br />

Institute <strong>of</strong> Animal Hygiene and Veterinary Public Health, Faculty <strong>of</strong> Veterinary Medicine, Leipzig, Germany<br />

Porcine leptospirosis, LipL32, ELISA<br />

Introduction<br />

Leptospirosis is a widespread disease in the world with a high<br />

zoonotic potential. The momentary gold standard is the<br />

microscopic agglutination test (MAT). The specificity seems to be<br />

good, but the sensitivity is controversially discussed. It is lacking<br />

as a non-objective test procedure. Additionally, the detection <strong>of</strong><br />

antibodies depends on the used serovar panel. Apart from the<br />

sensitivity, there is a risk <strong>of</strong> laboratory infection, as the leptospires<br />

used in the MAT have to be alive.<br />

Therefore, current research focuses on the development <strong>of</strong> an<br />

ELISA for the serological diagnosis <strong>of</strong> leptospirosis. Whole-cell<br />

based ELISA did not provide the desired results in sensitivity and<br />

specificity. Recombinant proteins, especially those that are only<br />

expressed in pathogenic leptospires, seem to be a more<br />

promising agent as an antigen. The goal <strong>of</strong> this study was to find<br />

a serovar-independent antigen which can be used in a screening<br />

ELISA in comparison to the MAT.<br />

Materials & methods<br />

Microscopic agglutination test (MAT):<br />

8 serovars (L. Bratislava, Canicola, Grippotyphosa, Pomona,<br />

Tarassovi, Sejroe, Copenhageni, Saxkoebing) were used in the<br />

MAT panel. The highest titer where there were still less than 50%<br />

<strong>of</strong> free leptospires to be seen in the dark-field-microscopy, was<br />

the endtiter.<br />

Samples with a titer ≥ 1:100 were considered positive.<br />

Porcine serum samples were screened at dilutions <strong>of</strong> 1:50, 1:100<br />

and 1:200. Samples that were still positive at 1:200 were further<br />

diluted.<br />

ELISA:<br />

The recombinant protein LipL32 was used as antigen. It is highly<br />

conserved among the pathogenic serovars <strong>of</strong> Leptospira (1) and<br />

is not expressed in apathogenic leptospires. LipL32 contributes to<br />

a large amount <strong>of</strong> the outer membrane proteins <strong>of</strong> leptospires. It<br />

has already been tested in an ELISA for human, canine and<br />

bovine leptospirosis (2,3).<br />

Flat-bottom microtiter plates were coated with LipL32 and<br />

incubated over night at 4°C. The optimal antigen concentration<br />

was determined by checkerboard titration. The range was from<br />

1,17 g to 0,55 ng / well.<br />

The plate was blocked the next day with skim milk and again<br />

incubated over night at 4°C. After washing the plate, serum was<br />

added at a dilution <strong>of</strong> 1:100 and incubated for 1 hour at room<br />

temperature on a shaker. After the next washing step, the<br />

conjugate (a goat anti-pig antibody that was conjugated to<br />

horseradish-peroxidase) was added. The plate was incubated for<br />

1 hour at room temperature on a shaker, washed, and the<br />

substrate (TMB) was added. The reaction was stopped after 5<br />

minutes with 0,5M H 2 SO 4 and read at 450/630nm.<br />

Control sera:<br />

The positive control serum was gained from a pig that was<br />

vaccinated 3 times in intervals <strong>of</strong> 2 weeks with 10 9 CFU (Colony<br />

forming units) <strong>of</strong> formalin inactivated Leptospira Pomona.<br />

The serum <strong>of</strong> a specific pathogen free pig (that was negative in<br />

the MAT, too) was used as negative control.<br />

Results<br />

Optimum antigen concentration:<br />

The titration <strong>of</strong> LipL32 did not show a plateau with additional<br />

decrease <strong>of</strong> OD values. Changes <strong>of</strong> the conjugate and serum<br />

dilutions as well as different blocking agents and microtiter plates<br />

did not improve the titration curve. Hence, the antigen dilution<br />

with the lowest OD for the negative control and still the biggest<br />

difference between the ODs <strong>of</strong> the negative and positive control<br />

was chosen. Weak positive samples were tested to verify that<br />

they could still be detected.<br />

It is becoming apparent that MAT negative porcine serum<br />

samples can be differentiated from MAT positive samples with the<br />

recombinant protein LipL32.<br />

OD‐value<br />

1,600<br />

1,400<br />

1,200<br />

1,000<br />

0,800<br />

0,600<br />

0,400<br />

0,200<br />

0,000<br />

0 1 2 3<br />

1= positive, 2 = weak positive, 3= negative sera<br />

Figure 1: 27 porcine sera with different MAT titers tested in the<br />

LipL32-ELISA<br />

Discussion & conclusions<br />

Leptospirosis is a livestock disease with a serious impact on<br />

public health. It is important to find a reliable, inexpensive and<br />

hazardous-free diagnostic test. This study’s target is to test the<br />

possible utilisation <strong>of</strong> the recombinant protein LipL32 in an ELISA<br />

for porcine leptospirosis.<br />

LipL32 seems to be a potential candidate for an ELISA. The<br />

serum samples tested until now show different OD values,<br />

depending on having negative or positive MAT titers. There is a<br />

correlation <strong>of</strong> the qualitative results (positive / negative), but there<br />

seems to be no correlation <strong>of</strong> the quantitative results (MAT titers /<br />

OD values). Figure 1 shows that samples with weak positive MAT<br />

titers (1:100 / 1:200) are in the same range <strong>of</strong> OD values as<br />

samples with strong positive MAT titers (≥ 1:800).<br />

The goal <strong>of</strong> the ELISA wasn’t to establish a replicate <strong>of</strong> MAT<br />

titers, but to give reliable results whether an animal has been<br />

exposed to leptospires or not.<br />

The sensitivity and specificity <strong>of</strong> the ELISA have to be examined<br />

in further experiments.<br />

Acknowledgements<br />

The authors thank Dr. K. Nöckler and E. Luge, Federal Institute<br />

for Risk Assessment, Berlin, Germany for assistance with the<br />

MAT.<br />

References<br />

1. Haake,D.A., Chao,G. Zuerner, R.L. Barnett, J.K. Barnett, D., Mazel, M.,<br />

Matsunga, J., Levett, P.N., Bolin, C.A., (2000). The leptospiral major outer<br />

membrane protein LipL32 is a lipoprotein expressed during mammalian<br />

infection. Infect. Immun. 68, 2276-2285<br />

2. Dey,S., Madhan Mohan,C. (2004). Recombinant LipL32 antigen-based<br />

single serum dilution ELISA for detection <strong>of</strong> canine leptospirosis. Vet.<br />

Microbiol.103, 99-106.<br />

3. Bomfim, M.R.Q., Ko, A., Cota Koury, M. (2005). Evaluation <strong>of</strong> the<br />

recombinant LipL32 in enzyme-linked immunosorbent assay for the<br />

serodiagnosis <strong>of</strong> bovine leptospirosis. Vet. Microbiol. 109, 89-94


S4 - O - 02<br />

BIOLOG GENERATION III, MATRIX ASSISTED LASER DESORPTION/IONISATION TIME-OF-FLIGHT<br />

(MALDI-TOF) MASS SPECTROMETRY AND 16S rRNA GENE SEQUENCING FOR THE<br />

IDENTIFICATION OF BACTERIA OF VETERINARY INTEREST<br />

Peter Wragg 1 , Luke Randall 2 , Adrian Whatmore 3<br />

1 Animal Health and Veterinary Laboratories Agency, Laboratory Services Department, Penrith, United Kingdom<br />

2,3 Animal Health and Veterinary Laboratories Agency, Department <strong>of</strong> Bacteriology, Weybridge, United Kingdom<br />

Biolog, Maldi-ToF, 16S rRNA<br />

Introduction<br />

The veterinary bacteriologist is currently presented with a wide<br />

variety <strong>of</strong> technologies <strong>of</strong>fering potential solutions to problems in<br />

the identification <strong>of</strong> bacteria.<br />

16S rRNA sequencing is perhaps the most familiar <strong>of</strong> the genomic<br />

approaches. Nonetheless, genomic methods are not without<br />

limitations and may suffer similar database validation issues to<br />

phenotypic systems (1,2). Automated or semi-automated<br />

metabolic methods have developed rapidly in recent years, with<br />

multi-analyte systems <strong>of</strong>fering extended databases, although<br />

frequently less well populated with veterinary taxa. Biolog’s<br />

Generation III identification system employs a novel method <strong>of</strong><br />

detecting substrate utilisation and an extensive database,<br />

including veterinary isolates (3). Matrix-assisted laser desorption/<br />

ionization time-<strong>of</strong>-flight mass spectrometry (MALDI-TOF) has<br />

become widely recognised as one <strong>of</strong> the most adaptable <strong>of</strong> the<br />

chemotaxonomic methods (4). Veterinary strains are wellrepresented<br />

on manufacturer’s databases. Database<br />

enhancement initiatives are ongoing for all three technologies in a<br />

number <strong>of</strong> research facilities.<br />

This study was designed to assess the performance <strong>of</strong> the above<br />

methods for the identification <strong>of</strong> a limited but typical range <strong>of</strong><br />

referral isolates <strong>of</strong> veterinary significance (n=66) by comparison to<br />

partial 16S rRNA DNA sequencing, with a view to indicating<br />

potential suitability as a ’front-line’ identification tool.<br />

Materials & methods<br />

The isolates used in this study consisted <strong>of</strong> field strains referred<br />

for determinative bacteriology by Regional Laboratories <strong>of</strong> the<br />

Animal Health and Veterinary Laboratories Agency (AHVLA),<br />

although a number <strong>of</strong> strains from the National Culture Type<br />

Collection (NCTC, Colindale, London) and the American Type<br />

Culture Collection (ATCC, Manassas, Virginia) were also included.<br />

Each field isolate had previously been identified by conventional<br />

biochemical approaches. Cultures were prepared on 5% sheep<br />

blood-Columbia agar (Oxoid, Basingstoke, UK), incubated either<br />

aerobically or in 7.5% carbon dioxide at 37 °C for 18-24h,<br />

according to cultural characteristics, and tested blind by each <strong>of</strong><br />

the three methods as follows.<br />

Biolog Generation 3:<br />

Inocula were prepared according to manufacturer’s instructions<br />

(Biolog, Hayward, CA). Growth patterns were measured using a<br />

Microstation reader and analysed using Microlog s<strong>of</strong>tware.<br />

Matrix-assisted laser desorption ionization-time <strong>of</strong> flight mass<br />

spectrometry (MALDI-TOF):<br />

Samples were prepared using standard Bruker protocols (Bruker<br />

Daltonik GmbH, Bremen, Germany) (4) and analysed using a<br />

Bruker Aut<strong>of</strong>lex II machine. Spectra were analysed with MALDI<br />

Biotyper s<strong>of</strong>tware version 2.0.10.0 in comparison with reference<br />

spectra (MALDI Biotyper reference library version 3).<br />

16S rRNA gene sequencing:<br />

Ribosomal RNA sequence identification was based on a minimum<br />

600bp sequence corresponding to the V2-V4 regions <strong>of</strong> the gene.<br />

Sequences were compared with both NCBI and RDP databases<br />

with criteria <strong>of</strong> a >99% similarity to the type strain <strong>of</strong> a<br />

taxonomically valid species being used as identification criteria for<br />

species. If matches were below 99% or there were matches >99%<br />

to multiple taxonomically valid species identification was given to<br />

the genus level only.<br />

In the majority <strong>of</strong> cases, referral for determinative bacteriology<br />

within AHVLA requires identification to species level. A scoring<br />

system was devised, using either 16S sequencing or the<br />

NCTC/ATCC identity as the ‘gold standard’. Where sequencing<br />

lacked species information for comparison, identification was<br />

scored at genus level only in all methods.<br />

Results<br />

Scores <strong>of</strong> 3, 1 and 0 were assigned for identifications to species,<br />

genus and ’ no identification’, respectively.<br />

Table 1 summarises the performance <strong>of</strong> each system.<br />

Table 1: Performance <strong>of</strong> identification systems<br />

Numbers <strong>of</strong> strains identified Biolog MALDI 16S sequencing<br />

To genus 28 30 29<br />

To species 27 27 36<br />

No ID 11 9 1<br />

Incorrect genus 0 0 0<br />

Incorrect species 0 0 0<br />

Overall score 109 111 137<br />

Scores for MALDI and Biolog may have improved if alternative<br />

phenotypic data had been considered in determining true identity.<br />

Discussion & conclusions<br />

Scores for MALDI and Biolog were broadly comparable, although<br />

a number <strong>of</strong> taxa were identified more specifically by particular<br />

techniques. A number <strong>of</strong> isolates proved refractory either by<br />

MALDI-TOF or Biolog, although local enhancements to databases<br />

would minimise these occurrences.<br />

Whilst each methodology <strong>of</strong>fers unique advantages, other factors<br />

which require consideration are the accessibility <strong>of</strong> the technology,<br />

including start-up and running costs, the quality and ease <strong>of</strong><br />

supplementation <strong>of</strong> the manufacturer’s database and the<br />

requirement for centralisation <strong>of</strong> resources and expertise which<br />

may be implicit in the choice <strong>of</strong> technique.<br />

Acknowledgements<br />

We thank the AHVLA Regional Laboratories at Bury St Edmunds,<br />

Starcross and Sutton Bonington for provision <strong>of</strong> field strains, Mark<br />

Koylass and the Central Sequencing Unit at AHVLA Weybridge.<br />

References<br />

1.Janda, J.M., Abbott, S.L. (2007). 16S rRNA gene sequencing for bacterial<br />

identification in the diagnostic laboratory: pluses, perils and pitfalls. J. Clin.<br />

Microbiol. 45: 2761-2764.<br />

2. Woo, P.C., Lau, S.K., Teng, J.L., Tse, H., Yuen, K-Y. (2008) Then and<br />

now: use <strong>of</strong> 16S rDNA gene sequencing for bacterial identification and<br />

discovery <strong>of</strong> novel bacteria in clinical microbiology laboratories. Clin.<br />

Microbiol. Infect., 14: 908-934.<br />

3. Morgan, M.C., Boyette, M., G<strong>of</strong>orth, C., Sperry, K. V., Greene, S. R.,<br />

(2009). Comparison <strong>of</strong> the Biolog Omnilog Identification System and 16S<br />

ribosomal RNA gene sequencing for accuracy in identification <strong>of</strong> atypical<br />

bacteria <strong>of</strong> clinical origin. J. Microbiol. Meth. 79: 336-343.<br />

4. Eigner, U., Holfelder, M., Oberdorfer, K. et al. (2009) Performance <strong>of</strong> a<br />

matrix-assisted laser desorption ionization-time-<strong>of</strong>-flight mass spectrometry<br />

system for the identification <strong>of</strong> bacterial isolates in the clinical routine<br />

laboratory. Clin. Lab., 55: 289-296.


S4 - O - 03<br />

EVALUATION OF THE DIAGNOSTIC PERFORMANCE OF PEPTIDE COCKTAILS IN THE INTERFERON<br />

GAMMA ASSAY FOR DIAGNOSIS OF TUBERCULOSIS IN CATTLE<br />

Björn Schröder 1 , Roland Hardegger 1 , Marcus G. Doherr 2 , Jean-Louis Moyen 3 , Eamonn Gormley 4 and<br />

Alex J. Raeber 1<br />

1 Prionics AG, Schlieren, Switzerland, 2 Vetsuisse Faculty, University <strong>of</strong> Bern, Switzerland, 3 Laboratoire Départemental d’Analyse et de Recherche,<br />

Coulounieix-Chamiers, France, 4 University College Dublin, Ireland,<br />

Tuberculosis, Bovigam, defined antigens, peptide cocktail<br />

Introduction<br />

Cattle infected with bovine tuberculosis (bTB) still represent a<br />

serious regulatory and health concern in a many countries. For<br />

more than two decades, eradication programs for bTB have<br />

relied on early detection and removal <strong>of</strong> infected cattle using the<br />

intradermal skin test as primary test and the in vitro interferon<br />

gamma (IFN-) assay as an ancillary assay. The interferon<br />

gamma assay is generally considered to be more sensitive than<br />

the skin test while the specificity is slightly lower. To increase the<br />

specificity <strong>of</strong> the IFN- assay, defined mycobacterial antigens<br />

such as ESAT-6 and CFP-10 have been evaluated as alternative<br />

stimulation antigens in blood cultures and have shown promising<br />

results although sensitivities were slightly lower than with<br />

tuberculin stimulation. Combining additional specific<br />

mycobacterial antigens with ESAT-6 and CFP-10 has been<br />

proposed to bring the sensitivity <strong>of</strong> antigen cocktails to the levels<br />

obtained with tuberculin and at the same time achieving<br />

specificities comparable to the skin test. We have developed<br />

peptide cocktails based on ESAT-6 and CFP-10 (1, 2, 3) and<br />

compared their performance to tuberculins in the IFN- assay for<br />

diagnosis <strong>of</strong> bTB in field studies in Ireland and France.<br />

Materials & methods<br />

Lelystad tuberculin PPD antigens (avian and bovine) (Prionics,<br />

The Netherlands) or a synthetic peptide cocktail were used for<br />

stimulation <strong>of</strong> whole blood cultures. Following stimulation, plasma<br />

was harvested and IFN-γ was measured with the BOVIGAM ® 2G<br />

(Prionics, Switzerland) sandwich enzyme immunoassay. The<br />

peptide cocktail Prionics ® PC-HP (Prionics, Switzerland)<br />

contained overlapping peptides derived from ESAT-6 and CFP-<br />

10 and in addition peptides derived from Rv3615c and 3 other<br />

mycobacterial proteins. Diagnostic sensitivity was assessed in<br />

naturally bTB-exposed animals in Ireland. Diagnostic specificity<br />

was evaluated in France in animals from 12 herds free <strong>of</strong> bTB<br />

according to OIE criteria. Estimates for the test characteristics in<br />

the absence <strong>of</strong> a true gold standard were derived by a Bayesian<br />

model as described by Branscum et al. 2005 (4).<br />

Results<br />

Estimates <strong>of</strong> sensitivity and specificity (median and 95%<br />

confidence intervals) are shown in Table 1. In France 390<br />

animals have been included into the specificity trial. Specificity<br />

estimates for the BOVIGAM ®<br />

2G with tuberculin PPD from<br />

Lelystad was calculated with 84% (95%CI: 81% - 87%) whereas<br />

using the peptide cocktail PC-HP for whole blood stimulation a<br />

specificity estimate <strong>of</strong> 94% (95%CI: 92% - 96%) was computed.<br />

The difference in the specificity estimates <strong>of</strong> PPD and PC-HP<br />

was highly significant (p < 0.05; two-tailed exact Fisher test).<br />

Sensitivity was evaluated from the sample <strong>of</strong> 201 animals from<br />

Ireland, with an apparent prevalence (depending on test)<br />

between 16% and 31%. Sensitivity in this trial was calculated for<br />

Lelystad PPD with 89% (95%CI: 75% - 97%) whereas with PC-<br />

HP a comparable sensitivity <strong>of</strong> 85% (95%CI: 63% - 96%) was<br />

obtained.<br />

Table 1: Comparison <strong>of</strong> diagnostic performance estimates <strong>of</strong><br />

BOVIGAM ® 2G using PPD or antigen peptide cocktail PC-HP for<br />

stimulation <strong>of</strong> blood.<br />

Sensitivity<br />

Estimates<br />

95% CI<br />

Specificity<br />

Estimates<br />

95% CI<br />

PPD 89% 75 - 97% 84% 81 - 87%<br />

PC-HP 85% 64 - 96% 94% 92 - 96%<br />

Discussion & conclusions<br />

Since 1991 when the BOVIGAM ® IFN- test was approved as an<br />

<strong>of</strong>ficial test for bTB in Australia and later on in the European<br />

Union and the US, numerous field trials were undertaken to<br />

determine its diagnostic performance. In a review published by<br />

de la Rua-Domenech et al. (5) the sensitivity <strong>of</strong> the IFN- test<br />

varied between 73.0% and 100%, with a median value <strong>of</strong> 87.6%<br />

while its median specificity was calculated with 96.6%, with a<br />

range <strong>of</strong> 85.0–99.6%. Using a Bayesian modelling approach, we<br />

have compared the diagnostic performance <strong>of</strong> the peptide<br />

cocktail Prionics ® PC-HP with tuberculin PPD for the stimulation<br />

<strong>of</strong> blood cultures in the BOVIGAM ® 2G IFN- assay. Our results<br />

suggest that the sensitivity <strong>of</strong> the peptide cocktail PC-HP is not<br />

significantly different from the sensitivity with PPD whereas<br />

peptide cocktail PC-HP resulted in a specificity that was<br />

significantly higher than stimulation with PPD. Overall, we<br />

conclude from these studies that peptide cocktails provide<br />

superior performance compared to tuberculin PPD in the<br />

BOVIGAM ® 2G assay which opens up new possibilities for highly<br />

specific diagnostic tools in the eradication <strong>of</strong> bTB.<br />

References<br />

1. Buddle, BM, Ryan, TJ, Pollock, JM, Andersen, JM, and de Lisle, GW<br />

(2001). Use <strong>of</strong> ESAT-6 in the interferon- test for diagnosis <strong>of</strong> bovine<br />

tuberculosis following skin testing. Vet Microbiol, 80, 37-46<br />

2. Sidders, B, Pirson, C, Hogarth, PJ, Hewinson, RG, Stoker, NG, and<br />

Vordermeier, HM (2008). Screening <strong>of</strong> highly expressed mycobacterial<br />

genes identifies Rv3615c as a useful differential diagnostic antigen for the<br />

Mycobacterium tuberculosis complex. Infect Immun, 76, 3932-3939<br />

3. Schiller, I, Vordermeier, HM, Waters, WR, Palmer, M, Thacker, T,<br />

Whelan, A, Hardegger, R, Marg-Haufe, B, Raeber, A and Oesch, B (2009).<br />

Assessment <strong>of</strong> Mycobacterium tuberculosis OmpATb as a novel antigen<br />

for the diagnosis <strong>of</strong> bovine tuberculosis. Clin Vaccine Immunol, 16,1314-21<br />

4. Branscum, AJ, Gardner, IA and Johnson WO (2005). Estimation <strong>of</strong><br />

diagnostic-test sensitivity and specificity through Bayesian modeling. Prev<br />

Vet Med 68, 145-163<br />

5. De la Rua-Domenech, R, Goodchild, A.T., Vordermeier, H.M.,<br />

Hewinson, R.G., Christiansen, K.H., Clifton-Hadley, R.S. (2006) Ante<br />

mortem diagnosis <strong>of</strong> tuberculosis in cattle: a review <strong>of</strong> the tuberculin tests,<br />

gamma-interferon assay and other ancillary diagnostic techniques." Res<br />

Vet Sci 81: 190-210


S4 - O - 04<br />

MATRIX-ASSISTED LASER DESORPTION IONIZATION-TIME OF FLIGHT MASS SPECTROMETRY<br />

(MALDI-TOF MS) IN A VETERINARY DIAGNOSTIC LABORATORY<br />

Annet E. Heuvelink, Refke Peerboom, Erik van Engelen, Abdul A. Hassan<br />

GD Animal Health Service, Deventer, The Netherlands<br />

MALDI-TOF, bacterial identification, veterinary laboratory<br />

Introduction<br />

Matrix-Assisted Laser Desorption Ionization-Time <strong>of</strong> Flight Mass<br />

Spectrometry (MALDI-TOF MS) has emerged as a new tool for<br />

fast and reliable identification <strong>of</strong> microorganisms (). We evaluated<br />

the performance <strong>of</strong> MALDI-TOF MS for the routine identification<br />

<strong>of</strong> bacteria in our veterinary diagnostic laboratory.<br />

Materials & methods<br />

Mass spectra were acquired using a Micr<strong>of</strong>lex LT mass<br />

spectrometer and analysed by MALDI Biotyper 3.0 s<strong>of</strong>tware<br />

(Bruker Daltonik GmbH, Bremen, Germany).<br />

First, we determined the accuracy <strong>of</strong> MALDI-TOF MS<br />

identification by testing 66 bacterial strains <strong>of</strong> culture collections,<br />

in duplicate spots on the MALDI target plate. Second, we<br />

performed a prospective validation study, including 262 isolates<br />

collected in the routine laboratory: 156 isolates from post-mortem<br />

examinations <strong>of</strong> mammals and 106 mastitis pathogens from<br />

cow’s milk. The isolates were identified by conventional<br />

biochemical methods in parallel with MALDI-TOF MS, measured<br />

in duplicate spots. A selection <strong>of</strong> Gram-positive cocci, including<br />

coagulase-negative staphylococci (CNS) and Streptococcus<br />

uberis isolates, were tested by both direct colony testing and the<br />

extended direct transfer method described by the manufacturer,<br />

comprising a rapid partial extraction with 70% aqueous formic<br />

acid. Finally, the repeatability and the robustness <strong>of</strong> identification<br />

<strong>of</strong> bacteria by MALDI-TOF MS were evaluated. The repeatability<br />

was determined with five reference strains, by testing 10 cfu from<br />

one plate, in one run, by one technician. The robustness was<br />

evaluated by testing the effect <strong>of</strong> (a) refrigerated storage <strong>of</strong> fresh<br />

cultures grown on agar plates (for 24 or 48 h), (b) the use <strong>of</strong><br />

selective solid media, and (c) prolonged incubation (for 48 or 72<br />

h) either or not followed by a refrigeration step. For each <strong>of</strong> the<br />

species included in the robustness test, one reference strain and<br />

five field isolates were tested.<br />

some isolates, especially those <strong>of</strong> S. uberis, more not reliable<br />

identifications were obtained when performed with cultures stored<br />

for 48 h in the refrigerator or incubated for 48 h, and especially<br />

for 72 h prior to identification.<br />

Discussion & conclusions<br />

The present results show that MALDI-TOF MS is a reliable and<br />

rapid method for identification <strong>of</strong> most bacterial species<br />

encountered in our routine veterinary laboratory. No<br />

misidentifications occurred, only “no reliable identifications”. For<br />

the majority <strong>of</strong> the isolates direct colony testing can be performed<br />

without extraction. For identification <strong>of</strong> presumptive CNS isolates,<br />

it is recommended to use the simple partial extraction procedure<br />

with 70% aqueous formic acid following the instructions <strong>of</strong> the<br />

manufacturer.<br />

One <strong>of</strong> the additional benefits <strong>of</strong> MALDI-TOF MS analysis is the<br />

identification <strong>of</strong> CNS species. Species level identification <strong>of</strong> CNS<br />

will help to elucidate sources, transmission mechanisms and the<br />

impact <strong>of</strong> different CNS species on cow health, productivity and<br />

milk quality, and this knowledge may lead to species-specific<br />

infection control measures.<br />

References<br />

1. Bizzini A, Greub G. Matrix-assisted laser desorption ionization<br />

time-<strong>of</strong>-flight mass spectrometry, a revolution in clinical microbial<br />

identification. Clin. Microbiol. Infect. 2010, 16(11), 1614-1619.<br />

2. Wieser A, Schneider L, Jung J, Schubert S. MALDI-TOF MS<br />

in microbiological diagnostics-identification <strong>of</strong> microorganisms<br />

and beyond (mini review). Appl. Microbiol. Biotechnol. <strong>2012</strong>,<br />

93(3), 965-974.<br />

Results<br />

Of the 66 reference strains tested, 79% were correctly identified<br />

to species level. The remaining strains were identified to genus<br />

level (14%) or could not be identified (7%). The latter strains<br />

belonged to species not included in the BioTyper 3.0 database.<br />

For 90% <strong>of</strong> the 156 isolates from post-mortem examinations <strong>of</strong><br />

mammals matching identifications at species level were obtained<br />

between MALDI-TOF MS and biochemical methods, 8% were<br />

correctly identified to genus level, and for 2% no reliable<br />

identifications were achieved. Eighty-nine% <strong>of</strong> the MALDI-TOF<br />

MS identifications <strong>of</strong> 106 mastitis pathogens were concordant<br />

with standard biochemical identifications. Ten% <strong>of</strong> the mastitis<br />

pathogens were identified to species level by MALDI-TOF MS<br />

(mainly CNS), while standard biochemical identifications <strong>of</strong> these<br />

isolates were at genus level only. For one isolate the MALDI-TOF<br />

MS result diverged from the biochemical identification; MALDI-<br />

TOF MS identified the isolate as Klebsiella pneumoniae, a<br />

qualitatively good result at species level, while the API 20E<br />

(bioMérieux, Marcy l’Etoile, France) result was Klebsiella<br />

oxytoca, with a doubtful pr<strong>of</strong>ile. Most bacteria tested in this study<br />

could be successfully identified at species level using the direct<br />

colony testing method. However, for 70% <strong>of</strong> the CNS isolates the<br />

partial extraction procedure showed to be superior. For S. uberis<br />

the superiority <strong>of</strong> the extended direct transfer method appeared<br />

to be less pronounced, being observed for only 32% <strong>of</strong> the<br />

isolates.<br />

The repeatability <strong>of</strong> MALDI-TOF MS identification <strong>of</strong> the<br />

reference strains <strong>of</strong> Escherichia coli, Clostridium perfringens,<br />

Staphylococcus aureus, and Salmonella Typhimurium was 100%.<br />

For the Streptococcus suis reference strain it was 90%, with 10%<br />

being not reliably identified.<br />

In general, MALDI-TOF MS identification results were not altered<br />

when performed with cultures stored for 24 or 48 h at<br />

refrigeration temperatures or with cultures tested after 48 or 72 h<br />

<strong>of</strong> incubation, nor when selective agars were used. However, for


S4 - O - 05<br />

RAPID IDENTIFICATION OF BOVINE MASTITIS PATHOGENS USING MALDI TOF<br />

Gudrun Overesch 1 , Andreas Thomann 1 , Vincent Perreten 1<br />

1<br />

Institute <strong>of</strong> Veterinary Bacteriology, University <strong>of</strong> Bern, Bern, Switzerland<br />

Maldi T<strong>of</strong>, mastitis, identification, veterinary pathogens<br />

Introduction<br />

Correct and rapid identification <strong>of</strong> pathogens is the main objective<br />

<strong>of</strong> bacteriological diagnostic. Phenotypical identification<br />

procedures using biochemical parameters have been widely used<br />

in the last decades. Although automatic systems were developed,<br />

these methods are still time consuming. Therefore molecular<br />

identification methods, i. e. real time PCR, were established to<br />

replace biochemical approach when applicable. The exclusive<br />

use <strong>of</strong> molecular based diagnostic does not allow performing<br />

antibiotic susceptibility tests. The recently established matrixassisted<br />

laser desorption/ionization time-<strong>of</strong>-flight mass<br />

spectroscopy (MALDI TOF MS) approach overcomes the<br />

disadvantages <strong>of</strong> the methods mentioned above. It is faster than<br />

PCR and, while working with cultured strains, it is possible to<br />

perform antimicrobial susceptibility testing. The databases <strong>of</strong><br />

MALDI TOF MS are usually based on spectra generated from<br />

human isolates [1]. Its use in veterinary bacteriology needs<br />

validation with livestock derived strains and update <strong>of</strong> the<br />

databases with spectra from relevant pathogens <strong>of</strong> veterinary<br />

interest. This study presents the establishment <strong>of</strong> MALDI TOF<br />

MS technology for the direct identification to the species level <strong>of</strong><br />

relevant bovine mastitis pathogens.<br />

Materials & methods<br />

156 bovine mastitis milk samples from individual cows were<br />

included. Ten microliter <strong>of</strong> whole milk was cultured on trypticase<br />

soy agar plates with 5% sheep blood (Becton Dickinson) at 37 °C<br />

for 24 hours under aerobic conditions. Colonies were identified<br />

phenotypically by routine diagnostic procedures [2] [3]. In parallel,<br />

the same colonies were identified by MALDI TOF MS (Biotyper<br />

3.0, Bruker) using the direct transfer protocol recommended by<br />

the manufacturer. Brieflly, material from a single colony was<br />

taken with a toothpick, smeared on a steel plate in dublicates and<br />

overlaid with 1 μl <strong>of</strong> HCCA matrix solution. After air drying, the<br />

samples were measured using standard settings in the “Flex<br />

control” s<strong>of</strong>tware. For calibration and as an internal reference the<br />

“bacterial test standard” (BTS, Bruker) was applied in triplicate on<br />

every plate measured. Analysis <strong>of</strong> spectra with the Biotyper 3.0<br />

included a comparison against the internal commercial database<br />

in combination with the institute database. The latter was<br />

generated using veterinary field strains, which include coagulasenegative<br />

staphylococci (Table 1), S. pseudintermedius,<br />

Aerococcus viridans and Streptococcus uberis. All strains were<br />

identified with VITEK 2 (Biomérieux) and 16SrDNA sequence.<br />

New pr<strong>of</strong>iles were introduced into MALDI TOF MS database.<br />

Identification was accepted when a score value > 2.000 was<br />

achieved. All identified strains isolated from mastitis milk included<br />

in this study are listed in Table 1.<br />

Results<br />

After updating the commercial database with veterinary field<br />

strains (i. e. coagulase-negative staphylococci and. S. uberis)<br />

155 out <strong>of</strong> 156 biochemical defined strains were identified from a<br />

24 hour direct plating culture by MALDI TOF MS analyses (Table<br />

1). These strains include all highly relevant mastitis pathogens,<br />

i. e. staphylococci and streptococci species. MALDI TOF MS<br />

identified all alpha- double- and non-hemolytic Staphylococcus<br />

aureus. Trueperella (Arcanobacterium) pyogenes, Histophilus<br />

somni, E. coli and Klebsiella oxytoca were also clearly identified.<br />

Members <strong>of</strong> the Streptococcus-, Lactococcus-, Enterococcusgroup,<br />

which cannot be easily characterized using classical<br />

methods could also be identified to the species level using<br />

MALDI TOF MS. Only Corynebacterium bovis could not be<br />

directly identified by MALDI TOF MS, since this species has not<br />

yet been introduced in the new database.<br />

Discussion & conclusion<br />

MALDI-TOF MS was shown to represent an excellent method for<br />

rapid species identification <strong>of</strong> relevant bovine mastitis pathogens<br />

by direct analysis <strong>of</strong> overnight cultures. However, the commercial<br />

database needed to be expanded with pr<strong>of</strong>iles <strong>of</strong> important<br />

bovine mastitis pathogens. Further improvement such has<br />

extraction prior identification should also be engaged to increase<br />

the identification spectrum. A more detailed identification <strong>of</strong><br />

bacteria present in mastitis milk will also allow to increase the<br />

knowledge regarding species diversity involved in mastitis.<br />

Table 1: Biochemical identification and Maldi T<strong>of</strong> identification <strong>of</strong><br />

bacterial strains isolated from mastitis milk<br />

Isolates<br />

(N)<br />

Biochemical<br />

identification<br />

Maldi T<strong>of</strong> identification<br />

(number)<br />

39 Streptococcus uberis Streptococcus uberis<br />

39<br />

16<br />

15<br />

14<br />

9<br />

Non haemolytic<br />

staphylococci<br />

Double haemolytic<br />

S. aureus<br />

Streptococcus-,<br />

Lactococcus-,<br />

Enterococcus-group<br />

Alpha haemolytic<br />

staphylococci<br />

Streptococcus<br />

dysgalactiae subsp.<br />

dysgalactiae<br />

8 Enterococcus spp.<br />

5<br />

Trueperella<br />

(Arcanobacterium)<br />

pyogenes<br />

S*. xylosus (19)<br />

S. chromogenes (7)<br />

S. sciuri (4)<br />

S. epidermidis (2)<br />

S. vitulinus (2)<br />

S. equorum (2)<br />

S. warneri (1)<br />

S. devriesei (1)<br />

S. aureus (1)<br />

S. aureus (16)<br />

Lactococcus garvieae (9)<br />

Aerococcus viridans (3)<br />

Lactococcus lactis (1)<br />

Enterococcus devriesei (1)<br />

Streptococcus uberis (1)<br />

S. aureus (6)<br />

S. hemolyticus (5)<br />

S. xylosus (3)<br />

Streptococcus dysgalactiae<br />

subsp. dysgalactiae<br />

Enterococcus faecalis (4)<br />

Aerococcus viridans (2)<br />

Streptococcus equinus (1)<br />

Enterococcus faecium (1)<br />

Trueperella<br />

(Arcanobacterium) pyogenes<br />

4 Escherichia coli Escherichia coli<br />

2<br />

Streptococcus<br />

agalactiae<br />

Streptococcus agalactiae<br />

1 Histophilus somni Histophilus somni<br />

1 Klebsiella oxytoca Klebsiella oxytoca<br />

*S. = Staphylococcus<br />

Acknowledgments<br />

This study was partially financed by grant 1.11.21 <strong>of</strong> the Federal<br />

Veterinary Office (FVO), Switzerland.<br />

References<br />

1. Seng P, Drancourt M, Gouriet F, La Scola B, Fournier PE, Rolain JM, et<br />

al. Ongoing revolution in bacteriology: routine identification <strong>of</strong> bacteria by<br />

matrix-assisted laser desorption ionization time-<strong>of</strong>-flight mass<br />

spectrometry. Clin Infect Dis 2009;49(4):543-551.<br />

2. Guélat-Brechbuehl M, Thomann A, Albini S, Moret-Stalder S, Reist M,<br />

Bodmer M, et al. Cross-sectional study <strong>of</strong> Streptococcus species in<br />

quarter milk samples <strong>of</strong> dairy cows in the canton <strong>of</strong> Bern, Switzerland. Vet<br />

Rec 2010;167(6):211-215.<br />

3. Moret-Stalder S, Fournier C, Miserez R, Albini S, Doherr MG, Reist M,<br />

et al. Prevalence study <strong>of</strong> Staphylococcus aureus in quarter milk samples<br />

<strong>of</strong> dairy cows in the Canton <strong>of</strong> Bern, Switzerland. Prev Vet Med<br />

2009;88(1):72-76.


S4 - O - 06<br />

15 MINUTE ELISA USING A LOW COST COMMERCIAL BIOSENSOR<br />

Jason Sawyer 1 , Jennifer Cork 1 , Rebecca Jones 1<br />

1<br />

AHVLA (Weybridge), Technology Transfer Unit, New Haw, Addlestone, Surrey KT15 3NB, UK<br />

ELISA, IBR, Biosensor, Penside testing, Serology, Vantix<br />

Introduction<br />

The use <strong>of</strong> biosensors for diagnostic testing <strong>of</strong>fers several<br />

potential advantages, but to date their use for routine diagnostic<br />

testing has been limited to specific markets (e.g. glucose testing).<br />

We evaluated the use <strong>of</strong> a commercially available potentiometric<br />

biosensor platform - Vantix - to carry out a serological assay<br />

for antibodies to infectious bovine rhinotracheitis (IBR), caused<br />

by Bovine Herpes Virus 1 (BoHV-1).<br />

The Vantix system 1<br />

comprises a low cost reader unit and<br />

disposable potentiometric biosensors. The biosensors consist <strong>of</strong><br />

a working and reference electrode coated in a conductive<br />

polymer (polypyrrole) and covered in a protective plastic film.<br />

Biochemical or enzymatic activity taking place on the surface <strong>of</strong><br />

the working electrode as a result <strong>of</strong> immunocomplexes built up on<br />

the electrode, cause electrochemical changes in the conductive<br />

polymer layer on and around the working electrode thus<br />

generating a measurable change in electrical potential (measured<br />

in millivolts) relative to the reference electrode. The signal<br />

produced is proportional to the level <strong>of</strong> analyte under<br />

investigation.<br />

IBR is a highly contagious disease <strong>of</strong> cattle that is endemic in the<br />

UK and many other parts <strong>of</strong> the world 2 . ELISA is commonly used<br />

in the detection and control <strong>of</strong> this disease and AHVLA uses an<br />

in-house indirect ELISA. In this assay, BoHV-1 antigens, bound<br />

to the surface <strong>of</strong> a microtitre plate, capture BoHV-1 specific<br />

antibodies present in test samples. Bound antibodies are then<br />

detected using a horseradish peroxidase (HRP) labelled antibovine<br />

immunoglobulin and subsequent HRP catalysed oxidation<br />

<strong>of</strong> the chromogen substrate 3,3’,5,5’-tetramethylbenzidine (TMB)<br />

produces a colour change. The in-house ELISA for BoHV-1 used<br />

at AHVLA has a total assay time <strong>of</strong> over 3 hours which is typical<br />

<strong>of</strong> commercial ELISA tests.<br />

In this work, the well-characterised and established reagents<br />

used in the parent ELISA were used to construct an assay on the<br />

Vantix platform. A panel <strong>of</strong> serum samples, submitted for<br />

routine ELISA testing, were then tested with the Vantix assay and<br />

the results compared to the parent ELISA.<br />

Table 1: Comparison <strong>of</strong> methodology <strong>of</strong> the IBR antibody biosensor<br />

assay and parent ELISA.<br />

Biosensor assay<br />

Indirect ELISA<br />

Step<br />

Incubation<br />

Incubation<br />

Step<br />

Time<br />

Time<br />

Biosensors and ELISA plates pre coated with BoHV1 antigen<br />

and stored ready for use<br />

Incubation with diluted<br />

Incubation with diluted<br />

5 min<br />

sample<br />

sample<br />

2 h<br />

8 Washes no soak 0.5 min<br />

5 x washes 10 second<br />

soak<br />

5 min<br />

Incubation with diluted<br />

Rabbit anti bovine HRP 5 min<br />

Incubation with diluted<br />

Rabbit anti bovine HRP 1 h<br />

Conjugate<br />

Conjugate<br />

8 Washes no soak 0.5 min<br />

5 x washes 10 second<br />

soak<br />

5 min<br />

Probes added to TMB<br />

Plate incubated with<br />

5 – 20 min<br />

substrate and read<br />

TMB substrate.<br />

4 min<br />

immediately. Potential<br />

Addition <strong>of</strong> Stop 1 min<br />

difference reading.<br />

Colorimetric read 1 min<br />

Total assay time 15 min Total assay time >3.25 h<br />

Materials & methods<br />

Table 1 summarises the steps and timings involved in the<br />

performing the Biosensor assay compared to the ELISA.<br />

Biosensor probes were prepared by coating with BoHV-1 antigen,<br />

and were then stored at +4 o C for up to 12 weeks. To perform the<br />

assay, the electrodes <strong>of</strong> the antigen coated biosensors were<br />

briefly placed in diluted serum, washed, and briefly incubated<br />

with conjugate. After a final wash step, the sensors were<br />

inserted into the Vantix reader unit and the working and<br />

reference electrode placed into TMB substrate. Changes in<br />

electrochemical potential difference were observed over a period<br />

<strong>of</strong> four minutes and recorded using Vantix s<strong>of</strong>tware. The<br />

biosensor assay was used to test 194 serum samples,<br />

comprising 90 positive and 104 negative as designated by the in<br />

house ELISA.<br />

% signal relative to weak<br />

positive control<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

IBR Biosensor<br />

2 x 2 box analysis assay<br />

Serum Samples Positive Negative Totals<br />

IBR<br />

ELISA<br />

Negative<br />

Positive 86 4 90<br />

Negative 2 102 104<br />

Totals 88 104 194<br />

Positive<br />

True positive rate<br />

(Sensitivity)<br />

0<br />

0 25 50 75 100<br />

False positive rate<br />

(100 - Specificity)<br />

Fig.1 Comparison <strong>of</strong> results obtained when testing serum<br />

samples using the parent IBR antibody ELISA and the biosensor<br />

assay. 2x2 table (Top); Box and whisker plot showing biosensor<br />

results for ELISA negative and positive samples (bottom left) and<br />

ROC analysis (bottom right).<br />

Results<br />

Fig 1 shows the resulting 2X2 Table, Box and Whisker plot and<br />

ROC curve when the biosensor assay was compared to the<br />

ELISA. Using the optimal cut-<strong>of</strong>f value (determined by ROC<br />

analysis and based on a percentage signal relative to the weak<br />

positive control serum run alongside the test samples) the<br />

biosensor assay had a sensitivity <strong>of</strong> 98% and a specificity <strong>of</strong> 96%<br />

when compared to the indirect ELISA. Only 6 <strong>of</strong> 194 samples<br />

resulting in a different categorisation (positive or negative)<br />

compared to the ELISA results. Overall, the results demonstrate<br />

that the biosensor assay produces test results that closely match<br />

those obtained with the parent ELISA.<br />

Discussion & conclusion<br />

This work has shown that results equivalent to the ELISA can be<br />

obtained using the Vantix biosensor platform in 15 min. The<br />

relatively low cost <strong>of</strong> the biosensors (~ few Euro each) and reader<br />

(~ few thousand euro) make this an interesting and promising<br />

technology. The rapid and simple nature <strong>of</strong> the assays, and the<br />

fact that established ELISA reagents can readily be converted to<br />

the platform, suggest Vantix could be useful in a variety <strong>of</strong><br />

diagnostic testing scenarios. These may include rapid testing <strong>of</strong><br />

samples which require fast turnaround or testing in low tech<br />

laboratories. The generation <strong>of</strong> an electronic signal <strong>of</strong>fers the<br />

potential for transmission across mobile networks. Vantix are<br />

currently developing a hand held device which uses the same<br />

biosensors but <strong>of</strong>fers the potential for more rapid (


Poster presentations<br />

“General Session”<br />

(1 st session)


S1 - P - 01<br />

LAWSONIA INTRACELLULARIS IN BLUE FOXES IN FINLAND. A CASE REPORT<br />

Heli Kallio, Heikki Ahola<br />

Finnish Food Safety Authority Evira, Production Animal and Wildlife Health Research Unit, Seinäjoki, Finland<br />

Lawsonia intracellularis, blue fox, enteritis, diarrhoea, rectum prolapse<br />

Introduction<br />

Lawsonia intracellularis-bacteria is commonly detected in the<br />

enteritis in domestic pigs with proliferative enteropathy, intestinal<br />

adenomatosis and ileitis. In addition to pigs, Lawsonia<br />

intracellularis has been detected in wide variety <strong>of</strong> domestic and<br />

wild animals, including wild red foxes, and it is also suggested to<br />

be found in blue foxes reared in a fur farm. However,<br />

pathogenesis <strong>of</strong> Lawsonia intracellularis enteritis in fur farm<br />

animals is poorly studied. In this study we detected Lawsonia<br />

intracellularis for the first time in enteritis from blue fox in a<br />

Finnish fur farm.<br />

A 1 2 3 4<br />

Materials & methods<br />

A blue fox was submitted to laboratory for diagnostic because <strong>of</strong><br />

occurrence <strong>of</strong> diarrhoea in a single farm. Symptoms <strong>of</strong> the foxes<br />

were, in addition to diarrhoea with loose faeces, decreased<br />

weight gain, and rectal prolapses.<br />

Necropsy was performed for the fox. Then intestinal samples<br />

were prepared for HE- and WS staining. Nested-PCR for the<br />

epithelium <strong>of</strong> the intestine was performed as earlier described by<br />

Jones et al. to detect Lawsonia intracellularis -bacteria. Prior to<br />

PCR DNA was extracted by boiling procedure according to<br />

Moeller et al. Intestinal samples were cultured on sheep blood<br />

agar in aerobic and anaerobic atmosphere, as well as plated onto<br />

a Campylobacter blood-free selective medium. The Sheather's<br />

sugar flotation technique was used to detect parasites.<br />

Results<br />

In necropsy rectal prolapse, severe hyperemia, oedema and<br />

thickening <strong>of</strong> mucosa in the distal ileum, colon and rectum were<br />

detected. Signs <strong>of</strong> poor growth and loss <strong>of</strong> fluid were also seen.<br />

In histology the fox had proliferative enteritis, colitis and proctitis<br />

with severe villushyperemia and –necrosis, and hypertrophic<br />

dilated crypts with vigorously branching structure. By WS-staining<br />

curved organisms, in the apical cytoplasm <strong>of</strong> hyperplastic<br />

epithelium lining intestinal glands, were detected. Using nested-<br />

PCR, Lawsonia intracellularis was discovered from the intestinal<br />

samples. No other putative pathogens were identified in<br />

bacteriological and parasitological studies.<br />

Figure 2. Amplification <strong>of</strong> Lawsonia intracellularis after nested-<br />

PCR analysis from intestine. Negative amplification control (1),<br />

positive amplification control (2), two separate samples from the<br />

epithelium <strong>of</strong> the intestine (3, 4). DNA ladder, 100-1000 bp (A)<br />

Discussion & conclusions<br />

The results obtained by this study show the Lawsonia<br />

intracellularis caused diarrhoea for the first time in Finnish blue<br />

fox farm. The fox examined showed typical changes in necropsy<br />

and histology. Lawsonia intracellularis was detected by PCR and<br />

WS-staining. Eriksen et al. reported earlier adenomatosis in blue<br />

foxes reared in a fur farm, most likely caused by Lawsonia<br />

intracellularis. Lawsonia intracellularis has been detected also<br />

from faeces <strong>of</strong> wild red foxes. However, according to our<br />

knowledge, other reports about Lawsonia intracellularis in foxes<br />

have not been published. Significance <strong>of</strong> Lawsonia intracellularis<br />

for fox farming is still poorly understood and further studies are<br />

required.<br />

References<br />

1. Tomanova,K Literak,I, Klimes,J Pavlacik,L, Mrlik,V and Smola,J (<br />

2003). Lawsonia intracellularis in Wild Mammals in the Slovak<br />

Carpathians. Journal <strong>of</strong> Wildlife Diseases, 39(2), 407-411<br />

2. Eriksen,K, Landsverk,K.T and Bratberg,B. (1990). Morphology and<br />

immunoperoxidase studies <strong>of</strong> intestinal adenomatosis in the blue fox,<br />

Alopex lagopus. Journal <strong>of</strong> Comparative Pathology, 102, 265-278<br />

3. Jones,GF, Ward,GE, Murtaugh,MP, Lin G, Gebhart,CJ (1993)<br />

Enhanced detection <strong>of</strong> intracellular organism <strong>of</strong> swine proliferative enteritis,<br />

ileal symbiont intracellularis, in feces by polymerase chain reaction. J Clin<br />

Microbiol, 31(10), 2611-5<br />

4. Møller,K, Jensen,TK, Jorsal,SE, Leser,TD, Carstensen,B (1998)<br />

Detection <strong>of</strong> Lawsonia intracellularis, Serpulina hyodysenteriae, weakly<br />

beta-haemolytic intestinal spirochaetes, Salmonella enterica, and<br />

haemolytic Escherichia coli from swine herds with and without diarrhoea<br />

among growing pigs. Vet Microbiol, 30;62(1), 59-72<br />

Figure 1. Curved organisms in the apical cytoplasm <strong>of</strong><br />

hyperplastic epithelium lining intestinal glands stain positively in<br />

WS-staining.


S1 - P - 02<br />

DIAGNOSTIC PERFORMANCE OF A COMMERCIAL PRRS SERUM ANTIBODY ELISA ADAPTED TO<br />

ORAL FLUID: LONGITUDINAL RESPONSE IN EXPERIMENTALLY-INOCULATED POPULATIONS<br />

Apisit Kittawornrat 1 , John Prickett 1 , Chong Wang 1, 2 , Chris Olsen 1 , Yaowalak Panyasing 1 , Andrea Ballagi 3 ,<br />

Anna Rice 3 , Sergio Lizano 3 , Raymond Rowland 4 , Jeffrey Zimmerman 1<br />

1 Department <strong>of</strong> Veterinary Diagnostic and Production Animal Medicine, 2 Department <strong>of</strong> Statistics, Iowa State University, Ames, Iowa, United States, 3 IDEXX<br />

Laboratories, Westbrook, Maine, United States, 4 Department <strong>of</strong> Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas<br />

Introduction<br />

Previous work showed that a commercial PRRS ELISA<br />

(HerdChek® PRRS X3 ELISA) could be adapted to detect anti-<br />

PRRSV IgM, IgA, and IgG in oral fluid specimens. Further, the<br />

protocol for the IgG ELISA for oral fluid samples was readily<br />

amenable to the routine performance <strong>of</strong> the assay in highthroughput<br />

diagnostic laboratories. This suggested the possibility<br />

<strong>of</strong> a cost-effective method to routinely monitor commercial swine<br />

populations for maternal antibody, vaccination compliance, and<br />

herd immune parameters using oral fluid sampling. The purpose<br />

<strong>of</strong> the present study was to evaluate the ability <strong>of</strong> the PRRS oral<br />

fluid IgG ELISA to detect anti-PRRSV IgG antibody in pen-based<br />

oral fluid samples from experimentally inoculated pigs over time.<br />

Materials & methods<br />

In nine trials, ~200 pigs per trials were intramuscularly (IM)<br />

inoculated with PRRSV isolate NVSL 97-7895. Oral fluid<br />

samples were collected on 0, 5, 7, 9, 11, 14, 17, and 21 days<br />

post inoculation (DPI). All oral fluid samples were randomized<br />

and tested for anti-PRRSV antibodies using the PRRS ELISA<br />

protocol for oral fluids: 1:2 oral fluid sample dilution, 250µl sample<br />

volume, 16 hour incubation at 4°C, and detection <strong>of</strong> reaction<br />

using anti-pig IgM, IgA, and IgG FC (1).<br />

Results<br />

Anti-PRRSV antibody isotype (IgM, IgA, IgG FC ) kinetics in<br />

experimental samples are shown in Figure 1 for oral fluid<br />

samples collected on DPI 0, 5, 7, 9, 11, 14, 17, and 21. Mean<br />

IgM, IgA, and IgG S/P ratios on DPI 7 were 1.14 (95% confidence<br />

interval (CI) 1.04, 1.24), 0.09 (95% CI 0.06, 0.12), and 0.59 (95%<br />

CI 0.50, 0.68), respectively. The mean IgM S/P ratio peaked at<br />

DPI 9 (1.88, 95% CI 1.78, 1.97) and declined rapidly to a mean<br />

S/P <strong>of</strong> 0.10 (95% CI 0.09, 0.13) on DPI 21. Mean IgA and IgG<br />

S/P ratios peaked at DPI 11 (1.41, 95% CI 1.29, 1.53) and DPI<br />

14 (3.69, 95% CI 3.52, 3.85), respectively. Mean IgA and IgG<br />

S/P ratios remained relatively constant until the end <strong>of</strong> study at<br />

DPI 21.<br />

Oral fluid, ELISA, Experimental samples, PRRSV, Antibody<br />

Figure 1: Kinetics <strong>of</strong> anti-PRRSV antibody isotypes (IgM, IgA,<br />

IgG FC ) in experimental samples<br />

Discussion & conclusions<br />

These results indicated that anti-PRRSV antibodies in oral fluid<br />

are amenable to rapid detection post-infection. Testing based on<br />

oral fluids could provide an efficient, cost-effective approach to<br />

PRRSV monitoring in commercial herds and surveillance in<br />

elimination programs.<br />

References<br />

1. Kittawomrat, A et al.: <strong>2012</strong>. Detection <strong>of</strong> PRRSV antibodies in oral fluid<br />

specimens using a commercial PRRSV serum antibody ELISA. J Vet<br />

Diagn Invest 24 (2):262-269


S1 - P - 03<br />

DETECTION OF PRRSV ANTIBODY IN ORAL FLUID SPECIMENS FROM INDIVIDUAL BOARS USING A<br />

COMMERCIAL PRRSV SERUM ANTIBODY ELISA<br />

Apisit Kittawornrat 1 , Mark Engle 3 , John Johnson 1 , John Prickett 1 , Chris Olsen 1 , Trevor Schwartz 1 , Daniel<br />

Whitney 1 , Kent Schwartz 1 , Anna Rice 4 , Andrea Ballagi 4 , Sergio Lizano 4 , Chong Wang 1,2 , Jeffrey Zimmerman 1<br />

1 Department <strong>of</strong> Veterinary Diagnostic and Production Animal Medicine, 2 Department <strong>of</strong> Statistics, Iowa State University, 3 PIC North America,<br />

Hendersonville, Tennessee, USA, 4 IDEXX Laboratories, Westbrook, Maine, USA<br />

Oral fluid, Individual boar, ELISA, PRRSV<br />

Introduction<br />

Oral fluid specimens are used in human medicine for the<br />

detection <strong>of</strong> a variety <strong>of</strong> infectious agents, hormones, and drugs.<br />

Oral fluid samples are <strong>of</strong> interest in swine medicine because they<br />

are easily collected, yet highly efficacious for the surveillance <strong>of</strong><br />

PRRSV and other pathogens using PCR-based assays (1,3).<br />

Recent work showed that a commercial PRRS serum antibody<br />

ELISA (IDEXX Laboratories, Inc., Westbrook, ME USA) could be<br />

adapted to detect PRRSV antibody in oral fluid specimens (2). In<br />

the current study we describe the kinetics <strong>of</strong> the ELISAdetectable<br />

anti-PRRSV IgG response in oral fluids collected from<br />

individually-housed boars.<br />

Materials & methods<br />

The study was conducted in 72 boars ranging from 6 months to<br />

3.6 years in age. Boars were under the ownership <strong>of</strong> PIC North<br />

America (Hendersonville, TN USA) and housing, study<br />

procedures, and protocols were approved and supervised by the<br />

PIC USA Health Assurance and Welfare department.<br />

Boars were assigned to three trials (I, II, III). Boars (n = 24) in<br />

Trial I were intramuscularly (IM) inoculated with 2 ml <strong>of</strong> a<br />

modified live virus (MLV) vaccine (RespPRRS®, Boehringer<br />

Ingelheim Vetmedica, Inc., St. Joseph, MO USA). Boars (n = 22)<br />

in Trial II were IM inoculated with 2 ml <strong>of</strong> a Type 1 PRRSV field<br />

isolate. Boars (n = 24) in Trial III were IM inoculated with 2 ml <strong>of</strong> a<br />

PRRSV Type 2 isolate (MN-184).<br />

Boars were monitored for 21 days post inoculation (DPI). Oral<br />

fluid samples were collected daily using 5/8" 3-strand 100%<br />

cotton rope. Serum samples were collected from all boars on DPI<br />

-7, 0, 7, 14, 21 and from 4 randomly selected boars on DPIs 3, 5,<br />

10, and 17. Thereafter, serum and oral fluid were assayed for<br />

PRRSV antibody using the ELISA protocol appropriate for each<br />

sample type (serum or oral fluid).<br />

Results<br />

The ELISA S/P response in oral fluid and serum samples<br />

followed similar patterns over time (Figure 1). Individual boar oral<br />

fluid samples were ELISA positive from DPI 8 to DPI 21 (Table<br />

1). Overall, 96% <strong>of</strong> the results were in agreement, i.e., 145 oral<br />

fluid samples and 150 serum samples were ELISA positive.<br />

Table 1: Detection <strong>of</strong> PRRSV antibody in oral fluid samples from<br />

individually housed boars<br />

PRRSV<br />

ELISA-positive boars (%) by day post inoculation<br />

0 8 9 10 11 12 13 14 17 & 21<br />

MLV 0 0 38 71 88 96 100 100 100<br />

Type 1 0 17 42 59 82 82 81 85 100<br />

Type 2 0 17 65 83 96 96 100 100 100<br />

Total 0 11 48 71 89 91 91 93 100<br />

Discussion & conclusions<br />

The ring test results showed that the PRRS oral fluid IgG ELISA<br />

was highly reproducible across laboratories. These results<br />

support the routine use <strong>of</strong> this test in laboratories providing<br />

diagnostic service to pig producers. Thus, herd monitoring based<br />

on oral fluid sampling could be one part <strong>of</strong> a PRRSV control<br />

and/or elimination program. Further, the successful adaptation <strong>of</strong><br />

one assay to the oral fluid matrix suggests that this approach<br />

could provide the basis for monitoring specific health and welfare<br />

indicators in commercial swine herds using a "pig friendly"<br />

approach.<br />

References<br />

1. Kittawornrat A, et al. 2010. PRRSV in serum and oral fluid samples from<br />

individual boars. Virus Res 154:170-176.<br />

2. Kittawornrat A, et al. <strong>2012</strong>. Detection <strong>of</strong> PRRSV antibodies in oral fluid<br />

specimens using a commercial PRRSV serum antibody ELISA. J Vet<br />

Diagn Invest 24(2):262-269.<br />

3. Ramirez et al. <strong>2012</strong>. Efficient surveillance <strong>of</strong> pig populations using oral<br />

fluids. Prev Vet Med (Epub ahead <strong>of</strong> print).<br />

Figure 1: Mean PRRS antibody ELISA kinetics over time: ora<br />

fluid vs. serum samples


S1 - P - 04<br />

DIAGNOSIS OF MAIN HAEMOPARASITIC DISEASES OF CATTLE BY REAL-TIME PCR.<br />

Villa Aleida 1 ; Benito AA 1 ; Arnal JL 1 ; Serrano JD 1 and Baselga R 1 .<br />

1 EXOPOL Autovacunas y Diagnóstico. Pol. Río Gállego, 50840 San Mateo de Gállego Zaragoza, España. Tel 976 69 45 25, exopol@exopol.com<br />

Piroplasmosis, Anaplasmosis, Diagnostic, qPCR, cattle<br />

Introduction<br />

Piroplasmosis and Anaplasmosis are the most important blood<br />

parasitic diseases in cattle and responsible for significant<br />

economical losses and mortality in livestock from many countries.<br />

Although these diseases occur mainly in tropical and subtropical<br />

areas, they are also becoming an increasing and serious problem<br />

in Europe ( 1 ). Clinical signs in haemoparasitic diseases are quite<br />

similar and include anemia, pyrexia, weakness, weight loss and<br />

drop in production levels. Bovine Piroplasmosis is caused by<br />

pathogenic Babesia and Theileria species. Th. parva and Th.<br />

annulata are the most important pathogens in this genus,<br />

although only Th. Annulata has been described in Europe ( 2 ).<br />

There are several pathogenic Babesia species affecting cattle (B.<br />

major, B. bovis, B. divergens and B.bigemina), but B. bigemina<br />

and B. bovis are the most frequently described ( 3 ). Bovine<br />

Anaplasmosis is caused by rickettsia <strong>of</strong> genus Anaplasma. A.<br />

marginale is considered the most pathogenic specie while A.<br />

centrale has been implicated in mild or less severe cases <strong>of</strong> this<br />

disease ( 4 ) .<br />

Laboratory diagnosis <strong>of</strong> these pathogens included<br />

their microscopical identification in blood smears; however, this<br />

method requires highly qualified personnel and is not reliable for<br />

detecting pre-symptomatic or carrier animals. Serological tests<br />

can also be used; although problems <strong>of</strong> cross-reactivity between<br />

pathogens <strong>of</strong> same genus have been reported. Recently, some<br />

techniques <strong>of</strong> polymerase chain reaction (PCR) have been<br />

developed and allowed a specific and sensitive detection <strong>of</strong> these<br />

agents ( 5 ) .<br />

The main objective <strong>of</strong> this study was evaluate the<br />

feasibility <strong>of</strong> a Real-time PCR (qPCR) assay for the diagnosis <strong>of</strong><br />

Th. annulata, Th. parva, B. bigemina, B. bovis, A. marginale, A.<br />

centrale and A. phagocitophylum, in a multiparametric test for a<br />

specific identification and also quantification <strong>of</strong> these main tickborne<br />

pathogens <strong>of</strong> cattle.<br />

Materials & methods<br />

Samples: A total <strong>of</strong> 122 cases (330 blood, 58 serum and 9 tissue<br />

samples) submitted to our laboratory with suspect <strong>of</strong><br />

haemoparasitic disease were evaluated. Some <strong>of</strong> these cases<br />

(10% aprox) had macroscopical findings and histopathological<br />

lesions <strong>of</strong> haemolytic syndrome. Most <strong>of</strong> cases 93% (113/122)<br />

came from 21 provinces in Spain and the rest from Portugal.<br />

Whole blood, serum and tissue samples were subjected to DNA<br />

extraction individually or pooled up to five per case.<br />

DNA extraction: A commercial kit “Genomic mini Kit LGD 500<br />

LabTurbo” and an automated DNA/RNA extraction System<br />

“LabTurbo 36 compact system C3620” (Taigen Bioscience<br />

Corporation, Taiwan) were used for DNA extraction following the<br />

manufacturer's instructions. DNA purity (260/280) and DNA yields<br />

were determined using a micro-volume spectrophotometer<br />

(Quawell UV Q5000, s<strong>of</strong>tware 4.0).<br />

Quantitative Real-Time PCR (Genesig Ltd): The qPCR assays<br />

used in this study identified the following pathogen-specific<br />

targets: Tams1 gene (Merozoite-piroplasm surface antigen) for<br />

Th. annulata; p104 gene (Microneme-rhoptry antigen) for Th.<br />

Parva; 18S gene (18S ribosomal RNA) for B. bigemina; 18S<br />

gene (18S ribosomal RNA) for B. bovis; msp4 gene (Major<br />

surface protein 4) for A. marginale; gen; msp2 gene (Major<br />

surface protein 2) for A. centrale and msp4 gene (Major surface<br />

protein 4) for A. phagocytophylum. A unique qPCR protocol were<br />

performed for all these pathogens according manufacturer's<br />

instructions, briefly: 10µl <strong>of</strong> MasterMix, 1µl <strong>of</strong> pathogen-specific<br />

primer/probe (FAM labeled, BHQ quenched), 5µl <strong>of</strong> RNAse/<br />

DNAse free water and 5µl <strong>of</strong> DNA sample (> 5ng) or control<br />

template conformed the PCR reaction in a final volume <strong>of</strong> 20 µl.<br />

Amplification reactions were carried out on a MiniOpticon RT<br />

System CFB3120 thermal cycler (Bio Rad, s<strong>of</strong>tware CFX<br />

manager 2.0) using a unique thermal pr<strong>of</strong>ile for all qPCRs<br />

consisting <strong>of</strong> an initial enzyme activation step <strong>of</strong> 10 min a 95º C,<br />

followed by 50 cycles <strong>of</strong>: a denaturing step <strong>of</strong> 10 sec at 95º C,<br />

and an annealing/data collection step <strong>of</strong> 60 second at 60º C.<br />

Positive cut-<strong>of</strong>f value was established in ≤ 45 cycles. Detection<br />

limits and quantification were performed by 10 fold dilutions (10 6<br />

a 10 0 ) <strong>of</strong> specific DNA plasmids for each pathogen. Negative<br />

samples were evaluated for DNA integrity by amplification <strong>of</strong><br />

Beta-actina house-keeping gene.<br />

Results<br />

This multi-parametric qPCR assay was robust, specific and<br />

showed an analytical sensitivity for at less 10 2 copies <strong>of</strong> the target<br />

DNA. The Cq values for positive controls ranged from 25 to 35<br />

and the qPCR efficiencies were around 95% with R 2<br />

values<br />

above 0.990. A total <strong>of</strong> 122 cases were evaluated in this study<br />

and 49% (59/122) <strong>of</strong> them were positive by qPCR for at less one<br />

<strong>of</strong> these pathogens. Haemoparasites were detected in blood<br />

samples 52% (171/330), but also in tissue 22% (2/9) and serum<br />

38% (22/58) samples. Nearly 10% <strong>of</strong> cases had macroscopical<br />

and/or histopathological lesions compatible with haemolytic<br />

syndrome. Microscopical identification <strong>of</strong> Piroplasmids and<br />

Anaplasma were found in blood and/or tissue samples from 8 <strong>of</strong><br />

these cases and all <strong>of</strong> them had one or more positive result for<br />

haemoparasites by qPCR. From the overall cases Th. annulata<br />

27% (33/122) and A. marginale 27% (33/122) were the more<br />

frequent detected pathogens, followed by B. bigemina 16%<br />

(20/122), B. bovis 2% (3/122) and A. phaghocytophylum 2%<br />

(2/122). No positive cases were recorded for Th. Parva (0/122)<br />

and A. centrale (0/122). Concurrent infections with Th. annulata,<br />

A. marginale and B. bigemina were detected in 15% (9/57) <strong>of</strong> the<br />

positive cases.<br />

Discussion & conclusions<br />

Specific identification <strong>of</strong> haemoparasites infecting cattle is a<br />

crucial step for control <strong>of</strong> these diseases; because measures and<br />

treatment depend <strong>of</strong> the implicated agent. Although microscopical<br />

identification is possible it is not always reliable even for most<br />

expertice technicians. Clinical diagnosis is also difficult, by the<br />

similar symptomatology in these diseases. Our results clearly<br />

demonstrate the value <strong>of</strong> these qPCRs as a specifics and<br />

sensitive diagnostic tools for the rapid detection <strong>of</strong> main tickborne<br />

disease <strong>of</strong> cattle in blood and tissue sample from infected<br />

animals. The use <strong>of</strong> this qPCR multi-parametric assay allow a<br />

complete diagnosis <strong>of</strong> the main haemoparasitic diseases in a<br />

rapid way and reducing cost by pooling samples, up to five, per<br />

case. It also has the advantge for no competition <strong>of</strong> multiple<br />

primers by the target DNA, such ocurrs in a qPCR multiplex<br />

where a DNA depletion could happen mainly in animals with<br />

mixed infections. Our results showed a high percentage <strong>of</strong><br />

haemoparasitic infections, mainly Th annulata, A. marginale and<br />

B. bigemina, in the evaluated cases from the Iberian peninsula<br />

and justified the use <strong>of</strong> specific and sensitive diagnostic test, as<br />

this qPCR, to study the real epidemiological situation <strong>of</strong> these<br />

disease in the cattle population from these countries.<br />

References<br />

1. Paul Heyman, Christel Cochez, Agnetha H<strong>of</strong>huis, Joke van der Giessen,<br />

Hein Sprong, Sarah Rebecca Porter, et al. A clear and present danger:<br />

Tick-borne diseases in Europe. Expert Rev Anti Infect Ther. 2010; 8(1):33-<br />

50.<br />

2. R. Cassini, F. Marcer, F. di Regalbono, G. Cancrini, S. Gabrielli, A.<br />

Moretti, et al. New insights into the epidemiology <strong>of</strong> bovine piroplasmoses<br />

in Italy. Vet Parasit <strong>2012</strong>(184):77-82.2. R. Cassini, F. Marcer, F. di<br />

Regalbono, G. Cancrini, S. Gabrielli, A. Moretti, et al. New insights into the<br />

epidemiology <strong>of</strong> bovine piroplasmoses in Italy. Vet Parasit <strong>2012</strong>(184):77-<br />

82.<br />

3. S. Almeria, J. Castellà, D. Ferrer, A. Ortuño, A. Estrada-Peña, J.F.<br />

Gutiérrez. Bovine piroplasms in Minorca (Balearic Islands, Spain): a<br />

comparison <strong>of</strong> PCR-based and light microscopy detection. Vet Parasitol<br />

2001(99):249–259.<br />

4. L. Ceci, N. Decaro, E. Lorusso, P. Paradies, G. Elia, V. Martella, et al.<br />

First report <strong>of</strong> bovine anaplasmosis by Anaplasma centrale in Europe,<br />

molecular identification and phylogenetic analysis. Vet Res commun<br />

2008(32):S236-S266.<br />

5. Oficina Internacional de Epizootias (OIE). Manual de las Pruebas de<br />

Diagnóstico y de las Vacunas para los Animales Terrestres. 2008.


