12.07.2015 Views

Reproduction, Fertility and Development - the Society for ...

Reproduction, Fertility and Development - the Society for ...

Reproduction, Fertility and Development - the Society for ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Reproduction</strong>, <strong>Fertility</strong> <strong>and</strong> <strong>Development</strong>An international journal <strong>for</strong> <strong>the</strong> publication o<strong>for</strong>iginal work, review <strong>and</strong> comment in <strong>the</strong> fields ofreproductive biology, reproductiveendocrinology <strong>and</strong> development biology, including puberty, lactation <strong>and</strong> fetal physiology when <strong>the</strong>y fall within <strong>the</strong>sefields. The Journal will accept papers dealing with biochemistry, cell biology, molecular biology, endocrinology, immunology<strong>and</strong> genetic sex determination of animals including humans, but only if <strong>the</strong>se are related to reproduction, fertility ordevelopment.This journal is one of<strong>the</strong> Australian Journals ofScientific Research published by CSIRO PUBLISHING in cooperation with<strong>the</strong> Commonwealth Scientific <strong>and</strong> Industrial Research Organization (CSIRO) <strong>and</strong> <strong>the</strong> Australian Academy of Science.Editorial policy <strong>for</strong> <strong>the</strong> series is developed by a Board of St<strong>and</strong>ards appointed jointly by CSIRO <strong>and</strong> <strong>the</strong> Academy ofScience.CSIRO PUBLISHING requires that all authors of a multi-authored paper agree to its submission. The Journal has used itsbest endeavours to ensure that work published is that of <strong>the</strong> named authors except where acknowledged <strong>and</strong>, through itsreviewing procedures, that any published results <strong>and</strong> conclusions are consistent with <strong>the</strong> primary data. It takes no responsibility<strong>for</strong> fraud or inaccuracy on <strong>the</strong> part of<strong>the</strong> contributors.Editorial Advisory CommitteeAcceptance ofpapers is <strong>the</strong> responsibility of<strong>the</strong> Managing Editor in consultation with <strong>the</strong> Editorial Advisory Committee onbehalfof<strong>the</strong> Board of St<strong>and</strong>ards. Committee members may advise <strong>the</strong> Managing Editor on <strong>the</strong> selection ofreferees <strong>and</strong> mayadjudicate in <strong>the</strong> case of conflicting or adverse reports. All papers are peer reviewed.Chairperson:G. B. Martin, University ofWestern AustraliaMembers: D. T. Armstrong, University ofAdelaideS. G. Mat<strong>the</strong>ws, University ofToronto1. E. Harding, University ofAuckl<strong>and</strong>1. Hinds, CSIRO Sustainable Ecosystems, Canberrap. 1. Kaye, University of Queensl<strong>and</strong>, BrisbaneW M. C. Maxwell, University of SydneyM. F. Pera, Monash Institute of<strong>Reproduction</strong> <strong>and</strong> <strong>Development</strong>, MelbourneAdvisory PanelA. M. Carter (Denmark); 1. R. G. Challis (Canada); Y. Combarnous (France); B. Fadem (USA); A. P. F. Flint (UK); W W Hay(USA); K. M. Henderson (New Zeal<strong>and</strong>); 1. 1. Guillette (USA); 1. Huhtaniemi (Finl<strong>and</strong>); B. Jegou (France); P. M. Motta(Italy); T. Nagai (Japan); E. Nieschlag (Germany); C. Pholpramool (Thail<strong>and</strong>); 1. D. Skinner (South Africa); 1. F. Smith (NewZeal<strong>and</strong>); D. G. Whittingham (UK); R. Yanagimachi (USA)Managing Editor:Subscription rates <strong>for</strong> 2002Institution print & online:Institution online only:Personal print & online:Personal online only:C. M. MyersAustralian <strong>and</strong> New Zeal<strong>and</strong> customers pay in AU$. All o<strong>the</strong>rcustomers pay in US$. Prices include air freight delivery.Special rates are available <strong>for</strong> society members. Visit:www.publish.csiro.auljournals/rfd8 issues per year; 4 archival print issues plus supplement.ISSN 1031-3613© CSIRO Australia 2002.A$$550$480$120$115US$$625$550$150$115The Instructions to Authors are published in <strong>the</strong> first issue ofeachvolume. Copies are available on request. In<strong>for</strong>mation about <strong>the</strong>Journal, including <strong>the</strong> Instructions to Authors, is available onWorld Wide Web at URLhttp://www.pubIish.csiro.au/journals/rfdAll inquiries <strong>and</strong> manuscripts should be <strong>for</strong>warded to:The Managing Editor<strong>Reproduction</strong>, <strong>Fertility</strong> <strong>and</strong> <strong>Development</strong>CSIRO PUBLISHINGPO Box 1139 (150 Ox<strong>for</strong>d Street)Collingwood, Victoria 3066, AustraliaInternational telephone 613 9662 7629; facsimile 613 9662 7611Email: publishing.rfd@csiro.auSOCIETY FORREPRODUCTIVE BIOLOGYPROGRAM AND ABSTRACTSTHIRTY-THIRDAnnual Scientific Meeting22-25 September 2002Adelaide Convention Centre, Adelaide, SAFivnt cover images. Main photograph: injection of DNA into <strong>the</strong> pronucleusofa centrifuged porcine zygote; courtesy ofAndrew French, Centre <strong>for</strong> EarlyHuman <strong>Development</strong>, Institute of <strong>Reproduction</strong> <strong>and</strong> <strong>Development</strong>, MonashUniversity. Inset: newborn tammar wallaby, Maeropus eugenii, attached to <strong>the</strong>teat in <strong>the</strong> pouch; courtesy ofLyn Hinds <strong>and</strong> CSIRO Sustainable Ecosystems.4:~i~1~- ~C SIR 0 Academy of SciencePermission to photocopy items from this journal is granted byCSIRO to libraries <strong>and</strong> o<strong>the</strong>r users registered with <strong>the</strong> CopyrightClearance Center (CCC), provided that <strong>the</strong> appropriate fee <strong>for</strong>each copy is paid directly to <strong>the</strong> CCC, 222 Rosewood Drive,Danvers, MA 01923, USA. For copying in Australia, CSIROPUBLISHING is registered with Copyright Agency Limited,Level 19, 157 Liverpool Street, Sydney, NSW 2000. Specialrequests should be addressed to <strong>the</strong> Managing Editor,<strong>Reproduction</strong>, <strong>Fertility</strong> <strong>and</strong> <strong>Development</strong>, PO Box 1139 (150Ox<strong>for</strong>d Street), Collingwood, Victoria 3066, Australia.Copyright © <strong>Society</strong> <strong>for</strong> Reproductive Biology, 2002ISSN 1031-3613


The <strong>Society</strong> <strong>for</strong> Reproductive Biology(SRB)acknowledges support of <strong>the</strong> following 2002 Conference Sponsors:<strong>Society</strong> <strong>for</strong>Reproductive Biology (SRB)PEST ANIMAL CONTROL CRCCRC FOR INNOVATIVEDAIRY PRODUCTSTABLE OF CONTENTSPrevious SRB/ASRB Office Bearers/Awardees 4CRCCORPORATE SPONSORSNovo Nordisk Pharmaceuticals(Satchel <strong>and</strong> Dinner Sponsor)GlaxoSmithKline (Dinner Sponsor)GlaxoSmithKline2002 SRB Office Bearers 5SRB Student In<strong>for</strong>mation 6Adelaide Convention Centre Floorplan <strong>and</strong> DelegateIn<strong>for</strong>mation 7Trade Delegate In<strong>for</strong>mation 10Eli Lilly (Welcome Function Sponsor)Joint SRB/ESA Program Details 15Serono (Junior Scientist Award Sponsor)(SeroncVSRB Program - Session Details 33Abstracts (in order of presentation) 43<strong>Reproduction</strong>, <strong>Fertility</strong> <strong>and</strong> <strong>Development</strong>Author Index 93Joint SRB/ESA Program at a Glance 96CONFERENCE SECRETARIATAustralian Science Network Pty Ltd (ASN)Unit 21 B Balnarring Village Centre(PO Box 200) Balnarring, VIC 3926Tel: +61 3 5983 2400 Fax: +61 3 5983 2223Email: mp@asnevents.net.au23


<strong>Society</strong> <strong>for</strong> Reproductive BiologylAustralian <strong>Society</strong> <strong>for</strong> Reproductive Biology<strong>Society</strong> <strong>for</strong> Reproductive BiologyPast Office Bearers & AwardeesGODING /JUNIORYEAR CHAIRMAN TREASURER SECRETARY FOUNDERS SCIENTISTLECTURE WINNER 2002 Office Bearers1969-701970-71RG Wales JR GodingTJ Robinson1971-72 Chairman John Hearn1972-73 Honorary Secretary Rob Gilchrist1973-74Honorary TreasurerChris OINeiliIG White BM Bindon1974-75IACummin Committee Members Graham Jenkin (Deputy Chair)CWEmmens1975-76 WHansel Peter Kaye1976-77 DT Baird Kate Lovel<strong>and</strong>1977-78TD GloverCamilla Myers (ad hoc)GM Stone IACumming1978-79CH T ndale-BiscoeAnne TurnerDM de Kretser1979-80GMH ThwaitesPostgraduate Representative Christine White1980-81 GHMcDowell KPMcNattRJ ScaramuzziCommun. OfficerlWebmaster Jim Cumminsl Michael Sinosich1981-82BK Follett PJ LufenJSA Committee Chair Alan Tilbrook1982-83 NWMoore RJ Rodger &BG Miller J Wilson SRB Secretariat Australian Convention <strong>and</strong> Travel ServiceTK RobertsCBGow1983-84BM Bindon SP FlahertJKFindlay1984-85 A Bellve C O'NeillJMCummins1985-86 CD NancarrowBM Bindon1986-87 WHansel LJ WiltonG Evans1987-88HG Bur er A Sto'anoff Chair Kate Lovel<strong>and</strong>BP Setchell LA Hinds1988-89FW Bazer MB HarveMembers Arun Dharmarajan1M Shelton1989-90 GD Thorburn AHTorne Ann DrummondJC Rodger GB Martin1990-91 RMMoor HMassa Sarah MeachemPLKaye1991-92CR Austin DKGardner Sarah RobertsonRJ Scarammuzzi CG Tsonis1992-93JK Findlay SWWalkden- Andrew SinclairLABrownSalamonsenAnne Turner1993-94 GCLi ins CMMarkeAO Trounson LJ Wilton1994-95 I Huhtaniemi MJ Hotzel & SMPHedgerMcDou all1995-96RF Seamark JD Curlewis1996-97No Lecture S Robinson1997-98JGThompson F Bronson M Jas erMB RenfreeChair Ray RodgersMB Harvey1998-99 DM de Kretser M Panteleon Members Rob Gilchrist2000-01 R Short C Gargett Wendy IngmanJHeam PKaye T Lavranos D Albertini WIngman Hamish Eaton2001-02 R Gilchrist A TrounsonTony RobertsJeremy ThompsonESA Program Organising Committee Chair: Leon BachBP Setchell BJWaddellSRB Program Organising Committee 2002RF Seamark I van Wezel Local Organising Committee (ESAlSRB) 20024 5


SOCIETY FOR REPRODUCTIVE BIOLOGYStudent MembersINFORMATION FOR DELEGATES AND PRESENTERSThere are three events at <strong>the</strong> 2002 SRB Annual Conference that students should not miss!All events are being held on Monday 23 rd September.RIVE.RIU\.HKCARPARKfESTIVAL DRIVE1pm Student Lunch Lecture Adelaide Convention CentreProfessor Alan Trounson will be participating in a roundtable discussion specifically <strong>for</strong> students,following his Founders Lecture. All students are welcome to attend, <strong>and</strong> a light lunch will beprovided.7pm SRB Students Annual Meeting University ofAdelaide Staff ClubThis is a great opportunity to meet fellow students working in <strong>the</strong> field of reproductive biology, <strong>and</strong>voice your opinions on how we can improve your experience of SRB student membership. Themeeting will be held right be<strong>for</strong>e <strong>the</strong> Student BBQ at <strong>the</strong> University of Adelaide Staff Club.7.30pm SRB/ESA Student BBQ University ofAdelaide Staff ClubAs students, we know <strong>the</strong> value of a cheap meal <strong>and</strong> night out! For only $30 per person, <strong>the</strong>SRB/ESA Local Organising Committee is providing a full buffet dinner, drinks <strong>and</strong> entertainment.Donlt miss this chance to meet <strong>and</strong> have a great night with o<strong>the</strong>r SRB <strong>and</strong> ESA students, <strong>and</strong>network with o<strong>the</strong>r conference delegates working in your field.WANT TO BECOME AN SRB STUDENT MEMBER?For only $35 per year, membership gives you:• Member registration discount at SRB Annual Scientific Meetings• Free SRB Abstract Booklets <strong>and</strong> Newsletters• Eligibility <strong>for</strong> SRB <strong>and</strong> RFD Travel Awards <strong>and</strong> <strong>the</strong> Junior Scientist Award• Reduced purchase price on: <strong>Reproduction</strong>, <strong>Fertility</strong> <strong>and</strong> <strong>Development</strong> <strong>and</strong> <strong>Reproduction</strong>• Access to a professional network in <strong>the</strong> field of Reproductive BiologyPlease see your Postgraduate Student Representative, Christine White, <strong>for</strong> an application<strong>for</strong>m. Email: christine.white@med.monash.edu.auNORTH TERRACELEVEL ONE//-- -"/ "",,-'-.../ (/ \iHVATT )OO"COUOT Il'MW.__------------') ~-----"REPRODUCTION, FERTILITY & DEVELOPMENTTravel Award 2003Presented annually to an SRB member PhD student or junior postdoc with <strong>the</strong> best eligiblemanuscript published in RFD in <strong>the</strong> year prior to 31 st March 2003 (application closing date).Comprises $1000 towards overseas conference travel <strong>and</strong> a year's subscription to RFD.Applications <strong>for</strong>ms at http://www.publish.csiro.au/journals/rfdMore info at Students Annual Meeting.The Speaker Preparation RoomConference Room 3 is being used as <strong>the</strong> speaker preparation room throughout <strong>the</strong> event.Networked computers in this room will allow MS Powerpoint presentations loaded here to beshown in any of <strong>the</strong> session rooms. Technicians <strong>and</strong> assistants will be in attendance in <strong>the</strong> room,<strong>and</strong> speakers are encouraged to load <strong>the</strong>ir presentations as soon as possible to avoid last minuterushes. Carousels will also be in this room <strong>for</strong> <strong>the</strong> pre-loading of any 35mm slides but <strong>the</strong>se mustbe reserved in advance, as it is not <strong>the</strong> st<strong>and</strong>ard or preferred means of presentation.6 7


Abbott Laboratories Site Nos: 73&74KURNELL NSW 2230Phone: 02 9668 1850www.abott.comAbbott Laboratories is a global diversifiedcompany dedicated to <strong>the</strong> discovery,development, manufacture <strong>and</strong> marketing ofhealthcare products <strong>and</strong> services. Abbott withReductil, a new weight management drug waslaunched into <strong>the</strong> Australian market in February2002, <strong>and</strong> Gopten, an ACE Inhibitor, are pleasedto be associated with <strong>the</strong> ESA & SRB <strong>and</strong> ADS &ADEA conferences 2002.Alphapharm Site Nos: 50-53GLEBE NSW 2037Phone: 02 9298 3953Fax: 02 9566 4686www.alphapharm.com.auAlphapharm is one of Australia's leadingpharmaceutical companies. Alphapharm develop,manufacture, market <strong>and</strong> distribute prescription<strong>and</strong> pharmacy-only medicines. While Alphapharmspecialises in bringing patent-expired medicinesto market, which allow people save money,Alphapharm have a pipeline of innovativeproducts in development. Recent introductionsinto <strong>the</strong> Australian market include Campral, Bicor<strong>and</strong> Diabex 1000. Alphapharm is part of MerckKGaA.Astrazeneca Pty Ltd Site Nos: 7&8NTH RYDE NSW 2113Phone: 02 9978 3882Fax: 02 99783730www.astrazeneca.comAstraZeneca is one of <strong>the</strong> world1s leadingpharmaceutical companies. It has a strongresearch base <strong>and</strong> an extensive product portfoliodesigned to meet <strong>the</strong> medical needs of a broadrange of patients. The current AZ CardiovascularPortfolio includes Atac<strong>and</strong>, Zestril, Plendil,Betaloc, Tenormin, Inderal <strong>and</strong> Imdur. A majorresearch <strong>and</strong> development ef<strong>for</strong>t is currentlyunderway aiming to bring to market exciting new<strong>the</strong>rapies in <strong>the</strong> areas of hypercholesterolaemia<strong>and</strong> thrombosis. AstraZeneca is committed tofinding tomorrow's medical cures.CONFERENCE TRADE EXHIBITORSAtCor Medical Pty Ltd Site No: 17WEST RYDE NSW 2114Phone: 02 9874 8761Fax: 02 9874 9022www.atcormedical.comAtCor Medical market <strong>the</strong> innovative SphygmoCorrange of cardiovascular diagnostic devices. TheSphygmoCor system allows you to non-invasivelydetect diabetic patients who are at risk of heartattack <strong>and</strong> stroke. Now you can see, monitor <strong>and</strong>manage <strong>the</strong> key central processes driving <strong>the</strong>cardiovascular risk of your patients.Aventis Pharma Site Nos: 41-49LANE COVE NSW 2066Phone: 02 9422 6416Fax: 02 9422 6418www.aventis.comAventis Pharma is one of <strong>the</strong> world1s leadingpharmaceutical companies. With a strong focuson Diabetes, Aventis Australia has invested over$16 Million in <strong>the</strong> past 2 years to diabetesresearch alone, <strong>and</strong> has planned to contributeano<strong>the</strong>r $15Million in <strong>the</strong> next 2yrs again solely to<strong>the</strong> prevention <strong>and</strong> management of diabetes.Bristol Myers Squibb Site No: 10.2NOBLE PARK VIC 3174Phone: 03 9213 4000www.bms.comBristol-Myers Squibb is a leading diversifiedworldwide health care company with <strong>the</strong> missionto develop <strong>and</strong> market innovative products thatextend <strong>and</strong> enhance human life. The company isa leading maker of innovative <strong>the</strong>rapies <strong>for</strong>cardiovascular, metabolic <strong>and</strong> infectious diseases,central nervous system <strong>and</strong> dermatologicaldisorders, <strong>and</strong> cancer.DSL Australia Pty Ltd Site No: 21BAULKHAM HILLS NSW 2153Phone: 02 9680 7200Fax: 0296807255www.DSLabs.comDSL (Diagnostic Systems Laboratories) is arecognized leader in endocrine diagnosticsinvolved in <strong>the</strong> development, manufacture <strong>and</strong>distribution of immuno-diagnostic test kits. DSLmanufacture <strong>and</strong> distribute approximately 120products which can be classified into <strong>the</strong> follOWingareas: <strong>and</strong>rogen assessment, fertility <strong>and</strong>reproductive function, disease markers, thyroidfunction, bone <strong>and</strong> mineral metabolism, growthfactors, metabolism, <strong>and</strong> infectious diseases.Eli Lilly Site No: 25-30WEST RYDE NSW 2114Phone: 02 9325 4486Fax: 02 9325 4573www.lilly.comEli Lilly <strong>and</strong> Company is a leader in diabetesresearch <strong>and</strong> has been since producing <strong>the</strong> firstcommercially available insulin in 1923. This yearEli Lilly introduced Actos (pioglitazone) toAustralia, a type 2 diabetes treatment that targetsinsulin resistance. Also included in <strong>the</strong> diabetesportfolio is <strong>the</strong> Humalog range of rapid actinginsulin analogs. Eli Lilly Australia is part of <strong>the</strong>global Eli Lilly <strong>and</strong> Company, a research-basedpharmaceutical corporation dedicated to creating<strong>and</strong> delivering innovative health care solutionsaimed at improving quality of life. Eli Lilly Australiawas established in 1960, employs 403 people <strong>and</strong>invests more than $22.7 million annually inAustralian research in <strong>the</strong> critical disease states inwhich it operates; neuroscience, oncology,infectious diseases, endocrinology, includingosteoporosis <strong>and</strong> diabetes, <strong>and</strong> cardiovasculardisease.Genzyme Australasia Site No: 9PO Box 6207, Baulkham Hills Business CentreBAULKHAM HILL BIC NSW 2153Phone: 02 9680 8383Fax: 0296342777www.genzyme.com.auThyrogen® (thyrotropin alfa - rch) is arecombinant <strong>for</strong>m of TSH developed <strong>for</strong> use inthyroid cancer patients that allows patients toremain on thyroid hormone suppression <strong>the</strong>rapythroughout <strong>the</strong> course of periodic testing <strong>for</strong>recurrence of cancer or metastases, <strong>the</strong>rebyavoiding <strong>the</strong> debilitating symptoms ofhypothyroidism associated with thyroid hormonewithdrawal.GlaxoSmithKline Site Nos: 58-63BORONIA VIC 3155Phone: 03 9721 4314Fax: 039721 4333www.gsk.comGlaxoSmithKline (GSK) Australia is one ofAustralia1s largest pharmaceutical <strong>and</strong> healthcarecompanies. GSK has four main sites in Australia,employing more than 1500 people. It is Australia'slargest vaccine manufacturer <strong>and</strong> a leadingsupplier of medicines <strong>for</strong> asthma, bacterial <strong>and</strong>viral infections, depression, migraine,gastroenterological disease, epilepsy, smokingcessation <strong>and</strong> pain relief. More than 16 millionAustralians rely on at least one of GSK'smedicines, vaccines or consumer healthcareproducts. The company invests more than $25million in R&D each year, making it one ofAustralia's top 20 R&D investors. GSK Australiaplays a significant role in <strong>the</strong> globalpharmaceutical supply chain - exporting 60% ofproduction to more than 50 countries, with totalexport earnings totalling $197 million in 2001.IPSEN Pty Ltd Site No: 11MT WAVERLEY VIC 3150Phone: 03 9550 1843Fax: 03 9545 3513www.ipsen.com.auIpsen's strong history of creative <strong>and</strong> successfuldrug discovery illustrates its commitment to <strong>the</strong>development of highly specialised products tomeet specific needs in areas such as neurology<strong>and</strong> endocrinology. An international companybased in Europe, Ipsen represents a relativelynew name in <strong>the</strong> Australian pharmaceuticalindustry <strong>and</strong> currently markets Somatuline® LA(Ianreotide) - a somatostatin analogue indicated<strong>for</strong> <strong>the</strong> treatment of acromegaly. Somatuline LAprovides an alternative in somatostatin analogues<strong>and</strong> has been used extensively in <strong>the</strong> UK <strong>and</strong>Europe over recent years.1011