S1 - P - 05<br />

DEVELOPMENT AND EVALUATION OF A NEW AND ORIGINAL EXTRACTION PROTOCOL TO<br />

DETECT MYCOBACTERIUM AVIUM SUBSP PARATUBERCULOSIS IN BOVINE FECES BY REAL TIME<br />

PCR<br />

Blanchard Béatrice. 1 , Versmisse Yann. 1 , Rouillard Tony. 2<br />

1 ADIAGENE, 38 rue de Paris 22000 Saint Brieuc, FRANCE<br />

2<br />

AES CHEMUNEX, Rue Maryse Bastié, 35172 Bruz, FRANCE<br />

Mycobacterium paratuberculosis (Map), real-time PCR, diagnosis, low shedder.<br />

Introduction<br />

Mycobacterium avium ssp paratuberculosis (MAP) is the<br />

etiological agent <strong>of</strong> Johne’s disease, a granulomatous enteritis<br />

that can affect cattle, sheep and goats and other non ruminant<br />

wildlife species. Numerous countries have set up programmes to<br />

control the disease. It requires the development <strong>of</strong> highthroughput<br />

sensitive diagnostic methods for the direct detection<br />

<strong>of</strong> infected animals. The performance <strong>of</strong> the PCR methods was<br />

<strong>of</strong>ten hampered by low sample input and inhibition. To increase<br />

the sensitivity <strong>of</strong> diagnostic test, we developed an original<br />

protocol to concentrate Map from 6g <strong>of</strong> faecal samples and<br />

compared the real time PCR results obtained with classical DNA<br />

extraction from 1g.<br />

Materials & methods<br />

A negative faeces collected from a paratuberculosis-free herd<br />

was spiked with serial dilutions <strong>of</strong> a MAP titrated suspension to<br />

4670, 2333, 1167, 233 and 117 bacteria per g. DNA extraction <strong>of</strong><br />

each sample spiked was repeat four times using the new protocol<br />

(NP) developed by Adiagene. Briefly, 6g +/- 0.2 g <strong>of</strong> faeces were<br />

placed in a bottle with 40 ml <strong>of</strong> sterile distilled water, mixed 15<br />

seconds and allowed to settle 10 to 20 minutes. 10 ml <strong>of</strong> the<br />

obtained supernatant were transferred on an ADIAFILTER and<br />

centrifuged 5 minutes at 3 000 g. The obtained pellet was<br />

resupended then disrupted 10 minutes at 30 Hz with a mixer mill<br />

and 300 mg <strong>of</strong> glass beads and centrifuge 5 minutes at 15 000 g.<br />

Then DNA was extracted from 200 μl <strong>of</strong> the supernatant.<br />

5µL <strong>of</strong> each DNA solution was analyzed with the ADIAVET ®<br />

PARATB REALTIME PCR kit (Adiagene).<br />

The detection limit <strong>of</strong> the diagnostic method is the smallest<br />

amount <strong>of</strong> MAP per sample generating 100% positive results.<br />

Evaluation <strong>of</strong> the new protocol on field samples<br />

329 faecal samples were collected from different veterinary<br />

diagnostic laboratories. The common point among these samples<br />

is that they were sent to the laboratory for MAP analyses; they<br />

came from different area, from different herds with or without<br />

history <strong>of</strong> paratuberculosis.<br />

All the samples were analysed in parallel with two DNA extraction<br />

protocols.<br />

This first was performed using Adiapure ®<br />

(Adiagene) DNA<br />

extraction protocol (SP). The analyses were carried out from 1g<br />

<strong>of</strong> faecal sample as recommended by the manufacturer.<br />

The second DNA extraction was realised using the new protocol<br />

(NP). It was achieved with 6g +/- 0.2 g <strong>of</strong> faeces as previously<br />

described.<br />

5 µl <strong>of</strong> the each DNA solution obtained from the 2 different<br />

protocols was analysed with ADIAVET ® PARATB REALTIME kit.<br />

difficult to obtain a good sensitivity <strong>of</strong> the test due to the presence<br />

<strong>of</strong> inhibitor and the biology <strong>of</strong> the bacteria.<br />

Foremost, it was important to increase the quantity <strong>of</strong> treated<br />

faecal sample. Most <strong>of</strong> the works done for MAP detection like<br />

culture or PCR treat up to 1g <strong>of</strong> faeces. Even the new technique<br />

combining magnetic beads separation and PCR uses a minimum<br />

amount <strong>of</strong> samples.<br />

The originality <strong>of</strong> our protocol is to be able to treat up to 10g <strong>of</strong><br />

faeces in order to concentrate the MAP present in the sample. In<br />

the standard protocol (SP) the DNA <strong>of</strong> 1g <strong>of</strong> faeces are treated.<br />

After homogenization and DNA purification, 0.15mg are used for<br />

PCR analyse; With the new protocol (NP), 6g are extracted and<br />

15mg <strong>of</strong> feces are used for PCR analyse. So, we are able to<br />

concentrate 100 times the sample and increase the sensitivity <strong>of</strong><br />

the whole method. Prior study have suggested that high<br />

shedders yield > 50CFU/tube and low shedders


S1 - P - 06<br />

VALIDATION OF REAL-TIME PCR TEST FOR THE DETECTION AND QUANTIFICATION OF COXIELLA<br />

BURNETII FOR ABORTION CONTROL PROGRAM<br />

Leborgne Maëlle 1 , Blanchard Béatrice 1 , Gracieux Patrice 1 , Rouillard Tony 2<br />

1<br />

ADIAGENE, 38 rue de Paris 22000 Saint Brieuc, FRANCE<br />

2<br />

AES CHEMUNEX, Rue Maryse Bastié, 35172 Bruz, FRANCE<br />

Coxiella burnetii, Quantitative PCR, Diagnosis, Zoonosis<br />

Introduction<br />

Q fever, a zoonosis caused by the obligate intracellular bacterium<br />

Coxiella burnetii, is endemic throughout the world and infects<br />

arthropods, birds, pets, domestic and wild mammals, and<br />

humans.<br />

The recent developments in the EU, especially the increase in<br />

confirmed human cases <strong>of</strong> Q fever in the Netherlands, call for<br />

special consideration as regards the risks posed by Q fever for<br />

humans and animals. The European Commission requested<br />

further scientific advices and human risk assessment, as regards<br />

Q fever in animals (1). For EU it’s important to assess the<br />

significance <strong>of</strong> the occurrence <strong>of</strong> Q fever in the EU Member<br />

States for a better understanding <strong>of</strong> the scale and distribution <strong>of</strong><br />

the disease and infection. In this context, French authorities<br />

organise a national program <strong>of</strong> Q fever surveillance. The control<br />

measures in infected herds consist first in preventing<br />

contamination <strong>of</strong> susceptible animals in reducing Coxiella<br />

shedding in infected ruminants. So, it is necessary to detect and<br />

quantify the amount <strong>of</strong> bacteria in infected samples in order to<br />

eliminate or separate the heaviest shedders from the herds.<br />

To reach this goal it is important to improve and standardize the<br />

diagnostic method in laboratories. We have developed a<br />

quantitative real time PCR test according to the guidelines for the<br />

development and validation <strong>of</strong> veterinary PCR (AFNOR, XP U47-<br />

600-2) (2). The new ADIAVET ® kit was validated by the French<br />

national reference laboratory for Q fever (ANSES, Sophia-<br />

Antipolis, France).<br />

Materials & methods<br />

One-well quantitative real time PCR was developed to detect<br />

simultaneously the C. burnetii IS1111 and a housekeeping gene<br />

GAPDH as an endogenous internal control <strong>of</strong> extraction and<br />

amplification.<br />

The specificity <strong>of</strong> the PCR test was tested against a panel <strong>of</strong><br />

bacteria and virus commonly found in bovine, caprine or ovine<br />

samples.<br />

The detection limit <strong>of</strong> the PCR (LD PCR ) generating a positive result<br />

in 95% <strong>of</strong> cases and the quantification limit <strong>of</strong> the PCR (LQ PCR )<br />

were calculated with a standard genomic DNA solution provided<br />

by the French national reference laboratory for Q fever (ANSES-<br />

Sophia Antipolis).<br />

The national control program recommended to collect vaginal<br />

swabs or placenta swabs after abortion.<br />

Four PCR methods, including DNA extraction from sample and<br />

PCR, were evaluated. The detection limit and the field <strong>of</strong><br />

quantification <strong>of</strong> each method were calculated.<br />

Table 1: Determination <strong>of</strong> the detection limit and <strong>of</strong> the field <strong>of</strong><br />

quantification for each method. The results are expressed in<br />

C.burnetii/ml.<br />

Vaginal<br />

swab<br />

Placenta<br />

swab<br />

QIAamp ® DNA Mini Kit<br />

Detection limit<br />

<strong>of</strong> method<br />

Field <strong>of</strong><br />

quantification<br />

NucleoSpin ® Tissue<br />

Detection limit<br />

<strong>of</strong> method<br />

Field <strong>of</strong><br />

quantification<br />

300 500 à 1.10 6 300 500 à 1.10 6<br />

300 500 à 1.10 6 300 500 à 1.10 6<br />

Discussion & conclusions<br />

To assess the sanitary status <strong>of</strong> the herds, the animals are<br />

considered as clinically infected if the swab contains more than<br />

10 4 C. burnetii/ml. Thus, the performances <strong>of</strong> the real-time PCR<br />

developed match with the requirements for the management <strong>of</strong><br />

C. burnetii infection.<br />

This quantitative real time PCR test was validated by the French<br />

national reference laboratory for Q fever. Currently, it’s used in<br />

France for a national program <strong>of</strong> Q fever surveillance. In addition,<br />

this kit may be also used for the epidemiologic diagnostic <strong>of</strong> C.<br />

Burnetii in faeces, milk, tissues and foetal liquid.<br />

References<br />

1. EFSA (European Food Safety Authority) (2010). Panel on Animal<br />

Health and Welfare (AHAW): More, S., Stegeman, al. Scientific Opinion on<br />

Q Fever. EFSA Journal, 8(5):1595. [114 pp.].<br />

doi:10.2903/j.efsa.2010.1595. May.<br />

2. AFNOR XP U47-600-2 (juin 2011), Méthodes d’analyse en santé<br />

animale – PCR (réaction de polymérisation en chaîne) - Partie 2 :<br />

exigences et recommandations pour le développement et la validation de<br />

la PC<br />

Results<br />

This PCR test was assessed against a panel <strong>of</strong> 130 samples<br />

issues from organisms preferencially found in the same ecologic<br />

niche and/or close phylogeneticaly, among which Legionella. No<br />

cross reaction was observed.<br />

The analysis Probit showed that LD PCR was to 1.5 Genome<br />

Equivalent/reaction (5µL). LQ PCR was calculated to 2 Genome<br />

Equivalent/reaction (5µL).<br />

The results <strong>of</strong> the evaluation <strong>of</strong> different methods are showed in<br />

the following table.


S1 - P - 07<br />

ANALYTICAL EVALUATION OF A SWINE INFLUENZA VIRUS PCR ON AVIAN SAMPLES<br />

J. C. Pedersen 1 , C. Boss 2 , A. Burrell 3 , C. O’Connell 3<br />

1<br />

National Veterinary Services Laboratory (NVSL), Ames, Iowa. USA<br />

2<br />

Life Technologies,Darmstadt, Germany<br />

3<br />

Life Technologies, Austin, USA<br />

Introduction<br />

Swine influenza is a highly contagious viral infection <strong>of</strong> pigs that<br />

has a significant economic impact on affected herds. The disease<br />

is caused by Swine Influenza Virus (SIV) which is a group <strong>of</strong><br />

specified viruses that are members <strong>of</strong> the family<br />

Orthomyxoviridae and placed in the genus influenzavirus A.<br />

Pigs play a unique role in the ecology <strong>of</strong> Influenza Type A, in that<br />

they have receptors in their respiratory tract that will bind swine,<br />

human, and avian influenza viruses. Consequently, pigs have<br />

been called the ‘mixing vessels’ for the development <strong>of</strong> new<br />

influenza viruses when swine, avian, and/or human influenza<br />

viruses undergo genetic reassortment in pigs. Subtypes <strong>of</strong> SIV<br />

that are most frequently identified in pigs include classical and<br />

avian H1N1, human (hu) H1N1 and H1N2, reassortant (r) H3N2,<br />

and rH1N2.<br />

In theory, a PCR based test which detects RNA coding for the<br />

matrix and nucleoprotein genes <strong>of</strong> SIV, an Influenzavirus A,<br />

should be able to detect similar viral RNA sequences in avian<br />

samples. The USDA licensed Swine Influenza Virus RNA Test Kit<br />

(VetMAX ® -Gold SIV Detection Kit) was used to test RNA<br />

extracted from a panel <strong>of</strong> clinical Avian Influenza Virus samples<br />

representing all 16 HA subtypes in order to assess the<br />

performance <strong>of</strong> the test in species other than swine.<br />

Materials & methods<br />

A panel <strong>of</strong> 34 cultured isolates (stock virus) representing all 16<br />

HA Influenzavirus A subtypes was assembled at the NVSL. RNA<br />

was extracted from the samples using the MagMAX ® Viral RNA<br />

Isolation Kit following manufactures’ instructions. Undiluted RNA<br />

extracted from the stock viruses was tested using the VetMAX ® -<br />

Gold SIV Detection Kit real time PCR from Life Technologies<br />

following the product insert instructions. The test detects RNA<br />

representing two different targets on the SIV matrix gene as well<br />

as an additional target on the nucleoprotein gene.<br />

Results<br />

Strong amplification signals were obtained (Ct range: 13.2-20.9)<br />

for each <strong>of</strong> the panel members tested. The quality standards<br />

including positive and negative extraction conotrols, positive kit<br />

control and no template controls performed as expected.<br />

Discussion & conclusions<br />

This preliminary study gives an indication that further validation<br />

studies are warranted to support additional claims for use <strong>of</strong> this<br />

product for the detection <strong>of</strong> Influenzavirus A in avian samples.<br />

On a global level, the use diagnostic assays for the detection <strong>of</strong><br />

certain reportable diseases is becoming more highly regulated in<br />

order to ensure that clinical decisions are being made from high<br />

quality laboratory results. In light <strong>of</strong> this, the advantages listed<br />

above must be understood along with the limitations <strong>of</strong> these<br />

reported data.<br />

Limitations: The VetMAX ® -Gold SIV Detection Kit is a USDA<br />

licensed product for the detection <strong>of</strong> Swine Influenza Virus (SIV)<br />

RNA isolated from nasal swab samples. Off-label uses <strong>of</strong> the<br />

product in the United States are not supported by the<br />

manufacturer and any additional claims for performance <strong>of</strong> the kit<br />

must be supported by rigorous studies which must be submitted<br />

and approved by the USDA before any additional claims can be<br />

made in the US. Usage claims in other countries outside the US<br />

are subject to the regulatory requirement(s) within each country<br />

<strong>of</strong> use.<br />

References<br />

OIE Terrestrial Manual 2010, Chapter 2.8.8. Swine Influenza, accessed 30<br />

Mar <strong>2012</strong><br />

SIV, PCR, Avian Samples


S1 - P - 08<br />

QUALITY CONTROL OF PRRSV VACCINATION BY PARAMETERS OF IMMUNITY<br />

Böttcher J 1 , Janowetz B 1 , Alex M 1 , Seidenspinner M 2 , Schuh H 2 , Strutzberg-Minder K 3 , Wendt M 4 , Leibold W 5<br />

1<br />

Bavarian Animal Health Service, Poing, Germany,<br />

2<br />

Dr. Hermann Schuh, Veterinary Practice, Ipsheim, Germany<br />

3<br />

IVD GmbH, Hannover, Germany,<br />

University <strong>of</strong> Veterinary Medicine, Foundation, 4 Klinik für kleine Klauentiere, 5 Institute <strong>of</strong> Immunology, Hannover, Germany<br />

PRRS, vaccination, immunity<br />

Introduction<br />

Control <strong>of</strong> porcine reproductive and respiratory syndrome (PRRS)<br />

mainly relies on vaccination. Quality control <strong>of</strong> vaccination should<br />

focus on diagnostic parameters which are associated with<br />

immunity, i.e. neutralizing antibodies (nab) and Interferon-gamma<br />

(IFN-γ) response. The level <strong>of</strong> maternally derived nabs (MDNab)<br />

has to be included as an additional variable.<br />

Materials & methods<br />

Six sows were tested for nabs 2 weeks (w) post partum. Piglets<br />

were classified by MDNab in “0.05, i.e. the lowest<br />

value <strong>of</strong> the IFN-γ-standard titration, or OD SC = 4<br />

< 4<br />

1 2 3 4<br />

vaccination group<br />

NT_US<br />

0<br />

Fig.2: Genotype-specificity <strong>of</strong> NT (n=58) and IFN-γ (n=29) in w27.<br />

Interferon-gamma (ng/ml)<br />

1000<br />

100<br />

10<br />

IFN_EU<br />

Vaccination group 2<br />

IFN_US<br />

Fig.3: PRRSV-EU-IFN-γresponse.<br />

Data for 12, 4, 1<br />

and 8 piglets are shown for<br />

groups 1, 2, 3 and 4,<br />

respectively. Groups 2 and<br />

3 showed marked IFN-γresponse<br />

against Marc145<br />

(NC). The effect <strong>of</strong> MDNab<br />

in group 1 was significant<br />

(p=0.036, Mann-Whitneytest,<br />

two-tailed probability).<br />

Acknowledgements<br />

This study was financially supported by the Free State <strong>of</strong> Bavaria and the<br />

Bavarian Joint Founding Scheme for the Control and Eradication <strong>of</strong> contagious<br />

Livestock Diseases. This study was performed as a part <strong>of</strong> the thesis <strong>of</strong> Mr. Marc<br />

Seidenspinner.<br />

References<br />

1. Böttcher, J., M. Ritzmann, A. Gangl (2006): Felduntersuchungen mit einem<br />

„Porcine Reproductive and Respiratory Syndrome Virus“ (PRRSV)<br />

Neutralisationstest. Tierärztl. Umschau, 61, 550 – 559.<br />

Neutralization titer (PRRSV EU)<br />

Neutralization titer (PRRSV EU)<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Nab-titer <strong>of</strong> sows<br />

>= 4<br />

= 4<br />

< 4<br />

2 7 10 13 16 21 25 26 27<br />

weeks <strong>of</strong> life


S1 - P - 09<br />

PTS FOR BVDV ANTIGEN DETECTION BY GD - ANIMAL HEALTH SERVICE<br />

Rianne Buter 1 , Jet Mars 1 , Wim Swart 1 , Huub van de Sande 1 , Kees van Maanen 1<br />

1<br />

GD-Animal Health Service, Diagnostics,Research and Epidemiology,Deventer , the Netherlands<br />

Pr<strong>of</strong>iciency tests, nucleic acid amplification techniques, ISO/IEC 17043:2010, BVDV, PCR<br />

Introduction<br />

Quality control should be an integral element in the<br />

implementation <strong>of</strong> nucleic acid amplification techniques (NATs)<br />

e.g. PCR in diagnostic laboratories. Dealing with test<br />

characteristics like specificity and sensitivity, variations in<br />

qualitative and quantitative results, contamination and inhibition<br />

control mechanisms remains an on-going challenge. External<br />

quality controls are a useful tool to improve a laboratories quality<br />

system.<br />

At this moment ,different pr<strong>of</strong>iciency tests for nucleic amplification<br />

techniques (NAT’s) e.g.PCR are available at GD, such as for<br />

Bovine Viral Diarrhoea Virus (BVDV), mastitis pathogens,<br />

Porcine Circovirus type 2 (PCV-2), Porcine Reproductive and<br />

Respiratory Syndrome Virus (PRRSV) and Brachyspira<br />

hyodysenteriae.<br />

In general, the purpose <strong>of</strong> pr<strong>of</strong>iciency testing is to determine the<br />

performances <strong>of</strong> individual laboratories for specific tests. All PTS<br />

organized by GD are performed according to ISO/IEC<br />

17043:2010 guidelines. This abstract presents the (anonimized)<br />

results <strong>of</strong> the BVDV antigen and genome detection PTS that was<br />

organized in 2011.<br />

Materials & methods<br />

The BVDV antigen detection PTS consisted <strong>of</strong> 11 inactivated,<br />

freeze-dried sera and 1 inactivated and freeze dried tissue<br />

sample, which were sent to each <strong>of</strong> the participants with the<br />

request to test the samples for BVDV antigen/genome using all<br />

PCR and antigen ELISA techniques in operation at the time. The<br />

samples were identified by sample numbers (#1 - #12) only.<br />

Before shipment homogeneity and stability <strong>of</strong> the samples were<br />

checked. The samples were sent by TNT courier to participants<br />

all over the world but mainly in Europe and the deadline for<br />

testing and reporting was 6 weeks. The participants were asked<br />

to analyse the samples in duplicate in two different test runs.<br />

Results<br />

Statistical calculations were performed when there were at least<br />

6 laboratories participating within one test system. The boxplot<br />

represents the measurements for each sample <strong>of</strong> all laboratories<br />

participating in a commercial BVD ag ELISA (figure 1) and the<br />

XY-diagram represents the average z-scores <strong>of</strong> all laboratories<br />

using the commercial ELISA. (figure 2)<br />

Figure 1 BVDV ELISA boxplot<br />

For the BVDV real time PCR only the within lab reproducibility<br />

could be calculated (table 1), since there were too many<br />

differences in RNA isolation methods, PCR mixtures, PCR<br />

cyclers e.g.<br />

Table 1: BVDV Real-time PCR within-lab-reproducibility<br />

Lab<br />

Mean value<br />

Within Lab<br />

reproducibility sd<br />

A 28.20 0.10<br />

B 36.15 2.39<br />

C 28.85 1.54<br />

D 30.66 1.53<br />

E 30.96 0.19<br />

F 29.78 0.24<br />

G* 37.25 .<br />

H 30.54 0.39<br />

I 32.18 0.24<br />

J 28.53 0.37<br />

K 29.29 0.95<br />

M 30.27 2.29<br />

N* 29.03 .<br />

O 29.84 0.41<br />

P 30.11 1.48<br />

Q 28.36 0.44<br />

R 29.23 0.87<br />

S 30.94 0.60<br />

Labs marked with * reported only single results.<br />

Discussion & conclusions<br />

In this PTS specificity did not seem to be a problem for both<br />

ELISA and PCR, since almost all labs scored a BVDV negative<br />

sample and a serum sample spiked with a heterologous RNA<br />

virus correctly negative.<br />

Detection limits <strong>of</strong> the ELISAs used in this PTS, as measured by<br />

TCID 50 /mL, were relatively high with all spiked samples up to a<br />

titre <strong>of</strong> 10 4,05 TCID 50 /mL being scored negative by all participants.<br />

Also (maternal) antibodies can interfere with virus detection by<br />

ELISA, as demonstrated by the results for samples #6 and #10.<br />

Since such interference does not occur with PCR, critical<br />

evaluation <strong>of</strong> discrepant test results is always needed when PCR<br />

and ELISA are both used in the context <strong>of</strong> BVDV diagnosis and<br />

compulsory or voluntary control and eradication programmes.<br />

Most <strong>of</strong> the laboratories that participated in this PTS used realtime<br />

PCR for BVDV detection. The detection limit <strong>of</strong> real-time and<br />

conventional PCRs as compared with the antigen detection<br />

ELISAs used in this PTS is much lower as demonstrated by the<br />

fact that allmost all serum samples spiked with BVDV-1 virus with<br />

a titre <strong>of</strong> 10 1,9 TCID50/mL or higher scored positive. The overall<br />

results <strong>of</strong> the laboratories that participated in this first GD BVDV<br />

antigen and genome PTS were reasonably good. Most labs have<br />

reliable assays in place for detection <strong>of</strong> BVDV PI-animals.<br />

Acknowledgements<br />

We thank our product-sales manager Annemiek Slothouber for<br />

her support and enthusiasm.<br />

We thank all participants for their enthusiasm, their quick<br />

response and their valuable comments.<br />

Figure 2 BVDV ELISA scatterplot


S1 - P - 10<br />

PRESENTATION OF THE RESULTS OF THE ANNUAL PROFICIENCY TEST FOR THE SEROLOGICAL<br />

DIAGNOSIS OF EQUINE INFECTIOUS ANEMIA CONDUCTED IN ITALY BETWEEN 2006-2010.<br />

Ricci I 1 , Gasperetti L. 1 , D'Alonzo A. 1 , Nardini R. 1 , Giusti C. 1 , Caprioli A. 1 , Forletta R. 1 .<br />

1<br />

National reference centre for equine infectious Anemia, Istituto Zoopr<strong>of</strong>ilattico Sperimentale Lazio e Toscana<br />

Keywords: Equine infectious anemia, agar gel immunodiffusion, pr<strong>of</strong>iciency test<br />

Introduction<br />

Equine infection anemia (EIA) is a viral infection <strong>of</strong> equidae,<br />

caused by a virus (EIAV) <strong>of</strong> the Lentivirus genus, Retroviridae<br />

family. Since 2006, Italy has adopted a national surveillance plan<br />

based on the serological screening <strong>of</strong> all animals older than 6<br />

months except for meat horses. According to the Italian<br />

Regulations, the <strong>of</strong>ficial confirmatory test is the agar gel<br />

immunodiffusion (AGID), which detects antibodies against the<br />

p26, a highly conserved and primary immunogenic structural<br />

protein <strong>of</strong> the virus (1). The serological analysis <strong>of</strong> the samples,<br />

collected for the surveillance plan, can only be performed by the<br />

network <strong>of</strong> the national <strong>of</strong>ficial laboratories, belonging to the<br />

Isitituti Zoopr<strong>of</strong>ilattici Sperimentali, mostly operating according to<br />

the ISO IEC 17025 and annually participating to pr<strong>of</strong>iciency tests.<br />

In this context, since 2002, the national reference centre for<br />

equine infectious anemia (CRAIE) has conducted inter-laboratory<br />

agid pr<strong>of</strong>iciency tests (2) with an increase in the number <strong>of</strong><br />

participating laboratories.<br />

The aim <strong>of</strong> this paper is to present and discuss the results <strong>of</strong> the<br />

pr<strong>of</strong>iciency tests conducted in Italy during the years 2006-2010<br />

for the diagnosis <strong>of</strong> EIA using the AGID test.<br />

Materials & methods<br />

Over the period 2006-2010, 65 laboratories participated to the<br />

annual pr<strong>of</strong>iciency test, performing both methods prescribed for<br />

AGID for AIE: “the Coggins test” which uses two layers <strong>of</strong> agar<br />

(1.5% and 0.7% respectively and wells <strong>of</strong> 7mm in diameter and 3<br />

mm apart) and the OIE prescribed by the OIE (3). The panel<br />

distributed to each laboratory was each year made up <strong>of</strong> 10 sera,<br />

3 negatives and 7 positives, with different level <strong>of</strong> antibodies<br />

against the P26 antigen (high to weak positives). Only in 2008,<br />

the panel was constituted <strong>of</strong> 2 negative and 8 positive sera.<br />

Before dispatching the samples to each laboratory, they were<br />

subjected to homogeneity and stability tests for their reactivity (2),<br />

so as to correctly evaluate reproducibility and repeatability<br />

parameters. Statistical analysis was carried out using K Cohen<br />

statistic, which was calculated for each laboratory and for all<br />

laboratories gathered together (4).<br />

Results<br />

Of the 65 participating laboratories, 27 constantly had a K value<br />

equal to 1. The results <strong>of</strong> the different laboratories per year are<br />

reported in table 1.<br />

Table 1: K value results per year.<br />

Results <strong>of</strong> the 65 laboratories per year<br />

2006 2007 2008 2009 2010<br />

N. <strong>of</strong> laboratories<br />

0.21-0.40 8 1 2 1 0<br />

0.42-0.60 5 0 0 0 1<br />

0.61-0.80 7 2 0 2 1<br />

0.81-1 45 62 63 62 63<br />

K Value<br />

On the other hand when considering the samples not correctly<br />

identified (K < 1) in both methods for all laboratories, those<br />

mostly misclassified were weak positive, 7.6%, against the 1.4%<br />

<strong>of</strong> the strong positive sera.<br />

When taking into account the concordance <strong>of</strong> samples for the two<br />

different methods, the percentages <strong>of</strong> error for different AGID<br />

methods were respectively 5.2 for the Coggins test and 3.8 for<br />

the OIE method.<br />

The K analysis conducted to evaluate the concordance among<br />

the different laboratories for each year is reported in table 2. The<br />

results using the two different AGID methods are not significantly<br />

different.<br />

Table 2: K value results for all raters per year using both AGID<br />

methods.<br />

K value results among the raters per year<br />

YEAR 2006 2007 2008 2009 2010<br />

AGID Coggins 0.78 0.94 1 0.94 0.95<br />

AGID OIE 0.77 0.98 1 0.97 0.98<br />

Discussion & conclusions<br />

The constant increase <strong>of</strong> participating laboratories to the<br />

pr<strong>of</strong>iciency tests during the years is an important element for<br />

evaluating the efficiency <strong>of</strong> the diagnostic system at national level<br />

for detecting EIA infection. These results stress the importance <strong>of</strong><br />

performing such trials to achieve efficient diagnostic standards for<br />

each <strong>of</strong>ficial laboratory as well as for the national diagnostic<br />

network.<br />

Furthermore, the increase in the number <strong>of</strong> laboratories during<br />

the years, having a K value > 0.8, demonstrates an improvement<br />

<strong>of</strong> the diagnostic performances, as also the achievement <strong>of</strong> good<br />

quality standards, with very satisfactory reproducibility and<br />

repeatability parameters for both AGID tests, even if better for the<br />

AGID OIE. It is important to underline that some difficulties<br />

occurred in the interpretation <strong>of</strong> positive samples with lower<br />

levels <strong>of</strong> antibody against the p26 antigen (weak positive sera).<br />

This represents a problem especially because this error during<br />

routine performance is expected to be higher in consideration <strong>of</strong><br />

an expected higher attention in reading a pr<strong>of</strong>iciency test. This<br />

may constitute a potential bias <strong>of</strong> the results obtained, generating<br />

the calculation <strong>of</strong> a better pr<strong>of</strong>iciency than the effective.<br />

In this respect it our opinion that a more sensible technique such<br />

as the ELISA should be introduced to improve the efficacy and<br />

efficiency <strong>of</strong> the surveillance system. The introduction <strong>of</strong> such a<br />

screening test, with its characteristic <strong>of</strong> high sensitivity, rapidity<br />

and objectiveness is surely a more useful instrument for the<br />

laboratories performing EIA diagnosis, especially within an <strong>of</strong>ficial<br />

surveillance and control programme. Another important issue<br />

emerging from our results is that the laboratories having lower<br />

level <strong>of</strong> concordance were those testing fewer samples in their<br />

diagnostic routine. This further remarks the importance <strong>of</strong> the<br />

experience in the execution and interpretation <strong>of</strong> a test such as<br />

the AGID.<br />

References<br />

1. Ordinanza 8 agosto 2010 Piano di sorveglianza nazionale per l'anemia<br />

infettiva degli equidi. G.U. Serie Generale n. 219 del 18 settembre 2010<br />

2. Guidelines OIE 1998. Guidelines <strong>of</strong> the Office International des<br />

Epizooties for laboratory quality evaluation, for international reference<br />

standards for antibody assays and for laboratory pr<strong>of</strong>iciency testing. Rev.<br />

Sci. Tech. Off. Int. Epiz., 17 (2), 600-609<br />

3. Manual <strong>of</strong> Diagnostic Tests and Vaccines for Terrestrial Animals 2010,<br />

Chapter 2 . 5 . 6 .Equine infectious anaemia<br />

4. Langton S.D., Chevennement R., Nagelkerke N., Lombard B., 2002.<br />

Analysing collaborative trials for qualitative microbiological methods:<br />

accordance and concordance. International Journal <strong>of</strong> Food Microbiology<br />

79: pp. 175-181<br />

5. Charles J. Issel, R. Frank Cook (1993)REVIEW ARTICLE A review <strong>of</strong><br />

techniques for the serologic diagnosis <strong>of</strong> equine infectious anemia J Vet<br />

Diagn Invest 5: 137-141


S1 - P - 11<br />

ANTIMICROBIAL SUSCEPTIBILITY OF AVIBACTERIUM (HAEMOPHILUS) PARAGALLINARUM<br />

ISOLATES RECOVERED IN THE RUSSIAN TERRITORY<br />

A.V. Chernyshov 1 , O.I. Ruchnova 1 , O.P. B’yadovskaya 1<br />

1 FGBI ARRIAH, Vladimir, Russian Federation<br />

Avibacterium paragallinarum, Haemophilus paragallinarum, Infectious Coryza, antimicrobial susceptibility<br />

Introduction<br />

One <strong>of</strong> the most widely spread respiratory infectious diseases<br />

is infectious coryza. The disease is caused by the Gramnegative<br />

bacterium <strong>of</strong> Avibacterium paragallinarum species<br />

previously known as Haemophilus paragallinarum (1, 2, 5).<br />

The disease belongs to highly contagious diseases. It is<br />

manifested by catarrhal inflammation <strong>of</strong> nasal mucosa and air<br />

cells, as well as by the swollen-head-like syndrome and in rare<br />

cases by pneumonia. Chicks and chickens are susceptible to<br />

infectious coryza. Economic losses are caused by the stunting<br />

<strong>of</strong> chicks (up to (10-40%), drop in egg production (up to 41%)<br />

and mortality (up to 10%). (1, 5)<br />

Avian infectious coryza is reported from many countries <strong>of</strong> the<br />

world (2, 5).<br />

Currently, different antibacterial preparations are used in<br />

industrial poultry production but the violation <strong>of</strong> requirements to<br />

their use can lead to the emergence <strong>of</strong> resistant bacterial<br />

strains. Therefore, study <strong>of</strong> susceptibility <strong>of</strong> microorganisms to<br />

antibiotics has a vital significance for the bacterial infection<br />

therapy.<br />

The present work was focused on the study <strong>of</strong> antimicrobial<br />

susceptibility pattern A. paragallinarum field isolates from<br />

poultry from the RF farms.<br />

Materials & methods<br />

Pieces <strong>of</strong> lungs, tracheal and infraorbital sinus washings<br />

received from poultry <strong>of</strong> different age between 2009-2011 were<br />

tested for the presence <strong>of</strong> infectious coryza agent. Thirty-five<br />

A. paragallinarum field isolates were used in the given study.<br />

A. paragallinarum was identified based on the results <strong>of</strong><br />

studying morphological, cultural, biochemical and genetic<br />

properties <strong>of</strong> the isolates (1, 2, 5).<br />

Susceptibility to antimicrobial preparations was determined<br />

using Mueller-Hinton agar supplemented with 5% <strong>of</strong> sheep red<br />

blood cells by Kirby-Bauer disk diffusion test (4) using a kit <strong>of</strong><br />

disks impregnated with different antibiotics (Research Center<br />

<strong>of</strong> Pharmacotherapy, St. Petersburg). The degree <strong>of</strong><br />

resistance <strong>of</strong> the isolates was determined according to the<br />

instruction to the kit.<br />

Results<br />

During 2009-2011 224 samples <strong>of</strong> pathological materials from<br />

broiler chicks and chickens were tested for avian infectious<br />

coryza. Total 35 isolates <strong>of</strong> A. paragallinarum were isolated<br />

from chickens and broiler chicks including 57.1% <strong>of</strong> isolates<br />

from infraorbital sinus swabs, 34.3% - from lung and 8.6% -<br />

from tracheal swabs.<br />

The recovered bacteria possessed tinctorial, morphologic,<br />

cultural and biochemical properties typical <strong>of</strong> A. paragallinarum<br />

and consistent with Bergey’s manual <strong>of</strong> systematic<br />

bacteriology and works by P.J. Blackall [et al.] (1, 2).<br />

Results <strong>of</strong> the study <strong>of</strong> A. paragallinarum susceptibility to<br />

antibacterial preparations are presented in Table 1.<br />

It was established that most isolates <strong>of</strong> A. paragallinarum were<br />

characterized by a wide range <strong>of</strong> resistance to antibacterial<br />

preparations. In particular, to lincosamide group (clindamycin)<br />

(94.5%), tetracycline (tetracycline (82.9%), doxycycline<br />

(87.9%)), co-trimoxazole (trimethoprim) (82.9%), natural<br />

penicillins (benzylpenicillin) (82.9%), third generation<br />

cephalosporins (cefixime) (71.4 %), second generation<br />

fluoroquinolones (<strong>of</strong>loxacin (60 %), cipr<strong>of</strong>loxacin (intermediate<br />

susceptibility 42.9 % <strong>of</strong> isolates), norfloxacin (51.4 %)) and<br />

macrolide group (erythromycin) (62.9 %).<br />

A. paragallinarum isolates had high susceptibility mainly to<br />

inhibitor-protected penicillins (amoxicillin<br />

clavulanate/amoxiclav) – 77.1% <strong>of</strong> strains, first and second<br />

generation aminoglycosides (gentamicin, amikacin<br />

respectively) and nitr<strong>of</strong>uran group (furadonin) – more than 60<br />

%.<br />

Table 2. Antimicrobial susceptibility <strong>of</strong> A, paragallinarum<br />

isolates. Number <strong>of</strong> isolates in % <strong>of</strong> total number. R –<br />

resistant, S – susceptible, I – intermediate susceptibility.<br />

No. Antibiotic name S I R<br />

1 Ofloxacin 34,3 5,7 60,0<br />

2 Cipr<strong>of</strong>loxacin 31,4 42,9 25,7<br />

3 Norfloxacin 34,3 14,3 51,4<br />

4 Lev<strong>of</strong>loxacin 34,3 22,9 42,9<br />

5 Tetracycline 0,0 17,1 82,9<br />

6 Doxycycline 12,1 0,0 87,9<br />

7 Clindamycin 0,0 5,7 94,3<br />

8 Trimethoprim 17,1 0,0 82,9<br />

9 Benzylpenicillin 17,1 0,0 82,9<br />

10 Amoxiclav 77,1 5,7 17,1<br />

11 Ampicillin 28,6 25,7 45,7<br />

12 Laevomycetin 62,9 17,1 20,0<br />

13 Erythromycin 5,7 31,4 62,9<br />

14 Gentamicin 60,0 22,9 17,1<br />

15 Amikacin 65,7 22,9 11,4<br />

16 Furadonin 57,1 25,7 17,1<br />

17 Cefuroxime 48,6 17,1 34,3<br />

18 Cefoperazone 48,6 22,9 28,6<br />

19 Сefixime 11,4 17,1 71,4<br />

Discussion & conclusions<br />

A high rate <strong>of</strong> resistance to erythromycin (62,9%) in our<br />

research according with Yuan-Man Hsu [et. al.] (1) results. He<br />

also marked the resistance <strong>of</strong> the majority <strong>of</strong> tested field strains<br />

to this preparation. Data on tetracycline resistance are similar to<br />

P. J. Blackall (4) investigations.<br />

Our investigation, on the contrary, demonstrated high<br />

percentage <strong>of</strong> isolates resistant to aminopenicillin, erythromycin<br />

and benzylpenicillin in contrast to the results presented by E.S.<br />

Vargas and H.R. Terzolo (10).<br />

In conclusion, A. paragallinarum isolates have wide resistance<br />

range to large spectrum <strong>of</strong> antimicrobials, in particular<br />

tetracyclines (82.9 %), lincosamides (94.3 %), co-trimoxazole<br />

(82.9 %), natural penicillines (82.9 %). The majority <strong>of</strong> bacteria<br />

were susceptible to aminoglycosides (57.1 %), laevomycetin<br />

(62.9 %), inhibitor-protected penicillins (77.1 %) and nitr<strong>of</strong>urans<br />

(57.1 %).<br />

These data on antibiotic resistance should be helpful in<br />

planning strategies for the control <strong>of</strong> A. paragallinarum infection<br />

in poultry.<br />

References<br />

1. Blackall, P.J. (1989) The Avian Haemophili. Clin. Microbiology<br />

Reviews. Vol.2(3). P. 270-277.<br />

2. Blackall, P.J. (1999) Infectious Coryza: Overview <strong>of</strong> the disease and<br />

new diagnostic options. Clin. Microbiology Reviews. Vol. 12, №4. P. 627-<br />

632.<br />

3. Hsu, Y.-M., Shieh, H.K., Chen, W.-H. [et al.] (2007) Antimicrobial<br />

susceptibility, plasmid pr<strong>of</strong>iles and haemocin activities <strong>of</strong> Avibacterium<br />

paragallinarum strains. Vet. Microbiology. Vol.124. P. 209–218.<br />

4. Jorgensen, J.H., Turnidge, J.D. (2007) Susceptibility test methods:<br />

dilution and disk diffusion methods. In P. R. Murray, E. J. Baron, J. H.<br />

Jorgensen, M. L. Landry, and M. A. Pfaller (ed.), Manual <strong>of</strong> clinical<br />

microbiology, 9th ed. ASM Press, Washington, D.C. P. 1152–1172.<br />

5. Vargas, E.S., Terzolo, H.R. (2004) Haemophilus paragallinarum:<br />

Etiology <strong>of</strong> infectious coryza. Vet. Mex. Vol. 35, №3. P. 245–259.


S1 - P - 12<br />

COMPARISON BETWEEN ANALYTICAL SENSITIVITY OF RABIES VIRUS ISOLATION IN<br />

NEUROBLASTOMA CELL CULTURE WITH ANALYTICAL SENSITIVITY OF MOUSE INOCULATION<br />

TEST<br />

E.V. Chernyshova 1 , N.A. Nazarov 1 , A.E. Metlin 1<br />

1 FGBI "Federal Centre for Animal Health"(FGBI "ARRIAH"), Vladimir, Russian FederationRabies,<br />

diagnosis, virus isolation in neuroblastoma cell culture, mouse inoculation test, analytical sensitivity<br />

Introduction<br />

Rabies is an acute viral zoonotic infection characterized by<br />

lesions <strong>of</strong> the central nervous system impairment which almost<br />

always results in death.<br />

The main scheme <strong>of</strong> rabies laboratory diagnosis in animals<br />

adopted in the Russian Federation (RF) (State Standard 26075-<br />

84) consists <strong>of</strong> pathological material testing (brain tissue) using<br />

fluorescent antibody test (FAT). When FAT is negative mouse<br />

inoculation test (MIT) should be carried out (4).<br />

In laboratories with appropriate conditions and staff competence<br />

the OIE recommends using virus isolation in neuroblastoma cell<br />

culture instead <strong>of</strong> MIT (2). The reasons for such a<br />

recommendation are the following:<br />

Both methods are <strong>of</strong> high diagnostic sensitivity and specificity;<br />

Minimum and maximum time for diagnosis using virus isolation in<br />

neuroblastoma cell culture is from 4 to 12 days respectively and<br />

MIT performance takes from 12 to 30 days in average;<br />

It is necessary to confirm the results <strong>of</strong> MIT using FAT in addition;<br />

Virus isolation in neuroblastoma cell culture is more humane and<br />

less cost-consuming.<br />

In the EU countries when FAT is negative the virus isolation in<br />

cell culture is performed only in case <strong>of</strong> animal-human contact<br />

(France, Germany). In some countries (Lithuania) in case <strong>of</strong><br />

absence <strong>of</strong> such contacts pathological materials are just<br />

passaged once in cell culture.<br />

Currently, many regional veterinary laboratories <strong>of</strong> the RF are<br />

capable <strong>of</strong> running diagnostic tests using cultural methods.<br />

Introduction the “Methodical instructions on rabies virus isolation<br />

in mouse neuroblastoma cell culture” into veterinary laboratory<br />

practice in the RF is <strong>of</strong> great importance.<br />

The study is aimed at the comparison <strong>of</strong> analytical sensitivity<br />

between methods <strong>of</strong> rabies virus isolation in cell culture and<br />

rabies virus isolation in white mice (1, 3).<br />

neuroblastoma cell culture. As for our experiments, the difference<br />

was 1 lg10.<br />

Thus, our results provide one more basis to replace a<br />

MIT the method <strong>of</strong> rabies virus isolation in mouse neuroblastoma<br />

cell culture in laboratories and research institutions dealing with<br />

rabies diagnosis.<br />

References<br />

1. Anthony R. Fooks. New Diagnostic Tools for Rabies in Animals (2009)<br />

http://www.oie.int/eng/A_RABIES/presentations_rage/S21%20NewDiagno<br />

sticTools_Pr<strong>of</strong>Fooks.pdf 40 p.<br />

2. OIE. Manual <strong>of</strong> Diagnostic Tests and Vaccines for Terrestrial Animals.<br />

Vol. 1. – 6nd ed. - Paris, (2008). - Chap. 2.1.13. - p. 304-323.<br />

3. Rabies General Aspects &Laboratory DiagnosticTechniques (2007)<br />

http://www.whoindia.org/LinkFiles/Communicable_Diseases_Rabies_-<br />

General_Aspects_&_Laboratory_Diagnostic_Techniques.pdf 72 p.<br />

4. State Standard 26075-84 Methods <strong>of</strong> laboratory diagnosis for rabies<br />

(1984), 9 p.<br />

Materials & methods<br />

The following materials were used for the study: mouse<br />

neuroblastoma N-2a cell line; white outbred mice (6-8 g); mouse<br />

brain-adapted rabies virus (CVS strain; 5.66 lg TCID 50 /ml); antirabies<br />

fluorescent immunoglobulin produced by the FGBI<br />

“ARRIAH”.<br />

Analytical sensitivities <strong>of</strong> the methods were determined by<br />

titration <strong>of</strong> rabies virus CVS strain in mouse neuroblastoma N-2a<br />

cell line and in white mice.<br />

The titre was expressed as the highest virus dilution at<br />

which specific fluorescence in cell culture or deaths <strong>of</strong> mice were<br />

observed.<br />

Specific deaths <strong>of</strong> mice were confirmed by FAT.<br />

Results<br />

A 10% suspension <strong>of</strong> original mouse-brain-adapted CVS strain <strong>of</strong><br />

rabies virus was prepared in RPMI medium for inoculation.<br />

Rabies virus was preliminary titrated using a series <strong>of</strong> tenfold<br />

dilutions <strong>of</strong> the prepared suspension. The highest dilution <strong>of</strong> the<br />

virus suspension with the specific fluorescence corresponded to<br />

1.66 lg TCID50/ml titer.<br />

In order to determine sensitivity more accurately five tw<strong>of</strong>old<br />

dilutions <strong>of</strong> the virus suspension with 1.66 lg TCID50/ml titer were<br />

prepared. Test results demonstrated that the last dilution with the<br />

specific fluorescence corresponded to 1.358 TCID50/ml titer.<br />

Seven tw<strong>of</strong>old dilutions <strong>of</strong> the virus suspension with 2.66<br />

TCID50/ml titer were prepared to determine sensitivity <strong>of</strong> the MIT.<br />

Test results demonstrated that the last dilution accompanied by<br />

specific deaths in mice corresponded to 2.358 TCID50/ml titer.<br />

Discussion & conclusions<br />

A conclusion was made after comparison <strong>of</strong> the obtained results<br />

that the MIT had a lower analytical sensitivity in comparison to<br />

analytical sensitivity <strong>of</strong> rabies virus isolation in mouse


S1 - P - 13<br />

IDENTIFICATION OF GLOBICATELLA SANGUINIS ISOLATED FROM PNEUMONIA IN A GOAT<br />

M. Cocchi , S. Deotto, T. Di Giusto, M. Bregoli<br />

Istituto Zoopr<strong>of</strong>ilattico Sperimentale delle Venezie, Sezione territoriale di Udine, Basaldella di Camp<strong>of</strong>ormido (UD), Italy<br />

Globicatella sanguinis, pneumonia, goat<br />

Introduction<br />

Globicatella (G.) sanguinis was described in 1992. This<br />

bacterium is a catalase-negative, facultative anaerobic, gram<br />

positive coccus. It has been associated with various diseases in<br />

humans, such as bacteraemia, meningitis and infections <strong>of</strong> the<br />

urinary tract. In veterinary medicine, G. sanguinis was described<br />

in meningoencephalitis in lambs (1, 3). The aim <strong>of</strong> this study was<br />

to describe an episode <strong>of</strong> pneumonia in a goat produced by G.<br />

sanguinis.<br />

Materials & methods<br />

Two 1-year-old Tibetan goats out <strong>of</strong> a total <strong>of</strong> four goats<br />

belonging to a farm located in Friuli Venezia Giulia region (North<br />

east <strong>of</strong> Italy) showed unspecific clinical signs such as anorexia,<br />

depression, and permanent decubitus. These goats died after<br />

four days by the onset <strong>of</strong> the symptoms, and one <strong>of</strong> them was<br />

submitted for a necropsy. Bacteriological examinations were<br />

performed from lungs, brain and liver as follows: swabs were<br />

streaked onto blood agar base (BAB) and eosin-methylene blue<br />

agar (EMB). Brain Heart Infusion broth (BHI) was inoculated, too.<br />

Plates and tubes were incubated at 371°C for 48 hrs, in<br />

aerobiosis. BAB was incubated in anaerobic condition, too. From<br />

intestine a Perfringens agar base (PAB) other than EMB plate<br />

was used, incubated in anaerobiosis. It was obtained by using<br />

ANAEROGEN System, with an anaerobic indicator (Oxoid).<br />

Depending on morphology <strong>of</strong> the colonies, Gram staining,<br />

catalase and oxidase tests were performed. Miniaturized<br />

commercial tests were also used for identification. Agar plates<br />

and tubes were supplied by the service for media <strong>of</strong> the Istituto<br />

Zoopr<strong>of</strong>ilattico Sperimentale delle Venezie. Antimicrobial test was<br />

performed with disk diffusion method, in accordance with Clinical<br />

and Laboratory Standards Institute (CLSI) guidelines (2). Tested<br />

antimicrobials were: penicillin (10 g, Becton Dickinson),<br />

ampicillin (10 g, Becton Dickinson), oxacillin (10 g, Becton<br />

Dickinson), tetracycline (30 g, Oxoid), sulfamethoxazoletrimethropim<br />

(23,75-1,25 g –Oxoid-) and cephalothin (30 g –<br />

Oxoid-). Reading was done using the established diameter for<br />

Streptococcus (S.) spp other than S. pneumoniae.<br />

Results<br />

Necropsy. Macroscopic lesions recovered from lungs referred to<br />

acute bronchopneumonia and tracheal and laryngeal oedema.<br />

Catarrhal enteritis and hepatomegaly were observed, too.<br />

Bacteriological tests. Cultures from lungs and liver revealed pure<br />

culture <strong>of</strong> -haemolytic, pin-pointed (2-3 mm <strong>of</strong> diameter), circular<br />

colonies. Subsequently, analyses revealed a gram positive,<br />

catalase-negative, coccus-shaped organism.<br />

Figure 1 shows the morphology <strong>of</strong> the bacterium after 24 hrs <strong>of</strong><br />

incubation, on BAB. Biochemical identification was performed<br />

with a commercial kit, API rapid ID 32 Strep (Biomerieux). Both<br />

the gram positive strains showed identical biochemical pr<strong>of</strong>iles<br />

(22274063110), with a percentage <strong>of</strong> identification <strong>of</strong> 99.9%,<br />

T=0.92. Only fermentation <strong>of</strong> ribose resulted as an atypical test<br />

(negative in our reaction). Cultures from the intestine showed E.<br />

coli and Clostridium perfringens.<br />

Antimicrobial test. G. sanguinis revealed resistance to oxacillin,<br />

and to tetracycline, while it was sensitive to other tested drugs.<br />

Discussion & conclusions<br />

Pneumonia is one <strong>of</strong> the most common respiratory problems in<br />

small ruminants throughout the world. In goat herds, pneumonia<br />

increases production costs associated with expensive treatments.<br />

Infectious and non-infectious agents can cause inflammation <strong>of</strong><br />

the lungs. In goats the most frequent causes <strong>of</strong> respiratory<br />

infection and death are Pasteurella multocida and Mannheimia<br />

haemolytica. They are commonly found in the upper respiratory<br />

tract <strong>of</strong> healthy goats, too. Transportation stress, viral infections,<br />

parasites, poor management conditions, increase goats'<br />

susceptibility to these agents <strong>of</strong> pneumonia. In our case, the<br />

identification <strong>of</strong> G. sanguinis in pure culture from diseased lungs<br />

could be indicative <strong>of</strong> the pathological role <strong>of</strong> this bacterium.<br />

The performed analysis on other organs revealed the presence <strong>of</strong><br />

G. sanguinis in liver, too. Differences are in reference to the<br />

growth in different atmospheres: culture from lungs showed G.<br />

sanguinis both in aerobic and in anaerobic conditions. Cultures<br />

from liver revealed the presence <strong>of</strong> the bacterium only in aerobic<br />

incubation. The fact that all the goats in the farm showed identical<br />

clinical signs is strongly indicative that this bacterium was also<br />

responsible for the pathology in the other goats.<br />

In veterinary medicine, G. sanguinis was isolated in a case <strong>of</strong><br />

suppurative meningoencephalitis in lambs (1). In our case<br />

macroscopic analysis revealed an acute bronchopneumonia. Any<br />

lesion was recovered from the brain.<br />

Regarding identification, in this paper we named the bacterium in<br />

accordance with the pr<strong>of</strong>ile given by a miniaturized system. The<br />

phenotypic findings are consistent with those described for this<br />

bacterium species, except for pyrrolidonil arylamidase test. It is<br />

pertinent to note that G. sanguinis belongs to a broad range <strong>of</strong><br />

Gram-positive, catalase-negative new taxa recently founded in<br />

human and animal clinical samples. Some authors revealed that<br />

misidentification can occur between Aerococcus viridans and G.<br />

sanguinis, because <strong>of</strong> very similar biochemical pr<strong>of</strong>iles (1,5).<br />

Moreover, looking into literature only a few number <strong>of</strong> isolates<br />

were tested, with some differences in the results (1,3). For these<br />

reasons, subsequent investigations using different methods, such<br />

as molecular genetic tools should accompany the identification <strong>of</strong><br />

this bacterium.<br />

The improvement <strong>of</strong> the identification systems <strong>of</strong> these unusual<br />

gram-positive, catalase-negative bacteria may contribute to<br />

making progress in current epidemiological knowledge on host<br />

distribution and range <strong>of</strong> clinical conditions.<br />

Data about antimicrobial susceptibility test showed resistance<br />

varied significantly among the tested strains (5). In our report<br />

resistance to tetracycline does not agree with the data <strong>of</strong><br />

Seegmuller et al (4).<br />

To the author’s knowledge this is the first report about G.<br />

sanguinis in an episode <strong>of</strong> pneumonia in a goat.<br />

References<br />

1. Vela, A.I., Fernandez E., Las Heras A., Lawson, P.A., Dominguez L.,<br />

Collins M.D., and Fernandez-Garayzabal J. (2000). Meningoencephalitis<br />

associated with Globicatella sanguinis infection in lambs. J Clin Microbiol<br />

vol 38, 4254-4255.<br />

2. Clinical and laboratory standards institute (CLSI). M31-A3. Performance<br />

standards for antimicrobial disk and dilution susceptibility tests for bacteria<br />

isolated from animals. Vol 28. n 8.<br />

3. Collins, M.D., Aguirre, M., Faclam, R.R., Shallcross J., and Williams<br />

A.M. (1992) Globicatella sanguis gen.nov., sp. Nov., a gram-positive<br />

catalase-negative bacterium fropm human sources. J Appl. Bacteriol.<br />

73:433-437.<br />

4. Seegmuller I., Van der Linden M., Heeg C., and Reinert R.R. (2007).<br />

Globicatella sanguinis is ana etiological agent <strong>of</strong> ventriculoperitoneal<br />

shunt-associated meningitis. J Clinical Microbiol. Febb. 666-667.<br />

5. Shewmaker, P.L., Steigerwalt, A.G., Shealey, L., Weyant, R., and<br />

Facklam, R. (2001). DNA relatedness, phenotypic characteristics, and<br />

antimicrobial susceptibilities <strong>of</strong> Globicatella sanguinis strains. J Clinic<br />

Microbiol. Nov 4052-4057.<br />

Figure 1: G. sanguinis after 24h incubation on blood agar base, in<br />

aerobic atmosphere


S1 - P - 14<br />

KAIZEN PROCESS APPLIED AT THE DIAGNOSTIC SERVICE OF FACULTÉ DE MÉDICINE<br />

VÉTÉRINAIRE, UNIVERSITÉ DE MONTREAL<br />

Estela Cornaglia, Véronique Boyer, Jean-François Brodeur, Guy Fontaine<br />

Université de Montréal, Faculté de médicine vétérinaire, Diagnostic Service, Saint-Hyacinthe, Canada<br />

Continuous improvement, Kaizen<br />

Introduction<br />

The diagnostic service (SD) at the Faculté de medicine<br />

vétérinaire de l’Université de Montréal, include 20 laboratories or<br />

services unities. First line laboratories as bacteriology, virology,<br />

parasitology, clinical pathology, pathology, molecular diagnosis,<br />

serology; and specialized unites as ichtyopathology, food safety,<br />

genomics, wildlife, OIE reference EcL laboratory, Actinobacillus<br />

pleuropneumoniae serology and serotyping.<br />

Our staff is around 90 people, 26 veterinarians and 14<br />

pr<strong>of</strong>essionals, among them 19 PhD and 13 graduated ACVP.<br />

Continuous improvement and accreditations are a priority for us<br />

to assure the quality <strong>of</strong> our service. We’ve chosen Kaizen as a<br />

principal tool to realize them.<br />

Kaizen means "continuous improvement" or "change for the<br />

better". It refers to philosophy or practices that focus upon<br />

continuous improvement <strong>of</strong> processes in manufacturing,<br />

engineering, supporting business processes, and management.<br />

It comes from Japan.<br />

Improvement can be broken down between innovation and<br />

Kaizen. Innovation involves a drastic improvement in the<br />

existing process and requires large investments. Kaizen<br />

signifies small improvements as a result <strong>of</strong> coordinated<br />

continuous efforts by all employees. Management works<br />

continuously towards revising the current standards, once they<br />

have been mastered, and establishing higher ones. Kaizen<br />

methodology includes making changes and monitoring results,<br />

then adjusting. Large-scale pre-planning and extensive project<br />

scheduling are replaced by smaller experiments, which can be<br />

rapidly adapted as new improvements are suggested.<br />

Kaizen is a process-oriented thinking versus a result-oriented<br />

thinking. Kaizen concentrates on improving the process rather<br />

than on achieving certain results.<br />

Materials & methods<br />

The following Kaizen’s methodologies and concepts were<br />

applied:<br />

- 5S activity. 5S is the name <strong>of</strong> a methodology that uses a list<br />

<strong>of</strong> 5 Japanese words which are seiri, seiton, seiso, seiketsu<br />

and shitsuke. In English: sorting, straightening, systematic<br />

cleaning, standardizing, and sustaining<br />

- Activities based on teamwork. It involved technicians, support<br />

staff, quality assurance manager, pr<strong>of</strong>essionals and directors<br />

- This small multidisciplinary group worked in improving their<br />

own work environment and productivity.<br />

- This group was guided through the kaizen process by an<br />

external consultant.<br />

- This group prepared a Value Stream Mapping to select<br />

specific areas <strong>of</strong> improvement that would be addressed<br />

through Kaizen events and “just-do-it” activities.<br />

- The consultant facilitated an ideation/brainstorming processes<br />

to identify improvement options and obtained participant<br />

feedback.<br />

- The group reported Kaizen results to all employees and<br />

celebrated success<br />

Figure 1: Workplace before Kaizen<br />

Figure 1: Workplace after Kaizen<br />

Results<br />

Kaizen increased employee satisfaction by added-value activities<br />

and improvement <strong>of</strong> the work environment).<br />

By improving standardized activities and processes, kaizen<br />

eliminated waste (lean manufacturing).<br />

Kaizen process identified expected measurable improvements.<br />

It prepared an action plan with a list <strong>of</strong> activities required to<br />

realize those improvements.<br />

Discussion & conclusions<br />

Kaizen process allowed improving work environment and working<br />

distribution. It helped to eliminate waste and to recover time for<br />

value added activities.<br />

References<br />

1. Imai, M (2007). Gemba Kaizen, L’art de manager avec bon sens.


S1 - P - 15<br />

ACCURACY OF PARATUBERCULOSIS ANTIBODY ELISA AT PREDICTING FECAL SHEDDING OF<br />

MYCOBACTERIUM AVIUM SUBSP PARATUBERCULOSIS IN CATTLE<br />

Rakel T. Fernández 1 , Carmen Eiras 2 , Carmen Calvo 2 , Ignacio Arnaiz 2 , Fco. Javier Diéguez 1,3<br />

1<br />

Santiago de Compostela University (Veterinary Faculty), Department <strong>of</strong> Anatomy and Animal Production, Lugo, Spain<br />

2<br />

Animal Health and Production Laboratory, Lugo, Spain<br />

3<br />

Santiago de Compostela University (Veterinary Faculty), Institute <strong>of</strong> Food Analysis and Research, Lugo, Spain<br />

Paratuberculosis, bovine, ELISA, fecal culture<br />

Introduction<br />

Paratuberculosis, also called Johne's disease, is chronic<br />

granulomatous enteritis caused by Mycobacterium avium subsp.<br />

paratuberculosis (MAP). This infectious disease has a worldwide<br />

distribution affecting mainly ruminants, both domestic and wild,<br />

but has also been isolated from other animal species. It causes<br />

great economic losses in cattle farming 1 .<br />

ELISA is an essential tool in paratuberculosis control programs,<br />

although bacterial culture remains the reference confirmatory<br />

test. In this line, the aim <strong>of</strong> this study was to determine the<br />

accuracy <strong>of</strong> the ELISA test as a predictor <strong>of</strong> the MAP fecal<br />

elimination "status" (determined by means <strong>of</strong> fecal culture) <strong>of</strong><br />

individual animals.<br />

Materials & methods<br />

The study was carried out in Galicia (north-west Spain). Galicia is<br />

the major cattle-farming region <strong>of</strong> Spain. It was responsible for<br />

35% <strong>of</strong> the milk and 12% <strong>of</strong> the beef produced in Spain,<br />

constituting approximately 1.7% <strong>of</strong> the milk and 1.3% <strong>of</strong> the beef<br />

produced in the European Union.<br />

Bacteriological culture was performed in fecal samples from 239<br />

animals that were collected during the period 2007-2010. 181<br />

animals were negative to fecal culture and 58 were culture<br />

positive animals. Serological analysis (ELISA) was also<br />

performed in serum samples from the same animals.<br />

Bacterial culture was performed as described by the World<br />

Organization for Animal Health (OIE) (2008) 2 . Briefly, 1 g <strong>of</strong> feces<br />

was added to 20 mL <strong>of</strong> sterile distilled water, and tubes were<br />

shaken for 30 min and then allowed to stand undisturbed for 30<br />

min. Five millilitres <strong>of</strong> the supernatant were added to 0.75%<br />

hexadecylpyridinium chloride (HPC) (Sigma). Tubes were<br />

inverted several times and allowed to stand undisturbed for 18 h<br />

at room temperature for decontamination. Triplicate Herrold’s egg<br />

yolk medium (HEYM) culture slopes containing amphotericin B,<br />

vancomycin and nalidixic acid were inoculated with 0.1 mL <strong>of</strong> the<br />

undisturbed sediment, incubated at 37º C and observed at 2-<br />

week intervals for 16 weeks. Suspect colonies were evaluated for<br />

mycobactin dependence along with morphology and acid-fast<br />

staining. Positive-staining colonies were confirmed by PCR. For<br />

every nine fecal samples a positive control (a positive field<br />

sample) was included.<br />

The ELISA used was “PARATUBERCULOSIS ANTIBODY<br />

SCREENING” (Institute Pourquier, France). False-positive results<br />

were reduced by pre-absorbing the samples with sonicates <strong>of</strong> the<br />

environmental mycobacterium Mycobacterium phlei. Samples<br />

were considered positive at a % sample:positive ratio <strong>of</strong> 55% or<br />

more.<br />

Receiver operating characteristic (ROC) procedure was used to<br />

evaluate the overall diagnostic accuracy to estimate the antibody<br />

titer that was the best cut-<strong>of</strong>f point in terms <strong>of</strong> sensitivity and<br />

specificity as a predictor <strong>of</strong> the bacteriological "status" <strong>of</strong> the<br />

animals (culture positive/negative).<br />

Results<br />

The ROC curve -as measure <strong>of</strong> the accuracy <strong>of</strong> ELISA antibody<br />

titers to predict the bacteriological “status”- indicated a good<br />

diagnostic discrimination (Figure 1). The area under the curve<br />

was 0.878. The best overall diagnostic precision was in the cut<strong>of</strong>f<br />

point 155.7 with sensitivity <strong>of</strong> 84.5% and specificity <strong>of</strong> 78.9%<br />

(Table 1).<br />

Figure 1: ROC curve for overall accuracy <strong>of</strong> antibody titers as a<br />

predictor <strong>of</strong> MAP bacteriological “status”<br />

Table 1: Sensitivities and specificities obtained from ROC<br />

procedure<br />

Cut <strong>of</strong>f point Sensitivity 1-Specificity<br />

71.4 1.00 0.81<br />

133.1 0.91 0.34<br />

147.2 0.88 0.26<br />

155.7 0.84 0.22<br />

160.1 0.83 0.20<br />

164.2 0.81 0.18<br />

167.7 0.78 0.17<br />

309.2 0.00 0.01<br />

Discussion & conclusions<br />

Traditionally the interpretation <strong>of</strong> ELISA is dichotomous (positive<br />

or negative) based on the cut-<strong>of</strong>f designed to optimize the<br />

sensitivity and specificity to detect animals with antibodies. The<br />

use <strong>of</strong> antibody titers on a continuous scale can improve the<br />

amount <strong>of</strong> diagnostic information obtained by this technique. The<br />

ELISA evaluated in this study, as is the case <strong>of</strong> previous studies<br />

with different ELISAs 3 could allow a more accurate estimate <strong>of</strong><br />

the situation <strong>of</strong> the animal, in this case related to the<br />

bacteriological “status”.<br />

References<br />

1. Kennedy, DJ, Benedictus, G (2001). Control <strong>of</strong> Mycobacterium avium<br />

subsp. paratuberculosis infection in agricultural species. Scientific and<br />

Technical Reviews, 20, 151-79.<br />

2. World Organization for Animal Health (OIE) (2008). Manual <strong>of</strong> diagnostic<br />

test & vaccines for terrestrial animals, Chapter 2.1.11.<br />

3. Collins, MT (2002). Interpretation <strong>of</strong> a commercial bovine<br />

paratuberculosis enzyme-linked immunosorbent assay by using likelihood<br />

ratios. Clinical and Diagnostic Laboratory Immunology, 9, 1367-71.


S1 - P - 16<br />

EVALUATION OF AN ELISA ASSAY AS AN ANCILLARY METHOD IN A HERD WITH A NATURAL<br />

TUBERCULOSIS INFECTION<br />

I. Arnaiz 1 , C. Eiras 1 , M.L. Luaces 2 , J. Fraga 3 , M. Rodríguez 3 , C. Calvo 1<br />

1 Laboratorio de Sanidade e Produción Animal de Galicia, Consellería do Medio Rural e do Mar, Xunta de Galicia, Lugo, España<br />

2<br />

Área Veterinaria de Lugo, Servicio Provincial de Ganadería de Lugo, Consellería do Medio Rural e do Mar, Xunta de Galicia, Lugo, España<br />

3<br />

Área Veterinaria de Vilalba, Servicio Provincial de Ganadería de Lugo, Consellería do Medio Rural e do Mar, Xunta de Galicia, Lugo, España<br />

Keyword: Tuberculosis, ELISA, IFN-g, SIT, isolation<br />

Introduction<br />

The single intradermal tuberculin (SIT) test and the Interferongamma<br />

assay (IFN-g) are the tuberculosis diagnostic tools in<br />

the tuberculosis eradication program currently used in Spain.<br />

These tests based on cell-mediated response can detect<br />

animals in the early stages <strong>of</strong> infection (1), although not all<br />

affected animals are detected with these tests. Humoral<br />

immune response is found in cattle with natural infection and<br />

this response could be detected by the use <strong>of</strong> an ELISA assay.<br />

ELISA testing is not routinely used in bovine tuberculosis<br />

control programs mainly due to a reduced sensitivity (2)<br />

although it has been suggested that it might be used as a<br />

complement to the tuberculin test, especially for the detection<br />

<strong>of</strong> anergic tuberculous cattle (3).<br />

The aim <strong>of</strong> this study is the evaluation <strong>of</strong> the use <strong>of</strong> SIT test<br />

and the IFN-g assay together with an ELISA assay in order to<br />

obtain the most complete detection <strong>of</strong> tuberculosis infected<br />

animals in a herd previously confirmed to have tuberculosis.<br />

Material & methods<br />

314 animals from a tuberculosis infected beef herd from<br />

Galicia, in north-west Spain, were tested using the SIT test.<br />

The IFN-g detection assay (BovigamTM, Prionics AG,<br />

Switzerland) was used in parallel with the SIT test. The 16<br />

positive SIT animals (<strong>of</strong> which 15 animals were also detected<br />

by IFN-g assay) and 17 cattle that were only detected by IFN-g<br />

assay were slaughtered and their tissues were collected for<br />

Mycobacterium tuberculosis complex (MTC) isolation. At the<br />

same time, 20 cows older than 10 years were slaughtered too.<br />

Tissues from the 53 slaughtered animals were inoculated into<br />

the selective media (4). The MTC was recovered and<br />

confirmed by real-time PCR (5) in 18 cattle.<br />

The plasma collected for the IFN-g assay was also used on<br />

the ELISA assay for M. bovis antibody (IDEXX Laboratories).<br />

18 cattle were detected with the ELISA assay.<br />

The agreement and kappa statistic measurements were<br />

calculated by comparing the MTC isolation with SIT test, IFN-g<br />

and ELISA assays respectively.<br />

References<br />

1. Rua-Domenech R, Goodchild AT, Vordermeier HM, Hewinson RG,<br />

Christiansen KH, Clifton-Hadley RS: Ante mortem diagnosis <strong>of</strong><br />

tuberculosis in cattle: a review <strong>of</strong> the tuberculin tests, gamma-interferon<br />

assay and other ancillary diagnostic techniques. Res Vet Sci 2006,<br />

81:190-210.<br />

2.Ritacco V, Lopez B, Barrera L, Nader A, Fliess E, de Kantor: Further<br />

evaluation <strong>of</strong> an indirect enzyme-linked immunosorbent assay for the<br />

diagnosis <strong>of</strong> bovine tuberculosis. Zentralbl Veterinarmed B 1990, 37:19-<br />

27.<br />

3. Plackett P, Ripper J, Corner LA, Small K, de Witte K, Melville L,<br />

Hides S, Wood PR: An ELISA for the detection <strong>of</strong> anergic tuberculous<br />

cattle. Aus Vet J 1989, 66:15-19.<br />

4. Idigoras P., Beristain X., Iturzaeta A., Vicente D., Pérez-Trallero E.<br />

2000. Comparison <strong>of</strong> the automated nonradiometric BACTEC MGIT<br />

960 system with Löwestein-Jensen, Coletsos, and Middlebrook 7H11<br />

solid media for recovery <strong>of</strong> mycobacteria. European Journal <strong>of</strong> Clinical<br />

Microbiology Infections Diseases 19: 350-354.<br />

5. Huard R.C., Oliveira-Lazzarini L.C., Butler W.R., van Soolingen D.,<br />

Ho J.L. 2003. PCR-Basaed method to differentiate the subspecies <strong>of</strong><br />

the Mycobacterium tuberculosis complex on the basis <strong>of</strong> genomic<br />

deletions. Journal <strong>of</strong> Clinical Microbiology, vol 41, n 4: 1637-1650.<br />

Results<br />

10 bovine with MTC recovered were SIT test positive. 16 were<br />

positive in the IFN-g assay and the ELISA detected 13 <strong>of</strong> the<br />

18 cattle with MTC recovered (one <strong>of</strong> them was negative in<br />

SIT test and IFN-g assay). One bovine with MTC recovered<br />

was negative in the three tests; this animal was slaughtered<br />

because it was more than10 years old.<br />

The kappa statistic shows a lower agreement for the SIT test<br />

(0.39) and the IFN-g assay (0.36) with the MTC isolation than<br />

the ELISA assay (0.58).<br />

Discussion & conclusions<br />

The results suggest that the use <strong>of</strong> IFN-g and ELISA assays<br />

as ancillary techniques might help to detect a larger number <strong>of</strong><br />

tuberculosis infected animals in a TB-confirmed herd. These<br />

assays complement each other because they may detect<br />

animals in different stages <strong>of</strong> tuberculosis.<br />

Acknowledgements<br />

We thanks IDEXX Laboratories, Inc., for providing the ELISA<br />

test for detection <strong>of</strong> M. bovis antibodies. We also would like to<br />

thank the Official Veterinary Services from Vilalba for assisting<br />

with abattoir sampling, the Regional Animal Health Veterinary<br />

Services for their support and the Laboratory <strong>of</strong> Animal Health<br />

<strong>of</strong> Galicia.


S1 - P - 17<br />

VIROLOGICAL SURVEILLANCE AND CHARACTERIZATION OF AVIAN-LIKE REASSORTANT H1N2<br />

SWINE INFLUENZA VIRUSES IN SWEDEN<br />

Giorgi Metreveli , Eva Emmoth, Lena Renström<br />

National Veterinary Institute, Department <strong>of</strong> Virology, Immunobiology and Parasitology, Uppsala, Sweden<br />

Introduction<br />

The influenza A virus subtypes H1N1, H1N2 and H3N2 are<br />

prevalent in pig populations worldwide. In 2009 and 2010 there<br />

was the first reported isolations and demonstrations <strong>of</strong> natural<br />

reassortants <strong>of</strong> H1N2 viruses in pigs in Sweden (1,2).<br />

Characterized Swedish isolates possessed avian-like SIV H1N1<br />

HA and European H3N2 SIV-like NA. They were compared<br />

regarding their molecular composition and biological<br />

characteristics.<br />

Materials & methods<br />

Virus was isolated from the clinical material by infecting Madin<br />

Darby Canine Kidney (MDCK) (ATCC CCL-34) cells, following<br />

standard cell culture procedures. Viruses were analyzed by endpoint<br />

titration through cpe using 96-well plates containing MDCK<br />

cells, in 10-fold dilutions assaying eight replicates <strong>of</strong> 50 µl per<br />

dilution, and the virus titres after 6-8 days were calculated<br />

according to Kärber (3). NA enzyme activity and drug inhibition<br />

assays were performed with methylumbelliferone<br />

nacetylneuraminic acid (MUNANA) as the substrate. Genetic<br />

analyses were conducted on the clinical material and on the<br />

MDCK cell isolates. Total RNA was prepared from virus-infected<br />

MDCK cells by the QiagenRNeasy Mini kit, according to the<br />

manufacturer’s instructions (Qiagen, Hilden Germany).<br />

Sequencing templates were generated by amplification <strong>of</strong> each<br />

gene segment using one-step RT-PCR (QIAGEN OneStep RT-<br />

PCR Kit). The PCR products were purified using the ‘Purification<br />

Kit from Promega’ and sequenced using the fluorescent dye<br />

terminator method with an ABI PRISM® Big Dye Terminator<br />

Cycle Sequencing v3.1 Ready Reaction kit (Perkin Elmer,<br />

Waltham, MA, USA) on an ABI PRISM® 310 genetic analyzer<br />

according to the manufacturer’s recommendations (Applied<br />

Biosystems).Sequence data analyses, multiple alignments <strong>of</strong> the<br />

DNA sequences <strong>of</strong> each gene, were performed using CLC, Main<br />

Workbench 5.0.2 (CLC bio A/S, Aarhus, Denmark).<br />

Results<br />

The most remarkable result was a truncated coding region for<br />

PB1-F2 in the earlier isolates and a full length coding region in<br />

the more recent isolates. Concerning biological properties, these<br />

viruses reached lower titre and showed reduced<br />

cytopathogenicity in MDCK cells compared with an avian-like<br />

H1N1 isolate A/swine/Lidkoping/1193/2002 belonging to the<br />

same lineage as the 2009 and 2010 isolates.<br />

Figure 1: Always put legend below the figure<br />

Discussion & conclusions<br />

Recent serological investigations in Swedish pig population has<br />

also shown that this uncommon avian-like reassortant H1N2 SIV<br />

variant appears to be gaining a stronger foothold among Swedish<br />

pig populations and producing more clinical disease. The<br />

molecular genomic differences found here indicate that the virus<br />

population is steadily evolving. In order to determine the effect on<br />

the swine industry and influenza ecology, further surveillance<br />

investigations and detailed analyses are needed.<br />

References<br />

1 Bálint, A., et al., The first Swedish H1N2 swine influenza virus isolate represents<br />

an uncommon reassortant. Virology Journal, 2009. 6 Oct 28(180).<br />

2. Metreveli, G., et al. Comparison <strong>of</strong> two H1N2 swine influenza A viruses from<br />

disease outbreaks in pigs in Sweden during 2009 and 2010.Virus Genes, 2011.<br />

Apr;42(2):236-44<br />

3. Kärber, G., Beitrag zur kollektiven Behandlung pharmakologischer<br />

Reihenversuche. Archiv für experimentelle Pathologie und Pharmacologie, 1931.<br />

162: p. 480-483.<br />

Influenza A, swine, genetic characterization


S1 - P - 18<br />

A TOOL TO ASSESS ANTIMICROBIAL RESISTANCE PATTERNS IN DAIRY FARMS<br />

Gerolimetto E. 1 , Cibin V. 1 , Marafin E. 1 , Zavagnin P. 1 , Longo A. 1 , Ramon E. 1 , Ruffa M. 1 , Menin R. 2 , Carraro A. 2 ,<br />

Pozza G. 1 , Lettini A.A. 1 ,Ricci A. 1<br />

1<br />

Istituto Zoopr<strong>of</strong>ilattico Sperimentale delle Venezie, Legnaro (PD), Italy<br />

2 ASL 15 Alta Padovana, Cittadella (PD), ITALY<br />

Antimicrobial resistance, E. coli, Enterococcus spp., dairy farms<br />

Introduction<br />

Resistance to antimicrobials is <strong>of</strong> major public health concern.<br />

The inappropriate use <strong>of</strong> antimicrobials may contribute to the<br />

emergence <strong>of</strong> antibiotic resistant commensal bacteria, with the<br />

subsequent risk <strong>of</strong> widespread dissemination <strong>of</strong> antibiotic<br />

resistance determinants (1). To our knowledge few studies<br />

describing antimicrobials resistance patterns <strong>of</strong> indicator bacteria<br />

in dairy farms have been published (2). Aim <strong>of</strong> this study is to<br />

evaluate a tool to identify antimicrobial resistance patterns <strong>of</strong><br />

commensal bacteria at dairy herd-level highlighting differences<br />

among animal categories.<br />

Material & methods<br />

30 dairy farms in the north-eastern part <strong>of</strong> Italy were sampled<br />

between September 2010 and January 2011. Individual faecal<br />

samples were collected from 4 groups <strong>of</strong> animals taking into<br />

account the intra-herd animal population distribution: 15, 5, 5 and<br />

up to 5 samples from lactating cows (LA), dry cows (DC), heifers<br />


S1 - P - 19<br />

CHARACTERIZATION OF EXTENDED-SPECTRUM ß-LACTAMASE (ESBL)-PRODUCING<br />

ESCHERICHIA COLI BOVINE ISOLATES IN NORTHWEST SPAIN.<br />

V. Gómez 1,2 , A. Mora 2 , A. Méndez 1 , R. Mamani 2 , C. López 2 , A. Lamas 1 , C. Eiras 1 , J. Blanco 2<br />

1 Laboratorio de Sanidade e Produción Animal de Galicia, Consellería do Medio Rural e do Mar, Xunta de Galicia, Lugo, España<br />

2<br />

Departamento de Microbioloxía e Parasitoloxía. Laboratorio de Referencia de Escherichia coli. Facultade de Veterinaria. USC, Lugo, España<br />

Keyword: Escherichia coli, ESBLs, bovine, CTX-M<br />

Introduction<br />

Extended-spectrum ß-lactamases (ESBLs) confer bacterial<br />

resistance to all beta-lactams except carbapenems and<br />

cephamycins. In 1989, a new family <strong>of</strong> ESBLs called CTX-M<br />

was characterized with a potent hydrolytic activity against<br />

cephalosporins such as cefuroxime, cefotaxime and cefepime.<br />

At least 65 ß-lactamases CTX-M are currently known and<br />

spread all over the world (1).<br />

The emergence and wide dissemination <strong>of</strong> ESBLs among<br />

clinical Escherichia coli (E. coli) isolates in hospitals,<br />

community patients, food-producing animals and household<br />

pets in recent years are <strong>of</strong> great concern and represent a<br />

problem for the treatment <strong>of</strong> infectious diseases (2).<br />

The aim <strong>of</strong> this study was the genotypic and phenotypic<br />

characterization <strong>of</strong> E. coli strains isolated from bovine.<br />

3. Locatelli C, Caronte I, Scaccabarozzi L, Migliavacca R, Pagani L,<br />

Moroni P. 2009. Extended-spectrum β-lactamase production in E. coli<br />

strains isolated from clinical bovine mastitis. Vet. Res. Commun. 33:<br />

141-144.<br />

4. López-Cerero L, Egea P, Serrano L, Navarro D, Mora A, Blanco J,<br />

Doi Y, Paterson DL, Rodríguez-Baño J, Pascual A. Characterisation <strong>of</strong><br />

clinical and food animal Escherichia coli isolates producing CTX-M-15<br />

extended-spectrum β-lactamase belonging to ST410 phylogroup A. Int J<br />

Antimicrob Agents. 2011 Apr;37(4):365-7. Epub 2011 Feb 16. PubMed<br />

PMID: 21330111.<br />

5. Cortés P, Blanc V, Mora A, Dahbi G, Blanco JE, Blanco M, López C,<br />

Andreu A, Navarro F, Alonso MP, Bou G, Blanco J, Llagostera M.<br />

Isolation and characterization <strong>of</strong> potentially pathogenic antimicrobialresistant<br />

Escherichia coli strains from chicken and pig farms in Spain.<br />

Appl Environ Microbiol. 2010 May;76(9):2799-805. Epub 2010 Mar 12.<br />

PubMed PMID: 20228098; PubMed Central PMCID: PMC2863447.<br />

Material and Methods<br />

From 2003 to 2009, 38 strains <strong>of</strong> ESBL-producing E. coli were<br />

isolated from clinical cases <strong>of</strong> bovine mastitis and diarrhoea<br />

from dairy farms in Galicia, northwest Spain.<br />

The phenotypic characterization was determined by BD<br />

PhoenixTM Automated Microbiology System and double-disk<br />

synergy test.<br />

Genotypic ESBL confirmation, virulence gene, phylogenetic<br />

groups, and amplification <strong>of</strong> fragment blaCTX-M were carried<br />

out by PCR. Finally, the strains were serotyped, and molecular<br />

characterized by multilocus sequence typing (MLST) and<br />

pulsed-field gel electrophoresis (PFGE).<br />

Results<br />

Analysis <strong>of</strong> the 38 isolates showed that 74% (28 strains) and<br />

26% (10 strains) produced CTX-M-14 and CTX-M-32<br />

enzymes, respectively. A statistical significant association was<br />

found between CTX-M-14 and resistance to gentamicin, and<br />

CTX-M-32 and resistance to aztreonam and ceftazidime. Of<br />

the 38 E. coli isolates, 30 (79%) carried at least two virulence<br />

gene typical <strong>of</strong> extraintestinal pathogenic E. coli strains<br />

(ExPEC).<br />

The determination <strong>of</strong> phylogenetic groups showed that the E.<br />

coli isolates belonged to A (50%), B1 (31.5%) and D (18.4%)<br />

phylogroups. None <strong>of</strong> the bovine isolates belonged to the<br />

phylogenetic group B2 associated with human virulence in<br />

ExPEC.<br />

In accordance with the high diversity identified by serotyping,<br />

PFGE molecular analysis revealed highly heterogeneous<br />

pr<strong>of</strong>iles. Only one successful clonal group, the O20:H8/HNT-<br />

B1-ST448 CTX-M-14-producing, was detected.<br />

Discussion & conclusions<br />

Our survey demonstrates that Galician bovine is a reservoir <strong>of</strong><br />

CTX-M-14 and CTX-M-32-producing E. coli strains, with<br />

important associated resistances to aminoglycoside,<br />

tetracycline, sulphonamide, fluoroquinolone and<br />

chloramphenicol.<br />

Although person-to person spread is recognised as the main<br />

vehicle <strong>of</strong> spread <strong>of</strong> ESBL-producing E. coli in hospital and<br />

the community, the primary reservoirs <strong>of</strong> such organisms are<br />

still in discussion. ESBL-producing E. coli have been isolated<br />

from food animals in many countries, particularly poultry and<br />

cattle (3, 4, 5), so farm animals are now recognised as<br />

important carriers <strong>of</strong> BLEEs. Further studies are required to<br />

determine the true zoonotic potential <strong>of</strong> these ESBL-producing<br />

strains.<br />

References<br />

1. Cantón R, Coque TM. 2006. The CTX-M beta-lactamase pandemic.<br />

Curr. Opin. Microbiol. 9:466-475.<br />

2. Carattoli A. 2008. Animal reservoirs for extended-spectrum ß-<br />

lactamase producers. Clin. Microbiol. Infect. 14:117-123.


S1 - P - 20<br />

OVINE ABORTION CAUSED BY YERSINIA PSEUDOTUBERCULOSIS – A CASE REPORT<br />

Varpu Hirvelä-Koski, Minna Nylund<br />

Finnish Food Safety Authority, Research Department, Oulu, Finland<br />

Ovine abortion, pseudotuberculosis<br />

Introduction<br />

Yersinia pseudotuberculosis is a gram-negative organism that<br />

causes various syndromes including mesenteric lymphadenitis<br />

and septicaemia in wild and domestic animals as well as in<br />

humans. Wild birds and rodents are considered to be<br />

reservoirs <strong>of</strong> the bacterium. Sporadic abortions due to<br />

placentitis have been reported in sheep, goats and cows (1,2).<br />

We describe here a case <strong>of</strong> ovine abortion caused by Yersinia<br />

pseudotuberculosis.<br />

Material & methods<br />

The samples were obtained from an organic farm keeping<br />

about 300 ewes in central Finland. The ewes were traditional<br />

Finnish homebred sheep. Two <strong>of</strong> the ewes aborted before the<br />

lambing season with an interval <strong>of</strong> two weeks, approximately<br />

one month before the estimated lambing. Samples from the<br />

second abortion were sent to the laboratory, unfortunately<br />

frozen.<br />

Two fetuses and the afterbirth were obtained to the laboratory.<br />

The samples were examined using gross pathology and<br />

histopathology (HE stain). Lung, liver and contents <strong>of</strong><br />

abomasum as well as the afterbirth were cultivated on the<br />

following media: trypticase soy agar incorporated with 5 %<br />

bovine blood (aerobic incubation), fastidious anaerobe agar<br />

(FAA) (microaerophilic), FAA (anaerobic), selective FAA for the<br />

detection <strong>of</strong> Campylobacter fetus (microaerophilic), Sabouraud<br />

agar (aerobic), salmonella enrichment media (pre-enrichment<br />

broth, modified semi-solid Rappaport Vassiliadis medium).<br />

STAMP stain was used for the detection <strong>of</strong> brucellosis using<br />

material from lung, liver and afterbirth. All plates were<br />

incubated at 37⁰+ 1⁰C. The identification <strong>of</strong> Yersinia<br />

pseudotuberculosis was based on colony morphology, gramstain,<br />

oxidase reaction, production <strong>of</strong> acid from glucose, typical<br />

growth on MacConkey agar and API 20 E pr<strong>of</strong>ile.<br />

Wild rodents are considered to be one reservoir <strong>of</strong> Yersinia<br />

pseudotuberculosis. The population densities <strong>of</strong> wild voles has<br />

been high in Finland in recent years. It is possible that this<br />

phenomenon has increased the infection pressure caused by<br />

Yersinia pseudotuberculosis.<br />

References<br />

1. Karbe, E, Erickson, ED (1984). Ovine abortion and stillbirth due to<br />

purulent placentitis caused by Yersinia pseudotuberculosis. Vet. Pathol.<br />

21, 601-606<br />

2. Juste, RA, Minguijón, E, Arranz, J, Fuertes, M, Beltrán de Heredia, I<br />

(2009). Lamb mortality in an outbreak <strong>of</strong> Yersinia pseudotuberculosis<br />

mastitis, as a collateral effect <strong>of</strong> colostrum feeding for Lentivirus-control.<br />

Small Ruminant Res. 86, 46-53.<br />

3. Hallanvuo, S (2009). Foodborne Yersinia. Identification and<br />

molecular epidemiology <strong>of</strong> isolates from human infections. Acad. Diss.,<br />

University <strong>of</strong> Helsinki, Finland. National Institute for Health and Welfare,<br />

Research series no 11, Helsinki University Print.<br />

Results<br />

The fetuses, a male and a female weighed 347,5 and 600,8 g,<br />

the fetal length being 21 and 26 cm, respectively. In gross<br />

pathology no specific lesions could be found. Both fetuses and<br />

the afterbirth had quite intense autolytic changes.<br />

Yersinia pseudotuberculosis was isolated as a pure growth<br />

from the afterbirth. Growth was observed on all non-selective<br />

media at all atmospheres. The bacterium was detected also<br />

from the inner organs <strong>of</strong> both fetuses. The growth was most<br />

abundant in cultivations from abomasum. Other pathogenic<br />

organisms were not detected.<br />

The bacterium was a gram-negative small rod, oxidase<br />

negative, acid but not gas was produced from glucose. There<br />

were small brown colonies on MacConkey. API 20 E pr<strong>of</strong>ile<br />

suggested identification to Yersinia pseudotuberculosis.<br />

Histopathological examination revealed suppurative placentitis<br />

and non-suppurative conjunctivitis. In the villi <strong>of</strong> the cotyledons<br />

there were multiple necrotic foci with neutrophilic and<br />

mononuclear infiltrates. There were masses <strong>of</strong> coccobacilli and<br />

infiltrations <strong>of</strong> mononuclear and plasma cells with a few<br />

granulocytes in the lamina propria <strong>of</strong> the conjunctiva <strong>of</strong> both<br />

fetuses. In other internal organs the autolytic changes were<br />

quite intense and any specific lesions could not be found.<br />

Discussion & conclusions<br />

Yersinia pseudotuberculosis is <strong>of</strong>ten observed as the cause <strong>of</strong><br />

septicemia in wild animals in Finland, e,g, hare. The bacterium<br />

has also caused several large outbreaks <strong>of</strong> food-borne<br />

illnesses in humans in Finland in the last decade. Yersinia<br />

pseudotuberculosis has been reported as a rare causative<br />

agent <strong>of</strong> sporadic abortion <strong>of</strong> sheep and goat. However, as far<br />

as we know, this is the first time that it was isolated from ovine<br />

abortion samples in Finland.


S1- P - 21<br />

ANALYSIS OF THE EFFICIENCY OF THE MCMASTER METHOD IN RAYNAUD’S MODIFICATION IN<br />

DETECTION OF TOXOCARA SP. AND TRICHURIS SP. EGGS IN CARNIVORES FAECES<br />

Maciej Kochanowski, Joanna Dąbrowska, Jacek Karamon, Tomasz Cencek<br />

National Veterinary Research Institute, Departament <strong>of</strong> Parasitology and Invasive Disease, Pulawy, Poland<br />

McMaster method, efficiency, carnivores, Toxocara, Trichuris<br />

Introduction<br />

Carnivores animals are the hosts <strong>of</strong> the important zoonotic<br />

intestinal parasites. Faeces <strong>of</strong> these animals, containing eggs <strong>of</strong><br />

dangerous parasites are a potential source <strong>of</strong> infection for<br />

humans. The particular importance in the spread <strong>of</strong> dangerous<br />

invasive parasitic forms may have a common use <strong>of</strong> this faeces<br />

as natural organic fertilizers. The aim <strong>of</strong> study was to determine<br />

the efficiency <strong>of</strong> McMaster method in Raynaud’s modification<br />

(Raynaund, 1970) in the detection <strong>of</strong> parasite eggs <strong>of</strong> the genera<br />

Toxocara and Trichuris present in the faeces <strong>of</strong> carnivores, using<br />

faeces samples enriched with known numbers <strong>of</strong> eggs.<br />

Materials & methods<br />

Four variants <strong>of</strong> this McMaster method (counting in one grid, two<br />

grids, the whole McMaster chamber and flotation slide) were<br />

used to examine dog faeces samples enriched with Toxocara sp.,<br />

Trichuris sp. and Ascaris suum eggs. A. suum eggs were the<br />

control <strong>of</strong> influence <strong>of</strong> the parasite eggs properties on the results.<br />

One hundred and sixty enriched samples were prepared from<br />

dog faeces (20 repetitions for each eggs level) containing 15, 25,<br />

50, 100, 150, 200, 250 and 300 eggs <strong>of</strong> the three types <strong>of</strong> eggs in<br />

1 g <strong>of</strong> faeces. Moreover, to compare the influence <strong>of</strong> kind <strong>of</strong><br />

faeces on the efficiency <strong>of</strong> the method 160 pig faecal samples<br />

were prepared and enriched with A. suum eggs.in the same way<br />

Results<br />

The highest (worst) limit <strong>of</strong> detection (Table 1) in all McMaster<br />

variants were obtained with eggs <strong>of</strong> Toxocara sp. (from 25 to 250<br />

eggs / g faeces, depending on the variant) - about twice higher<br />

than for Trichuris sp. and A. suum eggs (from 15 to 100 eggs / g<br />

feces). The highest (worst) limit <strong>of</strong> detection for flotation in the<br />

tube were obtained for Trichuris sp. eggs (100 eggs / g) - it was<br />

4-times higher than the results for other types <strong>of</strong> eggs (25 eggs /<br />

g). The best results <strong>of</strong> the limit <strong>of</strong> detection, sensitivity, the lowest<br />

coefficients <strong>of</strong> variation (providing about repeatability) were<br />

obtained with the use <strong>of</strong> whole McMaster chamber variant. There<br />

was no significant impact <strong>of</strong> faeces properties (dog and pig<br />

faeces) on the obtained results. Multiplication factors (to estimate<br />

the real number <strong>of</strong> eggs in 1 g <strong>of</strong> faeces) were calculated on the<br />

basis <strong>of</strong> the transformed equation <strong>of</strong> the trend line illustrating the<br />

relationship between number <strong>of</strong> eggs found by the examination<br />

and the number <strong>of</strong> eggs added to the sample. Multiplication<br />

factors for eggs <strong>of</strong> Toxocara sp. and Trichuris sp. were higher<br />

than planned by Raynaud, and for A. suum were comparable with<br />

them. The multiplication factor estimated for flotation in the tube<br />

(flotation slide variant) for all types <strong>of</strong> examined parasite eggs<br />

were significantly higher than that given by Raynaud (Table 2).<br />

Table 1: Limit <strong>of</strong> detection for all variants <strong>of</strong> the McMaster method<br />

in Raynaund’s modification<br />

Variants<br />

Dog faeces<br />

Pig faeces<br />

<strong>of</strong> the method Toxocara Trichuris Ascaris Ascaris<br />

one grid 250 100 100 50<br />

two grids 100 50 25 25<br />

whole chamber 25 15 15 15<br />

flotation slide 25 100 25 25<br />

Discussion & conclusions<br />

Most <strong>of</strong> the publications about detection <strong>of</strong> parasite eggs are<br />

based on material from natural or experimental infection. For that<br />

reason the real number <strong>of</strong> eggs in faeces can not be properly<br />

assessed (Vadlejch et al, 2011; Mes et al, 2007). Therefore, in<br />

order to obtain the most reliable results in our experiment the<br />

material enriched with a known number <strong>of</strong> parasite eggs was<br />

used. The results <strong>of</strong> experiments indicate that detection <strong>of</strong><br />

parasite eggs <strong>of</strong> carnivores by McMaster method in Raynaud’s<br />

modification differs from the basic parameters <strong>of</strong> this<br />

modification. There were significant differences in the efficiency<br />

<strong>of</strong> the method according to the type <strong>of</strong> parasite eggs. In the case<br />

<strong>of</strong> Toxocara sp. eggs examined in the McMaster chamber there<br />

were observed significantly lower numbers <strong>of</strong> detected eggs in 1<br />

g <strong>of</strong> faeces than obtained for Trichuris sp. and A. suum eggs.<br />

Most likely reason <strong>of</strong> the lowest detection Toxocara sp. eggs in<br />

the McMaster chamber is the specific shell structure <strong>of</strong> these<br />

eggs – namely, they have strong adhesive properties<br />

(Overgaauw, Knapen, 2008). It can result in higher losses <strong>of</strong><br />

parasite eggs during performing <strong>of</strong> the method. The variant in the<br />

tube (flotaion slide) was characterized by the lowest number <strong>of</strong><br />

detected eggs in the case <strong>of</strong> Trichuris sp. It is probably related to<br />

the high specific gravity <strong>of</strong> the Trichuris sp. eggs affects to less<br />

effective flotation process in tube. According to our study the<br />

detection <strong>of</strong> Toxocara sp. eggs in McMaster chamber is twice<br />

lower than planned by Raynaud. In the case <strong>of</strong> Trichuris sp. eggs<br />

it is slightly lower. However, A. suum eggs in pig and dog feces,<br />

were detected in number close to the assumptions <strong>of</strong> McMaster<br />

method in Raynaud’s modification. Detection <strong>of</strong> all three types<br />

parasite eggs as a result <strong>of</strong> the tube flotation (flotation slide<br />

variant) is several times lower than the assumptions <strong>of</strong> Raynaud's<br />

modification (which is at the level <strong>of</strong> detection obtained in the<br />

variant <strong>of</strong> two grids <strong>of</strong> McMaster chamber). Our results allowed<br />

us to assess the real efficiency <strong>of</strong> the McMaster method in<br />

Raynaud’s modification in carnivore feces samples enriched with<br />

a known number <strong>of</strong> parasite eggs. The investigation carried out<br />

with selected (important to the diagnostic and the zoonotic point<br />

<strong>of</strong> view) two types <strong>of</strong> nematodes (Toxocara sp. and Trichuris sp.)<br />

showed a significant effect <strong>of</strong> parasite genus on the detection<br />

efficiency. However, it is desirable to extend calibration <strong>of</strong> the<br />

method for other kinds <strong>of</strong> parasites whose eggs differ significantly<br />

in the morphology - in particular with regard to the eggs <strong>of</strong><br />

tapeworms.<br />

References<br />

1. Mes T., Eysker M., Ploeger H., 2007. A simple, robust and semiautomated<br />

parasite egg isolation protocol. Nature Protocols.<br />

2. Overgaauw P., Knapen F., 2008. Toxocarosis, an important zoonosis.<br />

European Journal <strong>of</strong> Companion Animal Practice Vol. 18 Issue 3 (2008).<br />

3. Raynaud J., 1970. Etude de l’efficacite d’une technique de coproscopie<br />

quantitative pour le diagnostic de routine et le controle des infestations<br />

parasitaires des bovins, ovins, equines et porcins. Annales de<br />

Parasitologie (Paris). 45 pp. 321-342<br />

4. Vadlejch J., Petrtyl M., Zaichenko I., Cadkova Z., Jankovska I.,<br />

Langrova I., Moravec M., 2011. Which McMaster egg counting technique is<br />

the most reliable? Parasitology Research 109(5): 1387-1394 (2011)<br />

Table 2: Multiplication factors for estimation <strong>of</strong> the number <strong>of</strong><br />

eggs in 1g <strong>of</strong> faeces, for all variants <strong>of</strong> the McMaster method in<br />

Raynaund’s modification<br />

Variants<br />

Dog faeces<br />

Pig faeces<br />

<strong>of</strong> the method Toxocara Trichuris Ascaris Ascaris<br />

one grid 244 145 108 111<br />

two grids 120 78 54 63<br />

whole chamber 40 26 22 22<br />

flotation slide 49 62 35 43


S1 - P - 22<br />

MILK AMYLOID A AND ITS USEFULNESS IN THE LABORATORY DIAGNOSIS OF MASTITIS<br />

Gabriel Kováč 1 , Csilla Tóthová 1 , Oskar Nagy 1 , Martin Kováč 2<br />

1 University <strong>of</strong> Veterinary Medicine and Pharmacy, Clinic for Ruminants, Košice, Slovak Republic<br />

2 Regional Veterinary and Food Administration, Košice, Slovak Republic<br />

Dairy cows, mastitis, milk amyloid A, haptoglobin, serum amyloid A<br />

Introduction<br />

Mastitis is recognized as one <strong>of</strong> the most important diseases<br />

affecting dairy cattle. Raised levels <strong>of</strong> major acute phase proteins<br />

in cattle, haptoglobin and serum amyloid A, have previously been<br />

shown in milk from cows with clinical mastitis as a result <strong>of</strong> the<br />

leakage <strong>of</strong> these proteins from the blood to the milk (Eckersall et<br />

al., 2001). Further investigations showed an extrahepatic<br />

synthesis <strong>of</strong> specific is<strong>of</strong>orm <strong>of</strong> amyloid A directly in mammary<br />

epithelial cells, the so called milk amyloid A (MAA) (McDonald et<br />

al., 2001).<br />

Therefore, this work was aimed at the evaluation <strong>of</strong> the influence<br />

<strong>of</strong> clinical and sub-clinical mastitis in dairy cows on the<br />

concentrations <strong>of</strong> MAA in milk samples, and selected acute<br />

phase proteins in blood serum.<br />

Materials & methods<br />

Into the evaluation we included 41 dairy cows <strong>of</strong> a low-land black<br />

spotted breed and its crossbreeds with various clinical findings on<br />

the mammary gland. These cows were in the 3rd – 4th lactation,<br />

but not in the period shortly after parturition. Clinical examination<br />

<strong>of</strong> the mammary gland was performed by visual inspection and<br />

palpation. Clinical mastitis was diagnosed by the presence <strong>of</strong><br />

observable signs <strong>of</strong> inflammation in the infected quarter such as<br />

swelling, heat, pain or redness, and by the presence <strong>of</strong> clots and<br />

flakes in the milk, or by its abnormal color or consistency. To<br />

detect sub-clinical mastitis the Californian Mastitis Test (CMT)<br />

was performed. According to the results <strong>of</strong> the clinical<br />

examination <strong>of</strong> the udder and to the results <strong>of</strong> CMT the animals<br />

were divided into 4 groups: Group I – cows without clinical<br />

changes on the mammary gland and with negative CMT (n = 7),<br />

Group II – cows without clinical changes on the mammary gland<br />

and with weakly positive CMT (n = 12), Group III – cows without<br />

clinical changes on the mammary gland and with strongly positive<br />

CMT (n = 13), Group IV – cows with clinical changes on the<br />

mammary gland and changes in milk appearance (n = 9).<br />

Milk samples were collected into plastic tubes by hand-stripping.<br />

Blood samples were collected by direct puncture <strong>of</strong> v. jugularis.<br />

Milk samples were used to assess the concentrations <strong>of</strong> milk<br />

amyloid A (M-SAA, ng/ml), and haptoglobin (Hp, mg/ml) and<br />

serum amyloid A (SAA, μg/ml) were assessed in blood samples.<br />

Haptoglobin was assessed using commercial colorimetric kits<br />

(Tridelta Development, Ireland) in microplates. SAA and M-SAA<br />

were analysed by commercial ELISA kits (Tridelta Development,<br />

Ireland). The optical densities were read on automatic microplate<br />

reader Opsys MR (Dynex Technologies, USA) at an optical<br />

density <strong>of</strong> 630 nm for Hp, and 450 nm using 630 as reference for<br />

SAA and M-SAA.<br />

The analysis <strong>of</strong> the significance <strong>of</strong> differences in measured<br />

values between cows with various clinical findings on the<br />

mammary gland was performed by Kruskal-Wallis nonparametric<br />

ANOVA test and Dunn's Multiple Comparisons Test.<br />

Results<br />

M-SAA concentrations in milk samples differed significantly<br />

between the groups (P < 0.001), with concentrations in samples<br />

from cows with clinical mastitis (group IV) being significantly<br />

higher than in samples from groups I and II (P < 0.001 and P <<br />

0.05, respectively, Table 1). The concentrations <strong>of</strong> M-SAA in milk<br />

samples increased with increasing CMT score.<br />

The serum concentrations <strong>of</strong> Hp showed also tendency <strong>of</strong><br />

gradual significant increase with increasing CMT score and<br />

clinical changes on the mammary gland (P < 0.05, Table 1). The<br />

highest mean Hp concentration we found in cows with clinically<br />

manifested signs <strong>of</strong> mastitis. Similarly, SAA concentrations<br />

differed significantly between the evaluated groups <strong>of</strong> cows (P <<br />

0.05), with the highest mean concentration in animals with clinical<br />

signs <strong>of</strong> mastitis. However, the differences in the obtained results<br />

<strong>of</strong> Hp and SAA concentrations between the evaluated groups <strong>of</strong><br />

cows were less significant (P < 0.05) compared with MAA<br />

concentrations. The mean SAA concentrations found in cows<br />

from group I and group II were roughly uniform.<br />

Table 1: Concentrations <strong>of</strong> evaluated variables in dairy cows<br />

Variable I II III IV P <<br />

M- x 325.7 A,B 1433.1 a 3910.4 A 6073.8 B,a<br />

SAA ±sd 173.8 949.2 2145.8 4414.0<br />

0.001<br />

Hp<br />

x 0.046 0.122 0.299 0.329<br />

±sd 0.053 0.263 0.314 0.339<br />

0.05<br />

SAA<br />

x 29.7 27.6 a 48.2 71.5 a<br />

±sd 27.6 28.0 42.5 31.5<br />

0.05<br />

The same indexes in lines mean significance <strong>of</strong> differences in values<br />

between the groups: a – P < 0.05; A, B – P < 0.01<br />

Discussion & conclusions<br />

The results <strong>of</strong> our study indicate that inflammatory diseases <strong>of</strong><br />

the mammary gland lead to an increase in the concentrations <strong>of</strong><br />

M-SAA. Compared to Hp and SAA, M-SAA is synthesized directly<br />

in the mammary epithelia <strong>of</strong> the udder in response to infection<br />

(Jacobsen et al. 2005). Therefore, M-SAA is believed to be a<br />

more sensitive indicator <strong>of</strong> mastitis; it accumulates in milk only<br />

during mammary inflammation. Petersen et al. (2005) reported<br />

that M-SAA concentrations, similarly to our results, were higher in<br />

quarters with mastitis compared to healthy quarters.<br />

Moreover, mastitis may activate systemic inflammatory reactions,<br />

including the induction <strong>of</strong> the synthesis <strong>of</strong> acute phase proteins<br />

by the liver. The results <strong>of</strong> our study showed that the<br />

concentrations <strong>of</strong> Hp and SAA were higher in serum from cows<br />

with clinical mastitis, and increased with increasing CMT score.<br />

These results are in line with previous studies (Eckersall et al.<br />

2001). It appears that localized severe inflammation <strong>of</strong> the udder<br />

is sufficiently intense to induce a measurable systemic acute<br />

phase response. The concentrations <strong>of</strong> measured acute phase<br />

proteins had a tendency to be higher in the serum from the cows<br />

with local signs <strong>of</strong> mastitis and also in cows without clinical<br />

changes on the mammary gland, but with positive CMT.<br />

However, the finding that the differences in Hp and SAA<br />

concentrations observed between the groups <strong>of</strong> cows were less<br />

significant than the differences in M-SAA concentrations means<br />

that the measuring <strong>of</strong> serum concentrations <strong>of</strong> some acute phase<br />

proteins would be less useful to the evaluation <strong>of</strong> the severity <strong>of</strong><br />

mastitis than the measuring <strong>of</strong> the concentrations <strong>of</strong> M-SAA<br />

directly in milk samples.<br />

Generated data suggest the usefulness <strong>of</strong> M-SAA for diagnosis<br />

<strong>of</strong> bovine sub-clinical mastitis, as well as in the determination <strong>of</strong><br />

the severity <strong>of</strong> mastitis, suggesting that its assessment as a part<br />

<strong>of</strong> the laboratory diagnosis would be a valuable supplementation<br />

to the proper clinical diagnosis and determination <strong>of</strong> other blood<br />

laboratory parameters.<br />

Acknowledgements<br />

This work was supported by VEGA Scientific Grant No 1/0592/12<br />

from the Ministry <strong>of</strong> Education, and by Slovak Research and<br />

Development Agency under contract No. APVV-0475-10.<br />

References<br />

1. Eckersall, PD, Young, FJ, McComb, C, Hogarth, CJ, Safi, S, Weber, A,<br />

McDonald, T, Nolan, AM, Fitzpatrick, JL (2011). Acute phase proteins in<br />

serum and milk from dairy cows with clinical mastitis. Vet Rec, 148, 35-41<br />

2. Jacobsen, S, Niewold, TA, Kornalijnslijper, E, Toussaint, MJM, Gruys, E<br />

(2005). Kinetics <strong>of</strong> local and systemic is<strong>of</strong>orms <strong>of</strong> serum amyloid A in<br />

bovine mastitic milk. Vet Immunol Immunopathol, 104, 21-31<br />

3. McDonald, TL, Larson, MA, Mack, DR, Weber, A (2001). Elevated<br />

extrahepatic expression and secretion <strong>of</strong> mammary-associated serum<br />

amyloid A3 (M-SAA3) into colostrums. Vet Immunol Immunopathol, 83,<br />

203-211<br />

4. Petersen, HH, Gardner, IA, Rossitto, P, Larsen, HD, Heegaard, PMH<br />

(2005). Accuracy <strong>of</strong> milk amyloid A (MAA) concentration and somatic cell<br />

count for diagnosing bovine mastitis. Proceedings <strong>of</strong> the 5 th International<br />

Colloquim on Animal Acute Phase Protein, Dublin, Ireland, March 14-15,<br />

2005, 43-44


S1 - P - 23<br />

RELIABLE AND EARLY DETECTION OF SALMONELLA IN PIGS<br />

Tilman Kühn 1 , Patrik Buholzer 2<br />

1<br />

Synlab.vet Leipzig, Leipzig, Germany<br />

2<br />

Prionics AG, Schlieren, Switzerland<br />

Introduction<br />

Salmonellosis is one <strong>of</strong> the most important zoonotic diseases and<br />

can cause serious clinical symptoms in humans. Pigs have been<br />

recognized as an important source <strong>of</strong> Salmonella infections. The<br />

existence <strong>of</strong> this disease presents great risks for human health.<br />

Salmonella infections <strong>of</strong> animals intended for the food industry<br />

play an important role in public health as these animals are<br />

considered to be the major source <strong>of</strong> human Salmonella<br />

infections. The PrioCHECK Salmonella Ab porcine 2.0 was<br />

developed for large-scale screening <strong>of</strong> pigs and for application in<br />

Salmonella control programs. Here we show that the test clearly<br />

discriminates positive and negative samples through its large<br />

dynamic range. As the test also allows earlier detection <strong>of</strong> a<br />

Salmonella infection in pigs, than competitor tests, it is the<br />

diagnostic tool <strong>of</strong> choice for Salmonella control programs.<br />

Materials & methods<br />

Two separate experiments were performed. In the first<br />

experiment serum samples <strong>of</strong> ten pigs (4 negative and 6<br />

Salmonella positive samples) were tested with both the<br />

PrioCHECK Salmonella Ab porcine 2.0 and a competitor test.<br />

Additionally, the Salmonella status was determined in ten meat<br />

juice samples with both tests.<br />

In the second experiment, 40 pigs were experimentally inoculated<br />

with live Salmonella vaccine. Serum samples were taken at the<br />

day <strong>of</strong> inoculation, 3, 6 and 9 weeks post inoculation and were<br />

tested with the PrioCHECK Salmonella Ab porcine 2.0 and two<br />

competitor tests both validated for use in Germany. The results <strong>of</strong><br />

the three tests were compared.<br />

Results<br />

In the first experiment, both tests identified all positive and<br />

negative samples correctly, but the positive samples gave a<br />

much higher values with the PrioCHECK Salmonella Ab porcine<br />

2.0 and therefore results were more clear-cut than those obtained<br />

with the competitor test. The average percentage positivity (PP)<br />

<strong>of</strong> positive samples <strong>of</strong> the PrioCHECK Salmonella Ab porcine 2.0<br />

exceeded that <strong>of</strong> the competitor test by over 70 %. However, the<br />

PP values <strong>of</strong> negative samples were equal to, or lower than those<br />

<strong>of</strong> the competitor test. This means that the test does not generally<br />

generate high values, but is specific for positive samples.<br />

The PrioCHECK Salmonella Ab porcine 2.0 also earlier identifies<br />

animals experimentally infected with Salmonella. In serum<br />

samples <strong>of</strong> experimentally infected pigs, the PrioCHECK<br />

Salmonella Ab porcine 2.0 identified 25 samples as positive at 3<br />

weeks post inoculation. 17 out <strong>of</strong> these 25 samples were missed<br />

by at least one competitor test (13 samples) at three weeks post<br />

inoculation, or only detected by the other tests at a later time post<br />

inoculation (4 samples).<br />

Discussion & conclusions<br />

The data show that the results obtained with PrioCHECK<br />

Salmonella Ab porcine 2.0 showed a large dynamic range and<br />

therefore allow reliable discrimination between positive and<br />

negative samples. This dynamic range means that the cut-<strong>of</strong>f <strong>of</strong><br />

40% PP -- implemented in most countries -- could easily be<br />

lowered to 20% without loss <strong>of</strong> specificity.<br />

The application <strong>of</strong> the PrioCHECK Salmonella Ab porcine 2.0<br />

reliably helps control Salmonella infections even early after<br />

infection.<br />

References<br />

1. B. Nielsen, D. Baggesen, F. Bager, J. Haugegaard, P. Lind Veterinary<br />

Microbiology 47 (1995) 205-218<br />

2. H.M.F. van der Heijden. First International Ring Trial <strong>of</strong> ELISA’s for<br />

Salmonella- antibody. Berl. Münch. Tierärztl Wschr. (2001) 389-392.<br />

3. PrioCHECK® Salmonella Ab porcine 2.0, Prionics AG<br />

http://www.prionics.com/diseases-solutions/salmonellosis/<br />

Food safety, diagnostic test, Salmonellosis


S1 - P - 24<br />

DETECTION OF BVD VIRUS ON PERSISTENTLY INFECTED CALVES BY REAL TIME REVERSE<br />

TRANSCRIPTION PCR ON EAR NOTCHES<br />

Damien MAGNEE 1 , Julie CHARROT 1 , Stéphane DALY 1 , Sandrine MOINE 1 , Eric SELLAL 1<br />

1<br />

LSI Laboratoire Service International, LISSIEU, France<br />

PCR, BVDV, Ear Notches, PI Calves, Magnetic Beads<br />

Introduction<br />

Bovine Viral Diarrhea Virus is a pestivirus <strong>of</strong> the Flaviviridae<br />

family. The virus is transmitted by direct contact between<br />

animals. Vertical transmission plays an important role in the<br />

epidemiology and infection pathogenesis. In cattle, fetus infection<br />

can cause abortions, stillbirths, teratogenic effect or persistent<br />

infection <strong>of</strong> newborn calf (PI). It is therefore in the interest <strong>of</strong><br />

farmer to detect PI animals earliest after birth and then prevent<br />

virus spreading in the herd. The BVD diagnosis is performed on<br />

blood and serum, but there is a doubt about the possibility <strong>of</strong><br />

virus detection in the presence <strong>of</strong> calf colostral antibodies, in the<br />

first day <strong>of</strong> life.<br />

In 2011, the French National Federation “Groupe de Défense<br />

Sanitaire” decided to study the feasibility <strong>of</strong> detecting the<br />

Persistently Infected (PI) calves by direct diagnosis on ear<br />

notches samples picked up while animals are tagged. In the<br />

frame <strong>of</strong> BVDV eradication, this system could become universally<br />

used if its sensitivity is equivalent to the detection on blood by RT<br />

rtPCR<br />

Materials & methods<br />

A study was conducted in Eastern France on 19 calves (3 days to<br />

4 weeks old) with confirmed PI status. Bloods and ear notches<br />

were collected for each animal.<br />

For each sample, nucleic acids were extracted with silica<br />

columns methods and with a magnetic beads method developed<br />

by LSI. Real-Time PCR was performed using the TaqVet<br />

BVDV Screening kit.<br />

The results obtained for bloods and ear notches were compared<br />

for each sample, to validate the using <strong>of</strong> skin samples for the<br />

BVDV diagnosis.<br />

The second part <strong>of</strong> the study consists to validate the using <strong>of</strong> pool<br />

<strong>of</strong> 10 samples <strong>of</strong> ear notches. Each positive eluate <strong>of</strong> skin sample<br />

for PI animals was pooled with 9 negative BVDV eluates, to<br />

obtain a pool <strong>of</strong> 10 samples. After RNA purification by column or<br />

magnetic beads, Real-Time PCR was performed with LSI kit.<br />

The last part consists <strong>of</strong> a study <strong>of</strong> the storage conditions <strong>of</strong> ear<br />

notches, from the sampling to the analysis. The samples were<br />

stored 7 days at 3 different conditions: -20°C, +4°C and room<br />

temperature. The RNA purification was performed with silica<br />

columns methods.<br />

Results<br />

For the comparison between blood and ear notches, the results<br />

showed a correlation <strong>of</strong> 100% (Figure 1). The mean difference<br />

between the both matrixes is about 0.7 Ct. Ear notch matrix<br />

shows an equivalent sensitivity than blood for PI calves detection.<br />

30<br />

25<br />

Blood<br />

Ear Notch<br />

Concerning the use <strong>of</strong> pool <strong>of</strong> 10 samples, the results show a<br />

good correlation between individual and pool <strong>of</strong> 10 results, with a<br />

Ct value difference <strong>of</strong> 3.2 (figure 2). This difference corresponds<br />

to the ear notches dilution factor <strong>of</strong> 10 (log [10] = 3.3Ct). Ear<br />

notch is then a reliable sample for PI detection in individual or in<br />

pool <strong>of</strong> 10 samples analysis.<br />

Figure 2: Ear Notch Analysis in pool <strong>of</strong> 10<br />

Concerning the last part <strong>of</strong> the study, for the storage conditions <strong>of</strong><br />

ear notches, no significative difference was observed on the PCR<br />

sensitivity at -20°C or at +4°C (∆Ct <strong>of</strong> 0.6, between D0 and D7).<br />

The storage at room T°C shows a significative variability on Ct<br />

values. So the room temperature is not adapted for the storage <strong>of</strong><br />

skin samples (∆Ct <strong>of</strong> 2.7, between D0 and D7). (Figure 3)<br />

Ct value<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

D0<br />

D7 at RT°<br />

D7 at +4°C<br />

D7 at ‐20°C<br />

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 18 19<br />

Calves<br />

Figure 4: Stability <strong>of</strong> Ear notch samples<br />

Discussion & conclusions<br />

Our results show that (i) ear notch matrix <strong>of</strong>fers the same<br />

sensitivity for PI calves detection than blood, (ii) the detection <strong>of</strong><br />

one PI calf amoung a pool <strong>of</strong> 10 samples can be performed on<br />

both matrixes, and (iii) the ear notch is a stable matrix (at +4°C<br />

and -20°C) for the diagnosis by PCR.<br />

The complete analysis method developed on ear notches<br />

(TaqVet BVDV Screening and MagVet Universal Isolation<br />

kit) is sensitive enough to detect a PI calf in pool <strong>of</strong> 10 samples or<br />

individual analysis. Then it could be used in large scale to control<br />

the disease and decrease the incidence <strong>of</strong> the PI in bovine herds.<br />

Ct value<br />

20<br />

15<br />

Acknowledgements<br />

A special thank to the LVD55-Segilab laboratory to their active<br />

participation in this project.<br />

10<br />

5<br />

0<br />

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 18 19<br />

Calves<br />

Figure 1: Comparison on blood and ear notches matrixes<br />

The results obtained with columns or magnetic beads are similar<br />

for BVD and IPC detection, with a mean difference <strong>of</strong> 1 Ct<br />

between both methods. Extraction with beads shows equivalent<br />

results on PCR sensitivity than columns.


S1 - P - 25<br />

GENOTYPIC VARIABILITY DETERMINATION OF PATHOGENIC PROTOTHECA<br />

Sara Marques 1 , Eliane Silva 1 , Gertrude Thompson 1 , Volker Huss 2<br />

1<br />

University <strong>of</strong> Porto, ICBAS – CIBIO, Porto, Portugal<br />

2<br />

University <strong>of</strong> Erlangen-Nuremberg, Biology Department – MPP, Erlangen, Germany<br />

Prototheca spp.; bovine mastitis; internal transcribed spacer (ITS); 16S rDNA; intergenic spacer (IGS)<br />

Introduction<br />

Members <strong>of</strong> the genus Prototheca are ubiquitous nonphotosynthetic<br />

green algae that can be associated with<br />

pathologies in humans and animals (1). The known pathogenic<br />

species in the genus are P. zopfii, P. wickerhamii and P.<br />

blaschkeae (2). Both P. zopfii and P. blaschkeae have been<br />

associated with bovine mastitis (1, 2). Presently this pathology is<br />

recognized as endemic worldwide and is considered a public<br />

health problem (3). Prototheca identification is generally achieved<br />

by phenotypic and molecular characterization, the latter being<br />

used to easily identify Prototheca species. Phylogenetic<br />

relationships <strong>of</strong> Prototheca can be inferred from 18S and 28S<br />

rDNA sequence comparisons (1, 2, 4). The ribosomal internal<br />

transcribed spacer (ITS) regions and plastid ribosomal RNA<br />

operon are less conserved than 18S and 28S rRNA genes and<br />

provide greater interspecific differences that are useful for the<br />

differentiation <strong>of</strong> strains (5). Thus, the aim <strong>of</strong> the present study<br />

was to determine the genotypic variability within P. zopfii and P.<br />

blaschkeae strains by amplification and sequencing <strong>of</strong> these<br />

regions.<br />

Materials & methods<br />

Prototheca isolates used in this study belong to a major collection<br />

<strong>of</strong> milk pathogens obtained from different dairy herds from the<br />

Northwest <strong>of</strong> Portugal. Thirty seven isolates were previously<br />

identified as P. zopfii genotype 2 and five as P. blaschkeae (1).<br />

These isolates were spread and grown on Sabouraud Dextrose<br />

Agar medium (Merck Laboratories, Darmstadt, Germany). In this<br />

study PCR amplification <strong>of</strong> the nuclear ITS region and plastid 16S<br />

rDNA, intergenic spacer (IGS) and partial 23S rDNA were<br />

performed. Briefly, genomic DNA preparation was performed as<br />

previously described (1). For amplification <strong>of</strong> the ITS region<br />

several sets <strong>of</strong> primers were applied and some additives were<br />

used to improve the reaction. The amplification products were<br />

analysed on a 0.8% (wt/vol) agarose gel after staining with<br />

ethidium bromide. Since some <strong>of</strong> the amplifications produced two<br />

bands, these were purified with the QIAquick ® PCR purification kit<br />

(Qiagen, Crawley, UK) and cloned into Topo ® XL PCR Cloning<br />

Kit (Invitrogen, Carlsbad, USA). As some sequences displayed<br />

similarities to the Prototheca plastid IGS region between 16S and<br />

23S rDNA, several other primers for the ITS region were tested<br />

and amplification <strong>of</strong> the 16S rDNA, IGS and part <strong>of</strong> the 23S rDNA<br />

were also performed.<br />

Results<br />

ITS amplification <strong>of</strong> Prototheca isolates retrieved bands <strong>of</strong> the<br />

expected size only in few cases (Fig. 1). Sequencing <strong>of</strong> the<br />

amplified products retrieved nuclear ITS <strong>of</strong> only 3 P. zopfii strains<br />

with some differences between them. No ITS sequences were<br />

obtained for P. blaschkeae.<br />

zopfii isolates had identical sequences, and the 5 P. blaschkeae<br />

isolates sequences could be assigned to 2 groups which differed<br />

in two positions within the 16S rRNA gene and one position in the<br />

tRNA-alanine gene.<br />

Figure 2: 16S rDNA amplification <strong>of</strong> some P. zopfii (Pz1-2) and P.<br />

blaschkeae (Pb1-2) isolates with 16S universal primers. All PCR<br />

fragments have a size close to the expected size <strong>of</strong> about 1,500<br />

bp. Lane EcoRI + HindIII – molecular weight standard, -C,<br />

negative control without DNA.<br />

Discussion & conclusions<br />

The variability <strong>of</strong> nuclear ITS and plastid rDNA sequences<br />

between and within P. zopfii and P. blaschkeae strains is shown<br />

for the first time. Within the ITS sequences <strong>of</strong> the P. zopfii<br />

isolates, similarities between 92 and 96% were found. The plastid<br />

sequences obtained for P. zopfii and P. blaschkeae isolates<br />

showed only 82.7% similarity between each other. Similarity <strong>of</strong><br />

the complete 16S rDNA alone was 85.3%, which was lower than<br />

that <strong>of</strong> the respective 18S rDNA (98.0%) (1). Solving the<br />

amplification problem and sequencing <strong>of</strong> the ITS region together<br />

with the plastid rRNA operon from all isolates will be <strong>of</strong> great<br />

value for Prototheca population genetics and epidemiology.<br />

Acknowledgements<br />

This work was supported by Fundação para a Ciência e<br />

Tecnologia, Portugal, grant SFRH/BD/28892/2006.<br />

References<br />

1. Marques, S, Silva, E, Kraft, C, Carvalheira, J, Videira, A, Huss, V,<br />

Thompson, G (2008). Bovine mastitis associated with Prototheca<br />

blaschkeae. J Clin Microbiol; 46, 1941-1945.<br />

2. Janosi, S, Ratz, F, Szigeti, G, Kulcsar, M, Kerenyi, J, Lauko, T, Katona,<br />

F, Huszenicza, G (2001). Review <strong>of</strong> the microbiological, pathological, and<br />

clinical aspects <strong>of</strong> bovine mastitis caused by the alga Prototheca zopfii. Vet<br />

Q, 23, 58-61.<br />

3. Malinowski, E, Lassa, H, Klossowska, A (2002). Isolation <strong>of</strong> Prototheca<br />

zopfii from inflamed secretion <strong>of</strong> udders. Bull Vet Inst Pulawy, 46, 295-299.<br />

4. Moller, A, Truyen, U, Roesler, U (2007). Prototheca zopfii genotype 2:<br />

the causative agent <strong>of</strong> bovine protothecal mastitis? Vet Microbiol, 120,<br />

370-374.<br />

5. Pröschold, T, Darienko, T, Silva, PC, Reisser, W, Krienitz, L (2011). The<br />

systematics <strong>of</strong> Zoochlorella revisited employing an integrative approach.<br />

Environ Microbiol, 13, 350-364.<br />

Figure 1: ITS amplification <strong>of</strong> some P. zopfii (Pz1-6) and P.<br />

blaschkeae (Pb) isolates with ITS primers. All PCR fragments<br />

should have a size <strong>of</strong> approximately 1,300 bp, however different<br />

sizes were found and in some isolates two bands were amplified.<br />

Lane ClaI – molecular weight standard, -C, negative control<br />

without DNA.<br />

The complete plastid 16S rDNA <strong>of</strong> most <strong>of</strong> the isolates was<br />

amplified (Fig. 2). Almost complete sequences <strong>of</strong> the plastid<br />

rRNA operon were obtained for 8 Prototheca isolates. Three P.


S1 - P - 26<br />

DETECTION OF SAXITOXINE (PSP MARINE BIOTOXIN) IN MOLLUSCS BY ELISA<br />

Miroslaw M. Michalski, Katarzyna Grądziel-Krukowska<br />

National Veterinary Research Institute, Department <strong>of</strong> Hygiene <strong>of</strong> Food <strong>of</strong> Animal Origin, Puławy, Poland<br />

Head <strong>of</strong> the Department: pr<strong>of</strong>. Jacek Osek<br />

Marine biotoxins, saxitoxine, Elisa, intoxication<br />

Introduction<br />

Marine biotoxins are a group <strong>of</strong> natural toxins that sometimes<br />

accumulate in shellfish. Many biotoxins are produced by marine<br />

algae (phytoplankton, including diatoms and din<strong>of</strong>lagellates) and<br />

can accumulate in fish or shellfish if they ingest these algae.<br />

There are several types <strong>of</strong> illnesses, caused by marine biotoxins,<br />

that are connected with the consumption <strong>of</strong> contaminated<br />

shellfish. They include Paralytic Shellfish Poisoning (PSP), and<br />

Diarrhetic Shellfish Poisoning (DSP), Amnesic Shellfish<br />

Poisoning (ASP). Saxitoxin is a naturally produced marine<br />

biotoxin by several gonyaulacoid or gymnodinioid din<strong>of</strong>lagellates,<br />

including Alexandrium, Gymnodinium, Pyrodinium, and has also<br />

been found in freshwater cyanobacterial strains such as<br />

Cylindrospermopsis raciborskii<br />

The toxins responsible for PSP are heterocyclic guanidines<br />

(saxitoxins) and there are over 21 known congeners. Substitution<br />

at R4 results in substantial changes in toxicity. Saxitoxin (STX)<br />

binds with a high affinity to site 1 on the voltage dependent<br />

sodium channel, inhibiting channel opening (1, 3)<br />

Table 1<br />

Type <strong>of</strong> shellfish<br />

1.Mule<br />

Mytilus edulis<br />

2.Oysters<br />

Crassostrea gigas<br />

3.Vongole<br />

Tapes<br />

semidecussatus<br />

4.Scallops<br />

Pecten spp.<br />

Number <strong>of</strong><br />

samples<br />

Number <strong>of</strong> samples:<br />

bld 1 /pos 2 /exc 800<br />

µg/ 3<br />

24 5/19/0<br />

13 3/10/0<br />

Country <strong>of</strong><br />

origin<br />

Holland,<br />

Norway,<br />

France<br />

Holland,<br />

France<br />

12 2/10/0 Italia<br />

10 2/7/0 Holland<br />

Total numbers <strong>of</strong><br />

sam-samples<br />

59<br />

1 - bellow limit <strong>of</strong> detection<br />

2 - range 50-800 µg/kg<br />

3 – concentartion in molluscs meat more than 800 µg/kg<br />

PSP toxic syndrome is due primarily to the consumption <strong>of</strong><br />

molluscs contaminated by PSP toxins as a result <strong>of</strong> filter-feeding<br />

by toxic din<strong>of</strong>lagellates. After intoxication <strong>of</strong> PSP, the effects are<br />

predominantly neurological and include tingling, burning,<br />

numbness, drowsiness, incoherent speech, and respiratory<br />

paralysis. Symptoms <strong>of</strong> the disease develop fairly rapidly, within<br />

0.5 to 2 hours after ingestion <strong>of</strong> the shellfish, depending on the<br />

amount <strong>of</strong> toxin consumed. In severe cases respiratory paralysis<br />

is common, and death may occur if respiratory support is not<br />

provided (1, 2, 3). In the EU law a limit for PSP toxins in bivalve<br />

molluscs is laid down at 80 g STX eq/100 g <strong>of</strong> shellfish meat (4).<br />

Materials & methods<br />

The RIDACSREEN ® FAST PSP SC test was used for the<br />

determination <strong>of</strong> PSP toxins in shellfish. This assay is a direct<br />

competitive Elisa for PSP and related algae toxins in mussels. All<br />

reagents are contained in the test kit. Detection limit <strong>of</strong> test is 50<br />

µg/kg <strong>of</strong> shellfish meat. A total <strong>of</strong> four different types <strong>of</strong> mussels<br />

were used for analysis. Live shellfish samples used in this study<br />

were collected from warehouses and markets. A total number <strong>of</strong><br />

59 samples were investigated. Preparation <strong>of</strong> samples and Elisa<br />

test was performed according to the test producer instruction.<br />

Discussion & conclusions<br />

Fifty nine samples <strong>of</strong> shellfish meat were analysed by the Elisa<br />

test. In 46 samples PSP biotoxins were found, expressed as<br />

saxitoxin-HCl, in range 50-800 µg/kg. In 12 samples the<br />

concentration <strong>of</strong> PSP toxins was bellow the limit <strong>of</strong> the detection<br />

<strong>of</strong> the test and no sample with more than 800 µg/kg <strong>of</strong> PSP toxins<br />

in mussels. The present experiment clearly showed that the<br />

shellfish available on Polish marked are, as a food, safe for<br />

consumers.<br />

References<br />

1. EFSA. 2009. Marine biotoxins in shellfish – Saxitoxin group.<br />

Scientific Opinion <strong>of</strong> the Panel on Contaminants in the Food<br />

Chain. The EFSA Journal, 2009, 1019, 1-76<br />

2. Michalski Miroslaw. Biotoksyny morskie - występowanie i metody<br />

analizy. Żywność.Nauka.Technologia.Jakość. 2006, 3(48), 16-22<br />

3. Michalski Mirosław: Paralityczne toksyny morskie jako zagrożenie dla<br />

zdrowia konsumenta. Medycyna Weterynaryjna 2007, 63(12), 1530-1533<br />

4. Regulation (EC) No 853/2004 <strong>of</strong> The European Parliament and <strong>of</strong> the<br />

Council <strong>of</strong> 29 April 2004 laying down specific hygiene rules for food <strong>of</strong><br />

animal origin.<br />

Results<br />

The content <strong>of</strong> saxitoxine determined by the Elisa test in the four<br />

species <strong>of</strong> shellfish is shown in table 1.


S1 - P - 27<br />

PROFICIENCY TESTING (PT) FOR CHEMICAL LABORATORIES PERFORMING ANALYSIS OF MEAT<br />

AND MEAT PRODUCTS ORGANISED BY NVRI IN PULAWY<br />

Michalski Miroslaw<br />

National Veterinary Research Institute, Department <strong>of</strong> Hygiene <strong>of</strong> Food <strong>of</strong> Animal Origin, Puławy, Poland<br />

Head <strong>of</strong> the Department: pr<strong>of</strong>. Jacek Osek<br />

Pr<strong>of</strong>iciency testing, z-score, chemical analysis<br />

Introduction<br />

The aim <strong>of</strong> this study is the qualification <strong>of</strong> laboratories with help<br />

<strong>of</strong> interlaboratory pr<strong>of</strong>iciency testing (PT) in range <strong>of</strong> analysed<br />

substances. Results <strong>of</strong> analyses should be credible with<br />

previously determined standard uncertainty <strong>of</strong> measurement.<br />

The harmonization <strong>of</strong> correct results <strong>of</strong> chemical analysis is<br />

secure by the validation <strong>of</strong> analytic methods, standard uncertainty<br />

for individual measurement or for the method <strong>of</strong> analysis, using<br />

certified reference materials <strong>of</strong> and participation in pr<strong>of</strong>iciency<br />

testing (3). Information on the precision and accuracy <strong>of</strong> the<br />

results are to be taken into consideration in the design <strong>of</strong> the<br />

assay (3,4,5). In case <strong>of</strong> lack <strong>of</strong> reference materials or reference<br />

certified materials, participation in PT is the only method for<br />

confirming the technical competences <strong>of</strong> the laboratory (1,5).<br />

Materials & methods<br />

Pasteurized meat product was used for analysis. Every<br />

participant received sample together with instruction <strong>of</strong><br />

transportation as well as a document which allows to introduce<br />

the sample in a quality system <strong>of</strong> laboratory. In this sample<br />

laboratory personnel should analyse at least one <strong>of</strong> parameters<br />

as sodium nitrate, salt (NaCl) by Mohr method, water, fat,<br />

phosphorus, proteins, ash, hydroxyproline, and starch. Deadline<br />

<strong>of</strong> realisation <strong>of</strong> analyses was appointed by organizer. The<br />

homogeneity <strong>of</strong> material was estimated on the basis investigation<br />

<strong>of</strong> eight random choosen samples from designed to analysis,<br />

applying the criterion <strong>of</strong> the homogeneity Ss0,3 .<br />

Statistical calculations and homogeneity assessments have been<br />

conducted in accordance with the principles stipulated in ISO<br />

13528:2005 “Statistical methods for use in pr<strong>of</strong>iciency testing by<br />

inter-laboratory comparisons”.<br />

In the pr<strong>of</strong>iciency test 25 laboratories participated (<strong>of</strong>ficial<br />

veterinary labs as well as private) and 31 analysts. Laboratory<br />

code numbers were known only for laboratory and organizer. The<br />

participants <strong>of</strong> PT applied the standardized method or such,<br />

which they use during normal analysis, after their validation.<br />

Reference values for each analysed parameter were calculated<br />

with a help <strong>of</strong> algorithm A, according to Standard PN-ISO 57255:<br />

2000, based on results from participating laboratories.<br />

The calculated reference values as well as standard deviations<br />

made it possible to calculate z-score accoprding to the formula:<br />

z <br />

*<br />

x<br />

X<br />

*<br />

s<br />

where,<br />

s*- standard deviation for pr<strong>of</strong>iciency assessment<br />

X*- assigned value<br />

x - participant result<br />

For the purposes <strong>of</strong> performance assessment for a single round,<br />

z scores are interpreted as follows:<br />

z≤ 2.00 satisfactory result<br />

2.00 < zand < 3.00 questionable result<br />

z≥ 3.00 unsatisfactory result<br />

Table1: Results <strong>of</strong> statistical calculation for tested samples<br />

NaNO 3<br />

[mg/kg]<br />

NaCl<br />

[g/100g]<br />

Nitrogen<br />

[g/100g]<br />

Water<br />

[g/100]<br />

Fat<br />

[g/100g]<br />

Phosphoru<br />

s [mg/kg]<br />

Ash<br />

[g/100g]<br />

Starch<br />

[g/100g]<br />

Hydroxypr<br />

oline<br />

[g/100g]<br />

Reference<br />

value 18,39 2,22 2,84 76,07 1,09 4308,3 2,91 1,78 0,040<br />

Std 3,39 0,05 0,03 0,12 0,12 100,92 0,03 0,16 0,005<br />

Coefficient<br />

<strong>of</strong> variation<br />

cv (%)<br />

18,43 2,25 1,06 0,16 11,01 2,34 1,03 8,99 12,50<br />

Table 2: The number <strong>of</strong> results for respective parameters, which<br />

z score carried out |z| > 2<br />

Number <strong>of</strong><br />

results<br />

2


S1 - P - 28<br />

SPECIFIED RISK MATERIAL REMOVAL PRACTICES:<br />

CAN WE REDUCE THE BSE HAZARD TO HUMAN HEALTH?<br />

D. Pitardi 1 , D. Meloni 1 , C. Maurella 1 , D. Di Vietro 1 , L. Nocilla 1 , A. Piscopo 2 , E. Pavoletti 3 , M. Negro 4 , M. Caramelli 1 ,<br />

E. Bozzetta 1 .<br />

1 Istituto Zoopr<strong>of</strong>ilattico Sperimentale del Piemonte Liguria e Valle d’Aosta, Via Bologna 148, 10154, Torino, Italy.<br />

2 Azienda Sanitaria Provinciale 1AG, Viale della Vittoria 321, 92100, Agrigento, Italy.<br />

3 Azienda Sanitaria Locale VC, Via Benadir 35, 13100, Vercelli, Italy.<br />

4 Azienda Sanitaria Locale CN1, Corso Francia 12, 12100, Cuneo, Italy.<br />

BSE; vCJD; Spinal cord; SRM Contamination; Alternative slaughter practices.<br />

Introduction<br />

Following the bovine spongiform encephalopathy (BSE)<br />

epidemics across Europe in the early 1990s, the removal <strong>of</strong><br />

designated BSE specified risk material (SRM) became<br />

mandatory to minimize the risk to consumers <strong>of</strong> exposure to the<br />

infectious agent. Despite this preventive measure, crosscontamination<br />

<strong>of</strong> edible meat with SRM can occur during<br />

conventional slaughter (1-4).<br />

Currently, there are no markers that can identify the presence <strong>of</strong><br />

SRM in meat as a whole. Nevertheless, some assays are able to<br />

detect traces <strong>of</strong> CNS, hence this parameter is <strong>of</strong>ficially taken as<br />

indicative <strong>of</strong> SRM contamination.<br />

In this two-stage study, we carried out a survey to estimate the<br />

prevalence <strong>of</strong> carcass contamination at two slaughterhouses, one<br />

large and the other medium-sized; we then compared three<br />

different methods (conventional vs. suction vs. water-jet) for<br />

spinal cord removal employed at the large slaughterhouse to<br />

assess their performance in preventing the spread <strong>of</strong> CNT over<br />

the carcass.<br />

Materials & methods<br />

Prevalence <strong>of</strong> CNS contamination<br />

The prevalence <strong>of</strong> CNS contamination by the conventional<br />

technique was estimated from a total <strong>of</strong> 216 carcasses from a<br />

large slaughterhouse and 196 from a medium-sized one. In both<br />

abattoirs the carcasses were split with a hand-guided belt-type<br />

saw and the spinal cord cut along its length was removed from<br />

each side <strong>of</strong> the carcass. Sampling was performed immediately<br />

after spinal cord removal.<br />

Alternative techniques<br />

Two alternative spinal cord removal techniques were compared<br />

to the conventional method. In both techniques, the SRM is<br />

extracted before the carcass is split (by suction or by water-jet).<br />

Comparative study<br />

The estimated sample size (50 carcasses) was sampled for each<br />

SRM removal method: conventional; suction; and water-jet. The<br />

specimens were withdrawn immediately after carcass splitting.<br />

Sample collection and preparation<br />

Bovine older than 12 months were included in the study. Samples<br />

were collected from a defined area on the medial surface <strong>of</strong> each<br />

half <strong>of</strong> the split carcass. The area was selected and marked <strong>of</strong>f<br />

on the paravertebral muscles.<br />

Testing activity<br />

A commercially available ELISA kit (Ridascreen® Risk Material<br />

10/5, R-Biopharm), which detects GFAP as a marker for CNS,<br />

was used to analyze the samples. In order to facilitate application<br />

<strong>of</strong> the test for screening purposes, in previous studies we<br />

validated its qualitative use by plotting an ROC curve to set a<br />

useful cut-<strong>of</strong>f value (2).<br />

Results<br />

Using a qualitative approach, samples were defined as positive if<br />

CNS tissue was detected at a concentration ≥0.049% (2).<br />

Samples tested positive in 130/216 carcasses (60.2%, 95% CI<br />

53.3-66.8) from the large slaughterhouse and in 152/196 (77.6%,<br />

95% CI, 71-83.2) <strong>of</strong> those from the medium-sized one (Table I).<br />

The conventional slaughter technique was associated with an<br />

overall prevalence <strong>of</strong> CNS contamination <strong>of</strong> 68.4% (95% CI, 53 -<br />

83).<br />

The comparative study showed a CNS contamination <strong>of</strong> 62%<br />

(95% CI, 47.2-75.3) associated with the conventional technique,<br />

60% (95% CI, 45.2-73.6) with the suction technique, and 36%<br />

(95% CI, 22.9-50.8) with the water-jet system (Table II). The<br />

difference among the three methods appeared to be significant<br />

(P=0.0047).<br />

Table I: Conventional method - positive samples stratified<br />

according to contamination level and type <strong>of</strong> abattoir.<br />

Large abattoir Medium abattoir<br />

CNS contamination positive samples positive samples<br />

(%)<br />

(%)<br />

Low ( ≥ 0.049 and


S1 - P - 29<br />

LOW PATHOGENIC AVIAN INFLUENZA VIRUS INFECTIONS ON POULTRY FARMS IN THE<br />

NETHERLANDS<br />

Sylvia Pritz-verschuren 1 , Jeanet van der Goot 1 , Johan Bongers 1 , Guus Koch 1<br />

1<br />

Central Veterinary Institute <strong>of</strong> Wageningen UR,Lelystad,The Netherlands<br />

Avian Influenza Virus, low pathogenic, poultry<br />

Introduction<br />

Outbreaks <strong>of</strong> highly pathogenic avian influenza (HPAI) have a<br />

devastating economic impact on the poultry industry and also<br />

causes social distress. In countries free <strong>of</strong> HPAI, epidemics start<br />

after introduction <strong>of</strong> low pathogenic AI virus which may evolves to<br />