Mayne Pharma Site No: 13PARKVILLE VIC 3052Phone: 03 8341 5000Fax: 03 8341 5050www.faulding.comMayne's pharmaceutical division is represented inmore than 50 countries worldwide, with a focus ondeveloping, manufacturing <strong>and</strong> marketing genericinjectable <strong>and</strong> oral pharmaceuticals, primarily <strong>for</strong><strong>the</strong> hospital market. Mayne Pharma Australia hasalso established a global reputation <strong>for</strong> marketinginnovative patent protected in-licensed products.Through our specialised care team we marketproducts in <strong>the</strong> following <strong>the</strong>rapeutic areas;Transplant Medicine, Oncology, Endocrinology,Urology, Infectious Diseases, Neurology <strong>and</strong>Psychiatry.Merck Sharp & Dohme Site No: 12SOUTH GRANVILLE NSW 2142Phone: 02 9795 9185Fax: 0297959602www.msd-australia.com.auMerck Sharp & Oohme (MSO) Australia is aresearch based pharmaceutical company <strong>and</strong> hasbeen looking after <strong>the</strong> health of Australians since1952, a history of which MSD is extremely proud.MSD invests a considerable amount intoAustralian research <strong>and</strong> development. Over 90%of MSD products are manufactured <strong>and</strong> packagedin Australia, which leads to new investment <strong>and</strong>infrastructure opportunities. The ongoingcommitment of MSO in Australia is evidenced byits achievement in becoming <strong>the</strong> largestpharmaceutical exporter in <strong>the</strong> country, exportingto more than 16 countries throughout <strong>the</strong> world.Novartis Pharmaceuticals Site No:10NTH RYDE NSW 2113Phone: 02 9805 3555Fax: 02 9888 3408www.pharma.novartis.comNovartis is a global life sciences company <strong>for</strong>medin 1997 through <strong>the</strong> merger of S<strong>and</strong>oz <strong>and</strong> Ciba.In Australia, Novartis Pharmaceuticals is <strong>the</strong>leading supplier of pharmaceutical products tohospitals. The company boasts a broad portfolioof p~oducts in <strong>the</strong> fields of neurology,transplantation, oncology, endocrinology <strong>and</strong>gastroenterology. Novartis has recently increasedits focus on <strong>the</strong> areas of Oncology,Transplantation <strong>and</strong> Opthalmics by <strong>the</strong> <strong>for</strong>mationof business units to manage <strong>the</strong>se specialtyproducts. The company is also committed toextensive clinical research within AustralianInstitutions. Some well known br<strong>and</strong>s of <strong>the</strong>Novartis Oncology business unit include Femara® (Ietrozole), S<strong>and</strong>ostatin LAR® (octreotide),Zometa® (zoledronic acid) <strong>and</strong> <strong>the</strong> recentlydeveloped tyrosine kinase inhibitior, GliveC®(imatinib). The Novartis representatives presentat this meeting would be happy to answer anyquestions related to Novartis Oncology products.NovoNordisk PharmaceuticalsSite Nos: 1-6 & 64-72PO Box 6086PARRAMATTA BUSINESS CENTRE NSW2150Phone: 02 9890 0723Fax: 02 9683 3029www.novonordisk.com.auNovo Nordisk recognizes <strong>the</strong> commitment thatmembers of <strong>the</strong> Endocrine <strong>Society</strong> of Australiamake towards patients with diabetes <strong>and</strong>endocrinological disease. The Novo Nordiskcorporate goal is to improve <strong>the</strong> way people withdiabetes live <strong>and</strong> work. We continue to be at <strong>the</strong><strong>for</strong>efront of diabetes discovery <strong>and</strong> are committedto <strong>the</strong> development of new <strong>and</strong> innovativetreatments, in partnership with sophisticatedpatient focused delivery systems. Our goal is tomeet <strong>the</strong> individual needs of patients byrecognizing that all patients are not <strong>the</strong> same <strong>and</strong>require different solutions to <strong>the</strong> everydaychallenges <strong>the</strong>y face.Pfizer Pty Ltd Site Nos: 31-34WEST RYDE NSW 2114Phone: 02 9850 3333Fax: 02 9850 3146www.pfizer.comPfizer is <strong>the</strong> worldls leading research based healthcare company. They discover, develop,manufacture <strong>and</strong> market innovative medicaltreatments <strong>for</strong> humans <strong>and</strong> animals. Ranked asone of <strong>the</strong> worldls top 10 companies, Pfizer willinvest US$5 billion in research <strong>and</strong> development<strong>for</strong> conditions including heart disease, arthritis <strong>and</strong>diabetes. Pfizer is dedicated to improving <strong>the</strong>management of people with CV risks <strong>and</strong>associated complications.Pharmacia Site Nos: 18&19RYOALMERE NSW 2116Phone: 02 9848-3150Fax: 02 9848 3333www.pharmacia.comPharmacia Corporation is committed to improvinghealth <strong>and</strong> wellbeing <strong>for</strong> people around <strong>the</strong> world<strong>and</strong> is recognised as an innovative leader in anumber of <strong>the</strong>rapeutic areas, includingarthritis/inflammation, gastroenterology, CNS,ophthalmology, oncology, neurology, women'shealth, pain management, infectious diseases <strong>and</strong>metabolic diseases. Pharmacia Australia isinvolved in <strong>the</strong> research, <strong>and</strong> development of newdrugs <strong>for</strong> <strong>the</strong> company <strong>and</strong> is passionate in itscommitment to providing <strong>the</strong> highest qualityproducts to foster good health <strong>and</strong> wellnessthroughout <strong>the</strong> community.Sanofi-Syn<strong>the</strong>labo Site No: 24LANE COVE NSW 2066Phone: 02 9937 1706Fax: 02 9937 1755www.sanofLcom.auResulting from a series of mergers with severalpharmaceutical companies, Sanofi-Syn<strong>the</strong>labo isa leading pharmaceutical group with itsoperational head office in France. It is one of <strong>the</strong>new French companies operating on a globalscale, which has achieved a high level ofexcellence developed over <strong>the</strong> years. Ranking 6thin Europe <strong>and</strong> amongst <strong>the</strong> top 20 pharmaceuticalcompanies worldwide, Sanofi-Syn<strong>the</strong>labo is <strong>the</strong>flagship of French pharmaceutical research <strong>and</strong>industry around <strong>the</strong> world.Seronosponsorwww.serono.com.auWe are one of <strong>the</strong> world's top three biotechnologycompanies focussing on four core <strong>the</strong>rapeuticareas; Growth & Metabolism, Reproductive Health& Multiple Sclerosis. Serono invests more than20% of its sales in research <strong>and</strong> development.We have over 30 projects in preclinical or clinicaldevelopment, 13 new molecules <strong>and</strong> over 20discovery projects aiming to identify new drugc<strong>and</strong>idates. We aim <strong>for</strong> continuous productimprovement <strong>and</strong> <strong>the</strong> support of growthprofessionals including medical <strong>and</strong> patienteducation. We are proud of supporting <strong>the</strong> workof Australian doctors in developing <strong>the</strong> "Hormones& Me" series of booklets. One.click®, Serono'snew autoinjection device <strong>for</strong> <strong>the</strong> delivery of Saizenis designed with patient convenience in mind. It is<strong>the</strong> only such device which injects <strong>and</strong> deliversgrowth hormone toge<strong>the</strong>r with only one action.Servier Laboratories Site Nos: 54-57HAWTHORN VIC 3122Phone: 03 8823 7333Fax: 03 9822 4546www.servier.comServier is a privately owned Frenchpharmaceutical company that has grown fromhumble beginnings in 1955 with 9 employees, tomore than 13,000 employees today. Servier'sproducts are <strong>the</strong> result of in-house discovery <strong>and</strong>development primarily in <strong>the</strong> areas of metabolic<strong>and</strong> cardiovascular disease. 250/0 of Servier'srevenues are reinvested annually in research <strong>and</strong>development.Servier Australia's commercial interests arepresently in hypertension <strong>and</strong> heart disease(Coversyl - perindopril, Coversyl Plus ­perindopril/indapamide <strong>and</strong> Natrilix SR 1.5mg ­indapamide), diabetes (Diamicron MR -gliclazide)<strong>and</strong> disseminated malignant melanoma(Muphoran - fotemustine).1213


ESA & SRB Annual Scientific Meeting 2002ESA & SRBJoint ProgramAdelaide Convention CentreSunday, 22 September 2002toWednesday, 25 September 2002Sunday, 22 September 2002New Horizons in Reproductive BiologySymposium Conference Room 24:00pm EPIGENETIC PHENOMENA IN REPRODUCTIVE BIOLOGYMichael Holl<strong>and</strong>4:30pm EPIGENETIC INHERITANCE IN MAMMALSEmma Whitelaw5:00pm SUPRAMOLECULAR SCIENCE TO NANOTECHNOLOGYLis<strong>and</strong>ra Martin5:30pm GRAPES AND WINE FLAVOURPatrick WilliamsExhibition FoyersRiverview 3Eli Lilly Welcome FunctionWomen in Endocrinology Function4:00pm - 6:00pm6:00pm - 7:30pm7:00pm - 8:30pmRiverview 2ESA Council dinner <strong>and</strong> Meeting8:00pm - 11 :30prn1415


Monday, 23 September 2002Japan-Australia PlenaryPlenary Lecture Conference Hall 0 8:45am - 9:45am8:45am THE DISCOVERY OF GHRELIN - GROWTH HORMONE AND APPETITE STIMULATING HORMONEMasayasu KojimaSRB Oral Presentations - Session 1 - Male <strong>Fertility</strong> <strong>and</strong> Function 1Conference Room 108:45am - 10:00am8:30am IRON IS HIGHLY GENOTOXIC TOWARD HUMAN SPERMATOZOADennis Sawyer8:45am MORPHOLOGICAL MANIFESTATION OF Y CHROMOSOMAL MICRODELETIONS IN THE HUMANDr Marc Luetjens9:00am LEPTIN INHIBITS BASAL BUT NOT STIMULATED TESTOSTERONE PRODUCTION BY THEIMMATURE MOUSE TESTISDr Jim McFarlane9:15am THE ROLE OF FGFR SIGNALLING IN SPERM TAIL DEVELOPMENTLeanne Cotton9:30am REDOX REGULATION OF SPERM FUNCTIONMark Baker9:45am DOES CALCIUM HAS A ROLE IN SPERM HEAD ROTATION TO T-SHAPE ORIENTATION DURINGCAPACITATION? A TALE OF TWO MARSUPIAL SPECIES, THE BRUSHTAIL POSSUM (TRICHOSURUSVULPECULA) AND THE TAMMAR WALLABY (MACROPUS EUGEN/~.Dr Kuldip Sidhu..) SRB Oral Presentations - Session 2 - Embryo <strong>Development</strong> <strong>and</strong> ImplantationConference Room 28:45am - 10:15am8:30am PROLACTIN IS A PREIMPLANTATION GROWTH FACTOR IN MICEDr Peter Kaye8:45am REAL-TIME PCR EXPRESSION OF HYPOXIA-INDUCIBLE FACTORS DURING BOVINEPREIMPLANTATION EMBRYO DEVELOPMENT9:00am9:15am9:30am9:45am10:00amAlex<strong>and</strong>ra HarveyEFFECT OF IN VITRO OXYGEN CONCENTRATION ON GLUCOSE TRANSPORTER-1 EXPRESSION INTHE MOUSE BLASTOCYSTKaren KindA COMPARISON OF STAINING METHODS USED TO DETERMINE NUMBERS OF CELLS IN PIGBLASTOCYSTSChristopher GrupenPARACRINE PERTURBATIONS ASSOCIATED WITH BOVINE NUCLEAR TRANSFER PREGNANCIESSusan RavelichSURVIVAL OF FRESH AND VITRIFIED SHEEP IVP AND CLONED EMBRYOS AFTER EMBRYOTRANSFERTeija PeuraABNORMAL TROPHOBLAST MHC-I EXPRESSION AND ASSOCIATED ENDOMETRIAL LYMPHOCYTICRESPONSE IN BOVINE NUCLEAR TRANSFER PREGNANCIES.Jonathan HillESA Servier Awardoral Conference Hall 0 9:45am - to:OOam9:45am SIGNALLING THROUGH THE SMAD PATHWAY BY INSULIN-LIKE GROWTH FACTOR BINDING PROTEIN­3 IN BREAST CANCERDr Susan FanayanExhibition Halls F & G16Morning Tea10:00am -10:30amESA Novartis Junior Investigator AwardConference Hall 010:30am - 12:00pm10:30am PROSTATE-SPECIFIC ANTIGEN (PSA) OVER-EXPRESSION INCREASES THE MIGRATORYPOTENTIAL OF THE PROSTATE CANCER CELLS, PC-3.10:45amTara Veveris-LoweA MOUSE MODEL OF SPINAL BULBAR MUSCULAR ATROPHY (SBMA)Patrick McNamanmy11 :OOam GENE-ENVIRONMENT INTERACTION INFLUENCES THE RELATIONSHIP BETWEEN ALCOHOLCONSUMPTION AND ABDOMINAL OBESITY IN HEALTHY FEMALE TWINS11:15amJerry GreenfieldSIGNAL TRANSDUCTION VIA 1-0-PHOSPHATIDYLINOSITOL-3 KINASE (PI3K) IS NECESSARY FORTHE SURVIVAL OF THE PREIMPLANTATION EMBRYO.Vashetharan Ch<strong>and</strong>rakanthan11 :30am INHIBITION OF LIVER RECEPTOR HOMOLOGUE-1 (LRH-1) BY SMALL HETERODIMER PARTNER(SHP) IN PREADIPOCYTES.Agnes Kovacic11 :45am ROLE OF b1-INTEGRIN IN SPERMIATION AND SPERMIATION FAILUREAm<strong>and</strong>a BeardsleySRB Oral Presentations - Session 3 - Gamete Biotechnology <strong>and</strong> BreedingConference Room 1010:30am - 12:00pm10:30am CONCENTRATION OF PROGESTERONE AND SUPEROVULATORY RESPONSE WITH DIFFERENTDOSAGES OF FSHLuciene Santiago10:45am11:00am11:15amEFFECT OF ADDING SEMINAL PLASMA TO BOAR SPERM BEFORE AND AFTERCRYOPRESERVATIONRoslyn BathgateTHE INFLUENCE OF ENVIRONMENTAL TEMPERATURE ON ELITE A.1. BULLS HOUSED IN ACOOLING SHED OR FIELD AS MEASURED WITH AN INFRARED THERMOMETERAndrew BrockwayFLOW CYTOMETRIC SORTING OF FROZEN-THAWED RAM SPERMATOZOAFiona Hollinshead11 :30am EFFECT OF TIME OF INSEMINATION AND DOSE OF SORTED, CRYOPRESERVED RAM SPERM ONFERTILITY IN EWESChis Maxwell11 :45am THE EFFECT OF NUTRITION DURING THE CYCLE OF MATING ON OVIDUCTAL FLUID AMMONIAAND UREA LEVELS IN THE EWEMr Muhammad Azam KakarSRB Oral Presentations - Session 4 - Female <strong>Fertility</strong> <strong>and</strong> Function 1Conference Room 210:30am - 12:00pm10:30am PITUITARY LH~- AND FSH~-SUBUNITmRNA AND LH AND FSHCONTENT IN HEIFERS DURING ANDAFTER TREATMENT WITH GnRH AGONIST,10:45amProf Michael D'OcchioEXPRESSION OF GERM CELL AND SOMATIC CELL MARKERS DURING GONADAL DEVELOPMENTIN A MARSUPIALGratiana E Wijayanti11 :OOam LEPTIN RECEPTOR mRNA EXPRESSION DURING FOLLICULAR DEVELOPMENT AND OVULATION INTHE IMMATURE PRIMED RAT OVARYNatalie Ryan11 :15am ANDROGEN RECEPTOR EXPRESSION IN OVARIAN EPITHELIUM IN MICE OF DIFFERING AGES ANDTOTAL OVULATION NUMBER.11:30amMs Olivia TanLOCALISATION OF EPITOPES ON BRUSHTAIL POSSUM (TRICHOSURUS VULPECULA) ZONAPELLUCIDA 2 PROTEINDr Xianlan Cui11 :45am EOSINOPHILS IN DEVELOPMENT AND FUNCTION OF REPRODUCTIVE TISSUESAm<strong>and</strong>a Sferruzzi-Perri17


SRB Founders LecturePlenary Lecture Conference Hall 012:00pm EMBRYO DEVELOPMENT AND BIOTECHNOLOGYAlan TrounsonLunch (includes SRB Student Lunch Roundtable & ESA poster viewing)Exhibition Halls F & G1:OOpm - 2:00pmTransplantation <strong>and</strong> <strong>Reproduction</strong> - Issues <strong>and</strong> Solutions (SRB)Symposium Conference Room 2 2:00pm - 3:30pm~ 2:00pm DISCRIMINATING SELF AND NON-SELF: THE IMMUNOLOGICAL PARADOX POSED BY MAMMALIAN.-:;--;# REPRODUCTIONBruce Lovel<strong>and</strong>2:20pm2:50pm3:10pm18PRESENT AND FUTURE OPTIONS FOR PRESERVATION OF TESTIS TISSUE AND FUNCTIONStefan SchlattDOES THE IMMUNE SYSTEM PLAY A ROLE IN REPRODUCTION? - A CLINICIANS PERSPECTIVEKelton Tremel/enFINDING SOLUTIONS: SEMEN AND MATERNAL IMMUNE TOLERANCEDr Sarah RobertsonESA Poster discussions 1 - Female <strong>Reproduction</strong>poster Exhibition Halls F & G 2:00pm - 3:15amUSE OF A HUMAN COMMERCIAL KIT TO ASSAY OESTROGEN DURING BOVINE CYCLES WITHDIFFERENT FOLLICULAR WAVESEduardo NogueiraOPTIMIZATION OF SOLID PHASE EXTRACTION PROCEDURE FOR ANALYSIS OFESTROGENS AND PROGESTOGENS IN BIOLOGICAL SAMPLESKathrin Kulhanek-~OLE OF LOCAL ESTROGEN METABOLISM IN THE CONTROL OF ESTROGEN ACTION IN HUMANPARTURITIONGemma MadsenGLUTAMATERGIC NEURONS IN THE ARCUATENENTROMEDIAL COMPLEX EXPRESS ESTROGENRECEPTOR AND PROJECT TO PREOPTIC AREA IN EWESAIda PereiraSEASONAL CHANGES IN EXPRESSION OF PREPROOREXIN (ppORX),MELANIN CONCENTRATING HORMONE (MCH) AND LEPTIN RECEPTOR (Ob-Rb) mRNA INOVARIECTOMISED (OVX) CORRIEDALE EWES IN RELATION TO APPETITE AND BREEDINGAlex<strong>and</strong>ra RaoREPRODUCTIVE HORMONE RESPONSES TO ACUTE EXERCISE IN YOUNG EUMENORRHEICLeanne RedmanESTROGEN HAS A ROLE IN REGULATING APOPTOSIS IN THE BRAINRachel HillIMPAIRED MAMMARY PTHrP CONTENT AND FUNCTION AND NEWBORN GROWTH DURINGLACTATION IN THE SHRDr Mary WlodekDEVELOPING OF AN ANALYTICAL METHOD BASED ON SUPRAMOLECULAR CHEMISTRY ANDHIGH·PERFORMANCE LIQUID CHROMATOGRAPHY FOR SEPARATION AND QUANTIFICATION OFSEX STEROIDSPawel Zarzycki12:00pm -1 :OOpmESA Poster discussions 2 - HPAposter Exhibition Halls F & G 2:00pm - 3:15amTHE ACUTE ADMINISTRATION OF THE SELECTIVE SEROTONIN REUPTAKE INHIBITOR (SSRI)ANTIDEPRESSANT, SERTRALlNE, ACTIVATES THE HPA AXIS IN FEMALE, BUT NOT MALE, SHEEP.Jill Broadbear.~ THE PHYSIOLOGICAL ROLE OF GLUCOCORTICOIDS DURING EMBRYONIC DEVELOPMENT:ANALYSIS IN GLUCOCORTICOID RECEPTOR NULL MICETim ColeCORTICOTROPIN RELEASING HORMONE CAUSES VASODILATION IN HUMAN SKIN VIA MAST CELLDEPENDENT PATHWAYSRenee CromptonEFFECT OF SIZE AT BIRTH ON SALIVARY CORTISOL IN THE ADULT GUINEA PIGSanita GroverCHANGES IN ADRENAL RESPONSIVENESS DURING RAT PREGNANCY: THE ROLES OF ACTHRECEPTOR AND STAR.Susan HishehPRENATAL ENDOTOXIN EXPOSURE REDUCES STRESS RESPONSE OF PRE-WEANED GUINEAPIGS IN SEX-SPECIFIC MANNER.Nicolette TapeNEONATAL BACTERIAL ENDOTOXIN EXPOSURE ALTERS PRO-INFLAMMATORY AND FEBRILERESPONSES IN ADULT RODENTSDr Rohan Walker___);:HE EFFECT OF METYRAPONE INFUSION ON ADRENAL STEROIDOGENESIS IN THE LATEGESTATION FETAL SHEEPKirsty WarnesESA Poster discussions 3 - Inhibinposter Exhibition Halls F & G 2:00pm - 3:15amINHIBIN INCREASES MDA-MB-468 BREAST CANCER CELL ADHESION TO FIBRONECTIN.Tali LangNEOINTIMAL HYPERPLASIA IN THE RAT CAROTID INJURY MODEL IS NOT REDUCED BY LOCALFOLLISTATIN ADMINISTRATIONDavid McGrawCELLULAR RELEASE OF ACTIVIN A IN ACUTE INFLAMMATORY EVENTS IS PROSTAGLANDININDEPENDENT AND MAY REQUIRE DIRECT INTERACTION WITH LIPOPOLYSACCHARIDE (LPS).Kristian JonesEXPRESSION OF MULTIPLE TRANSFORMING GROWTH FACTOR (TGF)-~ SUPERFAMILY MEMBERSAND FOLLISTATIN IN THE ANTERIOR PITUITARYYao WangSERUM INHIBIN B & PRO-aC LEVELS WITH EXPERIMENTAL MANIPULATION OF GONADOTROPHINLEVELS IN NORMAL MEN.Dr Kati MatthiessonESA Poster discussions 4 - Metabolismposter Exhibition Halls F & G 2:04pm - 3:15pmCLONING AND EXPRESSION OF MOUSE AND RAT HOMOLOGUES OF NOVEL G PROTEIN-COUPLEDRECEPTORS, LGR7 AND LGR8; PUTATIVE RECEPTORS FOR RELAXIN AND INSULIN 3Daniel ScottFETAL GROWTH RESTRICTION IMPAIRS INSULIN SENSITIVITY OF GLUCOSE METABOLISMINDEPENDENTLY OF ADIPOSITY AND CIRCULATING FREE FATIY ACIDS IN THE ADULT GUINEADane Michael HortonAGING AND THE INSULIN RESISTANCE SYNDROME IN THE GUINEA PIG: A ROLE FOR ALTEREDBODY COMPOSITIONPrema ThavaneswaranSEPARATION OF PROSTAGLANDIN (PG)E2 AND PGF2a FROM THEIR TISSUE METABOLITESUSING REVERSED·PHASE THIN-LAYER CHROMATOGRAPHYToni WelshSOX13, HIGH SOLT, NUCLEAR LOCALISATION AND INSULIN EXPRESSIONHelen Kasimiotis19