HPAI viruses. Early detection <strong>of</strong> AI infection is crucial for the<br />

control <strong>of</strong> outbreaks. Infections <strong>of</strong> LPAI viruses in laying birds is<br />

subclinical or causes mild symptoms such as a drop in egg<br />

production.<br />

Early detection <strong>of</strong> AI virus infection is crucial to prevent HPAI<br />

outbreaks. Therefore, in the Netherlands LPAI infections are<br />

detected by early warning and compulsory serological monitoring<br />

programmes <strong>of</strong> all poultry farms.<br />

In the period <strong>of</strong> 2006-2011, an increase in the number <strong>of</strong> LPAI<br />

virus infections in poultry was observed. A number <strong>of</strong> possible<br />

causes is discussed.<br />

Materials & methods<br />

Serological samples were screened in an competitive NP<br />

antibody ELISA and the positive samples were confirmed and<br />

typed by hemagglutination inhibition and neuraminidase inhibition<br />

test.<br />

For detection <strong>of</strong> the virus an in house developed PCR with the M-<br />

gene as target is used. Positive samples were tested in an H5<br />

and H7 specific PCR described by the community EU reference<br />

lab (AHVLA, Weybridge). H5 and H7 negative samples were<br />

subtyped using a generic, hemagglutinin and neuraminidasespecific<br />

RT-PCR (1) followed by sequencing the PCR product.<br />

Results<br />

The number LPAI introductions either detected by PCR and/or<br />

antibodies increased in the period 2006-2011, especially since<br />

2009 (Table 1). Although there is a marked increase in the<br />

number <strong>of</strong> detections, the period is too short to speak about a<br />

tendency.<br />

Table 1. Number <strong>of</strong> poultry farms where LPAI viruses and/or<br />

antibodies were detected in period 2006-2011<br />

Year<br />

Number <strong>of</strong> LPAI Number <strong>of</strong> primary<br />

detected farms infections<br />

2006 4 2<br />

2007 13 8<br />

2008 10 9<br />

2009 11 10<br />

2010 20 18<br />

2011 33 23<br />

Total 91 70<br />

A number <strong>of</strong> possible causes for the increase <strong>of</strong> LPAI detections<br />

are:<br />

More intensive search for LPAI viruses and introduction <strong>of</strong> the<br />

early warning.<br />

The early warning program was started in 2008/2009. However<br />

most farms (57/70) are detected by serological monitoring which<br />

was in place since 2004. So, this can’t be an explanation for the<br />

increase.<br />

Higher sensitivity <strong>of</strong> the serological test used for the monitoring.<br />

Sera are tested in an ELISA by the Dutch Animal Health Service<br />

(AHS). In January 2009 the AHS has changed to another test.<br />

However, the validation tests revealed no differences in<br />

sensitivity between the old and new test.<br />

<strong>of</strong> the Erasmus Medical Centre in Rotterdam there is no<br />

sufficient evidence to assume such an increased prevalence <strong>of</strong><br />

LPAI viruses in wild birds.<br />

An increase <strong>of</strong> the number wild birds in the Netherlands.<br />

Reports <strong>of</strong> Dutch Bird research group (SOVON) shows that the<br />

number <strong>of</strong> wild birds was more or less constant in the period<br />

2004-2009. Thus this also can’t explain the increase <strong>of</strong> the<br />

number <strong>of</strong> detection.<br />

More contact between poultry and wild birds due to the increased<br />

number <strong>of</strong> outdoor-layer farms.<br />

Data <strong>of</strong> the Product Boards for Livestock, Meat and Eggs (PVE)<br />

shows that the total number <strong>of</strong> outdoor-layer farms was lower in<br />

2008 and 2009 than in the years before and gradually increased<br />

again in 2010 and 2011.<br />

Different subtypes are found on the poultry farms (Table 2). In<br />

poultry H5, H7 and H8 viruses and/or antibodies are the most<br />

common in the period 2006-2011. For serology the<br />

neuraminidase inhibition test is used for typing, however the test<br />

is not suitable to test large numbers <strong>of</strong> sera samples and<br />

therefore the neuraminidase type is not yet determined for most<br />

farms, because 57/70 farms were detected in the serological<br />

monitoring programme. Therefore the most common N-type is<br />

not known.<br />

Table 2. Avian Influenza virus subtypes detected in poultry farms<br />

in the Netherlands in period 2006-2011<br />

HA N1 N2 N3 N4 N5 N6 N7 N8 N9 N?* Totaal<br />

H1 2 1 3 6<br />

H2 2 1 3<br />

H3 2 2<br />

H4<br />

H5 1 10 11<br />

H6 1 1 3 5<br />

H7 1 1 2 4 4 12<br />

H8 3 9 12<br />

H9 3 1 4<br />

H10 2 1 3<br />

H11<br />

H12 1 1<br />

H13<br />

H14 1 1<br />

H16<br />

H?* 1 13 14<br />

Totaal 4 5 3 5 2 6 49 74<br />

* Typing <strong>of</strong> the haemagglutinine and/or neuraminidase was not<br />

possible.<br />

Discussion & conclusions<br />

Every year in the Netherlands more infections with LPAI viruses<br />

are detected in poultry farms . Up to now we did not identify an<br />

obvious explanation for the increase <strong>of</strong> LPAI virus introduction in<br />

poultry. However, different subtypes are found. The most<br />

common H-types are H5, H7 and H8.<br />

References<br />

1. Gall,A, H<strong>of</strong>fmann, B, Harder, Timm, Grund, C, Ehricht, R, Beer, M<br />

(2009). Rapid an Highly Sensitive Neuramidase Subtyping <strong>of</strong> Avian<br />

Influenza Viruses by Use <strong>of</strong> a Diagnostic DNA Microarray. J. <strong>of</strong> Clin.<br />

Microbiol., 47, 2985-2988<br />

A increased prevalence <strong>of</strong> LPAI viruses in wild birds.<br />

The prevalence varies per season, year, wild bird and location,<br />

but from a wild bird surveillance studies performed by the group


S1 - P - 30<br />

DETECTION OF BRACHYSPIRA HYODYSENTERIAE AND BRACHYSPIRA PILOSICOLI IN SWISS PIGS<br />

USING A COMBINATION OF CULTURE AND PCR<br />

Sarah Prohaska 1 , Helen Huber 1 , Titus Sydler 2 , Max M. Wittenbrink 1<br />

1 Institute <strong>of</strong> Veterinary Bacteriology, Vetsuisse Faculty, University <strong>of</strong> Zurich, Switzerland<br />

2<br />

Institute <strong>of</strong> Veterinary Pathology, Vetsuisse Faculty, University <strong>of</strong> Zurich, Switzerland<br />

Introduction<br />

Swine dysentery and porcine intestinal spirochaetosis caused by<br />

pathogenic Brachyspira (B.) spp. (B. hyodysenteriae and<br />

B. pilosicoli) lead to considerable economic loss and are<br />

therefore <strong>of</strong> increasing importance to the pig industry.<br />

B. hyodysenteriae colonise the epithelium <strong>of</strong> the large intestine<br />

and subsequently induce severe mucohaemorrhagic colitis<br />

showing the typical signs <strong>of</strong> diarrhoea containing blood and plugs<br />

<strong>of</strong> mucus (1). Infections with B. pilosicoli however are<br />

characterised by moderate growth loss and mild diarrhoea (2).<br />

The goal <strong>of</strong> this study was to evaluate the presence <strong>of</strong><br />

B. hyodysenteriae and B. pilosicoli in Swiss pig herds by a<br />

combination <strong>of</strong> culture and PCR.<br />

Swine, Brachyspira spp., culture, PCR<br />

Materials & methods<br />

Over a time period <strong>of</strong> three years (2009-2011) ligated colon<br />

sections (n=140) from dissected pigs as well as faecal swabs<br />

(n= 661) collected on 228 Swiss pig farms were subjected to a<br />

cultural assay and subsequent PCR for the detection <strong>of</strong><br />

B. hyodysenteriae and B. pilosicoli. Faecal swabs were taken<br />

from pigs suffering from acute diarrhoea.<br />

For culture, the selective BJ-Agar (Trypticase soy agar<br />

supplemented with 5% cattle blood and Colistin, Vancomycin,<br />

Spectinomycin, Spriramycin and Rifampicin) was used (3). Plates<br />

were incubated under anaerobic conditions at 42°C for 4-6 days.<br />

To evaluate the presence <strong>of</strong> Brachyspira spp., native<br />

preparations <strong>of</strong> colony material were investigated by darkfield<br />

microscopy. If spirochaetes were found, DNA was extracted from<br />

suspicious colonies. Subsequently, PCR using two primer pairs<br />

for the detection <strong>of</strong> B. hyodysenteriae and B. pilosicoli was<br />

performed (4).<br />

Results<br />

In two <strong>of</strong> the 140 (1.4%) tested colon sections B. hyodysenteriae<br />

could be detected, whereas B. pilosicoli was not found in these<br />

samples (Figure 1).<br />

In terms <strong>of</strong> faecal swabs, cultivation and subsequent confirmation<br />

by PCR yielded B. hyodysenteriae in 122 (18.5%) samples and<br />

B. pilosicoli in 33 samples (5.0%). However, analysis <strong>of</strong> seven<br />

(1.0%) swabs revealed a co-infection <strong>of</strong> B. hyodysenteriae and<br />

B. pilosicoli (Figure 2).<br />

Figure 2: Results <strong>of</strong> faecal swabs<br />

Discussion & conclusions<br />

The combination <strong>of</strong> culture and PCR for diagnosis <strong>of</strong> Swine<br />

dysentery and intestinal spirochaetosis pro<strong>of</strong>ed to be a<br />

successful tool.<br />

The results <strong>of</strong> this study show a remarkable presence <strong>of</strong><br />

pathogenic Brachyspira spp. in Switzerland. B. hyodysenteriae<br />

was detected in samples taken from pigs from 12 <strong>of</strong> 26 cantons<br />

in Switzerland. Due to this situation, the Swiss pig health service<br />

implemented a monitoring system <strong>of</strong> pathogenic Brachyspira spp.<br />

starting <strong>2012</strong>.<br />

References<br />

1. Hampson D.J., Atyeo R.F., Combs B.G., 1997. Swine Dysentery 175-<br />

209. In: Intestinal Spirochaetes in Domestic Animals and Humans<br />

(Hampson D.J. and Stanton T.B.), CAB International, Wallingford, UK.<br />

2. Taylor D.J., Simmons J.R., Laird H.M., 1980. Production <strong>of</strong> diarrhoea<br />

and dysentery in pigs by feeding pure cultures <strong>of</strong> a spirochaete differing<br />

from Treponema hyodysenteriae. Vet. Rec. 106, 326-332.<br />

3. Dünser M., Schweighardt H., Pangerl R., Awad-Masalmeh M., Schuh<br />

M., 1997. Schweinedysenterie und Sprirochaetendiarrhoe – vergleichende<br />

Untersuchungen serpulinenbedingter Enteritiden. Wien. Tierärztl. Mschr.<br />

84, 151-161.<br />

4. La T., Philips N.D., Hampson D.J. 2003. Development <strong>of</strong> a Duplex PCR<br />

Assay for Detection <strong>of</strong> Brachyspira hyodysenteriae and Brachyspira<br />

pilosicoli in Pig Feces. J.Clin. Microbiol. 41, 3372-3375.<br />

Figure 1: Results <strong>of</strong> ligated colon samples


S1 - P - 31<br />

PRRSV DIAGNOSTICS BY RT-PCR AND DIFFERENT ELISA SYSTEMS IN SERUM AND ORAL FLUID<br />

OF PRRSV VACCINATED PIGS<br />

Tatjana Sattler 1 , Sandra Revilla-Fernández 2 , Adolf Steinrigl 2 , Manfred Berger 2 , Friedrich Schmoll 2<br />

1<br />

University Leipzig, Large Animal Clinic for Internal Medicine, Leipzig, Germany<br />

2<br />

AGES, Institute for Veterinary Disease Control, Mödling, Austria<br />

PRRSV, ELISA, PCR, oral fluid, pig<br />

Introduction<br />

PRRSV infection is widely distributed all over the world. PRRSV<br />

eradication programs have been developed and PRRSV negative<br />

herds and farms have been established. PRRSV diagnostic tools<br />

with high sensitivity and specificity are essential to control the<br />

disease and to guarantee the negative status <strong>of</strong> the herds (1).<br />

Furthermore, specimens like oral fluid are advantageous in terms<br />

<strong>of</strong> costs, working time and animal welfare, and therefore have<br />

been recently proposed for disease monitoring (2). The objective<br />

<strong>of</strong> this study was therefore to evaluate PRRSV RNA and antibody<br />

detection in pig oral fluid by RT-qPCR and commercial ELISA<br />

test systems, respectively. Furthermore, the performance <strong>of</strong><br />

different ELISA systems was compared both, in blood serum and<br />

oral fluid <strong>of</strong> pigs.<br />

Materials & methods<br />

Ten pigs (from 8 to 20 weeks <strong>of</strong> age) from a PRRSV negative<br />

farm were injected with an EU-type PRRSV life vaccine (Porcilis ®<br />

PRRS, Intervet, Unterschleißheim, Germany). Blood and oral<br />

fluid samples were taken individually from each pig before, and 4,<br />

7, 14 and 21 days after vaccination. After automated RNA<br />

extraction, serum and oral fluid samples were analysed with an<br />

in-house PRRSV RT-qPCR assay (3).<br />

PRRSV antibody detection in serum and oral fluid was performed<br />

with three different ELISA systems: a) IDEXX PRRS X3 Ab-Test<br />

(IDEXX GmbH, Ludwigsburg, Germany), b) INGEZIM PRRS<br />

UNIVERSAL, c) INGEZIM PRRS DR (both Ingenasa, Madrid,<br />

Spain). Serum samples were diluted according to manufacturer’s<br />

instructions, whereas oral fluid samples were tested without<br />

dilution.<br />

Results<br />

In serum samples (table 1), antibodies were detected in seven<br />

out <strong>of</strong> ten pigs by day 14 after vaccination. One pig<br />

seroconverted at day 21. Two pigs did not seroconvert at all<br />

throughout the study period and also remained RT-qPCR<br />

negative throughout the whole study. Three out <strong>of</strong> ten pigs were<br />

RT-qPCR positive in serum as early as day 4 after vaccination,<br />

while most pigs (7/10) became positive by day 7. One pig<br />

became RT-qPCR positive as late as day 14, while other pigs<br />

became negative in RT-qPCR at this time-point. This animal was<br />

also the last to seroconvert during the study period.<br />

Table 1: Number <strong>of</strong> serum samples positive by PRRSV RT-qPCR<br />

and by three different PRRSV antibody ELISA testkits (a, b, c –<br />

see material and methods)<br />

day 0 (no<br />

vaccination)<br />

day 4 after<br />

vaccination<br />

day<br />

7<br />

day<br />

14<br />

day<br />

21<br />

PCR 0/10 3/10 7/10 5/10 3/10<br />

ELISA<br />

a)<br />

ELISA<br />

b)<br />

ELISA<br />

c)<br />

0/10 0/10 0/10 7/10 8/10<br />

0/10 0/10 0/10 7/10 7/10<br />

0/10 0/10 3/10 7/10 8/10<br />

In oral fluid (table 2), both RT-qPCR and ELISA positive results<br />

(in two <strong>of</strong> the tested ELISA kits) were observed, although<br />

sensitivity was significantly reduced in comparison to serum. The<br />

INGEZIM PRRS DR ELISA (Kit c) did not show positive results at<br />

all in oral fluid.<br />

Table 2: Number <strong>of</strong> oral fluid samples positive by PRRSV RTqPCR<br />

and by three different PRRSV antibody ELISA testkits (a,<br />

b, c – see material and methods)<br />

day 0 (no<br />

vaccination)<br />

day 4 after<br />

vaccination<br />

day<br />

7<br />

day<br />

14<br />

day<br />

21<br />

PCR 0/10 0/10 2/10 2/10 0/10<br />

ELISA<br />

a)<br />

ELISA<br />

b)<br />

ELISA<br />

c)<br />

0/10 0/10 0/10 2/10 3/10<br />

0/10 0/10 0/10 2/10 4/10<br />

0/10 0/10 0/10 0/10 0/10<br />

Discussion & conclusions<br />

PRRSV-antibodies could be detected in serum <strong>of</strong> some pigs as<br />

early as day 7 after vaccination with the INGEZIM PRRS DR<br />

ELISA (Kit c), which was the most sensitive <strong>of</strong> the three ELISA<br />

systems. Results <strong>of</strong> the other two ELISA systems did not differ<br />

substantially from each other, although one pig remained<br />

antibody negative in serum at day 21 with the INGEZIM PRRS<br />

UNIVERSAL (Kit b), while it was found positive in both other<br />

ELISA systems. A seroconversion in this pig, tested with<br />

INGEZIM PRRS UNIVERSAL ELISA, could be expected later<br />

than day 21 after vaccination.<br />

Interestingly, some animals can remain seronegative despite<br />

vaccination, as was the case in this study in two out <strong>of</strong> ten pigs.<br />

The most likely reason was failure <strong>of</strong> the vaccine virus to<br />

replicate in these animals, as indicated by negative RT-qPCR<br />

results at all time-points tested. A similar finding was described in<br />

a previous vaccination study, where one out <strong>of</strong> 20 vaccinated<br />

gilts remained seronegative (4).<br />

Comparison <strong>of</strong> RT-qPCR and ELISA results obtained from<br />

matched oral fluid and serum samples suggest that testing <strong>of</strong>.oral<br />

fluid is much less sensitive. Lower amounts <strong>of</strong> both PRRSV RNA<br />

and antibodies and/or the presence <strong>of</strong> inhibitory factors can be<br />

expected in oral fluid (5). No positive results in oral fluid samples<br />

were found with the INGEZIM PRRS DR ELISA (kit c).<br />

It can be concluded that all tested ELISA systems were similarly<br />

able to detect PRRSV antibodies after PRRSV life virus<br />

vaccination, whereas INGEZIM PRRS DR ELISA seems to be<br />

the most sensitive. Reliable PRRSV antibody detection in oral<br />

fluid will require validation <strong>of</strong> test kits for this medium. RT-qPCR<br />

results confirmed the antibody detection results in both, serum<br />

and oral fluid.<br />

References<br />

1. große Beilage, E, Bätza, HJ. (2007): PRRSV-eradication: An option for<br />

pig herds in Germany? Berl Münch Tierärztl Wochenschr. 120, 11/12, 470-<br />

479.<br />

2. Kittawornrat, A, Prickett, J, Chittick, W, Wang, C, Engle, M, Johnson, J,<br />

Patnayak, D, Schwartz, T, Whitney, D, Olsen, C, Schwartz, K,<br />

Zimmerman, J. (2010): Porcine reproductive and respiratory syndrome<br />

virus (PRRSV) in serum and oral fluid samples from individual boars: will<br />

oral fluid replace serum for PRRSV surveillance Virus Res. 154, 1-2,170-6.<br />

3. Revilla-Fernández S, Wallner B, Truschner K, Benczak A, Brem G,<br />

Schmoll F, Mueller M, Steinborn R.(2005): The use <strong>of</strong> endogenous and<br />

exogenous reference RNAs for qualitative and quantitative detection <strong>of</strong><br />

PRRSV in porcine semen. J Virol Methods. 126, 1-2, 21-30.<br />

4. Sipos, W, Indik, S, Irgang, P, Klein, D, Schuh, M, Schmoll, F (2002):<br />

Transfer <strong>of</strong> EU-ML-PRRSV (Porcilis PRRS) from vaccinated to nonvaccinated<br />

gilts. Proc 17th IPVS Congress, Ames, Iowa, USA, 418<br />

5. Chittick, WA, Stensland, WR, Prickett, JR, Strait, EL, Harmon, K, Yoon,<br />

KJ, Wang, C, Zimmerman, JJ (2010): Comparison <strong>of</strong> RNA extraction and<br />

real-time reverse transcription polymerase chain reaction methods for the<br />

detection <strong>of</strong> Porcine reproductive and respiratory syndrome virus in<br />

porcine oral fluid specimens. J Vet Diagn Invest, 23, 248-53


S1 - P - 32<br />

RELIABLE DETECTION AND TYPING OF PRRSV USING MULTIPLEX REAL-TIME RT-PCR<br />

Christine Gaunitz, Carsten Schroeder, Marco Labitzke, Eva V. Knoop, Jörg Gabert<br />

Labor Diagnostik GmbH Leipzig, Leipzig, Germany<br />

PRRSV, High Pathogenic, real-time RT-PCR, simultaneous detection<br />

Introduction<br />

Infections with the Porcine Reproductive and Respiratory<br />

Syndrome Virus (PRRSV) in swine are very prevalent and<br />

economically most important for the swine industry. Clinical signs<br />

are respiratory disease in piglets and reproductive failure in<br />

pregnant sows.<br />

PRRSV is a single-stranded RNA virus <strong>of</strong> the family Arteriviridae,<br />

order Nidovirales, and classified into the European (EU/I) and<br />

North American (NA/II) genotype. These genotypes show only<br />

55-70 % homology <strong>of</strong> nucleotides in the different genes. In 2006<br />

in China a highly pathogenic (HP) NA-strain emerged, which<br />

shows a deletion <strong>of</strong> around 30 amino acids in the non-structural<br />

protein 2 (Nsp2). Infection with the HP-strain is characterized by<br />

high fever and high mortality in pigs <strong>of</strong> all ages. In the last years<br />

there have been detected also new isolates from Eastern Europe.<br />

Purpose <strong>of</strong> this study was to develop a real-time PCR which<br />

allows reliable detection and differentiation <strong>of</strong> PRRSV strains in<br />

one test run.<br />

Materials & methods<br />

To evaluate analytical sensitivity <strong>of</strong> VIROTYPE ® PRRSV, titration<br />

studies with in-vitro RNA were performed.<br />

Analytical specificity was proven by testing 12 PRRSV reference<br />

strains from cell culture.<br />

Diagnostic sensitivity was proven by testing the EPIZONE ring<br />

trial panel 2011.<br />

Serum, tissue and saliva samples were tested in comparison to<br />

other PCR assays. Two commercial PCR kits and VIROTYPE ®<br />

PRRSV (Labor Diagnostik GmbH Leipzig, Germany) were used<br />

according to the manufacturer´s recommendations.<br />

Results<br />

The high analytical sensitivity <strong>of</strong> the VIROTYPE ® PRRSV kit was<br />

proven by a titration series <strong>of</strong> in vitro RNA (PRRSV<br />

EU1/EU2/NA/HP [10 8 copies/well to 10 copies/well]), performed<br />

in triplicates <strong>of</strong> ten-fold dilutions. A high correlation between<br />

number <strong>of</strong> copies and amount <strong>of</strong> amplification product was<br />

demonstrated in the range <strong>of</strong> 10 8 to 10 3 and 10 8 to 10 2 copies,<br />

respectively (Fig. 1).<br />

PRRSV EU I<br />

PRRSV NA<br />

Figure 1: Evaluation <strong>of</strong> analytical sensitivity.<br />

PRRSV EU II<br />

PRRSV HP<br />

The analytical specificity was proven by testing a RNA-panel <strong>of</strong><br />

12 PRRSV reference strains comparatively with VIROTYPE ®<br />

PRRSV and two commercial available tests (Test I and Test II).<br />

All reference strains were detected well with the VIROTYPE ®<br />

PRRSV. In comparison to Test II, the VIROTYPE ®<br />

PRRSV kit<br />

detects the EU strains around two Ct-values and the NA strains<br />

around five Ct-values more sensitive. The results <strong>of</strong> commercial<br />

test I and VIROTYPE ® PRRSV are comparable (Tab. 1).<br />

Table 1: Evaluation <strong>of</strong> analytical specificity.<br />

VIROTYPE ® PRRSV commercial Test I commercial Test II<br />

Strain<br />

EU NA HP IC PRRSV EU NA IC EU NA IC<br />

Ct<br />

FAM<br />

Ct<br />

TEX<br />

Ct<br />

Cy5<br />

Ct<br />

HEX<br />

Ct<br />

FAM<br />

Ct<br />

FAM<br />

Ct<br />

FAM<br />

Ct HEX<br />

Ct<br />

FAM<br />

Ct<br />

Cy5<br />

Ct<br />

HEX<br />

EU Stendal V953 25.22 - - 34.61 24.62 24.08 - 29.89 24.95 - 35.66<br />

Intervet 22.02 - - 33.22 20.41 19.41 - 28.85 24.65 - -<br />

Cobbelsdorf 23.86 - - 32.65 22.91 22.20 - 29.32 26.02 - -<br />

Stendal V852 23.24 - - 39.96 23.50 21.86 29.68 26.16 - 36.98<br />

Stendal V1904 24.30 - - 39.06 24.48 23.47 29.30 27.63 - 37.41<br />

Stendal V1952/97 26.29 - - 35.41 26.32 24.51 31.32 28.12 - 36.06<br />

Stendal V1445/99 - 21.39 - 34.85 23.80 - 21.17 30.78 - 24.92 34.10<br />

USA VD28775/2 - 21.35 - 29.14 23.94 - 21.61 30.40 - 24.07 39.08<br />

USA VD29949/17 - 22.41 - 28.48 27.72 - 22.18 29.65 - 26.89 -<br />

USA 2 - 19.83 - 30.64 23.07 - 20.44 30.81 - 23.64 -<br />

USA 18 - 19.06 - 31.48 20.47 - 19.50 30.39 - 26.54 -<br />

China - 20.87 22.44 28.36 22.82 - 20.54 29.89 - 25.84 -<br />

Mean Value EU 24.16 23.71 22.59 26.26<br />

Mean Value NA 20.82 23.64 20.91 25.32<br />

In comparison to an in-house PCR (Kleiboeker mod.) performed<br />

at Friedrich-Loeffler-Institute, VIROTYPE ®<br />

PRRSV detects all<br />

samples <strong>of</strong> the Epizone Ring Trial 2011 more sensitive.<br />

VIROTYPE ®<br />

PRRSV reliably detects the samples <strong>of</strong> the EU-2,<br />

EU-3, EU-4 and the atypical EU genotype 1 (Tab. 2).<br />

Table 2: Results from the EPIZONE ring trial panel 2011.<br />

VIROTYPE® PRRSV Kleiboeker (mod.)<br />

PRRSV-strain Genotyp RNA Dil. EU NA HP IC EU NA<br />

EU Stendal V954 EU-1 10 -2 24.96 - - 26.88 28.24 -<br />

EU Stendal V954 EU-1 10 -3 28.41 - - 27.06 31.53 -<br />

EU Stendal V954 EU-1 10 -4 31.61 - - 26.91 34.12 -<br />

EU Stendal V954 EU-1 10 -5 34.63 - - 27.14 37.61 -<br />

EU Stendal V954 EU-1 10 -6 - - - 28.11 - -<br />

EU Stendal V1952/97 EU-1 10 -1 23.08 - - 27.23 25.82 -<br />

EU Stendal V1952/97 EU-1 10 -2 26.57 - - 27.66 29.17 -<br />

EU Stendal V1952/97 EU-1 10 -3 29.54 - - 26.99 32.19 -<br />

EU Stendal V1952/97 EU-1 10 -4 32.50 - - 26.86 35.25 -<br />

EU Stendal V1952/97 EU-1 10 -5 36.92 - - 27.87 40.31 -<br />

Cobbelsdorf EU-1 1:500 23.33 - - 27.26 26.13 -<br />

Stendal V852 EU-1 1:500 23.55 - - 27.54 25.78 -<br />

Stendal V1904 EU-1 1:500 24.37 - - 27.37 26.72 -<br />

Aus_LT3 EU-2 01:10 24.80 - - 27.59 25.93 -<br />

BY_Bor/59 EU (atypical) 01:10 32.81 - - 27.79 33.63 -<br />

BY_231/SZA EU-3 01:10 21.01 - - 27.74 23.78 -<br />

BY_8380/OKT_2p EU-4 01:10 25.70 - - 27.59 28.36 -<br />

China NA-HP 10 -2 - 20.01 22.14 26.22 - 21.83<br />

China NA-HP 10 -3 - 23.93 25.79 26.89 - 26.06<br />

China NA-HP 10 -4 - 27.61 29.25 27.56 - 28.43<br />

China NA-HP 10 -5 - 29.93 31.71 26.96 - 32.34<br />

China NA-HP 10 -6 - 37.1 34.96 27.50 - 35.20<br />

Stendal V1445/99 NA 1:500 - 21.63 - 27.22 - 23.17<br />

USA VD29949/17 NA 1:500 - 22.74 - 26.75 - 24.44<br />

USA 2 NA 1:500 - 21.12 - 27.51 - 22.43<br />

Mean Value 27.12 25.51 29.62 26.74<br />

Standard Deviation 4.19 5.77 4.17 4.88<br />

Discussion & conclusions<br />

To evaluate sensitivity and specificity <strong>of</strong> VIROTYPE ®<br />

PRRSV,<br />

titration studies with in-vitro RNA were performed. Serum, tissue<br />

and saliva samples were tested in comparison to other<br />

commercial PCR assays. Testing the EPIZONE PRRSV ring trial<br />

panel, all strains could be detected reliably. East European EU<br />

strains and the atypical EU strain scored positive.<br />

VIROTYPE ®<br />

PRRSV allows the reliable and simultaneously<br />

detection <strong>of</strong> PRRSV EU and NA genotype and the HP strain from<br />

porcine blood, serum, tissue, bronchial swabs, bronchial lavage,<br />

saliva, semen and also cell culture samples. The assay internal<br />

control guarantees the control <strong>of</strong> extraction as well as<br />

amplification. Due to the high sensitivity <strong>of</strong> VIROTYPE ® PRRSV<br />

pools <strong>of</strong> five samples can be tested. VIROTYPE ® PRRSV is easy<br />

to use with only one reaction-mix.<br />

Acknowledgements<br />

The EPIZONE ring trial panel and the panel <strong>of</strong> 12 PRRSV<br />

reference strains were kindly provided by K. Wernike and B.<br />

H<strong>of</strong>fmann, Federal Institute for Animal Health, Germany.<br />

References<br />

1. Wernike et al., Porcine reproductive and respiratory syndrome virus:<br />

inter-laboratory ring trial to evaluate real-time RT-PCR detection methods,<br />

submitted – under review


S1 - P - 33<br />

APPLICATION OF A HERD PROGRAMME BASED ON CONTROL MEASURES AND LABORATORY<br />

MONITORING TO ERADICATE A LEPTOSPIRA OUTBREAK IN A LARGE DAIRY CATTLE HERD<br />

M.T. Scicluna, G. Manna, F. Rosone, A. Caprioli, R. Frontoso, , G.L. Autorino<br />

Istituto Zoopr<strong>of</strong>ilattico Sperimentale delle Regioni Lazio e Toscana, 00178 Roma Italia<br />

Leptospira serovar hardjo, management, laboratory diagnosis<br />

Introduction<br />

In Italy, the application <strong>of</strong> severe sanitary measures, including<br />

animal movement restriction are mandatory on the diagnosis <strong>of</strong><br />

Leptospira infection and are lifted only after the removal <strong>of</strong> all<br />

seropositive animals. In the attempt <strong>of</strong> reducing economic losses<br />

deriving from these actions, a herd control programme was<br />

applied in a dairy intensive farm, in which the infection had been<br />

diagnosed, based on the combination <strong>of</strong> preventive, treatment<br />

and biosecurity measures, aiming at the sterilization <strong>of</strong> carriers<br />

and prevention <strong>of</strong> new cases <strong>of</strong> infection (1,2,3,4). The primary<br />

objective was to accelerate the control/eradication <strong>of</strong> the infection<br />

which was evaluated by monitoring the absence <strong>of</strong> its circulation<br />

with the aid <strong>of</strong> serological and molecular techniques and not as<br />

requested by the Italian Regulation, through the sole removal <strong>of</strong><br />

the seropositive animals. Following is the description <strong>of</strong> the<br />

supplementary measures adopted and the actions applied for the<br />

assessment <strong>of</strong> the efficacy <strong>of</strong> the control protocol.<br />

Materials & methods<br />

Following the notification <strong>of</strong> leptospira diagnosis, an extensive<br />

serological investigation was undertaken in the farm <strong>of</strong> 600<br />

producing cows, to verify if the infection was actively circulating:<br />

the random sampling scheme (Table 1) adopted, allowed the<br />

estimation <strong>of</strong> a prevalence <strong>of</strong> at least 50%, with a standard error<br />

<strong>of</strong> 10% and a confidence interval <strong>of</strong> 95%. The serological method<br />

used was the microagglutination test (MAT) described in the OIE<br />

manual, employing the serovar hardjo. A sample was considered<br />

positive when presenting a titre > to 1/100 (6).<br />

The supplementary control plan applied on the farm was based<br />

on the following points:<br />

- preventive measures, based on vaccination to limit new cases<br />

contributing to the further spread <strong>of</strong> leptospirosis. The vaccine<br />

employed, produced by the Istituto Zoopr<strong>of</strong>illatico Sperimentale<br />

<strong>of</strong> Umbria and Marche, contained serovar hardjo and was<br />

administered as prescribed to all animals.<br />

- antibiotic treatment was adopted to sterilize carrier animals and<br />

eliminate primary source <strong>of</strong> infection. Two different antibiotics<br />

were chosen for economical and management reasons. (1,6) The<br />

more economical, Penistrep ®, 25mg/kg b.w., was for 3<br />

consecutive days used in animals managed individually. To avoid<br />

any impact <strong>of</strong> treatment on the sale <strong>of</strong> milk, antibiotics were<br />

administered when the producing animals were dried <strong>of</strong>f.<br />

Treatment was conducted so as to ensure that all animals <strong>of</strong> the<br />

same group received it all together, to avoid infection <strong>of</strong><br />

seronegative subjects, after the disappearance <strong>of</strong> the effect <strong>of</strong><br />

treatment. These animals received a single dose <strong>of</strong> 20 mg/kg<br />

b.w., <strong>of</strong> Duphaciclina 300 LA®.<br />

- Biosecurity measures aimed at further limiting the presence <strong>of</strong><br />

leptospira in the environment, consisted in: isolation <strong>of</strong> the<br />

different units and their drinking systems in relation to whether<br />

these had undergone antibiotic treatment and complete<br />

vaccination; exclusive use <strong>of</strong> artificial insemination; systematic<br />

draining <strong>of</strong> paddocks to avoid water and urine stagnation;<br />

confinement, feeding and milking last <strong>of</strong> untreated animals.<br />

On conclusion <strong>of</strong> the control plan, actions were set up to verify its<br />

efficacy. These consisted in the microbiological examination <strong>of</strong><br />

272 urine samples <strong>of</strong> the animals in the different productive units,<br />

using the same sampling scheme described for the<br />

serosurveillance. These were examined in a SYBR Green Real<br />

Time PCR, specific for pathogenic Leptospira (7). Advantage <strong>of</strong><br />

this test is that it can identify the carrier state even in a<br />

serologically positive animal and compared to the microbiological<br />

isolation, is fast and highly specific. Although reaction inhibition<br />

could occur due to the type <strong>of</strong> sample, using the test at herd<br />

level, increases the sensitivity <strong>of</strong> the method.<br />

In addition, a serological control was carried out on 50 animals<br />

born after the conclusion <strong>of</strong> the programme, used also as<br />

seronegative sentinels, placed in the productive units for the<br />

detection <strong>of</strong> an active circulation. The number <strong>of</strong> sentinel animals<br />

examined aimed to reveal an infection prevalence <strong>of</strong> 5% (IC<br />

95%). These animals were retested at the end <strong>of</strong> their in-contact<br />

period, <strong>of</strong> two months, ensuring exposure, incubation period and<br />

also the production <strong>of</strong> a detectable serological response.<br />

Results & discussion<br />

The results <strong>of</strong> the serosurvey (table 1) demonstrate that<br />

leptospira infection was actively circulating in the farm. The<br />

supplementary measures adopted were considered as successful<br />

on the basis <strong>of</strong> the interruption <strong>of</strong> the leptospira infection in the<br />

different production units, as indicated by the negative results<br />

obtained for the urine samples and the seronegativity <strong>of</strong> the<br />

sentinels after their in-contact period with serological positive<br />

animals. Further evidence <strong>of</strong> the efficiency <strong>of</strong> the control<br />

programme was also the seronegativity <strong>of</strong> all animals born after<br />

the conclusion <strong>of</strong> the adoption <strong>of</strong> these measures.<br />

The adoption <strong>of</strong> such a herd control programme would be <strong>of</strong><br />

benefit for both the farmers as well as the Veterinary Authorities,<br />

in that they would have more instruments for the control <strong>of</strong> this<br />

infection.<br />

Table 1. Sample scheme for serological survey.<br />

Productive Consistency Examined Positive N° <strong>of</strong><br />

Unit<br />

(positive)<br />

animals<br />

(Titres)<br />

Heifers 60 34 (5) 5 1(1/100)<br />

1(1/200)<br />

3(1/400)<br />

Pregnant<br />

Heifers<br />

160 54 (38) 38 27 (1/100)<br />

3(1/400)<br />

8(1/200)<br />

Dry cows 140 51 (37) 37 29(1/100) 73<br />

8(1/200)<br />

Primipare 120 48 (26) 26 26 (1/100) 54<br />

Pluripare 160 60 (35) 35 34(1/100) 58<br />

1(1/200)<br />

Fresh 50 33 (12) 12 10(1/100) 36<br />

Lactating<br />

cows<br />

2(1/200)<br />

Problem 80 44 (25) 25 25 (1/100) 57<br />

animals<br />

Sick bay 20 17 (7) 7 7 (1/100) 41<br />

References<br />

1. Bolin CA, Alt DP. 2001. Am J Vet Res. 62(7):995-1000<br />

2. Cortese VS, et al., 2007. Vet Ther. 8(3):201-8<br />

3. Little TW, et al., 1992. Vet Rec. 1;131(5):90-2<br />

4. Little TW, et al., 1992. Vet Rec. 24;131(17):383-6<br />

5. Levett PN Clin. Microbiol. Rev. 2001 Apr.,: 296–32<br />

6. Alt DP, et al.,2001.J Am Vet Med Assoc.1;219(5):636-9<br />

7. Ahmed A, et. al. 2009. PLoS One 4: Vol 4, Issue 9:1-8<br />

Estimted<br />

prevalence %<br />

15<br />

70


S1 - P - 34<br />

THE SERUM PROTEIN ELECTROPHORETIC PATTERN IN CALVES WITH CHRONIC RESPIRATORY<br />

DISEASES<br />

Csilla Tóthová 1 , Oskar Nagy 1 , Gabriel Kováč 1<br />

1 University <strong>of</strong> Veterinary Medicine and Pharmacy, Clinic for Ruminants, Košice, Slovak Republic<br />

Calves, electrophoresis, respiratory diseases, protein fractions<br />

Introduction<br />

Together with diarrhoea, respiratory tract diseases constitute the<br />

main health problem and overall the most common cause <strong>of</strong><br />

morbidity and mortality in calves and young cattle. In case <strong>of</strong><br />

dairy calf pneumonia, diagnosis and treatment are mainly based<br />

on the observation <strong>of</strong> clinical symptoms. However, in many<br />

cases, the infected calves show only mild clinical symptoms that<br />

could be easily missed in a group <strong>of</strong> calves on a farm (Gänheim<br />

et al., 2007). Therefore, there is a need for objective parameters<br />

that are suitable as indicators <strong>of</strong> health or disease in calf herds<br />

applicable in the laboratory diagnosis <strong>of</strong> diseases. Analytical<br />

methods like serum protein electrophoresis could be useful for<br />

identifying diseased animals. Although it provides useful<br />

information on the pathological conditions associated with<br />

disorders <strong>of</strong> the protein pr<strong>of</strong>ile, the serum protein electrophoresis<br />

in cattle with various organ diseases is a rarely used laboratory<br />

method. Therefore, the objective <strong>of</strong> this study was to determine<br />

whether chronic respiratory diseases in calves cause changes in<br />

the serum protein electrophoretic pattern.<br />

Materials & methods<br />

Twenty-five calves with clinical signs <strong>of</strong> chronic respiratory<br />

diseases <strong>of</strong> various degrees were included into this study. The<br />

calves were <strong>of</strong> a Slovak spotted breed at the age <strong>of</strong> 4-6 months.<br />

Into the evaluation we included the calves with clinical signs <strong>of</strong><br />

the disease manifesting for more than 2 weeks. To compare the<br />

evaluated variables between sick and healthy animals, 29<br />

clinically healthy calves <strong>of</strong> the same age and breed in good<br />

general health were used as a group <strong>of</strong> controls.<br />

Blood samples were collected by a direct puncture <strong>of</strong> v. jugularis.<br />

The separated serum was analyzed for the total serum protein<br />

concentrations and identification <strong>of</strong> serum protein fractions.<br />

Total protein (TP, g/l) concentrations in blood serum were<br />

assayed in the automated biochemical analyser Alize (Lisabio,<br />

France) by the biuret method using commercial diagnostic kits<br />

(Randox). Serum protein fractions were separated by zone<br />

electrophoresis on the buffered agarose gel at pH 8.8 on an<br />

automated electrophoresis system Hydrasys (Sebia Corporate,<br />

France) using commercial diagnostic kits Hydragel 7 Proteine<br />

(Sebia Corporate, France) according to the procedure described<br />

by the manufacturer. The electrophoretic migration was<br />

performed for 15 minutes at 20 °C constantly at 10 W, 40 mA,<br />

and 240 V. After migration, the gels were stained in amidoblack<br />

staining solution, and then destained by acidic solutions and<br />

dried completely. The electrophoretic gels were scanned, and the<br />

serum protein fractions were visualized and displayed on the<br />

densitometry system Epson Perfection V700 (Epson America<br />

Inc., USA). Protein fractions were identified and quantified by<br />

computer s<strong>of</strong>tware Phoresis 5.50 (Sebia Corporate, France).<br />

Serum proteins were separated into the following fractions in the<br />

order from fastest to slowest mobilities: albumin, alpha 1 (α 1 )-,<br />

alpha 2 (α 2 )-, beta 1 (β 1 )-, beta 2 (β 2 )-, and gamma (γ)-globulins.<br />

Albumin: globulin ratios (A/G) were computed from the<br />

electrophoretic scan.<br />

Results<br />

The total serum protein concentrations obtained in the calves<br />

suffering from chronic respiratory diseases were significantly<br />

higher than the values recorded in healthy animals (P


S1 - P - 35<br />

DEVELOPMENT OF A BROADLY DETECTING ASSAY FOR FLAVIVIRUS USING A NOVEL CONCEPT<br />

Frederik Widén 1 , Mikael Leijon 1 , Alia Yacoub 1<br />

1<br />

SVA, VIP, Uppsala, Sweden<br />

2<br />

Affiliation Institute name, Department name, City, Country<br />

Introduction<br />

The flavivirus genus is a large group containing enveloped single<br />

stranded RNA viruses. At least 70 different species <strong>of</strong> flaviviruses<br />

have been identified, several cause disease in animals or man<br />

and many can be transmitted from animals to man by vectors.<br />

Climate change and globalisation may cause the virus to appear<br />

in new areas. This can be illustrated with the detection in Europe<br />

<strong>of</strong> Usutuvirus, WNV lineage 2 and Bagazavirus which were<br />

previously present only in Africa or very rarely in Europe.<br />

Because <strong>of</strong> a changing situation it is appropriate to test<br />

suspected cases with broadly detecting assays. Previously<br />

developed assay for broad detection <strong>of</strong> flavivirus have poor<br />

sensitivity and show strong variation in sensitivity depending on<br />

virus species. This is very unsatisfactory and therefore a PCR<br />

assay based on a novel approach was developed and tested on<br />

a panel <strong>of</strong> different flaviviruses.<br />

Materials & methods<br />

The following viruses were included in the test panel: WNV 1 and<br />

2, Usutuvirus, TBEV (3 strains, Yellow fever virus, Dengue virus<br />

1-4 Japanese Encephalitis virus (two strains) and unrelated virus<br />

(pestivirus and hepacivirus). Several other viruses were used to<br />

test the specificity. Amplification primers were designed using<br />

Jalview etc. The assay was run on a Corbett Rotorgene 3000<br />

real-time thermocycler.<br />

Results<br />

All tested flaviviruses were detected while the unrelated virus was<br />

not detected. The sensitivity <strong>of</strong> the assay was similar for all tested<br />

viruses. For WNV the sensitivity was similar to the specific WNV-<br />

PCR.<br />

Discussion & conclusions<br />

Our results clearly demonstrate that it is possible to detect many<br />

different flaviviruses with very good sensitivity. It is likely that this<br />

assay will also detect emerging variants <strong>of</strong> previously unknown<br />

flaviviruses. This assay is an important diagnostic tool for<br />

surveillance <strong>of</strong> flavivirus infections.<br />

Acknowledgements<br />

Thanks are due to the Swedish institute <strong>of</strong> infectious disease<br />

control, to Cecile Baronti at the European Virus Archive project<br />

and to Tamas Bakonyi and Norbert Novotny for providing virus<br />

strains.<br />

References<br />

1. Moreau et al.(2007) A Real-Time RT-PCR Method for the Universal<br />

Detection and Identification <strong>of</strong> Flaviviruses Vector borne and zoonotic<br />

diseases.7,467-477<br />

Flavivirus, PCR, WNV, detection


S1 - P - 36<br />

EXTENDING THE SHELF LIFE OF COMMERCIAL ELISA KITS<br />

Michael O’Connor, Goretti McDonagh and Ronan G. O’Neill<br />

Virology Division, Department <strong>of</strong> Agriculture, Food & the Marine Laboratories, Backweston, Celbridge, Co. Kildare, Ireland<br />

Kits, ELISA, shelf-life, longevity<br />

Introduction<br />

In <strong>2012</strong>, veterinary laboratories must use an ever expanding<br />

range <strong>of</strong> ELISAs to detect antibodies to, and antigens <strong>of</strong>, a large<br />

number <strong>of</strong> viruses which cause endemic, exotic and emerging<br />

diseases. The demand for such non-routine testing is impossible<br />

to anticipate, frequently resulting in kits or portions <strong>of</strong> kits<br />

exceeding their stated expiry dates. Veterinary virology has<br />

developed through in-house and non-commercial ELISAs,<br />

microtitre cell culture neutralisation tests and virus isolation<br />

techniques. Reagents and controls for these tests were produced<br />

in-house or received as gifts. They are in limited supply, valuable<br />

and <strong>of</strong>ten unique. The precedent has been that such reagents<br />

were stored under optimal conditions and used for as long as<br />

they worked. COULD THE SAME APPLY TO COMMERCIAL<br />

ELISAS?<br />

Materials & methods<br />

Competitive and indirect commercial ELISA kits for the antibodies<br />

to, or the antigens <strong>of</strong>, classical and African swine fever viruses,<br />

foot and mouth disease virus, swine vesicular disease virus,<br />

African horse sickness virus, bluetongue virus, porcine<br />

reproductive and respiratory syndrome virus, the ruminant<br />

pestiviruses, ovine herpesvirus 2 and Coxiella burnetii were<br />

evaluated. Kits stored beyond their stated expiry dates were<br />

compared at intervals with in-date kits. Control sera remaining<br />

from used kits and pr<strong>of</strong>iciency test sera were retested. Results<br />

obtained with the same samples using within-date and postexpiry<br />

date kits were compared. Anticipated shelf life on storage<br />

was assessed by accelerated degradation at 37 o C.<br />

Results<br />

Kits stored at 4 o C for up to 5 years beyond their expiry date<br />

satisfied all manufacturers’ validity criteria, detected all control<br />

samples and performed satisfactorily in EU and National<br />

Reference Laboratory pr<strong>of</strong>iciency tests. Positive control serum<br />

from a blocking ELISA for antibodies to classical swine fever<br />

virus, stored for 98 months (8 years) at 4 o C, reacted similarly to<br />

the control serum supplied with the current kit.<br />

Table 1: Effect <strong>of</strong> storage for 5 years on the performance <strong>of</strong> a<br />

blocking ELISA kit for antibodies to porcine reproductive and<br />

respiratory syndrome virus<br />

9/02/05<br />

Dates tested to 27/09/06 3/10/07 23/03/09 16/04/10<br />

18/05/05<br />

Time beyond<br />

kit expiry date - 9 to - 6 + 10 + 23 + 40 + 53<br />

in months<br />

Optical density<br />

<strong>of</strong> negative 1.5 1.4 1.7 1.3 1.4<br />

control serum<br />

Percentage<br />

inhibition <strong>of</strong><br />

positive<br />

control serum<br />

95 89 89 87 88<br />

Accelerated degradation at 37 o C for a simulated time <strong>of</strong> 27<br />

months had no effect on kits for African horse sickness virus<br />

antigen and African swine fever virus antibody, although a<br />

simulated time <strong>of</strong> 12 months significantly impaired the<br />

performance <strong>of</strong> an African horse sickness virus antibody kit.<br />

Table 2: Percentage inhibition (PI) <strong>of</strong> NRL pr<strong>of</strong>iciency test sera<br />

examined for antibodies to FMD serotype O using post-expiry<br />

date blocking ELISA and in-date kit. PI ≥ 50 is positive<br />

Serum<br />

ID<br />

Kit 10 months<br />

post expiry date<br />

Same kit 21<br />

months post expiry<br />

date<br />

New in-date kit<br />

B107 16 22 19<br />

B108 100 100 100<br />

B109 95 95 94<br />

B110 18 28 23<br />

Ready-to-use substrate/chromogen was sometimes found to<br />

have developed colour on prolonged storage at 4 o C. This was<br />

easily recognised and readily replaced by spare reagent from<br />

newer kits/different manufacturers or from commercial sources.<br />

Table 3: Time beyond expiry date (in months) <strong>of</strong> 5 different<br />

commercial ELISAs for antibodies to bluetongue virus, which all<br />

gave identical, correct results in NRL pr<strong>of</strong>iciency test<br />

ELISA Kit 1 Kit 2 Kit 3 Kit 4<br />

Kit<br />

5<br />

Indirect (I) or competitive (C) I C C C I<br />

Months beyond kit expiry date - 6 + 23 + 3 + 18 + 9<br />

Throughout this study, the only less than satisfactory ELISA kit<br />

detected was a recently purchased within-date one, which<br />

yielded atypical and much reduced, although still valid, control<br />

readings coupled with poor specificity. This kit was replaced free<br />

<strong>of</strong> charge by the manufacturer.<br />

Discussion & conclusions<br />

The demonstration <strong>of</strong> a shelf life <strong>of</strong> up to 5 years beyond the<br />

manufacturers’ expiry date means that ELISA kits from<br />

previous/occasional studies can be safely used. These could<br />

rapidly identify or exclude novel, rare, exotic or emerging<br />

infections, rather than waiting for fresh kits from laboratory<br />

suppliers. It can also reduce wastage, especially where kit usage<br />

has unexpectedly declined or kits have been stockpiled. Kit<br />

suppliers may also expand their diagnostic range without the<br />

need to continuously produce all items in their catalogues.<br />

Four primary antibodies to cell markers were found by 221<br />

laboratories to be effective beyond their expiry dates (1). Sixtyfive<br />

polyclonal and monoclonal antibodies, up to 131 months (11<br />

years) beyond their expiry date, showed no significant<br />

deterioration (2).<br />

The overall performance <strong>of</strong> the ELISA, rather than its expiry date,<br />

should be the absolute determinant <strong>of</strong> whether it may be used.<br />

Performance monitoring should include not only the meeting <strong>of</strong> all<br />

validity criteria but maximum and minimum ODs, sensitivity,<br />

specificity, trends, time for chromogen development, room<br />

temperature etc. Critical evaluation <strong>of</strong> the results by the<br />

laboratory worker will enable early detection <strong>of</strong> problems and<br />

rapid identification <strong>of</strong> significant results, enhancing the value <strong>of</strong><br />

performing ELISA tests.<br />

ELISA kits should be stored at the correct temperature, usually<br />

4 o C, throughout their life. Care should be exercised in<br />

withdrawing reagent aliquots (aseptically or in aliquots) and<br />

removing plate strips/sections. Unused materials/components<br />

should not be left at room temperature while awaiting test<br />

completion. The substrate/chromogen must be examined before<br />

use by decanting into a clear container. SOPs and Quality<br />

Manuals must take account <strong>of</strong> the likely greatly extended shelf<br />

life <strong>of</strong> ELISA kits.<br />

Every laboratory worker using an ELISA plate or strip has the<br />

immediate reassurance <strong>of</strong> seeing that validity criteria and control<br />

parameters have been met as each test is performed and<br />

therefore can have appropriate and logical confidence in the<br />

subsequent results.<br />

References<br />

1. Tubbs, RR, Nagle R, Leslie K, Pettigrew NM, Said JM, Corwin DJ,<br />

Rickert RR & Roche PC. Extension <strong>of</strong> useful reagent shelf life beyond<br />

manufacturers’ recommendations. Arch Pathol Lab Med 1998, 122, 1051–<br />

1052.<br />

2. Balaton AJ, Drachenberg CB, Rucker C, Vaury P & Papadimitriou JC.<br />

Satisfactory performance <strong>of</strong> primary antibodies beyond manufacturers’<br />

recommended expiration dates. Appl Immunohistochem Mol Morphol<br />

1999, 7, 221–225.


S1 - P - 37<br />

MONITORING OF BOVINE VIRAL DIARRHEA VIRUS (BVDV) IN DAIRY CATTLE HERDS IN POLAND<br />

Aleksandra Kuta, Mirosław P. Polak, Magdalena Larska, Jan F. Żmudziński<br />

National Veterinary Research Institute, Department <strong>of</strong> Virology, Puławy, Poland<br />

Bovine viral diarrhea, bulk tank milk, Ab ELISA, RT-PCR<br />

Introduction<br />

Bovine viral diarrhea virus (BVDV), a Pestivirus <strong>of</strong> the family<br />

Flaviviridae is an important pathogen <strong>of</strong> dairy cattle. BVDV<br />

infection can lead to: reduced milk production, diarrhea,<br />

respiratory disorders, interfility, abortion, stillbirth, birth <strong>of</strong><br />

persistently infected (PI) calves. Controlling BVDV is very<br />

complicated and time-consuming. PI animals may be missed if<br />

laboratory monitoring is not implemented at herd level. These<br />

animals shed BVD virus throughout their entire life and they<br />

are the main source <strong>of</strong> infection in a herd. This is the reason<br />

why we should constantly test cattle for the presence <strong>of</strong> BVDV<br />

infection. Farmers’ expectation is that this should be done at<br />

the lowest cost. The aim <strong>of</strong> the study was to assess the status<br />

<strong>of</strong> BVDV infection in selected dairy herds based on bulk tank<br />

milk serology and RT-PCR.<br />

Materials & methods<br />

The material for the study consisted <strong>of</strong> 162 samples <strong>of</strong> bulk<br />

tank milk collected in 60 dairy herds in Poland. The size <strong>of</strong><br />

these herds was between 15 and 565 cattle. Only one herd<br />

was vaccinated against BVDV and in another one herd clinical<br />

problems were observed.<br />

The SVANOVIR BVDV-Ab ELISA was used to detect<br />

antibodies in bulk tank milk and 35 samples with high<br />

Corrected Optical Density (COD) values were additionally<br />

tested by reverse transcriptase – polymerase chain reaction<br />

(RT-PCR). Viral RNA was extracted from the milk somatic cells<br />

using TRI Reagent (SIGMA-ALDRICH, USA), according to the<br />

manufacturer’s instructions. RT–PCR was carried out using<br />

a commercial kit (Titan One Tube RT – PCR System, Roche,<br />

Germany), following the manufacturer’s protocol. Amplification<br />

was done using primers specific for 5’UTR region, sequences<br />

<strong>of</strong> which were published by Vilcek et al. (2). RT-PCR products<br />

were visualized by electrophoresis in 1.5% agarose gel.<br />

The results <strong>of</strong> antibody ELISA were interpreted according to<br />

Swedish system <strong>of</strong> classification. Herds were allocated into 4<br />

different classes based on corrected optical density (COD)<br />

value. Class: 0 – (COD:


S1 - P - 38<br />

DETECTION OF BOVINE LEUKAEMIA VIRUS PROVIRAL DNA IN DENDRITIC CELLS BY IN SITU PCR<br />

Maria Szczotka, Jacek Kuźmak, Ewelina Iwan<br />

National Veterinary Research Institute, Biochemistry Department, Pulawy, Poland<br />

Bovine leukemia, dendritic cells, PCR in situ<br />

Introduction<br />

Bovine leukaemia virus (BLV), like human and simian T-<br />

lymphotropic viruses (HTLV-I/II, STLV-I/II, STLV-L), belongs to<br />

the Deltaretrovirus genus <strong>of</strong> Retroviridae family and causes<br />

enzootic bovine leukosis (EBL), a disease <strong>of</strong> the<br />

lymphoreticular system.<br />

Dendritic cells (DCs) are most potent antigen presenting cells<br />

(APCs) with unique ability take up and process antigens in the<br />

peripheral blood and tissues. They subsequently migrate to<br />

draining lymph nodes, where they present antigen to resting<br />

lymphocytes. They express higher levels <strong>of</strong> MHC class II and<br />

accesory molecules on their surface than other pr<strong>of</strong>essional<br />

APCs. DCs are generally considered as relatives <strong>of</strong> monocytes<br />

and macrophages and to be <strong>of</strong> myeloid origin. They appear to<br />

derive from progenitor also capable <strong>of</strong> forming granulocytes<br />

and macrophages, although more recently a committed DC<br />

progenitor, possibly a downstream precursor, has been<br />

identified. The myeloid mature DCs emphasize by the direct<br />

development <strong>of</strong> a form <strong>of</strong> DCs from blood monocytes. All these<br />

lines <strong>of</strong> evidence for a myeloid origin <strong>of</strong> DC derive from culture<br />

studies using GM-CSF. The aim <strong>of</strong> presented study was to<br />

develop in situ PCR for the detection <strong>of</strong> proviral DNA <strong>of</strong> BLV in<br />

DCs <strong>of</strong> naturally infected cattle. The results <strong>of</strong> provirus<br />

detection by in situ PCR were compared with those obtained<br />

using conventional PCR i.e. solution phase PCR<br />

Materials & methods<br />

Animals. Investigations were performed on the group <strong>of</strong> 9<br />

naturally infected with BLV leukaemic and 5 healthy cows.<br />

Samples. Blood samples, bone marrow, spleen and lymph<br />

nodes were examined in this experiment. Blood was collected,<br />

centrifuged in Histopaque gradient, density 1.077. Cells from<br />

interphase were collected and monocytes were separated with<br />

the use <strong>of</strong> magnetic microbeads labelled with MoAb CD14.<br />

Samples <strong>of</strong> lymphoid organs were cut, digested with<br />

collagenase-DNase, centrifuged and incubated with<br />

immunomagnetic microbeads coated with monoclonal<br />

antibody. Cells were passed through the magnetic sorter, the<br />

cells retendent on the magnet were eluted, washed and placed<br />

in cell culture for 2 weeks in RPMI in the presence <strong>of</strong> IL-4 and<br />

GM-CSF. Cells were grown in special labteks in incubator at<br />

37 o C in 5% <strong>of</strong> CO 2 . After that cells were washed, fixed and<br />

PCR in situ was performed in termocycler. Detection was<br />

performed with the use <strong>of</strong> anti-DIG peroxidase conjugate and<br />

AEC or alkaline phosphatase substrates.<br />

Immun<strong>of</strong>luorescence test (IF) and flow cytometry were<br />

performed with the use <strong>of</strong> MoAbs anti gp51 –BLV glycoprotein<br />

for determination <strong>of</strong> gp51 expression. Flow cytometry was<br />

used for determination <strong>of</strong> DCs immunophenotype in healthy<br />

and naturally infected with BLV cattle. Expression <strong>of</strong> cytokines:<br />

IL-6, IL-10, IL-12p40 and IL-12p70 was measured with<br />

commercial ELISA sets.<br />

Control. As positive control the foetal lamb kidney<br />

permanently infected with BLV - (FLK-BLV) cell line was used<br />

Results<br />

Generated from the blood and inner organs dendritic cells had<br />

typical shape, veils and processes. The expression <strong>of</strong> gp51<br />

glycoprotein was determined by flow cytometry.<br />

In dendritic cells infected with BLV we observed very high<br />

percentage <strong>of</strong> determinants: CD11a, CD11b, CD11c and<br />

MHC-II class and very high expression <strong>of</strong> IL-6, IL-10. IL-12p40<br />

and IL-12p70 in culture fluids. Leukemic DCs exhibited DCs<br />

morphology and had a phenotype <strong>of</strong> mature DCs. The<br />

expression <strong>of</strong> gp51 glycoprotein <strong>of</strong> BLV on leukaemic DCs was<br />

detected in flow cytometry and immun<strong>of</strong>luorescence. The cells<br />

<strong>of</strong> samples with positive reaction in in situ PCR were dark<br />

brown stained. The negative controls were unstained. In situ<br />

PCR results agreed with those <strong>of</strong> provirus detection using<br />

conventional PCR and localization <strong>of</strong> proviral material in cells<br />

was visible.<br />

The results <strong>of</strong> PCR in situ:<br />

DCs-blood DCs - spleen DCs bone marrow<br />

DCs – blood<br />

T<br />

The results <strong>of</strong> IF:<br />

DCs - spleen<br />

Discussion & conclusions<br />

In the present study, DCs generated from peripheral blood and<br />

different lymphoid tissues <strong>of</strong> cattle naturally infected with BLV,<br />

were examined for the presence <strong>of</strong> proviral DNA. In the first<br />

approach, called also direct in situ PCR, dNTP’s, labelled with<br />

digoxygenin, biotin or other markers, are incorporated into<br />

amplification products; the second approach involves two<br />

stage process which combines in situ PCR plus in situ<br />

hybridization with DNA probe. In our study we have used the<br />

first strategy and digoxigenin labelled dNTP’s. The presence <strong>of</strong><br />

gp51 expression was determined as well in IF as in flow<br />

cytometry. We obtained agreement in results <strong>of</strong> in situ PCR<br />

with results <strong>of</strong> other test. The DCs infected with BLV had more<br />

delicate morphology with many vacuoles in cytoplasm.<br />

Infection with BLV caused an increase in cytokines expression<br />

and changes in the percentage <strong>of</strong> surface molecules on<br />

dendritic cells. With the use <strong>of</strong> in situ PCR the localization <strong>of</strong><br />

proviral load could be detected.<br />

Application <strong>of</strong> in situ PCR would be recommended as a<br />

method to confirmation <strong>of</strong> infection with BLV.<br />

Acknowledgements<br />

The research was supported by the Polish State Committee for<br />

Scientific Research, grant No N N 308 622 138. The authors<br />

thank the Polish State Committee for Scientific Research for<br />

financial support.<br />

References<br />

1.Mirsky M.L., Olmstead C.A., Da Y., Lewin H.A. 1996: The prevalence<br />

<strong>of</strong> proviral bovine leukemia virus in peripheral blood mononuclear cells<br />

at two subclinical stages <strong>of</strong> infection. J Virol, 70, 2178-2183.<br />

2. Naif H.M., Brandon R.B., Daniel R.C.W., Lavin M.F. 1998: Bovine<br />

leukaemia proviral DNA detection in cattle using the polymerase chain<br />

reaction. Vet Microbiol, 1, 25, 117-129.<br />

3. Steinman R.M. 1991. The dendritic cell system and its role in<br />

immunogenicity. Annu Rev Immunol. 9, 271-296.<br />

4. Ballagi-Pordany A., Klintevall K., Merza M., Klingeborn B., Belak S.<br />

1992 : Direct detection <strong>of</strong> bovine leukemia virus infection: Practical<br />

applicability <strong>of</strong> a double polymerase chain reaction. J Vet Med B, 39,<br />

69-77.


S1 - P - 39<br />

BOVINE BLOOD DENDRITIC CELLS IN CATTLE INFECTED WITH BOVINE LEUKAEMIA VIRUS (BLV)<br />

Maria Szczotka, Jacek Kuźmak<br />

National Veterinary Research Institute, Biochemistry Department, Pulawy, Poland<br />

Bovine leukemia, dendritic cells, immunophenotype<br />

Introduction<br />

Dendritic cells (DCs) play an important role in the immune<br />

system. These cells are specialized in the presentation <strong>of</strong><br />

antigens. DCs are leukocytes derived from bone marrow and<br />

they are the only cells able to present antigen to naïve T cells.<br />

They initiate primary immune response, exist in all lymphoid and<br />

most non-lymphoid tissues and express both MHC class I and<br />

class II antigens. They not only have an enhancing effect on the<br />

acquired immunity development, but can induce tolerance and<br />

can be manipulated to prevent and treat allergic and autoimmune<br />

diseases. These cells are present in most tissues and comprise a<br />

small fraction <strong>of</strong> the leukocytes through the body, representing<br />

only about 1% <strong>of</strong> peripheral blood mononuclear cells. DCs are<br />

heterogenous cells that can be divided into the few functionalldistinct<br />

subsets on the basis <strong>of</strong> their phenotype, possibility <strong>of</strong><br />

migration and ability for cytokine production. They modulate both<br />

innate and adaptive immune response.<br />

The aim <strong>of</strong> the study was the isolation <strong>of</strong> monocytes from bovine<br />

blood with the use <strong>of</strong> immunomagnetic beads coated with<br />

monoclonal antibody, generation <strong>of</strong> DCs and determination <strong>of</strong><br />

immunophenotype and cytokine expression.<br />

Materials & methods<br />

Isolation <strong>of</strong> monocytes.<br />

Investigations were performed on a group <strong>of</strong> 9 cows naturally<br />

infected with BLV and 4 healthy, BLV-negative animals in ELISA<br />

and PCR that served as negative control. Peripheral blood<br />

mononuclear cells were isolated from whole blood treated with<br />

EDTA-K 2, by standard density centrifugation in Histopaque<br />

(density 1.077). The cells from interphase were collected,<br />

washed twice by centrifugation and incubated with<br />

immunomagnetic Microbeads (Miltenyi Biotec, Germany), coated<br />

with monoclonal mouse anti-human CD14 antibody. After<br />

incubation, cells were centrifuged, cell pellet was resuspended in<br />

AutoMacs Rinsing Solution and placed on a magnetic column<br />

MACS LS (Miltenyi Biotec). The magnetically labelled CD14+<br />

cells were retained on the column and eluted after removal <strong>of</strong> the<br />

magnetic column from the magnet. The viability <strong>of</strong> the cells was<br />

assessed by trypan blue exclusion. The purity <strong>of</strong> the monocyte<br />

fraction was higher than 90%, determined by FACS analysis.<br />

Monocyte culture.<br />

Isolated monocytes were cultured in RPMI 1640 medium<br />

(containing FCS, glutamax and antibiotics) at 37 0 C in humidified<br />

5%CO 2 atmosphere, in the presence <strong>of</strong> granulocyte-macrophage<br />

colony stimulating factor (GM-CSF) and recombinant bovine<br />

interleukin- 4. The cells were fed every 3 day and cultivated for 7-<br />

10 days.during this period, the monocytes transformed into DCs.<br />

Cytology <strong>of</strong> DCs<br />

After 7-10 days, the cultured cells were dislodged by gentle<br />

pipetting, washed, cytocentrifuged onto slides, air-dried at room<br />

temperature and stained with Giemsa’s stain. Morphological<br />

observations were performed under the light microscope<br />

Olympus, equipped with computer system Lucia (Nikon).<br />

Ultrastructure <strong>of</strong> DCs was examined using scanning electron<br />

microscopy (SEM). Briefly, the cells were fixed in 2,5%<br />

glutaralaldehyde in 0.1M cacodylate buffer, dehydrated in serially<br />

graded alcohols (70%-100%), and embedded. Ultrathin sections<br />

were cut, counterstained, and then observed under SEM.<br />

Immunophenotype <strong>of</strong> DCs in both groups <strong>of</strong> cows was<br />

determined in flow cytometer with the use <strong>of</strong> specific monoclonal<br />

antibodies for CD markers and fluorescent conjugates.<br />

Results<br />

In normal bovine blood, the number <strong>of</strong> monocytes was very low:<br />

1%-4%. After magnetic separation we obtained a fraction <strong>of</strong><br />

positively enriched (95%-97%) labelled cells. The CD14 positive<br />

cells had typical monocyte morphology with great nucleus and<br />

small cytoplasm. These cells cultured 24 h in the presence <strong>of</strong><br />

GM-CSF and IL-4 in a growth medium transformed to the<br />

dendritic cells. Some <strong>of</strong> them were elongated and had dendritelike<br />

processes or edges. After 72 h, almost all cells had typical<br />

dendrites: some <strong>of</strong> them were divided on the end. DCs <strong>of</strong> infected<br />

cows cultured in vitro had very strong expression <strong>of</strong> CD11a,<br />

CD11b, CD11c, MHC-I and MHC-II. Expression <strong>of</strong> gp51<br />

glycoprotein was determined in BLV infected cows.by flow<br />

cytometry<br />

Tab.1. The immunophenotype <strong>of</strong> dendritic cells in leukaemic and<br />

control cattle (% cells – mean values)<br />

BLV(+)<br />

CD<br />

14<br />

CD<br />

11a<br />

CD<br />

11b<br />

CD<br />

11c<br />

MHC<br />

I<br />

MHC<br />

II<br />

gp5<br />

1<br />

1 week 83,6 42,5 89,5 58,4 84,6 17,0 5,6<br />

2 weeks 87,9 53,6 84,9 30,0 79,5 16,4 4,1<br />

3 weeks 92,7 63,2 95,7 31,6 77,5 20,5 5,4<br />

BLV (-)<br />

CD<br />

14<br />

CD<br />

11a<br />

CD<br />

11b<br />

CD<br />

11c<br />

MHC<br />

I<br />

MHC<br />

II<br />

1 week 49,4 38,8 67,3 26,5 80,4 20,5 -<br />

2 weeks 41,6 27,3 75,7 26,5 51,3 9,2 -<br />

3 weeks 88,4 23,4 72,2 11,2 28,9 10,5 -<br />

4 weeks 12,7 10,4 41,3 2,1 1,4 30,7 -<br />

gp5<br />

1<br />

Discussion & conclusions<br />

The great amount <strong>of</strong> work performed in the recent years has led<br />

to the definition <strong>of</strong> DCs as cells identified by peculiar morphologic<br />

characteristics and specific functions, such as the ability to<br />

activate naive T cells. Very little is known about their origin and<br />

pathways <strong>of</strong> differentiation. Developing the immunogenic<br />

potential <strong>of</strong> DCs for vaccines – and what is very important for<br />

cancer therapy in humans – requires better access to this cell<br />

type. This report demonstrates that DCs can be derived from<br />

positively-selected monocytes in cattle. Large amounts <strong>of</strong> DCs<br />

can be obtained after the in vitro exposure <strong>of</strong> peripheral blood<br />

monocytes to the effects <strong>of</strong> cytokine combinations, mainly<br />

interleukin IL-4 and GM-CSF, but the ability <strong>of</strong> monocytes to<br />

convert to DCs in vivo has not been so far established. Although<br />

their origins are still controversial or remain unknown, myeloidderived<br />

and lymphoid-derived DCs lineages have been<br />

described. Through their ability to determine the differentiation <strong>of</strong><br />

either Th1 or Th2 cells, distinct DCs subsets may present<br />

antigens in vivo in an immunogenic or tolerogenic fashion. It is<br />

important that the immunostimulatory properties <strong>of</strong> DCs are<br />

dependent on their maturity state: immature DCs have been<br />

shown to be capable <strong>of</strong> inducing antigen-specific inhibition <strong>of</strong> in<br />

vivo T-cell function in humans. Moreover, DCs are potent to<br />

improve vaccine efficacy and are still being investigated as an<br />

important tool in cancer therapy. The availability <strong>of</strong> sufficient<br />

numbers <strong>of</strong> efficient antigen-presenting DCs facilitates effective<br />

vaccine development and the study <strong>of</strong> T-cell-mediated responses<br />

to tumour-associated antigens and cancer therapy.<br />

References<br />

1.Banchereau, J, Steiman, R.1998, Dendritic cells and the control <strong>of</strong><br />

immunity. Nature, 392, 245-252<br />

2. Dubois, B, Fayette, J.1999. Dendritic cells directly modulate B cell<br />

growth and differentiation. J Leukoc Biol, 66, 224-230.<br />

3. Pinchuk LM, Boyd BL, Kruger EF. 2003. Bovine dendritic cells<br />

generated from monocytes and bone marrow progenitors regulate<br />

immunoglobulin production in peripheral blood B cells. Comp Immunol<br />

Microbiol Infect Dis, 26, 233-249.


S1 - P - 40<br />

PORCINE CIRCOVIRUS TYPE 2 IN POSTWEANING PIGS WITH ANTIBIOTIC NON-RESPONSIVE<br />

DIARRHEA<br />

Anna Szczotka 1 , Jacek Żmudzki 1 , Tomasz Stadejek 2 , Zygmunt Pejsak 1<br />

1<br />

National Veterinary Research Institute, Department <strong>of</strong> Swine Diseases, Pulawy, Poland<br />

2<br />

Warsaw University <strong>of</strong> Life Sciences - SGGW, Faculty <strong>of</strong> Veterinary Medicine, Department <strong>of</strong> Pathology and Veterinary Diagnostics Warsaw, Poland<br />

Pigs, PCV2, diarrhea<br />

Introduction<br />

Porcine circovirus type 2 (PCV2) is a causative agent<br />

<strong>of</strong> postweaning multisystemic wasting syndrome (PMWS). The<br />

diseases affects 5 – 15 weeks old pigs and is characterized by<br />

wasting, dyspnea, enlarged lymph nodes, pallor and jaundice.<br />

Sometimes diarrhoea could also be observed and this clinical<br />

manifestation should be differentiated from other swine enteric<br />

diseases, like proliferative enteropathy (PE ) and swine dysentery<br />

(SD). The etiological agents <strong>of</strong> these diseases are, respectively:<br />

Lawsonia intracellularis (L. Intracellularis) and Brachyspira<br />

hyodysenteriae (B. hyodysenteriae).<br />

The aim <strong>of</strong> the study was to analyze the presence <strong>of</strong> porcine<br />

circovirus type 2 (PCV2) in cases <strong>of</strong> antibiotic non-responsive<br />

diarrhoea and to evaluate the possible role <strong>of</strong> the virus in<br />

development <strong>of</strong> enteritis in pigs. For differential diagnosis<br />

identification <strong>of</strong> L. Intracellularis and B. hyodysenteriae was<br />

performed.<br />

Materials & methods<br />

Internal organs and feces were collected from 76 pigs, 5 – 19<br />

weeks old, from 50 farrow-to-finish farms, PMWS-positive or<br />

PMWS-suspected. Sections <strong>of</strong> lymph nodes and intestines<br />

(ileum, caecum and colon) were analyzed for presence <strong>of</strong> PCV2<br />

DNA by in situ hybridization test (ISH) (2). They were also<br />

hematoxilin-eosin (HE) stained for standard histopathological<br />

examination. Addtionally, fecal samples were tested for presence<br />

<strong>of</strong> B. hyodysenteriae and L. intracellularis by PCR (3).<br />

Results<br />

In samples from 37 pigs, 10 – 17 weeks old, large amounts <strong>of</strong><br />

PCV2 DNA, typical for PMWS, were detected in lymph nodes by<br />

ISH. In this group, in samples from 19 pigs, PCV2 was also found<br />

in abundant amount in samples <strong>of</strong> ileum. The remaining 18 pigs,<br />

PCV2-positive in lymph nodes, were negative in ileum. In<br />

samples from only 1 animal lymph nodes were negative for PCV2<br />

in ISH, but virus was detected in considerble amount in ileum. In<br />

HE stained sections <strong>of</strong> lymph nodes histopathological lesions<br />

characteristic for PMWS were identified. Similar lesions were<br />

observed in PCV2-positive samples <strong>of</strong> ileum. Seventy samples <strong>of</strong><br />

feces were negative in PCR for B. hyodysenteriae and<br />

L. intracellularis. DNA <strong>of</strong> L. intracellularis was found in feces from<br />

3 pigs and mixed infection caused by both bacteria was detected<br />

in 3 animals<br />

Figure 2: Presence <strong>of</strong> multinucleated giant cells (arrows) in<br />

mesenteric lymph node (left); multiple intracytoplasmic inclusion<br />

bodies in ileum (right). Hematoxylin and eosin (HE), 200x.<br />

Discussion & conclusions<br />

According to the obtained results in PMWS-affected pigs similar<br />

lesions could be observed both in lymph nodes and in ileum and<br />

they correlate with clinical outcome <strong>of</strong> disease. Also, it was found<br />

that presence <strong>of</strong> PCV2 in ileum could be correlated with<br />

diarrhoea in PMWS- free animal, which confirms that the<br />

emergence <strong>of</strong> this virus as an intestinal pathogen may represent<br />

a new phenomenon (1). In the animals negative for<br />

B. hyodysenteriae and L. intracellularis and PCV2 other<br />

causative agents <strong>of</strong> diarrhoea should be considered.<br />

Acknowledgements<br />

This work was supported by the research grant N N308 075634<br />

from Polish Ministry <strong>of</strong> Science and Higher Education<br />

References<br />

1. Jensen TK, Vigre H, Svensmark B, Bille-Hansen V (2006) Distinction<br />

between porcine circovirus type 2 enteritis and porcine proliferative<br />

enteropathy caused by Lawsonia intracellularis. J Comp Pathol 135: 176–<br />

182.<br />

2. Stadejek T., Podgórska K., Kołodziejczyk P., Bogusz R., Kozaczyński<br />

W., Pejsak Z. (2006) Pierwszy przypadek poodsadzeniowego<br />

wielonarządowego zespołu wyniszczającego świń w Polsce. Medycyna<br />

Wet., 62 (3), 297-301. [First report <strong>of</strong> postweaning multisystemic wasting<br />

syndrome in pigs in Poland].<br />

3. Żmudzki J, Osek J, Stankevicius A, Pejsak Z (2004) Rapid detection <strong>of</strong><br />

Brachyspira hyodysenteriae and Lawsonia intracellularis in swine faecal<br />

and mucosal specimens by multiplex PCR. Bull Vet Inst Pulawy 48: 207-<br />

214.<br />

Figure 1: Large amount <strong>of</strong> PCV2 antigen in inguinal superficial<br />

lymph node (left) and in ileum (right). In situ hybridization (ISH),<br />

200x.


Poster presentations<br />

“Diagnostics<br />

at the point <strong>of</strong> interest”<br />

(2 nd session)


S2 - P - 01<br />

EVALUATION OF FIELD BASED METHODS FOR DETECTION OF CLASSICAL SWINE FEVER BY PCR<br />

Ann-Sophie Ol<strong>of</strong>son 1 , Frederik Widén 1 , Neil Leblanc 1<br />

1<br />

SVA, VIP, Uppsala, Sweden<br />

Introduction<br />

Classical Swine Fever is a disease <strong>of</strong> high economical<br />

importance. Outbreaks in pigs can be extremely costly due to<br />

mortality, production losses and trade restrictions. In Europe this<br />

disease is circulating among wild boars. This virus reservoir<br />

constitutes an important risk for spill over to domestic pigs and<br />

therefore it is important to detect outbreaks among wild boars as<br />

early as possible. Field based PCR detection <strong>of</strong> CSFV <strong>of</strong>fers the<br />

possibility <strong>of</strong> very early detection and is therefore an important<br />

tool for restricting and combating the outbreak.<br />

Materials & methods<br />

Several different PCR instruments, like Cepheid Smartcycler,<br />

Corbett Rotorgene and Tetracore T4 etc. were compared for<br />

detection <strong>of</strong> CSFV using wet kit, dry kit and conventional<br />

reagents for PCR and several different filter papers, handheld<br />

magnetic comb and conventional methods for RNA extraction.<br />

Results<br />

Using the field based methods for extraction <strong>of</strong> RNA and field<br />

based PCR it was possible to detect CSFV with good sensitivity.<br />

Specific data will be presented.<br />

Discussion & conclusions<br />

The results demonstrates that a truly field based detection <strong>of</strong><br />

CSFV can be accomplished using a battery driven PCR<br />

instrument an filter paper based extraction <strong>of</strong> RNA.<br />

Acknowledgements<br />

Thanks are due to Tetracore Inc, Rockville, USA for providing<br />

PCR instruments and reagents<br />

References<br />

1. ,Wakeley PR,Errington, J, Squirrell D.Use <strong>of</strong> a field-enabled extraction and<br />

PCR to detect BVDV. Veterinary Record 2010;166:238-239<br />

doi:10.1136/vr.b4783<br />

PCR, field based, penside, CSFV, Swine Fever


S2 - P - 02<br />

NEW IMMUNOASSAYS FOR DIAGNOSIS OF ASFV BASED ON VP72: CAPTURE ELISA FOR<br />

DETECTION OF IGM SPECIFIC ANTIBODIES AND PEN-SIDE TEST FOR BLOOD SAMPLES.<br />

Teresa Perez 2 , Carmina Gallardo, 1 Angel Venteo 2 , Ana Ranz 2 , Raquel Nieto 1 , Ana Simon 1 , Paloma Rueda 2 ,<br />

Patricia Sastre, P 2 M Luisa Arias 1 , Antonio Sanz 2<br />

1 CISA-INIA, MADRID, SPAIN;<br />

2<br />

INGENASA, MADRID, SPAIN<br />

ASFV, PEN-SIDE TEST, diagnostic, IgM<br />

Introduction<br />

African swine fever (ASF) is a complex and lethal disease <strong>of</strong><br />

swine caused by a large double-stranded DNA virus belonging<br />

to the Asfaviridae family. The acute forms <strong>of</strong> the disease are<br />

devastating and result in very high mortality that may reach<br />

100% producing a major negative effect on national, regional<br />

and international trade. ASF it has been endemic in most <strong>of</strong><br />

sub-Saharan African countries and in Sardinia (Italy) until<br />

2007. In April 2007 its remarkable potential for trans-boundary<br />

spread was amply demonstrated appearing in the Republic <strong>of</strong><br />

Georgia with subsequent outbreaks to Armenia, Azerbaijan<br />

and Russia. Currently ASF is considered as being established<br />

in the Russia Federation threatens to other regions <strong>of</strong> Europe,<br />

central Asia and even China, which has the largest pig<br />

population in the world. Since there is no vaccine available,<br />

rapid and specific diagnostic procedures are an essential<br />

component in affected countries. The early appearance and<br />

subsequent long-term persistence <strong>of</strong> antibodies make antibody<br />

detection techniques a key factor in the control <strong>of</strong> the disease.<br />

To this end, INGENASA in collaboration with the European<br />

Union Reference Laboratory for ASF (CISA-INIA), have been<br />

working in the development an initial standardization <strong>of</strong> a<br />

Capture ELISA for detection <strong>of</strong> specific IgM. In addition has<br />

been improved a rapid, one-step immunochromatographic strip<br />

(pen-side test) INGEZIM PPA CROM for its applicability in the<br />

field to detect specific anti-ASF antibodies directly in blood<br />

specimens besides serum.<br />

Discussion & conclusions<br />

Although more studies are required for further validation, these<br />

preliminary results indicate that the p72 based IgM-capture-<br />

ELISA could be very useful tool for early detection <strong>of</strong> ASFV<br />

infection.<br />

In addition, the analyses <strong>of</strong> blood samples using the<br />

immunochromatographic assay INGEZIM PPA CROM allow us<br />

to detect specific antibodies against ASFV with appropriate<br />

values <strong>of</strong> sensitivity and specificity showing the capability <strong>of</strong><br />

the assay for rapid diagnosis <strong>of</strong> ASF in the field and where<br />

laboratory support and skilled personnel are limited.<br />

References<br />

1. Sánchez-Vizcaíno et al. Scientific report submitted to EFSA on<br />

African Swine Fever. (2009), 1-141.<br />

2. J. M. Sánchez-Vizcaíno et al. African Swine Fever: An<br />

Epidemiological Update. Transboundary and Emerging Diseases, <strong>2012</strong><br />

(59), S1: 27–35.<br />

These studies have been made in collaboration with CISA-INIA<br />

(ASFRISK Project)<br />

Materials & methods<br />

Three different ASFV proteins were used as antigen for the<br />

IgM-ELISAs which comprised the fused p30-p15 protein, the<br />

p72 protein and the uncharacterized protein <strong>of</strong> 33kDa encodes<br />

by the K205R ORF. For initial standardization and validation <strong>of</strong><br />

the IgM-ELISAs, a panel <strong>of</strong> experimental serum samples was<br />

used. The samples were obtained from four independent<br />

experimental infections using the Kenyan ASF viruses<br />

belonging to genotype IX and X, the current East Europe<br />

circulating virus <strong>of</strong> genotype II and the attenuated and non<br />

haemadsorbing Portugal ASFV strain NH/P68 (NHV)<br />

belonging to p72 genotype I.<br />

To test the applicability <strong>of</strong> the immunochromatographic assay<br />

INGEZIM PPA CROM to detect antibodies in blood samples, in<br />

this study were included a wide panel <strong>of</strong> paired experimental<br />

and field porcine serum and blood samples available at CISA-<br />

INIA.<br />

The results were compared to those obtained using the OIE<br />

serological prescribed assays (OIE 2008) as gold standards.<br />

Results<br />

The results obtained in the initial standardization and validation<br />

<strong>of</strong> the IgM-ELISAs, showed that the new developed IgM<br />

capture ELISA, using p72 as antigen, detected specific IgM to<br />

ASFV at early times post infection. These positive samples<br />

remained negative using HT and K205R based IgM-ELISAs,<br />

as well as using the OIE prescribed serological assays.<br />

Results obtained with experimental infections using the<br />

attenuated and non haemadsorbing Portugal ASFV strain<br />

indicated that specific antibody response was detected on<br />

blood samples since day 6 p.i. using the<br />

immunochromatographic assay INGEZIM PPA CROM until the<br />

end <strong>of</strong> the experiment. Same result was obtained using the<br />

OIE (Indirect ELISA) and INGENASA (Competition ELISA)<br />

conventional assays.


S2 - P - 03<br />

PORTABLE PLATFORMS FOR THE DETECTION OF AFRICAN SWINE FEVER VIRUS TESTED IN<br />

FIELD CONDITIONS IN NORTHERN UGANDA<br />

Neil LeBlanc 1 , Edward Okoth 2 , Mikhayil Hakhverdyan 1 , Charles Masembe 3 , Richard Bishop 2 ,<br />

Sándor Belák 1 , Karl Ståhl 1<br />

1 National Veterinary Institute, Uppsala, Sweden<br />

2 International Livestock Research Institute, Nairobi, Kenya<br />

3 Makerere Univeristy, Kampala, Uganda<br />

African Swine Fever, Penside, Real-time PCR<br />

Introduction<br />

African swine fever virus (ASFV) is a serious transboundary<br />

animal disease in Suidae, causing high rates <strong>of</strong> morbidity and<br />

mortality. The objective <strong>of</strong> this study was to produce a portable<br />

molecular platform lab that is suitable for use in the field or<br />

modestly equipped laboratories.<br />

In Southern and Eastern Africa, the presence <strong>of</strong> the sylvatic<br />

cycle involving wild Suidae and s<strong>of</strong>t ticks within the Ornithodoros<br />

family means that the risk <strong>of</strong> introduction <strong>of</strong> ASFV into domestic<br />

swine is always present. However, ASFV can become<br />

established and maintained in domestic and wild pig populations<br />

for prolonged periods. In fact, due to low biosecurity, pig to pig<br />

transmission and contamination are considered the main factors<br />

in maintaining circulation <strong>of</strong> the virus. In Uganda, small pig<br />

holders form a key part <strong>of</strong> the food supply. The ability to detect<br />

ASFV at penside can be a key aspect in control <strong>of</strong> the disease<br />

and education <strong>of</strong> the pig owners to create an awareness <strong>of</strong><br />

biosecurity.<br />

Materials & methods<br />

The T-COR 4 portable real-time PCR thermocycler was used for<br />

amplification and detection (Figure 1). In addition, materials for<br />

three methods <strong>of</strong> DNA extraction from whole blood were brought<br />

in for testing. They included a magnetic bead extraction method<br />

(Nordiag AB), FTA elute indicating cards, and Tego sample<br />

cards.<br />

The real-time PCR assay (ASFV UPL) employed was a new,<br />

recently published assay based on the universal probe library 1<br />

(Roche).The reagents used for the assay included the TaqMan<br />

Fast Advanced Master Mix, as this provided a robust mix that<br />

was thermostable to deal with field conditions. The primary field<br />

location was in an area near Koch Goma, Gulu District, Uganda;<br />

which had no electricity or running water. The laboratory was run<br />

three times over an eight day period, twice in Koch Goma and<br />

once in Kampala.<br />

Results<br />

The assay was tested in ASFV ring trials from two European<br />

projects (Epizone and ASFRISK) as well as another panel <strong>of</strong><br />

isolates representing all 22 genotypes <strong>of</strong> ASFV. They were also<br />

tested with the 2011 clinical ring trial samples prepared by the<br />

EURL. The assay performed well and is designed for ease <strong>of</strong> use<br />

in the field. The ASFV UPL assay was pre-mixed for use in the<br />

field, so assay preparation consisted <strong>of</strong> simply pipetting the mix<br />

into reaction tubes. The results indicate that platforms now can<br />

be established at pen-side or in field laboratories that perform at<br />

a level comparable to sophisticated molecular laboratories.<br />

Discussions & Conclusions<br />

The field trial revealed that these methods have potential for<br />

application in research and monitoring.<br />

Figure 1. Real-time PCR results from field site in Koch Goma,<br />

Uganda<br />

Extraction<br />

The magnetic bead extraction method appears to be robust and<br />

the most efficient. However, it involves a good deal <strong>of</strong> pipetting<br />

and various reagents. The Tego extraction is extremely simple<br />

and requires little time but sensitivity is lower. The FTA method<br />

also works well but there is significant drying time a 30 minute<br />

heating step, which means that although the method is simple, it<br />

does take some time. The sensitivity appears in between the two<br />

former methods.<br />

Assay<br />

The ASFV UPL assay worked was sensitive, reliable and robust.<br />

It is simple to use because it can be pre-mixed before going in<br />

the field.<br />

Instrument<br />

The TCOR 4 instruments functioned very well and managed to<br />

do several runs in a genuine field setting without fail. Re-charging<br />

the instruments by plugging into a vehicle outlet using a 12V<br />

universal adapter was effective in keeping the field lab functional<br />

while <strong>of</strong>f the grid for several days.<br />

Future<br />

Further efforts are now ongoing in collaboration with the<br />

International Livestock Research Institute in Kenya and Makerere<br />

University in Uganda. This includes another trial to facilitate the<br />

use <strong>of</strong> a portable ASFV platform in a major field study.<br />

References<br />

1. Fernández-Pinero, J, Gallardo, C, Elizalde, A et al. (<strong>2012</strong>). Molecular<br />

Diagnosis <strong>of</strong> African Swine Fever by a New Real-Time PCR Using<br />

Universal Probe Library. Transbound. and Emerg. Dis.,<br />

DOI: 10.1111/j.1865-1682.<strong>2012</strong>.01317.x


Poster presentations<br />

“Emerging, re-emerging<br />

and wildlife diseases –<br />

diagnostic possibilities”<br />

(3 rd session)


S3 - P - 01<br />

BEAD-BASED SUSPENSION ARRAY FOR THE SIMULTANIOUS DETECTION OF ANTIBODIES<br />

AGAINS THE RIFT VALLEY FEVER VIRUS NUCLEOCAPSID AND GN GLYCOPROTEIN<br />

René Achterberg 1 , Fimme Jan van der Wal 1 , Matthijn de Boer 2 , Hani Boshra 3 , Alejandro Brun 3 , Kitty Maassen 1 ,<br />

Jeroen Kortekaas 1<br />

1 Central Veterinary Institute <strong>of</strong> Wageningen UR, Department <strong>of</strong> Bacteriology, Lelystad, The Netherlands<br />

2 Department <strong>of</strong> Infectious Diseases and Immunology, Virology Division, Faculty <strong>of</strong> Veterinary Medicine, Utrecht University, Utrecht, the Netherlands<br />

3 Centro de Investigación en Sanidad Animal (CISA-INIA), Carretera de Valdeolmos-El Casar s/n, Valdeolmos, Madrid, Spain<br />

Introduction<br />

Rift Valley fever virus (RVFV) is a mosquito borne virus that was<br />

first isolated in 1930 in Kenya, and has since caused devastating<br />

outbreaks throughout Africa.<br />

The glycoprotein Gn and the nucleocapsid protein N were used<br />

to develop a Luminex assay for the simultaneous detection <strong>of</strong><br />

antibodies against these proteins.<br />

The resulting assay was evaluated using a well characterized<br />

panel <strong>of</strong> sera. The assay correctly identifies seropositive serum<br />

samples. In addition, results show that the assay may be used to<br />

differentiate infected from vaccinated animals (DIVA) when new<br />

vaccines based on RVFV glycoproteins are were used.<br />

Materials & methods<br />

Recombinant Gn and N proteins were coupled to Luminex<br />

paramagnetic MagPlex beads using standard coupling chemistry.<br />

The assay was evaluated using ring trial sera (1), field sera, and<br />

experimental sera from lambs vaccinated with a recombinant<br />

NDV virus expressing the RVFV Gn and Gc proteins (2).<br />

The sets include sera from sheep, cattle and humans and were<br />

tested using anti-species IgG and anti-species IgM.<br />

Results<br />

Ring trial (1) sera (57 sheep, 13 cattle) were defined ‘positive’ if<br />

at least one <strong>of</strong> the ELISAs in the ring trial gave a positive result at<br />

all participating laboratories. Most <strong>of</strong> the sera that scored positive<br />

in the ring trial scored positive in the Gn/N IgG Luminex assay.<br />

Sera that scored positive in the ring trial, but negative in the IgG<br />

Luminex assay, were found positive in the IgM Luminex assay.<br />

The results obtained from the field sera (11 ovine, 5 bovine and<br />

16 human) using the Gn/N IgG Luminex assay corresponded with<br />

data (unpublished) from an IgG ELISA.<br />

Animals, vaccinated with a Gn vaccine that lacks N, over time<br />

developed an IgG response exclusively against Gn, as<br />

demonstrated with the Gn/N IgG Luminex assay.<br />

Discussion & conclusions<br />

The RVFV Gn/N Luminex assay can be used for the<br />

simultaneous detection <strong>of</strong> antibodies against the RVFV Gn and N<br />

proteins in a single sample. The assay is capable <strong>of</strong> detecting<br />

IgG and IgM antibodies in multiple species and performs<br />

comparable to existing ELISAs. The results further show that the<br />

assay may be used as DIVA test to accompany glycoprotein<br />

based RVFV vaccines.<br />

Acknowledgements<br />

The authors would like to thank Catherine Cêtre-Sossah, Philippe<br />

Marianneau, Michel Pépin, Martin Eiden, Francesc Xavier Abad<br />

Morejón de Girón, Christiaan A. Potgieter and Shirley J. Smith for<br />

sera, Jan Wichers and Rob Moormann for helpful discussions.<br />

This research was funded by the Dutch Ministry <strong>of</strong> Economic<br />

Affairs, Agriculture and Innovation.<br />

References<br />

1. Kortekaas et al., European ring trial to evaluate Rift Valley fever virus<br />

ELISAs, submitted<br />

2. Kortekaas, J., de Boer, S.M., Kant, J., Vloet, R.P., Antonis, A.F.,<br />

Moormann, R.J., 2010. Rift Valley fever virus immunity provided by a<br />

paramyxovirus vaccine vector. Vaccine 28, 4394-401<br />

RVFV, serology, Luminex, DIVA, suspension array


S3 - P - 02<br />

LIPOPOLYSACCHARIDE-CAPTURE ELISA FOR THE DETECTION OF LEPTOSPIRA<br />

BORGPETERSENII SEROVAR HARDJO IN SHEEP<br />

Zbigniew Arent, Colm Gilmore, William Ellis<br />

OIE Leptospirosis Reference Laboratory, Veterinary Sciences Division, AFBI, Belfast, Northern Ireland, UK<br />

Leptospira, leptospirosis, sheep, ELISA<br />

Introduction<br />

Endemic infection <strong>of</strong> sheep with Leptospira borgpetersenii<br />

serovar Hardjo (type Hardjo Bovis) is now recognised in many<br />

countries. Many authors demonstrated that infection in sheep is<br />

economically and clinically significant, particularly with regards to<br />

reproductive problems (1).This type <strong>of</strong> infection is <strong>of</strong>ten attributed<br />

to abortion, stillbirth, agalactia and death <strong>of</strong> weak newborn lambs.<br />

In addition, Hardjo infection is an occupational zoonosis <strong>of</strong> those<br />

who work with sheep. All the evidence points to sheep being an<br />

alternative maintenance host for this serovar. The efficacy <strong>of</strong><br />

epidemiological or diagnostic studies <strong>of</strong> leptospirosis in sheep<br />

relies mainly on the correct identification <strong>of</strong> animals which have<br />

been exposed to infection. However, seroprevalence studies in<br />

sheep are difficult to interpret because the microscopic<br />

agglutination test (MAT) - standard serological test, detects only<br />

short-live immunological responses and many previously<br />

exposed and/or infected animals are seronegative. . Therefore<br />

there is a need for more sensitive test than the MAT as an<br />

indicator <strong>of</strong> past exposure.<br />

Leptospiral lipopolysaccharide (LPS) has been identified as an<br />

immunodominant antigen, both in response to infection and to<br />

vaccination. It has proved useful for diagnosing previous<br />

exposure <strong>of</strong> cattle to Hardjo infection as it can be used to detect<br />

specific IgG antibodies to be suitable for diagnostic purposes as it<br />

is more specific and it may give some indication <strong>of</strong> the immune<br />

status <strong>of</strong> an animal.<br />

The aim <strong>of</strong> this study was the development <strong>of</strong> antibody-capture<br />

ELISA using a monoclonal antibody directly against LPS epitope<br />

and its optimisation for use in sheep<br />

Materials & methods<br />

Plates were coated with monoclonal antibody against Leptospira<br />

borgpetersenii serovar Hardjo lipopolysaccharide produced and<br />

characterized earlier in our laboratory (2).<br />

Carbohydrate (CH) antigen was extracted from culture <strong>of</strong><br />

Leptospira borgpetersenii serovar Hardjo type Bovis strains 0294<br />

using the hot-phenol–water method.<br />

Mouse monoclonal anti-ovine IgG antibodies prepared earlier in<br />

our laboratory were used to prepared conjugate. The monoclonal<br />

Ig’s were purified on a Protein G column and covalently attached<br />

to horseradish peroxidase (HRP) using periodate oxidation<br />

method. TMB substrate was used as colorimetric indicator.<br />

After reading the plate at 405 nm results were normalized by<br />

expressing the corrected optical density (Serum OD – Negative<br />

Control OD) as the percentage <strong>of</strong> positivity (%P) <strong>of</strong> the positive<br />

control serum. The optimal cut-<strong>of</strong>f value for the ELISA was<br />

determined by receiver operating characteristic (ROC) analysis.<br />

Correlation between MAT and ELISA was evaluated using 560<br />

field sheep serum samples<br />

Results<br />

A cut-<strong>of</strong>f point was calculated using 80 serum samples obtained<br />

from flocks with no history <strong>of</strong> Leptospirosis and which had no<br />

serological evidence <strong>of</strong> exposure to serovar Hardjo and 60<br />

Hardjo MAT-positive sheep sera. The ROC analysis estimated<br />

the optimized ELISA cut-<strong>of</strong>f as %P ≥ 4.5, with corresponding<br />

diagnostic sensitivity at 95% (95% Cl: 86.1 - 99.0) and specificity<br />

at 98.75% (95% Cl: 93.2 - 100.0) (Fig1).<br />

Sensitivity (%)<br />

100<br />

80<br />

60<br />

40<br />

20<br />

Fig 1 ROC curve analysis<br />

0<br />

cut-<strong>of</strong>f value: >4.5<br />

0 20 40 60 80 100<br />

100-Specificity (%)<br />

The percentage <strong>of</strong> positive results obtained by ELISA test and<br />

MAT applied to field sera are presented in Tab. 1. In the MAT,<br />

reactions to serovar Hardjo were with 28 ovine sera (5.0%)<br />

reacting at 1/100 or greater. When using ELISA, 82 (14.6%) <strong>of</strong><br />

the sera were positive. Only 5 (0.9%) sera were positive in MAT<br />

and negative in ELISA test.<br />

Tab. 1 Comparison <strong>of</strong> ELISA and MAT results<br />

ELISA<br />

Positive<br />

ELISA<br />

Negative<br />

MAT<br />

≥ 1:100<br />

MAT<br />

Negative<br />

TOTAL<br />

for ELISA<br />

14.6% 85.4%<br />

TOTAL<br />

for MAT<br />

4.6% 0.4% 5.0%<br />

10.0% 85.0% 95.0%<br />

Discussion & conclusions<br />

The results <strong>of</strong> the study demonstrated that the ELISA was more<br />

sensitive than MAT. The differences in correlation between the<br />

MAT and ELISA tests was to be expected since the tests<br />

measure different classes <strong>of</strong> antibody. The MAT detects both IgG<br />

and IgM antibodies whereas the ELISA used in this study detects<br />

only IgG. This shows that our ELISA has some limitation<br />

especially as an individual animal test, but despite this, the higher<br />

sensitivity, when compared with the MAT indicates obvious<br />

advantages in use <strong>of</strong> the ELISA as a flock test, where the aim is<br />

to estimate exposure and immunity levels in the flock. The<br />

ELISA seems to be a valuable complement to serological<br />

diagnosis <strong>of</strong> leptospirosis but cannot replace MAT, which<br />

represents the gold standard.<br />

Advantages <strong>of</strong> LPS-captured ELISA test:<br />

- Leptospira Hardjo specific<br />

- Better indicator <strong>of</strong> past exposure than MAT<br />

- Safe and easy to use<br />

- Suitable for screening large numbers <strong>of</strong> sera<br />

References<br />

1. Ellis et al. (1983) Possible involvement <strong>of</strong> leptospires in abortion,<br />

stillbirths and neonatal deaths in sheep. Vet Rec 26, 291-293.<br />

2. Yan K.-T. et al. 1999. Development <strong>of</strong> an ELISA to detect antibodies to<br />

a protective lipopolysaccharide fraction <strong>of</strong> Leptospira borgpetersenii<br />

serovar hardjo in cattle. Vet Microbiol 69, 173-187.


S3 - P - 03<br />

EVALUATION OF DIRECT BLOOD POLYMERASE CHAIN REACTION FOR RAPID DETECTION OF<br />

AFRICAN SWINE FEVER VIRUS<br />

Claudia Gabriel 1 , Marianna Khachatryan 1 , Immanuel Leifer 1,2 , Sandra Blome 1 , Johanna Zemke 1 ,<br />

Bernd H<strong>of</strong>fmann 1 , Martin Beer 1<br />

1<br />

Friedrich-Loeffler-Institut, Institute <strong>of</strong> Diagnostic Virology, Greifswald – Insel Riems, Germany<br />

2<br />

Present address: Institute <strong>of</strong> Virology and Immunoprophylaxis, Mittelhaeusern, Switzerland<br />

Introduction<br />

Most polymerase chain reaction (PCR) techniques for the<br />

detection <strong>of</strong> African swine fever virus (ASFV) require labourintensive<br />

and expensive DNA extraction steps. Nowadays, PCR<br />

kits are available that allow PCR directly from blood samples.<br />

Here, we report on the evaluation <strong>of</strong> such kits in combination with<br />

routine primers.<br />

Materials & methods<br />

In the initial phase <strong>of</strong> the study, two direct PCR kits were<br />

assessed using positive EDTA blood samples obtained from<br />

animals experimentally infected with a highly virulent ASFV<br />

isolate from Armenia: A) the Phusion® Blood Direct PCR Kit<br />

(Finnzymes Oy, Vantaa, Finland; now Thermo Fisher Scientific,<br />

Waltham, USA) and B) the KAPA Blood PCR Master Mix<br />

(PEQLAB Biotechnologie GmbH, Erlangen, Germany). For<br />

specific detection <strong>of</strong> ASFV, primers published by King et al.<br />

(2003) were used.<br />

Both test kits were optimised for gel-based PCR regarding<br />

sample volume and temperature pr<strong>of</strong>ile referring to the<br />

manufacturer’s recommendations. Subsequently, the direct PCR<br />

Kits were tested with negative samples, positive samples from<br />

animal trials, and dilution series <strong>of</strong> positive samples in<br />

comparison with the routinely used real-time PCR assay.<br />

Results<br />

After preliminary optimisation, ASFV was reliably detected in<br />

blood samples from experimentally infected pigs. Both with these<br />

experimental samples and dilution series <strong>of</strong> positive samples, the<br />

direct PCR proved to be as sensitive as established real-time<br />

PCR protocols.<br />

Discussion & conclusions<br />

Concluding, this assay is favourable in both economical and<br />

technological terms and allows the analysis <strong>of</strong> very small sample<br />

volumes. Moreover, direct blood PCR could be advantageous for<br />

pen-side diagnostic applications.<br />

References<br />

1. King DP, Reid SM, Hutchings GH, Grierson SS, Wilkinson PJ, Dixon LK,<br />

Bastos AD, Drew TW. (2003). Development <strong>of</strong> a TaqMan PCR assay with<br />

internal amplification control for the detection <strong>of</strong> African swine fever virus. J<br />

Virol Methods, 107(1):53-61<br />

African swine fever virus, Diagnosis, Direct PCR


S3 - P - 04<br />

DEVELOPMENT OF A SCHMALLENBERG VIRUS ANTIBODY ELISA<br />

Christian Schelp 1 , Yann Senechal 1 , San Pun 1 , Daniel Schumacher 1 , Dragos Gradinaru 2 , Christoph Egli 1 , Jean-<br />

Luc Troch 1 , John Lawrence 3 , Serge Leterme 3<br />

Introduction<br />

Schmallenberg virus was first identified in Germany in late 2011<br />

as a new Orthobunyavirus genetically highly similar to viruses <strong>of</strong><br />

the Simbu serogroup. The virus infects ruminants such as cattle,<br />

sheep and goats. Schmallenberg virus infections have been<br />

recently confirmed in several European countries including<br />

Germany 1 , The Netherlands, Belgium, France, United Kingdom,<br />

Luxembourg, Italy and Spain. Clinical signs include reduced milk<br />

yields, fever, diarrhea, abortions and malformed newborns. Virus<br />

detection by real time RT-PCR or virus cultivation is not the<br />

method <strong>of</strong> choice for infection monitoring due to the very short<br />

viremic period. Therefore, there is an urgent need for an antibody<br />

ELISA to identify infected animals.<br />

Materials & methods<br />

Hundreds <strong>of</strong> samples originating from clinically affected herds,<br />

from virus positive animals and from areas with no<br />

Orthobunyavirus presence are being collected. Samples will be<br />

further characterized by using an inhouse immun<strong>of</strong>luorescence<br />

assay, PCR and epidemiological and clinical data.<br />

An antibody ELISA will be described. Microtiter plates will be<br />

coated with inactivated Schmallenberg-virus antigen. Binding <strong>of</strong><br />

Schmallenbergvirus antibodies will be visualized by colour<br />

change in the wells <strong>of</strong> the microtiter plate. The diagnostic<br />

relevance <strong>of</strong> the result will be assessed by comparing the optical<br />

density (OD) <strong>of</strong> the samples with the OD <strong>of</strong> the positive control.<br />

Results<br />

Preliminary data will be presented. Specificity and sensitivity data<br />

will be calculated using characterized samples. The data will<br />

allow for setting a cut-<strong>of</strong>f to identify Schmallenbergvirus antibody<br />

positive and negative samples.<br />

Discussion & conclusions<br />

The data will be discussed and conclusions will be proposed<br />

according to obtained antibody test performance.<br />

Acknowledgements<br />

We kindly acknowledge the collaboration with the FLI (Friedrich-<br />

Loeffler-Institut), Insel Riems, Germany.<br />

We also would like to thank for the assistance by various<br />

institutes and veterinarians across Europe in helping to collect<br />

samples and additional data.<br />

References<br />

1. H<strong>of</strong>fmann, B, Scheuch, M, Höper, D, Jungblut, R, Holsteg, M,<br />

Schirrmeier, H, Eschbaumer, M, Goller, K.V., Wernike, K, Fischer, M,<br />

Breithaupt, A, Mettenleiter, T.C., Beer, M (<strong>2012</strong>). Novel Orthobunyavirus in<br />

Cattle, Europe, 2011. Emerging Infectious Diseases, vol. 18, 469-472<br />

1<br />

IDEXX Switzerland AG, Liebefeld, Switzerland<br />

2 IDEXX Montpellier SA, Montpellier, France<br />

IDEXX Laboratories, Westbrook, USA 3<br />

Schmallenberg virus, antibody ELISA, Orthobunyavirus


S3 - P - 05<br />

DETECTION OF ECHINOCOCCUS MULTILOCULARIS IN FOXES’ FAECES BY PCR WITH THE USE OF<br />

DILUTED DNA SAMPLES – COMPARISON OF DIFFERENT METHODS OF DNA ISOLATION<br />

Jacek Karamon, Jacek Sroka, Tomasz Cencek<br />

National Veterinary Research Institute, Department <strong>of</strong> Parasitology and Invasive Disease, Pulawy, Poland<br />

Key words: Echinococcus multiloculais, PCR, copro-DNA, inhibitors, foxes<br />

Introduction<br />

Echinococcus multioloculais infection is one <strong>of</strong> the most<br />

dangerous zoonoses and still remains a significant public health<br />

problem. Definitive hosts <strong>of</strong> this tapeworms are carnivores<br />

(especially foxes, but also dogs) which disperse the invasive<br />

eggs with faeces. A lot <strong>of</strong> prevalence study are conducted in<br />

populations <strong>of</strong> foxes and dogs, in order to estimate the infection<br />

risk for people. In wild carnivires post mortem examinations <strong>of</strong><br />

intestines are recommended - among them the SCT – regarded<br />

as the “gold standard”. But there are many situations when<br />

investigation must be carried out in vivo (for example examination<br />

<strong>of</strong> dogs populations). Therefore there are some techniques for<br />

detection <strong>of</strong> Echinococcus infection in definitive hosts by feaces<br />

examination and among them PCR for copro-DNA detection is<br />

one <strong>of</strong> the most usefull and sensitive. However, there are many<br />

problems connected with PCR inhibiting factors contained in<br />

faeces. The aim <strong>of</strong> this investigation was to compare the<br />

effectiveness <strong>of</strong> different methods <strong>of</strong> E. multilocularis DNA<br />

isolation from faeces for PCR performed with different DNA<br />

dillutions - to choose the variant minimizing the inhibiting influent<br />

<strong>of</strong> faeces.<br />

Materials & methods<br />

Intestines (small and large) used in investigation were obtained<br />

from 35 foxes. Small intestines were examined by sedimentation<br />

and counting technique (SCT). Samples <strong>of</strong> faeces (about 2 g)<br />

collected from the end <strong>of</strong> rectum were frozen (-20 o C) for<br />

molecular examination.,<br />

DNA from samples were isolated and purified with the use <strong>of</strong> 3<br />

different methods:<br />

(L)– with the use <strong>of</strong> QIAamp DNA Stool Mini Kit (Qiagen)<br />

according to protocol for larger volumes <strong>of</strong> stool (1g samples<br />

were used)<br />

(S)– with the use <strong>of</strong> QIAamp DNA Stool Mini Kit (Qiagen)<br />

according to standard protocol for (200 mg samples were used)<br />

(T)- with the use <strong>of</strong> QIAamp DNA Mini Kit (Qiagen) according to<br />

standard producer protocol (20 mg samples were used)<br />

DNA isolated in each method were tested by PCR in 6 variants:.<br />

one not diluted (1/1) and 5 diluted (1/2.5, 1/5, 1/10. 1/20, 1/40)<br />

A nested PCR method was used, as described by Dinkel et al.<br />

(1998) with some modifications concerning reaction mixture and<br />

time conditions <strong>of</strong> amplification (Karamon et al <strong>2012</strong>). The<br />

sequence for amplification was part <strong>of</strong> the E. multilocularis<br />

mitochondrial 12S rRNA gene. In a second stage <strong>of</strong> PCR the<br />

fragment specific for E. multilocularis (250 bp) was amplified.<br />

In case <strong>of</strong> 25 foxes additional faecal samples were collected and<br />

examinmed at once for detection od Taenidae eggs by McMaster<br />

flotatuion technique.<br />

Fig. 1. Percentage <strong>of</strong> E. multilocularis positive samples detected<br />

by SCT and PCR (3 variants <strong>of</strong> DNA isolation) and Taenidae<br />

positive samples found by flotation.<br />

.<br />

% <strong>of</strong> positive samples<br />

60,0<br />

50,0<br />

40,0<br />

30,0<br />

20,0<br />

10,0<br />

0,0<br />

51,4<br />

45,7<br />

40,0<br />

34,3<br />

20,0<br />

SCT PCR L PCR S PCR T Flotation<br />

(Taenidae<br />

eggs)<br />

Results<br />

E. multilocularis tapeworms were found in 18 from 35 (51,4%)<br />

intestines examined by SCT. The intensity was ranged from 2 to<br />

1055 worms per intestine.<br />

The highest number <strong>of</strong> positive results in PCR (16) were obtained<br />

in isolation variant (L) (isolation with the use <strong>of</strong> larger stool<br />

volume), and then 14 and 12 respectively for (S) and (T) variants<br />

<strong>of</strong> isolation. In one case (sample No. 19) sample negative in<br />

SCT was estimated as positive in PCR (L).<br />

In some samples positive results in PCR were obtained only in<br />

diluted DNA. From 25 faeces examined by flotation only in 5<br />

(20%) Taenidae eggs were detected<br />

Table 1. Selected results obtained by PCR for detection <strong>of</strong> E.<br />

multilocularis copro-DNA (in different DNA dilution and with the<br />

use <strong>of</strong> 3 different variants <strong>of</strong> DNA isolation) in comparison to<br />

microscopic sedimentation and counting technique (SCT).<br />

Discussion & conclusions<br />

Blocking <strong>of</strong> PCR by inhibitors contained in faeces is the known<br />

problem in molecular diagnostics. When the concentration <strong>of</strong><br />

inhibiting substances in the sample is high, the elimination <strong>of</strong><br />

them during DNA purification (also by using dedicated methods)<br />

may be insufficient In that cases significant amount <strong>of</strong> them leave<br />

in the DNA sample and can block the PCR - causing possibility <strong>of</strong><br />

false negative results. Some authors demonstrated that samples<br />

which block the PCR may be identified by repeated examination<br />

<strong>of</strong> all negative ones with adding to each one additional positive<br />

control (Dinkel et al 1998). But in such method all identified<br />

inhibiting samples had to be eliminated from further analysis at<br />

all, because there were still no answer which ones were really<br />

positive or negative.<br />

However, our investigation showed that in E. multilocularis PCR<br />

additional using diluted DNA (besides non diluted) could avoid<br />

false negative results causing by PCR inhibition. In the (L)<br />

method <strong>of</strong> purification using additionally 1/10 dilution <strong>of</strong> DNA<br />

seems to be enough. But, when DNA purification was done by<br />

the method not dedicated for faeces, DNA samples required<br />

more and higher dilutions (because <strong>of</strong> higher inhibitors<br />

concentration).<br />

References<br />

1. Dinkel, A. et al Echinococcus multilocularis in the definite host,<br />

coprodiagnosis by PCR is an alternative to necropsy. J. Clin. Microbiol. 36,<br />

1871–1876 (1998)<br />

2. Karamon et al. The first detection <strong>of</strong> Echinococcus multilocularis in<br />

slaughtered pigs in Poland. Vet Parasitol 185 327– 329 (<strong>2012</strong>)


S3 - P - 06<br />

COMPARISON OF CULTURE AND PCR FOR DETECTION OF MYCOBACTERIUM AVIUM SSP.<br />

PARATUBERCULOSIS IN BOVINE FAECES<br />

Selina Keller 1 , Nicole Cernela 2 , Roger Stephan 2 , Max M. Wittenbrink 1<br />

1<br />

Institute <strong>of</strong> Veterinary Bacteriology, Vetsuisse Faculty, University <strong>of</strong> Zurich, Switzerland<br />

2<br />

Institute for Food Safety and Hygiene, Vetsuisse Faculty, University <strong>of</strong> Zurich, Switzerland<br />

Introduction<br />

Paratuberculosis is an incurable granulomatous enteropathy<br />

caused by Mycobacterium avium ssp. paratuberculosis (MAP)<br />

affecting primarily wild and domestic ruminants with animals up to<br />

six months <strong>of</strong> age being most receptive (1). Common route <strong>of</strong><br />

infection is faecal-oral with faeces or milk and incubation time<br />

ranges from months to years. Together with the late onset <strong>of</strong><br />

symptoms, this makes diagnosis <strong>of</strong> paratuberculosis very<br />

challenging. The aim <strong>of</strong> this study was to compare a direct real<br />

time PCR with a cultural approach for screening faecal samples<br />

<strong>of</strong> healthy animals from different farms with a history <strong>of</strong><br />

paratuberculosis.<br />

Materials & methods<br />

This study comprised faecal samples <strong>of</strong> 1266 animals from 13<br />

dairy cattle and 10 sucking cow herds. In these herds at least one<br />

animal had been diagnosed with paratuberculosis in the last 5<br />

years. Herd sizes ranged from 11 to 130 animals.<br />

Paratuberculosis, culture, PCR, cattle<br />

Culture and PCR were performed from three pooled faecal<br />

samples at a time (2). For culture, a decontamination with 4%<br />

NaOH and 5% oxalic acid was used to reduce the accompanying<br />

bacterial flora before the 422 faecal pools were transferred onto<br />

selective media containing Mycobactin J, namely Löwenstein-<br />

Jensen (LJ) and Herrold’s Egg Yolk agar (HEYA) (3). Suspicious<br />

colonies were inoculated into Middlebrook 7H9 broth enriched<br />

with ADC and Mycobactin J (4). After one week <strong>of</strong> multiplication,<br />

Ziehl-Neelsen staining was performed for detection <strong>of</strong> acid fast<br />

bacilli. For confirmation <strong>of</strong> MAP, real time PCR based on the F57<br />

element was used (5). Moreover, for direct detection <strong>of</strong> the<br />

pathogen in pooled faecal samples, the same real time PCR<br />

protocol was performed.<br />

Results<br />

After incubation for 16 weeks, in 62 <strong>of</strong> the 422 (14.7%) faecal<br />

pools cultivation <strong>of</strong> MAP succeeded (Figure 1), <strong>of</strong> which 48<br />

(77.4%) grew on HEYA, 2 (3.2%) on LJ and 12 (19.6%) grew<br />

both on LJ and HEYA (Figure 2). However, only 9 <strong>of</strong> 422 (2.1%)<br />

faecal pools tested positive by direct real time PCR. In general,<br />

HEYA revealed positive cultures already after 9 weeks <strong>of</strong><br />

cultivation, whereas 11 weeks were necessary for visible growth<br />

on LJ.<br />

For identification <strong>of</strong> the MAP spreading animals, samples (n=186)<br />

<strong>of</strong> the 62 positive tested pools were individually subjected to<br />

culture and PCR. Cultivation <strong>of</strong> MAP succeeded in 59 <strong>of</strong> the 186<br />

(31.6%) faecal samples, whereas only 12 <strong>of</strong> 186 (6.4%) tested<br />

positive by direct real time PCR.<br />

Figure 2: Growth <strong>of</strong> MAP on HEYA and LJ<br />

Discussion & conclusions<br />

The results <strong>of</strong> this study show that for MAP detection in faecal<br />

samples from animals without clinical signs, the cultural assay is<br />

a more sensitive method compared to F57 real time PCR.<br />

Nevertheless, considering the long cultivation time and the workintensive<br />

procedure, the applicability in view <strong>of</strong> eradication<br />

systems remains restricted. The limited performance <strong>of</strong> PCR,<br />

however, may be due to the fact that in the cultural assay 1244-<br />

times more faecal material is used. Results from this study also<br />

highlight the impact <strong>of</strong> the medium chosen for cultivation since<br />

HEYA revealed a much higher prevalence <strong>of</strong> MAP compared to<br />

LJ.<br />

References<br />

1. Bögli-Stuber, K., et al. (2005): Detection <strong>of</strong> Mycobacterium avium<br />

subspecies paratuberculosis in Swiss dairy cattle by real-time PCR and<br />

culture: a comparison <strong>of</strong> the two assays,<br />

J Appl. Microbiol. 99:587–597.<br />

2. Kalis, C.H.J., et al. (2000): Culture <strong>of</strong> strategically pooled bovine fecal<br />

samples as a method to screen herds for paratuberculosis, Vet. Diagn.<br />

Invest. 12:547–551.<br />

3. Beerwerth, W. (1967): Die Züchtung von Mykobakterien aus dem Kot<br />

der Haustiere und ihre Bedeutung für die Epidemiologie und Bekämpfung<br />

der Tuberkulose, Prax. Pneumol. 21:189–202.<br />

4. Glanemann, B., et al. (2004): Detection <strong>of</strong> Mycobacterium avium<br />

subspecies paratuberculosis in Swiss dairy cattle by culture and serology,<br />

Schweiz. Arch. Tierheilk., 146:409-415.<br />

5. Bosshard, C., et al. (2006): Application <strong>of</strong> an F57 sequence-based realtime<br />

PCR assay for Mycobacterium paratuberculosis detection in bulk tank<br />

raw milk and slaughtered healthy dairy cows, J Food Prot. 69:1662-7.<br />

Figure 1: MAP detection in faecal pools by PCR and culture.


S3 - P - 07<br />

HEPATITIS E VIRUS IN DOMESTIC SWINE AND WILD BOAR FROM GERMANY<br />

Oliveira-Filho, E.F, Bank-Wolf, B.R., Thiel, H-J., König, M.<br />

Institute <strong>of</strong> Virology, Justus-Liebig University Gießen, Schubertstr. 81, 35392 Gießen<br />

Introduction<br />

Hepatitis E is an emerging zoonotic disease distributed<br />

worldwide. The causative agent Hepatitis E virus (HEV)<br />

belongs to the genus Hepevirus <strong>of</strong> the virus family<br />

Hepeviridae. Another member <strong>of</strong> the family not assigned to a<br />

genus so far is the avian HEV.<br />

HEV is non-enveloped with a single stranded RNA genome <strong>of</strong><br />

positive polarity. The genome encompassing approximately<br />

7.2 kb encodes three open reading frames. Members <strong>of</strong> HEV<br />

can be subdivided into at least 4 genotypes (1-4). Virus<br />

isolates from rats and rabbits may belong to additional new<br />

genotypes.<br />

HEV may cause acute hepatitis in humans but infection <strong>of</strong>ten<br />

remains subclinical. Besides man HEV was also found in<br />

domestic animals such as swine as well as in wildlife species<br />

like wild boar, deer, rabbit and rat. So far, no clinical disease<br />

has been associated with HEV infection in these animals.<br />

In this study we intended to investigate the presence <strong>of</strong> HEV in<br />

wild boar and domestic swine in Germany. Phylogenetic<br />

analyses were performed to form the basis for molecular<br />

epidemiology and the improvement <strong>of</strong> diagnostic tests.<br />

Materials & methods<br />

A total <strong>of</strong> 105 porcine faecal samples and 124 sera from wild<br />

boar were tested by conventional RT-PCR using different<br />

primer combinations. In addition a quantitative real time PCR<br />

(qPCR) test for HEV was set up.<br />

PCR amlicons were cloned and nucleic acid sequences<br />

obtained and compared to sequence data from GenBank.<br />

Phylogenetic analyses using partial as well as complete<br />

sequences <strong>of</strong> the HEV capsid gene were performed.<br />

Results<br />

A fragment <strong>of</strong> 241 nucleotides was detected in 13.7% <strong>of</strong> the<br />

sera from wild boar and in a single faecal sample from a<br />

domestic swine (0.95%). All viruses could be assigned to<br />

genotype 3 <strong>of</strong> HEV. Sequence diversity was significant with<br />

differences <strong>of</strong> up to 21.6% among the wild boar samples.<br />

The complete capsid protein gene was cloned and sequenced<br />

from three wild boar samples and the single positive domestic<br />

swine. Based on this sequence data the isolate from domestic<br />

swine was assigned to HEV subtype 3a and the three wild<br />

boar viruses to subtypes 3a and 3i.<br />

Additional analyses indicated recombination between<br />

members <strong>of</strong> HEV subtypes as one reason for sequence<br />

diversity.<br />

Discussion & conclusions<br />

Our study indicates that HEV is circulating among wild boar in<br />

Germany and may be transferred to domestic swine. These<br />

data support observations by others (1, 2).<br />

The close relationship <strong>of</strong> the animal and human HEV<br />

sequences implies the risk for zoonotic transmission <strong>of</strong> the<br />

virus. Phylogenetic analyses should be based on complete<br />

capsid gene sequences to allow a significant separation <strong>of</strong><br />

subtypes.<br />

Acknowledgements<br />

Samples from wild boar were kindly provided by Landesbetrieb<br />

Hessisches Landeslabor Giessen.<br />

References<br />

1. Schielke, A, Sachs, K, Lierz, M, Appel, B, Jansen, A, Johne, R<br />

(2009). Detection <strong>of</strong> hepatitis E virus in wild boars <strong>of</strong> rural and urban<br />

regions in Germany and whole genome characterization <strong>of</strong> an endemic<br />

strain. Virol J. 14;6:58.<br />

2. Wenzel JJ, Preiss J, Schemmerer M, Huber B, Plentz A, Jilg W.<br />

(2011). Detection <strong>of</strong> hepatitis E virus (HEV) from porcine livers in<br />

Southeastern Germany and high sequence homology to human HEV<br />

isolates. J Clin Virol. 52(1):50-4.<br />

HEV, zoonosis, wild boar, phylogenetic analysis


S3 - P - 08<br />

EMERGENCE OF SCHMALLENBERG VIRUS:<br />

DEVELOPMENT AND VALIDATION OF A REAL-TIME PCR DETECTION KIT<br />

Damien MAGNEE 1 , Stéphane DALY 1 , Sandrine MOINE 1 , Eric SELLAL 1<br />

1<br />

LSI Laboratoire Service International, LISSIEU, France<br />

Schmallenberg, Real-Time PCR, Outbreak,<br />

Introduction<br />

Between August and December 2011, outbreaks <strong>of</strong> disease in<br />

adult cattle, abortions and births <strong>of</strong> malformed animals in sheep,<br />

cattle and goats were reported in the Netherlands, Germany and<br />

Belgium. A new virus was identified as a cause <strong>of</strong> these problems<br />

and was named “Schmallenberg virus” after the place where it<br />

was first identified.<br />

This virus belongs to the Bunyaviridae family, genus<br />

orthobunyaviridae and is closely related to Akabane, Aino and<br />

Shamonda viruses. Preliminary studies have suggested that this<br />

virus affects mainly ruminants and must be transmitted by a<br />

vector (biting midges <strong>of</strong> the genus Culicoides, mosquitoes...).<br />

We present here the development <strong>of</strong> new real-time PCR kits for<br />

identification <strong>of</strong> Schmallenberg virus, by targeting the S segment,<br />

and the different validation steps leading to final authorisation for<br />

SBV diagnosis by the French National Reference Laboratory<br />

(ANSES Maisons-Alfort) and by the Friedrich-Loeffler-Insitut in<br />

Germany.<br />

Materials & methods<br />

Systems for specific detection <strong>of</strong> Schmallenberg Virus were<br />

designed on the basis <strong>of</strong> the sequence deposited on Genbank. A<br />

first system was designed on the L segment. Then, according to<br />

the FLI recommendations, we realised a new design, based on<br />

the detection <strong>of</strong> the S segment. The LSI SBVS kit allows the<br />

simultaneous detection <strong>of</strong> SBV target and an endogenous IPC.<br />

Detectability <strong>of</strong> the both kits was compared with the FLI design<br />

using serial dilution <strong>of</strong> SBV RNAs provided by the Friedrich-<br />

Loeffler Institut (Germany). Analytical specificity and sensitivity <strong>of</strong><br />

L and S segment kit were assessed using several field samples<br />

(brains), coming from France and Belgium.<br />

Specificity <strong>of</strong> prototype kits was evaluated on a panel <strong>of</strong> ruminant<br />

pathogens and other orthobunyaviridae.<br />

Both prototype kits were sent to the Friedrich Loeffler Institut and<br />

the S segment prototype kit was sent to the French NRL<br />

(ANSES-Maisons Alfort) for evaluation <strong>of</strong> their specificity,<br />

sensitivity, detectability and repeatability on field samples, in<br />

order to receive <strong>of</strong>ficial validation for diagnosis in France and<br />

Europe.<br />

SBV virus detection is preferably performed on brains <strong>of</strong> aborted<br />

fetus, but the virus can also be located in blood, serum and<br />

spleen. For brain and spleen samples, a grinding step is<br />

necessary.<br />

Results<br />

The both systems showed good specificity, with detection <strong>of</strong> all<br />

positive samples and no detection <strong>of</strong> other ruminant pathogens.<br />

These kits have equivalent detectability with the FLI designs, with<br />

better sensitivity for the S segment relative to the L segment (for<br />

FLI and LSI design).<br />

On 34 field samples (brains coming from stillbirths with<br />

malformations), we have better detectability with LSI systems,<br />

with 29 positives samples with LSI S segment PCR and 25<br />

positives with FLI PCR.<br />

Characteristics <strong>of</strong> the both kits obtained at LSI were confirmed at<br />

the French NRL and the Friedrich Loeffler Institut. Kits showed<br />

good sensitivity, specificity and detectability (Table 1).<br />

Table 1: Official evaluation <strong>of</strong> LSI SBV Segment S kit<br />

Sensitivity<br />

and<br />

specificity<br />

Detectability<br />

ANSES (France)<br />

37 samples with<br />

100% correlation<br />

2 negatives samples<br />

with FLI system are<br />

positive with LSI kit<br />

Gain <strong>of</strong> 1 log<br />

FLI (Germany)<br />

40 field samples<br />

with 100%<br />

correlation<br />

100% correlation<br />

with FLI system<br />

The SBV-S segment kit commercialised by LSI received <strong>of</strong>ficial<br />

authorisation for utilisation in the French network for diagnosis <strong>of</strong><br />

Schmallenberg virus and was also validated by the FLI, for use in<br />

European market.<br />

Discussion & conclusions<br />

We showed here all the steps leading to <strong>of</strong>ficial validation <strong>of</strong> the<br />

SBV LSI kit in Real-Time rt-PCR, by the French National<br />

Reference Laboratory and the Friedrich Loeffler Institut.<br />

The first step was to develop a PCR kit, targeting the L segment,<br />

with exogenous IPC. After recommendation by the FLI to target<br />

preferably the S segment and request <strong>of</strong> the French NRL to use<br />

an endogenous IPC, we develop a second kit with segment S<br />

detection, with endogenous IPC.<br />

The initial validation at LSI shows good results in sensitivity,<br />

specificity and detectability. A panel <strong>of</strong> 34 field samples show<br />

good results, for SBV target and for IPC (figure 1).<br />

Figure 1: Results on field samples for SBV-S LSI kit<br />

All characteristics obtained at LSI were confirmed at the French<br />

NRL and at the FLI, giving to this kit an <strong>of</strong>ficial authorisation for<br />

use in SBV diagnosis in France and Europe.<br />

Acknowledgements<br />

A special thank to the French NRL (ANSES Maisons-Alfort) and<br />

Bernd H<strong>of</strong>fmann (FLI) for their scientific expertise and their help,<br />

in this particular context.<br />

References<br />

1. H<strong>of</strong>fmann, B, Scheuch, M (<strong>2012</strong>). Novel Orthobunyavirus in Cattle,<br />

Europe, 2011. Center for Disease Control and Prevention, Vol 18 N° 3, p1-<br />

6


S3 - P - 09<br />

PRELIMINARY VALIDATION OF A SOLID- PHASE COMPETITIVE ELISA FOR THE DETECTION OF<br />

ANTIBODIES AGAINST WEST NILE DISEASE VIRUS IN HORSE SERA<br />

G. L. Autorino 1 , A. Caprioli 1 , F. De Simone 2 , R. Frontoso 1 , D. Lelli 2 , R. Nardini 1 , F. Rosone 1 , M. T. Scicluna 1 .<br />

1<br />

National Reference Centre for Equine Infectious Diseases, Istituto Zoopr<strong>of</strong>ilattico Sperimentale Lazio e Toscana, 00178 Roma Italia. 2 Istituto<br />

Zoopr<strong>of</strong>ilattico Sperimentale Emilia Romagna e Lombardia, Via A. Bianchi 9, 25124 Brescia Italia<br />

Keywords: West Nile Disease, serological diagnosis, competitive elisa, validation<br />

Introduction<br />

In 1998, Italy reported the first equine outbreaks <strong>of</strong> West Nile<br />

Disease (WND). From 2001, a national veterinary surveillance<br />

system was adopted in areas considered at risk <strong>of</strong> WNV<br />

introduction, based on the passive surveillance <strong>of</strong> avian mortality<br />

and on the repeated serological testing <strong>of</strong> sentinel horses first in<br />

screening ELISA tests available commercially, either in a<br />

competitive format or based on the detection <strong>of</strong> IgG and IgM.<br />

Positive samples are then examined in the confirmatory test<br />

represented by the plaque neutralization reduction test. A solidphase<br />

competitive ELISA was developed and a preliminarily<br />

validation was carried by an interlaboratory trial. The data <strong>of</strong> the<br />

this preliminary validation is presented and discussed.<br />

Materials & methods<br />

The procedure for the competitive ELISA (C-ELISA), object <strong>of</strong><br />

this validation, is briefly described as follows. 96 well microplate<br />

are pre-adsorbed with a monoclonal antibody (Mab), recognising<br />

the domain III (Ed III) <strong>of</strong> WND virus (WNDV), are prepared ready<br />

for use. The reaction is started by the addition <strong>of</strong> the antigen, a<br />

cell-culture cryolysate <strong>of</strong> inactivated virus, on to the plate and its<br />

incubation for 90 minutes at 37°C. In parallel to the first<br />

incubation, serum samples and controls are diluted 1:5 and 1:10<br />

on a separate plate. The samples and controls are then<br />

transferred on the adsorbed plate containing the antigen and<br />

incubated for 60 minutes at 37°C. After washing the plate, the<br />

same Mab used for adsorption is added conjugated with<br />

horseradish peroxidase. Following another incubation period <strong>of</strong><br />

90 minutes at 37°C, ortophenyl-diamine substrate is added and<br />

the plate is incubated at room temperature for 15 minutes in the<br />

dark. Sulphuric acid is used to stop the enzymatic reaction, which<br />

is read using an optical density (OD) <strong>of</strong> 492nm. Sera are<br />

categorized as positive or negative according to percentage<br />

inhibition (PI), calculated as the ratio between sample and<br />

reaction control.<br />

Validation was performed according to WOAH Manual guidelines.<br />

The aim for this ELISA test is for screening purposes.<br />

Nine laboratories were involved in this validation. Pre-adsorbed<br />

plates, reagents (Mab and antigen) and a panel <strong>of</strong> sera were sent<br />

to each <strong>of</strong> them. The latter was made up <strong>of</strong> 20 sera, each<br />

replicated twice, and numbered from 1 to 40. The panel derived<br />

from the sera <strong>of</strong> five horses, two experimentally infected, two<br />

repeatedly vaccinated and one negative. The order <strong>of</strong> the sera<br />

was different for each laboratory. Each participant was requested<br />

to carry out the test three times, each time carried out by a<br />

different operator and on different days. The OD <strong>of</strong> each test was<br />

registered and returned in an Excel file. Validation criteria for the<br />

ELISA were the following: mean OD <strong>of</strong> control reaction higher<br />

than 1.0; OD <strong>of</strong> negative control < 50% <strong>of</strong> OD <strong>of</strong> control reaction<br />

for both dilutions; OD <strong>of</strong> positive control > 50% <strong>of</strong> OD <strong>of</strong> control<br />

reaction in both dilutions. Runs that did not comply with these<br />

criteria were discarded.<br />

In analysing all data the following parameters were estimated:<br />

Qualitative accuracy, estimated by:<br />

Sensitivity (Se) and specificity (Sp), Cohen K value for each<br />

laboratory, Weighted Cohen K value for each laboratory,<br />

Cohen K value for all laboratories gathered together.<br />

K values were calculated by comparing the expected results,<br />

positive and negative, with those obtained, for Cohen K value<br />

and negative, weak, medium and strong positive for Cohen K<br />

value and weighted Cohen K value.<br />

In considering both qualitative and semi-quantitative<br />

characteristics <strong>of</strong> the ELISA test, repeatability and reproducibility<br />

were estimated using the following parameters:<br />

Coefficient <strong>of</strong> variation (CV), Accordance, Concordance,<br />

Concordance Odds Ratio (COR), K value <strong>of</strong> all laboratories<br />

results gathered together.<br />

For details regarding the calculation <strong>of</strong> these parameters, articles<br />

by Langton et al., Quatto and Soliani et al. can be consulted<br />

(1,2,3,4).<br />

Results<br />

1.Both sensitivity and specificity resulted 100%.<br />

2.K values are shown in figure 1. K for all laboratories resulted<br />

equal to 0.76.<br />

K VALUE<br />

1,2<br />

1<br />

0,8<br />

0,6<br />

0,4<br />

0,2<br />

0<br />

K (2 cat.) WEIGHTED K (4 cat.) K(4 cat.)<br />

LAB 1 LAB 2 LAB 3 LAB 4 LAB 5 LAB 6 LAB 7 LAB 8 LAB 9<br />

Figure 1: K values for each laboratory: not weighted with 2 and 4<br />

categories, and weighted with two categories.<br />

3.Values <strong>of</strong> accordance, concordance and COR are showed in<br />

table 1.<br />

Table 1: Values <strong>of</strong> Accordance, concordance and COR for each<br />

serum and all laboratories together<br />

Serum N° Accordance Concordance COR Serum N° Accordance Concordance COR<br />

1 86,71 81,75 1,46 11 62,14 45,11 2,00<br />

2 95,29 95,24 1,01 12 75,43 74,87 1,03<br />

3 92,43 90,48 1,29 13 64,00 41,27 2,53<br />

4 100 100 1 14 66,86 52,78 1,80<br />

5 76,29 56,88 2,44 15 100 100 1<br />

6 95,29 95,24 1,01 16 100 100 1<br />

7 65,86 49,74 1,95 17 100 100 1<br />

8 86,71 81,75 1,46 18 100 100 1<br />

9 100 100 1 19 100 100 1<br />

10 100 100 1 20 100 100 1<br />

K value <strong>of</strong> all laboratories together not compared with expected<br />

results resulted equal to 0.72.<br />

Discussion & conclusions<br />

From the results <strong>of</strong> the evaluated parameters the test is suitable<br />

for screening purposes. Accuracy is highly satisfactory since<br />

sensitivity and specificity resulted equal to 100%. Furthermore, K<br />

values indicate a degree <strong>of</strong> concordance almost perfect<br />

according to the classification <strong>of</strong> Landis et al. (5) Qualitative<br />

repeatability and reproducibility are also satisfactory since CV<br />

values are all less than 20%, value set as acceptable limit;<br />

accordance and concordance are also close to 100% in more<br />

than the half <strong>of</strong> sera. COR resulted very close to 1 for all sera<br />

except for two <strong>of</strong> them, however with a maximum value <strong>of</strong> 2.53.<br />

An additional advantage <strong>of</strong> this c-ELISA is that it can be extended<br />

to the testing <strong>of</strong> other species for which it has to be validated. To<br />

fulfil the remaining criteria set by WOAH, a second phase <strong>of</strong> the<br />

validation procedure is necessary in which further parameters<br />

need to be evaluated, among which are the performances <strong>of</strong> this<br />

test on field samples.<br />

References<br />

1.S.D. Langton et al ” Analysing collaborative trials for qualitative<br />

microbiological methods: accordance and concordance”, International<br />

Journal <strong>of</strong> Food Microbiology 79 (2002) 175-181<br />

2.http:/www.dsa.unipr.it/soliani/soliani.html<br />

3.J. Richard Landis e Gary G. Koch del 1977 “The measurement <strong>of</strong><br />

observer agreement for categorial data” Biometrics, Vol. 33, pp.159-174.<br />

4.P. Quatto (2004). “Un test di concordanza tra più esaminatori”. In:<br />

Statistica, vol. 64, n. 1, pp. 145-151<br />

5.J. Richard Landis e Gary G. Koch del 1977 “The measurement <strong>of</strong><br />

observer agreement for categorical data” Biometrics, Vol. 33, pp.159-174.