o SITE-SPECIFICESA Poster discussions 5 - Pregnancy/Parturitionposter Exhibition Halls F & G 2:00pm - 3:15pmMATHEMATICAL MODELLING OF HUMAN MYOMETRIAL TENSION TRAJECTORIES THROUGHOUTPREGNANCYPeter SokolowskiESA Concurrent Oral Communications - Clinical 1oral Conference Room 4 3:45pm - 5:45pm3:45pm4:00pm4:15pm4:30pm4:45pm5:00pm5:15pmEXPRESSSION OF GENES IN THE UPPER AND LOWER SEGMENTS OF THE HUMANUTERUS DURING PREGNANCYKylie BrownREGULATION OF 15-HYDROXYPROSTAGLANDIN DEHYDROGENASE (PGDH) mRNA LEVELS IN THEHUMAN CHORION LAEVE AT TERM.Renee Johnson'-...QONTROL OF HUMAN PARTURITION INVOLVES CHANGES IN RESPONSIVENESS OF THE-'-"-MYOMETRIUM TO PROGESTERONE AND ESTROGENDr Sam MesianoI--.~RH GENE EXPRESSION IS REPRESSED BY ESTROGEN AND STIMULATED BY THEESTROGEN-ANTAGONIST ICI182,780 IN PLACENTARick NicholsonGonadotrophin stimulation <strong>and</strong> Risk of Multiple PregnancyProf Robert NormanGENERATION OF REACTIVE OXYGEN SPECIES BY HUMAN PLACENTAL CELLSKellie-Ann TaylorPLACENTAL RESTRICTION AND PLASMA THYROID HORMONE IN THE NEONATAL LAMB.Miles De BlasioBIOMARKER TRAJECTORIES IN PREGNANCY AND THE DETECTION OF PATHOLOGYDr Shaun McGrathDOES BULK TRANSFER OF BOVINE BLASTOCYSTS COMPROMISE EARLY EMBRYODr Jim PetersonPURIFICATION AND CHARACTERISATION OF CYTOSOLIC THYROID HORMONE BINDING PROTEININ HUMAN PLACENTABrett McKinnonExhibition Halls F & GCORTICOSTEROID BINDING GLOBULIN POLYMORPHISMS IN CHRONIC FATIGUE SYNDROMEDavid TorpyGLUCOCORTICOID RECEPTOR POLYMORPHISMS AND POST TRAUMATIC STRESS DISORDERDrAnthony BachmannPLASMA FREE, SALIVARY AND TOTAL CORTISOL RESPONSES TO VESTIBULAR CALORICSTIMULATION IN NORMAL HUMANSDr Jeff GriceDIETARY COMPOSITION IN RESTORING REPRODUCTIVE AND METABOLIC PHYSIOLOGY INOVERWEIGHT WOMEN WITH POLYCYSTIC OVARY SYNDROMELisa MoranTHE EFFECT OF INCREASED DIETARY PROTEIN ON WEIGHT LOSS AND INSULIN RESISTANCE INOBESE MEN AND WOMEN.Natalie LuscombeTHE EXTRAOCULAR MUCLE THYROID STIMULATING HORMONE RECEPTOR (TSHR) AND ITSRELEVANCE TO GRAVES' OPTHALMOPATHYSteven KloproggeMANAGEMENT OF MULTINODULAR GOITRE IN AUSTRALIA: DIFFERENCES BETWEENENDOCRINOLOGISTS AND ENDOCRINE SURGEONSJohn WalshAfternoon Tea3:30pm - 4:00pm,.,- SRB Oral Presentations - Session 5 - Female <strong>Fertility</strong> <strong>and</strong> Function 11Conference Room 24:00pm - 6:00pm5:30pm ENDOCRINE RESPONSE TO TRAUMATIC HEAD INJURY: A PROSPECTIVE STUDY.Prof Jim Stockigt20 214:00pm4:15pm4:30pm4:45pm5:00pm5:15pm5:30pm5:45pmESTRADIOL-MEDIATED CYCLIN 02 EXPRESSION: A ROLE FOR ER??DrAnn DrummondPURIFICATION OF HUMAN GRANULOSA CELLS FROM FOLLICULAR FLUID SAMPLES FOR USE INSERIAL ANALYSIS OF GENE EXPRESSION (SAGE).Michael QuinnREMODELLING OF EXTRACELLULAR MATRIX IN BOVINE FOLLICLES AT OVULATIONHelen INing-RodgersPROTEOGLYCANS - OSMOTICALLY ACTIVE MOLECULES IN FOLLICULAR FLUIDHannah ClarkeTHE INFLUCENCE OF THE OOCYTE DURING IN VITRO MATURATION ON THE METABOLISM OFBOVINE CUMULUS-OOCYTE COMPLEXESMelanie SuttonEXPRESSION WITHOUT SECRETION OF TGF-B1 AND TGF-B2 BY BOVINE AND MURINE OOCYTESLesley RitterDEVELOPMENT AND ANTRUM FORMATION IN MARSUPIAL PREANTRAL FOLLICLES CULTURED INA SERUM-FREE MEDIUMNadine RitchingsINCORPORATION OF BRDU INTO THE OVARIAN SURFACE EPITHELIUM IN MATURE MOUSE OVARYSLICES MAINTAINED IN CULTURE.Peter HurstJoint ESAlSRB (SRB 6) Concurrent Free Orals - Male <strong>Reproduction</strong>oral Conference Hall D 4:00pm - 5:45pm4:00pm4:15pm4:30pm4:45pm5:00pm5:15pm5:30pmSEX DIFFERENCES IN THE NEURONAL INPUTS FROM THE HYPOTHALAMUS AND BRAINSTEM TOTHE REGION OF THE GONADOTROPHIN-RELEASING HORMONE (GnRH) NEURONES IN THEMEDIAL PREOPTIC AREADr Chris ScottIN VITRO REGULATION OF ACTIVIN / INHIBIN SUBUNITS, FOLLISTATIN AND BAMBI IN THE 3 DAYOLD RAT TESTIS BY ACTIVIN, FOLLISTATIN AND FSHDr Kate Lovel<strong>and</strong>REGULATION OF SERTOLI CELL ACTIVIN AND INHIBIN SECRETION BY HORMONES, CYTOKINESAND THE SEMINIFEROUS EPITHELIAL CYCLEMark HedgerCHARACTERISATION OF PLASMA INHIBIN FORMS IN FERTILE AND INFERTILE MENDr David RobertsonContraceptive Efficacy Of A Depot Androgen And Progestin Combination In MenProf David H<strong>and</strong>elsmanPRESENCE OF IMMUNE MODULATING MOLECULES IN BOAR SEMINAL PLASMASean O'LearyINFLUENCE OF OXYTOCIN ON OXYTOCIN RECEPTOR AND 5 a-REDUCTASE TRANSCRIPTION INTHE RAT PROSTATEMr Stephen Assinder


ESA Concurrent Oral Communications - GH/IGFConference Room 104:00pm4:15pm4:30pm4:45pm5:00pm5:15pm5:30pmMeetingMeeting4:00pm - 5:45pmTHE IDENTIFICATION AND EXPRESSION PATTERN OF A GHRELIN VARIANT THAT ENCODES ANOVEL PEPTIDE IN THE HUMAN AND MOUSEPenny JefferyOESTROGEN AND SELECTIVE OESTROGEN RECEPTOR MODULATORS EXERT DIFFERENTIALEFFECTS ON GROWTH HORMONE SIGNALLINGKin LeungINSULIN-LIKE GROWTH FACTOR-I PREVENTS APOPTOSIS IN GLUCOSE DEPLETED NEURONALCELLSDr Kisho KobayashiUROKINASE TYPE PLASMINOGEN ACTIVATOR RECEPTOR (UPAR) IS INVOLVED IN INSULIN-LIKEGROWTH FACTOR I AND II-INDUCED MIGRATION OF RHABDOMYOSARCOMA (RMS) CELLS INMarisa Gal/icchioFGF-2 PROMOTES DIFFERENTIATION OF NEUROBLASTOMA CELLS BY INHIBITION OF IGFMITOGENIC ACTIVITY AND MODULATION OF THE APOPTOTIC SIGNALING PATHWAYS.Dr Vince RussoTNFaAND IGF SYSTEM CROSS-TALK IN A HUMAN KERATINOCYTE CELL LINE: IMPLICATIONS FOREPI DERMAL HOM EOSTASIS.Dr Stephanie EdmondsonSERUM INSULIN-LIKE GROWTH FACTOR BINDING PROTEIN-5 IS RE-DISTRIBUTED IN HUMANDIABETES MELLITUS.Dr Stephen TwiggConference Hall DAdelaide University ClubAdelaide University Club22ESA Annual General MeetingSRB Student MeetingStudent BBQ5:45pm - 6:45pm7:00pm - 7:30pm7:30pm - 11 :30pmTuesday, 24 September 2002Fetal Programming (ESA)Symposium Conference Hall D 9:00am - 11 :OOam9:00am9:30am10:00am10:30amNEUROENDOCRINE AND CARDIOVASCULAR ADAPTATIONS OF THE FETUS TO ALTEREDNUTRITION- THE FETAL ORIGINS OF ADULT DISEASE?Prof Caroline McMillenPERINATAL PROGRAMMING OF SYNDROME X IN THE ADULTJulie OwensNEUROENDOCRINE PROGRAMMING OF CANCERDr Deborah HodgsonTHE PLACENTAL GLUCOCORTICOID BARRIER & FETAL PROGRAMMING: A LEPTIN CONNECTION?Dr Brendan WaddellESA Concurrent Oral Communications· Clinical 2Conference Room 119:00am9:15am9:30am9:45am10:00am10:15am10:30am10:45am9:00am - 11 :OOamTHE MORBIDITY AND MORTALITY IN GH-DEFICIENT ADULTS: EFFECTS OF LONG-TERM GHREPLACEMENT THERAPYGudmundurJohannssonUSE OF LONG ACTING SOMATOSTATIN IN ACROMEGALY: AN AUDITTien-Ming HngLONG-TERM EFFECTS OF ANDROGEN REPLACEMENT THERAPY ON BONE MINERAL DENSITY(BMD) IN MENAshrafAminorroayaACTIVATING AND INACTIVATING MUTATIONS IN THE CALCIUM SENSING RECEPTORASSOCIATED WITH DISORDERS OF CALCIUM HOMEOSTASISAaron MagnoHYPOVITAMINOSIS D - A FORGOTTEN EPIDEMIC IN THE ELDERLY WITH OSTEOPOROTICFRAGILITY FRACTURESChanna PereraSEVERE VITAMIN D DEFICIENCY IN YOUNGER ADULTS - EFFECTS ON THE SPINEDr Walter PlehweSEX-SPECIFIC CHANGES IN PLACENTAL FUNCTION AND FETAL GROWTH AND DEVELOPMENT INPREGNANCIES COMPLICATED BY ASTHMAMs Vanessa MurphyPREVALENCE AND PREDICTORS OF ANDROGEN ABUSE IN AUSTRALIAN HIGH SCHOOLSPaula AndersonSRB Oral Presentations· Session 7 • Male <strong>Fertility</strong> <strong>and</strong> Function IIoral Conference Room 1 9:00am - 11 :OOam9:00am9:15am9:30am9:45am10:00am10:15am10:30am10:45amINTROMISSION FAILURE LEADS TO INFERTILITY IN TGFB1 NULL MICEWendy IngmanIMMUNOREGULATORY CYTOKINES IN SEMINAL PLASMA OF MEN WITH INFLAMMATION ORSPERM AUTOIMMUNITYMark HedgerSEASONAL CHANGES IN THE ABUNDANCE OF 14 kDa SEMINAL PLASMA PROTEINS SHOWN TOBIND TO RAM SPERMATOZOADr John F SmithIDENTIFICATION AND LOCALISATION OF A MESOTOCIN RECEPTOR IN THE PROSTATE OF THEBRUSHTAIL POSSUM.Helen NicholsonTYROSINE PHOSPHORYLATION AND EPIDIDYMAL MATURATION IN MURINE SPERMATOZOAKelly AsquithUSE OF THE YEAST TWO-HYBRID SYSTEM TO CHARACTERISE THE FUNCTION OF SPERMPROTEIN, TPX-1Deborah HickoxGENE EXPRESSION OF ADENYLYL CYCLASE IN RAT TESTIS AND CAUDAL EPIDIDYMAL SPERMMargaret WadeSPERM SURFACE PROTEIN PH-20 EXPRESSION DURING SHEEP SPERMATOGENESISFu Yu23


ESAlSRB (SRB 8) Joint Concurrent Oral Communications - Female <strong>Reproduction</strong>oral Conference I;loom 2 9:00am - 11 :OOam9:00am OVARIAN FOLLICULAR DEVELOPMENT IS COMPROMISED IN TRANSGENIC (mREN-2)27 RATS ANDANGIOTENSIN II-INFUSED SPRAGUE DAWLEY RATSTanyth de Gooyer9:15am9:30am9:45am10:00am10:15am10:30am10:45amEFFECT OF SUB-BURSAL FAS ANTIBODY ADMINISTRATION ON FOLLICULAR CASPASE-3ACTIVATION AND OVARIAN VOLUMEMr Mark FenwickGRANULOSA CELLS MATURE BEFORE DEATH IN BASAL ATRESIADr Ray RodgersESTROGEN PROMOTES ANGIOGENESIS IN ER?-EXPRESSING HUMAN MYOMETRIALMICROVASCULAR ENDOTHELIAL CELLS BY UPREGULATING VEGF RECEPTOR-2Caroline GargettDIETARY ESTROGENS HAVE SELECTIVE EFFECTS ON THE REPRODUCTIVE AXIS IN ESTROGENDIFFICIENT ARKO MICEKara BrittA LONG TERM FOLLOW-UP OF A FEMALE WITH AROMATASE DEFICIENCYYoko MurataLIVER RECEPTOR HOMOLOGUE-1 REGULATES AROMATASE EXPRESSION IN PREADIPOCYTES.Colin ClyneMECHANISMS UNDERLYING THE REDUCED SENSITIVITY TO PROLACTIN NEGATIVE FEEDBACKDURING LACTATIONDr Stephen AndersonExhibition Halls F & GMorning TeaHarrison LecturePlenary Lecture Conference Hall DChaired by Prof. Ken Ho11 :30am ENDOCRINE-IMMUNE INTERACTIONSGeorge ChrousosExhibition Halls F & GLunch <strong>and</strong> Poster Viewing11 :OOam - 11 :30am11 :30am - 12:30pm12:30pm - 1:30pmApoptosis <strong>and</strong> Signalling (SRB)Symposium Conference Hall D 1:30pm - 3:30pm2:00pm2:10pm2:30pm3:00pm3:15pmSIGNALLING MECHANISMS DURING APOPTOSISProf Arun DharmarajanDROSOPHILA AS A MODEL SYSTEM TO STUDY STEROID HORMONE REGULATED CELL DEATHSharad KumarNEGATIVE REGULATION OF PROLACTIN RECEPTOR SIGNALLING BY SOCS PROTEINS: A NEWMECHANISM IN LUTEOLYSIS?AlProfJon CurlewisPARACRINE SIGNALLING WITHIN THE OVARIAN FOLLICLERob GilchristEXPRESSION TGFI3 SUPERFAMILY MEMBERS AND THEIR SIGNALLING COMPONENTRY BY THE RATOVARYDrAnn DrummondESA Poster discussions 6 - boneposter Exhibition Halls F & G 1:30pm - 3:00pmCYCLICAL INTRAVENOUS PAMIDRONATE IN THE TREATMENT AND PREVENTION OFOSTEOPOROSISDr Sophie ChanEXPERIENCE WITH INTRAVENOUS PAMIDRONATE THERAPY FOR THE PROPHYLAXIS ANDTREATMENT OF OSTEOPOROSIS.AlProf Duncan ToplissUP-REGULATION OF THE VITAMIN D 24-HYDROXYLASE GENE PROMOTER BY 1,25DIHYDROXYVITAMIN D: RAPID ACTIVATION AND SPECIFIC ROLES OF MAP KINASE PATHWAYS INTHE INDUCTION MECHANISM.Dr Prem DwivediESA Poster discussions 7 - cancerposter Exhibition Halls F & G 1:30pm - 3:00pmCHARACTERISATION OF A STEROID RECEPTOR-SPECIFIC TRANSCRIPTIONAL RESISTANCE IN AGRANULOSA TUMOUR CELL LINE.Simon ChuINHIBITION OF ANDROGEN SIGNALLING IN PROSTATE CANCER CELLS BY DOMINANT NEGATIVEANDROGEN RECEPTORSLisa ButlerINCREASED ANDROGEN RECEPTOR AND HER2 IMMUNOREACTIVITY IN CLINICALLY LOCALISEDPROSTATE CANCER IS ASSOCIATED WITH DISEASE PROGRESSION FOLLOWING RADICALPROSTATECTOMYDr Carmela RicciardelliLACK OF RESPONSE TO THE SYNTHETIC PROGESTIN, MEDROXYPROGESTERONE ACETATE, INADVANCED BREAST CANCER IS ASSOCIATED WITH EXPRESSION OF NON-FUNCTIONALANDROGEN RECEPTORSMiao YangESA Poster discussions 8 - clinicalposter Exhibition Halls F & G 1:30pm - 3:00pmRECOVERY OF PITUITARY FUNCTION IN XANTHOMATOUS HYPOPHYSITIS: A CASE REPORTMorton BurtINOPERABLE INTRACARDIAC PHEOCHROMOCYTOMA IN A YOUNG WOMAN WITH SUBSEQUENTCOMPLICATED BY SUBSEQUENT PREGNANCYBernard ChampionPULSED TESTOSTERONE THERAPY: PHARMACOKINETICS AND SAFETY OF INHALEDTESTOSTERONE IN POSTMENOPAUSAL WOMEN.Dr Sonia DavisonFACTORS UNDERLYING THE RISING INCIDENCE OF PAPILLARY THYROID CARCINOMA INTASMANIA (1988-1998).John BurgessFAMILIAL NEUROHYPOPHYSEAL DIABETES INSIPIDUS (FNDI) ASSOCIATED WITH A MISSENSEMUTATION ENCODING CYS58?ARG IN NEUROPHYSIN II (NPII)John Peng2425


ESA Poster discussions 9 - comparativeposter Exhibition Halls F & G 1:30pm - 3:00pmREGULATION OF RELAXIN GENE EXPRESSION IN THE CORPUS LUTEUM OF THE TAMMARWALLABY.Helen GehringSTEROID PRODUCTION BY ADRENALS AND GONADS FROM EMBRYONIC SOUTHERN SNOWSKINKS, NIVEOSCINCUS MICROLEPIDOTUS, DURING THEIR PROTRACTED GESTATION PERIOD.Dr Jane GirlingDOES HABITAT DETERMINE HORMONAL REGULATION OF METAMORPHOSIS IN AUSTRALIANANURANS?Natalie PlantLOCAL EMBRYO-DERIVED FACTORS INFLUENCE UTERINE OXYTOCIN RECEPTORS IN THETAMMAR WALLABY.Andrew SiebelTHE EFFECT OF L-CARNITINE ON ENERGY BALANCE IN THE MARSUPIAL SMINTHOPSISCRASSICAUDATA.Zoe HolthouseESA Poster discussions 10 - GH/IGFposter Exhibition Halls F & G 1:30pm - 3:00pmGROWTH HORMONE TREATMENT OF MEN WITH ABDOMINAL OBESITY REDUCES FATACCUMULATION IN THE LIVER.GudmundurJohannssonFUNCTIONAL DIFFERENCE OF SOMATOTROPES FROM AROMATASE KNOCKOUT AND WILD-TYPEMOUSE PITUITARIESDr Chen ChenEVIDENCE THAT INSULIN-LIKE GROWTH FACTOR BINDING PROTEINS HAVEINDEPENDENT AND IGF-I DEPENDENT ENDOCRINE ACTIONS IN PREGNANCYPatricia GrantMDA-MB-468 BREAST CANCER CELLS ADHESION TO FIBRONECTIN IS REGULATED BYINSULIN-LIKE GROWTH FACTOR BINDING PROTEIN-3 (IGFBP-3).Kathryn HalePRESENCE OF GROWTH HORMONE SECRETAGOGUES RECEPTOR (GHS-R) AND GHRELIN INENDOMETRIAL ADENOCARCINOMANeveen TawadrosREGULATION OF THE SOCS3 PROMOTER ACTIVITY BY GROWTH HORMONE AND OESTROGEN.Gary LeongESA Poster discussions 11 - male reproductionposter Exhibition Halls F & G 1:30pm - 3:00pmTHE MOLECULAR EFFECTS OF ANDROGENS ON SKELETAL MUSCLE CELLS IN VITROAnnaAxell26CONTROL OF SERTOLI CELL PROLIFERATION AND DIFFERENTIATIONJeremy BuzzardEXPRESSION OF 17HSD31N MOUSE PRE-ADIPOCYTES AND THE REGULATION OFANDROSTENEDIONE TO TESTOSTERONE CONVERSION IN VITRO.Lisa De C<strong>and</strong>iaTESTICULAR ACTIONS OF AN ACTIVATING MUTATION OF THE HUMAN FSH RECEPTOR INTRANSGENIC MICEAlvara GarciaDEVELOPMENT OF TRANSGENIC MICE TO EXPLORE THE FUNCTION OFPHOSPHATIDYLETHANOLAMINE BINDING PROTEINS ON MALE FERTILITYGerard GibbsA GENOME WIDE SCREEN FOR GENES ESSENTIAL TO MALE FERTILITY USING ENUClaire KennedyREDUCED N/C INTERACTIONS BUT INCREASED LIGAND INDUCED TRANSCRIPTIONAL ACTIVITY OFAN ANDROGEN RECEPTOR VARIANT CONTAINING A DISRUPTED POLYGLUTAMINE TRACTNicole MooreESA Concurrent Oral Communications - HPAConference Room 23:00pm - 5:00pm3:00pm ANALYSIS OF TRANSCRIPTION FACTORS REGULATING PLACENTAL CRH THROUGH THE CRE.Kristy Shipman3:15pm3:30pm3:45pm4:00pm4:15pm4:30pm4:45pmCORTICOSTERONE BINDING GLOBULIN EXPLOITS SERPIN MECHANISMS TO DELIVER CORTISOLTO TARGET CELLS.Dr Sinan AliREGULATION OF P450c17 mRNA BY TGF-13 SUPERFAMILY MEMBERS IN A MOUSE ADRENAL CELLLINEGuckOoiMETABOLISM OF SYNTHETIC GLUCOCORTICOIDS USED FOR THE TREATMENT OF ASTHMA BYPLACENTAL 11 ?-HYDROXYSTEROID DEHYDROGENASE TYPE 2Rebecca FittockTRANSFORMING GROWTH FACTOR B1 (TGFB1) - PUTTING THE BRAKES ON ADRENALSTEROIDOGENESIS BEFORE BIRTHDr Ca<strong>the</strong>rine CoulterNOVEL ASPECTS OF CRH AND AVP SIGNALLING IN PITUITARY CELLSDr Giavanna AngeliIDENTIFICATION OF AMINO ACID RESIDUES WITHIN THE TETRATRICOPEPTIDE REPEAT DOMAINOF CYCLOPHILIN 40 THAT MEDIATE HSP90 RECOGNITIONDr Bryan WardSPECIFIC LEUCINE RESIDUES WITHIN AN HSP90 C-TERMINAL MOTIF TARGETED BY THE HSP90ANTAGONIST NOVOBIOCIN ARE ESSENTIAL FOR HSP90 DIMERISATION AND INTERACTION WITHSTEROID RECEPTOR-ASSOCIATED IMMUNOPHILINSRudi AllanExhibition Halls F & GESA Concurrent Oral Communications - Male & Female <strong>Reproduction</strong> sessionSymposium Conference Hall D 3:00pm - 5:00pm3:00pm3:15pm3:30pm3:45pm4:00pmZONAL AND DEVELOPMENTAL VARIATION OF PROGESTERONE- AND GLUCOCORTICOIDRECEPTOR MRNA EXPRESSION IN THE RAT PLACENTA.Peter MarkHUMAN MYOMETRIAL GENES ARE DIFFERENTIALLY EXPRESSED IN LABOUR: A SUPPRESSIONSUBTRACTIVE HYBRIDISATION (SSH) STUDYEng-Cheng ChanINTERLEUKIN 11 SECRETION BY HUMAN ENDOMETRIAL STROMAL CELLS IS STIMULATED BYPROSTAGLANDIN E2 IN PART VIA A CYCLIC ADENOSINE MONOPHOSPHATE DEPENDENTPATHWAY.Dr Evodokia DimitriadisIDENTIFICATION OF THE TRANSCRIPTION FACTOR E2F4 AS A NOVEL BINDING PARTNER FORTHE GONADOTROPIN-RELEASING HORMONE RECEPTORAylin HanyalogluTHE INTRACELLULAR CONSEQUENCES OF THE CAG EXPANSION AND ANDROGEN RECEPTORTRUNCATION ON THE PATHOGENESIS OF KENNEDY'S DISEASEJanifer Wong4:15pm EXPRESSION AND REGULATION OF CYCLO-OXYGENASE ISOFORMS IN THE ADULT RAT TESTISUgurAIi4:30pm THE NOVEL G PROTEIN-COUPLED RECEPTOR LGR8 IS A POTENTIAL RECEPTOR FOR INSULIN 3Ross Bathgate4:45pm STEREOLOGICAL EVALUATION OF SPERMATOGENESIS INDUCED BY TESTOSTERONE (T) ORHUMAN CHORIONIC GONADOTROPHIN (hCG) IN GONADOTROPHIN DEFICIENT (hpg) MICE.Jennifer SpalivieroSRB Afternoon Tea3:00pm - 3:30pm27


ESA Concurrent Oral Communications· MetabolismSymposium Conference Room 10 3:00pm· 5:00pm3:00pm3:15pm3:30pm3:45pm4:00pm4:15pm4:30pm4:45pmDOES BETACELLULIN-INDUCED ERBB ACTIVATION INHIBIT CYTOKINE-INDUCED APOPTOSIS OFPANCREATIC b-CELLS?Natasha LloydPREDIALYSIS CHRONIC RENAL INSUFFICIENCY IS ASSOCIATED WITH ELEVATED GLUCOSELEVELS AND INSULIN RESISTANCEDr Anthony O'SuJlivanCHOLESTEROL FEEDING RESCUES THE FATTY LIVER PHENOTYPE OF THE AROMATASEKNOCKOUT (ARKO) MOUSE.Kylie HewittTHE EFFECT OF FETAL SERUM ON THE SYNTHETIC FUNCTION OF THE BRONCHIAL SMOOTHMUSCLEAnnette Osei-KumahPERIPUBERTAL INCREASES IN PLASMA LEPTIN LEVELS IN FEMALE MERINO SHEEPDr Jim McFarlanePLACENTAL RESTRICTION AND ONTOGENY OF INSULIN-REGULATED GLUCOSE HOMEOSTASIS INSHEEPDr Kathy Gaf<strong>for</strong>dPROGRAMMING OF APPETITE AND BODYWEIGHT BY PERINATAL DIETARY OMEGA-3 FATTYACIDS AND ANGIOTENSIN.Michael MathaiTHE EFFECT OF COMMON VARIANTS IN MTHFR ON TWINNING AND EMBRYO LOSS.Mr Grant MontgomeryESA Concurrent Oral Communications - Clinical Sex SteroidsSymposium Conference Room 11 3:00pm· 5:00pm3:00pm3:15pm3:30pm3:45pm4:00pm4:15pm4:30pm4:45pmTHE THRESHOLD FOR ANDROGEN DEFICIENCY SYMPTOMS IN MEN WITH HYPOGONADISM OFDIFFERING ETIOLOGIESMs Sharyn KelleherTHE EFFECT OF 12 MONTHS ORAL TESTOSTERONE UNDECANOATE ON BODY COMPOSITION,MUSCLE STRENGTH AND PROSTATE FUNCTION IN ELDERLY MEN.Mat<strong>the</strong>w HarenTHE ACUTE EFFECTS OF HIGH DOSE TESTOSTERONE ON SLEEP IN AGEING MENDr. Peter LiuTHE RELATIONSHIP OF SERUM TOTAL TESTOSTERONE TO BODY COMPOSITION IN HEALTHYAGEING MENCarolyn AJlanTESTOSTERONE ENHANCES THE ANTINATRIURETIC AND METABOLIC EFFECTS OF GROWTHHORMONEDr James GibneyENDOGENOUS OESTROGENS BENEFICIALLY INFLUENCE ENDOTHELIAL FUNCTION IN HEALTHYYOUNG MENDr Paul KomesaroffPURIFIED ISOFLAVONES IMPROVE ARTERIAL FUNCTION IN MEN AND WOMEN.Dr Helena TeedeASSESSING THE PROGNOSTIC POTENTIAL OF CD82, A METASTASIS SUPPRESSOR, IN PRIMARYPROSTATE CANCERA/ProfAlbert FraumanSRB Junior Scientist Awards (SRB)Conference Room 13:30pm - 5:00pm3:30pm DIFFERENTIAL GENE EXPRESSION IN INTERLEUKIN-11 RECEPTOR a DEFICIENT MICE COMPAREDTO WILD TYPE DURING DECIDUALISATION3:45pm4:00pm4:15pm4:30pm4:45pmChristine WhiteMANIPULATION OF BOVINE OOCYTE-CUMULUS CELL GAP JUNCTIONAL COMMUNICATION DURINGIN VITRO MATURATION BY MODULATING CELL-SPECIFIC CAMP LEVELSRebecca ThomasLEPTIN DYNAMICS IN THE PREGNANT RAT: CHANGES IN CLEARANCE RATE ANDTRANSPLACENTAL PASSAGE LATE IN GESTATIONJeremy SmithSEMINAL CYTOKINE CONCENTRATION AND HUMAN REPRODUCTIVE OUTCOMEDavid SharkeyTHE EFFECT OF P53 DELETION ON EMBRYO VIABILITYAiqing LiINSEMINATION AND UTERINE LYMPHOCYTE TRAFFICKING IN MICEJohn BromfieldTaft Lecture (ESA)Plenary Lecture Conference Hall D 5:00pm· 6:00pm5:00pm PRE-RECEPTOR METABOLISM OF CORTISOL BY 11 ~-HYDROXYSTEROID DEHYDROGENASES:IMPLICATIONS FOR HUMANProf Paul StewartSRB Annual General MeetingMeeting Conference Room 15:00pm· 6:00pmStam<strong>for</strong>d Gr<strong>and</strong>, GlenelgESAISR Conference DinnerSponsored by Novo Nordisk7:00pm - 11 :30pm28 29


Thyroid Update (ESA)Symposium Conference Room 1 10:00am - 12:00pm10:00am10:30amWednesday, 25 September 2002Stem Cells (SRB)Symposium Conference Hall 0 8:30am - 9:30am8:30am9:00amGENERATING PITUITARY CELLS FROM EMBRYONIC STEM CELLS - A ROUTE TO PITUITARY CELLTHERAPYPaul ThomasTHERAPEUTIC APPLICATION OF STEM CELL TECHNOLOGIESPeter RathjenPlenaryPlenary Lecture Conference Hall B & C8:30am COMPARATIVE 3-D STRUCTURES OFTHE IGF-1 AND EGF RECEPTORSColin WardExhibition Halls F & GGrowth Factors/Cytokines (ADS/ESA)Symposium Conference Hall B & C 10:00am - 12:00pm10:00am10:30amINSULIN-LIKE GROWTH FACTORS IN DIABETES - FOR BETTER OR FOR WORSE?Prof George Wer<strong>the</strong>rSUPPRESSORS OF CYTOKINE SIGNALLINGDoug HiltonMorning Tea11 :OOam CONNECTIVE TISSUE GROWTH FACTORS IN DIABETIC COMPLICATIONSDr Stephen Twigg11 :30am GROWTH FACTORS OF THE VEGF FAMILY INVOLVED IN ANGIOGENESIS ANDLYMPHANGIOGENESISSteve StackerTHE ROLE OF THE TSH RECEPTOR IN THYROID ASSOCIATED OPHTHALMOPATHYA/ProfAlbert FraumanSIGNIFICANCE OF EYE MUSCLE AUTOIMMUNITY IN THYROID-ASSOCIATED OPHTHALMOPATHYJack Wall11 :OOam GENE THERAPY FOR MEDULLARY THYROID CANCERBruce Robinson11 :30amSteve Boyages8:30am - 9:30am9:30am - 10:00amHormones <strong>and</strong> <strong>Reproduction</strong> (ESA/SRB) SymposiumSymposium Conference Room 2 10:00am - 12:00pm10:00am10:30amOOCYTE-DERIVED GROWTH FACTORS AND OVULATION RATE IN MAMMALSKen McNattyINTRAOVARIAN REGULATION OF FOLLICULOGENESIS: SIGNAL MOLECULES AND THEIR CELLULARSOURCESDavid Armstrong11 :OOam OESTROGEN TARGET SITES IN HUMAN TESTISPhillipa Saunders11 :30am MALE CONTRACEPTION - APPROACH TO HORMONAL METHODSRobert McLachlanExhibition Halls F & G30Lunch <strong>and</strong> Poster Viewing12:00pm - 1:30pmPlenary Lecture1:00pm1:OOpm - 2:00pmSRB Oral Presentations - Session 9 - Models of <strong>Fertility</strong> <strong>and</strong> Infertilityoral Conference Room 1 1:OOpm - 3:00pm1:00pm1:15pm1:30pmSRB Oral Presentations - Session 10 - Uterine Biologyoral Conference Room 2 1:OOpm :' 3:00pm1:OOpm1:15pm1:30pm1:45pm2:00pm2:15pm2:30pmNeuroendocrinology of stress (with Neuroendocrinology Interest Group)Symposium Conference Room 1 2:30pm - 4:30pm2:30pm3:00pm3:30pm4:00pmFOLLICULAR DEVELOPMENT IN WALLABY OVARIAN TISSUE XENOGRAFTS IS NOT ALTERED BYTHE HORMONAL ENVIRONMENT OF THE RECIPIENTMelanie SnowDEVELOPMENT OF ANTRAL FOLLICLES IN RABBIT OVARIAN XENOGRAFTS IN IMMUNODEFICIENTMICEKarla MuirheadACUTE FSH WITHDRAWAL IMPAIRS SERTOLI AND GERM CELL DEVELOPMENT IN THE IMMATURERATDr Sarah Meachem1:45 pm EVALUATION OF TESTICULAR COOLING FOR GERM CELL TRANSPLANTATION2:00pm2:15pmZhenZhangDELIVERY OF ANTIBODIES FOR MALE IMMUNOCONTRACEPTIONAssoc Prof Russell JonesIMMUNOCONTRACEPTIVE EFFECT OF RECOMBINANT PORCINE ZONA PELLUCIDA C PROTEIN INRABBITSDr Eileen McLaughlinEFFECT OF ANGIOGENESIS INHIBITORS ON OESTROGEN MEDIATED ENDOMETRIALENDOTHELIAL CELL PROLIFERATION IN THE OVARIECTOMISED MOUSEBambang HeryantoMOLECULAR RESPONSE OF MYOMETRIAL MICROVASCULAR ENDOTHELIAL CELLS TO VEGF ANDPROGESTERONE ASSESSED BY MICROARRAYPeter RogersANALYSIS OF REPRODUCTIVE PHENOTYPE AND EXPRESSION OF CALCIUM BINDING PROTEINSIN CABP-D28K NULL MICEKien LuuMAST CELL NUMBER AND SPATIAL LOCATION IN THE VAGINAL CUL-DE-SAC OF THE BRUSHTAILPOSSUM AFTER ADMINISTRATION OF EXOGENOUS OESTRADIOLTrish MahoneyIMMUNOLOCALISATION OF INTERLEUKIN 11 AND ITS RECEPTOR IN CYCLING ENDOMETRIUM ANDIMPLANTATION SITES OF THE RHESUS MONKEY.Dr Evodokia DimitriadisREPEATED MATERNAL GLUCOCORTICOID TREATMENTS IN SHEEP IMPAIR PLACENTALSTRUCTURAL MATURATIONClaire RobertsTREATMENT WITH A GLUCOCORTICOID RECEPTOR ANTAGONIST PREVENTS BIRTH IN TAMMARWALLABIES.Geoff ShawADS/ESA Joint Plenary LectureConference Hall B & CPPARS AND THE METABOLIC SYNDROMESBart StaelsTHE NEUROENDOCRINOLOGY OF STRESSGeorge ChrousosSTRESS & STRESSORS: A CONCEPTUAL FRAMEWORKTrevor DaySTRESS & THE REPRODUCTIVE AXIS: SEX DIFFERENCES AND CONSEQUENCES FOR FERTILITYDr Alan TilbrookNEUROENDOCRINE REGULATION AND THE METABOLIC SYNDROMERol<strong>and</strong> Rosmond31


Symposium Conference Hall 8 & C2:30pm Bart Staels3:00pm Ken Kraegen3:30pm Dr John Prins4:00pm Assoc.ProfJoe ProiettoNuclear signalling (ESA/ADS)Sponsored by GlaxoSmithKline2:30pm· 4:30pmESA & SRB Annual Scientific Meeting 2002SRB PROGRAMESA! SRB Late Breaking ScienceChaired by Kate Lovel<strong>and</strong> <strong>and</strong> Leon BachConference Room 23:00pm· 4:00pmAdelaide Convention Centre22-25 September 2002Sunday, 22 September 2002ISymposium - New Horizons in Reproductive BiologyChairs: Ray Rodgers <strong>and</strong> John MorrisonConference Room 24:00pm 1. EPIGENETIC PHENOMENA IN REPRODUCTIVE BIOLOGYMichael Holl<strong>and</strong>4:30pm 2. EPIGENETIC INHERITANCE IN MAMMALSEmma Whitelaw5:00pm 3. SUPRAMOLECULAR SCIENCE TO NANOTECHNOLOGYLis<strong>and</strong>ra Martin5:30pm 4. GRAPES AND WINE FLAVOURPatrick Williams4:00pm - 6:00pmI Eli Lilly Welcome Function I'-;E;:::X-:;::h:-;;ib:-;it;;-io---n-F=o-y--e-rs--------:!...---=------=-=-...::...:.......:..-=..::..::...:...:.::..=...:~-----6-:O-O-p-m-.-7-:3-0-p-m--;::;:~-:---::.- w__o_m_e_n_·_1n_E_nd..::..:..o.:.cr:...:.i.:..:.n..=.o.:...:lo:...::g~y~Fu=..::..:.nc=_t::io=_n:..:- 1Riverview 37:00pm· 8:30pm3233


IMonday, 23 September 2002SRB PROGRAMSRB Oral Presentations - Session 1 - Male <strong>Fertility</strong> <strong>and</strong> Function IChairs: Gerard Gibbs <strong>and</strong> Chris O'NeillConference Room 108:30am 5. IRON IS HIGHLY GENOTOXIC TOWARD HUMAN SPERMATOZOADennis Sawyer8:45am9:00am9:15am9:30am9:45am8:45am -10:00am6. MORPHOLOGICAL MANIFESTATION OF Y CHROMOSOMAL MICRODELETIONS IN THE HUMANDr Marc Luetjens7. LEPTIN INHIBITS BASAL BUT NOT STIMULATED TESTOSTERONE PRODUCTION BY THEIMMATURE MOUSE TESTISDrJim McFarlane8. THE ROLE OF FGFR SIGNALLING IN SPERM TAIL DEVELOPMENTLeanne Cotton9. REDOX REGULATION OF SPERM FUNCTIONMark Baker10. DOES CALCIUM HAS A ROLE IN SPERM HEAD ROTATION TO T-SHAPE ORIENTATION DURINGCAPACITATION? A TALE OF TWO MARSUPIALS SPECIES, THE BRUSHTAIL POSSUM (TRICHOSURUSVULPECULA) AND THE TAMMAR WALLABY (MACROPUS EUGEN/~.Dr Kuldip SidhuSRB Oral Presentations - Session 2 - Embryo <strong>Development</strong> <strong>and</strong>ImplantationChairs: Eva Dimitriadis <strong>and</strong> Graham JenkinConference Room 28:30am 11. PROLACTIN IS A PREIMPLANTATION GROWTH FACTOR IN MICEDr Peter Kaye8:45am9:00am9:15am9:30am9:45am10:00am12. REAL-TIME PCR EXPRESSION OF HYPOXIA-INDUCIBLE FACTORS DURING BOVINEPREIMPLANTATION EMBRYO DEVELOPMENTAlex<strong>and</strong>ra Harvey8:45am - 10:15am13. EFFECT OF IN VITRO OXYGEN CONCENTRATION ON GLUCOSE TRANSPORTER-1 EXPRESSIONIN THE MOUSE BLASTOCYSTKaren Kind14. A COMPARISON OF STAINING METHODS USED TO DETERMINE NUMBERS OF CELLS IN PIGBLASTOCYSTSChristopher Grupen15. PARACRINE PERTURBATIONS ASSOCIATED WITH BOVINE NUCLEAR TRANSFER PREGNANCIESSusan Ravelich16. SURVIVAL OF FRESH AND VITRIFIED SHEEP IVP AND CLONED EMBRYOS AFTER EMBRYOTRANSFERTeija Peura17. ABNORMAL TROPHOBLAST MHC-I EXPRESSION AND ASSOCIATED ENDOMETRIALLYMPHOCYTIC RESPONSE IN BOVINE NUCLEAR TRANSFER PREGNANCIES.Jonathan HillIMonday, 23 September 2002SRB PROGRAMSRB Oral Presentations - Session 3 - Gamete Biotechnology <strong>and</strong>BreedingChairs: Teija Peura <strong>and</strong> Graeme MartinConference Room 1010:30am -12:00pm10:30am 18. CONCENTRATION OF PROGESTERONE AND SUPEROVULATORY RESPONSE WITH DIFFERENTDOSAG ES OF FSHLuciene Santiago10:45am19. EFFECT OF ADDING SEMINAL PLASMA TO BOAR SPERM BEFORE AND AFTERCRYOPRESERVATIONRoslyn Bathgate11 :OOam 20. THE INFLUENCE OF ENVIRONMENTAL TEMPERATURE ON ELITE A.1. BULLS HOUSED IN ACOOLING SHED OR FIELD AS MEASURED WITH AN INFRARED THERMOMETERAndrew Brockway11 :15am 21. FLOW CYTOMETRIC SORTING OF FROZEN-THAWED RAM SPERMATOZOAFiona Hollinshead11 :30am 22. EFFECT OF TIME OF INSEMINATION AND DOSE OF SORTED, CRYOPRESERVED RAM SPERM ONFERTILITY IN EWESChis Maxwell11 :45am 23. THE EFFECT OF NUTRITION DURING THE CYCLE OF MATING ON OVIDUCTAL FLUID AMMONIAAND UREA LEVELS IN THE EWEMuhammad Azam KakarSRB Oral Presentations - Session 4 - Female <strong>Fertility</strong> <strong>and</strong> Function IChairs: Ann Drummond <strong>and</strong> Philippa SaundersConference Room 210:30am - 12:00pm10:30am 24. PITUITARY LH~- AND FSH~-SUBUNIT mRNA AND LH AND FSH CONTENT IN HEIFERS DURINGAND AFTER TREATMENT WITH GnRH AGONISTMichael D'Occhio10:45am25. EXPRESSION OF GERM CELL AND SOMATIC CELL MARKERS DURING GONADAL DEVELOPMENTIN A MARSUPIALGratiana E Wijayanti11 :OOam 26. LEPTIN RECEPTOR mRNA EXPRESSION DURING FOLLICULAR DEVELOPMENT AND OVULATIONIN THE IMMATURE PRIMED RAT OVARY1Natalie Ryan11:15am27. ANDROGEN RECEPTOR EXPRESSION IN OVARIAN EPITHELIUM IN MICE OF DIFFERING AGESAND TOTAL OVULATION NUMBER.Olivia Tan11 :30am 28. LOCALISATION OF EPITOPES ON BRUSHTAIL POSSUM (TRICHOSURUS VULPECULA) ZONAPELLUCIDA 2 PROTEINXianlan Cui11 :45am 29. EOSINOPHILS IN DEVELOPMENT AND FUNCTION OF REPRODUCTIVE TISSUESAm<strong>and</strong>a Sferruzzi-PerriI Morning Tea I'-;E;:::x"':';h::";ib;::-;i::;ti'":-o-=-n7 H =-a7: n- s -=F:-:&=-=G-----------:::::.-----------1-0-:0-0-a-m---1-0-:3-0-a-m-3435


IChair: John HearnConference Hall D12:00pmMonday, 23 September 2002SRB PROGRAMSRB FOUNDERS LECTUREProfessor Alan TrounsonMonash University30. EMBRYO DEVELOPMENT AND BIOTECHNOLOGY12:00pm .. 1:OOpmLunch (includes SRB Student Lunch Roundtable with Professor Trounson)Exhibition Halls F & G1:OOpm .. 2:00pmI Symposium - Transplantation <strong>and</strong> <strong>Reproduction</strong> - Issues <strong>and</strong> Solutions IChairs: Mark Hedger <strong>and</strong> Wendy IngmanConference Room 22:00pm .. 3:30pm2:00pm 31. DISCRIMINATING SELF AND NON-SELF: THE IMMUNOLOGICAL PARADOX POSED BYMAMMALIAN REPRODUCTIONBruce Lovel<strong>and</strong>2:20pm 32. PRESENT AND FUTURE OPTIONS FOR PRESERVATION OF TESTIS TISSUE AND FUNCTIONStefan Schlatt2:50pm 33. DOES THE IMMUNE SYSTEM PLAY A ROLE IN REPRODUCTION? - A CLINICIANS PERSPECTIVEKelton Tremel/en3:10pm 34. FINDING SOLUTIONS: SEMEN AND MATERNAL IMMUNE TOLERANCE.Sarah Robertson1--;;::::;::7.:7.:7:::-;-::::-;;:--;:-:::-:::- A__ ft:...:,e....:rn..::..o.:..o:.,:n:...:...-=.T-=e.=.a 1Exhibition Halls F & G3:30pm .. 4:00pmISRB Oral Presentations - Session 5 - Female <strong>Fertility</strong> <strong>and</strong> Function IIChairs: Rob Gilchrist <strong>and</strong> Eileen McLaughlinConference Room 24:00pm .. 6:00pm4:00pm 35. ESTRADIOL-MEDIATED CYCLIN D2 EXPRESSION: A ROLE FOR ERa?Ann Drummond4:15pm 36. PURIFICATION OF HUMAN GRANULOSA CELLS FROM FOLLICULAR FLUID SAMPLES FOR USE INSERIAL ANALYSIS OF GENE EXPRESSION (SAGE)4:30pm4:45pmMichael Quinn37. REMODELLING OF EXTRACELLULAR MATRIX IN BOVINE FOLLICLES AT OVULATIONHelen Irving-Rodgers38. PROTEOGLYCANS - OSMOTICALLY ACTIVE MOLECULES IN FOLLICULAR FLUIDHannah ClarkecontinuedI5:00pm5:15pm5:30pm5:45pmMonday, 23 September 2002SRB PROGRAM39. THE INFLUCENCE OF THE OOCYTE DURING IN VITRO MATURATION ON THE METABOLISM OFBOVINE CUMULUS-OOCYTE COMPLEXESMelanie Sutton40. EXPRESSION WITHOUT SECRETION OF TGF-B1 AND TGF-B2 BY BOVINE AND MURINE OOCYTESLesley Ritter41. DEVELOPMENT AND ANTRUM FORMATION IN MARSUPIAL PREANTRAL FOLLICLES CULTURED IN A SERUM-FREE MEDIUMNadine Ritchings42. INCORPORATION OF BRDU INTO THE OVARIAN SURFACE EPITHELIUM IN MATURE MOUSEOVARY SLICES MAINTAINED IN CULTURE.Peter HurstJoint ESAlSRB Session 6 Oral Communications - Male <strong>Reproduction</strong>Chairs: John Aitken <strong>and</strong> Peter LiuConference Hall D4:00pm .. 5:45pm4:00pm 43. SEX DIFFERENCES IN THE NEURONAL INPUTS FROM THE HYPOTHALAMUS AND BRAINSTEMTO THE REGION OF THE GONADOTROPHIN-RELEASING HORMONE (GnRH) NEURONES IN THEMEDIAL PREOPTIC AREA4:15pm4:30pm4:45pm5:00pm5:15pm5:30pmChris Scott44. IN VITRO REGULATION OF ACTIVIN / INHIBIN SUBUNITS, FOLLISTATIN AND BAMBI IN THE 3 DAYOLD RAT TESTIS BY ACTIVIN, FOLLISTATIN AND FSHKate Lovel<strong>and</strong>45. REGULATION OF SERTOLI CELL ACTIVIN AND INHIBIN SECRETION BY HORMONES, CYTOKINESAND THE SEMINIFEROUS EPITHELIAL CYCLEMark Hedger46. CHARACTERISATION OF PLASMA INHIBIN FORMS IN FERTILE AND INFERTILE MENDavid Robertson47. CONTRACEPTIVE EFFICACY OF A DEPOT ANDROGEN AND PROGESTIN COMBINATION IN MENDavid H<strong>and</strong>elsman48. PRESENCE OF IMMUNE MODULATING MOLECULES IN BOAR SEMINAL PLASMASean O'Leary49. INFLUENCE OF OXYTOCIN ON OXYTOCIN RECEPTOR AND 5 a-REDUCTASE TRANSCRIPTION INTHE RAT PROSTATEStephen Assinder1 S_R_B_S_tu_d_e_n_t_M_e_e_t_in-=9:..-- ----IIAdelaide University Club7:00pm .. 7:30pmI'-- S_t_ud_e_n_tB_8_0 1Adelaide University Club7:30pm .. 11 :30pm3637