S3 - P - 10<br />

SIMULTANEOUS DETECTION AND DIFFERENTIATION OF INFLUENZA A VIRUS<br />

AND NEWCASTLE DISEASE VIRUS BY ONE STEP RT-PCR.<br />

D. Nidzworski 1 , L. Rabalski 1 , B. Szewczyk 1 , K. Śmietanka 2 , Z. Minta 2<br />

1<br />

Intercollegiate Faculty <strong>of</strong> Biotechnology University <strong>of</strong> Gdansk and Medical University <strong>of</strong> Gdansk, Department <strong>of</strong> Molecular Virology, Gdansk, Poland<br />

2<br />

National Veterinary Research Institute in Pulawy, Department <strong>of</strong> Poultry Diseases, Pulawy, Poland<br />

Influenza virus, Newcastle disease virus, diagnosis, RT-PCR,<br />

Introduction<br />

Newcastle disease Virus (NDV), a member <strong>of</strong> the<br />

Paramyxoviridae family and Influenza A virus (IAV), from the<br />

Orthomyxoviridae family, are two main avian pathogens causing<br />

serious economic problems in poultry farming (3,4). NDV strains<br />

are classified into three major pathotypes: velogenic, mesogenic<br />

and lentogenic (2). Avian Influenza (AI) viruses are also divided<br />

into low pathogenic (LPAI) and highly pathogenic (HPAI) strains<br />

(1).Both viruses are enveloped, single stranded, negative-sense<br />

RNA viruses which give similar symptoms ranging from subclinical<br />

infections to severe disease, including loss in egg<br />

production, acute respiratory syndrome and high mortality,<br />

depending on their level <strong>of</strong> pathogenicity. This hinders the<br />

diagnosis based on clinical and post mortem examination only.<br />

Most <strong>of</strong> the currently available molecular detection methods are<br />

also pathogen-specific and require to perform more than one RT-<br />

PCR to confirm or exclude the presence <strong>of</strong> both pathogens.<br />

To overcome this disadvantage, we have applied One Step<br />

Duplex RT-PCR method to distinguish between those two<br />

pathogens. The main objective <strong>of</strong> the project was to develop new,<br />

fast and non-expensive method, which could be used in any<br />

veterinary laboratory.<br />

Materials & methods<br />

Thirty six viruses: sixteen ND Polish pigeon strains, four ND<br />

reference strains, four Influenza and twelve other (negative<br />

control strains) viruses were isolated from allantoic fluids <strong>of</strong> SPF<br />

embryonated eggs. The 4-week-old SPF chickens were also<br />

infected with both viruses (NDV – LaSota; IV – H7N1) Swabs<br />

from cloaca and trachea were collected and examined (Tab.1).<br />

Table 1: The experimental design<br />

Actions<br />

Day <strong>of</strong> experiment<br />

Inoculation <strong>of</strong> birds (4 week old) with<br />

1<br />

LaSota and H7N1 strains<br />

Collection <strong>of</strong> cloacal and oral swabs 4<br />

Collection <strong>of</strong> cloacal and oral swabs 5<br />

RNA was extracted using an RNeasy Nini Kit (Qiagen, Valencia,<br />

CA, USA). After RNA isolation, One Step RT-PCR were carried<br />

out.<br />

Two sets <strong>of</strong> primers based on conserve fragments <strong>of</strong> the M gens<br />

<strong>of</strong> NDV and IV were used in this study:<br />

NDV-F-4011: 5’ GTCCCAAATACCGGAGACCT 3’<br />

NDV-R-4142: 5’ TTGTTTGCCACAACCCTACAG 3’<br />

IV-F-VII/25: 5’ AGATGAGTCTTCTAACCGAGGTCG 3’<br />

IV-R-VII/124: 5’ TGCAAAAACATCTTCAAGTCTCTG 3’<br />

The reaction was carried out according to the Transcriptor One-<br />

Step RT-PCR Kit protocol (Roche Diagnostics, Mannheim,<br />

Germany). The reaction mixture contained: viral RNA, both sets<br />

<strong>of</strong> primers (0,3µM each), buffer (1x), Transcriptor Enzyme Mix<br />

and water. Condition <strong>of</strong> reaction presents Tab. 2<br />

Table 2. Conditions <strong>of</strong> One Step RT-PCR reaction.<br />

Step Temp. Time Cycles<br />

Reverse<br />

transcription<br />

50ºC 30 min 1<br />

Initial<br />

denaturation<br />

94ºC 7 min 1<br />

Denaturation 94ºC 10 sec<br />

Annealing 53 ºC 30 sec 10<br />

Elongation 68 ºC 105 sec<br />

PCR pr<strong>of</strong>ile<br />

Denaturation 94 ºC 10 sec<br />

Annealing 60 ºC 30 sec 25<br />

Elongation 68 ºC 105 sec<br />

Final<br />

Elongation<br />

68 ºC 7 min 1<br />

Two products were expected: one for NDV – 152 base pair<br />

fragment contained conserve M gene fragment; and one for IV –<br />

102 base pair fragment also contained conserve M gene<br />

fragment. The results were visualized in 2,5% agarose gel.<br />

Results<br />

All NDV and Influenza virus strains isolated from eggs and from<br />

oral or cloacal swabs were detected by One Step RT-PCR<br />

method. The results <strong>of</strong> analysis are presented in Figure 1 and<br />

Tab. 3.<br />

NDV IV NDV+IV ntc M<br />

152 bp<br />

102 bp<br />

Figure. 1. Analysis <strong>of</strong> One Step RT-PCR reaction. 2,5% Agarose<br />

gel with EtBr. ntc – no template control, M – Mass marker (Hyper<br />

Ladder II, BioLine)<br />

Table 3. Results <strong>of</strong> analysis <strong>of</strong> infected birds and comparison with<br />

other previously described methods.<br />

Chicken no 41 Chicken no 31<br />

AIV NDV AIV NDV<br />

A OS B OS A OS B OS<br />

4 dpi O 20.48 + n.d. n.d. 20.71 + 29.82 +<br />

4 dpi C 22.76 + 29.74 + 24.80 + >35 -<br />

5 dpi O 20.68 + 27.34 + 21.69 + 33.69 +<br />

5 dpi C 22.07 + n.d. n.d. 23.40 + >35 -<br />

Chicken no 42 Chicken no 10<br />

AIV NDV AIV NDV<br />

A OS A OS A OS B OS<br />

4 dpi O 21.68 + n.d. n.d. 21.98 + 35.00 +<br />

4 dpi C 23.94 + n.d. n.d. 19.55 + 33.39 +<br />

5 dpi O 21.86 + 34.55 + 23.34 + >35 -<br />

5 dpi C 25.01 + 34.97 + 20.72 + n.d. n.d<br />

AIV – Avian Influenza Virus; NDV – Newacastle Disease Virus; dpi – day<br />

post infection; O – Oral swab; C – Cloacal swab; n.d. – not detected; A –<br />

method <strong>of</strong> detection based on Spackman et al., 2002 (6); B – method <strong>of</strong><br />

detection based on Mia Kim et al., 2008 (5); OS – One Step RT-PCR<br />

Method; “+” – positive; “-“ – negative.<br />

Discussion & conclusions<br />

Birds infected by Newcastle Disease or Influenza virus give very<br />

similar symptoms, hard to distinguish by veterinarians even<br />

during post-mortem examination.<br />

For this reason, the new One Step RT-PCR method for direct<br />

detection and differentiation <strong>of</strong> NDV and IV was developed. One<br />

Step RT-PCR assay can be applied by veterinary diagnosticians<br />

for preliminary differentiation between NDV and Influenza virus.<br />

The ability to detect both major avian pathogens in a very easy<br />

procedure makes this method very attractive for application in<br />

veterinary laboratories.<br />

References<br />

1. Alexander, D.J. – Orthomyxovirus infections. in: Viral Infections <strong>of</strong> Birds,<br />

red: J.B. McFerran, M.S. McNulty – Elsevier Science, London 1993, 287-<br />

316<br />

2. Beard, C, Hanson, R (1984). Newcastle Disease. In: H<strong>of</strong>stad, M,<br />

Barnes, H, Calnek, B, Reid, W, Yoder, H. (Eds.) Disease <strong>of</strong> Poultry, 8 th ed.<br />

Iowa State University Press, Ames, 452-470.<br />

3. Capua, I., Alexander, D.J. - Avian influenza and human health - Acta<br />

Tropica 83 (2002), 1–6<br />

4. Lamb, R, Collins,P, Kolak<strong>of</strong>sky, D, Melero J, Nagai,Y, Oldstone, M,<br />

Pringle,C, Rima, B. Family paramyxoviridae. In: Fauquet, C, Mayo M,<br />

Manil<strong>of</strong>f J, Desselberger, U, Ball, L. (Eds.) Virus Taxonomy, VIII th Report <strong>of</strong><br />

the International Committee on Taxonomy <strong>of</strong> Viruses. Elsevier Academic<br />

Press, San Diego, 655-668.<br />

5. Mia Kim, L., Suarez, D.L., Afonso, C.L., 2008. Detection <strong>of</strong> a broad<br />

range <strong>of</strong> class I and II Newcastle disease viruses using a multiplex real<br />

time reverse transcription polymerase chain reaction assay. J. Vet. Diagn.<br />

Invest. 20, 414-425.<br />

6. Spackman, E, Senne, DA, Myers, TJ, Bulaga, LL, Garber, LP, Perdue,<br />

ML, Lohman, K, Daum, LT, Suarez, DL , 2002. Development <strong>of</strong> a real-time<br />

reverse transcriptase PCR assay for type A influenza virus and the avian<br />

H5 and H7 hemagglutinin subtypes.J. Clin Microbiol., 40, 3256


S3 - P - 11<br />

DOLPHIN MORBILLIVIRUS INFECTION IN A CAPTIVE HARBOR SEAL (PHOCA VITULINA)<br />

Sandro Mazzariol 1 , Francesco Grande 2 , Cristina Pilenga 2 , Paola Modesto 3 , Cristina Biolatti 3 , Giovanni Di<br />

Guardo 4 , Alessandra Mondin 5 , Simone Peletto 3 , Pier Luigi Acutis 3<br />

1 Department <strong>of</strong> Comparative Biomedicine and Nutrition (BCA), University <strong>of</strong> Padua, Legnaro, Italy<br />

2 Zoomarine Italia, Rome, Italy<br />

3 Istituto Zoopr<strong>of</strong>ilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Turin, Italy<br />

4 Department <strong>of</strong> Comparative Biomedical Sciences, University <strong>of</strong> Teramo, Teramo, Italy<br />

5<br />

Department <strong>of</strong> Animal Medicine and Health, University <strong>of</strong> Padua, Legnaro, Italy<br />

morbillivirus, DMV, harbor seal, transmission, phylogeny<br />

Introduction<br />

During the last 25 years morbilliviral infections have caused<br />

dramatic mortalities among several aquatic mammal species and<br />

populations worldwide (1). In particular, Dolphin Morbillivirus<br />

(DMV) represents a major biological threat for free-ranging<br />

cetaceans. In the Mediterranean Sea, DMV has been responsible<br />

for two epidemics, the first <strong>of</strong> which caused a dramatic mortality<br />

among striped dolphins (Stenella coeruleoalba) between 1990<br />

and 1992, while the second one was firstly reported in the<br />

western Mediterranean in 2006-2008, moving thereafter eastward<br />

along the French and the Italian coastlines. In June 2011, one<br />

bottlenose dolphin (Tursiops truncatus) and one striped dolphin<br />

stranded along the Tyrrhenian coastline, close to Rome. Both<br />

animals were found molecularly (RT-PCR) positive to<br />

Morbillivirus. Personnel from a marine park helped the stranding<br />

network with the bottlenose dolphin, which stranded alive and<br />

died after one day <strong>of</strong> rehabilitation. Standard quarantine protocols<br />

were applied, in order to avoid any contact with cetaceans kept in<br />

the park. On July 25 th , 2011, an 8-years-old male harbor seal<br />

(Phoca vitulina) hosted inside the aforementioned park was<br />

submitted for post-mortem examination after a short history <strong>of</strong><br />

anorexia, tremors, abdominal contractions, and polyuria with<br />

hypothermia and vomit in the last phases.<br />

Materials & methods<br />

A complete post-mortem investigation was carried out the day<br />

after death. Samples from main organs (brain, lungs, heart, liver,<br />

kidneys, GI tract, spleen, lymph nodes) were fixed in 10% neutral<br />

buffered formalin, paraffin-embedded and routinely processed for<br />

histopathology. Samples from intestine and kidneys were<br />

submitted for routine microbiological exams while cerebral,<br />

hepatic, cardiac, splenic, pulmonary and lymph node tissues<br />

were preserved frozen for molecular analyses.<br />

Molecular investigations were carried out to look for the presence<br />

<strong>of</strong> Canine Distemper Virus (CDV), Phocine Distemper Virus<br />

(PDV) and Cetacean Morbillivirus (CeMV) nucleic acids. Multiple<br />

sequence alignment was performed with the s<strong>of</strong>tware BioEdit<br />

using CLUSTAL W. An identity matrix was created to compare<br />

sequenced products with morbillivirus sequences deposited in<br />

GenBank. Phylogenetic analysis was performed using MEGA 5.0<br />

s<strong>of</strong>tware. The tree topology was inferred by the maximum<br />

likelihood method with Kimura 2-parameter model. The reliability<br />

<strong>of</strong> the topology was tested by bootstrapping 1,000 replicates.<br />

Results<br />

Necropsy revealed a mild gastro-enteritis and splenitis due to<br />

Aeromonas hydrophyla, along with hepatic and renal necrosis<br />

presumably due to endotoxemia. Besides these pathological<br />

findings, a generalized and diffuse lymph node enlargement was<br />

observed, along with a severe meningeal hyperemia and<br />

choroidal edema. Histologically, a severe and diffuse reactive<br />

lymphadenopathy was apparent, with lesions being characterized<br />

by lymphoid cell depletion, hyalinosis, and several multinucleate<br />

syncytia. A mild, multifocal, chronic choriomeningitis with white<br />

matter spongiosis and demyelination were also apparent, with<br />

eosinophilic inclusion bodies being also found in the cytoplasm <strong>of</strong><br />

brain neurons and glial cells. Furthermore, the lung showed a<br />

mild, chronic bronchointerstitial pneumonia, with simultaneous<br />

evidence <strong>of</strong> bronchiolar epithelial cell hyperplasia/hypertrophy.<br />

Pathological findings suggested a morbilliviral infection, which<br />

was confirmed by RT-PCR, with CeMV-specific genome<br />

sequences being subsequently amplified from the brain <strong>of</strong> this<br />

seal. CDV and PDV molecular analyses were negative. Similarity<br />

was 97.6-99.5% with known DMV sequences and 85.9-89.6%<br />

with other CeMV members (Porpoise Morbillivirus and the<br />

tentatively named Pilot Whale Morbillivirus). Phylogenetic<br />

analysis confirmed clustering <strong>of</strong> the harbor seal CeMV sequence<br />

with previously reported DMV sequences from cetacean species<br />

(Figure 1).<br />

53<br />

75<br />

DMV AJ608288<br />

95 DMV AY586536<br />

DMV Z36978<br />

DMV AJ224705<br />

94<br />

DMV Sc/2007 HQ829973<br />

75 DMV Gme/2007 HQ829972<br />

Harbor seal<br />

PWMV 20E FJ842382<br />

90<br />

PMV IRL88 FJ648457<br />

PMV 2990 AY586537<br />

MeV UK140H94 U29285<br />

69<br />

RPV LA96 JN234010<br />

PPRV ICV89 EU267273<br />

PDV AF479277<br />

CDV EU716337<br />

SeV X56131<br />

0.2<br />

Figure 1: Maximum Likelihood dendrogram, based on the Kimura<br />

2-parameter model, illustrating haemagglutinin gene (H)<br />

nucleotide sequence relationships <strong>of</strong> morbilliviruses.<br />

Discussion & conclusions<br />

During the last epidemic outbreak, morbilliviral infection was<br />

reported in several species besides striped dolphins, namely pilot<br />

whales (Globicephala melas), bottlenose dolphins, Risso’s<br />

dolphins (Grampus griseus), and fin whales (Balaenoptera<br />

physalus). Although the susceptibility <strong>of</strong> these cetacean species<br />

was already known, no reports other than the present one have<br />

ever described DMV transmission to harbor seals. Indeed,<br />

infection <strong>of</strong> pinniped species with morbilliviruses <strong>of</strong> cetacean<br />

origin was previously speculated on the basis <strong>of</strong> serological<br />

cross-reactivity and sequence homology (2). On a global scale,<br />

mortality events in both this and in other pinniped species such<br />

as grey seals (Halichoerus grypus), Bajkal seals (Pusa siberica),<br />

and Caspian seals (P. caspica) have been respectively linked to<br />

PDV and to CDV, with the former agent being more closely<br />

related to CDV than to any other known morbillivirus, while DMV<br />

appears to be more closely related to Rinderpest Virus (RPV).<br />

Apart from their diverging phylogeny and their phylogenetic<br />

distances, DMV and CDV could share similar epidemiologic<br />

features. This hypothesis is strongly supported by the<br />

simultaneous evidence <strong>of</strong> morbilliviral infection in the two freeranging<br />

dolphins found beached ashore near Rome before this<br />

captive seal died. Therefore, a possible route <strong>of</strong> virus (DMV)<br />

entry into the facility could have been represented by either the<br />

personnel and/or the instruments used for the rehabilitation <strong>of</strong> the<br />

Morbillivirus-infected bottlenose dolphin that was found stranded<br />

alive one month before. In conclusion, this report emphasizes<br />

that, apart from cetaceans, also pinniped species such as harbor<br />

seals are susceptible to DMV infection and related mortality.<br />

Therefore, strict quarantine procedures should be enforced also<br />

in structures where pinnipeds are kept when injured or stranded<br />

cetaceans are hospitalized within these facilities.<br />

References<br />

1. Di Guardo G, Mazzariol S, Fernández A. (2011). Biologically threatened<br />

dolphins and whales. Environmental Microbiology, 13(11), 2833-4.<br />

2. Van de Bildt MWG, Martina BEE, Vedder EJ et al. (2000). Identification<br />

<strong>of</strong> morbilliviruses <strong>of</strong> probable cetacean origin in carcases <strong>of</strong> Mediterranean<br />

monk seals (Monachus monachus). Veterinary Record, 10, 691-4.


S3 - P - 12<br />

PRELIMINARY VALIDATION OF THE ID SCREEN AFRICAN SWINE FEVER INDIRECT ELISA BASED<br />

ON THREE RECOMBINANT ASF PROTEINS<br />

Pourquier, P. 1 , Loic, C. 1,<br />

1<br />

IDvet, Montpellier, France<br />

African Swine Fever, diagnostics, serology, ELISA<br />

Introduction<br />

African swine fever (ASF) is a highly contagious, viral<br />

haemorrhagic disease <strong>of</strong> pigs, warthogs, European wild boar and<br />

American wild pigs caused by the African Swine Fever Virus<br />

(ASFV). There is neither treatment nor vaccine for ASF. To<br />

prevent introduction and spread <strong>of</strong> the disease, countries<br />

implement strict surveillance, control and eradication programs<br />

which require accurate and reliable diagnostic tests.<br />

In this context, IDvet has launched the ID Screen ® African Swine<br />

Fever Indirect ELISA. This test detects anti-ASFV antibodies in<br />

domestic or wild pigs, in serum, plasma or blood filter paper<br />

samples.<br />

Unique features <strong>of</strong> the ID Screen ® African Swine Fever Indirect<br />

ELISA include the coating <strong>of</strong> three recombinant ASFV antigens<br />

(P32, P62, and P72), and the ability to use the test with blood<br />

filter paper samples as well as serum and plasma.<br />

This study presents the preliminary validation data obtained for<br />

this ELISA kit on both serum and filter paper samples.<br />

References<br />

1. Barderas MG et al, 2000. Serodiagnosis <strong>of</strong> African Swine Fever using<br />

the recombinant protein p30 expresses in insect larvae. Journal <strong>of</strong><br />

virological methods, 89:129-136.<br />

2. Gallardo C et al, 2009. Recombinant antigen targets for serodiagnosis <strong>of</strong><br />

African Swine Fever. 16 :1012-1020.<br />

3. Gallardo C et al, 2006. Antigenic properties and diagnostic potential <strong>of</strong><br />

African Swine Fever virus protein p62 expressed in insect cells. Journal <strong>of</strong><br />

clinical microbiology, 44 : 950-956.<br />

4. Hutching GH et al, 2006. Indirect sandwich ELISA for antigen detection<br />

<strong>of</strong> African Swine Fever virus : comparison <strong>of</strong> polyclonal and monoclonal<br />

antibodies. Journal <strong>of</strong> virological methods, 131:213-217.<br />

5. OIE, 2008. Manuel terrestre de l’OIE : African Swine Fever. Chapitre<br />

2.8.1.<br />

6. Sanz et al, 1985. Monoclonal antibodies specific for African Swine Fever<br />

Virus proteins. Journal <strong>of</strong> virology, 54:199-206.<br />

Material & methods<br />

The ID Screen ® African Swine Fever Indirect is based on three<br />

recombinant ASFV antigens (P32, P62, and P72) and an antiswine-HRP<br />

conjugate. Analyses were performed as per<br />

manufacturer’s specifications.<br />

Results<br />

Specificity<br />

- 556 disease-free sera from domestic pigs (France and Norway),<br />

wild boars (France), and Iberian pigs (Spain) were tested. All<br />

samples were found negative.<br />

- 90 negative animals were tested by both the serum and filter<br />

paper protocols. All sera were found negative by both protocols.<br />

Sensitivity<br />

- 3 sera were tested from pigs vaccinated on day 0 and day 24<br />

with the ASF strain Ourt88/3, challenged on day 42 with a mild<br />

ASF strain (ANSES, Ploufragan, France), and bled on day 62 or<br />

63. After challenge, all three animals gave strong positive results<br />

with the ID Screen ® African Swine Fever Indirect ELISA.<br />

- 8 reference sera provided by the Community Reference<br />

Laboratory (CRL), CISA-INIA, Spain were analysed. The ID<br />

Screen® African Swine Fever Indirect ELISA accurately identified<br />

all sera.<br />

- 92 sera from infected herds (Sassari, Sardinia, Italy) were<br />

tested in parallel by the ID Screen ® ELISA and the CRL ELISA<br />

reagents (CISA-INIA, Spain). Test correlation was found to be<br />

95.7%. The relative sensitivity and specificity were 97.67% and<br />

97.83%, respectively.<br />

- 3 sera were titrated and tested by both the serum and filter<br />

paper protocols. The measured analytical sensitivity was similar<br />

regardless <strong>of</strong> the sample type tested (serum, Whatman 1 or<br />

Whatman 3 filter paper).<br />

Discussion & conclusions<br />

The ID Screen ® African Swine Fever Indirect ELISA is an efficient<br />

and reliable tool for the diagnosis <strong>of</strong> ASF in wild and domestic<br />

species.<br />

It is the only commercial ELISA based on 3 recombinant<br />

antigens. Both the serum and filter paper test protocols show<br />

excellent sensitivity and specificity<br />

Collecting blood using filter paper facilitates sample collection in<br />

the field. With the ID Screen protocol, filter paper samples may<br />

be tested in a 96-well deep-well plate, making sample processing<br />

faster and less prone to mix-ups.


S3 - P - 13<br />

ONE STEP RT-PCR FOLLOWED BY MSSCP AS A NOVEL METHOD FOR A FAST AND RELIABLE<br />

DETECTION AND DIFFERENTIATION OF MIXED INFECTION WITH NEWCASTLE DISEASE VIRUS OF<br />

DIFFERENT ORIGIN.<br />

L. Rabalski 1 , D. Nidzworski 1 , B. Szewczyk 1 , K. Śmietanka 2 , Z. Minta 2<br />

1<br />

Intercollegiate Faculty <strong>of</strong> Biotechnology University <strong>of</strong> Gdansk and Medical University <strong>of</strong> Gdansk, Department <strong>of</strong> Molecular Virology, Gdansk, Poland<br />

2<br />

National Veterinary Research Institute in Pulawy, Department <strong>of</strong> Poultry Diseases, Pulawy, Poland<br />

MSSCP, differentiation <strong>of</strong> strands, RT-PCR, Newcastle disease<br />

Introduction<br />

Newcastle disease virus (NDV) is an avian enveloped virus<br />

containing linear non-segmented single-stranded RNA (Lamb et<br />

al., 2005). It belongs to the Mononegavirales order,<br />

Paramyxoviridae family and Avulavirus genus. NDV strains are<br />

classified into three major pathotypes: velogenic, mesogenic and<br />

lentogenic (Beard and Hanson, 1984). Vaccination (with live<br />

virus) does not protect against the spread <strong>of</strong> virulent form <strong>of</strong> the<br />

virus. Detection <strong>of</strong> the field virus in immunized population <strong>of</strong> birds<br />

by means <strong>of</strong> conventional diagnostic methods is costly and time<br />

consuming. To overcome this disadvantage, we have applied<br />

One Step RT-PCR and Multitemperature Single-Strand<br />

Conformational Polymorphism (MSSCP) methods in our<br />

laboratories to distinguish between pathogenic and vaccine<br />

strains <strong>of</strong> the NDV. Classical single-strand conformational<br />

polymorphism (SSCP) analysis is based on the observation that<br />

single stranded DNA fragments attain a number <strong>of</strong><br />

conformational forms which may be separated by native<br />

polyacrylamide gel electrophoresis giving a characteristic pattern<br />

<strong>of</strong> electrophoretic bands. Point mutations, other minor changes in<br />

nucleotide sequence, as well as physico-chemical conditions like<br />

ionic strength, pH and temperature may have significant effect on<br />

electrophoretic pattern <strong>of</strong> single stranded DNA (Orita et al.,<br />

1989). By sequential changes <strong>of</strong> separation temperature it is<br />

possible to increase the resolution <strong>of</strong> single-strand DNA bands;<br />

this technique was named MSSCP (where M stands for<br />

“multitemperature”) (Kaczanowski et al., 2001).<br />

Materials & methods<br />

SPF chickens housed in isolation were infected oculonasally with<br />

three different strains <strong>of</strong> NDV (LaSota – lentogenic, Roakin –<br />

mesogenic and Italy – velogenic). Swabs from cloaca and<br />

oropharynx were collected at different time-points post infection<br />

(Tab.1). After RNA isolation and One Step RT-PCR, 123bp DNA<br />

<strong>of</strong> viral fusion (“F”) gene fragments were obtained (Fig. 1). PCR<br />

products were denatured (3’ in 97C) and subjected to MSSCP<br />

electrophoresis where, after silver staining, they gave specific<br />

ssDNA band patterns.<br />

Table 1: The experimental design<br />

Actions<br />

Inoculation <strong>of</strong> birds with<br />

LaSota and Roakin NDV<br />

strains<br />

Inoculation <strong>of</strong> birds with Italy<br />

NDV strain<br />

Collection <strong>of</strong> cloacal and<br />

oropharyngeal swabs<br />

Day <strong>of</strong> experiment<br />

1<br />

3<br />

3, 4 ,5 ,6 ,7, 10<br />

Results<br />

Three embryo-grown NDV strains (separately and mixed<br />

together) were tested for the level <strong>of</strong> detection. We found it<br />

possible to determine the presence <strong>of</strong> the minor variant <strong>of</strong> the<br />

virus, even when its contribution was less than 0.1% <strong>of</strong> total<br />

sample. After MSSCP analysis, characteristic ssDNA band<br />

patterns were obtained for each NDV strain. Both artificially<br />

mixed and swab samples showed a combined pattern <strong>of</strong> bands<br />

making it easy to identify the composition <strong>of</strong> samples (Fig. 2).<br />

Figure 1: 123bp PCR product at 7 days after infection (digit -<br />

identification <strong>of</strong> the chicken, G - oropharyngeal swabs,<br />

K – cloacal swabs, M - HyperLadder IV - 100bp-1000bp)<br />

Figure 2: MSSCP patterns <strong>of</strong> 123bp PCR product (M -<br />

HyperLadder IV - 100bp-1000bp, L – LaSota, R – Roakin,<br />

I – Italy, number – level <strong>of</strong> matrix concentration)<br />

Discussion & conclusions<br />

MSSCP combined with One Step RT-PCR can be applied for the<br />

detection and preliminary characterization <strong>of</strong> NDV without the<br />

need for nucleotide sequencing. The ability to diagnose multistrain<br />

infections and to detect the minor viral variant are the main<br />

advantages <strong>of</strong> the proposed method. Additionally, we confirm the<br />

observation that vaccination does not protect against virus<br />

replication and shedding in chickens.<br />

The developed method can be especially helpful in the detection<br />

<strong>of</strong> field infection in flocks immunized with live vaccines provided<br />

that we know the MSSCP pattern <strong>of</strong> an NDV strain used for<br />

vaccination. In such cases an RT-PCR method alone yields<br />

postitive results irrespective <strong>of</strong> the number <strong>of</strong> strains present in<br />

the tested sample while the sequencing shows the overlapping<br />

peaks <strong>of</strong> fluorsecence. If the MSSCP patterns <strong>of</strong> NDV present in<br />

the sample are not known, there is a possibility to cut the bands<br />

from the gel and sequence them separately.<br />

References<br />

1. Orita, M, Iwahana, H, Kanazawa, H, Hayashi, K, Sekiya, T (1989).<br />

Detection <strong>of</strong> polymorphisms <strong>of</strong> human DNA by gel electrophoresis as<br />

single-strand conformation polymorphisms. Proc. Natl. Acad. Sci.USA, 86,<br />

2766-70.<br />

2. Kaczanowski, R, Trzeciak, L, Kucharczyk, K (2001). Multitemperature<br />

single-strand conformation polymorphism. Electrophoresis, 22, 3539-45.<br />

3. Lamb, R, Collins,P, Kolak<strong>of</strong>sk, D, Melero J, Nagai,Y, Oldstone, M,<br />

Pringle,C, Rima, B. Family paramyxoviridae. In: Fauquet, C, Mayo M,<br />

Manil<strong>of</strong>f J, Desselberger, U, Ball, L. (Eds.) Virus Taxonomy, VIII th Report <strong>of</strong><br />

the International Committee on Taxonomy <strong>of</strong> Viruses. Elsevier Academic<br />

Press, San Diego, 655-668.<br />

4. Beard, C, Hanson, R (1984). Newcastle Disease. In: H<strong>of</strong>stad, M,<br />

Barnes, H, Calnek, B, Reid, W, Yoder, H. (Eds.) Disease <strong>of</strong> Poultry, 8 th ed.<br />

Iowa State University Press, Ames, 452-470.


S3 - P - 14<br />

DEVELOPMENT OF MULTISPECIES SEROLOGICAL ASSAYS TO DETECT ANTIBODIES SPECIFIC<br />

FOR Mycobacterium bovis IN SERUM SAMPLES<br />

Lisset López 1 , Ana Ranz 1 , Angel Venteo 1 , Teresa Pérez 1 , Tamara Ruiz 1 , L DeJuan 2 , Beatriz Romero 2 , Mariana<br />

Boadella 3 , Christian Gortázar 3 , Beatrice Boniotti 4 , Paolo Pasquali 5 , Paloma Rueda 1<br />

1 1 INGENASA, Madrid Spain; 2 VISAVET, Madrid, Spain; 3 IREC, Barcelona, Spain; 4 I.Z.S. della Lombardia e dell'Emilia, Brescia, Italy ; 5 Istituto Superiore di<br />

Sanità di Roma, Roma, Italy<br />

Tuberculosis, PEN-SIDE TEST, DIAGNOSTIC, ELISA DR<br />

Introduction<br />

Bovine tuberculosis (TB) is a chronic bacterial disease <strong>of</strong><br />

animals and humans caused by M. bovis. In a large number <strong>of</strong><br />

countries bovine tuberculosis is a major infectious disease<br />

among cattle, other domesticated animals, and certain wildlife<br />

populations. Transmission to humans constitutes a public health<br />

problem. For years eradication programmes have been carried<br />

out. Traditional mycobacterium culture remains the gold<br />

standard method for routine confirmation <strong>of</strong> infection but<br />

Delayed Hypersensitivity test and gamma-interferon test are the<br />

ones used in these programmes. Detection <strong>of</strong> antibodies<br />

specific could be a useful alternative for TB diagnosis. Different<br />

antibodies detection assays have been described although all <strong>of</strong><br />

them present sensitivity problems<br />

The aim <strong>of</strong> this study has been to develop and validate<br />

multispecies assays based on Double Recognition ELISA<br />

(ELISA-DR) and Immunochromatography (IC) techniques for<br />

detection <strong>of</strong> antibodies specific <strong>of</strong> M. bovis. using the<br />

recombinant protein MPB83, expressed in insect cells.<br />

Material & methods<br />

Recombinant proteins ESAT6, CFP10, MPB64, MPB70 y MPB<br />

83 were obtained, expressed and purified. Finally, MPB83 was<br />

selected as the antigen for serological assays (ELISA and IC<br />

assays)<br />

To validate these assays, sera from different species previously<br />

catalogued by gamma interferon assay (72 bovine sera), by<br />

culture (46 buck, 205 wild boar) or by necropsy (205 deer and<br />

138 wild boar experimental sera) were used. Moreover, cross<br />

reactivity with M. avium subsp. paratuberculosis was<br />

determined. For this propose sera and plasma <strong>of</strong> 9 cows and 9<br />

goats positive to Paratuberculosis and negative to TB were<br />

used.<br />

Samples from different wild species such as alpaca, elephant<br />

and American bison are going to be tested. In addition sera<br />

collected from wild boar and deer are being evaluated.<br />

Table 2.<br />

Ref: ELISA DR<br />

SPECIES N Accuracy<br />

CATTLE 13 77%<br />

WILD BOAR 200 90%<br />

BUCK 46 89%<br />

DEER 22 77%<br />

Discussion & conclusions<br />

INGENASA has developed two multispecies assays based on<br />

the use <strong>of</strong> the recombinant protein MPB83 expressed in insect<br />

cells. More than 300 samples from different species have been<br />

analyzed giving high percentages <strong>of</strong> sensitivity and specificity.<br />

Although higher number <strong>of</strong> samples is necessary, preliminary<br />

results suggest that both diagnostic approaches are very useful<br />

methods for control and surveillance <strong>of</strong> TB infection.<br />

The immunochromatographic assay can become a useful tool in<br />

situations where laboratory support and skilled personnel are<br />

limited.<br />

References<br />

1. Schiller et al. Transboundary and Emerging Diseases. (2010),<br />

57:205-220<br />

2. Boadella et al. Preventive Veterinary Medicine (<strong>2012</strong>), 104:160-164<br />

3. Waters et al. Clinical and Vaccine Immunology (2011), 18:1882-1888<br />

(KBBE-2007-1-3-04) Strategies for the eradication <strong>of</strong> bovine tuberculosis<br />

(TB-STEP)<br />

Results.<br />

Preliminary results show that both assays detect antibodies<br />

specific <strong>of</strong> TB. None <strong>of</strong> them showed cross-reaction with<br />

antibodies to PTB. Depending on the species analyzed,<br />

sensitivity and specificity <strong>of</strong> ELISA-DR range between 70-100%<br />

and 92-97%, respectively (Table I). Concerning to IC, accuracy<br />

with respect to ELISA DR ranged between 77 and 90%<br />

depending on the species studied (TABLE II).<br />

Table 1<br />

ELISA-DR<br />

SPECIES N Sensitivity Specificity REFERENCE<br />

CATTLE 72 88% 96% Bovigam®<br />

WILD BOAR 138 100% 97% Necropsy<br />

WILD BOAR 200 82% 92% Culture<br />

BUCK 46 70% 94% Culture<br />

DEER 205 93% 92% Necropsy


S3 - P - 15<br />

FIRST REPORTED LOW PATHOGENCITY AVIAN INFLEUNZA VIRUS SUBTYPE H9 INFECTION OF<br />

DOMESTIC FOWL IN ENGLAND<br />

C. Daniel Parker 1 , Scott M. Reid 2 , Allan Ball 1 , William J. Cox 2 , Steve C. Essen 2 , Amanda Hanna 2 ,<br />

Marek J. Slomka 2 , Richard M. Irvine 2 , Ian H. Brown 2<br />

1 Slate Hall Veterinary Practice Ltd, Unit 7 Highgate Farm, Over Road, Willingham, Cambridge, CB24 5EU<br />

2 Animal Health and Veterinary Laboratories Agency-Weybridge, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB<br />

Avian influenza virus, H9 real-time RT-PCR, broiler breeders, UK poultry, Egg drop in poultry<br />

Introduction<br />

Poultry outbreaks, caused by avian influenza (AI) viruses <strong>of</strong><br />

H9N2 subtype, have been reported in many parts <strong>of</strong> Asia and the<br />

Middle East since the 1990s where they may be considered as<br />

being endemic (1, 2). Although H9 virus infections are typically<br />

characterised as being <strong>of</strong> low pathogenicity (LP) and are not<br />

classified as notifiable disease (3), LPAI H9N2 poultry outbreaks<br />

in these regions continue to be characterised clinically by<br />

respiratory disease and a reduction in egg production (4).<br />

However, since the more recent emergence <strong>of</strong> a significant H9N2<br />

poultry problem in Asia and the Middle East, the present paper<br />

presents the first detailed epidemiological investigation <strong>of</strong> a H9<br />

poultry outbreak in the UK and indeed in Europe. This occurred in<br />

the East Anglia region in December 2010 in a broiler breeder<br />

flock, and an important aspect <strong>of</strong> the outbreak investigation was<br />

the successful use <strong>of</strong> molecular methods to identify and<br />

characterise the AIV subtype.<br />

Materials & methods<br />

Disease suspicion was based on acute drops in egg production in<br />

two <strong>of</strong> four sheds, poor egg shell quality and evidence <strong>of</strong><br />

diarrhoea. Cloacal and oropharyngeal swabs from 60 chickens<br />

from each <strong>of</strong> two affected houses were pooled (n=48) into groups<br />

<strong>of</strong> five swabs obtained from the same anatomical site. All swab<br />

pools were inoculated into 10-day-old embryonated SPF fowls’<br />

eggs, and allantoic fluids were harvested and tested for the<br />

presence <strong>of</strong> any haemagglutinating agent (5). Four AI real-time<br />

reverse transcription polymerase chain reaction (RRT-PCR)<br />

assays were used to test the RNA extracts: (i) the Matrix (M)-<br />

gene assay for generic AI detection; (ii) H5 and H7 AI virus RRT-<br />

PCR assays to test for notifiable avian influenza (NAI); (iii) an H9<br />

RRT-PCR; (iv) selected RNA extracts were tested by an N1 RRT-<br />

PCR. The RNA extracts were simultaneously screened for<br />

Newcastle disease virus (NDV) using primers and probes<br />

targeting the L-gene <strong>of</strong> NDV. RNA extracted from the positive<br />

cloacal swab specimen A/chicken/England/1415-51184/10 was<br />

used for nucleotide sequencing. The HA and neuraminidase (NA)<br />

genes were amplified by conventional reverse transcription (RT)-<br />

PCR, sequenced and analysed phylogenetically.<br />

poultry for 40 years vindicates the need for continued vigilance<br />

and surveillance <strong>of</strong> AI viruses in poultry populations.<br />

Acknowledgements<br />

The authors thank Andrew Gibson, Mark Spelman and Simon<br />

Rednall from the farm management team, Sue Graham <strong>of</strong> Slate<br />

Hall Veterinary Practice Ltd for her laboratory assistance and Dr<br />

Dennis Alexander <strong>of</strong> AHVLA for technical assistance. This study<br />

was supported by Defra contract ED1300: Scanning Surveillance<br />

for Disease in Poultry and Game Birds in England, Scotland and<br />

Wales.<br />

References<br />

1.Alexander, DJ (2007). An overview <strong>of</strong> the epidemiology <strong>of</strong> avian<br />

influenza. Vaccine 25:5637–5644.<br />

2.Capua, I, Alexander, DJ (2007) Avian influenza infections in birds – a<br />

moving target. Influenza and Other Respiratory Viruses 1:11–18.<br />

3.OIE (World Organisation for Animal Health). (2008). World Health<br />

Organisation for Animal Health, Terrestrial Animal Health Code, chapter<br />

10.4 “Avian influenza”. OIE: Paris. Available at:<br />

http://www.oie.int/eng/normes/mcode/en_chapitre_1.10.4.pdf<br />

4.Swayne, DE, Halvorson, DA (2003). Influenza. In ”Diseases <strong>of</strong> Poultry”,<br />

pp135-160. Eds: YM Saif, HJ Barnes, JR Glisom, AM Fadly, LR<br />

McDougald and DE Swayne. Ames, Iowa: Iowa State University Press.<br />

5.OIE (World Organisation for Animal Health). (2010). Manual <strong>of</strong><br />

Diagnostic Tests and Vaccines for Terrestrial Animals, chapter 2.3.4.<br />

“Avian influenza”. Paris: OIE. Available at:<br />

http://www.oie.int/fileadmin/Home/eng/Health_standards/tahm/2.03.04_AI.<br />

p<br />

Results<br />

Attempted virus isolation in embryonated SPF fowls’ eggs was<br />

unsuccessful. H9N1 LPAI virus infection was confirmed by RRT-<br />

PCR. Sequencing <strong>of</strong> the haemagglutinin and neuraminidase<br />

genes revealed high nucleotide identity <strong>of</strong> 93.6% and 97.9% with<br />

contemporary North American H9 and Eurasian N1 genes,<br />

respectively. Epidemiological investigations were conducted to<br />

identify the source <strong>of</strong> infection and any onward spread.<br />

Discussion & conclusions<br />

This is the first known report <strong>of</strong> an H9 subtype avian influenza<br />

infection in poultry in England. The infecting subtype was H9N1<br />

and phylogenetic analysis <strong>of</strong> the HA and NA genes indicated<br />

North American and Eurasian origins for these two genes, which<br />

suggested that this AIV may be a reassortment that consists <strong>of</strong><br />

genetic segments <strong>of</strong> diverse geographic origin. The source <strong>of</strong> the<br />

influenza virus infection could not clearly be established.<br />

Following the infection, epidemiological investigations failed to<br />

identify any human, inter- or intra-company contacts that could<br />

have introduced the virus onto the farm. No other breeder or<br />

poultry farms in the area were known to be infected. Infection<br />

followed a period <strong>of</strong> extremely cold weather and snow which<br />

could have compromised disease security. Wild bird activity<br />

around the farm buildings was significantly higher during this cold<br />

weather period as birds foraged for feed. In the present episode,<br />

clinical signs were relatively mild in the poultry with no mortality.<br />

However, this first reported detection <strong>of</strong> H9 LPAI virus in UK


S3 - P - 16<br />

DETECTION OF IBV QX IN COMMERCIAL POULTRY FLOCKS IN THE UNITED KINGDOM<br />

Richard M. Irvine 1 , Rebecca M. Jones 1 , Richard J. Ellis 1 , Jennifer L. Cork 1 ,<br />

Jane Errington 2 , William J. Cox 1 , Vanessa Ceeraz 1 , Alisdair M. Wood 3 , Scott M. Reid 1 ,<br />

Ian H. Brown 1<br />

1 Animal Health and Veterinary Laboratories Agency – Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK;<br />

2 Animal Health and Veterinary Laboratories Agency – Penrith, Merrythought, Calthwaite, Penrith, Cumbria CA11 9RR, UK;<br />

3 Animal Health and Veterinary Laboratories Agency – Lasswade, International Research Centre, Pentland Science Park, Bush Loan, Penicuik, Midlothian<br />

EH26 0PZ, UK.<br />

Infectious bronchitis virus, IBV QX strain, IBV real-time RT-PCR, IBV S1 genotyping, UK commercial poultry flocks<br />

Introduction<br />

The QX strain <strong>of</strong> infectious bronchitis virus (IBV) was first<br />

detection in China between 1995 and 2004, associated with<br />

nephritis, mortality and egg production problems in IBVvaccinated<br />

flocks (1). Strains <strong>of</strong> IBV showing a close relationship<br />

with the so-called Chinese QX strain <strong>of</strong> IBV (IBV QX) have<br />

recently been detected in the UK. Following the initial isolation <strong>of</strong><br />

a QX-like variant strain serologically unrelated to the major UK<br />

contemporary IBV strains from a small backyard flock during<br />

2007, European IBV QX-like strains have been detected in<br />

backyard and hobby flocks. Since 2009, such strains have also<br />

been detected in samples submitted for diagnostic investigation<br />

from commercial poultry flocks (2).<br />

Materials & methods<br />

Diagnostic samples testing positive by IBV real-time reverse<br />

transcription polymerase chain reaction were subject to partial<br />

sequencing <strong>of</strong> the S1 gene (3). Primers and probes were<br />

optimised for sequencing a fragment <strong>of</strong> the S1 gene (c. 450 bp).<br />

For an initial genotyping test, a 140bp region was compared to a<br />

library <strong>of</strong> IBV S1 gene sequences.<br />

Results<br />

Comparison <strong>of</strong> the entire 450bp region for the QX strains allowed<br />

separate sub-lineages to be identified. Whilst the European IBV<br />

QX-like genotypes detected from commercial poultry flocks in<br />

Great Britain (GB) and Northern Ireland are closely related,<br />

differences are apparent in the S1 sequence, revealing separate<br />

sub-lineages (Figure 1). The greatest differences occur between<br />

European IBV QX-like strains detected from backyard poultry and<br />

commercial flocks. Furthermore, strains from some commercial<br />

poultry flocks in GB share a higher degree <strong>of</strong> S1 sequence<br />

homology with each other and a strain detected from poultry in<br />

The Netherlands.<br />

SA12- 008180<br />

SA12- 013762<br />

SA12- 007589<br />

SA12- 009984<br />

SA12- 009987 SA12-013717<br />

SA12-013719<br />

SA12-013721<br />

SA12-013722<br />

SA12-011754<br />

SA12-011755<br />

SA12-006209<br />

SA12-006211<br />

SA12- 010211<br />

SA12- 010573<br />

SA12- 010575<br />

SA12- 010825<br />

SA12- 010827<br />

SA12-005257<br />

SA12-010215<br />

SA12-004866<br />

SA12-006034<br />

Jun- 164-S3<br />

AVP - 09- 023587<br />

AVP - 09- 023588<br />

SA12- 011173<br />

Poulvac-QX -Vaccine<br />

QX AV2150/07<br />

SA12-014043<br />

AV418-trachea<br />

AVI-08- 035383<br />

AVI-08-022254<br />

SA12-011670<br />

Discussion & conclusions<br />

Together, these data revealed differences by sector, suggesting<br />

that separate routes <strong>of</strong> introduction may have occurred.<br />

Emergence <strong>of</strong> a novel variant such as IBV QX might threaten<br />

commercial poultry production as currently-used vaccines might<br />

not be fully protective and the severity <strong>of</strong> disease caused by<br />

novel strains may differ from that caused by extant strains. This<br />

may result in substantial losses. Experimental studies have<br />

demonstrated that protection may be afforded against infection<br />

with IBV QX-like strains in young chickens by existing vaccines<br />

and vaccine combinations (4). However, further work is required<br />

to fully understand the epidemiology <strong>of</strong> IBV QX in UK poultry<br />

populations.<br />

Acknowledgements<br />

This study was supported by Defra contract ED1300: Scanning<br />

Surveillance for Disease in Poultry and Game Birds in England,<br />

Scotland and Wales.<br />

References<br />

1. Liu, SW, Zhang, QX, Chen, JD, Han, ZX, Liu, X, Feng, L, Shao, YH,<br />

Rong, JG, Kong, XG, Tong, GZ (2006). Genetic diversity <strong>of</strong> avian<br />

infectious bronchitis coronavirus strains isolated in China between 1995<br />

and 2004. Archives <strong>of</strong> Virology 151: 1133-1148.<br />

2. Irvine, RM, Cox, WJ, Ceeraz, V, Reid, SM, Ellis, RJ, Jones, RM,<br />

Errington, J, Wood, AM, McVicar, C, Clark, MI (2010). Detection <strong>of</strong> IBV QX<br />

in commercial broiler flocks in the UK. Veterinary Record 167:877-879.<br />

http://veterinaryrecord.bmj.com/content/167/22/877.2.full.pdf<br />

3. Jones, RM, Ellis, RJ, Cox, WJ, Errington, J, Fuller, C, Irvine, RM,<br />

Wakeley, PR (2011). Development and Validation <strong>of</strong> RT-PCR Tests for the<br />

Detection and S1 Genotyping <strong>of</strong> Infectious Bronchitis Virus and other<br />

Closely Related Gammacoronaviruses Within Clinical Samples.<br />

Transboundary and Emerging Diseases 58 (5): 411–420.<br />

http://onlinelibrary.wiley.com/doi/10.1111/j.1865-<br />

1682.2011.01222.x/abstract.<br />

4 Terregino, C, T<strong>of</strong>fan, A, Beato, MS, Nardi, R, Vascellari, M, Meini, A,<br />

Ortali, G, Mancin, M, Capua, I (2008). Pathogenicity <strong>of</strong> a QX strain <strong>of</strong><br />

infectious bronchitis virus in specific pathogen free and commercial broiler<br />

chickens, and evaluation <strong>of</strong> protection induced by a vaccination<br />

programme based on the Ma5 and 4/91 serotypes. Avian Pathology 37<br />

(5) : 487-493.<br />

0.01<br />

Broilers; Broiler breeders; Layer; Backyard<br />

Figure 1: Evolutionary relationships


S3 - P - 17<br />

APPLICATION OF REVERSE TRANSCRIPTION REAL-TIME PCR TO DETECT THE<br />

SCHMALLENBERG VIRUS GENOME<br />

Nikolai Salnikov, Helena Nikitina, Sodnom Tsybanov, Denis Kolbasov<br />

National research institute for veterinary virology and microbiology <strong>of</strong> Russia, Pokrov, Russia<br />

Schmallenberg disease, virus, PCR<br />

Introduction<br />

The disease Schmallenberg was registered among cattle, sheep<br />

and goats in Germany, the Netherlands, Belgium, Britain, France,<br />

Italy, Luxembourg and Spain (3,4). The Schmallenberg virus is<br />

a member <strong>of</strong> the genus Orthobunyavirus family Bunyaviridae.<br />

Established the pathogenicity <strong>of</strong> the Schmallenberg virus for<br />

sheep, goats and cattle. Clinical signs <strong>of</strong> the disease in<br />

cattle include fever, drop in milk yield (50%), loss <strong>of</strong> appetite and<br />

sometimes diarrhea. If an adult ewe or heifer was infected in the<br />

early stages <strong>of</strong> pregnancy, the fetal infection can occur,<br />

leading to serious consequences: abortion, birth <strong>of</strong> premature or<br />

dead fetuses and lambs, goats and calves with<br />

various malformations. At detection in animals the characteristic<br />

clinical signs <strong>of</strong> the Schmallenberg disease carry out<br />

activities for virus isolation in cell cultures, detection <strong>of</strong> the<br />

virus genome by PCR and antibodies to it in the neutralization<br />

test (1,2). The aim <strong>of</strong> this work was the development the test -<br />

system to detect the Schmallenberg virus genome by real-time<br />

quantitative reverse transcription PCR (RT-qPCR).<br />

Materials & methods<br />

In our work the referent RNA samples <strong>of</strong> the Schmallenberg virus<br />

were used. Also we used the different strains <strong>of</strong> such viruses, as<br />

Nairobi sheep disease, Akabane, Rift Valley fever and<br />

bluetongue. Extraction <strong>of</strong> viral RNA was performed using "Trizol''<br />

(Trizol Reagent, Life Technology, USA). RT-qPCR was carried<br />

out on the thermocycler Rotor Gene 6000 (Corbett Research,<br />

Australia). Cloning <strong>of</strong> the amplified Schmallenberg virus<br />

genome's fragment was performed using the "Ins TAclone PCR<br />

cloning kit"(Fermentas, Latvia). The synthesis <strong>of</strong> RNA from<br />

cloned DNA inserts was performed using the kit "RiboMAX Large<br />

Scale RNA Production System T7" (Promega, USA).<br />

preparations <strong>of</strong> heterologous viruses and RNA extracted from the<br />

intact samples has not been received positive results. This<br />

proves the specificity <strong>of</strong> the primers and the probe included in<br />

the test - system. The analytical sensitivity <strong>of</strong> the developed test -<br />

system is 7,178*10^5 RNA copies / mkl.<br />

Acknowledgements<br />

We thank for the referent RNA samples <strong>of</strong> the Schmallenberg<br />

virus Dr. Martin Beer and Dr. Bernd H<strong>of</strong>fman, Institute <strong>of</strong><br />

Diagnostic Virology, Friedrich Loeffler Institute, Germany.<br />

References<br />

1. Gibbens, N. Schmallenberg virus: a novel viral disease in northern<br />

Europe. <strong>2012</strong>. Vet. Rec.17: 58.<br />

2. H<strong>of</strong>fmann B, Scheuch M, Höper D, Jungblut R, Holsteg M, Schirrmeier<br />

H, et al. Novel orthobunyavirus in cattle, Europe, 2011. Emerg. Infect.<br />

Dis. <strong>2012</strong> Mar.<br />

3. Latest posts on Pro Med-mail. International society for infectious<br />

diseases, 2011-<strong>2012</strong>. http://www.promedmail.org.<br />

4. Weekly disease information/WAHID Interface/OIE, 2011-<strong>2012</strong>.-<br />

http://www.oie.int.<br />

Results<br />

With order to select primers the available in Gen Bank nucleotide<br />

sequences <strong>of</strong> Schmallenberg virus Genome were analyzed using<br />

the programs Bio Edit 7.0 and Oligo 6.0. As a result a pair <strong>of</strong> the<br />

primers flanking the fragment <strong>of</strong> 130 bp within the gene N <strong>of</strong><br />

Schmallenberg virus was designed. For detection <strong>of</strong> amplification<br />

products in real-time the probe Taq-man, which contains at the<br />

5'-end the fluorophores HEX and at the 3'-end – quencher BHQ2<br />

was designed. Analytical sensitivity <strong>of</strong> the method was<br />

determined using in vitro transcripts synthesized on the template<br />

<strong>of</strong> the recombinant plasmid with the Schmallenberg virus<br />

genome fragment's insertion. Ten-fold serial dilutions <strong>of</strong> in<br />

vitro transcribed RNA (<strong>of</strong> known concentration) were used to<br />

determine analytical sensitivity <strong>of</strong> RT-q PCR. The limit <strong>of</strong><br />

sensitivity considered the maximum dilution at which a positive<br />

result was registered.<br />

The calculated value <strong>of</strong> analytical sensitivity <strong>of</strong> RT-PCR in realtime<br />

was 7.178*10^5 RNA copies /mkl, which corresponds<br />

to 3,698*10^6 copies <strong>of</strong> RNA per reaction. To evaluate<br />

the specificity <strong>of</strong> the test - system the RNA samples <strong>of</strong> Nairobi<br />

sheep disease, Akabane, Rift Valley fever and bluetongue<br />

viruses were examined. We also examined the RNA samples<br />

extracted from the intact cultures <strong>of</strong> kidney cells <strong>of</strong> antelope,<br />

BHK-21/13, CV-1, blood samples and organs from intact animals<br />

(goats, sheep, cattle, mice). With any <strong>of</strong> the examined RNA<br />

samples has not been a received positive result in RT-qPCR. By<br />

using the developed method over 500 blood samples and 15<br />

samples <strong>of</strong> placenta and meconium from clinically<br />

healthy pregnant heifers imported from Germany and the<br />

Netherlands in 2011-<strong>2012</strong> in the farms <strong>of</strong> Nizhny Novgorod,<br />

Moscow and Kaluga regions were examined. This also was<br />

not obtained positive results.<br />

Discussion & conclusions<br />

The designed test -system is suitable to detect the<br />

Schmallenberg virus genome in blood samples and<br />

organs <strong>of</strong> sheep and cattle. In the investigation <strong>of</strong> RNA


S3 - P - 18<br />

PREVALENCE OF COXIELLA BURNETII ANTIBODIES IN SHEEP AND GOATS IN FINLAND<br />

Teresa Skrzypczak 1 , Tiina Autio 2 , Jaana Seppänen 1 , Sinikka Pelkonen 1,2<br />

1<br />

Finnish Food Safety Authority, Evira, Research and Laboratory Department, Veterinary Bacteriology, Helsinki, Finland<br />

2<br />

Finnish Food Safety Authority, Evira, Research and Laboratory Department, Veterinary Bacteriology, Kuopio, Finland<br />

Coxiella burnetii, Q-fever, sheep, goat, prevalence<br />

Introduction<br />

Coxiella burnetii is an intracellular pathogen which causes a<br />

highly contagious zoonotic disease, Q fever. The infection is<br />

endemic worldwide and has been detected in several European<br />

countries in goat, sheep and cattle populations (2, 4). The<br />

transmission <strong>of</strong> the infection from goats to humans has caused<br />

severe zoonotic epidemic in the Netherlands in the last few years<br />

(3, 5). In Finland, C. burnetii infection was detected for the first<br />

time in 2008 in a dairy herd. Seropositive animals were found in<br />

ELISA tests made for export control. A succeeding PCR test <strong>of</strong><br />

the bulk tank milk sample was positive for C. burnetii. No clinical<br />

signs <strong>of</strong> Q fever were observed in the herd. A later performed<br />

nationwide prevalence study <strong>of</strong> dairy herds in 2009 indicated that<br />

the prevalence <strong>of</strong> C. burnetii infection in cattle population was<br />

extremely low. Only two out <strong>of</strong> 1238 tested herds were<br />

seropositive (1). In this study we assessed the prevalence <strong>of</strong> C.<br />

burnetii antibodies in sheep and goat populations in Finland.<br />

References<br />

1. Autio, T., Skrzypczak, T., Pohjanvirta, T., Pelkonen, S. 2010.<br />

Low prevalence <strong>of</strong> Coxiella burnetii (Q-fever) antibodies in dairy herds in<br />

Finland. 25th World Buiatrcis Congress, Chile.<br />

2. Guatteo, R., Seegers, H., Taurel, A.-F., Joly, A., Beaudeau, F. 2011.<br />

Prevalence <strong>of</strong> Coxiella burnetii infection in domestic ruminants: A critical<br />

review. Vet. Microbiol. 149:1-16.<br />

3. Roest, H.I.J., Tilburg, J.J.H.C., van Der Hoek, W., Vellema, P., van Zijderveld,<br />

F.G., Klaassen, C.H.W., Raoult, D. The Q fever epidemic in The Netherlands:<br />

history, onset, response and reflection. 2011. Epidemiol. Infect. 139: 1–12.<br />

4. Ryan, E, Kirby, M, Clegg, T, Collins, D,M. 2011. Seroprevalence <strong>of</strong><br />

Coxiella burnetii antibodies in sheep and goats in the Republic <strong>of</strong> Ireland.<br />

Vet. Record, 169, 280b<br />

5. Schimmer,B, Luttikholt, S, Hautvast, J.L.A, Graat, E.A.M, Vellema, P,<br />

van Duynhoven, Y.T.H.P.2011. Seroprevalence and risk factors <strong>of</strong> Q fever<br />

in goats on commercial dairy goat farms in the Netherlands, 2009-2010.<br />

BMC Vet. Res. 2011,7:81<br />

Materials & methods<br />

We used serum samples collected for sheep Meadia-Visna-virus<br />

(MV) and goat Caprine arthritis-encephalitis-virus (CAE)<br />

surveillance study in year 2010. According to the legislation, all<br />

herds having 20 eves or more must be tested for MV and CEA.<br />

The smaller herds remained out <strong>of</strong> this study.<br />

In total, we obtained 8545 ovine sera from 246 farms and 819<br />

caprine sera from 24 farms. From the herds with less than 50<br />

animals, all animals were included in the study. From herds with<br />

more than 50 animals, 50 sera were chosen randomly. The<br />

samples <strong>of</strong> individual herds were combined into pools <strong>of</strong> ten<br />

samples. Altogether 740 pooled samples from sheep were<br />

tested. Caprine samples were mainly studied individually, except<br />

from one herd with more than 200 animals. In case <strong>of</strong> a positive<br />

test result, all samples from a positive pool were tested<br />

separately.<br />

The samples were tested using CHEKIT Q-fever antibody ELISAtest<br />

(IDEXX, Liebefeld-Bern, Switzerland).<br />

Results<br />

All the tested samples were negative for C. burnetii antibodies.<br />

The negative results indicate that the seroprevalence <strong>of</strong> C.<br />

burnetii in the ovine population was less than 0.0004 % and<br />

caprine population less than 0.0048 %, with 95 % confidence.<br />

Discussion & conclusions<br />

This was the first time that Q fever antibodies were analyzed in<br />

sheep and goats in Finland. All tested farms and animals were<br />

negative. There were altogether 129 091 sheep on 1349 farms<br />

and 4 890 goat on 165 farms at the time <strong>of</strong> sampling in 2010. The<br />

sampling covered 6.6 % <strong>of</strong> sheep and 16.7 % <strong>of</strong> goat populations<br />

in the country, and represented 18.2% <strong>of</strong> ovine and 14.5% <strong>of</strong><br />

caprine farms situated in different parts <strong>of</strong> the country. The study<br />

shows extremely low prevalence <strong>of</strong> C. burnetii infection in sheep<br />

and goats in Finland. The finding is in line with the detected low<br />

prevalence <strong>of</strong> the infection in dairy cattle herds in 2009 (1). The<br />

Nordic climate, small herd size <strong>of</strong> sheep and goat farms and<br />

small density <strong>of</strong> these farms may have accounted for the<br />

favorable situation. We conclude that C. burnetii infection is very<br />

rare in domestic ruminants in Finland.<br />

Acknowledgements<br />

We thank Riikka Mäkinen for excellent technical assistance.


S3 - P - 19<br />

DETECTION OF SCHMALLENBERG VIRUS IN BOVINE SEMEN BY ONE-STEP REAL-TIME RT-PCR<br />

Remco Dijkman 1 , Jet Mars 1 , Gerard Wellenberg 1<br />

1<br />

Animal Health Service, Deventer, The Netherlands<br />

2<br />

Affiliation Institute name, Department name, City, Country<br />

Schmallenberg virus, semen, real-time PCR<br />

Introduction<br />

For the detection <strong>of</strong> the Schmallenberg virus (SBV) in serum,<br />

tissues and body fluids, a real-time PCR, based on the detection<br />

<strong>of</strong> specific regions <strong>of</strong> the L-segment <strong>of</strong> the Schmallenberg virus,<br />

was developed (1). Recently, two real-time PCR methods, based<br />

on the detection <strong>of</strong> S-segment sequences, were developed too.<br />

However, these PCR methods have not been validated for<br />

semen samples.<br />

In this study, we have examined whether the L-PCR, S3-PCR<br />

and an “in-house” S-PCR were suitable for the detection <strong>of</strong> SBV<br />

in bovine semen.<br />

Materials & methods<br />

Besides the L1 and S3 PCR, which were developed by FLI<br />

(Germany), we also used an “in-house” pan-orthobunyavirus S-<br />

PCR (OBV-S PCR) real-time PCR targetting the orthobunyavirus<br />

S-segment for the detection <strong>of</strong> SBV in bovine semen samples.<br />

RNA was extracted with the AM1836 extraction kit (Life<br />

Technologies) using the MagMAX express 96 system (Life<br />

Technologies). The AgPath-ID one-step RT-PCR kit (Life<br />

Technologies) was used for amplification. PCR amplification was<br />

performed on a ABI7500 sequence detection system from<br />

Applied Biosystems (Life Technologies).<br />

The validation <strong>of</strong> the three PCR tests was based on the<br />

determination <strong>of</strong> the detection limit (lowest detectable amount <strong>of</strong><br />

SBV per mL semen) for undiluted (fresh semen) and diluted<br />

semen (straw) samples.<br />

Therefore, ten-fold dilutions from a Schmallenberg virus stock<br />

solution with a known titer (10 4.9 TCID50/mL) were used for<br />

spiking semen samples and subsequently used for analyses.<br />

This study was also focused on spiking experiments to determine<br />

the negative influences <strong>of</strong> inhibitory compounds in semen<br />

samples as they may lead to false-negative results.<br />

As it is unknown whether SBV is excreted by semen, and natural<br />

SBV infected semen samples were not available, spiking<br />

experiments have been performed with undiluted (fresh semen)<br />

and diluted semen (straw) samples. Undiluted semen from 8<br />

different bulls (bulls A t / H) and diluted semen from 14 bulls<br />

(bulls I t / m V) were used. The recovery <strong>of</strong> the SBV in undiluted<br />

and diluted semen samples was determined by PCR and<br />

compared with the results <strong>of</strong> control samples.<br />

Results<br />

In silico analyses showed that the primers and probes <strong>of</strong> the L-<br />

and S3-PCR are specific for SBV, and that the selected primers<br />

and probe <strong>of</strong> the OBV S-PCR recognize conserved regions within<br />

the S-segment <strong>of</strong> SBV, also present in other orthobunyaviruses.<br />

The results <strong>of</strong> the detection limits <strong>of</strong> the three real-time PCR<br />

methods are reported in Table 1.<br />

straws were comparable or to a maximum <strong>of</strong> 4 Ct-values higher<br />

than the Ct-value <strong>of</strong> the Positive Reference sample (PBS matrix).<br />

The semen samples without spiking were negative in all three<br />

PCR methods (Figure 1).<br />

Figure 1: Ct-values <strong>of</strong> 1:3 diluted (fresh) semen and diluted<br />

semen (straws)spiked with SBV as obtained by L-, S3- and the<br />

OBV-S PCR.<br />

Discussion & conclusions<br />

This validation study showed that the L-, the S3-, and the OBV-S<br />

PCR are able to detect SBV in 1:3 diluted fresh semen and<br />

diluted (straw) semen samples. The detection limits <strong>of</strong> the S3-<br />

and the OBV-S PCR are lower compared to the detection limit <strong>of</strong><br />

the L-PCR.<br />

In addition, the spiking experiments, to examine the influence <strong>of</strong><br />

inhibitory compounds in bovine semen samples, showed that no<br />

negative influences were recorded in 1:3 diluted semen and in<br />

diluted semen (straw) samples. Therefore, fresh semen needs to<br />

be diluted prior to the RNA-extraction and semen straws can be<br />

examined by PCR without further dilution.<br />

The use <strong>of</strong> an internal control, to check for inhibitory effects <strong>of</strong> the<br />

matrix, was not examined in this study, but can be the scope for<br />

further validation.<br />

As fresh semen can be diluted 1:3, and semen in straws are<br />

diluted approximately between 1:20 and 1:30, the screening <strong>of</strong><br />

fresh semen is more sensitive than straws.<br />

It might be useful to use the OBV-S PCR as a general screening<br />

PCR, and the S3-PCR can be used as a screening and/or<br />

confirmatory PCR.<br />

References<br />

1. H<strong>of</strong>fmann B, Scheuch M, Höper D, Jungblut R, Holsteg M, Schirrmeier<br />

H, et al. Novel orthobunyavirus in cattle, Europe, 2011. Emerg Infect Dis<br />

(epub).<br />

Table 1: Detection limits <strong>of</strong> the three real-time SBV PCR methods<br />

Test method<br />

Detection limit (TCID50/ml)<br />

Undiluted semen Diluted semen<br />

Straws<br />

L-PCR 10 1.9 10 0.9<br />

S3-PCR 10 -0.1 10 -1.1<br />

OBV-S PCR 10 -0.1 10 -0.1<br />

Results showed that inhibitory effects were recorded for undiluted<br />

(fresh) semen, while no inhibitory effects were recorded by PCR<br />

when these semen samples were 1:3 pre-diluted. Ct-values <strong>of</strong><br />

the 1:3 diluted fresh semen samples and the diluted semen


S3 - P - 20<br />

INTERFERON-GAMMA ASSAY TO DIAGNOSE Mycobacterium bovis INFECTION IN PIGS<br />

Michele Pesciaroli 1 , Piera Mazzone 2 , Cinzia Marianelli 1 , Sara Corneli 2 , Miriam Russo 3 , Vincenzo Aronica 3 , Michele<br />

Fiasconaro 3 , Massimo Biagetti 2 , Marcella Ciullo 2 , Monica Cagiola 2 , Paolo Pasquali 1 and Vincenzo Di Marco 3<br />

1 Istituto Superiore di Sanità, Dipartimento di Sanità Pubblica Veterinaria e Sicurezza Alimentare, Italy,<br />

2 Istituto Zoopr<strong>of</strong>ilattico Sperimentale dell’Umbria e delle Marche, Italy, 3 Istituto Zoopr<strong>of</strong>ilattico Sperimentale della Sicilia,Italy<br />

Mycobacterium bovis, pig, γ-IFN test<br />

Introduction<br />

Different species <strong>of</strong> animals are susceptible to Mycobacterium<br />

bovis (MB), the causative agent <strong>of</strong> bovine tuberculosis (bTB) and<br />

may represent both "reservoir" <strong>of</strong> infection or "spillover host".<br />

Swine, until a few years ago, was considered only a spillover host<br />

(1). In areas where MB is still present in cattle which share the<br />

pasture with the pig population, the marginal role attributed to the<br />

pig in the maintenance <strong>of</strong> infection <strong>of</strong> MB is strongly questioned.<br />

When high rates <strong>of</strong> infection are observed also in pigs, this<br />

species may play the role <strong>of</strong> true "reservoir" (2). The diagnostic<br />

procedures used to identify in vivo MB infected cattle, including<br />

skin test (IDT) and γ-interferon (γ-IFN) test, are also adopted in<br />

other animal species (3,4) but their application in pigs has not<br />

been completely evaluated. The aim <strong>of</strong> this study was to<br />

calculate the sensitivity <strong>of</strong> the γ-IFN test in pigs infected with MB,<br />

compared with the results <strong>of</strong> post-mortem diagnostic tests<br />

(pathological examination and culture).<br />

Materials & methods<br />

Animals: The Black Sicilian Pig is a traditional Italian breed native<br />

<strong>of</strong> the Nebrodi Park (Sicily). These pigs are usually reared in freerange<br />

farms and share the pasture with cattle. In this area the<br />

prevalence <strong>of</strong> TB in cattle ranges from 5.36% to 8.58% (data<br />

2010 Italian Ministry <strong>of</strong> Health). For the study, 100 Blacks Sicilian<br />

Pigs, coming from farms with bTB positive cases (2), aging from<br />

12 to 24 months, were randomly sampled at the abattoir <strong>of</strong> Mirto<br />

(Sicily). In addition, 15 healthy pigs from tuberculosis-free herds<br />

were sampled at the same abattoir and used to set up the<br />

method and as negative controls.<br />

Protocol and dose response curves <strong>of</strong> IFN-γ: Samples <strong>of</strong> whole<br />

blood <strong>of</strong> the 15 healthy pigs were stimulated with 5μl/ml <strong>of</strong><br />

Pokeweed mitogen (PWM) (Sigma-Aldrich, MS, USA) and 5μl/ml<br />

<strong>of</strong> "Phosphate Buffered Saline" (0.01M PBS, pH 7.2). The<br />

production <strong>of</strong> γ-IFN by lymphocytes was evaluated using a<br />

sandwich ELISA (Porcine IFN-gamma Quantikine ELISA Kit,<br />

R&D Systems, Mn, USA) on the supernatant collected after 24<br />

hours <strong>of</strong> incubation at 37 °C at 5% CO 2 . The stimulation with PBS<br />

did not induce any production <strong>of</strong> IFN-γ; on the contrary, the blood<br />

samples stimulated with PWM, showed an average production <strong>of</strong><br />

γ-IFN <strong>of</strong> 2633 µg/ml, with a standard deviation <strong>of</strong> 688.2 µg/ml.<br />

The samples stimulated with PWM, with a value below 1250<br />

µg/ml (calculated as mean - 2 SD), were excluded from the<br />

analysis because considered unresponsive.<br />

γ-IFN test: Heparinized blood samples, taken at the<br />

slaughterhouse, were dispensed in aliquots <strong>of</strong> 1.5 ml in 24-well<br />

plates (Costar, Corning Incorporated, Corning, NY, USA). The<br />

lymphocyte stimulation was performed in duplicate: PBS, for the<br />

evaluation <strong>of</strong> the basal value <strong>of</strong> γ-IFN (N); PWM (5 μl/ml); bovine<br />

PPD (10μg/ml); avian PPD (10μg/ml). The PPDs (purified protein<br />

derivatives), used in the experiment, were both produced by the<br />

Istituto Zoopr<strong>of</strong>ilattico dell’Umbria e delle Marche (IZSUM),<br />

according to the <strong>of</strong>ficial protocols (EC Regulation N o 1226/2002).<br />

The values obtained from each sample, expressed in units <strong>of</strong><br />

Optical Density (OD), were interpreted as reported below.<br />

Table 1. Possible outcomes following lymphocytes stimulation<br />

with avian and bovine PPDs (OD 450 nm )<br />

AOD & BOD < 2N<br />

Negative<br />

AOD or BOD > 2N<br />

AOD 2 N Positive for M. avium<br />

BOD 2 N Positive for M. bovis<br />

BOD/AOD 0,9 Positive for M. avium<br />

AOD & BOD > 2N BOD/AOD 1,1 Positive for M. bovis<br />

0.9 BOD/AOD 1,1 Doubt<br />

N = baseline value (value obtained with PBS only)<br />

AOD = value obtained after stimulation with avian PPD<br />

BOD = value obtained after stimulation with bovine PPD<br />

Pathological examination (EAP): The macroscopic examination<br />

was conducted at the slaughterhouse in accordance with<br />

conventional inspection methods. Samples <strong>of</strong> lymph nodes (LN)<br />

from the head (mandibular, retropharyngeal, parotid LN), the<br />

thorax (bronchial LN), and the abdomen (hepatic, gastric,<br />

intestinal LN) were also collected from all the pigs.<br />

Microbiology: Bacterial culture for MB was performed according<br />

to the Italian Ministerial Decree (DM 592/1995) on each lymph<br />

node. Suspect colonies, testing positive for acid fast staining,<br />

were further identified through bio-molecular techniques (3).<br />

Results<br />

Of the 100 pigs involved in the study, 2 subjects did not respond<br />

to stimulation with PWM; these animals were excluded from the<br />

final evaluation. EAP and culture identified respectively 26<br />

(26,5% ) and 19 (19,4%) positive pigs . γ–IFN assay identified 22<br />

positive pigs among the 26 with tubercular lesions and 15 among<br />

the 19 with positive bacterial culture (Table 2). The sensitivity <strong>of</strong><br />

the test, assuming as gold standard the traditional tests, resulted<br />

84,6% (95% CI 52.6-100%) and 78,9% (95% CI 44.8-100%)<br />

respectively.<br />

Table 2: Comparison between the results obtained in γ-IFN test,<br />

EAP and culture for M. bovis. P: Positive N: Negative.<br />

Lesion<br />

Culture for M. bovis<br />

P N P N<br />

P 22 4 15 11<br />

γ- IFN<br />

N 4 68 4 68<br />

Total 26 72 19 79<br />

Discussion & conclusions<br />

Our results demonstrate the circulation, in the Nebrodi Park, <strong>of</strong><br />

MB also in the swine population that inhabits this area (2),<br />

supporting the hypothesis <strong>of</strong> an epidemiological role <strong>of</strong> the Black<br />

Sicilian pig as a significant reservoir <strong>of</strong> infection. Therefore, the<br />

implementation <strong>of</strong> future control strategies also in this species is<br />

needed. In this perspective, γ-IFN test could represent a good<br />

tool for intra vitam diagnosis <strong>of</strong> bTB in pigs. The study results<br />

show, in general, an acceptable level <strong>of</strong> agreement between<br />

traditional diagnostic tests and γ-IFN test. In particular, as shown<br />

in Table 2, there is a better agreement with the presence <strong>of</strong><br />

tubercular lesions rather than with the MB isolation. This feature<br />

can be explained with a generally low sensitivity <strong>of</strong> the cultural<br />

test (4). Animals with positive reaction at γ-IFN test showing<br />

neither macroscopic lesions nor MB infections could be assumed<br />

as false positive. However, they could also be ascribed to the<br />

capacity <strong>of</strong> this immunoassay to identify recently infected<br />

subjects with still unapparent lesions at least during the routine<br />

post-mortem inspection. These preliminary results encourage us<br />

to plan future research to optimize γ-IFN test performance in<br />

swine. This study was founded by the Research Project <strong>of</strong> the<br />