Tuesday, 24 September 2002SRB PROGRAMSRB Oral Presentations - Session 7 - Male <strong>Fertility</strong> <strong>and</strong> Function IIChairs: Sarah Meachem <strong>and</strong> Mark BakerConference Room 19:00am 50. INTROMISSION FAILURE LEADS TO INFERTILITY IN TGFB1 NULL MICEWendy Ingman9:15am9:30am9:45am10:00am10:15am10:30am10:45am9:00am - 11 :OOam51. IMMUNOREGULATORY CYTOKINES IN SEMINAL PLASMA OF MEN WITH INFLAMMATION ORSPERM AUTOIMMUNITYMark Hedger52. SEASONAL CHANGES IN THE ABUNDANCE OF 14 kDa SEMINAL PLASMA PROTEINS SHOWN TOBIND TO RAM SPERMATOZOAJohn F Smith53. IDENTIFICATION AND LOCALISATION OF A MESOTOCIN RECEPTOR IN THE PROSTATE OF THEBRUSHTAIL POSSUM.Helen Nicholson54. TYROSINE PHOSPHORYLATION AND EPIDIDYMAL MATURATION IN MURINE SPERMATOZOAKelly Asquith55. USE OF THE YEAST TWO-HYBRID SYSTEM TO CHARACTERISE THE FUNCTION OF SPERMPROTEIN, TPX-1Deborah Hickox56. GENE EXPRESSION OF ADENYLYL CYCLASE IN RAT TESTIS AND CAUDAL EPIDIDYMAL SPERMMargaret Wade57. SPERM SURFACE PROTEIN PH-20 EXPRESSION DURING SHEEP SPERMATOGENESISFuYuJoint ESAlSRB Session 8 Oral Communications - Female <strong>Reproduction</strong>Chairs: Rob Norman <strong>and</strong> Jock FindlayConference Room 29:00am - 11 :OOam9:00am 58. OVARIAN FOLLICULAR DEVELOPMENT IS COMPROMISED IN TRANSGENIC (mREN-2)27 RATSAND ANGIOTENSIN II-INFUSED SPRAGUE DAWLEY RATSTanyth de Gooyer9:15am9:30am9:45am10:00am10:15am10:30am10:45am59. EFFECT OF SUB-BURSAL FAS ANTIBODY ADMINISTRATION ON FOLLICULAR CASPASE-3ACTIVATION AND OVARIAN VOLUMEMark Fenwick60. GRANULOSA CELLS MATURE BEFORE DEATH IN BASAL ATRESIARay Rodgers61. ESTROGEN PROMOTES ANGIOGENESIS IN ER?-EXPRESSING HUMAN MYOMETRIALMICROVASCULAR ENDOTHELIAL CELLS BY UPREGULATING VEGF RECEPTOR-2Caroline Gargett62. DIETARY ESTROGENS HAVE SELECTIVE EFFECTS ON THE REPRODUCTIVE AXIS IN ESTROGENDIFFICIENT ARKO MICEKara Britt63. A LONG TERM FOLLOW-UP OF A FEMALE WITH AROMATASE DEFICIENCYYoko Murata64. LIVER RECEPTOR HOMOLOGUE-1 REGULATES AROMATASE EXPRESSION IN PREADIPOCYTESColin Clyne65. MECHANISMS UNDERLYING THE REDUCED SENSITIVITY TO PROLACTIN NEGATIVE FEEDBACKDURING LACTATIONStephen AndersonTuesday, 24 September 2002SRB PROGRAMI Morning Tea ILE-X-h-i-bi-ti-o-n-H-a-II-S-F-&-G--------=.::.::.=..:.:..:.:.:..:.s!--=...::...:.::...---------:;:11;-:::~OO~a::m=--";1~1::-;;3;;-O=am=-IChair: Ken HoConference Hall DHarrison Lecture11 :30am 66. THE NEUROENDOCRINOLOGY OF STRESSGeorge Chrousos11 :30am -12:30pmI Lunch I-E-X-h-ib-i-ti-o-n-H-a-II-S-F-&-G----------...:=::...::.::.::..:..-------------::;1;-;;2;:-::3:;;O;:p=m:--~1:;-:::;;30n;p::m=-IChairs: Arun Dharmarajan <strong>and</strong> David PhillipsConference Hall D2:00pm2:10pm2:30pm3:00pm3:15pmSymposium - Apoptosis <strong>and</strong> Signalling1:30pm - 3:30pm67. SIGNALLING MECHANISMS DURING APOPTOSISArun Dharmarajan68. DROSOPHILA AS A MODEL SYSTEM TO STUDY STEROID HORMONE REGULATED CELL DEATHSharad Kumar69. NEGATIVE REGULATION OF PROLACTIN RECEPTOR SIGNALLING BY SOCS PROTEINS: A NEWMECHANISM IN LUTEOLYSIS?Jon Curlewis70. PARACRINE SIGNALLING WITHIN THE OVARIAN FOLLICLERob Gilchrist71. EXPRESSION TGF~ SUPERFAMILY MEMBERS AND THEIR SIGNALLING COMPONENTRY BY THERAT OVARYAnn DrummondI SRB Afternoon Tea ILE-X-h-i-bi-ti-o-n-H-a-II-S-F-&---=G-------=.:....:..=..:...::.::..=.~::..=.;::....:.......::-.:.....:....:...---------:;3~:On;O;:p::m:-:--;;'3:;-;j3'i\Op~m;:-3839


Tuesday, 24 September 2002SRB PROGRAMWednesday, 25 September 2002SRB PROGRAMIChair: John HearnConference Room 13:30pm3:45pm4:00pm4:15pm4:30pm4:45pmSRB Junior Scientist Awards72. DIFFERENTIAL GENE EXPRESSION IN INTERLEUKIN-11 RECEPTOR a DEFICIENT MICECOMPARED TO WILD TYPE DURING DECIDUALISATIONChristine White3:30pm· 5:00pm73. MANIPULATION OF BOVINE OOCYTE-CUMULUS CELL GAP JUNCTIONAL COMMUNICATIONDURING IN VITRO MATURATION BY MODULATING CELL-SPECIFIC CAMP LEVELSRebecca Thomas74. LEPTIN DYNAMICS IN THE PREGNANT RAT: CHANGES IN CLEARANCE RATE ANDTRANSPLACENTAL PASSAGE LATE IN GESTATIONJeremy Smith75. SEMINAL CYTOKINE CONCENTRATION AND HUMAN REPRODUCTIVE OUTCOMEDavid Sharkey76. THE EFFECT OF P53 DELETION ON EMBRYO VIABILITYAiqing Li77. INSEMINATION AND UTERINE LYMPHOCYTE TRAFFICKING IN MICEJohn BromfieldI SRB Annual General Meeting I~C:::-o-n--'f;-e-re-n-c-e-=R:-o-o-m-1::----------------_---':~:""'------5:-O-O-pm-.-s-:O-O-p-m-Stam<strong>for</strong>d Gr<strong>and</strong>, GlenelgESA/SRB Conference DinnerSponsored by Novo Nordisk7:00pm - 11 :30pmSymposium - Stem CellsSponsored by <strong>the</strong> CRC <strong>for</strong> Innovative Dairy ProductsChairs: Michael Holl<strong>and</strong> <strong>and</strong> Craig Smith8:30am - 9:30am78. GENERATING PITUITARY CELLS FROM EMBRYONIC STEM CELLS: A ROUTE TO PITUITARY CELLTHERAPY?Paul ThomasConference Hall 08:30am9:00am79. THERAPEUTIC APPLICATION OF STEM CELL TECHNOLOGIESPeter Rathjen1~ M_o_rn_i_n..;;:;g_T_e_a :-:-__~:-:-_1Exhibition Halls F & G9:30am - 10:00amIESAlSRB Joint Symposium - Hormones <strong>and</strong> <strong>Reproduction</strong>Chairs: Guck Ooi <strong>and</strong> David H<strong>and</strong>elsmanConference Room 210:00am - 12:00pm10:00am 80. OOCYTE-DERIVED GROWTH FACTORS AND OVULATION RATE IN MAMMALSKen McNatty10.30am 81. INTRAOVARIAN REGULATION OF FOLLICULOGENESIS: SIGNAL MOLECULES AND THEIRCELLULAR SOURCESDavid Armstrong11.00am 82. OESTROGEN TARGET SITES IN HUMAN TESTISPhillipa Saunders11 :30am 83. MALE CONTRACEPTION - APPROACH TO HORMONAL METHODSRobert McLachlan1~ Lu_n_ch --:-:--=-=-_:-=-:-_1Exhibition Halls F & G12:00pm - 1:30pmISRB Oral Presentations - Session 9 - Models of <strong>Fertility</strong> <strong>and</strong> InfertilityChairs: Stefan Schlatt <strong>and</strong> Jeremy ThompsonConference Room 11:OOpm - 3:00pm1:00pm 84. FOLLICULAR DEVELOPMENT IN WALLABY OVARIAN TISSUE XENOGRAFTS IS NOT ALTERED BYTHE HORMONAL ENVIRONMENT OF THE RECIPIENTMelanie Snow1:15pm 85. DEVELOPMENT OF ANTRAL FOLLICLES IN RABBIT OVARIAN XENOGRAFTS INIMMUNODEFICIENT MICEKarla Muirhead1:30pm 86. ACUTE FSH WITHDRAWAL IMPAIRS SERTOLI AND GERM CELL DEVELOPMENT IN THEIMMATURE RATSarah Meachem1:45 pm 87. EVALUATION OF TESTICULAR COOLING FOR GERM CELL TRANSPLANTATIONZhenZhang2:00pm 88. DELIVERY OF ANTIBODIES FOR MALE IMMUNOCONTRACEPTIONRussell Jones2:15pm 89. IMMUNOCONTRACEPTIVE EFFECT OF RECOMBINANT PORCINE ZONA PELLUCIDA C PROTEININ RABBITSEileen McLaughlin4041


IIWednesday, 25 September 2002SRB PROGRAMSRB Oral Presentations - Session 10 - Uterine BiologyChairs: Sarah Robertson <strong>and</strong> Anne TurnerConference Room 2 , 1:00pm - 3:00pm1:00pm 90. EFFECT OF ANGIOGENESIS INHIBITORS ON OESTROGEN MEDIATED ENDOMETRIALENDOTHELIAL CELL PROLIFERATION IN THE OVARIECTOMISED MOUSEBambang Heryanto1:15pm 91. MOLECULAR RESPONSE OF MYOMETRIAL MICROVASCULAR ENDOTHELIAL CELLS TO VEGFAND PROGESTERONE ASSESSED BY MICROARRAYPeter Rogers1:30pm 92. ANALYSIS OF REPRODUCTIVE PHENOTYPE AND EXPRESSION OF CALCIUM BINDING PROTEINSIN CABP-D28K NULL MICEKien Luu1:45pm 93. MAST CELL NUMBER AND SPATIAL LOCATION IN THE VAGINAL CUL-DE-SAC OF THE BRUSHTAILPOSSUM AFTER ADMINISTRATION OF EXOGENOUS OESTRADIOLTrish Mahoney2:00pm 94. IMMUNOLOCALISATION OF INTERLEUKIN 11 AND ITS RECEPTOR IN CYCLING ENDOMETRIUMAND IMPLANTATION SITES OFTHE RHESUS MONKEYEva Dimitriadis2:15pm 95. REPEATED MATERNAL GLUCOCORTICOID TREATMENTS IN SHEEP IMPAIR PLACENTALSTRUCTURAL MATURATIONClaire Roberts2:30pm 96. TREATMENT WITH A GLUCOCORTICOID RECEPTOR ANTAGONIST PREVENTS BIRTH IN TAMMARWALLABIESGeoff ShawChairs: Leon Bach <strong>and</strong> Kate Lovel<strong>and</strong>Conference Room 2ESA! SRB Late Breaking Science3:00pm - 4:00pm<strong>Society</strong> <strong>for</strong> Reproductive BiologyThirty-third Annual Scientific MeetingABSTRACTS(Nos 1-96 in order of presentation)1. EPIGENETIC PHENOMENA IN REPRODUCTIVE BIOLOGYMichael K Holl<strong>and</strong>Monash Institute of<strong>Reproduction</strong> & <strong>Development</strong>, Monash University, Clayton, Vic 3168At many levels from monozygotic twins, to st<strong>and</strong>ard laboratory animals, to cultures of differentiated cells, it is clear thatgenotype <strong>and</strong> phenotype are not synonymous. This is in part because of epigenetic changes. These are heritable changesin gene function involving modification in chromatin or in gene function but not structure. Epigenetic changes are seenat many points in development including development of <strong>the</strong> mammalian germ line. These modifications includedemethylation <strong>and</strong> selective remethylation of <strong>the</strong> genome, chromatin remodelling, erasure of allele specific methylationof imprinted loci <strong>and</strong> re-activation of <strong>the</strong> silent X-chromosome. Imprinting is <strong>the</strong> epigenetic modification that marks agamete's haploid genome as maternal or paternal. Imprinted genes have roles in prenatal growth, development ofparticular lineages, in behavior <strong>and</strong> in some diseases. Imprints are initiated during gametogenesis, inherited by maturegametes <strong>and</strong> transmitted to embryos. Whilst we do not as yet completely underst<strong>and</strong> all <strong>the</strong> aspects of <strong>the</strong> linkagesbetween methylation, chromatin remodelling <strong>and</strong> imprinting it is clear <strong>the</strong>se epigenetic phenomena function in concert ifdevelopment is to proceed normally. One example of where this may go awry is in cloning by nuclear transfer. Genes,which are silenced in somatic cell nuclei used as nuclear donors in cloning, must be reactivated <strong>for</strong> clones to developnormally. This requires reprogramming in an entirely different context than that applying during gametogenesis <strong>and</strong>within a much shorter time span. It is <strong>the</strong>re<strong>for</strong>e not surprising that partial or total failure of reprogramming occurs. Thiswill result in a range of phenotypes in clones with only a small percentage showing complete reprogramming. Thiscorrelates with <strong>the</strong> low percentage of healthy clones. Clearly more complete underst<strong>and</strong>ing of epigenetic processeswould contribute substantially to our underst<strong>and</strong>ing of normal development <strong>and</strong> improve our of efficiency cloning. Thisis of particular importance ifhealthy cloned animals are to be produced <strong>for</strong> commercial purposes.2. EPIGENETIC INHERITANCE IN MAMMALSE. Whitelaw, V. Rakyan, J. Preis, M. Blewitt, R. Druker <strong>and</strong> S. ChongDepartment ofBiochemistry, University ofSydney, NSW 2006, AustraliaIt is well recognised that <strong>the</strong>re is a surprising degree of phenotypic variation among genetically identical individualseven when <strong>the</strong> environmental influences, in <strong>the</strong> strict sense of <strong>the</strong> word, are identical. Genetic textbooks acknowledgethis fact <strong>and</strong> use different terms such as "intangible variation" or "developmental noise" to describe it. We believe thatthis intangible variation results from <strong>the</strong> stochastic establishment of epigenetic modifications to <strong>the</strong> DNA nucleotidesequence. These modifications, which may involve cytosine methylation <strong>and</strong> chromatin remodelling, result in alterationsin gene expression that, in tum, affect <strong>the</strong> phenotype of <strong>the</strong> organism. Recent evidence, from our work <strong>and</strong> that of o<strong>the</strong>rsin mice, suggests that <strong>the</strong>se epigenetic modifications, which in <strong>the</strong> past were thought to be cleared <strong>and</strong> reset on passagethrough <strong>the</strong> germline, may sometimes be inherited to <strong>the</strong> next generation. This is termed epigenetic inheritance, <strong>and</strong>while this process has been well recognised in plants, <strong>the</strong> recent findings in mice <strong>for</strong>ce us to consider <strong>the</strong> implications ofthis type of inheritance in mammals. At this stage we do not know how extensive this phenomenon is in humans but itmay well tum out to be <strong>the</strong> explanation <strong>for</strong> some diseases which appear to be sporadic or show only weak geneticlinkage.Morgan H., Su<strong>the</strong>rl<strong>and</strong> H., Martin D.I.K. <strong>and</strong> Whitelaw E. (1999) Epigenetic inheritance at <strong>the</strong> agouti locus in <strong>the</strong>mouse. Nature Genetics, 23, 314-317.Whitelaw E <strong>and</strong> Martin D.I.K (2001) Retrotransposons as epigenetic mediators of phenotypic variation in mammals.Nature Genetics, 27, 361-365.Rakyan V K, Blewitt M, Droker R, Preis J, <strong>and</strong> Whitelaw E (2002) Metastable epialleles in mammals. Trends inGenetics (Vol 18, in press).4243


3. SUPRAMOLECULAR SCIENCE TO NANOTECHNOLOGYLisa MartinSchool ofChemistry, Physics <strong>and</strong> Earth Sciences, Flinders University, South AustraliaMany processes in biological <strong>and</strong> medicinal science are characterised by supramolecular chemistry. This chemistry isdefined as non-molecular with non-covalent interactions or associations between biological molecules. Examplesinclude gene regulation <strong>and</strong> transcription, protein-protein, protein-antibody interactions. Fur<strong>the</strong>rmore, <strong>the</strong> syn<strong>the</strong>sis ofproteins or cofactors is often achieved by use of chaperone proteins assisting <strong>the</strong> "intermediates" to exist in o<strong>the</strong>rwise"reactive" environments prior to delivery or assembly of<strong>the</strong> mature biomolecule.The most recent development in scientific technology has been <strong>the</strong> decrease in size from micro-scale to nano-scale.This has drawn into sharp focus <strong>the</strong> possible use of biological molecules as components in nano-scale devices. Thetypes of technology envisaged includes photonic devices, such as Rhodopsin, molecular motors such as, ATPase,memory chips <strong>for</strong> quantum computers based on DNA, single molecule sensitive biosensors <strong>and</strong> targeted drug delivery<strong>and</strong> gene <strong>the</strong>rapies. One of <strong>the</strong> major scientific developments that has lead to this new nanotechnology is <strong>the</strong>availability of new materials such as microporus coatings, high tensile carbon tubes, <strong>and</strong> nanocomposite ceramics thatcan be used in bioelectronics. In addition, <strong>the</strong> development of Scanning Probe Microscopy has resulted in routineimaging at <strong>the</strong> molecular <strong>and</strong> atomic scale. This talk aims to demystify some of <strong>the</strong>se components of nano-science <strong>and</strong>provide an awareness ofpossible applications in <strong>the</strong> bio-medical sciences.Proteins <strong>and</strong> enzymes are particularly well designed to per<strong>for</strong>m specific reactions with high selectivity <strong>and</strong> <strong>the</strong>re<strong>for</strong>e<strong>the</strong>y are ideal biological elements <strong>for</strong> biosensors. Of particular focus in this talk is <strong>the</strong> use of supramolecular strategiestoge<strong>the</strong>r with immobilised techniques <strong>for</strong> metalloproteins <strong>and</strong> enzymes. These methods enable <strong>the</strong> direct, real timemonitoring of redox behaviour, which can be related to enzyme activity or function. One aim in this research is to use<strong>the</strong> molecular properties of proteins <strong>and</strong> enzymes with high specificity <strong>and</strong> selectivity to underpin <strong>the</strong> developments ofa new generation of biosensors. The attachment of metalloproteins to an electrode surface also allows one to probe <strong>the</strong><strong>the</strong>rmodynamic <strong>and</strong> kinetic controls of <strong>the</strong>ir function. The methods used <strong>for</strong> enzyme immobilisation will be presentedas well as examples of <strong>the</strong> possible scope <strong>for</strong> application to o<strong>the</strong>r areas of biological <strong>and</strong> medicinal science.potential <strong>for</strong> new bio-medicinal diagnostics will be discussed.4. GRAPES AND WINE FLAVOURPatrick J WilliamsThe Australian Wine Research Institute, PO Box 197 Glen Osmond South Australia 5064A brief overview of research into wine <strong>and</strong> grape flavour highlighting selected developments in <strong>the</strong> field over <strong>the</strong> lastthree decades. The talk will be concerned with factors that shaped <strong>the</strong> direction <strong>and</strong> evolution of wine flavour researchra<strong>the</strong>r than with details of <strong>the</strong> techniques that have been employed. For example, <strong>the</strong> focus away from wine <strong>and</strong> onto<strong>the</strong> grape <strong>for</strong> an underst<strong>and</strong>ing of varietal flavours, <strong>the</strong> recognition of a multiplicity of flavour active components, <strong>and</strong><strong>the</strong> absence of single flavour impact compounds, <strong>and</strong> a better underst<strong>and</strong>ing of <strong>the</strong> origins of flavour components, arejust some of <strong>the</strong> features that define our present knowledge of wine flavour composition. Some applications of <strong>the</strong>knowledge gained from <strong>the</strong> research <strong>and</strong> <strong>the</strong> potential of this to fur<strong>the</strong>r enhance Australia's position as a premium wineproducer will be discussed.The5. IRON IS HIGHLY GENOTOXIC TOWARD HUMAN SPERMATOZOADE Sawyer <strong>and</strong> RJ AitkenDiscipline ofBiological Sciences, School ofEnvironmental <strong>and</strong> Life Sciences, The University ofNewcastle, Callaghan,NSW, Australia, <strong>and</strong> The Hunter Medical Research Institute, NSWAustraliaHuman spermatozoa have been reported to be susceptible to <strong>the</strong> DNA damaging effects of pro-oxidants such ashydrogen peroxide (HzOz). However, DNA damage in individual genes has not been investigated in <strong>the</strong>se cells. Thus,we have applied a quantitative PCR assay (QPCR) to measure gene-specific DNA damage in human spermatozoatreated with <strong>the</strong> pro-oxidants HzOz <strong>and</strong> iron. Methods: Human spermatozoa were treated in vitro with hydrogenperoxide (0-5 mM) or Fe(II)S04 (0-400 I-LM) <strong>for</strong> one hour at room temperature. After DNA purification, gene-specificDNA damage was measured by QPCR. DNA damage (e.g., str<strong>and</strong> breaks, bulky adducts) blocks <strong>the</strong> progression of <strong>the</strong>PCR polymerase, resulting in decreased amplification of <strong>the</strong> target genes. The presence of single-str<strong>and</strong> DNA breaks<strong>and</strong> alkali-labile sites was determined by alkaline gel electrophoresis. Results: Millimolar concentrations of HzOz wererequired to induce DNA damage in human spermatozoa as assessed by QPCR. After 5 mM HzOz, amplification wasreduced by 40-50%, depending on <strong>the</strong> particular gene. The nature of this DNA damage was revealed by alkaline gelelectrophoresis to be single-str<strong>and</strong> breaks <strong>and</strong> alkali-labile sites. In contrast, iron induced a significant amount of DNAdamage after treatment with as little as 100 I-LM (25-40% decreased amplification), while 400 !-LM iron reducedamplification of target genes by greater than 90%. Interestingly, <strong>the</strong> DNA damage induced by iron was not due tostr<strong>and</strong> breads or alkali-labile sites as assessed by alkaline gel electrophoresis. Discussion: These results indicate thatnuclear DNA of mature spermatozoa is relatively protected from <strong>the</strong> damaging effects of hydrogen peroxide; however,iron appears to be highly genotoxic to nuclear DNA of human spermatozoa. The two genotoxins also appear to inducedistinctly different lesions. HzOz induces str<strong>and</strong> breaks <strong>and</strong>/or alkali-labile sites, while iron does not induce <strong>the</strong>selesions. We hypo<strong>the</strong>size that iron is stimulating <strong>the</strong> release of genotoxic lipid peroxides, which <strong>the</strong>n <strong>for</strong>m covalentadducts with DNA bases. These results are <strong>the</strong> first to show that DNA damage can be measured in specific DNAsequences in male germ cells, <strong>and</strong> provide new insight into <strong>the</strong> susceptibility of human spermatozoa to genotoxic a~ents.6. MORPHOLOGICAL MANIFESTATION OF Y CHROMOSOMALMICRODELETIONS IN THE HUMAN TESTISeM Luetjens, M Simoni, E Gromoll <strong>and</strong> E NieschlagInstitute <strong>for</strong> Reproductive Medicine of<strong>the</strong> University Muenster; Domagkstr. 11; D-48129 Muenster, GermanyInfertility affects 10-15% of couples seeking offspring [1]. Between 2 - 20% of <strong>the</strong> severe cases carry Y-chromosomalmicrodeletions in <strong>the</strong> azoospermia factor (AZF) region [2]. These chromosomal microdeletions are associated withmorphological disorders of spermatogenesis. Although <strong>the</strong> missing genes are described, no principle causing germ celldepletion is known. The aim of this study is to characterise biopsies of AZF patients more intensively to find similaritiesamong <strong>the</strong> pathologies of such patients. Testicular biopsy specimens from 17 patients with. AZFc deletions wereanalysed. In a r<strong>and</strong>omised study <strong>the</strong> diameter of <strong>the</strong> tubules, <strong>the</strong> lumina, <strong>the</strong> lamina propria <strong>and</strong> <strong>the</strong> epi<strong>the</strong>lia weremeasured <strong>and</strong> compared with those of patients with idiopathic Sertoli-cell-Only (SeO) syndrome (n=II), mixed atrophy(n=10) <strong>and</strong> complete spermatogenesis (n=II). The biopsies were also stained <strong>for</strong> Sertoli cell-specific antigens. Additionally<strong>the</strong> distal <strong>and</strong> proximal regions of <strong>the</strong> microdeletion were analysed with new primers in order to characterise <strong>the</strong> microdeletionposition <strong>and</strong> length [3]. By comparing <strong>the</strong> testis parameters of <strong>the</strong> AZFc deletion group with <strong>the</strong> control patients, anintermediate state between total sea <strong>and</strong> complete spermatogenesis emerged. The diameters of <strong>the</strong> average tubule of patientswith an AZFc deletion <strong>and</strong> patients with idiopathic sea were significantly smaller <strong>the</strong>n those of patients with completespermatogenesis. The lumen diameter was significantly smaller compared to <strong>the</strong> control group with complete spennatogenesis<strong>and</strong> larger than in idiopathic sca. Analysis of <strong>the</strong> chromosomal microdeletions demonstrated that all patients with a deletionsolely in <strong>the</strong> AZFc-region have identical breakpoints. The immunohistochemical examination of <strong>the</strong> Sertoli cell proteins did notreveal any specific expression differences among <strong>the</strong> patient groups. AZFc microdeletions lead to alterations in testismorphology different from those in patients with mixed atrophy, although <strong>the</strong> germ cell status of both are identical. Patents withY chromosome microdeletions show an intermediate status between idiopathic seo <strong>and</strong> complete spermatogenesis. Sertoli cellfunction seems not to be altered [1] Nieschlag, E. (2000) Scope <strong>and</strong> goals of <strong>and</strong>rology. In: Nieschlag E. <strong>and</strong> Behre H. M.(eds.) Andrology: male reproductive health <strong>and</strong> dysfunction. 2nd ed. Springer, Heidelberg; 4-5.[2] Vogt et al. (1996).Human Molecular Genetics, 5, 933-943.[3] Kuroda-Kawaguchi et al. (2001) Nature Genetics, 29. 279-286.4445


7. LEPTIN INHffiITS BASAL BUT NOT STIMULATED TESTOSTERONEPRODUCTION BY THE IMMATURE MOUSE TESTISMurren Herrid <strong>and</strong> Jim McFarlaneSchool ofBiomedical,Biological <strong>and</strong> Molecular Science, University ofNew Engl<strong>and</strong>, Armidale, NSW 2351Leptin is secreted into <strong>the</strong> blood by adipose tissue <strong>and</strong> was originally identified as a regulator of appetite <strong>and</strong> energyexpenditure. It has subsequently been shown to be involved in many o<strong>the</strong>r physiological systems includingreproduction. In humans <strong>and</strong> mice, plasma leptin levels are lower in males than in females even when corrected <strong>for</strong>body fat. Some authors have attributed this difference to a suppressive effect of testosterone on leptin production.However, we have recently shown that plasma leptin levels in both intact <strong>and</strong> castrate male sheep are lower than thatfound in females, (3) suggesting that <strong>the</strong> lower levels in males are not induced by testosterone. In contrast it has alsobeen suggested that elevated leptin may suppress testosterone production, since obese men tend to have reducedfertility <strong>and</strong> reduced plasma testosterone. Recently it has been reported that leptin suppresses testosterone productionin hCG stimulated adult rat testis slices (1) <strong>and</strong> purified Leydig cells (2). In contrast, we have reported that leptin has noeffect on adult mouse Leydig cells so <strong>the</strong> aim ofthis study was to investigate <strong>the</strong> effect ofleptin on testicular slicesfrom immature (14 days old) <strong>and</strong> adult (60 days old) Swiss mouse testes. A biphasic response was observed with leptinsuppressing testosterone production by up to 60% in <strong>the</strong> unstimulated immature testis at a dose of 6.25 ng/ml. In <strong>the</strong>hCG stimulated testis leptin had no effect on testosterone production. In <strong>the</strong> adult testis leptin had no significant effecton ei<strong>the</strong>r stimulated or unstimulated testosterone production. In contrast purified Leydig cells from both immature <strong>and</strong>adult testes were not affected by leptin. These data suggest that physiological doses of leptin do not directly affectLeydig cell steroidogenesis but acts via paracrine signaling from o<strong>the</strong>r cell types within <strong>the</strong> testis.(1) Tena-Sempere et. al., 1999 Journal ofEndocrinology 161: 211-218(2) Caprio et. ai., 1999 Endocrinology 140: 4939-4947(3) Kauter etal. 2000. Journal ofEndocrinology 166: 127-1358. THE ROLE OF FGFR SIGNALLING IN SPERM TAIL DEVELOPMENTLeanne M Cotton, Gerard M Gibbs, John R Morrison, Kim Sebire, David M de Kretser <strong>and</strong> Moira K O'BryanMonash Institute of<strong>Reproduction</strong> <strong>and</strong> <strong>Development</strong>, Monash University, Victoria 3168Unusual sperm tail morphology is present in a high percentage of infertile males, <strong>and</strong> because motile sperm areessential <strong>for</strong> unassisted fertilisation, research into sperm tail proteins will have an important impact on <strong>the</strong> way in whichinfertility is treated. A major focus of our group is to characterise <strong>the</strong> function of <strong>the</strong> outer dense fibres (ODF) of <strong>the</strong>sperm tail. ODF provide rigidity <strong>and</strong> directionality to tail movement, but <strong>the</strong> recent isolation of ODF proteins wi<strong>the</strong>nzymatic or regulatory characteristics, suggests <strong>the</strong>y have a more active role in motility. Screening of a rodent testislibrary using an ODF specific antisera <strong>and</strong> 3'RACE identified SNT-2 (Suc-l associated associated Neurotrophic FactorTarget 2) as a potential ODF component. The SNTs are known FGF (Fibroblast Growth Factor) signalling adaptors<strong>and</strong> key components in <strong>the</strong> activation of <strong>the</strong> MAP kinase <strong>and</strong> PI3-kinase pathways via <strong>the</strong> FGF receptor. SubsequentlyNor<strong>the</strong>rn blot analysis revealed that SNT-2 is highly expressed in <strong>the</strong> testis <strong>and</strong> up-regulated concordant with <strong>the</strong>appearance of spermatids <strong>and</strong> sperm tail development. Fur<strong>the</strong>r, Western blot analysis has demonstrated that rat spermtails are immunopositive <strong>for</strong> FGFR-1, <strong>and</strong> <strong>the</strong>re are large concentrations of FGFs within <strong>the</strong> epididymis. Collectivelythis data suggests that FGF signalling through SNT-2 is involved in sperm tail development/function. In order to testthis hypo<strong>the</strong>sis we have created transgenic mice that express a dominant-negative variant of <strong>the</strong> FGFR-1 in haploidgerm cells. The transgene is a fusion of cDNAs encoding a truncated FOFR-1 (extracellular <strong>and</strong> transmembranedomains) <strong>and</strong> a flag-tag, driven by <strong>the</strong> mouse protamine promoter. Mice have been generated by microinjection <strong>and</strong> arecurrently undergoing genotyping prior to assessing spermatogenesis <strong>and</strong> fertility.9. REDOX REGULATION OF SPERM FUNCTIONMark A. Baker, Anton Krutskikh <strong>and</strong> R. John AitkenDiscipline ofBiological Sciences, University ofNewcastle, Australia 2308.Mammalian sperm function is redox regulated. Using a variety of chemiluminescent <strong>and</strong> colorimetric probes we havediscovered multiple sources of redox activity in human spermatozoa. Thus NADH- <strong>and</strong> NADPH-dependentoxidoreductases have been detected in human sperm that generate profound chemiluminescent responses whenlucigenin is used as <strong>the</strong> probe, but not luminol, isoluminol or MCLA. The activities observed when NADH or NADPHis used as substrate are highly correlated, but biochemically distinct. For example, <strong>the</strong> NADPH-dependent activity canbe inhibited with diphenylene iodonium, while <strong>the</strong> NADH response cannot. On <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, both oxidoreductasescan be suppressed with <strong>the</strong> thiol reactive agent, pCMBS <strong>and</strong> both enzyme systems are competent to reduce tetrazoliumsalt derivatives such as NBT <strong>and</strong> WST-1. The NADH-dependent oxidoreductase has been isolated by FPLC <strong>and</strong> aprotein b<strong>and</strong> correlating with enzyme activity was analysed by MALDI TOF. The isolated protein was identified ascytochrome bs reductase (R.2). Transient transfection of COS7 cells with this enzyme reveal an increase in NADHdependentlucigenin chemiluminescence, consistent with a redox role <strong>for</strong> cytochrome bs in human spermatozoa. TheNAD(P)H lucigenin reductase activity in human spermatozoa is significantly correlated with <strong>the</strong> induction of lucigenindependentchemiluminescent responses with 12-myristate, 13-acetate phorbol ester (PMA). Moreover, both <strong>the</strong> PMA<strong>and</strong> NAD(P)H responses are significantly elevated in populations of defective human spermatozoa recovered from <strong>the</strong>low density region of PercoIl gradients. In contrast, <strong>the</strong> calcium-dependent chemiluminescent responses typical ofNOX5 are similar in both Percoll fractions. These observations shed light on <strong>the</strong> biochemical nature of <strong>the</strong> lesionspresent in male infertility <strong>and</strong> will help resolve <strong>the</strong> aetiology of this condition.10. DOES CALCIUM HAVE A ROLE IN SPERM HEAD ROTATION TO T-SHAPEORIENTATION DURING CAPACITATION? A TALE OF TWO MARSUPIALS SPECIES,THE BRUSHTAIL POSSUM (TRICHOSURUS VULPECULA) AND THE TAMMARWALLABY (MACROPUS EUGENIl)K.S.Sidhu, K.E. Mate <strong>and</strong> J.C.RodgerMarsupial CRC, Macquarie University, Sydney, Australia 2109Australian marsupial sperm attain T-shape orientation in <strong>the</strong> oviduct (1,2) as also during in vitro oviduct co-culturesystems (3-5). The T-shape sperm in <strong>the</strong> brushtail possum were able to bind, penetrate <strong>and</strong> fuse with eggs (6,7). Herewe purpose a hypo<strong>the</strong>sis explaining <strong>the</strong> mechanism by which sperm turn T-shape during capacitation. MATERIALSAND METHODS The preparation of oviduct-conditioned media (CM) <strong>and</strong> induction of T-shape orientation in spermwere as described previously (2-6). Ionic calcium in CM was estimated using iSTAT chip. The effects of tyrosinephosphorylation <strong>and</strong> various stimulators of cAMP pathways on sperm were according to Sidhu et aI. (unpublished).RESULTS AND DISCUSSION The calcium levels in <strong>the</strong> possum oviduct is maintained at ~lmM as also in <strong>the</strong>oviductal CM produced in vitro. Estimating calcium in batches of CM indicated that <strong>the</strong> low levels of calcium areessential <strong>for</strong> optimal fertilization in <strong>the</strong> possum. Ejaculated wallaby semen contains 1.45-mM of calcium <strong>and</strong> sperm arestreamlined, unable to fertilize eggs. Resuspending <strong>the</strong> wallaby ejaculated sperm in calcium-free buffer i.e. TALP <strong>and</strong><strong>the</strong> possum epididymal sperm in low calcium buffer i.e. (pSOF media, calcium -0.7 mM) turned <strong>the</strong>m to T-shapewithin 2 h <strong>and</strong> that was reversible with 1.45 mM calcium. Increasing cAMP or tyrosine phosphorylation in sperm alsoturned <strong>the</strong>m to T-shape orientation within 1 h. In <strong>the</strong> possum, >80% sperm turn T-shape in pSOF media <strong>and</strong> <strong>the</strong>sesperm were unable to penetrate zona. Thus T-shape in sperm is not <strong>the</strong> sole criterion <strong>for</strong> capacitation. These resultsdemonstrate that low levels of calcium are essential <strong>for</strong> obtaining T-shape orientation in sperm <strong>and</strong> <strong>the</strong> latter involvescAMP <strong>and</strong> tyrosine phosphorylation pathways <strong>and</strong> are essential <strong>for</strong> fertilization. ACKNOWLEDGEMENT Thisstudy was supported by <strong>the</strong> Marsupial CRC Australia <strong>and</strong> MAF Policy New Zeal<strong>and</strong>. REFERENCES IBed<strong>for</strong>d, J.M.& Breed, W.O. (1994). BioI. Reprod. 50, 845-854. 2Molinia, et aI. (1998). J.Reprod. Fertil. 112, 9-17. 3Sidhu, et aI.(1998). lReprod. Fertil. 114, 55-61. 4Sidhu, et aI. (1999a). BioI. Reprod. 61, 1356-1361. 5Sidhu, et al. (1999b).Reprod. FertiI. Dev. 11, 329-336. 6Mate et aI. (2000). Zygote 8, 189- 196. 7Sidhu et aI. (2001). Proc. Aust. Soc. Reprod.BioI. 32, 22. ksidhu@possum.bio.mq.edu.au4647


11. PROLACTIN IS A PREIMPLANTATION GROWTH FACTOR IN MICEK. Markham <strong>and</strong> P.L. KayeSchool ofBiomedical Sciences, University ofQueensl<strong>and</strong>, Brisbane QLD 4074Many hormones, cytokines <strong>and</strong> growth factors support optimal development of <strong>the</strong> preimplantation embryo. The closephysiological relationship (1) between growth hormone, which has already been identified as a preimplantation growthfactor (2) <strong>and</strong> prolactin, <strong>and</strong> evidence from gene knock-out models, implicated prolactin (3) <strong>and</strong> its receptor (4) inpreimplantation development. The effects of prolactin on growth of mouse embryos in vitro were <strong>the</strong>re<strong>for</strong>einvestigated. Two-cell embryos were collected from superovulated Quackenbush mice 48 h post-hCG <strong>and</strong> incubatedwith ovine prolactin <strong>and</strong>/or antibodies against prolactin or <strong>the</strong> prolactin receptor <strong>for</strong> approximately 2 days. Grossmorphological development <strong>and</strong> <strong>the</strong> number of cells in blastocysts <strong>and</strong> <strong>the</strong>ir component tissues were assessed byspreading 96 h post hCG blastocysts <strong>and</strong> nuclear staining. Confocal laser scanning immunohistochemistry (CLSIH) asdescribed (2) demonstrated expression <strong>and</strong> localisation of prolactin <strong>and</strong> <strong>the</strong> prolactin receptor. Prolactin increased <strong>the</strong>total number of cells in <strong>the</strong> blastocyst. Concentration studies showed prolactin to be extremely potent in this role withEC50 < 1 pgfml <strong>and</strong> concentrations above 10 pgfml to be less effective. Addition of ei<strong>the</strong>r of <strong>the</strong> blocking antibodiesreduced cell number in blastocysts developing in <strong>the</strong> absence of exogenous prolactin <strong>and</strong> blocked <strong>the</strong> effects of addedprolactin. CLSIH showed prolactin to be present in blastocysts that had been cultured 24 h. The receptors were presentin oocytes <strong>and</strong> embryos throughout <strong>the</strong> preimplantation developmental stages, coincident with mRNA expression.This suggests that embryos express prolactin receptors that are responsive to prolactin secreted in an embryonicautocrine or paracrine regulation of cell number during preimplantation development. The predominantlytrophectodermal location of prolactin receptors in <strong>the</strong> late blastocyst suggests that <strong>the</strong>se receptors may be associatedwith regulation ofimplantation a major function of <strong>the</strong>se cells.1. Waters MJ <strong>and</strong> Kaye PL (2002). Growth hormone & IGF research. (in press)2. Pantaleon M, et al, (1997) Proc Natl Acad Sci USA. 94(10):5125-30.3. Horseman ND, et aI, (1997) EMBO J. 16(23):6926-35.4. Orm<strong>and</strong>y CJ, et aI, 1997 Genes Dev. 11(2):167-78.Supported by NHMRC.13. EFFECT OF IN VITRO OXYGEN CONCENTRATION ON GLUCOSETRANSPORTER-1 EXPRESSION IN THE MOUSE BLASTOCYSTKaren L. Kind, Rachael A. Collett, Alex<strong>and</strong>ra J. Harvey <strong>and</strong> Jeremy G. ThompsonReproductive Medicine Unit, Department ofObstetrics <strong>and</strong> Gynaecology, University ofAdelaide, The Queen ElizabethHospital, Woodville, SA, 5011.Preimplantation embryos develop in vivo under conditions of low oxygen. Oviductal <strong>and</strong> uterine oxygen concen~ationsare less than half that of atmospheric levels, with uterine O 2 concentrations of 3-5% reported around <strong>the</strong> time ofblastocyst <strong>for</strong>mation in several species (1). Decreasing <strong>the</strong> concentration of oxygen during in vitro culture, to moreclosely resemble in vivo conditions, facilitates embryo development (2) <strong>and</strong> <strong>the</strong>se effects maybe mediated throughaltered gene expression. Mouse preimplantation embryos express mRNA <strong>for</strong> hypoxia-inducible factor-la (3), anoxygen-regulated transcription factor which activates <strong>the</strong> expression of genes involved in <strong>the</strong> adaptation to low oxygen,including glucose transporter-l (GLUTl). The aim of this study was to determine whe<strong>the</strong>r oxygen concentra~onregulates expression of GLUTI in <strong>the</strong> mouse embryo. One-cell embryos were collected from CBAxC57B16 Fll111ce22 hours post hCG injection. Embryos were cultured in KSOM media under 7% O 2 ,6% CO 2 , 87% N 2 • After 48 hours,compact morulae were transferred to· 2%, 7% or 20% O 2 <strong>and</strong> cultured <strong>for</strong> a fur<strong>the</strong>r 40 hours to <strong>the</strong> blastocyst stage.Embryos were snap frozen <strong>for</strong> RNA extraction. In vivo developed blastocysts were collected 88 hours post hCG.Extracted RNA was reverse transcribed <strong>and</strong> <strong>the</strong> abundance of GLUT1 mRNA <strong>and</strong> 18S rRNA was analysed by real-timeRT-PCR using SyBr Green Master Mix in a GeneAmp 5700 Sequence Detection System (Applied Biosystems).GLUT1 results were normalised to 18S rRNA abundance. Oxygen concentration influenced GLUTI gene expression(P