Seventh Framework Program “Strategies for the eradication <strong>of</strong><br />

bovine tuberculosis” (Contract Number 212414 – Acronyms TB-<br />

STEP).<br />

References<br />

1. O’Reilly L. M., Daborn C. J., (1995) The epidemiology <strong>of</strong> Mycobacterium<br />

bovis infections in animals and man: a review. Tubercle and lung disease.<br />

The <strong>of</strong>ficial journal <strong>of</strong> the International Union against Tuberculosis and<br />

Lung Disease, 76 Suppl. 1: 1-46.<br />

2. V. Di Marco, P. Mazzone, MT. Capucchio, MB. Boniotti, V. Aronica, M.<br />

Russo, M. Fiasconaro, N. Cifani, S. Corneli, E. Biasibetti, M. Biagetti, ML.<br />

Pacciarini, M. Cagiola, P. Pasquali, and C. Marianelli. “Epidemiological<br />

Significance <strong>of</strong> the Domestic Black Pig (Sus scr<strong>of</strong>a) in Maintenance <strong>of</strong><br />

Bovine Tuberculosis in Sicily” J Clin Microbiol. <strong>2012</strong> Apr;50(4):1209-18.<br />

Epub <strong>2012</strong> Feb 8.<br />

3. OIE Manual <strong>of</strong> Diagnostic Tests and Vaccines for Terrestrial Animals<br />

2010 Chapter 2.4.7. Bovine tuberculosis (Version adopted in May 2009)<br />

4. Boadella M., Lyashchenko K., Greenwald R., Esfandiari J., Jaroso R.,<br />

Carta T., Garrido J. M., Vicente J., de la Fuente J., Gortázar C. (2011)<br />

Serologic tests for detecting antibodies against Mycobacterium bovis and<br />

Mycobacterium avium subspecies paratuberculosis in Eurasian wild boar<br />

(Sus scr<strong>of</strong>a scr<strong>of</strong>a). J Vet Diagn Invest. 23 (1): 77-83


S3 - P - 21<br />

USE OF EXPERIMENTAL JOHNIN IN THE GAMMA-INTERFERON TEST<br />

IN CATTLE INFECTED BY Mycobacterium avium subsp. paratuberculosis: PRELIMINARY DATA<br />

Piera Mazzone 1 , Nicoletta Vitale 2 , Matteo Ricchi 3 , Sara Corneli 1 , Piermario Mangili 1 , Paola Papa 1 , Massimo<br />

Biagetti 1 , Carla Sebastiani 1 , Angela Caporali 1 , Alex Raber 4 , Giovanni Pezzotti 1 , Norma Arrigoni 3 , Monica Cagiola 1<br />

1<br />

Istituto Zoopr<strong>of</strong>ilattico Sperimentale dell’Umbria e delle Marche, Italy<br />

2<br />

Istituto Zoopr<strong>of</strong>ilattico Sperimentale Piemonte, Liguria e Valle d’Aosta,Italy<br />

3<br />

Centro di<br />

Referenza Nazionale per la Paratubercolosi - Istituto Zoopr<strong>of</strong>ilattico Sperimentale della Lombardia ed Emilia Romagna,Italy 4 Prionics AG Switzerland<br />

Paratuberculosis, γ-IFN test, Johnin<br />

Introduction<br />

The diagnosis <strong>of</strong> Paratuberculosis (PTB), due to Mycobacterium<br />

avium subsp. paratuberculosis (MAP), is usually based on<br />

serology and fecal culture, detecting advanced stages <strong>of</strong><br />

infection. The Gamma Interferon (γ-IFN) test, used for antemortem<br />

diagnosis <strong>of</strong> bovine Tuberculosis (bTB), due to<br />

Mycobacterium bovis (MB), together with Skin Test (IDT) (1),<br />

may be used as additional test also for PTB diagnosis. Moreover,<br />

the development <strong>of</strong> an immunological test that, at the same time,<br />

may distinguish between animals infected with MB or MAP and<br />

enables an early diagnosis <strong>of</strong> PTB, could support the control <strong>of</strong><br />

both diseases. The γ-IFN test detects the amount <strong>of</strong> cytokine<br />

produced by T lymphocytes <strong>of</strong> infected animals, in response to a<br />

stimulation carried out with the traditional bovine and avian<br />

purified protein derivatives, respectively extracted by MB (PPDB)<br />

and by M. avium (PPDA). Similarly, the Johnin PPD (PPDJ) is<br />

obtained from a culture <strong>of</strong> MAP (2). In this preliminary study, in<br />

order to achieve an early diagnosis <strong>of</strong> PTB, 3 new PPDJs were<br />

produced, used in the γ-IFN test and compared to classic PPDs,<br />

avoiding false positive reactions for bTB.<br />

Materials & methods<br />

Johnin production: 20 Italian MAP strains, candidates to be used<br />

for production <strong>of</strong> PPDJs, were genotyped by amplification <strong>of</strong> Mini<br />

and Microsatellite loci (3). Two strains were selected: the one<br />

with the most and the one with the least frequent genetic pr<strong>of</strong>ile<br />

observed in Italy, identified as Strain A and Strain B respectively.<br />

The strain ATCC 19698 (Strain C) was used for the third batch <strong>of</strong><br />

Johnin. The three MAP strains were cultured for 4 months at<br />

37°C in Watson-Reid modified broth, and then the bottles were<br />

autoclaved at 100°C for 3 hours. Cells were removed and the<br />

proteins extracted by precipitation with trichloroacetic acid. The<br />

precipitate was then washed and dissolved in phosphate<br />

phenolate buffer and glycerine, with a final protein content <strong>of</strong> 1<br />

mg/ml, as required for PPDB by the Decree <strong>of</strong> the Italian Ministry<br />

<strong>of</strong> Health June, 26 th 1981 and Regulation (EC) N o . 1226/2002.<br />

Field trials: 24 IDT tuberculosis negative bovines from 3 bTB<br />

<strong>of</strong>ficially free (OF) dairy herds were selected. In these herds,<br />

clinical cases <strong>of</strong> PTB had been reported. The animals were<br />

subjected in parallel to ELISA test for PTB (IDVet), PCR (4) and<br />

culture (2) for MAP from faeces. Animals with positive results in<br />

at least one <strong>of</strong> the tests were considered positive for PTB. Of the<br />

24 animals, 16 were positive to at least one <strong>of</strong> the tests and 8<br />

were negative for all.<br />

In vitro stimulation and γ-IFN test: The heparinized blood<br />

samples <strong>of</strong> the 24 bovines were dispensed in aliquots <strong>of</strong> 1 ml and<br />

stimulated respectively with: "Phosphate Buffered Saline" (PBS<br />

0.01 M, pH 7.2), considered as blank; 30 μg <strong>of</strong> PPDB and 30 μg<br />

<strong>of</strong> PPDA (AgriQuality Australia Pty Ltd, Victoria, Australia); 10 μg<br />

<strong>of</strong> Italian PPDB and 10 μg <strong>of</strong> Italian PPDA; three experimental<br />

PPDJ (strains A, B, C) with three different dilutions (1:5, 1:10 and<br />

1:15). The evaluation <strong>of</strong> γ-IFN was performed with the Elisa kit<br />

BOVIGAM ® in the plasma collected after 24 hours <strong>of</strong> incubation.<br />

The samples have been considered positive when the Optical<br />

Density value (OD450 nm) was, at least, twice the OD <strong>of</strong> the<br />

blank. To evaluate the "dilution factor" <strong>of</strong> Johnin (1:5, 1:10; 1:15)<br />

and the "strain factor" (A, B, C), a 2-way analysis <strong>of</strong> variance<br />

using the procedure Proc GLM <strong>of</strong> SAS ®<br />

v9.2 s<strong>of</strong>tware was<br />

conducted. The relative specificity <strong>of</strong> PPDB and PPDJ in the<br />

diagnosis <strong>of</strong> bTB (bTB OF farms) was calculated using the IDT<br />

as Gold Standard (GS). The relative sensitivity <strong>of</strong> PPDJ in the<br />

diagnosis <strong>of</strong> PTB was calculated using as GS positivity in ELISA<br />

and/or PCR and/or Bacteriological test.<br />

Results<br />

The results <strong>of</strong> the lymphocyte stimulation with the different PPDs<br />

are shown in Figure 1. The analysis <strong>of</strong> variance showed<br />

statistically significant differences between the mean OD 450 <strong>of</strong><br />

PPDJ for both the "dilution factor" <strong>of</strong> Johnin (F test = 28.44,<br />

p


S3 – P - 22<br />

COMPARISON OF THE PERFORMANCE OF FIVE DIFFERENT IMMUNOASSAYS TO DETECT SPECIFIC<br />

ANTIBODIES AGAINST EMERGING ATYPICAL BOVINE PESTIVIRUS<br />

Magdalena Larska 1,2 , Lihong Liu 3 , Mirosław P. Polak 1 , Stefan Alenius 2 , Åse Uttenthal 5<br />

1 National Veterinary Research Institute (NVRI), Virology Department, Puławy, Poland, 2 Department <strong>of</strong> Clinical Sciences, Swedish University <strong>of</strong><br />

Agricultural Sciences (SLU), Uppsala, Sweden; 3 Department <strong>of</strong> Virology, Immunology and Parasitology (VIP), National Veterinary Institute (SVA),<br />

Uppsala, Sweden; 5 National Veterinary Institute, Technical University <strong>of</strong> Denmark, Lindholm, Denmark<br />

Atypical bovine pestivirus, antibodies, ELISA, Microsphere-based immunoassay (MIA), diagnosis<br />

Introduction<br />

Atypical bovine pestiviruses are a group <strong>of</strong> viruses which together<br />

with two bovine viral diarrhoea virus species (BVDV-1 and<br />

BVDV-2), border disease virus (BDV) and classical swine fever<br />

virus (CSFV) comprise the Pestivirus genus <strong>of</strong> the Flaviviridae<br />

family. The group was previously referred to as Hobi-like<br />

pestiviruses from the precursor strain isolated from Brazilian<br />

foetal bovine serum (FBS) (2). Increasing evidence <strong>of</strong><br />

intercontinental spread <strong>of</strong> the atypical pestivirus raised the<br />

discussion over the emergence <strong>of</strong> the new BVDV species and<br />

prompts the search for the tools supporting testing and control<br />

programs. Serological test has not been validated to detect<br />

specific antibodies against those pestiviruses yet.<br />

0.0001) with ρ values between 0.82 for b-ELISA-2 and 0.92 for i-<br />

ELISA-2.<br />

Materials & methods<br />

The study was conducted using 140 sera from calves<br />

experimentally inoculated with Asian atypical pestivirus<br />

Th/04_KhonKaen strain (Group A); BVDV-1a strain Horton 916<br />

(Group B), with a mixture <strong>of</strong> both viruses (Group C) and<br />

uninfected controls (1). Serum samples were collected from all<br />

animals on 0, 7, 14, 21, 28, 35, 42 days post inoculation (dpi).<br />

Five serological tests including two commercial indirect BVDV<br />

ELISAs (i-ELISA-1 and 2), two blocking BVDV ELISAs (b-ELISA-<br />

1 and 2) and newly developed microsphere immunoassay (MIA)<br />

(3) using recombinant E rns protein <strong>of</strong> Th/04_KhonKaen pestivirus<br />

were evaluated for their robustness, sensitivity, specificity and<br />

receiver operating characteristic (ROC) using virus neutralization<br />

test (VNT) as ’gold’ standard.<br />

Results<br />

According to VNT using homologous to inoculum viruses the<br />

animals from groups A and B started to seroconvert against<br />

homologous viruses at 14 dpi, while Group C seroconverted<br />

independently to both viruses before 14 dpi. The earliest<br />

detection <strong>of</strong> antibodies in the Group A was observed at 14 dpi in<br />

the i-ELISA-1, b-ELISA-2, MIA. Using i-ELISA-2 the antibodies<br />

were detected first in two animals from Group A at 21 dpi and in<br />

all animals from 28 dpi. Detection <strong>of</strong> specific antibodies in Group<br />

A was the most delayed to the end <strong>of</strong> experiment in b-ELISA-2.<br />

100% sensitivity in detecting Th/04_KhonKaen virus specific<br />

antibodies in Group A was obtained by b-ELISA-2 and MIA, while<br />

their specificities were 82% and 91%, respectively. Other assays<br />

were characterised by 100% specificity, however the sensitivity<br />

varied from only 25% for b-ELISA-2 and BVDV-1 VNT to 83% for<br />

i-ELISA-1 when testing calves from Group A. Most assays<br />

detected 100% <strong>of</strong> seropositive animals from Group B which were<br />

categorised as positive in BVDV-1 VNT, except for b-ELISA-1<br />

which had only 72% sensitivity. The specificity <strong>of</strong> the tests was<br />

lower in respect to VNT test, possibly due to the lower sensitivity<br />

<strong>of</strong> the latter. The AUC for the methods detecting atypical<br />

pestivirus antibodies were the highest reaching 0.93 for MIA and<br />

the lowest (0.62) for b-ELISA-1 and BVDV-1 VNT. Evaluation <strong>of</strong><br />

positive-negative cut-<strong>of</strong>f values by ROC analysis showed that<br />

optimizing the recommended by manufacturer or SOP values<br />

could increase senstitivities and specificities <strong>of</strong> some<br />

immunoassays. Two tests: MIA and b-ELISA-2 showed more<br />

flexible cut-<strong>of</strong>fs which allowed the values to fluctuate within a<br />

wider range without influencing their performance.<br />

The values <strong>of</strong> all ELISA tests were significantly correlated with<br />

the neutralizing titres against atypical pestivirus and BVDV-1 (P <<br />

0.0001) when tested by the Spearman rank correlation<br />

coefficients (ρ). Anti-Th/04_KhonKaen antibody titres showed the<br />

comparable correlations with all ELISAs tested, with the highest<br />

correlation to b-ELISA-2 (ρ= 0.84). The lowest correlation was<br />

observed between Th/04_KhonKaen antibody titres and VNT<br />

using BVDV-1 strain (ρ= 0.65). The correlations between BVDV-1<br />

antibody titres and ELISA results were also significant (P <<br />

Figure 1: Comparison <strong>of</strong> the sensitivity (Se) and specificity (Sp)<br />

(A) and AUC (area under ROC curve) values (B) <strong>of</strong> all<br />

immunoassays.<br />

Discussion & conclusions<br />

The results have shown that all the tested immunoassays were<br />

suitable to detect antibodies raised in calves inoculated with<br />

atypical pestivirus, however only some could be used in the early<br />

detection <strong>of</strong> the seroconversion. Two ELISAs (i-ELISA-1 and b-<br />

ELISA-2) and novel MIA were considered to be superior (even in<br />

respect to BVDV-1 VNT) for the routine detection <strong>of</strong> atypical<br />

bovine pestivirus antibodies in cattle. The analysis <strong>of</strong> optimal cut<strong>of</strong>f<br />

values suggested that cautious result interpretation using the<br />

recommended cut-<strong>of</strong>f values diminish the possibility <strong>of</strong> obtaining<br />

false results. Here the focus is on early detection <strong>of</strong> antibodies,<br />

as the immune system will initially produce IgM antibodies there<br />

is a natural preference <strong>of</strong> assays that are independent <strong>of</strong> IgG.<br />

Therefore, despite the conservative nature <strong>of</strong> NS3 protein,<br />

blocking b-ELISA-1 performed with the lowest sensitivity and the<br />

seroconversion appeared at the latest. Development <strong>of</strong> MIA<br />

using microspheres conjugated with mAbs against atypical<br />

pestivirus E rns protein resulted in high sensitivity and specificity <strong>of</strong><br />

detection <strong>of</strong> antibodies against homologous virus comparable to<br />

VNT, which however did not promise much possibility for the<br />

pestivirus specific discrimination as it detected BVDV-1<br />

antibodies equally efficiently.<br />

References<br />

1. Larska et al., <strong>2012</strong>. Kinetics <strong>of</strong> single and dual infection <strong>of</strong> calves with an<br />

Asian atypical bovine pestivirus and a highly virulent strain <strong>of</strong> bovine viral<br />

diarrhoea virus 1.Comp Immunol Microbiol Infec Dis. {Epub ahead <strong>of</strong> print]<br />

2. Schirrmeier et al., 2004. Genetic and antigenic characterization <strong>of</strong> an<br />

atypical pestivirus isolate, a putative member <strong>of</strong> a novel pestivirus species.<br />

J Gen Virol. 85, 3647–3652<br />

3. Xia et al., 2010. A microsphere-based immunoassay for rapid and<br />

sensitive detection <strong>of</strong> bovine viral diarrhoea virus antibodies. J Virol<br />

Methods 168, 18-21.


S3 - P - 23<br />

SCHMALLENBERG VIRUS (SBV) IN POLAND – PRELIMINARY RESULTS<br />

Magdalena Larska, Mirosław P. Polak, Jan F. Żmudziński<br />

National Veterinary Research Institute (NVRI), Virology Department, Puławy, Poland<br />

Schmallenberg virus, testing, Culicoides<br />

Introduction<br />

SBV was first detected in Germany by metagenomics analysis <strong>of</strong><br />

samples collected from sick cows from the city <strong>of</strong> Schmallenberg<br />

in the autumn 2011 (1). Until now over 4000 cases were<br />

described in Northern Europe. Because Poland is a neighbouring<br />

country to the area covered by SBV epidemic, the risk <strong>of</strong> further<br />

spread to our territory is possible. European SBV is a reassortant<br />

<strong>of</strong> Sathuperi and Shamonda viruses belonging to<br />

Orthobunyavirus <strong>of</strong> the family Bunyaviridae which emerged<br />

around a decade ago in Japan (2). The source <strong>of</strong> SBV infection in<br />

Europe is not know, however the virus is known to spread by<br />

Culicoides sp. biting midges, similarly to bluetongue virus (BTV),<br />

what may suggest climate change or involvement <strong>of</strong> redistribution<br />

<strong>of</strong> vectors as a possible cause.<br />

Materials & methods<br />

Tissue and blood samples from fifteen animals from two cattle<br />

and one goat herds from different parts <strong>of</strong> Poland were submitted<br />

to NVRI by district <strong>of</strong>ficial veterinarians under the suspicion <strong>of</strong><br />

Schmallenberg virus infection. The clinical signs included<br />

abortions (no malformation observed), fever, depression and in<br />

older cows respiratory disorders with sudden death. Mortality in<br />

older cows was later confirmed to occur due to antibiotic resistant<br />

Mannheimia haemolytica infection. Additionally, 36 pools <strong>of</strong><br />

midges including C. obsoletus, C. punctatus, C. chiropterus and<br />

C. pulicaris from the border zone with Germany and from northeastern<br />

part <strong>of</strong> Poland collected in September/October 2011<br />

were tested. Midge pools were homogenised in Ribolyzer tubes<br />

(Hybaid, UK) containing 0.6g beads with TRI Reagent (Sigma<br />

Adrich) for 80 sec. at 5m/sec. speed. Viral RNA was extracted<br />

using TRI Reagent. Real time RT-PCR was performed according<br />

to Friedrich-Loeffler Institut (FLI) protocol using primers and<br />

probes specific to S and L segment <strong>of</strong> SBV genome with β-actin<br />

as internal control. AgPath-ID One-Step RT-PCR Reagents<br />

(Ambion-Applied Biosystems) were used and the reaction was<br />

performed in StepOnePlus machine (Applied Biosystems). SBV<br />

RNA standard used as positive control and to determine<br />

sensitivity/specificity was acquired from FLI.<br />

Figure 2: Standard curve (correlation <strong>of</strong> Ct value and log dilution<br />

<strong>of</strong> SBV RNA standard)<br />

Discussion & conclusions<br />

More samples have to be tested to verify the possibility <strong>of</strong> SBV<br />

introduction into Polish herds. However, Polish population <strong>of</strong><br />

sheep and goats is scarce (approx. 250 000 and 130 000,<br />

respectively), the disease may already be present in the country,<br />

but SBV is not detected because <strong>of</strong> its mostly asymptomatic<br />

course in cattle. More research including some seroprevalence<br />

studies is planned.<br />

References<br />

1. H<strong>of</strong>fmann et al., <strong>2012</strong>. Novel orthobunyavirus in Cattle, Europe, 2011.<br />

Emerg Infect Dis. 18, :469-7<br />

2. Yanase et al., <strong>2012</strong>. Genetic reassortment between Sathuperi and<br />

Shamonda viruses <strong>of</strong> the genus Orthobunyavirus in nature: implications for<br />

their genetic relationship to Schmallenberg virus. Arch Virol. [Epub ahead<br />

<strong>of</strong> print]<br />

Results<br />

All samples were negative for the presence <strong>of</strong> SBV genetic<br />

material. In all animal and insect samples amplification <strong>of</strong> β-actin<br />

was positive with Ct values ranging from 25 to 34. Specificity <strong>of</strong><br />

PCR products was confirmed by sequencing. The specificity <strong>of</strong><br />

the assays was 100%, while sensitivity was comparable to FLI<br />

(detection limit at 10 -7<br />

SBV RNA). The efficiency determined<br />

using the standard RNA was approx. 98% with the mean slope <strong>of</strong><br />

3.38.<br />

Figure 1: Amplification plots <strong>of</strong> SBV RNA standard with segment<br />

S specific primers and TaqMan probe.


S3 - P - 24<br />

DEVELOPMENT OF A VIRUS NEUTRALISATION TEST TO DETECT ANTIBODIES AGAINST<br />

SCHMALLENBERG VIRUS AND FIRST RESULTS IN SUSPECT AND INFECTED HERDS<br />

W.L.A. Loeffen 1 , S. Quak 1 , E. de Boer-Luijtze 1 , W.H.M. van der Poel 1 , R. Bouwstra 2 , R. Maas 1<br />

1<br />

Central Veterinary Institute, <strong>of</strong> Wageningen University and Research Centre (CVI-Lelystad), Department <strong>of</strong> Virology, Lelystad, The Netherland.<br />

2<br />

Central Veterinary Institute, <strong>of</strong> Wageningen University and Research Centre (CVI-Lelystad), Department <strong>of</strong> Epidemiology, Crisis Management and<br />

Diagnostics, Lelystad, The Netherland.<br />

Schmallenberg, VNT, sensitivity, specificity<br />

Introduction<br />

On the 18th <strong>of</strong> November 2011, a new Orthobunya virus was<br />

reported to be found in Germany (3). This virus was closely<br />

related to Shamonda-, Aino- and Akabane-virus, which all belong<br />

to the Simbu serogroup <strong>of</strong> the genus Orthobunyavirus, family<br />

Bunyaviridae (1). The virus, provisionally called Schmallenberg<br />

virus (SBV), has since been associated with clinical signs <strong>of</strong><br />

decreased milk production, watery diarrhoea, fever and<br />

congenital malformations (3,4,5).<br />

To determine the prevalence <strong>of</strong> the disease and carry out<br />

epidemiological studies, there was a need for a reliable and<br />

robust serological assay. A virus neutralisation test (VNT) was<br />

developed, as this also allows for a (semi)quantitative detection<br />

<strong>of</strong> antibodies against the virus. An initial validation (specificity,<br />

sensitivity, repeatability, reproducibility, and robustness) was<br />

carried out and the test was used in suspect (congenital<br />

malformation, no positive RT-PCR) and infected herds (positive<br />

RT-PCR).<br />

Materials & methods<br />

The VNT was carried out in flat bottomed 96 well micro titre<br />

plates. Serum samples were inactivated for 30 min at 56°C and<br />

from a starting dilution <strong>of</strong> 1:4, two-fold dilutions were made.<br />

Subsequently 500 TCID 50 <strong>of</strong> virus was added to each well. Serum<br />

and virus were pre-incubated at 37°C for 1 to 2 hours to allow<br />

neutralisation <strong>of</strong> the virus. Thereafter, 20,000 cells (VERO cells)<br />

per well were added. Plates were then incubated for 5 days at<br />

37°C and under 5% CO2.<br />

After 5 days, the plates were stained with amido black and read<br />

macroscopically. Wells with 25-100% staining were considered to<br />

have no or limited CPE only. Wells with less than 25% staining<br />

were considered to have extensive or full CPE. In each dilution<br />

series the last well with no or limited CPE was identified and the<br />

sample was assigned a titre that was the reciprocal <strong>of</strong> the dilution<br />

in that well. Control samples were included and a back titration <strong>of</strong><br />

the virus was carried out for each test run.<br />

Initially, 351 blood samples (160 sheep, 191 cattle) from noninfected<br />

animals were tested in the final version <strong>of</strong> the assay.<br />

These samples originated from the Netherlands, from the period<br />

<strong>of</strong> 2000-2010, before the introduction <strong>of</strong> the Schmallenberg virus.<br />

They were used to set the initial cut-<strong>of</strong>f value in the assay.<br />

Subsequently, 366 samples from suspect herds (being herds in<br />

which congenital malformations were observed, with no<br />

confirmation in the RT-PCR), and infected herds (herds where<br />

the infection was confirmed by RT-PCR).<br />

Results<br />

In serum samples from non-infected sheep, a higher background<br />

was seen than in sera from non-infected cattle (figure 1).<br />

Figure 1: Distribution <strong>of</strong> titres in non-infected sheep (n=160) and<br />

cattle (n=191).<br />

Based on testing 160 sheep and 191 cows, the cut-<strong>of</strong>f values<br />

were set at a titre <strong>of</strong> 16 for sheep and 8 for cattle. Given this cut<strong>of</strong>f<br />

value, a specificity <strong>of</strong> 99.4% in sheep and 99.5% in cattle was<br />

estimated.<br />

Ninety-two percent <strong>of</strong> the cattle and 94% <strong>of</strong> the sheep from<br />

suspect and infected herds scored positive in the VNT. In sheep<br />

no titres <strong>of</strong> 8 and in cattle no titres <strong>of</strong> 4 were observed, thus<br />

clearly separating the peak <strong>of</strong> the positive samples from the peak<br />

<strong>of</strong> the negative samples (figure 2). This again corresponds to a<br />

cut-<strong>of</strong>f value <strong>of</strong> 16 and 8 respectively from sheep and cattle, as<br />

determined already by testing serum samples from non-infected<br />

animals. Sensitivity <strong>of</strong> the assay is therefore at least 92% and<br />

94% in cattle and sheep respectively, assuming the worst case<br />

scenario that all were seropositive in reality. However, the clear<br />

separation <strong>of</strong> the negative and positive peak suggests that the<br />

sensitivity for both species is close to 100%.<br />

Figure 2: Distribution <strong>of</strong> titres in goats, sheep and cattle from<br />

suspect (in utero malformations, RT-PCR negative), and infected<br />

herds (infection confirmed by RT-PCR).<br />

Discussion & conclusions<br />

A very robust VNT was developed with a very high specificity and<br />

sensitivity, both close to 100%, in sheep and cattle. The assay<br />

was shown to be highly repeatable and reproducible (results not<br />

shown). The VNT is easy to perform and can be read<br />

macroscopically, as a result <strong>of</strong> the amido black staining. With the<br />

current, optimized protocol, this staining results most <strong>of</strong> the times<br />

in a very sharp transition between wells with and without CPE.<br />

This makes it easy and very fast to read the plates, with a high<br />

reliability and reproducibility.<br />

In suspect and infected herds more than 90% <strong>of</strong> the sheep and<br />

cattle tested seropositive, suggesting that within herds with<br />

congenital malformations, the infection is wide spread with a high<br />

within-herd seroprevalence. High seroprevalences, both on a<br />

regional level, but also within herds, with and without obvious<br />

clinical signs, are not uncommon for viruses related to SBV. A<br />

very high overall seroprevalence <strong>of</strong> 70% in cattle was recently<br />

also reported in the Netherlands, using this VNT (2).<br />

References<br />

1. Causey OR, Kemp GE, Causey CE, Lee VH. 1972. Ann Trop Med<br />

Parasitol 66: 357-62.<br />

2. Elbers ARW, Loeffen WLA, Quak S, et al. <strong>2012</strong>. Emerg Infect Dis.<br />

3. H<strong>of</strong>fmann B, Scheuch M, Höper D, et al. <strong>2012</strong>. Emerg Infect Dis.<br />

4. Muskens J, Smolenaars AJG, Van der Poel WHM, et al. <strong>2012</strong> Tijdschr<br />

Diergeneeskd. 137: 112-5.<br />

5 .Van den Brom R, Luttikholt SJM, Lievaart-Peteron K,et al. <strong>2012</strong><br />

Tijdschr Diergeneeskd 137: 106-11.


S3 - P - 25<br />

VIRAL DIAGNOSIS USING TRANSMISSION ELECTRON MICROSCOPY SCHMALLENBERG VIRUS<br />

David J. Everest and W. A. Cooley<br />

1<br />

Central Animal Health and Veterinary Laboratories Agency-Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK<br />

Most viruses are very small and visualisation by Transmission<br />

Electron Microscopy (TEM) has provided a major contribution in<br />

the discovery and detection <strong>of</strong> viral infections. The AHVLA’s Bioimaging<br />

unit provides a rapid viral diagnostic service, analysing<br />

fresh unfixed scab, lesion, wart and faecal materials by TEM.<br />

Species range from domestic farm animals, to wildlife, marine<br />

mammals and Zoo animals, covering red squirrels to Elephants.<br />

Historically, this form <strong>of</strong> microscopy has been a key application in<br />

the diagnosis and surveillance <strong>of</strong> several wildlife viral infections.<br />

These have included Squirrel pox and adenovirus, both<br />

contributing to the decline <strong>of</strong> the UK Red Squirrel, calicivirus from<br />

Rabbits and Hares, the causative agent <strong>of</strong> rabbit haemorrhagic<br />

disease and pox viruses in various marine mammal species.


S3 - P - 26<br />

DETECTION OF EQUINE FOAMY VIRUS INFECTIONS IN HORSES<br />

Magdalena Materniak, Jacek Kuzmak<br />

National Veterinary Research Institute, Department <strong>of</strong> Biochemistry, Pulawy, Poland<br />

Equine foamy virus, diagnostics, PCR, ELISA<br />

Introduction<br />

Foamy viruses (FVs) are the least known complex retroviruses.<br />

FVs infections are highly prevalent among several animal species<br />

including non-human primates (chimpanzees, gorillas, etc.), cats<br />

and cattle. Another foamy virus was isolated from horses (Equine<br />

foamy virus - EFV) (1). Similarly to other FVs, EFV shows<br />

characteristic ultrastructure and susceptibility to infect large<br />

number <strong>of</strong> cell lines. Furthermore, its molecular analysis exhibits<br />

genome organization typical for other known foamy viruses, but<br />

its sequence shows the highest similarity to bovine foamy virus.<br />

Although the first isolation <strong>of</strong> EFV was reported over ten years<br />

ago, still nothing is known about the prevalence <strong>of</strong> EFV infections<br />

in horses. Therefore we aimed to develop diagnostic tools and<br />

apply them to investigate the EFV infection in horses from<br />

Poland.<br />

References<br />

1. Tobaly-Tapiero J., Bittoun P., Neves M., Guillemin M.-C., Lecellier Ch.-<br />

H., Puvion-Dutilleul F., Gicquel B., Zientara S., Giron M.-L., The H. de,<br />

Saϊb A. (2000). Isolation and characterization <strong>of</strong> an eqiune foamy virus. J.<br />

Virol., 74, 4064-4073.<br />

2. Romen F., Backes P., Materniak M., Sting R., Vahlenkamp T. W., Riebe<br />

R., Pawlita M., Kuzmak J., Löchelt M. (2007). Serological detection<br />

systems for identification <strong>of</strong> cows shedding bovine foamy virus via milk.<br />

Virology, 364, 123-131.<br />

Materials & methods<br />

Blood samples were collected from 175 horses (4 -18 years old),<br />

including common breeds, Hucul ponies and randomly selected<br />

samples from slaughtered horses. Semi-nested PCR was<br />

developed and applied to detect EFV DNA in DNA extracted from<br />

peripheral blood leukocytes (PBLs). Selected primers flanked 275<br />

bp fragment within pol gene, the most conservative EFV region.<br />

GST ELISA with BFV specific Gag-GST fusion protein (2) was<br />

used for detection <strong>of</strong> foamy virus specific antibodies in horse<br />

serum samples. To confirm ELISA results immunoblotting with<br />

antigen prepared from Cf2Th/BFV 100 cells was applied.<br />

Additionally, attempts were made to isolate EFV from peripheral<br />

blood leukocytes <strong>of</strong> PCR positive animals by co-cultivation with<br />

BHK-21 and RK-13 cells.<br />

Results<br />

12.5% <strong>of</strong> samples were positive in semi-nested PCR. Specificity<br />

<strong>of</strong> amplified fragments was confirmed by sequencing and parwise<br />

identity <strong>of</strong> particular sequences ranged from 95.3% to 98.3%,<br />

when compared to EFV reference sequence (GenBank Acc. No.<br />

NC_002201). In contrast, almost 19% <strong>of</strong> the samples showed<br />

seroreactivity to BFV Gag protein. Most <strong>of</strong> PCR positive samples<br />

were detected in Hucul ponies, while only several in samples<br />

randomly collected from slaughtered animals. All attempts to<br />

isolate EFV from PBLs <strong>of</strong> PCR positive horses failed.<br />

Table 1: Summary <strong>of</strong> results.<br />

Origin<br />

PCR (+) ELISA (+)<br />

samples samples<br />

Hucul ponies 15 14 0<br />

Saddle-horses 3 6 0<br />

Slaughtered<br />

horses<br />

TOTAL<br />

4<br />

22<br />

13<br />

33<br />

Co-cultivation (+)<br />

samples<br />

Discussion & conclusions<br />

This study presents the first report on prevalence <strong>of</strong> EFV infection<br />

in horses. The discordance between PCR and serological results<br />

can be due to application <strong>of</strong> BFV specific tests instead <strong>of</strong> EFV<br />

specific serological assays. Therefore further studies are<br />

essential to develop EFV specific serological tests to confirm<br />

obtained results. Nevertheless, our study clearly shows that EFV<br />

infections are present among horses in Poland.<br />

Acknowledgements<br />

The authors would like to thank Dr. Ali Saïb and Dr. Joelle<br />

Tobaly-Tapiero for providing EFV positive DNA and serum<br />

controls.<br />

0<br />

0


Poster presentations<br />

“New techniques in bacteriology,<br />

parasitology and pathology”<br />

(4 th session)


S4 - P - 01<br />

MOLECULAR CHARACTERIZATION OF MYCOBACTERIUM AVIUM SUBSP. PARATUBERCULOSIS<br />

STRAINS IN A NATIONAL PARATUBERCULOSIS CONTROL PROGRAM<br />

Michaela Altmann 1 , Eva Sodoma 1 , Petra Möbius 2 , Michael Dünser 1<br />

1<br />

Institute for Veterinary Disease Control, Austria Agency for Health and Food Safety, Linz, Austria<br />

2<br />

Institute for Molecular Pathogenesis, Friedrich Loeffler Institute, Jena, Germany<br />

Mycobacterium avium subsp. paratuberculosis, RFLP, MIRU, VNTR, paratuberculosis<br />

Introduction<br />

In 2006 Austria started a national paratuberculosis (PTB) control<br />

programme based on government regulation which affects cattle,<br />

sheep, goat and farmed deer. PTB was declared a notifiable<br />

disease, so animals showing clinical signs have to be tested for<br />

MAP at the national reference laboratory. MAP strains isolated<br />

from different herds between 2006 and 2011 and strains from red<br />

deer were selected for molecular characterization.<br />

Materials & methods<br />

165 MAP field strains from 164 clinically diseased ruminants<br />

were analysed by IS900 RFLP with two restriction enzymes<br />

(BstEII, PstI) and MIRU-VNTR at 8 genomic loci (MIRU292,X3;<br />

VNTR25,47,3,7,10,32) according to MÖBIUS et al. (2008) and<br />

THIBAULT et al. (2007). These field strains included 141 MAP<br />

cattle isolates from 73 farms and different breeds (Angus,<br />

Aquitaine Blonde, Austrian Brown Mountain, Fleckvieh<br />

(Simmental), Tyrolean Grey Mountain, Holstein Friesian, Jersey,<br />

Limousin, Murbodner, Piemontese, Red Friesian, Danish<br />

Melkrace), 19 isolates from goat and 4 isolates from red deer.<br />

Results<br />

The combination <strong>of</strong> RFLP and MIRU-VNTR analysis allowed a<br />

differentiation <strong>of</strong> 18 MAP strain types classified AT1 to AT18.<br />

Detailed results and frequency in animals and herds are shown in<br />

Table 1. The occurrence <strong>of</strong> MAP strain types in different cattle<br />

breeds, goats and red deer is listed in Table 2.<br />

Table 1: RFLP and MIRU-VNTR results <strong>of</strong> 165 MAP field strains<br />

(INMV: INRA NOUZILLY MIRU-VNTR)<br />

RFLP INMV<br />

MIRU VNTR<br />

Σ<br />

292,X3 25,47,3,7,10,32 animals<br />

AT1 C1P1 1 4,2 3,3,2,2,2,8 16<br />

AT2 C1P1 2 3,2 3,3,2,2,2,8 59<br />

AT3 C18 2 3,2 3,3,2,2,2,8 18<br />

PnewIII<br />

AT4 C1P1 17 3,1 3,3,2,2,2,8 8<br />

AT5 C18 2 4,2 3,3,2,2,2,8 4<br />

PnewII<br />

AT6 C1 2 4,2 3,3,2,2,2,8 9<br />

PnewV<br />

AT7 C1P7 1 4,2 3,3,2,2,2,8 1<br />

AT8 C1P1 5 4,2 3,3,2,2,1,8 2<br />

AT9 C18 1 4,2 3,3,2,2,2,8 15<br />

PnewIII<br />

AT10 C1P1 new 2,2 5,2,2,2,2,6 1<br />

AT11 CnewI 2 4,2 3,3,2,2,2,8 1<br />

PnewIII<br />

AT12 C18P1 17 3,1 3,3,2,2,2,8 1<br />

AT13 C28 1 4,2 3,3,2,2,2,8 1<br />

PnewI<br />

AT14 C18P1 2 4,2 3,3,2,2,2,8 5<br />

AT15 C1 2 4,2 3,3,2,2,2,8 1<br />

PnewIV<br />

AT16 C1P1 12 2,2 5,2,2,2,2,8 3<br />

AT17 CnewIII 2 4,2 3,3,2,2,2,8 19<br />

PnewIV<br />

AT18 C14<br />

PnewIII<br />

2 4,2 3,3,2,2,2,8 1<br />

Table 2: The occurrence <strong>of</strong> MAP strain types AT1-18<br />

Breed<br />

AT MAP strain type (number)<br />

Angus 2(3)<br />

Aquitaine Blonde 5(2),6(3)<br />

Austrian Braun Mountain 1(1),2(3),6(1)<br />

Fleckvieh 1(2),2(10),3(1),4(5),5(1),8(2)<br />

Austrian Grey Mountain 1(2)<br />

Holstein Friesian 1(1),2(9),5(1),6(3),16(3)<br />

Jersey 2(3),3(3),7(1),15(1)<br />

Limousin 1(5),2(25),3(12),4(3),9(15),11(1)<br />

13(1),14(5),18(1)<br />

Murbodner 3(1),12(1)<br />

Piemontese 1(4)<br />

Red Friesian 2(2),3(1),6(1),<br />

Danish Melkrace 2(1),6(1),10(1)<br />

Goat<br />

Red Deer<br />

18(19)<br />

1(1), 2(3)<br />

Discussion & conclusions<br />

Despite the fact that 18 different MAP types were detected in a<br />

comparatively large and diverse sample selection, 42% <strong>of</strong> all<br />

isolates were classified as MAP type AT2 (RFLP C1-P1; INMV<br />

2). MAP type AT2 was found in 8 <strong>of</strong> 12 PTB affected cattle<br />

breeds and in 2 <strong>of</strong> 4 isolates from red deer and is therefore the<br />

predominating MAP type strain in Austria. 9 new RFLP patterns<br />

and 1 new INMV pr<strong>of</strong>ile could be determined within the 165<br />

analysed field strains. Some unique occurring strains showed<br />

association with certain cattle breeds, goat and red deer (Table<br />

2). This is the first study carried out in Austria providing data<br />

about MAP strain types on a national scale. Additional<br />

information about routes <strong>of</strong> transmission between herds and the<br />

epidemiology <strong>of</strong> PTB in Austria could be achieved by using the<br />

national cattle data base and the results <strong>of</strong> MAP typing.<br />

References<br />

1. MÖBIUS, P., LUYVEN, G., HOTZEL, H., KÖHLER, H. (2008): High<br />

Genetic Diversity among Mycobacterium avium subsp. paratuberculosis<br />

Strains from German Cattle Herds Shown by Combination <strong>of</strong> IS900<br />

Restriction Fragment Length Polymorphism Analysis and Mycobacterial<br />

Interspersed Repetitive Unit Variable Number Tandem Repeat Typing. J.<br />

Clin. Microbiology, 46, 972-980<br />

2. THIBAULT, V., GRAYON, M., BOSCHIROLI, M. L., HUBBANS, C.,<br />

OVERDUIN, P., STEVENSON, K., GUTIERREZ, M.C., SUPPLY, P., BIET,<br />

F. (2007): New Variable Number Tandem Repeat Markers for Typing<br />

Mycobacterium avium subsp. paratuberculosis and M. avium Strains:<br />

Comparison with IS900 and IS1245 Restriction Fragment Length<br />

Polymorphism Typing. J. Clin. Microbiology, 45, 2404-2410


S4 - P - 02<br />

DEVELOPMENT AND VALIDATION OF A REAL-TIME PCR ASSAY FOR THE DETECTION OF<br />

LEPTOSPIRA BORGPETERSENII SEROVAR HARDJO IN URINE AND KIDNEY<br />

Zbigniew Arent 1 , Caroline Frizzell 2 , Colm Gilmore 1 , William Ellis 1<br />

1<br />

OIE Leptospirosis Reference Laboratory, Veterinary Sciences Division, AFBI, Belfast, Northern Ireland<br />

2<br />

Department <strong>of</strong> Veterinary Science, School <strong>of</strong> Agriculture and Food Science, The Queen’s University <strong>of</strong> Belfast<br />

Leptospira, leptospirosis, real-time PCR<br />

Introduction<br />

Leptospira borgpetersenii serovar Hardjo is the most common<br />

cause <strong>of</strong> leptospirosis in cattle throughout the world. Infection <strong>of</strong><br />

cattle with Hardjo can cause abortion, stillbirth, birth <strong>of</strong> weak<br />

calves and decreased milk production (1, 2). Infected animals<br />

can act as lifelong renal carriers without clinical signs <strong>of</strong> the<br />

disease (3) and shed live organisms over a long period <strong>of</strong> time<br />

(4).<br />

There is no satisfactory method available for the rapid<br />

identification <strong>of</strong> carrier animals. Culturing <strong>of</strong> leptospires is the<br />

most definitive test. However this is possible only on fresh<br />

samples where viable leptospires are present. . PCR assays<br />

have the potential to detect both viable and non-viable<br />

leptospires and in recent years, several real-time PCR assays<br />

have been described. Levett et al. (5) developed a real-time<br />

assay using SYBR green chemistry, in which the target was the<br />

gene for the outer-membrane protein LipL32. Expression <strong>of</strong> the<br />

LipL32 gene is a unique virulence factor for pathogenic<br />

leptospires. LipL32 is an outer membrane lipoprotein that is<br />

conserved genetically and immunologically in pathogenic<br />

leptospires. It is one <strong>of</strong> the most abundant proteins in Leptospira.<br />

In this study we developed and validated a TaqMan PCR using<br />

novel primers which target a 200 bp fragment <strong>of</strong> the LipL32 gene.<br />

Materials & methods<br />

Eight animals that had no evidence <strong>of</strong> exposure to serovar Hardjo<br />

were experimentally infected with L. borgpetersenii serovar<br />

Hardjo type Bovis via conjunctival instillation <strong>of</strong> 2.5 x 10 6<br />

organisms on 3 consecutive days. A total <strong>of</strong> 8 urine samples<br />

were collected from each animal prior to and at intervals up to 40<br />

days after infection. In addition , 24 culture negative urine<br />

samples obtained from herds with no evidence <strong>of</strong> leptospirosis<br />

and 72 kidney samples collected post mortem from animals with<br />

serological evidence <strong>of</strong> Leptospira serovar Hardjo exposure were<br />

used in the study. All samples were cultured using Leptospira<br />

semisolid Tween 80/40 medium and biphasic culture medium<br />

with different variants <strong>of</strong> inhibitory substance addition.<br />

DNA was extracted from bacterial cultures and kidneys using the<br />

QIAamp DNA Mini kits (Qiagen) and from urines using the<br />

QIAamp Viral RNA Mini Kit (Qiagen) according to the<br />

manufacturer’s recommendations.<br />

The TaqMan assay to amplify a 200 bp fragment <strong>of</strong> the LipL32<br />

gene was optimized (primer/probe concentration, annealing<br />

temperature and incubation time with the QuantiTec Probe PCR<br />

Kit (Qiagen) for urine samples and kidney according to the<br />

manufacturer’s instructions. As a control for PCR inhibitors,<br />

specific primers and probe were designed to a housekeeping<br />

gene, β-actin, in cattle for kidney samples but in case <strong>of</strong> urine<br />

samples TaqMan Exogenous Internal Positive Control reagent<br />

(Applied Biosystems) was added to the amplification reaction.<br />

The limit <strong>of</strong> detection was ten template copies. Thirty two urine<br />

samples were used to determined diagnostic specificity (DSp). All<br />

<strong>of</strong> them were culture negative. Also thirty two urine samples and<br />

20 kidney samples that contained serovar Hardjo, as determined<br />

by culture were used to determine diagnostic sensitivity (DSe).<br />

The table below gives the results.<br />

Table 1: Diagnostic sensitivity and specificity <strong>of</strong> the LipL32 realtime<br />

PCR.<br />

Sensitivity % Specificity %<br />

Urine 84.4 90.6<br />

Kidney 95.0 Not tested<br />

Discussion & conclusions<br />

This study evaluated the real-time PCR assay targeting LipL32<br />

gene for detection <strong>of</strong> Leptospira spp in bovine kidney and urine.<br />

The results indicate that the sensitivity <strong>of</strong> the diagnostic assay<br />

varies for kidney and urines. Presumably, some false negative<br />

results were caused by substances in the urine that inhibited<br />

extraction <strong>of</strong> the DNA, as the exogenous internal positive control<br />

did not show inhibition <strong>of</strong> the amplification reaction.<br />

In view <strong>of</strong> the difficulties associated with isolation <strong>of</strong> leptospires<br />

and the fact that serology does not imitate the carrier or shedding<br />

status, the LipL32 real-time PCR assay may be used as method<br />

for the detection <strong>of</strong> pathogenic leptospires in bovine urine and<br />

kidney samples<br />

References<br />

1. Ellis, W.A., (1984) Bovine Leptospirosis in the tropics: Prevalence,<br />

Pathogenesis and Control. Pre Vet Med 2: 411-421.<br />

2. Ellis, W.A., O’Brien, J.J., Bryson, D.G., Mackie, D.P., (1985) Bovine<br />

leptospirosis: some clinical features <strong>of</strong> serovar Hardjo infection. Vet Rec<br />

117: 101-104.<br />

3. Ellis, W.A., Songer, J.G., Montgomery, J., Cassells, J.A., (1986)<br />

Prevalence <strong>of</strong> Leptospira interrogans serovar Hardjo in the genital and<br />

urinary tracts <strong>of</strong> non-pregnant cattle. Vet Rec 118: 11-13.<br />

4. Ellis, W.A., (1994) Leptospirosis as a cause <strong>of</strong> reproductive failure. Vet<br />

Clin North Am Food Anim Pract 10: 463-478.<br />

5. Levett, P.N., Morey R.E., Galloway R.L., Turner D.E., Steigerwalt A.G.,<br />

Mayer L.W. (2005) Detection <strong>of</strong> pathogenic leptospires by real-time<br />

quantitative PCR. J Med Microbiol 54: 45-49.<br />

The real time PCR was tested against a panel <strong>of</strong> bacterial<br />

species known to be ruminant pathogens to determine the<br />

analytical specificity <strong>of</strong> the assay. The analytical sensitivity was<br />

calculated using a dilution series <strong>of</strong> target DNA. A genome size <strong>of</strong><br />

3.9 Mb was used to determine the genomic equivalent (GE) per<br />

microlitre <strong>of</strong> the purified DNA. The diagnostic sensitivity and<br />

specificity <strong>of</strong> the assay was determined by testing samples with<br />

only known culture status.<br />

Results<br />

The LipL32 target was amplified from 35 strains tested which<br />

belonged to 6 pathogenic leptospiral species (L.interrogans,<br />

L.borgpetersenil, L.kirschneri, L. santarosai, L.noguchii, L. weilii).<br />

The non-pathogenic Leptospira biflexa, intermediate L. fainei and<br />

other 20 bovine pathogens included were not amplified.


S4 - P - 03<br />

USEFULNESS OF LIVE/DEAD BACLIGHT BACTERIAL VIABILITY KIT TYPE 7007 MOLECULAR<br />

PROBES FOR EVALUATION OF VIABILITY ASCARIS SP., TOXOCARA SP. AND TRICHURIS SP.<br />

EGGS ISOLATED FROM SEWAGE SLUDGE<br />

Joanna Dąbrowska, Maciej Kochanowski, Krzyszt<strong>of</strong> Stojecki, Jolanta Zdybel, Tomasz Cencek<br />

National Veterinary Research Insitute, Departament <strong>of</strong> Parasitology and Invasive Disease, Pulawy, Poland<br />

Viability, sludge, Ascaris, staining, fluorescence<br />

Introduction<br />

Sewage sludge and treated sewage from wastewater treatment<br />

plant despite the use <strong>of</strong> new ways <strong>of</strong> decontamination may<br />

contain live parasite eggs, which after entering the watering<br />

place, drinking water or into the fields as fertilizer can be a source<br />

<strong>of</strong> infection <strong>of</strong> humans and animals. For this reason, there is duty<br />

to monitor the safety <strong>of</strong> these substances, including the obligation<br />

to parasitological research. According the current rules<br />

parasitological examination includes detection <strong>of</strong> the Ascaris<br />

suum, Trichuris sp., and Toxocara sp. eggs and assess their<br />

viability.The assessment <strong>of</strong> their viability based only on the<br />

incubation and observation <strong>of</strong> isolated egg is long, time- and<br />

labour-consiuming and imprecise.<br />

The aim <strong>of</strong> this study was to develop sensitive and less labourintensive<br />

methods for assessing viability <strong>of</strong> Ascaris suum,<br />

Toxocara sp. and Trichuris sp. eggs in sewage sludge and<br />

organic fertilizers.<br />

Material & methods<br />

Dehydrated sludge obtained from municipal wastewater<br />

treatment plant. Eggs <strong>of</strong> parasites isolated from the female<br />

nematode Ascaris suum, Trichuris ovis and Toxocara canis were<br />

used in the investigation (parasites were obtained from the<br />

intestines <strong>of</strong> animals killed in a slaughterhouse or excreted with<br />

faeces). Some <strong>of</strong> the eggs were inactivated by incubation in a<br />

water bath in 60 0 C for 30min. Eggs viability was confirmed by<br />

incubation in a moist chamber at 27 0 C. For differential staining <strong>of</strong><br />

live and dead eggs were used diagnostic kit Live/Dead Bacterial<br />

<strong>of</strong> Molecular Probes, type 7007.<br />

The study was conducted in two experiments.<br />

Experiment 1. Live and inactivated parasite eggs were<br />

suspended in distilled water in separate test tubes. Then the<br />

eggs were stained using a set <strong>of</strong> Live/Dead Bacterial Kit 7007<br />

using the ratio: 1 ml <strong>of</strong> eggs suspended in water and 6 ml dye<br />

mixture (3 ml dye SYTO 9 and 3 ml <strong>of</strong> dye propiodium iodide).<br />

After thorough mixing, the suspension was incubated for 15 min<br />

at room temperature in the dark. Then it filtered through a<br />

polycarbonate filter, and so prepared sample coated with<br />

glycerol. The sample was viewed under a fluorescence<br />

microscope with suitable filters for wavelength settings excitation<br />

470 nm, emission 490 at the area (magnifcation 400x).<br />

Experiment 2. Live and dead eggs were added to separate<br />

samples <strong>of</strong> dewatered sewage sludge. Then the eggs were<br />

isolated from samples using own methods which is the<br />

combination <strong>of</strong> flotation and sedimentation methods (3) which is<br />

the combination <strong>of</strong> flotation and sedimentation methods. Before<br />

filtering stage <strong>of</strong> the match after flotation and sedimentation <strong>of</strong><br />

sludge contained in the eggs <strong>of</strong> parasites were stained with<br />

Live/Dead Bacterial, type 7007 Molecular Probes. In the<br />

experiment used the proportion: in 20 ml <strong>of</strong> sludge was added 8<br />

ml mixture <strong>of</strong> dyes (4 ml SYTO 9 and 4 ml <strong>of</strong> dye propidium<br />

iodide). Then the sample was proceeded as in experiment 1.<br />

Results<br />

Experiment 1. The significant differences in staining <strong>of</strong> live and<br />

dead eggs <strong>of</strong> all three species <strong>of</strong> parasites were obtained. Strong<br />

green color <strong>of</strong> the live eggs and red color <strong>of</strong> inactivated eggs<br />

were observed under the fluorescence microscope (Figure 1).<br />

Experiment 2. With regard to the presence <strong>of</strong> organic matter<br />

derived from sewage sludge generated staining <strong>of</strong> eggs was<br />

similar as in experiment 1 although the colours were less<br />

intensive. Moreover, live eggs <strong>of</strong> Toxocara sp. took a green or<br />

blue than in experiment 1. In addition, live eggs <strong>of</strong> Toxocara sp.<br />

took a green or blue colour. Exemplary stained live and dead<br />

eggs obtained from the sewage sludge observed in fluorescence<br />

compared with the same eggs in bright field are shown in Figure<br />

2.<br />

Fig. 1 Example <strong>of</strong> stained live and dead eggs <strong>of</strong> Trichuris sp.<br />

suspended in water observed in fluorescence microscope<br />

Live egg in water<br />

Dead egg in water<br />

Fig. 2 Stained live and dead eggs obtained from sewage sludge<br />

observed in fluorescence and bright field microscope<br />

Live eggs<br />

Dead eggs<br />

Method <strong>of</strong> observation<br />

Method <strong>of</strong> observation<br />

Bright field Fluorescence Bright field Fluorescence<br />

1 1<br />

2 2<br />

3 3<br />

As is apparent from the images the biggest difference in the<br />

staining <strong>of</strong> live and dead eggs were obtained for eggs A. suum<br />

(images 1) and Trichuris (images 3). Slightly weaker results were<br />

obtained when stained eggs <strong>of</strong> nematodes- Toxocara (some <strong>of</strong><br />

the living eggs stained in blue, and dead eggs were pink-blue,<br />

images 3).<br />

Discussion & conclusions<br />

In most methods used in the world for parasitological<br />

examinations <strong>of</strong> sewage sludge eggs viability assessment is<br />

made by direct evaluation <strong>of</strong> the morphology <strong>of</strong> the eggs or<br />

observation and assessment their development during<br />

incubation. However, these methods are very subjective and in<br />

case <strong>of</strong> incubation very time and labour-consuming. Moreover<br />

the addition <strong>of</strong> glycerol (required to observe the microscopic filter<br />

papers) strongly inhibits the development <strong>of</strong> eggs. Diagnostic Kit<br />

Live/Dead Bacterial, the type <strong>of</strong> company Molecular Probes<br />

7007, composed <strong>of</strong> two components: SYTO 9 giving a green<br />

fluorescence, and propidium iodide (PI) - red fluorescence make<br />

use <strong>of</strong> a different ability <strong>of</strong> the two dyes to penetrate membrane<br />

<strong>of</strong> bacteria (1).<br />

The study showed that both dyes behave similarly also in relation<br />

to the cells <strong>of</strong> worm eggs. It was found that this method after the<br />

introduction <strong>of</strong> minor modifications by the nature <strong>of</strong> the test<br />

samples can be successfully used to distinguish between<br />

live and dead eggs <strong>of</strong> intestinal parasites in sewage sludge. This<br />

method is less dependent from subjective assessment <strong>of</strong> the<br />

investigator and less time consuming than other methods used to<br />

assess the viability <strong>of</strong> eggs <strong>of</strong> parasitic nematodes.<br />

References<br />

1. Biggerstaff J.P., Le Puil M., Weidow B.L., Prater J., Glass K.,<br />

Radosevich M., White D. New methodology for viability testing in<br />

environmental samples. Mol Cell Probes. 2006 Apr;20(2):141-6.<br />

Epub 2006 Feb 14.<br />

2. Zdybel J, Karamon J., Cencek T.. Występowanie jaj nicieni<br />

pasożytniczych z rodzajow Ascaris, Trichuris i Toxocara<br />

w nawozach organicznych i organiczno-mineralnych oraz<br />

osadach ściekowych. Życie Weterynaryjne, rocznik 84 2009,<br />

rocznik 84


S4 - P - 04<br />

AUTOMATION OF A CAPTURE PROBE MAGNETIC BEAD DNA EXTRACTION METHOD FOR 3 GRAM<br />

FAECAL SAMPLES FROM RED FOX INTENDED FOR PCR-DETECTION OF ECHINOCOCCUS<br />

MULTILOCULARIS<br />

Isaksson, M 1 , Hagström, Å 1 , Lukacs, M 2 , Holmberg, A 3 , Juremalm, M 1<br />

1<br />

National Veterinary Institute, Department <strong>of</strong> Virology, Immunobiology and Parasitology, Uppsala, Sweden<br />

2<br />

Nordiag ASA, Research and development, Oslo, Norway<br />

3<br />

Nordiag AB, Research and development, Stockholm, Sweden<br />

Automation, capture probe, DNA extraction, faecal samples, Echinococcus multilocularis<br />

Introduction<br />

As part <strong>of</strong> the Echinococcus multilocularis (EM) surveillance in<br />

Sweden, 4000 red fox faecal samples will be collected and<br />

analyzed by PCR. This necessitates an automated DNA<br />

extraction method. Due to the heterogeneous presence <strong>of</strong> EMeggs<br />

in faeces, 3 grams <strong>of</strong> sample material is needed for the<br />

DNA extraction <strong>of</strong> this analysis. Earlier findings show that a<br />

capture probe magnetic bead extraction method (1) increases<br />

sensitivity <strong>of</strong> the assay compared to the standard egg-flotation<br />

and DNA extraction previously used at the National Veterinary<br />

Institute in Sweden (not published). In this study, we show how<br />

automation <strong>of</strong> the manual capture probe magnetic bead DNA<br />

extraction method capable <strong>of</strong> handling 3 grams <strong>of</strong> faeces per<br />

sample can increase the sample throughput capacity compared<br />

to manual extraction.<br />

Materials & methods<br />

The robot used in this study is a Nordiag Bullet (Nordiag ASA). It<br />

is a flexible robot for microplate format and includes a gripper for<br />

plate transfers, a splittable 1ml eight channel pipette head,<br />

magnetic separation station, bar code capability and external<br />

waste. To be able to automate the large sample volume <strong>of</strong> the<br />

three grams <strong>of</strong> faeces in lysis buffer, 10ml 24-well deepwell<br />

plates were used together with two high power magnet stations.<br />

Results<br />

Essentially the automation consists <strong>of</strong> the five wash steps <strong>of</strong> the<br />

magnetic beads and the magnetic pelleting between washes,<br />

eliminating the need to manually pipette the wash steps <strong>of</strong> the<br />

samples. The robot can be loaded with up to 48 individual<br />

samples consisting <strong>of</strong> 3 grams <strong>of</strong> homogenized fox faeces in<br />

buffer with biotinylated hybridization probes hybridized to the<br />

target and attached to streptavidin coated magnetic beads. When<br />

the robot is finished, the target DNA is melted <strong>of</strong>f the probes and<br />

the probe/magnetic bead pellet is discarded. The manual capture<br />

probe magnetic bead DNA extraction method allows a laboratory<br />

technician to finish 12-20 samples in one day. The automation<br />

allows the same technician to finish 48 samples in one day, with<br />

time to do other things while the robot is working. 96 samples in<br />

one long day are also possible, but in this case with more or less<br />

100% hands on time, preparing the next batch <strong>of</strong> samples while<br />

the robot is processing the first batch.<br />

Tab 1. Throughput <strong>of</strong> samples per week per technician, time for<br />

weighing and division <strong>of</strong> samples not included.<br />

Samples per<br />

week per<br />

technician<br />

Manual extraction<br />

Automated extraction<br />

100 336<br />

Fig 1. The Nordiag Bullet robot used in the study<br />

Discussion & conclusions<br />

The preparation <strong>of</strong> faecal samples before extraction, manual or<br />

automated, is difficult to automate. The samples have to be<br />

divided and put in the correct format tube or well before the<br />

extraction method itself can be initiated. Automating the washing<br />

steps <strong>of</strong> this method means that the sample throughput is<br />

increased enough for the hybridization capture probe method to<br />

be implemented as a routine, large scale DNA extraction method<br />

for the PCR detection <strong>of</strong> EM eggs in red fox faecal samples.<br />

Current work is aimed at modifying the automated capture probe<br />

DNA extraction method to be usable also for RNA targets in a<br />

standardized format, mixing RNA and DNA targets in the same<br />

robot run, or even in the same sample well. This would make the<br />

automated capture probe extraction method even more<br />

interesting for a number <strong>of</strong> difficult sample materials such as<br />

organ material, soil, sewage water etc.<br />

Acknowledgements<br />

This project is funded by the Swedish Civil Contingencies<br />

Agency.<br />

References<br />

1. Opsteegh, M, Langlelaar, M, Sprong, H, den Hartog, L, De Craeye, S,<br />

Bokken, G, Ajzenberg, D, Kijlstra, A, van der Giessen, J. 2010. Direct<br />

detection and genotyping <strong>of</strong> Toxoplasma gondii in meat samples using<br />

magnetic capture and PCR. International Journal <strong>of</strong> Food Microbiology,<br />

139(3), 193-201.


S4 - P - 05<br />

EVALUATION OF THREE ELISAs FOR DETECTING SERUM ANTIBODIES AGAINST MYCOPLASMA<br />

HYOPNEUMONIAE<br />

Martos-Raich, Alba, Porquet-Garanto, Lourdes, Rebordosa-Trigueros, Xavier<br />

HIPRA, 17170 Amer, Girona, Spain<br />

1 Keywords: Mycoplasma hyopneumoniae, ELISA, vaccinated/challenged animals<br />

Introduction<br />

Mycoplasma hyopneumoniae (Mhyo) is an important component<br />

<strong>of</strong> the so called Porcine Respiratory Disease Complex (PRDC).<br />

Mhyo infection in pigs is usually monitored through serology. In<br />

this study, we compared three ELISAs developed to detect<br />

antibodies to Mhyo in swine serum. The main objectives <strong>of</strong> this<br />

study were to differentiate serum antibody pr<strong>of</strong>iles <strong>of</strong> four groups<br />

<strong>of</strong> animals: non-challenged/vaccinated, challenged/vaccinated,<br />

challenged/non-vaccinated and non-challenged/non-vaccinated<br />

animals.<br />

Materials & methods<br />

Seventy young piglets (10-day-old) serologically negative to<br />

Mhyo were divided into 7 groups <strong>of</strong> 10 animals each. Groups 1-5<br />

were vaccinated with 5 different commercial vaccines against<br />

enzootic pneumonia on D0, and D21, and subsequently<br />

challenged on D70 with Mhyo strain 3371. Groups 6 and 7 were<br />

used as non-vaccinated/challenged and non-vaccinated/nonchallenged<br />

control groups, respectively. Blood samples were<br />

taken from all animals at D0, D21, D42, D70 and D98. All<br />

samples were tested using the CIVTEST SUIS MHYO ELISA kit,<br />

a commercial indirect ELISA (ELISA-I), and a commercial<br />

competition ELISA based on a monoclonal antibody against the<br />

p74 (ELISA-C).<br />

Results<br />

All the kits showed good performance in the analysis <strong>of</strong> specificity<br />

(100%) (2). In regard to sensitivity, it varies depending on the<br />

type <strong>of</strong> sample analyzed: ELISA-C detected more positive<br />

samples from vaccinated animals (72,2%), followed by CIVTEST<br />

(41,67%) and the ELISA-I (14,81%). No differences in sensitivity<br />

were observed between ELISA-C and the CIVTEST (87,5% in<br />

both cases) in the samples from challenged animals (vaccinated<br />

and non-vaccinated); ELISA-I was less sensitive (50%) (Figure<br />

1). All three kits showed some common performance<br />

characteristics: they displayed maximum sensitivity at D42 after<br />

two doses <strong>of</strong> vaccine and sensitivity diminishes at D70. Also,<br />

sensitivity was lower when dealing with only challenged samples<br />

(2); in this case, CIVTEST and ELISA-C displayed the same<br />

sensitivity (33.3%) and detected all samples from<br />

vaccinated/challenged animals (100%); ELISA-I was the less<br />

sensitive (0% and 61.54%, respectively).<br />

Table 1. Kappa values obtained in the different comparisons.<br />

Comparison All Vacc. Chall.<br />

CIVTEST vs ELISA-C 0.7 0.4 1.0<br />

ELISA-I vs ELISA-I 0.54 0.39 0.25<br />

ELISA-C vs ELISA-I 0.34 0.12 0.25<br />

Figure 1. Quantitative representation <strong>of</strong> the Mhyo ELISA<br />

serology using three commercial kits. The results are shown as a<br />

mean (central point) and standard desviation (vertical bars).<br />

Discussion & conclusions<br />

The most similar kits were the CIVTEST and the ELISA-C<br />

because both <strong>of</strong> them have a high sensitivity (Table 1). CIVTEST<br />

has better performance differentiating quantitatively samples from<br />

vaccinated animals and samples from vaccinated/challenged<br />

animals. The high sensitivity <strong>of</strong> the ELISA-C when detecting<br />

vaccination seems to make this test have more problems when<br />

distinguishing between samples from vaccinated animals and<br />

samples from vaccinated/challenged animals (Figure 1).<br />

References<br />

1. Ameri-Mahabadi, Mehrdad et al: 2055, J Vet Diagn Invest 17:61-65.<br />

2. Earlandson, KR et al: 2005, Journal <strong>of</strong> Swine Health and Production<br />

198:203.