15. PARACRINE PERTURBATIONS ASSOCIATED WITH BOVINE NUCLEARTRANSFER PREGNANCIESSusan Ravelich a , Bernhard Breier a , Jim Peterson b , Jeffery Keelan a , David Wells b , Rita Lee baThe Liggins Institute, Faculty ofMedical <strong>and</strong> Health Sciences, University ofAuckl<strong>and</strong>, b Reproductive Technologies,AgResearch, Ruakura Research Centre, Hamilton, New Zeal<strong>and</strong>The regulation of fetal <strong>and</strong> placental growth <strong>and</strong> differentiation involve a variety of endocrine, autocrine/<strong>and</strong> orparacrine signals, including <strong>the</strong> insulin-like growth factor family (IGFs) <strong>and</strong> leptin. Increasing evidence suggests thatparacrine signalling plays a fundamental role in placental growth <strong>and</strong> development. Alterations in <strong>the</strong> paracrine systemin <strong>the</strong> endometrium may underlie pathological processes such as infertility, preeclampsia or endometrial neoplasia(l)·Abnormal placental development <strong>and</strong> function <strong>and</strong> abnormal fetuses have been associated with in vitro derivedembryos <strong>and</strong> especially embryos generated by nuclear transfer (NT) (2). The aim of <strong>the</strong> research was to explore <strong>the</strong>hypo<strong>the</strong>sis that paracrine factors involved in <strong>the</strong> regulation of normal placental growth, development <strong>and</strong> function aredysregulated in cattle pregnant with embryos generated by NT. Reproductive tracts from pregnant Friesian cows wereobtained at days 50, 100 <strong>and</strong> 150 of gestation following embryo transfer with NT or in vitro produced (IVP) embryosor following artificial insemination (AI). Placental localisation of insulin like growth factor binding proteins 2 <strong>and</strong> 3(IGFBP-2, -3) <strong>and</strong> leptin was per<strong>for</strong>med by immunohistochemistry. Spatial <strong>and</strong> temporal differences in <strong>the</strong> localisationof IGFBP-2, -3 <strong>and</strong> leptin were observed in placental tissues from NT compared with AIIIVP pregnancies. At day 50<strong>and</strong> 100 of gestation, increased immunostaining <strong>for</strong> IGFBP-2 was seen in <strong>the</strong> stromal tissue below <strong>the</strong> p1acentomes inNT pregnancies: Increased staining <strong>for</strong> IGFBP-3 was observed in uterine stromal tissue in NT pregnancies at all timepoints examined. Leptin immunostaining in <strong>the</strong> fetal villi in NT pregnancies was markedly increased at day 50 <strong>and</strong> 150of gestation. In summary, <strong>the</strong> spatial <strong>and</strong> temporal patterns of expression of a number of growth factors involved inplacental <strong>and</strong> fetal growth is altered in placental tissues from NT pregnancies. It is likely that placental dysfunction inNT pregnancies is associated with paracrine perturbations via a mechanism of action as yet unknown. (1) Reis FM,Cobellis L, Luisi S, Driul L, Florio P, Faletti A, Petraglia F. (2000) Paracrine/autocrine control of female reproduction.Gynecol Endocrinol 2000 Dec;14(6):464-75 (2) Wells, D. N., Misica, P. M., <strong>and</strong> Tervit, H. R. (1999) Production ofcloned calves following nuclear transfer with cultured adult mural granulosa cells. Biology of<strong>Reproduction</strong> 60 (4), 996­1005.16. SURVIVAL OF FRESH AND VITRIFIED SHEEP IVP AND CLONED EMBRYOSAFTER EMBRYO TRANSFERTeija T. Peura, Katrina M. Hartwich, Jennifer M. Kelly, Hamish M. Hamilton, David O. Kleemann, Skye R.Rudiger, Simon K. WalkerSouth Australian Research <strong>and</strong> <strong>Development</strong> Institute, Turretfield Research Centre, Rosedale 5350, SAThe aim of this study was to examine embryo survival rates following <strong>the</strong> transfer of in vitro produced (IVP) <strong>and</strong>cloned sheep embryos cryopreserved using <strong>the</strong> Open Pulled Straw (OPS) vitrification method (1). Initially, vitrifiedDay 6 IVP blastocysts were transferred to recipients (two embryos per recipient) ei<strong>the</strong>r within 1-2 h (=DELAYED; 34embryos) or 2 min (=DIRECT; 30 embryos) of warming. No significant differences in pregnancy or fetal survival rates(76.5 <strong>and</strong> 66.7%; 52.9 <strong>and</strong> 46.7%, respectively) were observed between <strong>the</strong> methods. Both procedures were used incloning experiments involving ei<strong>the</strong>r serum-starved or actively growing adult granulosa <strong>and</strong> fibroblast donor cells <strong>and</strong>both in vivo <strong>and</strong> in vitro derived oocytes. Recipient pregnancy rates <strong>and</strong> fetuses going to term <strong>for</strong> in vivo-fresh (178embryos), in vivo-OPS-DELAYED (15 embryos), in vitro-fresh (103 embryos), in vitro-OPS-DELAYED (113embryos) <strong>and</strong> in vitro-OPS-DlRECT (28 embryos) groups were 53.7,83.3,43.1,35.4 <strong>and</strong> 14.3% <strong>and</strong> 12.9,33.3, 15.5,5.8 <strong>and</strong> 0%, respectively. With in vivo derived embryos, fresh embryos yielded surprisingly significantly poorer resultsthan vitrified embryos, although <strong>the</strong>re were very small numbers in <strong>the</strong> latter group. Vnlike <strong>the</strong> initial observations onIVP-embryos, <strong>the</strong> pregnancy rates achieved with in vitro-derived cloned embryos were significantly lower with OPS­DIRECT method as compared with OPS-DELAYED or fresh transfers, <strong>and</strong> fetal survival rates were significantly lowerwith both OPS-methods compared with fresh transfers. These results suggest that in vivo-derived cloned embryosendure vitrification better than in vitro-derived clones, <strong>and</strong> that <strong>the</strong> delayed transfer after OPS-vitrification is <strong>the</strong>preferred option <strong>for</strong> somatic cell cloned sheep embryos. (1) Vajta G, Kuwayama M, Holm P, Booth P, Jacobsen H,Greve T, Callesen H. A new way to avoid cryoinjuries of mammalian ova <strong>and</strong> embryos: <strong>the</strong> OPS-vitrification. MolReprod Dev 1998; 51: 53-5817. ABNORMAL TROPHOBLAST MHC-I EXPRESSION AND ASSOCIATEDENDOMETRIAL LYMPHOCYTIC RESPONSE IN BOVINE NUCLEAR TRANSFERPREGNANCIESJonathan R. Hill l , Patricia J. Fisher 2 , Christopher J. Davies 2 <strong>and</strong> Donald H Schlafer 2Departments ofClinical Science/ <strong>and</strong> Biomedical Sciencei, Cornell University, Ithaca, New York, USA.Somatic cell nuclear transfer (NT) has produced healthy "cloned" offspring in 7 mammalian species. Few NT embryoshowever, ever reach this stage as less than 2 out of every 100 NT embryos transferred into recipient females result inviable offspring (compared to 601100 naturally fertilized embryos). In cattle, many of <strong>the</strong>se losses occur around <strong>the</strong>time of placental attachment from <strong>the</strong> fourth week of gestation. Expression of major histocompatibility complex class I(MHC-I) by trophoblast cells <strong>and</strong> distribution of endometrial T-Iymphocyte numbers were investigated as part of anassessment of <strong>the</strong> immunological status of NT pregnancies. Six 5 week old NT pregnancies were generated, all derivedfrom <strong>the</strong> same fetal cell line. All 6 NT placentas displayed trophoblast MHC-I expression. None of 8 controls (4-7weeks old) showed any MHC-I expression. Numbers of T lymphocytes (CD3 positive) were significantly higher in <strong>the</strong>endometrium of <strong>the</strong> majority of NT pregnancies compared with controls. Trophoblast MHC-I expression is nonnallysuppressed during early gestation, <strong>and</strong> <strong>the</strong> observed MHC-I expression in <strong>the</strong> cloned pregnancies is likely to haveinduced a maternal lymphocytic response that would be detrimental to maintaining viability of <strong>the</strong> NT pregnancy.18. CONCENTRATION OF PROGESTERONE AND SUPEROVULATORY RESPONSEWITH DIFFERENT DOSAGES OF FSHLuciene Lomas Santiago l ,2, Ciro Alex<strong>and</strong>re Alves Torres!, Eduardo Terra Nogueira l ,2, Antonio Bento Mancio 1<strong>and</strong> Graeme B. Martin 2IDepartment ofAnimal Science, University Federal ofVit;osa, Vifosa - Brazil.2 School ofAnilnal Biology, University ofWestern Australia, Crawley 6009.Seventeen Nelore (Bos indicus indicus) heifers were superovulated from Day 9 to 12 of <strong>the</strong> estrous cycle using twodifferent dosages of FSH (250 or 500 VI). The ojective was to measure <strong>the</strong> plasma concentration of progesteroneduring superovulation <strong>and</strong> embryo collection, <strong>and</strong> link <strong>the</strong> values to follicular dynamics <strong>and</strong> superovulatory responsethat were monitored daily by ultra-sound until <strong>the</strong> first insemination <strong>and</strong> embryo collection. Concentration ofprogesterone was analyzed every three days from Day 0 (estrous) until Day 12, at first artificial insemination <strong>and</strong> atembryo collection. Plasma was only analyzed in heifers that responded to superovulation. Heifers that displayed a lowconcentration of progesterone,. suggesting <strong>the</strong> absence of a functional corpus luteum in <strong>the</strong> beginning of <strong>the</strong>superovulatory protocol, didn't have satisfactory results to FSH administration. However, more viableembryos/collection were not associated with higher plasma levels of progesterone on <strong>the</strong> day of first administration ofFSH, as would be expected, but were related to individual heifer response. The plasma concentration of progesteroneon <strong>the</strong> day of insemination was similar between heifers superovulated with 250 or 500 VI ofFSH (Tab. 1). However, on<strong>the</strong> day of embryo collection (d=7), <strong>the</strong> levels were inversely correlated to FSH dosage <strong>and</strong> directly related to <strong>the</strong>number of corpora lutea following superovulation (11.25 x 8.25 CL/donor). We conclude that superovulation with 250VI of FSH was more efficient <strong>for</strong> stimulating more responsiveness follicles <strong>and</strong> <strong>the</strong> appearance of more corpora lutea;as a result, <strong>the</strong> secretion of progesterone was higher. Associated with this, all animals superovulated with 250 VI ofFSH responded to <strong>the</strong> superovulatory protocol.Table 1 - Plasma concentration of progesterone in Nelore heifers on <strong>the</strong> day of artificial insemination <strong>and</strong> <strong>the</strong> day ofembryo collection.Progesterone(ng/mL)Cycle day 250 VI FSH 500 VI FSHd=O (Artificial InsemmatlOn . .) 051 .'aA 0.35 a ,Ad-7 (Embryos collection) 41.29 a ,B 17.40 b ,BAverages followed by different lower case letter in same row are different (p


19. EFFECT OF SEMINAL PLASMA ON FROZEN·THAWED BOAR SPERMR Bathgate, BM Eriksson, WMC Maxwell <strong>and</strong> G EvansFaculty ofVeterinary Science, The University ofSydney, NSW 2006Under optimal conditions, fertility after insemination with frozen boar spenn can be close to that achieved with chilledsemen but, in <strong>the</strong> field, <strong>the</strong> levels are often substantially lower. We investigated <strong>the</strong> addition of seminal plasma (SP),reported to have beneficial effects in o<strong>the</strong>r species, be<strong>for</strong>e <strong>and</strong> after freeze-thawing, with <strong>the</strong> aim of improving postthawquality of spenn. The sperm-rich fraction of 3 ejaculates from each of 4 boars were collected <strong>and</strong> frozen,packaged in 0.5ml straws, according to <strong>the</strong> method of Bwanga et a1. (1). The experimental groups had cooling extendersupplemented with 100, 50 or 0% SP (st<strong>and</strong>ard protocol). After thawing, <strong>the</strong> samples were diluted inBTS containing0, 5, 10, 20, 40, 80 or 100% SP. Post-thaw addition of SP in <strong>the</strong> range of 5-40% had a beneficial effect on initialmotility <strong>and</strong> after 3h incubation at 37°C (p


23. THE EFFECT OF NUTRITION DURING THE CYCLE OF MATING ONOVIDUCTAL FLUID AMMONIA AND UREA LEVELS IN THE EWEM. A. Kakar\ S. Maddocks\ M. F. Lorimer 2 , D. o. Kleemann 3 , S. R. Rudiger 3 <strong>and</strong> S. K. Walker 3lDepartment ofAnimal Science, Adelaide University Roseworthy Campus, Roseworthy, SA 5371;2Biometrics SA, Adelaide University, Private Mail Bag1.GlenOsmond.SA 5064;3South Australian Research <strong>and</strong> <strong>Development</strong> Institute, Turretfield Research Centre, Rosedale, SA 5350This study examined <strong>the</strong> effect of changes in nutrition during <strong>the</strong> cycle of mating in <strong>the</strong> ewe on <strong>the</strong> concentrations ofammonia <strong>and</strong> urea in oviductal fluid. Twelve mature ewes (4-5 years, 58-67 kg) of comparable body condition werefed rations that provided ei<strong>the</strong>r 1.5 x daily energy needs <strong>for</strong> maintenance (H) or 0.5 x maintenance needs (L).Nutritional treatments were imposed from 18 d be<strong>for</strong>e until 6 d after <strong>the</strong> expected time of ovulation. Oviducts wereca<strong>the</strong>terised <strong>for</strong> four days be<strong>for</strong>e <strong>the</strong> start of <strong>the</strong> experiment <strong>and</strong> oviduct fluid was collected from day 0 until day 6 afterovulation. Half of <strong>the</strong> ewes were treated to superovulate (S) (using progestagen, FSH, GnRH treatment) while <strong>the</strong>remainder (NS) ovulated spontaneously following progestagen treatment. The data were analysed by ANOVA as a 2x2factorial design. Body weight was significantly (P


27. ANDROGEN RECEPTOR EXPRESSION IN OVARIAN EPITHELIUM IN MICE OFDIFFERING AGES AND TOTAL OVULATION NUMBER.Olivia L. Tan, Peter R. Hurst, Jean S. FlemingDepartment ofAnatomy & Structural Biology, Otago School ofMedical Sciences, PO Box 913, Dunedin, New Zeal<strong>and</strong>.One hypo<strong>the</strong>sis of ovarian surface epi<strong>the</strong>lial (OSE) carcinogenesis is that high levels of progesterone are protective,whereas high levels of <strong>and</strong>rogen increase <strong>the</strong> risk of ovarian cancer (1). Epi<strong>the</strong>lial-lined inclusion cysts are common inall women <strong>and</strong> are thought to be precursors of ovarian cancer (2). This study aimed to investigate spatial differences inexpression of <strong>and</strong>rogen receptor (AR) in <strong>the</strong> OSE <strong>and</strong> inclusion cysts in mice of varying ages <strong>and</strong> total lifetimeovulation number (total OV#).Ovaries from Swiss Webster mice (total OV# median: range [n mice]) were collected from 3 month virgins (3 m;113: 11-235 [55]), from 12 month old breeders (12 m; 217: 97-386 [21]) <strong>and</strong> from 8 month virgin mice, housed in splitcages alongside a male, to induce continuous oestrous cycles (8 m; 629: 456-908 [16]). Ovaries were also collectedfrom prepubertal mice (4 w; 0[9]) (3). AR immunohistochemistry was per<strong>for</strong>med on 4 /lm sections, cut frompara<strong>for</strong>maldehyde-fixed, paraffin-embedded tissue. Invaginations were defined as where OSE descended into <strong>the</strong>ovarian stroma, but remained connected to <strong>the</strong> OSE, while inclusion cysts were enclosed structures within <strong>the</strong> ovary,lined with epi<strong>the</strong>lium. Inclusion cysts were observed only in 8 m <strong>and</strong> 12 m animals.AR expression was low in OSE from 3 m <strong>and</strong> 12 m animals, with very few squamous cells staining. However in 4w <strong>and</strong> 8 m animals <strong>the</strong>re were very few positive cuboidal cells also. In contrast <strong>the</strong> majority of cells lining inclusioncysts, with <strong>the</strong> exception of attenuated cells, were stained <strong>for</strong> AR. Low levels of AR expression were seen ininvaginated OSE in 12 m animals. No differences were seen between invaginated epi<strong>the</strong>lium <strong>and</strong> OSE in 3 m or 8 manimals. The strong upregulation of AR expression in cyst epi<strong>the</strong>lial cells suggests that <strong>and</strong>rogen responsiveness mayplaya role in <strong>the</strong> aetiology of ovarian cancer.1. Risch HA (1998) Journal National Cancer Institute, 90, issue 23, 1774-17862. Blaustein A (1981) Pathology Annual, part 1,247-2493. Clow OL et al (2002) Molecular <strong>and</strong> Cellular Endocrinology 191(1), 105-11129. EOSINOPHILS IN DEVELOPMENT AND FUNCTION OF REPRODUCTIVETISSUESAm<strong>and</strong>a Sferruzzi-Perri, Ann Hallett, Sarah Robertson, Lindsay DentDepartments ofMolecular Biosciences <strong>and</strong> Obstetrics <strong>and</strong> Gynaecology University ofAdelaide, Adelaide SA 5000Eosinophils are a distinctive feature of some mucosal tissues, where <strong>the</strong>y may be important in defence againstinfectious agents, but may also have a role in normal physiology by contributing to development <strong>and</strong> maintenance ofepi<strong>the</strong>lial integrity. Several recent studies have indicated that deficiencies in eosinophils or defects in eosinophilrecruitment are associated with delayed postnatal mammary gl<strong>and</strong> morphogenesis [1] <strong>and</strong> perturbations in <strong>the</strong> estrouscycle [2], [3]. These reports led us to examine <strong>the</strong> affect of an over-abundance of eosinophils on <strong>the</strong>se processes. Asinterleukin 5 (IL-5) is central to <strong>the</strong> regulation of eosinophil growth <strong>and</strong> differentiation, eosinophilic IL-5 transgenic(IL-5 Tg) mice <strong>and</strong> normal wild type (WT) mice were compared to determine <strong>the</strong> impact of eosinophils on mammarygl<strong>and</strong> development <strong>and</strong> reproductive function. Eosinophils were increased 13-fold <strong>and</strong> 4-fold in <strong>the</strong> uterus <strong>and</strong>mammary gl<strong>and</strong>, respectively in IL-5 Tg mice compared to WT littermates. High numbers of eosinophils were alsonoted in <strong>the</strong> regressing corpora lutea of ovaries in IL-5 Tg mice whereas eosinophils were rarely seen in WT ovaries.Mammary gl<strong>and</strong> development was transiently delayed in IL-5 Tg mice, with significantly less ductal extension <strong>and</strong>branching at 7 weeks of age. IL-5 Tg mice had significantly shorter estrous cycles than WT mice (4.7±0.4 days versus6.4±0.5 days, respectively, p = 0.03). At day 18 of pregnancy, fetal resorptions are increased by 75% in IL-5 Tgheterozygote matings, <strong>and</strong> a decrease in fetal: placental ratio was observed (mean + SEM =7.54 ± 0.09 in IL-5 Tgmatings versus 8.26 ± 0.10 <strong>for</strong> WT, p < 0.001). Parturition was not compromised in IL-5 Tg mice, as judged by <strong>the</strong>gestation length <strong>and</strong> pregnancy outcomes. The birth weights <strong>and</strong> growth trajectories of pups were not significantlyinfluenced by maternal IL-5 transgene expression. Our studies suggest that IL-5 transgenesis <strong>and</strong>/or eosinophilia mayalter <strong>the</strong> estrous cycle <strong>and</strong> at least transiently delay ductal morphogenesis in <strong>the</strong> murine mammary gl<strong>and</strong>. Eosinophilsrecruited into <strong>the</strong> ovary of IL-5 Tg mice may impact indirectly on both of <strong>the</strong>se parameters. Despite <strong>the</strong>se observations,<strong>the</strong> outcomes of pregnancy, parturition <strong>and</strong> lactation were not compromised by maternal over-expression of IL-5.References: [1] <strong>Development</strong>, 2000. 127(11): p. 2269-82. [2] J Reprod Fertil, 2000. 120(2): p. 423-32. [3]Endocrinology, 2001. 142(10): p. 4515-21.28. LOCALISATION OF EPITOPES ON BRUSHTAIL POSSUM (TRICHOSURUSVULPECULA) ZONA PELLUCIDA 2 PROTEINXianlan. Cui*, Janine Duckworth*, Susie Scobie*, Denise Jones*, <strong>and</strong> Phil Cowan tL<strong>and</strong>care Research, *PO Box 69, Lincoln 8152, New Zeal<strong>and</strong>; tprivate Bag 11052, Palmerston North, New Zeal<strong>and</strong>Immunisation of female possums (Trichosurus vulpecula) with recombinant possum zona pellucida 2 constructs (rZP2)led to 72-75% reduction in fertility, presumably through interference in secondary sperm-egg binding. In order todefine those regions of <strong>the</strong> protein crucial to sperm-egg binding, antigenic regions on possum ZP2 protein weremapped. The amino acid sequence of full-length possum ZP2 protein was used to syn<strong>the</strong>sise a complete set ofoverlapped 15-mer peptides. The peptides were used in a modified enzyme-linked immunosorbent assay (ELISA) toidentify continuous epitopes recognized by antibodies in <strong>the</strong> sera of possums immunised with rZP2 constructs. Thirteenepitopes were located on possum ZP2 protein. Comparisons of <strong>the</strong> ELISA binding patterns of peptides to antibodies in<strong>the</strong> individual sera with <strong>the</strong> fertility status of<strong>the</strong> immunised possums revealed three significant peptide epitopes. One of<strong>the</strong> three epitopes bound to fixed possum spermatozoa from <strong>the</strong> caudal epididymis. Ano<strong>the</strong>r corresponded to a similarregion that has been shown to be integral to fertilisation in <strong>the</strong> mouse. The significance of <strong>the</strong>se three individualepitopes in relation to fertilisation in <strong>the</strong> possum is currently under investigation.5657


SRB FOUNDERS' LECTURE30. EMBRYO DEVELOPMENT AND BIOTECHNOLOGYAlan TrounsonCentre <strong>for</strong> Early Human <strong>Development</strong>, Institute of<strong>Reproduction</strong> <strong>and</strong> Develop11'lent <strong>and</strong> National Biotehnology CentreofExcellence - Centre <strong>for</strong> Stem Cells <strong>and</strong> Tissue Repair, Monash University, Clayton, Vic. 3168Mammalian preimplantation embryos are <strong>the</strong> only stage of development that may be grown in vitro independent of <strong>the</strong>maternal environment. The grown oocyte <strong>for</strong>med in <strong>the</strong> maturing follicle can be extracted from <strong>the</strong> ovary <strong>and</strong> ei<strong>the</strong>rmatured in vitro, or recovered after maturation in vivo, to produce viable embryos with a high developmentalcompetence. This has enabled <strong>the</strong> in vitro production of mammalian embryos that may be fertilized <strong>and</strong> grown to <strong>the</strong>blastocyst stage in culture. While control of <strong>the</strong> germ cell, primary oocyte <strong>and</strong> growing oocyte in <strong>the</strong> preantral follicleis less well understood, it is possible to manipulate ovarian follicular dynamics by ovarian transplantation <strong>and</strong> may becontrolled in <strong>the</strong> future by interference in <strong>the</strong> normal selection mechanisms of primordial follicle recruitment. Data isbeing generated from expression libraries in <strong>the</strong> human <strong>and</strong> o<strong>the</strong>r species to identify <strong>the</strong> key regulatory molecules <strong>for</strong>follicular <strong>and</strong> oocyte recruitment. This may be used to alter <strong>the</strong> reproductive life-span <strong>for</strong> <strong>the</strong> female.The production of oocytes <strong>and</strong> embryos provides <strong>the</strong> basis <strong>for</strong> reproductive biotechnologies in domestic animal species<strong>and</strong> <strong>the</strong> human. Cattle reproductive technologies are well developed <strong>for</strong> production <strong>and</strong> human IVF is <strong>the</strong> foundation<strong>for</strong> <strong>the</strong> treatment of human infertility. The cattle industry accepts embryo production, embryo cryopreservation <strong>and</strong>embryo transfer as an integral part of <strong>the</strong> breeding strategies used. More recently, <strong>the</strong> cloning of animals, particularlycattle, has been explored with variable outcomes. The reprogramming of somatic cells by ooplasm doesn't completelyreset <strong>the</strong> epigenetic regulators of gene expression, so that failure to erase <strong>the</strong> somatic epigenetic signature can result inabnormalities of development, including both placental <strong>and</strong> fetal compartments. This results in high rates ofimplantation failure, fetal growth <strong>and</strong> both anatomical <strong>and</strong> functional abnormalities which are often lethal. However,around 50% of offspring are viable <strong>and</strong> healthy despite <strong>the</strong> altered epigenetics. In <strong>the</strong> second generation <strong>the</strong> epigeneticirregularities are corrected during gametogenesis.The ability to clone animals enables <strong>the</strong> production of transgenic animals in one generation. O<strong>the</strong>r methods oftransgenesis in animals include sperm-mediated transfection, which involves intracytoplasmic sperm injection (ICSI).O<strong>the</strong>r new <strong>and</strong> interesting methods are being designed that involve <strong>the</strong> manipulation of gametogenesis.Human IVF has evolved into a successful treatment <strong>for</strong> female <strong>and</strong> male infertility. IeSI may used <strong>for</strong> testicular sperm<strong>and</strong> elongated spermatids. However, success with round spermatids or <strong>the</strong>ir precursors has not been clinically useful.The new <strong>and</strong> rapidly growing area of genetic diagnosis of point mutations <strong>and</strong> trinucleotide repeat disease inpreimplantation embryos is an important new biotechnology in human medicine. These new molecular techniques arealso being applied <strong>for</strong> aneuploid screening of IVF embryos, reducing <strong>the</strong> need <strong>for</strong> multiple embryo transfer <strong>for</strong>maintaining high pregnancy success rates. It is now possible to fingerprint sibling embryos to test new culture systemsetc. With <strong>the</strong>se capabilities, it is possible to identify genes that correlate with breast cancer <strong>and</strong> HLA type embryos <strong>for</strong>compatible transplants. Expression screening <strong>and</strong> apoptosis markers may also be used <strong>for</strong> determining embryo viability.Preimplantation embryos are also a source of pluripotential embryonic stem (ES) cells. These may be derived fromfertilized oocytes, par<strong>the</strong>nogenetic embryos <strong>and</strong> nuclear transfer embryos. It is of interest that embryos that have littleor no developmental competence can produce ES cell lines with apparently complete ability to produce all types ofterminal tissue types <strong>and</strong> to integrate in vivo into all tissues of interest. This may be similar to <strong>the</strong> ability of some adultcell types to transdifferentiate under experimental conditions in vitro <strong>and</strong> in vivo. These extraordinary cells may enablea new medical biotechnology of cell <strong>the</strong>rapy <strong>for</strong> tissue repair <strong>and</strong> regeneration. These cells may also allow <strong>for</strong> gene<strong>the</strong>rapy to be applied safely <strong>for</strong> <strong>the</strong> treatment of a wide range of genetic diseases.These have been exciting times <strong>for</strong> biological sciences <strong>and</strong> promise a wonderful future research opportunity <strong>for</strong> youngscientists. I thank all <strong>the</strong> scientists who have worked with me over <strong>the</strong> decades <strong>for</strong> <strong>the</strong> joy it has brought me <strong>for</strong> <strong>the</strong>contributions <strong>the</strong>y have' all made. I also acknowledge <strong>the</strong> funding of our creativity by numerous agencies <strong>and</strong>commercial bodies over <strong>the</strong> decades.31. THE IMMUNOLOGICAL PARADOX OF PREGNANCY - SURVIVAL OF THEULTIMATE ALLOGRAFTBruce E. Lovel<strong>and</strong>The Austin Research Institute, Heidelberg, VIC 3084A successful pregnancy requires that <strong>the</strong> mo<strong>the</strong>r accepts a <strong>for</strong>eign body. This involves paradoxes <strong>for</strong> <strong>the</strong> immunesystem. The extensive genetic diversity between mo<strong>the</strong>r <strong>and</strong> offspring provides protein differences that, givenexposure to <strong>the</strong> immune system, range from weakly to very strongly immunogenic. Thus <strong>the</strong> placenta <strong>and</strong> foetusrepresent "<strong>the</strong> ultimate allograft". The highly active metabolic exchange across <strong>the</strong> placenta <strong>and</strong> dynamicdevelopmental changes provide an opportunity <strong>for</strong> strong maternal immunity to develop. Establishment <strong>and</strong>maintenance of pregnancy involves immune tolerance, immunosuppression <strong>and</strong> immunomodulation. A state ofselective maternal-anti-foetal immunosuppression exists which would be <strong>the</strong> dream of organ transplanters to mimic.Global immunosuppression obviously would be detrimental to <strong>the</strong> mo<strong>the</strong>r <strong>and</strong> a strong local anti-microbial immunitymust be maintained.The mammalian immune system has evolved to cohabitat with microbes <strong>and</strong> parasites. It is perceived to consist ofdifferent but overlapping compartments, described as <strong>the</strong> Innate, Humoral <strong>and</strong> Cellular. These provide, respectively,extremely rapid containment <strong>and</strong> destruction of infecting organisms, an induced mechanism to recognise <strong>and</strong> removeparticulate matter, <strong>and</strong> an induced mechanism to recognise <strong>and</strong> destroy infected cells in <strong>the</strong> body. These are achievedthrough quite distinct "pattern recognition" processes. It is apparent that selective control of each of <strong>the</strong>se is crucial tosuccessful pregnancy. A central feature of <strong>the</strong> immune system is <strong>the</strong> need to distinguish "self' from "<strong>for</strong>eign" <strong>and</strong> <strong>the</strong>provision of a variety of mechanisms to achieve this at very subtle levels. Its critical function to screen out mutating,potentially cancerous cells, is also relevant to our consideration of pregnancy <strong>and</strong> immunity. So how is <strong>the</strong> specificsuppression induced?There are several mechanisms thought to be crucial to <strong>the</strong> maternal / foetal immune state - perhaps more to beuncovered. Blocking <strong>the</strong> function of indoleamine 2,3-dioxygenase (IDO), a widely expressed enzyme responsible <strong>for</strong>tryptophan metabolism, results in maternal-anti-foetal immune responses. IDO is currently <strong>the</strong> most fashionablec<strong>and</strong>idate to explain maternal immunosuppression. Secondly, control of <strong>the</strong> complement cascade, central to <strong>the</strong> innateimmune system, is also essential to healthy pregnancy.This presentation will introduce <strong>the</strong> immune mechanisms enabling <strong>the</strong> ultimate allograft, a foetus, to survive <strong>and</strong> thrive.32. PRESENT AND FUTURE OPTIONS FOR PRESERVATION OF TESTIS TISSUE ANDFUNCTIONStefan SchlattInstitute ofReproductive Medicine, University Munster, Domagkstr. 11, 48129 Munster, GermanyThe testis releases two major products: male gametes <strong>and</strong> male sex steroids. While absent or insufficient secretion ofsteroids can easily be treated by administration of exogenous hormones, no cure has been established <strong>for</strong> a defect ingerm cell development. The introduction of ICSI, however, has drastically changed <strong>the</strong> prospects <strong>for</strong> treatment, asgeneration of even a single postmeiotic male germ cell offers a chance of achieving a pregnancy. There<strong>for</strong>e, newapproaches based on male germ cell transplantation <strong>and</strong> testicular tissue grafting might be optimised to work <strong>for</strong> <strong>the</strong>generation of progeny. Interspecies transfer of germ cells showed that <strong>the</strong> recolonisation by stem cells is robust, asspermatogonial stem cells from phylogenetically distant species can recolonise <strong>the</strong> mouse testis. In contrast to <strong>the</strong> factthat spermatogonial stem cells are able to recolonise <strong>the</strong> seminiferous tubules, complete spermatogenesis is onlyobserved when germ cells are transplanted between phylogenetically closely related species. We have developed anapproach to infuse germ cells into monkey <strong>and</strong> human testes <strong>and</strong> tested whe<strong>the</strong>r germ cell transplantation can beapplied as a tool <strong>for</strong> gonadal protection in non-human primates. However, due to <strong>the</strong> risk of tumour cell contaminationgerm cell transplantation will not eliminate <strong>the</strong> cryopreservation of sperm as a widely used tool <strong>and</strong> well acceptedoption <strong>for</strong> gonadal protection in postpubertal oncological patients. It appears likely that optimised techniques willinstead become available to generate male gametes from preserved testicular material. One of <strong>the</strong> promising newapproaches, <strong>and</strong> an alternative to germ cell transplantation, is grafting of testicular tissue. We have shown that testiculartissue from various newborn species grafted ectopically into <strong>the</strong> backskin of immunodeficient mice is capable ofdeveloping qualitatively complete spermatogenesis. Sperm obtained from <strong>the</strong>se grafts have been used <strong>for</strong> <strong>the</strong> generationof progeny. Like <strong>the</strong> more advanced status of ovarian tissue grafting as a tool to protect fertility <strong>and</strong> substitutehormones in female patients, grafting of testicular tissue might become an important tool <strong>for</strong> fertility preservation <strong>and</strong>hormonal replacement <strong>the</strong>rapy in <strong>the</strong> male.5859