S4 - P - 06<br />

SEPRION-COATED MICROCANTILEVER SENSORS FOR PRP SC DETECTION<br />

Daniela Meloni¹, Danilo Pitardi¹, Maria Mazza¹, Riccardo Castagna 2 , Ivan Ferrante 2 , Carlo Ricciardi 2 , Elena<br />

Bozzetta¹.<br />

¹Istituto Zoopr<strong>of</strong>ilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Istopatologia e Test Rapidi, Turin, Italy;<br />

2<br />

Politecnico di Torino, Scienza dei Materiali e Ingegneria Chimica, Turin, Italy.<br />

Cantilever, Seprion, TSE, PrP sc detection<br />

Introduction<br />

The current state <strong>of</strong> technology to detect animal TSEs relies upon<br />

the post-mortem detection <strong>of</strong> prions, considering the EU trend in<br />

the age limit for testing [1] and in order to improve surveillance<br />

and food safety, new technologies should be developed to allow<br />

the ante mortem tests for PrP sc proteins. The new tests should be<br />

body fluids-based and performed on presymptomatic animals, but<br />

to be effective they would need to detect small amounts <strong>of</strong> PrP sc<br />

and to differentiate PrP c from PrP sc .<br />

Mechanical biosensors appear to be one <strong>of</strong> the most promising<br />

techniques for molecular diagnostics [2]. Dynamic-mode<br />

mechanical biosensors are oscillating devices with a resonance<br />

frequency that changes when molecules land on the cantilever.<br />

Thanks to the monitoring <strong>of</strong> different resonance modes over<br />

arrays <strong>of</strong> several cantilever-based microbalances, the selective<br />

identification and quantification <strong>of</strong> adsorbed mass <strong>of</strong> the order <strong>of</strong><br />

few picograms with very high precision was recently<br />

demonstrated [3].<br />

Here we propose the development <strong>of</strong> a biosensing platform for<br />

PrP sc<br />

detection based on microcantilever arrays coated with a<br />

selective polymer (Seprion from IDEXX Lab. Inc.).<br />

Materials & methods<br />

Microcantilever arrays were fabricated in a clean room making<br />

use <strong>of</strong> standard microtechnologies such as KOH bulk etching,<br />

optical lithography and Reactive Ion Etching; they were dipped<br />

into coating solution for few minutes inside a laminar flow bench,<br />

washed with PBS and MilliQ water, and finally dried on a plate<br />

dryer.<br />

An optical lever setup is used to monitor cantilever resonance<br />

curve while vibrating in vacuum environment; a piezoactuator is<br />

employed for the excitation. We calculated the arithmetic mean<br />

and uncertainty <strong>of</strong> relative frequency deviation Δf/f over the<br />

modes for each cantilever, and then we used the “weighted<br />

average”method to have the best estimation <strong>of</strong> the true value Δf/f<br />

<strong>of</strong> the array [3].<br />

PrP sc<br />

was extracted from scrapie positive samples, brainstems<br />

were homogenised in lysis buffer and centrifuged at 22.000 g for<br />

20 minutes, the supernatant was collected and incubated with<br />

proteinase K (20 μg/ml) for 1 hour at 37° C. After centrifugation at<br />

215.000 g for 1 hour, the pellet was dissolved in distilled water.<br />

The coated arrays were incubated for 2h in a water-based<br />

solution containing pathologic PrP sc or simply water as negative<br />

control experiment (“blank”). Samples were then rinsed in DI<br />

water and finally dried in a steam <strong>of</strong> nitrogen.<br />

Fig. 2. Micro-cantilever array<br />

Fig3. PrP sc capture<br />

Results<br />

Microcantilever arrays were successfully treated with both 0.1%<br />

and 0.2% coating solution: the calculated mass surface densities<br />

seemed to be rather similar over the arrays and dilution, showing<br />

a good homogeneity <strong>of</strong> the functionalization procedure.<br />

Preliminary data exhibited a relevant negative relative frequency<br />

deviation Δf/f up to nearly 10 times higher respect to blank<br />

signals, showing that the polymer remained active in immobilizing<br />

the target molecule and thus an increment <strong>of</strong> mass occurred on<br />

cantilever surfaces when exposed to PrP sc solutions.<br />

Discussion & conclusions<br />

Cantilever-based microbalances were successfully coated with<br />

Seprion polymer to selectively immobilize pathogenic PrP sc .<br />

Preliminary data showed that the technique looks promising, but<br />

more focused experiments are needed to exploit its potentiality.<br />

References<br />

1. European Commission, Brussels, 16.7.2010, COM (2010) 384 final.<br />

Communication from the Commission to the European Parliament and the<br />

Council. The TSE Road map 2, a Strategy paper on Transmissible<br />

Spongiform Encephalopathies for 2010-2015.<br />

2. Arlett, J.L., Myers, E.B., Roukes, M.L. (2011). Comparative advantages<br />

<strong>of</strong> mechanical biosensors. Nat. Nanotechnol. 6, 4:203-15.<br />

3. Ricciardi, C., Fiorilli, S., Bianco, S., Canavese, G., Castagna, R.,<br />

Ferrante, I., Digregorio, G., Marasso, S.L., Napione, L., Bussolino, F.<br />

(2010). Development <strong>of</strong> microcantilever based biosensor array to detect<br />

Angiopoietin-1, a marker <strong>of</strong> tumor angiogenesis. Biosens. Bioelect.,<br />

25:1193-1198<br />

Figure 1: Flow chart explaining the new technique


S4 - P - 07<br />

EVALUATION OF DIFFERENT EXTRACTION METHODS FOR THE DETECTION OF MYCOBACTERIUM<br />

AVIUM SUBSP. PARATUBERCULOSIS IN BOVINE FAECES<br />

Jean-Louis Moyen 1 , Laure Brugère 1 , Julie Charrot 2 , Eric Sellal 2 , Line Kirsty 3 , Adrian McGoldrick 3<br />

1<br />

Laboratoire départemental d’Analyse et de Recherche de la Dordogne,24660 Coulounieix-Chamiers France<br />

2<br />

Laboratoire Service International, Lisieux, France<br />

3 Animal Health and Veterinary Laboratories Agency Starcross, Exeter, United Kingdom<br />

M; avium susp. paratuberculosis, MAP,<br />

Introduction<br />

Johne’s disease or bovine paratuberculosis occurs throughout<br />

the world. It is a chronic, untreatable, intestinal disease <strong>of</strong><br />

ruminants caused by Mycobacterium avium subspecies<br />

paratuberculosis (MAP) 1 . The pr<strong>of</strong>ile and interest in the disease<br />

has risen in recent years, and consequently, it will be beneficial to<br />

have accurate information about prevalence in order to formulate<br />

policy and appropriate control strategies. Central to gaining<br />

accurate prevalence data in any country, is the availability <strong>of</strong><br />

good diagnostic methods. In order to control the disease, MAP<br />

has to be diagnosed ante-mortem by either detecting the<br />

causative agent itself or by the detection <strong>of</strong> specific antibodies.<br />

The need <strong>of</strong> a rapid, low cost, and sensitive diagnostic test would<br />

be beneficial in the detection and control <strong>of</strong> the organism. The<br />

methods currently available include solid and liquid culture,<br />

serological methodologies and more recently, polymerase chain<br />

reaction (PCR) based tests.<br />

Culture (liquid and solid) is sensitive but the time to detection<br />

ranges from 6 to 24 weeks. Serological methods tend to be<br />

used in screening programmes since they are quick and cheap,<br />

however the sensitivity is low. Available PCR methods allow fast<br />

results (2 days) but reported sensitivity also remains low.<br />

Sensitivity and the time taken to reach diagnosis are critical<br />

points in MAP management control programmes. Indeed, these<br />

disease control strategies can <strong>of</strong>ten fail because new clinical<br />

cases are found even after culling <strong>of</strong> positive diagnosed animals.<br />

In this study, with specific reference to PCR, we hypothesise that<br />

the optimization <strong>of</strong> sample preparation and nucleic acids<br />

extraction can improve the sensitivity making PCR a viable option<br />

in the control <strong>of</strong> the disease.<br />

Materials & methods<br />

Faecal material from cows belonging to certified MAP free herds<br />

were used as negative controls. Positive samples originated<br />

from naturally infected animals as well as from spiking negative<br />

faeces with MAP homogenates.<br />

Naturally infected samples, as well as those spiked, were used to<br />

prepare a dilution series <strong>of</strong> samples. Each set <strong>of</strong> samples were<br />

prepared in duplicate. All samples, both naturally infected and<br />

those spiked, were thoroughly mixed to ensure homogeneity (as<br />

much as practically possible).<br />

For a field trial (epidemiological study), sampling was made from<br />

undiluted samples. The heterogeneity between samples is<br />

increased but the aliquots are more representative using real<br />

faeces.<br />

Sample preparation conditions were comparable (size, dilution,<br />

shaking, bead beating, pK incubation and spinning)<br />

In this study we compared 3 PCR methods. The first was an<br />

IS900 real time PCR test from LSI (Taqman) 2,3 using a Qiagen<br />

spin column extraction method with 1g faeces and 4 ml H2O.<br />

The alternative methods were:<br />

An ambion magnetic beads method on Magmax KF96<br />

developped by AHVLA 4 and an LSI magnetic beads (MV384)<br />

method on Magmax KF96 optimized by the LDAR24.<br />

Results<br />

Spiked and diluted faeces:<br />

All negative samples were negative<br />

Detection <strong>of</strong> higher dilutions depends on the methods<br />

Tab.1. spiked samples – CT values for serial dilutions for each<br />

method<br />

10 -2 1/2 10 -2 1/4 10 -2 1/8<br />

10 -2<br />

1/16<br />

10 -2<br />

1/32<br />

10 -2<br />

1/64<br />

Qiagen<br />

Spin<br />

columns<br />

33.52 34.44 34.73 undet 37.81 38.75<br />

35.54 35.03 35.57 35.53 34.59 undet<br />

33.26 33.97 36.09 37.85 undet 37.39<br />

33.58 38.09 36.97 38.13 undet undet<br />

mean 33.98 35.38 35.84 37.17 36.20 38.07<br />

s 1.05 1.86 0.94 1.43 2.28 0.96<br />

Soil kit<br />

24.96 26.39 27.73 28.26 29.33 31.24<br />

25.36 26.14 27.2 28.01 29.77 31.06<br />

25.3 25.81 27.16 28.17 29.4 33.1<br />

26.02 26.49 27.31 28.09 29.64 34.42<br />

mean 25.41 26.21 27.35 28.13 29.54 32.46<br />

s 0.44 0.30 0.26 0.11 0.21 1.60<br />

Ambion<br />

AHVLA<br />

28.94 28.08 30.09 30.46 31.3 33<br />

27.21 27.11 31.17 31.89 31.54 32.96<br />

26.9 29.23 30.04 31.31 31.2 32.72<br />

26.96 27.63 31.05 33.06 33.44 33.08<br />

mean 27.50 28.01 30.59 31.68 31.87 32.94<br />

s 0.97 0.90 0.61 1.09 1.06 0.15<br />

Discussion & conclusions<br />

The heterogeneity <strong>of</strong> samples for field trial is important.<br />

The best method sensitivity is obtained with AHVLA<br />

method.<br />

This method is requires more time and needs a larger<br />

centrifuge.<br />

The LSI magnetic beads method gives a very good<br />

sensitivity too.<br />

The classic spin column method gives the worst<br />

sensitivity<br />

The epidemiological sensitivity is much better with the<br />

optimized methods and allows better eradication<br />

program.<br />

The choice between these methods depends on the customer’s<br />

requirement. (cost versus sensitivity)<br />

References<br />

1. N. Beth Harris, R. G. Barletta Clin. Microbiol. Rev. 2001 14(3): 489-512<br />

Mycobacterium avium ssp paratuberculosis in veterinary medicine<br />

2. E.P. Green, M.L.V. Tizard, M.T. Moss, J. Thompson, D.J. Winterhouse,<br />

J.J. McFadden and J. Hermon-Taylor: 1989, Sequence and characteristics<br />

<strong>of</strong> IS900, an insertion element identiefied in a human Crohn’s disease<br />

isolate <strong>of</strong> Mycobacterium paratuberculosis. Nucleic acid research volume<br />

17 number 22 9063-9073<br />

3.. LSI Kit TaqVetM. paratuberculosis advanced V003<br />

4. A. McGoldrich, K. Line <strong>2012</strong> (personnal communication)<br />

<br />

<br />

Sample preparation (sample size, dilution, shaking,<br />

bead beating and spinning conditions) is very important<br />

Soil kit (Fast Dna MPBIO) gives the best sensitivity but<br />

is time consuming and is not usable for field diagnosis


S4 - P - 08<br />

REAL-TIME PCR FOR DETECTION OF CAMPYLOBACTER JEJUNI, COLI AND LARI IN FOODS: TEST<br />

VALIDATION ACCORDING TO ISO16140:2003<br />

Nogarol C. 1 , Vencia W. 1 , Bianchi DM. 1 , Gallina S. 1 , Adriano D. 1 , Zuccon F. 1 , Decastelli L. 1<br />

1<br />

Istituto Zoopr<strong>of</strong>ilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, S.C. Controllo Alimenti e Igiene delle Produzioni, Turin, Italy<br />

Campylobacter, ISO 16140:2003, Real Time PCR<br />

Introduction<br />

Several countries with foodborne outbreaks (FBO) reporting<br />

systems documented significant increases over the past few<br />

decades in the incidence <strong>of</strong> diseases caused by microorganisms<br />

in food, including pathogens such as Salmonella, Campylobacter<br />

jejuni and enterohaemorrhagic Escherichia coli (3).<br />

Changes in food habits, such as a preference for fresh and<br />

minimally processed foods and the longer shelflife <strong>of</strong> products,<br />

contributed to the increased incidence <strong>of</strong> FBO ascribed to<br />

microbiological organisms.<br />

The recent EU-policy bases the surveillance <strong>of</strong> foodborne<br />

zoonoses on laboratory isolation <strong>of</strong> bacteria considered sources<br />

<strong>of</strong> outbreaks (3).<br />

Althought standardized by international bodies, cultural methods,<br />

due to execution time, not always allow the pathogen isolation<br />

and identification before outbreak conclusion, in particular when<br />

sources <strong>of</strong> FBO are represented by slow growing bacteria. On<br />

the contrary, the identification <strong>of</strong> bacteria responsible <strong>of</strong> FBO<br />

should be ended in shortly, in order to guarantee a specific<br />

therapy <strong>of</strong> ill patients and to correctly implement report database<br />

systems.<br />

For this reason, the use <strong>of</strong> alternative methods able to rapidly<br />

identify the source <strong>of</strong> FBO is recommended: European<br />

regulations accept the use <strong>of</strong> these rapid methods as long as<br />

they have been validated towards standardized reference method<br />

according to ISO 16140:2003 (1).<br />

Data here reported describe the results <strong>of</strong> validation <strong>of</strong> a<br />

qualitative Real-Time PCR commercial kit for the detection <strong>of</strong><br />

Campylobacter jejuni, coli and lari (JCL) in the food category<br />

“fruit and vegetable based products”.<br />

Materials & methods<br />

From January to August 2011, the Laboratorio Controllo Alimenti<br />

e Igiene delle Produzioni, in Istituto Zoopr<strong>of</strong>ilattico Sperimentale<br />

<strong>of</strong> Piemonte, Liguria and Valle d’Aosta (North-Western Italy) led a<br />

study aimed to verify the performances and to validate a<br />

Campylobacter JCL Real-Time detection kit.<br />

The commercial kit consists in two steps occurring in a semiautomated<br />

closed system: extraction <strong>of</strong> bacterial DNA and<br />

detection <strong>of</strong> Campylobacter JCL target genes. Reagents<br />

necessary for these two steps are included in the kit. According<br />

to the ISO 16140:2003, the validation was performed against the<br />

reference method (ISO 10272:2006) (2) and the following<br />

parameters were evaluated: Limit <strong>of</strong> Detection (LOD); relative<br />

accuracy, relative specificity and sensitivity; relative detection<br />

level (RDL); inclusivity and exclusivity.<br />

To evaluate LOD, a spiking inoculum was prepared with a<br />

concentration <strong>of</strong> Campylobacter jejuni <strong>of</strong> 1,5*10 7<br />

CFU/ml: eight<br />

decimal dilutions were prepared and one millilitre <strong>of</strong> each dilution<br />

was used to spike samples; after incubation, pre-enriched<br />

samples were tested with commercial detection kit.<br />

In order to determine relative accuracy, relative specificity and<br />

sensitivity a total <strong>of</strong> number <strong>of</strong> 60 test portions (50% positive, with<br />

three contamination levels and 50% negative samples) were<br />

analyzed.<br />

The relative detection level (RDL) is the smallest number <strong>of</strong><br />

culturable microorganism that can be detected in the sample in<br />

50% <strong>of</strong> occasions by the alternative and reference methods: it<br />

was evaluated throught the contamination <strong>of</strong> 72 food products<br />

with the three different target microorganisms (C.jejuni, coli and<br />

lari), at three concentration levels (24 negative control, 24<br />

samples spiked with theoretical detection level and 24 samples<br />

spiked with a bacteria concentration just above LOD level). Each<br />

combination (food product-level <strong>of</strong> contamination) was replicated<br />

six times by both the alternative and reference methods.<br />

In order to evaluate the inclusivity <strong>of</strong> the alternative method, 50<br />

pure culture <strong>of</strong> target microorganism were tested. The inoculum<br />

level used was 10 to 100 times greater than the minimum RDL <strong>of</strong><br />

the alternative method.<br />

The exclusivity was verified analyzing 30 pure cultures <strong>of</strong> nontarget<br />

strains (50% <strong>of</strong> Gram positive and 50% <strong>of</strong> Gram negative)<br />

Finally, to perform a complete validation according to ISO<br />

16140:2003 an interlaboratory study was organized, in order to<br />

determine the variability <strong>of</strong> the results obtained in different<br />

laboratories using identical samples.<br />

A total <strong>of</strong> 10 collaborative laboratories were included in the study:<br />

each <strong>of</strong> them received 24 blind samples, spiked with three<br />

contaminations levels (8 negative controls, 8 slightly above the<br />

LOD level and 8 samples 10 times greater than LOD level).<br />

Contamination was performed with reference material (lenticules<br />

discs) from HPA containing lyophilized C. jejuni pure culture.<br />

Guidelines and requirements for the interlaboratory study are<br />

given in the Annex H <strong>of</strong> ISO 16140:2003.<br />

Results<br />

The results obtained in our laboratory defined the LOD <strong>of</strong> Real-<br />

Time PCR protocol at 4 CFU/ 25 g or ml <strong>of</strong> food sample; the<br />

Table 1 resumes the results about relative accuracy, relative<br />

specificity and sensitivity.<br />

Table 1: Criteria percentage obtained with paired tabulate; LCL:<br />

lower confidence limit at 95%<br />

PARAMETERS % LCL 95%<br />

Relative Accuracy 98.33 96<br />

Relative Specificity 100 > 98<br />

Relative Sensitivity 96,66 93<br />

Only one sample resulted positive with alternative method and<br />

negative with the reference one.<br />

Interpretation <strong>of</strong> results obtained for the determination <strong>of</strong> RDL<br />

were calculated for each level and for each food/strain<br />

combination, comparing both methods performing exact Fisher<br />

tests. Results gave a p value=1 for each contamination levels:<br />

the probability that methods are able to detect the inoculum<br />

levels is 100% in all tested samples. Results <strong>of</strong> inclusivity,<br />

exclusivity and interlaboratory study were satisfactory, confirming<br />

all expected determinations.<br />

Discussion & conclusions<br />

The alternative Real-Time PCR method was successfully<br />

validated according to ISO 16140:2003. All the criteria and<br />

parameters tested gave excellent values and allowed to consider<br />

this commercial kit suitable for <strong>of</strong>ficial control on fruit and<br />

vegetable food samples. Finally the study also confirmed that this<br />

commercial kit is easy to use and handle by laboratory<br />

personnel.<br />

Acknowledgements<br />

This study was founded by the Italian Health Ministry: Ricerca Corrente<br />

Ministero della Salute, anno 2009, IZS PLV 12/09.<br />

References<br />

1. ISO 16140:2003, Microbiology <strong>of</strong> food and animal feeding stuffs –<br />

Protocol for the validation <strong>of</strong> alternative methods.<br />

2. ISO 10272:2006, Microbiology <strong>of</strong> food and animal feeding stuffs-<br />

Horizontal method for detection and enumeration <strong>of</strong> Campylobacter spp.<br />

3. EFSA Journal <strong>2012</strong>;10(3):2597


S4 - P - 09<br />

VALIDATION OF THE ID SCREEN SALMONELLA DUBLIN COMPETITIVE ELISA<br />

Pourquier, P. 1 , Loic, C. 1 , Pozet, F. 2 , Buthod-Garçon, M.P. 2<br />

1<br />

IDvet, Montpellier, France<br />

2<br />

LVD39, Poligny, France,<br />

Salmonella Dublin, diagnostics, serology, ELISA<br />

Introduction<br />

The most common type <strong>of</strong> Salmonella affecting cattle in many<br />

countries is Salmonella enteritica, subspecies enteritica serotype<br />

Dublin (Salmonella Dublin). Salmonella Typhimurium and<br />

Salmonella Montevideo are also frequently isolated from cattle.<br />

While antigens O:1 and O:12 are common to serotypes Dublin<br />

and Typhimurium, antigen O:9 characterises infection by S.<br />

Dublin.<br />

Salmonella Dublin is a bacteria which is host-adapted to cattle<br />

and which causes both economical and welfare losses to the<br />

cattle industry. It can potentially spread to humans, particularly<br />

through the consumption <strong>of</strong> contaminated, unpasteurised milk<br />

and cheese.<br />

Unlike most other types <strong>of</strong> Salmonella bacteria, Salmonella<br />

Dublin has a tendency to reside in the herds for years or<br />

decades, mainly due to its ability to produce persistently infected<br />

carriers that shed bacteria to the environment either continuously<br />

or periodically. In order to control infection in such endemically<br />

infected herds, it is necessary to cull the carriers and prevent<br />

infection <strong>of</strong> new carriers.<br />

Serology is therefore an important tool in order to limit the spread<br />

<strong>of</strong> the disease by identifying disease carriers, and eliminate<br />

excretors before contamination <strong>of</strong> bulk milk.<br />

IDvet, in collaboration with the Jura Departmental Veterinary<br />

Laboratory (LVD 39), has developed two ELISAs for the detection<br />

<strong>of</strong> antibodies against Salmonella Dublin in bovine milk, serum<br />

and plasma: ID Screen ® Salmonella Dublin Competition, and ID<br />

Screen ® Salmonella Dublin Indirect.<br />

Discussion & conclusions<br />

The specificity <strong>of</strong> the test ID Screen ®<br />

Salmonella Dublin<br />

Competition was found to be (100%, IC95% 99.5-99.99%). No<br />

false positive results due Salmonella Typhimurium were<br />

observed.<br />

The ID Screen ® competitive ELISA is a cost-effective Salmonella<br />

Dublin diagnostic method which facilitates identification <strong>of</strong> herds<br />

at risk and transitory excretors.<br />

It should be noted that the analytical sensitivity <strong>of</strong> competitive<br />

ELISA method on bulk milk is limited by degree <strong>of</strong> excretion <strong>of</strong><br />

the animal, the size <strong>of</strong> the bulk milk, and the fact that milk<br />

contains less antibodies than sera.<br />

A suggested testing protocol would be to screen bulk milks with<br />

the ID Screen Indirect ELISA, and test any positive samples<br />

individually with the competitive ELISA in order to maximize<br />

analytical sensitivity.<br />

References<br />

1. Hoorfar J, Feld NC, Schirmer AL, Bitsch V, Lind P. Serodiagnosis <strong>of</strong><br />

Salmonella dublin infection in Danish dairy herds using O-antigen based<br />

enzyme-linked immunosorbent assay. Can J Vet Res. 1994 Oct;58(4):268-<br />

74.<br />

2. Hoorfar J, Lind P, Bitsch V. Evaluation <strong>of</strong> an O antigen enzyme-linked<br />

immunosorbent assay for screening <strong>of</strong> milk samples for Salmonella dublin<br />

infection in dairy herds. Can J Vet Res. 1995 Apr;59(2):142-8.<br />

3. Nielsen LR, Ersbøll AK. Age-stratified validation <strong>of</strong> an indirect<br />

Salmonella Dublin serum enzyme-linked immunosorbent assay for<br />

individual diagnosis in cattle. J Vet Diagn Invest. 2004 May;16(3):212-8.<br />

This study presents the test validation data for the competitive<br />

ELISA.<br />

Materials & methods<br />

The ID Screen ®<br />

Salmonella Dublin Competition ELISA is a<br />

competitive ELISA based on microplates coated with S. Dublin<br />

LPS and an anti-O:9 monoclonal antibody conjugate.<br />

The ID Screen ®<br />

Salmonella Dublin Indirect ELISA is based on<br />

microplates coated with S. Dublin LPS and an anti-ruminant-HRP<br />

conjugate.<br />

Specificity:<br />

176 individual milks, 84 bulk milks, and 180 sera from a diseasefree<br />

region (Brittany, France) were tested with both kits.<br />

Exclusivity:<br />

111 individual milks from a herd infected with Salmonella<br />

Typhimurium (bacteria were isolated from several animals), were<br />

tested.<br />

Sensitivity:<br />

158 paired milk and sera samples from 20 herds with a history <strong>of</strong><br />

S. Dublin infection were tested. A bacteriological study <strong>of</strong><br />

seropositive animals allowed for the identification <strong>of</strong> excretors in<br />

Herd 2.<br />

Results<br />

Specificity and exclusivity studies: All animals were found<br />

negative with the ID Screen ®<br />

Salmonella Dublin competitive<br />

ELISA.<br />

Sensitivity study:<br />

- Correlation between milk and serum results was excellent<br />

(100%).<br />

- The average herd seroprevalence was found to be: 12.9 %<br />

(3,8%-30,2%).<br />

- 89% (16/18) <strong>of</strong> excretors were also seropositive.


S4 - P - 10<br />

ID SCREEN ® INTERFERON GAMMA ELISA : IMPROVEMENT OF ANALYTICAL SENSITIVITY AND<br />

INTRODUCTION OF A STANDARD REFERENCE CONTROL TO IMPROVE RESULT INTERPRETATION<br />

Pourquier, P. 1 , Loic, C. 1 , Olagnon, L. 1 , Marion, S. 1,<br />

1<br />

IDvet, Montpellier, France<br />

Interferon gamma, diagnostics, ELISA<br />

Introduction<br />

Detection <strong>of</strong> Interferon gamma (IFN-g) by the sandwich ELISA<br />

method is widely used to detect the cellular response to<br />

pathogens such as Mycobacterium avium subsp paratuberculosis<br />

(Map) or Mycobacterium bovis by measuring the difference<br />

between activated and non-activated whole blood or peripheral<br />

Blood Mononuclear Cells (PBMC) IFN-g signals. Results,<br />

expressed as the difference between raw optical densities, are<br />

not generally linked to a stable reference control.<br />

The ID Screen ®<br />

Ruminant IFN-g ELISA, contrary to other<br />

commercial ELISAs, expresses the level <strong>of</strong> IFN-g with respect<br />

to a standardized, freeze-dried positive reference control.<br />

This relative expression <strong>of</strong> the measured quantity <strong>of</strong> IFN-g<br />

guarantees the standardization <strong>of</strong> results between runs and kit<br />

batches.<br />

Results are expressed as sample / positive reference control<br />

(S/P) ratios.<br />

Materials & methods<br />

ID Screen ® Interferon gamma Capture ELISA: Wells are coated<br />

with an anti-IFN-γ monoclonal antibody (Mab). The conjugate is<br />

anti-IFN-γ–HRP conjugate monoclonal antibody.Creation <strong>of</strong><br />

standard reference controls: A freeze-dried positive control<br />

containing native bovine IFN-g was created and introduced onto<br />

each plate. All results were calculated with respect to this<br />

standard. A standard positive plasma reference sample was also<br />

created.<br />

Results<br />

Test baseline: Bovine and ovine sera and PBS-activated bovine<br />

and caprine plasmas were tested and found to have a low<br />

baseline with few heterophilic reactions.<br />

Analytical sensitivity: 5 bovine, 1 caprine and 1 ovine blood<br />

sample were non-specifically activated with 5 µg/ml <strong>of</strong> lectin, 24h<br />

at 37°C. Plasma was collected and each sample was serially<br />

diluted in non-activated plasma. Samples were then tested in<br />

parallel with the ID Screen ®<br />

ELISA and another commerciallyavailable<br />

IFN-g sandwich ELISA (kit A).<br />

- The ID Screen ® ELISA and Test A showed similar analytical<br />

sensitivity for ovine IFN-g, while the ID Screen ELISA<br />

demonstrated superior analytical sensitivity for bovine and<br />

caprine IFN-g. In addition, the ID Screen ® ELISA showed clearer<br />

separation <strong>of</strong> positive and negative samples.<br />

Multi-site field validation: 3 French laboratories tested previouslyactivated<br />

samples (n=216) using the ID Screen ®<br />

ELISA and<br />

another commercial ELISA.<br />

- Excellent correlation (98.6%) was found between the two tests,<br />

with the ID Screen ® test giving higher OD values for the positive<br />

samples.<br />

Field data from a Map-infected goat herd: 180 caprine blood<br />

samples from herd with a history <strong>of</strong> clinical paratuberculosis were<br />

activated with 9 different antigens, including 3 different Map<br />

extracts from a field caprine isolate, as well as PPD avium.<br />

- One <strong>of</strong> the IDvet caprine Map antigen preparations gave a<br />

similar result pr<strong>of</strong>ile to the PPD avium antigen tested.<br />

Discussion & conclusions<br />

The new ID Screen® Ruminant IFN-g ELISA has improved<br />

upon current IFN-g ELISA technology by incorporating a standard<br />

positive reference control for standardized result calculation and<br />

interpretation.<br />

The improved analytical sensitivity allows for the efficient<br />

detection <strong>of</strong> small quantities <strong>of</strong> native IFN-g (threshold dilution on<br />

other techniques), giving optical densities clearly distinct from the<br />

test baseline, thus avoiding false positive results. The very low<br />

level <strong>of</strong> heterophilic reactions and the low baseline guarantee that<br />

result interpretation is possible for all samples to be tested.<br />

In conclusion, the ID Screen ® Ruminant IFN-g ELISA is a reliable<br />

tool for the measurement <strong>of</strong> ruminant interferon gamma.<br />

References<br />

1. Buddle BM, et al. Advances in ante-mortem diagnosis <strong>of</strong> tuberculosis in<br />

cattle. New Zeland Veterinary Journal. 2009, 57(4):173-180.<br />

2. Denis M, Weldock DN, McCarthy AR, Parlane NA, Cockle PJ,<br />

Vordermeier HM, Hewinson RG, Buddles BM, 2007. Enhancement <strong>of</strong> the<br />

sensitivity <strong>of</strong> the whole-blood gamma interferon assay for diagnosis <strong>of</strong><br />

Mycobacterium bovis infections in cattle. Clinical and vaccine immunology,<br />

14(11): 1483-1489.<br />

3. Gormley E, et al. The effect <strong>of</strong> the tuberculin test and the consequences<br />

<strong>of</strong> a delay in blood culture on the sensitivity <strong>of</strong> a gamma-interferon assay<br />

for the detection <strong>of</strong> Mycobacterium bovis infection in cattle. Vet Immunol<br />

Immunopathol. 2004, Dec 28;102(4):413-20<br />

4. Lardizabal AA, Reichman BL, 2006. Interferon-gamma release assay for<br />

detection <strong>of</strong> Tuberculosis Infection. US respiratory Disease, 59-61.<br />

5. Pai M, Riley LW, Colford Jr JM, 2004. Interferon-g assays in the<br />

immunodiagnosis <strong>of</strong> tuberculosis: a systematic review. Infectious diseases,<br />

4:761-776.<br />

6. Robbe-Austermann S, et al. Time delay, temperature effects and<br />

assessment <strong>of</strong> positive controls on whole blood for the gamma interferon<br />

ELISA to detect Paratuberculosis. J Vet Med B Infect Dis Vet Public<br />

Health. 2006, 53(5):213-7.<br />

7. Ryan JT, et al. An evaluation <strong>of</strong> the gamma interferon test for detecting<br />

bovine tuberculosis in cattle 8 to 28 days after tuberculin skin testing.<br />

Research in Veterinary Science. 2000, 69(1) :57-61.


S4 - P - 11<br />

EFFECTIVE TESTING STRATEGY WITH PRIMAGAM ® FOR TUBERCULOSIS IN HUMAN PRIMATES<br />

Björn Schröder 2 , Christian Wenker 1 , Stefan Hoby 1 , Bettina Bernhard 2 , Alex J. Raeber 2<br />

1 Zoo Basel, Switzerland and 2 Prionics AG, Schlieren, Switzerland<br />

Introduction<br />

Primates infected with tuberculosis (TB) represent a serious<br />

regulatory, ethical and health concern as TB control requires<br />

accurate diagnostic methods. However, the primary test, the<br />

tuberculin skin test has serious limitations. Both false negative<br />

and false positive reactions are common, resulting in a spread <strong>of</strong><br />

infection and devastating TB outbreaks in colonies. As a gold<br />

standard for TB diagnostic is not available, a combination <strong>of</strong> test<br />

systems has been suggested to reliably diagnose the disease. In<br />

primates the skin test should be conducted as a primary test but<br />

should always be accompanied in the early and mid term phases<br />

<strong>of</strong> the disease by the IFN- release assay as the most<br />

appropriate test system. As late stage diagnostic tools,<br />

serological and direct tests may provide additional diagnostic<br />

benefits.<br />

In 2010, the Basel ZOO relocated all human primates to an<br />

external facility during the construction phase <strong>of</strong> a new primate<br />

house. The occasion was used to test all animals for TB before<br />

movement <strong>of</strong> the animals and following their resettlement in the<br />

new primate house. The Guideline for the prevention and control<br />

<strong>of</strong> tuberculosis in non-human primates 1 was used as the basis for<br />

the test schedule<br />

Materials & methods<br />

Lelystad tuberculin PPD antigens (Prionics, The Netherlands)<br />

were used for stimulation <strong>of</strong> whole blood cultures. The release <strong>of</strong><br />

IFN- from PBMCs was measured with the PRIMAGAM ®<br />

(Prionics, Switzerland) sandwich enzyme immunoassay (EIA).<br />

Diagnostic sensitivity was assessed in primate colonies <strong>of</strong><br />

unknown Tb status.<br />

Results<br />

Here we report the TB test results from three human primate<br />

colonies from the Basel ZOO. Seven Orangutans, six Gorillas<br />

and eight Chimpanzees have been initially tested with two<br />

different skin tests, two IFN- tests (one was PRIMAGAM ® ), two<br />

antibody tests and thoracic x-ray. Animals which were negative<br />

for all test results were considered as TB free. Animals that<br />

tested positive in at least one test system were required to<br />

undergo further monitoring. One Chimpanzee was tested positive<br />

in more than one test. After relocation all reactors were tested a<br />

second time, whereas all animals were negative tested with<br />

PRIMAGAM ® .<br />

Discussion & conclusions<br />

We have compared the diagnostic performance <strong>of</strong> tuberculin<br />

PPD for the stimulation <strong>of</strong> blood cultures in PRIMAGAM ® IFN-<br />

assay. In the presentation we will highlight the differences <strong>of</strong><br />

different test systems and point out how and when the different<br />

tests should be conducted and how PRIMAGAM ®<br />

can help to<br />

optimize the TB diagnostic in primates.<br />

References<br />

1. Guideline for the prevention and control <strong>of</strong> tuberculosis in non-human<br />

primates: recommendations <strong>of</strong> the European primate veterinary<br />

association working group on Tuberculosis, M. Bushmitz; A. Lecu; F.<br />

Verreck; E. Preussing; S. Rensing; K. Rensing; J. Med Primatol, 38, 59-69,<br />

2009)<br />

Tuberculosis, Primagam, tuberculin,


S4 - P - 12<br />

COMPARISON OF THE EFFICIENCY OF DIFFERENT PARASITOLOGICAL DIAGNOSTIC METHODS<br />

USED IN ANALYSIS OF DEHYDRATED SEWAGE SLUDGES<br />

Jolanta Zdybel, Tomasz Cencek, Jacek Karamon<br />

National Veterinary Research Institute, Departament <strong>of</strong> Parasitology and Invasive Disease, Pulawy, Poland<br />

Ascaris, Toxocara, Trichuris, eggs, sewage sludge, fertilizers<br />

Introduction<br />

The use <strong>of</strong> sewage sludge as fertilizer is connected with<br />

microbiological and parasitological hazards in terms <strong>of</strong> public<br />

health safety. Therefore, it is necessary to carry on proper<br />

parasitological investigations <strong>of</strong> this type <strong>of</strong> samples. Majority, <strong>of</strong><br />

current parasitological diagnostic procedures using in sewage<br />

sludge surveys are derived from soil samples investigation. This<br />

kind <strong>of</strong> procedure are characterized by low efficiency, due to<br />

occurrence <strong>of</strong> polyelectrolytes in sewage sludge samples.<br />

Therefore, new method <strong>of</strong> sewage sludge parasitological<br />

diagnostics was developed. The methods is focused on detection<br />

<strong>of</strong> parasitic nematodes eggs, belonging to Ascaris, Trichuris and<br />

Toxocara genus. It is combination <strong>of</strong> floatation and sedimentation<br />

method, preceded by sample prolonged dispersion. The purpose<br />

<strong>of</strong> this study is to compare the efficiency <strong>of</strong> the method developed<br />

in Department <strong>of</strong> Parasitology and Invasive Diseases to other<br />

methods which are used in laboratories worldwide.<br />

References<br />

1. Zdybel J., Karamon J., Cencek C.: The occurrence <strong>of</strong> eggs <strong>of</strong><br />

parasitic roundworms (genera: Ascaris, Trichuris, Toxocara) in<br />

organic and mineral-organic fertilizers and in sewage sludge.<br />

Zycie Wet.2009,84(12),992-996.<br />

2. Simonart T., Roussel S., Gireaudot-Liepman M.F.: desk study<br />

on European standard for the enumeration <strong>of</strong> diable helminth ova<br />

in sludge, soil and solid waste. HORIZONTAL-WP3-5, France<br />

2003.<br />

Materials & methods<br />

For this study we used randomly chosen 10 dehydrated sewage<br />

sludge samples derived from Polish waste water treatment<br />

plants. Each sample weighed 10 grams (dry weight) and was<br />

examined by our own method, PN-Z-19000-4 method, flotation<br />

method described by Quinn, method according to Environmental<br />

Protection Agency (EPA) and Triple flotation (TF) method<br />

according to ANFOR XP X33-017 . Microscopy (X20) was used<br />

to estimate the number <strong>of</strong> nematode eggs.<br />

Results<br />

Results concerning percentage <strong>of</strong> positive samples and numbers<br />

<strong>of</strong> detected parasite eggs obtained with the use <strong>of</strong> different<br />

methods were presented in Table 1.<br />

Table. 1. Mean number <strong>of</strong> parasite eggs and % <strong>of</strong> positive<br />

samples detected in sewage sludges samples with the use <strong>of</strong> 5<br />

different methods.<br />

Methods<br />

Mean number <strong>of</strong> detected eggs<br />

(% <strong>of</strong> positive samples)<br />

Ascaris Toxocara Trichuris<br />

Quinn<br />

2.2<br />

(70%)<br />

2.1<br />

(90%)<br />

0.3<br />

(30%)<br />

PN-Z-19000-4<br />

0.3<br />

(40%)<br />

0.15<br />

(30%)<br />

0<br />

Triple flotation<br />

0.6<br />

(10%<br />

3.3<br />

(40%)<br />

0<br />

EPA<br />

1.7<br />

(80%)<br />

1.3<br />

(100%)<br />

1.3<br />

(20%)<br />

Own method<br />

16.4<br />

(100%)<br />

11.1<br />

(100%)<br />

1.7<br />

(100%)<br />

Discussion & conclusions<br />

The results <strong>of</strong> the present study demonstrated that method<br />

developed in Department <strong>of</strong> Parasitology and Invasive Diseases<br />

is six-fold more efficient than flotation method devised by Quinn,<br />

seven-fold more efficient than TF method and EPA method, 65-<br />

fold more efficient then PN-Z-19000-4 method. The adaptation <strong>of</strong><br />

described method in other parasitological laboratories is highly<br />

recommended.


List <strong>of</strong> <strong>Abstract</strong>s (in order <strong>of</strong> number)<br />

S1-K-01 VAN DER POEL WIM H. M.<br />

S1-K-02 SCHWARZ STEFAN<br />

S1-O-01 BOSS CHRISTINA<br />

S1-O-02<br />

STRUTZBERG-<br />

MINDER<br />

KATRIN<br />

S1-O-03 BOUWSTRA RUTH<br />

S1-O-04 GÓRNA KAMILA<br />

S1-O-05 NARDINI ROBERTO<br />

S1-O-06 BALLAGI ANDREA<br />

S1-O-07 VAN MAANEN KEES<br />

S1-O-08<br />

REVILLA-<br />

FERNÁNDEZ<br />

SANDRA<br />

S1-O-09 COOLEY WILLIAM<br />

S1-O-10 LANGEVELD JAN<br />

S1-O-11<br />

REBORDOSA-<br />

TRIGUEROS<br />

XAVIER<br />

S1-O-12 O'NEILL RONAN<br />

S1-O-13 SCHIRRMEIER HORST<br />

S1-O-14 BOSS CHRISTINA<br />

S1-O-15 PORQUET-GARANTO LOURDES<br />

S1-O-15 VILLA ALEIDA<br />

S1-O-17 BALLAGI ANDREA<br />

S1-O-18 LOPEZ PABLO<br />

S2-K-01 SOLDAN ANDREW<br />

S2-O-01 NORTH SARAH<br />

THE EMERGENCE OF SCHMALLENBERG VIRUS – HOW TO<br />

RESPOND TO NEW EPIZOOTICS IN EUROPE<br />

ASSESSING THE ANTIMICROBIAL SUSCEPTIBILITY OF<br />

BACTERIA OBTAINED FROM ANIMALS<br />

ORAL FLUIDS – SAMPLE MATRIX FOR EFFECTIVE HERD<br />

HEALTH MONITORING<br />

DIAGNOSTIC PROPERTIES OF SEVERAL PRRS ANTIBODY<br />

ELISAS IN THE CONTEXT OF A LONGITUDINALSTUDY OF<br />

A FARM USING DIFFERENT VACCINATION STRATEGIES<br />

INTRODUCTION RATE OF A LOW PATHOGENIC AVIAN<br />

INFLUENZA VIRUS INFECTION IN DIFFERENT DUTCH<br />

POULTRY SECTORS<br />

A DUPLEX ONE-STEP REAL TIME RT-PCR FOR DIAGNOSIS<br />

OF FOOT AND MOUTH DISEASE<br />

VALIDATION OF A COMPETITIVE ELISA FOR THE<br />

DETECTION OF ANTIBODIES ANTI-P26 OF EQUINE<br />

INFECTIOUS ANEMIA VIRUS IN EQUINE SERA<br />

DIAGNOSTIC PERFORMANCE OF A COMMERCIAL PRRS<br />

SERUM ANTIBODY ELISA ADAPTED TO ORAL FLUID<br />

SPECIMENS: FIELD SAMPLES<br />

SUBTYPING OF SWINE INFLUENZA VIRUSES BY<br />

MULTIPLEX REAL-TIME PCR<br />

IMPORTANCE OF CONTINUOUS VALIDATION OF<br />

MOLECULAR METHODS FOR ROUTINE DIAGNOSIS OF<br />

PRRSV RNA IN CLINICAL SAMPLES<br />

VIRAL DIAGNOSIS USING TRANSMISSION ELECTRON<br />

MICROSCOPY<br />

A BEAD BASED MULTIPLEX IMMUNOFLUOROMETRIC<br />

ASSAY FOR SCREENING AND CONFIRMATION OF ALL<br />

MAJOR PRION TYPES IN SHEEP<br />

USING ORAL FLUID FOR THE SEROLOGICAL<br />

MONITORIZATION OF PRRSV CIRCULATION IN A GROUP<br />

OF INFECTED GILTS<br />

ENHANCED DETECTION OF BOVINE RESPIRATORY<br />

VIRUSES BY INCLUSION OF COHORT ANIMALS<br />

THE FIRST YEAR OF OBLIGATORY BVD CONTROL IN<br />

GERMANY – DIAGNOSTIC STRATEGIES, RESULTS AND<br />

EXPERIENCES<br />

EUROPEAN PRRSV – DIAGNOSTIC SOLUTIONS FOR A<br />

RAPIDLY MUTATING VIRUS<br />

SEROLOGICAL ANALYSIS AND MONITORING OF IBR IS IT<br />

POSSIBLE TO CONTROL IBRGE ANTIBODIES IN A BULK<br />

TANK MILK?<br />

DEVELOPMENT AND VALIDATION OF A REAL-TIME PCR<br />

ZEN GEL MIX FOR THE DIAGNOSIS AND QUANTIFICATION<br />

OF COXIELLA BURNETII<br />

RING TEST EVALUATION FOR THE DETECTION OF PRRSV<br />

ANTIBODIES IN ORAL FLUID SPECIMENS USING A<br />

COMMERCIAL PRRSV SERUM ANTIBODY ELISA<br />

REAL TIME PCR, MYCOPLASMA GALLISEPTICUM,<br />

MYCOPLASMA SYNOVIAE, MYCOPLASMA MELEAGRIDIS<br />

WHEN DO YOU WANT THE RESULT? HOW MUCH DO YOU<br />

WANT TO PAY!<br />

DEVELOPMENT OF A RAPID ISOTHERMAL ASSAY TO<br />

DETECT TAYLORELLA EQUIGENITALIS, THE CAUSATIVE<br />

AGENT OF CONTAGIOUS EQUINE METRITIS


S2-O-02 VRANCKEN ROBERT<br />

S2-O-03 SOCHA WOJCIECH<br />

S2-O-04 VALLS LAURA<br />

S3-K-01 GAVIER-WIDÉN DOLORES<br />

S3-O-01 BOUWSTRA RUTH<br />

S3-O-02 KOOI BART<br />

S3-O-03 BÖTTCHER JENS<br />

S3-O-04 CARTER CRAIG N.<br />

S3-O-05 EGLI C.<br />

LABORATORY VALIDATION OF AN<br />

IMMUNOCHROMATOGRAPHIC TEST FOR THE RAPID<br />

DETECTION OF KOI HERPESVIRUS (CYHV-3) IN GILL<br />

SWABS<br />

EVALUATION OF RAPID HRSV STRIP TESTS FOR<br />

DETECTION OF BOVINE RESPIRATORY SYNCYTIAL VIRUS<br />

EVALUATION OF A LATEX AGGLUTINATION TEST FOR THE<br />

IDENTIFICATION OF CLOSTRIDIUM DIFFICILE OF PORCINE<br />

ORIGIN<br />

EMERGING AND RE-EMERGING WILDLIFE DISEASES:<br />

PATHOLOGY AND RELATED TECHNIQUES<br />

SCHMALLENBERG VIRUS OUTBREAK IN THE<br />

NETHERLANDS: ROUTINE DIAGNOSTICS AND TEST<br />

RESULTS<br />

25 YEARS OF PASSIVE SURVEILLANCE OF BATS IN THE<br />

NETHERLANDS. MOLECULAR EPIDEMIOLOGY AND<br />

EVOLUTION OF EBLV-1<br />

DIAGNOSIS OF Q FEVER IN DAIRY CATTLE BY PHASE-<br />

SPECIFIC MILK-SEROLOGY<br />

EQUINE NOCARDIOFORM PLACENTITIS & ABORTION<br />

OUTBREAK AND FARM-BASED RISK FACTOR STUDY,<br />

2010-2011<br />

A NEW DIAGNOSTIC TOOL FOR BOVINE TUBERCULOSIS-<br />

IDEXX M.BOVIS ANTIBODY TEST KIT<br />

S3-O-06 LA ROCCA ANNA DETECTION OF SCHMALLENBERG VIRUS IN THE UK<br />

S3-O-07 CHAINTOUTIS SERAFEIM C.<br />

S3-O-08 SCHIRRMEIER HORST<br />

S3-O-09 FOERSTER CHRISTINE<br />

S3-O-10 STEINRIGL ADOLF<br />

S3-O-11 POURQUIER PHILIPPE<br />

DETECTION OF WNV ENZOOTIC CIRCULATION IN HORSES<br />

WITH NEUROLOGICAL SIGNS AND IN CAPTIVE SENTINEL<br />

CHICKENS IN THE PREFECTURE OF THESSALONIKI,<br />

GREECE<br />

SCHMALLENBERGVIRUS: SEROLOGICAL STUDIES IN<br />

GERMAN HOLDINGS<br />

DIFFERENT DIAGNOSTIC TOOLS FOR A BROAD RANGE<br />

OF MACAVIRUSES AND THEIR RESERVOIR AND<br />

SUSCEPTIBLE HOSTS<br />

DIAGNOSTIC ASPECTS OF SUID HERPESVIRUS 1<br />

INFECTION IN WILD BOAR<br />

PRELIMINARY VALIDATION OF THE ID SCREEN®<br />

SCHMALLENBERG VIRUS INDIRECT ELISA<br />

S4-K-01 KOSTRZEWA MARCUS MALDI-TOF AND OTHER NEW DIAGNOSTIC<br />

S4-O-01 RIPP ULRIKE<br />

S4-O-02 WRAGG PETER<br />

S4-O-03 RAEBER ALEX<br />

S4-O-04 HEUVELINK ANNET<br />

S4-O-05 OVERESCH GUDRUN<br />

S4-O-06 SAWYER JASON<br />

S1-P-01 AHOLA HEIKKI<br />

S1-P-02 BALLAGI ANDREA<br />

SUITABILITY OF RECOMBINANT PROTEINS FOR THE<br />

DIAGNOSIS OF LEPTOSPIROSIS IN PIGS<br />

BIOLOG GENERATION III, MATRIX ASSISTED LASER<br />

DESORPTION/IONISATION TIME-OF-FLIGHT (MALDI-TOF)<br />

MASS SPECTROMETRY AND 16S RRNA GENE<br />

SEQUENCING FOR THE IDENTIFICATION OF BACTERIA OF<br />

VETERINARY INTEREST<br />

EVALUATION OF THE DIAGNOSTIC PERFORMANCE OF<br />

PEPTIDE COCKTAILS IN THE INTERFERON GAMMA ASSAY<br />

FOR DIAGNOSIS OF TUBERCULOSIS IN CATTLE<br />

MATRIX-ASSISTED LASER DESORPTION IONIZATION-TIME<br />

OF FLIGHT MASS SPECTROMETRY (MALDI-TOF MS) IN A<br />

VETERINARY DIAGNOSTIC LABORATORY<br />

RAPID IDENTIFICATION OF BOVINE MASTITIS<br />

PATHOGENS USING MALDI TOF<br />

15 MINUTE ELISA USING A LOW COST COMMERCIAL<br />

BIOSENSOR<br />

LAWSONIA INTRACELLULARIS IN BLUE FOXES IN<br />

FINLAND. A CASE REPORT<br />

DIAGNOSTIC PERFORMANCE OF A COMMERCIAL PRRS<br />

SERUM ANTIBODY ELISA ADAPTED TO ORAL FLUID:<br />

LONGITUDINAL RESPONSE IN EXPERIMENTALLY-<br />

INOCULATED POPULATIONS


S1-P-03 BALLAGI ANDREA<br />

S1-P-04 BENITO ALFREDO<br />

S1-P-05 BLANCHARD BEATRICE<br />

S1-P-06 BLANCHARD BEATRICE<br />

S1-P-07 BOSS CHRISTINA<br />

S1-P-08 BÖTTCHER JENS<br />

S1-P-09 VAN MAANEN KEES<br />

S1-P-10 NARDINI R.<br />

S1-P-11 CHERNYSHOV ANATOLIY<br />

S1-P-12 CHERNYSHOVA ELENA<br />

S1-P-13 COCCHI MONIA<br />

S1-P-14 CORNAGLIA ESTELA<br />

S1-P-15 EIRAS CARMEN<br />

S1-P-16 EIRAS CARMEN<br />

S1-P-17 METREVELI GIORGI<br />

S1-P-18 GEROLIMETTO ELISA<br />

S1-P-19 EIRAS CARMEN<br />

S1-P-20 HIRVELÄ-KOSKI VARPU<br />

S1-P-21 KOCHANOWSKI MACIEJ<br />

S1-P-22 KOVAC GABRIEL<br />

S1-P-23 KÜHN TILMAN<br />

S1-P-24 MAGNEE DAMIEN<br />

DETECTION OF PRRSV ANTIBODY IN ORAL FLUID<br />

SPECIMENS FROM INDIVIDUAL BOARS USING A<br />

COMMERCIAL PRRSV SERUM ANTIBODY ELISA<br />

DIAGNOSIS OF MAIN HAEMOPARASITIC DISEASES OF<br />

CATTLE BY REAL-TIME PCR<br />

DEVELOPMENT AND EVALUATION OF A NEW AND<br />

ORIGINAL EXTRACTION PROTOCOL TO DETECT<br />

MYCOBACTERIUM AVIUM SUBSP PARATUBERCULOSIS IN<br />

BOVINE FECES BY REAL TIME<br />

VALIDATION OF REAL-TIME PCR TEST FOR THE<br />

DETECTION AND QUANTIFICATION OF COXIELLA<br />

BURNETII FOR ABORTION CONTROL PROGRAM<br />

ANALYTICAL EVALUATION OF A SWINE INFLUENZA VIRUS<br />

PCR ON AVIAN SAMPLES<br />

QUALITY CONTROL OF PRRSV VACCINATION BY<br />

PARAMETERS OF IMMUNITY<br />

PTS FOR BVDV ANTIGEN DETECTION BY GD - ANIMAL<br />

HEALTH SERVICE<br />

PRESENTATION OF THE RESULTS OF THE ANNUAL<br />

PROFICIENCY TEST FOR THE SEROLOGICAL DIAGNOSIS<br />

OF EQUINE INFECTIOUS ANEMIA CONDUCTED IN ITALY<br />

BETWEEN 2006-2010<br />

ANTIMICROBIAL SUSCEPTIBILITY OF AVIBACTERIUM<br />

(HAEMOPHILUS) PARAGALLINARUM ISOLATES<br />

RECOVERED IN THE RUSSIAN TERRITORY<br />

COMPARISON BETWEEN ANALYTICAL SENSITIVITY OF<br />

RABIES VIRUS ISOLATION IN NEUROBLASTOMA CELL<br />

CULTURE WITH ANALYTICAL SENSITIVITY OF MOUSE<br />

INOCULATION TEST<br />

IDENTIFICATION OF GLOBICATELLA SANGUINIS<br />

ISOLATED FROM PNEUMONIA IN A GOAT<br />

KAIZEN PROCESS APPLIED AT THE DIAGNOSTIC SERVICE<br />

OF FACULTÉ DE MÉDICINE VÉTÉRINAIRE, UNIVERSITÉ DE<br />

MONTREAL<br />

ACCURACY OF PARATUBERCULOSIS ANTIBODY ELISA AT<br />

PREDICTING FECAL SHEDDING OF MYCOBACTERIUM<br />

AVIUM SUBSP PARATUBERCULOSIS IN CATTLE<br />

EVALUATION OF AN ELISA ASSAY AS AN ANCILLARY<br />

METHOD IN A HERD WITH A NATURAL TUBERCULOSIS<br />

INFECTION<br />

VIROLOGICAL SURVEILLANCE AND CHARACTERIZATION<br />

OF AVIAN-LIKE REASSORTANT H1N2 SWINE INFLUENZA<br />

VIRUSES IN SWEDEN<br />

A TOOL TO ASSESS ANTIMICROBIAL RESISTANCE<br />

PATTERNS IN DAIRY FARMS<br />

CHARACTERIZATION OF EXTENDED-SPECTRUM ß-<br />

LACTAMASE (ESBL)-PRODUCING ESCHERICHIA COLI<br />

BOVINE ISOLATES IN NORTHWEST SPAIN<br />

OVINE ABORTION CAUSED BY YERSINIA<br />

PSEUDOTUBERCULOSIS – A CASE REPORT<br />

ANALYSIS OF THE EFFICIENCY OF THE MCMASTER<br />

METHOD IN RAYNAUD’S MODIFICATION IN DETECTION OF<br />

TOXOCARA SP. AND TRICHURIS SP. EGGS IN<br />

CARNIVORES FAECES<br />

MILK AMYLOID A AND ITS USEFULNESS IN THE<br />

LABORATORY DIAGNOSIS OF MASTITIS<br />

RELIABLE AND EARLY DETECTION OF SALMONELLA IN<br />

PIGS<br />

DETECTION OF BVD VIRUS ON PERSISTENTLY INFECTED<br />

CALVES BY REAL TIME REVERSE TRANSCRIPTION PCR<br />

ON EAR NOTCHES


S1-P-25 MARQUES SARA<br />

S1-P-26 MICHALSKI MIROSLAW<br />

S1-P-27 MICHALSKI MIROSLAW<br />

S1-P-28 MELONI D.<br />

S1-P-29 PRITZ-VERSCHUREN SYLVIA<br />

S1-P-30 PROHASKA SARAH<br />

S1-P-31 SATTLER TATJANA<br />

S1-P-32 SCHROEDER CARSTEN<br />

S1-P-33<br />

SCICLUNA<br />

MARIA<br />

TERESA<br />

S1-P-34 KOVAC GABRIEL<br />

S1-P-35 WIDÉN FREDERIK<br />

GENOTYPIC VARIABILITY DETERMINATION OF<br />

PATHOGENIC PROTOTHECA<br />

DETECTION OF SAXITOXINE (PSP MARINE BIOTOXIN) IN<br />

MOLLUSCS BY ELISA<br />

PROFICIENCY TESTING (PT) FOR CHEMICAL<br />

LABORATORIES PERFORMING ANALYSIS OF MEAT AND<br />

MEAT PRODUCTS ORGANISED BY NVRI IN PULAWY<br />

SPECIFIED RISK MATERIAL REMOVAL PRACTICES: CAN<br />

WE REDUCE THE BSE HAZARD TO HUMAN HEALTH<br />

LOW PATHOGENIC AVIAN INFLUENZA VIRUS INFECTIONS<br />

ON POULTRY FARMS IN THE NETHERLANDS<br />

DETECTION OF BRACHYSPIRA HYODYSENTERIAE AND<br />

BRACHYSPIRA PILOSICOLI IN SWISS PIGS USING A<br />

COMBINATION OF CULTURE AND PCR<br />

PRRSV DIAGNOSTICS BY RT-PCR AND DIFFERENT ELISA<br />

SYSTEMS IN SERUM AND ORAL FLUID OF PRRSV<br />

VACCINATED PIGS<br />

RELIABLE DETECTION AND TYPING OF PRRSV USING<br />

MULTIPLEX REAL-TIME RT-PCR<br />

APPLICATION OF A HERD PROGRAMME BASED ON<br />

CONTROL MEASURES AND LABORATORY MONITORING<br />

TO ERADICATE A LEPTOSPIRA OUTBREAK IN A LARGE<br />

DAIRY CATTLE HERD<br />

THE SERUM PROTEIN ELECTROPHORETIC PATTERN IN<br />

CALVES WITH CHRONIC RESPIRATORY DISEASES<br />

DEVELOPMENT OF A BROADLY DETECTING ASSAY FOR<br />

FLAVIVIRUS USING A NOVEL CONCEPT<br />

S1-P-36 O'NEILL RONAN EXTENDING THE SHELF LIFE OF COMMERCIAL ELISA KITS<br />

S1-P-37 POLAK MIROSŁAW P.<br />

S1-P-38 SZCZOTKA MARIA<br />

S1-P-39 SZCZOTKA MARIA<br />

S1-P-40 SZCZOTKA ANNA<br />

S2-P-01 OLOFSON ANN-SOPHIE<br />

S2-P-02 SANZ ANTONIO<br />

S2-P-03 LEBLANC NEIL<br />

S3-P-01 ACHTERBERG RENÉ<br />

S3-P-02 ARENT ZBIGNIEW<br />

S3-P-03 BLOME SANDRA<br />

S3-P-04 SCHELP CHRISTIAN<br />

S3-P-05 KARAMON JACEK<br />

MONITORING OF BOVINE VIRAL DIARRHEA VIRUS (BVDV)<br />

IN DAIRY CATTLE HERDS IN POLAND<br />

DETECTION OF BOVINE LEUKAEMIA VIRUS PROVIRAL<br />

DNA IN DENDRITIC CELLS BY IN SITU PCR<br />

BOVINE BLOOD DENDRITIC CELLS IN CATTLE INFECTED<br />

WITH BOVINE LEUKAEMIA VIRUS (BLV<br />

PORCINE CIRCOVIRUS TYPE 2 IN POSTWEANING PIGS<br />

WITH ANTIBIOTIC NON-RESPONSIVE DIARRHEA<br />

EVALUATION OF FIELD BASED METHODS FOR<br />

DETECTION OF CLASSICAL SWINE FEVER BY PCR<br />

NEW IMMUNOASSAYS FOR DIAGNOSIS OF ASFV BASED<br />

ON VP72: CAPTURE ELISA FOR DETECTION OF IGM<br />

SPECIFIC ANTIBODIES AND PEN-SIDE TEST FOR BLOOD<br />

SAMPLES<br />

PORTABLE PLATFORMS FOR THE DETECTION OF<br />

AFRICAN SWINE FEVER VIRUS TESTED IN FIELD<br />

CONDITIONS IN NORTHERN UGANDA<br />

BEAD-BASED SUSPENSION ARRAY FOR THE<br />

SIMULTANIOUS DETECTION OF ANTIBODIES AGAINS THE<br />

RIFT VALLEY FEVER VIRUS NUCLEOCAPSID AND GN<br />

GLYCOPROTEIN<br />

LIPOPOLYSACCHARIDE-CAPTURE ELISA FOR THE<br />

DETECTION OF LEPTOSPIRA BORGPETERSENII SEROVAR<br />

HARDJO IN SHEEP<br />

EVALUATION OF DIRECT BLOOD POLYMERASE CHAIN<br />

REACTION FOR RAPID DETECTION OF AFRICAN SWINE<br />

FEVER VIRUS<br />

DEVELOPMENT OF A SCHMALLENBERG VIRUS ANTIBODY<br />

ELISA<br />

DETECTION OF ECHINOCOCCUS MULTILOCULARIS IN<br />

FOXES’ FAECES BY PCR WITH THE USE OF DILUTED DNA<br />

SAMPLES – COMPARISON OF DIFFERENT METHODS OF<br />

DNA ISOLATION


S3-P-06 KELLER SELINA<br />

S3-P-07 KÖNIG MATTHIAS<br />

COMPARISON OF CULTURE AND PCR FOR DETECTION<br />

OF MYCOBACTERIUM AVIUM SSP. PARATUBERCULOSIS<br />

IN BOVINE FAECES<br />

HEPATITIS E VIRUS IN DOMESTIC SWINE AND WILD BOAR<br />

FROM GERMANY<br />

S3-P-08 MAGNEE DAMIEN EMERGENCE OF SCHMALLENBERG VIRUS:<br />

S3-P-09 NARDINI ROBERTO<br />

S3-P-10 ŚMIETANKA KRZYSZTOF<br />

S3-P-11 PELETTO SIMONE<br />

S3-P-12 POURQUIER PHILIPE<br />

S3-P-13 RABALSKI LUKASZ<br />

S3-P-14 RANZ ANA<br />

S3-P-15 REID SCOTT M.<br />

S3-P-16 REID SCOTT M.<br />

S3-P-17 KOLBASOV DENIS<br />

S3-P-18 SKRZYPCZAK TERESA<br />

S3-P-19 WELLENBERG GERARD<br />

S3-P-20 MAZZONE PIERA<br />

S3-P-21 MAZZONE PIERA<br />

S3-P-22 LARSKA MAGDALENA<br />

S3-P-23 LARSKA MAGDALENA<br />

S3-P-24 LOEFFEN W.L.A.<br />

S3-P-25 COOLEY W.A.<br />

S3-P-26 MATERNIAK MAGDALENA<br />

S4-P-01 ALTMANN MICHAELA<br />

S4-P-02 ARENT ZBIGNIEW<br />

PRELIMINARY VALIDATION OF A SOLID- PHASE<br />

COMPETITIVE ELISA FOR THE DETECTION OF<br />

ANTIBODIES AGAINST WEST NILE DISEASE VIRUS IN<br />

HORSE SERA<br />

SIMULTANEOUS DETECTION AND DIFFERENTIATION OF<br />

INFLUENZA A VIRUS AND NEWCASTLE DISEASE VIRUS BY<br />

ONE STEP RT-PCR<br />

DOLPHIN MORBILLIVIRUS INFECTION IN A CAPTIVE<br />

HARBOR SEAL (PHOCA VITULINA)<br />

PRELIMINARY VALIDATION OF THE ID SCREEN AFRICAN<br />

SWINE FEVER INDIRECT ELISA BASED ON THREE<br />

RECOMBINANT ASF PROTEINS<br />

ONE STEP RT-PCR FOLLOWED BY MSSCP AS A NOVEL<br />

METHOD FOR A FAST AND RELIABLE DETECTION AND<br />

DIFFERENTIATION OF MIXED INFECTION WITH<br />

NEWCASTLE DISEASE VIRUS OF DIFFERENT ORIGIN<br />

DEVELOPMENT OF MULTISPECIES SEROLOGICAL<br />

ASSAYS TO DETECT ANTIBODIES SPECIFIC FOR<br />

MYCOBACTERIUM BOVIS IN SERUM SAMPLES<br />

FIRST REPORTED LOW PATHOGENCITY AVIAN<br />

INFLEUNZA VIRUS SUBTYPE H9 NFECTION OF DOMESTIC<br />

FOWL IN ENGLAND<br />

DETECTION OF IBV QX IN COMMERCIAL POULTRY<br />

FLOCKS IN THE UNITED KINGDO<br />

APPLICATION OF REVERSE TRANSCRIPTION REAL-TIME<br />

PCR TO DETECT THE SCHMALLENBERG VIRUS GENOME<br />

PREVALENCE OF COXIELLA BURNETII ANTIBODIES IN<br />

SHEEP AND GOATS IN FINLAND<br />

DETECTION OF SCHMALLENBERG VIRUS IN BOVINE<br />

SEMEN BY ONE-STEP REAL-TIME RT-PCR<br />

INTERFERON-GAMMA ASSAY TO DIAGNOSE<br />

MYCOBACTERIUM BOVIS INFECTION IN PIGS<br />

USE OF EXPERIMENTAL JOHNIN IN THE GAMMA-<br />

INTERFERON TEST IN CATTLE INFECTED BY<br />

MYCOBACTERIUM AVIUM SUBSP. PARATUBERCULOSIS:<br />

PRELIMINARY DATA<br />

COMPARISON OF THE PERFORMANCE OF FIVE<br />

DIFFERENT IMMUNOASSAYS TO DETECT SPECIFIC<br />

ANTIBODIES AGAINST EMERGING ATYPICAL BOVINE<br />

PESTIVIRUS<br />

SCHMALLENBERG VIRUS (SBV) IN POLAND –<br />

PRELIMINARY RESULTS<br />

DEVELOPMENT OF A VIRUS NEUTRALISATION TEST TO<br />

DETECT ANTIBODIES AGAINST<br />

VIRAL DIAGNOSIS USING TRANSMISSION ELECTRON<br />

MICROSCOPY SCHMALLENBERG VIRUS<br />

DETECTION OF EQUINE FOAMY VIRUS INFECTIONS IN<br />

HORSES<br />

MOLECULAR CHARACTERIZATION OF MYCOBACTERIUM<br />

AVIUM SUBSP. PARATUBERCULOSIS STRAINS IN A<br />

NATIONAL PARATUBERCULOSIS CONTROL PROGRAM<br />

DEVELOPMENT AND VALIDATION OF A REAL-TIME PCR<br />

ASSAY FOR THE DETECTION OF LEPTOSPIRA<br />

BORGPETERSENII SEROVAR HARDJO IN URINE AND<br />

KIDNEY


S4-P-03 DĄBROWSKA JOANNA<br />

S4-P-04 ISAKSSON MATS<br />

S4-P-05 MARTOS RAICH ALBA<br />

S4-P-06 MELONI DANIELA<br />

S4-P-07 MOYEN JEAN-LOUIS<br />

S4-P-08 NOGAROL CHIARA<br />

S4-P-09 POURQUIER PHILIPPE<br />

S4-P-10 POURQUIER PHILIPPE<br />

S4-P-11 SCHROEDER BJOERN<br />

S4-P-12 ZDYBEL JOLANTA<br />

USEFULNESS OF LIVE/DEAD BACLIGHT BACTERIAL<br />

VIABILITY KIT TYPE 7007 MOLECULAR PROBES FOR<br />

EVALUATION OF VIABILITY ASCARIS SP., TOXOCARA SP.<br />

AND TRICHURIS SP. EGGS ISOLATED FROM SEWAGE<br />

SLUDGE<br />

AUTOMATION OF A CAPTURE PROBE MAGNETIC BEAD<br />

DNA EXTRACTION METHOD FOR 3 GRAM FAECAL<br />

SAMPLES FROM RED FOX INTENDED FOR PCR-<br />

DETECTION OF ECHINOCOCCUS MULTILOCULARIS<br />

EVALUATION OF THREE ELISAS FOR DETECTING SERUM<br />

ANTIBODIES AGAINST MYCOPLASMA HYOPNEUMONIAE.<br />

SEPRION-COATED MICROCANTILEVER SENSORS FOR<br />

PRPSC DETECTION<br />

EVALUATION OF DIFFERENT EXTRACTION METHODS<br />

FOR THE DETECTION OF MYCOBACTERIUM AVIUM<br />

SUBSP. PARATUBERCULOSIS IN BOVINE FAECES<br />

REAL-TIME PCR FOR DETECTION OF CAMPYLOBACTER<br />

JEJUNI, COLI AND LARI IN FOODS: TEST VALIDATION<br />

ACCORDING TO ISO16140:2003<br />

VALIDATION OF THE ID SCREEN SALMONELLA DUBLIN<br />

COMPETITIVE ELIS<br />

ID SCREEN® INTERFERON GAMMA ELISA :<br />

IMPROVEMENT OF ANALYTICAL SENSITIVITY AND<br />

INTRODUCTION OF A STANDARD REFERENCE CONTROL<br />

TO IMPROVE RESULT INTERPRETATION<br />

EFFECTIVE TESTING STRATEGY WITH PRIMAGAM® FOR<br />

TUBERCULOSIS IN HUMAN PRIMATES<br />

COMPARISON OF THE EFFICIENCY OF DIFFERENT<br />

PARASITOLOGICAL DIAGNOSTIC METHODS USED IN<br />

ANALYSIS OF DEHYDRATED SEWAGE SLUDGES


List <strong>of</strong> abstracts (alphabetical order <strong>of</strong> presenting authors)<br />

ACHTERBERG RENÉ S3-P-01<br />

AHOLA HEIKKI S1-P-01<br />

ALTMANN MICHAELA S4-P-01<br />

ARENT ZBIGNIEW S3-P-02<br />

ARENT ZBIGNIEW S4-P-02<br />

BALLAGI ANDREA S1-O-06<br />

BALLAGI ANDREA S1-O-17<br />

BALLAGI ANDREA S1-P-02<br />

BALLAGI ANDREA S1-P-03<br />

BENITO ALFREDO S1-P-04<br />

BLANCHARD BEATRICE S1-P-05<br />

BLANCHARD BEATRICE S1-P-06<br />

BLOME SANDRA S3-P-03<br />

BOSS CHRISTINA S1-O-01<br />

BOSS CHRISTINA S1-O-14<br />

BOSS CHRISTINA S1-P-07<br />

BÖTTCHER JENS S3-O-03<br />

BÖTTCHER JENS S1-P-08<br />

BOUWSTRA RUTH S1-O-03<br />

BOUWSTRA RUTH S3-O-01<br />

CARTER CRAIG N. S3-O-04<br />

CHAINTOUTIS SERAFEIM C. S3-O-07<br />

CHERNYSHOV ANATOLIY S1-P-11<br />

CHERNYSHOVA ELENA S1-P-12<br />

COCCHI MONIA S1-P-13<br />

COOLEY WILLIAM S1-O-09<br />

COOLEY W.A. S3-P-25<br />

CORNAGLIA ESTELA S1-P-14<br />

DĄBROWSKA JOANNA S4-P-03<br />

EGLI C. S3-O-05<br />

EIRAS CARMEN S1-P-15<br />

EIRAS CARMEN S1-P-16<br />

EIRAS CARMEN S1-P-19<br />

FOERSTER CHRISTINE S3-O-09<br />

GAVIER-WIDÉN DOLORES S3-K-01<br />

GEROLIMETTO ELISA S1-P-18<br />

GÓRNA KAMILA S1-O-04<br />

HEUVELINK ANNET S4-O-04<br />

HIRVELÄ-KOSKI VARPU S1-P-20<br />

ISAKSSON MATS S4-P-04<br />

KARAMON JACEK S3-P-05<br />

KELLER SELINA S3-P-06<br />

KOCHANOWSKI MACIEJ S1-P-21<br />

KOLBASOV DENIS S3-P-17<br />

KÖNIG MATTHIAS S3-P-07<br />

KOOI BART S3-O-02<br />

KOSTRZEWA MARCUS S4-K-01<br />

KOVAC GABRIEL S1-P-22<br />

KOVAC GABRIEL S1-P-34<br />

KÜHN TILMAN S1-P-23<br />

LA ROCCA ANNA S3-O-06<br />

LANGEVELD JAN S1-O-10<br />

LARSKA MAGDALENA S3-P-22<br />

LARSKA MAGDALENA S3-P-23<br />

LEBLANC NEIL S2-P-03<br />

LOEFFEN W.L.A. S3-P-24<br />

LOPEZ PABLO S1-O-18<br />

MAGNEE DAMIEN S1-P-24<br />

MAGNEE DAMIEN S3-P-08<br />

MARQUES SARA S1-P-25<br />

MARTOS RAICH ALBA S4-P-05<br />

MATERNIAK MAGDALENA S3-P-26<br />

MAZZONE PIERA S3-P-20<br />

MAZZONE PIERA S3-P-21<br />

MELONI D. S1-P-28<br />

MELONI DANIELA S4-P-06<br />

METREVELI GIORGI S1-P-17<br />

MICHALSKI MIROSLAW S1-P-26<br />

MICHALSKI MIROSLAW S1-P-27<br />

MOYEN JEAN-LOUIS S4-P-07<br />

NARDINI ROBERTO S1-O-05<br />

NARDINI R. S1-P-10<br />

NARDINI ROBERTO S3-P-09<br />

NOGAROL CHIARA S4-P-08<br />

NORTH SARAH S2-O-01<br />

OLOFSON ANN-SOPHIE S2-P-01<br />

O'NEILL RONAN S1-O-12<br />

O'NEILL RONAN S1-P-36<br />

OVERESCH GUDRUN S4-O-05<br />

PELETTO SIMONE S3-P-11<br />

POLAK MIROSŁAW P. S1-P-37<br />

PORQUET-<br />

GARANTO LOURDES S1-O-15<br />

POURQUIER PHILIPPE S3-O-11<br />

POURQUIER PHILIPE S3-P-12<br />

POURQUIER PHILIPPE S4-P-09<br />

POURQUIER PHILIPPE S4-P-10<br />

PRITZ-<br />

VERSCHUREN SYLVIA S1-P-29<br />

PROHASKA SARAH S1-P-30<br />

RABALSKI LUKASZ S3-P-13<br />

RAEBER ALEX S4-O-03


RANZ ANA S3-P-14<br />

REBORDOSA-<br />

TRIGUEROS XAVIER S1-O-11<br />

REID SCOTT M. S3-P-15<br />

REID SCOTT M. S3-P-16<br />

REVILLA-<br />

FERNÁNDEZ SANDRA S1-O-08<br />

RIPP ULRIKE S4-O-01<br />

SANZ ANTONIO S2-P-02<br />

SATTLER TATJANA S1-P-31<br />

SAWYER JASON S4-O-06<br />

SCHELP CHRISTIAN S3-P-04<br />

SCHIRRMEIER HORST S1-O-13<br />

SCHIRRMEIER HORST S3-O-08<br />

SCHROEDER CARSTEN S1-P-32<br />

SCHROEDER BJOERN S4-P-11<br />

SCHWARZ STEFAN S1-K-02<br />

SCICLUNA MARIA TERESA S1-P-33<br />

SKRZYPCZAK TERESA S3-P-18<br />

SOCHA WOJCIECH S2-O-03<br />

SOLDAN ANDREW S2-K-01<br />

STEINRIGL ADOLF S3-O-10<br />

STRUTZBERG-<br />

MINDER KATRIN S1-O-02<br />

SZCZOTKA MARIA S1-P-38<br />

SZCZOTKA MARIA S1-P-39<br />

SZCZOTKA ANNA S1-P-40<br />

ŚMIETANKA KRZYSZTOF S3-P-10<br />

VALLS LAURA S2-O-04<br />

VAN DER POEL WIM H. M. S1-K-01<br />

VAN MAANEN KEES S1-O-07<br />

VAN MAANEN KEES S1-P-09<br />

VILLA ALEIDA S1-O-15<br />

VRANCKEN ROBERT S2-O-02<br />

WELLENBERG GERARD S3-P-19<br />

WIDÉN FREDERIK S1-P-35<br />

WRAGG PETER S4-O-02<br />

ZDYBEL JOLANTA S4-P-12

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!