33. DOES THE IMMUNE SYSTEM PLAYA ROLE IN REPRODUCTION?A CLINICIANS PERSPECTIVEKelton TremellenReproductive Medicine Unit, Adelaide, South Australia.Human reproduction is an enigma from an immunological perspective since <strong>the</strong> immune system can be responsible <strong>for</strong>infertility <strong>and</strong> pregnancy complications, while facilitating a successful reproductive outcome in <strong>the</strong> majority. In thispresentation, a brief overview of <strong>the</strong> immune systems role in each stage of human reproduction will be presented, alongwith an example of <strong>the</strong> clinical consequences when things go wrong.Cytokines are involved in <strong>the</strong> control of all aspects of ovarian function. including follicle growth, steroid production<strong>and</strong> ovulation. Around <strong>the</strong> time of ovulation a surge in local production of TNF-a, IL-I ~ <strong>and</strong> GM-CSF results in <strong>the</strong>recruitment of macrophages into <strong>the</strong> follicle, which inturn release proteases <strong>and</strong> prostagl<strong>and</strong>ins producing rupture of <strong>the</strong>dominant follicle <strong>and</strong> ovulation. Inhibition of <strong>the</strong>se processes with non-steroidal anti-inflammatory drugs has beenshown to inhibit ovulation <strong>and</strong> potentially result in infertility.The presence of allogenic sperm in <strong>the</strong> female reproductive tract provides an immunological challenge to <strong>the</strong> female. Inmost cases, repeated exposure of a woman to sperm antigens will tollerise her towards her partners antigens, whichinturn will enhance placental invasion <strong>and</strong> decrease her chances of developing pre-eclampsia. Alternatively, adversematernal immune responses to sperm can lead to infertility. Examples include <strong>the</strong> ability of anti-sperm antibodies toblock fertilisation or destruction of sperm by peritoneal macrophages activated by cytokines liberated fromendometriosis.The interaction between <strong>the</strong> embryo, endometrium <strong>and</strong> immune system has been extensively studied in rodents.Cytokines such as II-I, IL-ll, LIF, GM-CSF <strong>and</strong> CSF seem to benefit embryo development <strong>and</strong> implantation, whileIFNy <strong>and</strong> TNFa have a negative influence. While <strong>the</strong>se cytokines may mediate <strong>the</strong>ir action directly on <strong>the</strong> embryoitself, many mediate <strong>the</strong>ir influence indirectly by modifying uterine leukocyte numbers <strong>and</strong> activity. Womenexperiencing recurrent miscarriage of immune aetiology have been identified as having abnormal endometrial cytokineexpression <strong>and</strong> Natural Killer cell activity.The immune system continues to playa role in human reproductive processes long after conception <strong>and</strong> earlyembryonic development. Inappropriate activation of <strong>the</strong> maternal immune system can lead to pre-term labour, fetalgrowth restriction <strong>and</strong> pre-eclampsia. Evidence will also be presented linking adverse maternal immune responses <strong>and</strong>cerebral palsy.34. FINDING SOLUTIONS: SEMEN AND MATERNAL IMMUNE TOLERANCESarah A. RobertsonDepartment ofObstetrics <strong>and</strong> Gynaecology <strong>and</strong> Reproductive Medicine Unit, The University ofAdelaide, SA 5005.Successful pregnancy requires a state of maternal immune 'tolerance' to accommodate antigens expressed by <strong>the</strong>conceptus. Implantation failure <strong>and</strong> placental pathologies such as recurrent miscarriage <strong>and</strong> pre-eclampsia largelyreflect insufficiencies in maternal immune adaptation, but progress in devising <strong>the</strong>rapeutic strategies to treat <strong>the</strong>seconditions is stalled because <strong>the</strong> mechanisms underlying induction <strong>and</strong> maintenance of maternal tolerance are unknown.Increasingly, clinical <strong>and</strong> experimental data support <strong>the</strong> proposal that insemination has consequences <strong>for</strong> <strong>the</strong>reproductive process beyond delivery of male gametes. An emerging hypo<strong>the</strong>sis is that insemination comprises <strong>the</strong>'inductive phase' of<strong>the</strong> maternal immune response to pregnancy, since it is in <strong>the</strong> context of semen that <strong>the</strong> female tractis first <strong>and</strong> most frequently exposed to paternal transplantation antigens shared by <strong>the</strong> conceptus. Our studies in miceindicate (1) that at insemination, <strong>the</strong> maternal tract is better poised to process antigens than at any o<strong>the</strong>r stage of <strong>the</strong>cycle or pregnancy; (2) that semen contains high concentrations of 'immune-deviating' cytokines, includingtrans<strong>for</strong>ming growth factor (TGF)~ <strong>and</strong> prostagl<strong>and</strong>in E, known to induce immune tolerance in o<strong>the</strong>r mucosae; (3) thatexposure to semen is sufficient to induce a transient state of functional immune tolerance to paternal transplantationantigens, <strong>and</strong> (4) that insemination is causally linked to <strong>the</strong> activation <strong>and</strong> expansion of populations of lymphocytesrecruited into <strong>the</strong> implantation site. Recent studies in women show that comparable events occur in <strong>the</strong> human cervixafter intercourse. Toge<strong>the</strong>r, <strong>the</strong>se studies indicate that exposure to semen may promote <strong>the</strong> success of pregnancythrough orchestrating a maternal immune response conducive to optimal placental invasion <strong>and</strong> function. Since it is<strong>the</strong> inductive phase that principally determines <strong>the</strong> strength <strong>and</strong> quality of any immune response <strong>and</strong> is most amenableto <strong>the</strong>rapeutic manipulation, we expect that unravelling <strong>the</strong> cellular <strong>and</strong> molecular processes linking insemination withimplantation will facilitate development ofnew clinical interventions <strong>for</strong> treatment of immune-based fertility disorders.35. ESTRADIOL-MEDIATED CYCLIN D2 EXPRESSION: A ROLE FOR ERa?Ann E. Drummond, Mitzi Dyson <strong>and</strong> Jock K. FindlayPrince Henry's Institute ofMedical Research, PO Box 5152 Clayton, Victoria 3168 AustraliaCyclin D2, a cell cycle regulator, plays a key role in <strong>the</strong> proliferation of ovarian granulosa cells (1) <strong>and</strong> thus follicledevelopment. Follicle stimulating hormone (FSH) <strong>and</strong> estradiol (E2) have been shown to enhance cyelin D2 mRNAexpression in vivo (2). It is unclear however, which estrogen receptor (ER), ERa. or ER~, mediates this response. Weaddressed this question using cultured granulosa cells (3), which exclusively express cyclin D2 (no o<strong>the</strong>r cyclins).Granulosa cells were isolated from <strong>the</strong> ovaries of diethylstilboestrol (DES)-treated, 21 day old Sprague-Dawley rats.Cells in triplicate wells, were plated overnight (day 1), <strong>the</strong> media was changed (day 2) <strong>and</strong> treatments were added onday 3 <strong>for</strong> 2 h. FSH (100ng/ml) <strong>and</strong> E2 (1 <strong>and</strong> IOnM) were added separately <strong>and</strong> in combination. RNA purified fromfreshly isolated <strong>and</strong> cultured granulosa cells, was reverse transcribed <strong>and</strong> subjected to real time PCR analysis of cyclinD2, ERa, ER~ <strong>and</strong> GAPDH (data normalisation) mRNAs. Experiments were repeated three times. Cyclin D2 mRNAexpression by freshly isolated granulosa cells was elevated 4-fold in comparison to control cells plated <strong>for</strong> 2 days. Inresponse to FSH, cyclin D2 mRNA expression was stimulated 2-fold (compared to control cells). In contrast, E2 at 1<strong>and</strong> IOnM, failed to influence cyclin D2 expression. The combined FSH <strong>and</strong> E2 treatment gave results consistent with<strong>the</strong> stimulation of cyclin D2 mRNA by FSH alone. ER~ mRNA expression by granulosa cells did not change with timein culture or in response to treatment. ERa mRNA expression by granulosa cells, down-regulated by in vivo DEStreatment (3), was not restored with time in culture. We hypo<strong>the</strong>sise that ERa not ER~ is involved in <strong>the</strong> regulation ofcyclin D2 expression by E2. The down-regulation of ERa mRNA, induced by <strong>the</strong> in vivo exposure to DES <strong>and</strong> itscontinued decline in vitro, may account <strong>for</strong> <strong>the</strong> inability of E2 to enhance cyclin D2 mRNA expression. These findingsare consistent with our belief that ERs play different roles in <strong>the</strong> ovary: ER~ mediates differentiated functions <strong>and</strong> ERacellular proliferation. [1] Sicinski et aI., (1996) Nature 384: 470-474. [2]Robker et aI., (1998) Molec. Endocrinol.12:924-939. [3]Drummond et aI., Molec. Cell. EndocrinoI. 149: 153-161. Supported by <strong>the</strong> NH&MRC of Australia(Regkey 983212).36. PURIFICATION OF HUMAN GRANULOSA CELLS FROM FOLLICULAR FLUIDSAMPLES FOR USE IN SERIAL ANALYSIS OF GENE EXPRESSION (SAGE)Michael C .I Quinn!, J L Stanton!, P A Hessian 2 , D P L Green!IMolecular Embryology Group, Department ofAnatomy & Structural Biology, University ofOtago, New Zeal<strong>and</strong>2Leukocyte Inflammation Research Laboratory, Department ofPhysiology University ofOtago, New Zeal<strong>and</strong>Follicular fluid collected at <strong>the</strong> time of IVF is a prime source of human granulosa cells. However, follicular fluid oftencontains red <strong>and</strong> white blood cells. This contamination must be removed <strong>for</strong> meaningful in vitro or molecular studies.Previous studies have purified granulosa cells by a combination of density gradients (1), <strong>the</strong> use of magnetic anti-CD45beads (2) <strong>and</strong> also by flow cytometry (3). We propose a purification methodology which exploits clumps of muralgranulosa cells present in follicular fluid. Follicular fluid samples were collected from female patients undergoing IVF<strong>for</strong> male factor infertility at <strong>the</strong> Otago <strong>Fertility</strong> Center, Healthcare Otago. Centrifugation through a 50% Percollgradient (600g) was used to remove red blood cells. Cells were collected from <strong>the</strong> Percoll interface <strong>and</strong> viewed under adissecting microscope. Clumps of granulosa cells were mechanically removed with a glass pipette <strong>and</strong> washed in HamsFlO media. To determine white blood cell content cells were stained with antibodies (CD3, CD14, CD16, CD19) <strong>and</strong>quantified by flow cytometry. White blood cell contamination was found to be between 2 <strong>and</strong> 4%. An FSH antibodyconfirmed purified cells were positive <strong>for</strong> FSH. In addition, cell clumps were examined by histology <strong>and</strong> electronmicroscopy <strong>and</strong> found to contain homogenous cells, which were identified as granulosa cells. This method provides arelatively simple <strong>and</strong> fast method to purify human granulosa cells, which is applicable to molecular studies.(1) Best, C. L., Pudney, J., Anderson, D. J., Hill, J. A. 1994. Modulation of human granulosa cell steroid production invitro by tumor necrosis factor alpha: implications of white blood cells in culture. Obstetrics & Gynecology, 84, 121­127. (2) Choi, D., Lee, E, Y., Yoon, S., Hwang, S., Yoon, B, & Lee, J. 2000. Clinical correlation of cyclin D2 mRNAexpression in human luteinized granulosa cells. Journal ofAssisted <strong>Reproduction</strong> <strong>and</strong> Genetics, 17, 574-579. (3) DeNeubourg, D., Robins, A., Fishel, S., & Gibbon, L. 1996. Flow cytometric analysis of granulosa cells from follicularfluid after follicular stimulation. Human <strong>Reproduction</strong>, 11,2211-2214.6061


37. REMODELLING OF EXTRACELLULAR MATRIX IN BOVINE FOLLICLES ATOVULATIONHelen F. Irving-Rodgers!, Michael J. D'Occhio 2 , William J. Aspden 2 , Raymond J. Rodgers l1Reproductive Medicine Unit, Department ofObstetrics <strong>and</strong> Gynaecology, University ofAdelaide <strong>and</strong> 2Animal Sciences<strong>and</strong> Production Group, Central Queensl<strong>and</strong> University, Rockhampton.Within <strong>the</strong> membrana granulosa we have identified a unique extracellular matrix (fociemix) consisting of basal laminalike material that develops as follicles enlarge (l). To elucidate its role we examined its fate following ovulation, byimmunohistochemical analysis of <strong>the</strong> composition of fociemix <strong>and</strong> <strong>the</strong> follicular basal lamina. Ovulation in Bos indicuscattle (426-562 kg) was synchronized as follows: Day 1; intra-vaginal progesterone releasing device (CIDR) plus 2 mg17~-oestradiol (Cidirol); Day 9; prostagl<strong>and</strong>in (Estrumate); Day 10; removal of CIDR. Animals were. r<strong>and</strong>omlyallocated to 3 groups: Group 1, Control (n =6), no fur<strong>the</strong>r treatment; Group 2 (n =8), injection of 25mg LH (Lutropin)Lm. on Day 11; Group 3 (n = 8), 25 mg i.m. LH on Day 10. Ovaries were collected on Day 12, such that Group 2ovaries were collected 12 - 14 h <strong>and</strong> Group 3 38 - 40.5 h after LH. Pre- or recently-ovulatory follicles were dissectedfrom <strong>the</strong> ovary, in an apical-basal orientation, <strong>and</strong> frozen in OCT compound or fixed in 2.5% glutaraldehyde.Immunohistochemistry was per<strong>for</strong>med on OCT embedded tissue <strong>and</strong> <strong>the</strong> following basal lamina components localised:collagen type IV (a1), laminin chains ~1, ~2, yl, nidogen, perlecan <strong>and</strong> <strong>the</strong> extracellular matrix proteoglycan versican.Group 1 follicles were 13-20 mm in diameter <strong>and</strong> Group 2 follicles 6-18 mm. Pre-ovulatory follicles in Group 3measured 10, 11 <strong>and</strong> 18 mm, <strong>and</strong> o<strong>the</strong>r follicles had ovulated. Following ovulation <strong>the</strong> follicular basal lamina remainedimmunopositive <strong>for</strong> laminin chains ~2, yl <strong>and</strong> nidogen. Laminin ~1 was not detected. Staining <strong>for</strong> <strong>the</strong> a1 chain ofcollagen type IV was reduced compared with basal lamina staining of smaller antral follicles (2), <strong>and</strong> discontinuous inrecently ovulated follicles, indicating degradation of <strong>the</strong> basal lamina. Perlecan was absent from <strong>the</strong> membranagranulosa <strong>and</strong> follicular basal lamina of most Group 3 pre- <strong>and</strong> post-ovulatory follicles as was versican. However,granulosa cells (especially Group 2 follicles) showed intense cytoplasmic staining <strong>for</strong> versican, suggesting it was upregulated. The findings indicate that matrix in <strong>the</strong> basal lamina <strong>and</strong> within <strong>the</strong> membrana granulosa of follicles activelychanges at <strong>the</strong> time of ovulation. (1)Biol Reprod 2001: 64, Suppl 1, Abstract 101. (2)Biol Reprod 1998: 59, 1334­1341.39. THE INFLUCENCE OF THE OOCYTE DURING IN VITRO MATURATION ON THEMETABOLISM OF BOVINE CUMULUS-OOCYTE COMPLEXESMelanie Sutton!, Pablo Cetica 2 , Robert Gilchrist!, Jeremy Thompson l1 The Reproductive Medicine Unit, Department ofObstetrics <strong>and</strong> Gynaecology, University ofAdelaide, The QueenElizabeth Hospital, Woodville, South Australia.2 Department ofBiochemistry, School ofVeterinary Science, University ofBuenos Aires, Argentina.Interactions between <strong>the</strong> oocyte <strong>and</strong> surrounding cumulus cells are bi-directional <strong>and</strong> oocyte-secreted factors haveprofound effects on cell growth <strong>and</strong> differentiation. The aims of this study were to determine <strong>the</strong> influence of <strong>the</strong>oocyte on bovine cumulus cell metabolism during in vitro maturation. Cumulus-oocyte complexes (COC) wereaspirated from antral follicles from abattoir-derived ovaries. Complexes were oocytectomised (OOX), leaving <strong>the</strong>cumulus <strong>and</strong> zona pellucida intact. Intact COC, OOX <strong>and</strong> OOX plus denuded oocytes (OOX+DO) were culturedindividually in 10JlI drops ofIVM media (TCM 199 + pyruvate + BSA + LH + FSH) at 39°C in 5% CO 2 in air. Oxygenconsumption measurements were taken at 1-4h, 11-14h <strong>and</strong> 21-24h of culture using a non-invasive microfluorescencemethod [1]. Spent media was collected <strong>and</strong> analysed <strong>for</strong> glucose <strong>and</strong> pyruvate consumption <strong>and</strong> L-lactate production[2]. The DNA content of individual complexes was quantified using PicoGreen fluorescence dye to enable metabolicmeasurements to be expressed per ng of DNA. There were no significant differences between COC, OOX <strong>and</strong>OOX+DO in any of <strong>the</strong> metabolic parameters measured, providing fur<strong>the</strong>r evidence that oocyte-specific factors, whileregulating cell proliferation, have few measurable effects on bovine cumulus mass expansion [3]. However, regardlessof treatment, metabolic profiles changed over <strong>the</strong> 24h maturation period. There were significant increases (P500 kDa, indicating that large molecular weightmolecules contribute significantly to <strong>the</strong> overall osmotic potential of follicular fluid. Pools were <strong>the</strong>n treated wi<strong>the</strong>nzymes (DNAse, proteinase K, hyaluronidase, chondroitinase ABC, keratanase, collagenase, or heparinase) <strong>and</strong> <strong>the</strong>ndialysed at 100 kDa cut off at 37C in 2M NaCI to prevent aggregation of macromolecules, followed by dialysis at 37°Cagainst H 2 0. Osmotic pressure was reduced 42 ± 2% <strong>and</strong> 33 ± 3% by digestion of hyaluronic acid (0.5u/201l1hyaluronidase from Streptomyces hyalurolyticus, 4 h, 37°C, pH 6) in healthy <strong>and</strong> atretic follicles, repectively. Digestionof GAG chains (0.02u/20IlI, chondroitinase ABC from Proteus vulgaris, 4 h, 37°C, pH 8) resulted in a reduction in OPof 54 ± 14% <strong>and</strong> 20 ± 3.6% in fluid from healthy <strong>and</strong> atretic follicles. This suggests that PGs in follicular fluid, toolarge to readily cross <strong>the</strong> follicular wall, can exert osmotic pressure.accumulation of follicular fluid.These PGs may <strong>the</strong>re<strong>for</strong>e contribute to <strong>the</strong>40. EXPRESSION WITHOUT SECRETION OF TGF-PI AND TGF·p2 BY BOVINE ANDMURINE OOCYTESLesley J Ritter, Megan P Morrissey, David T Armstrong <strong>and</strong> Robert B GilchristReproductive Medicine Unit, Department ofObstetrics <strong>and</strong> Gynaecology, The University ofAdelaide,TQEH, Woodville, South Australia.It is now well established that oocytes are potent regulators of cumulus cell function <strong>and</strong> <strong>the</strong>reby <strong>the</strong>ir ownmicroenvironment. This activity is commonly attributed to members of <strong>the</strong> TGF-~ superfamily because of <strong>the</strong> capacityof <strong>the</strong>se growth factors to mimic oocyte-secreted factors (1). The aims of this study were to examine in bovine <strong>and</strong>murine oocytes (1) <strong>the</strong> expression pattern of TGF-~l <strong>and</strong> ~2 mRNA throughout meiotic maturation, <strong>and</strong> (2) whe<strong>the</strong>roocytes secrete TGF-~1 <strong>and</strong> ~2. Follicular cells <strong>and</strong> oocytes were collected from abattoir derived bovine ovaries <strong>and</strong>from eCG-primed immature mice. RNA was extracted from follicular cells <strong>and</strong> from oocytes at defined stages ofmeiotic maturation; in mouse from both in vivo <strong>and</strong> in vitro matured oocytes at 0, 3, 6, 9 <strong>and</strong> 24h, <strong>and</strong> in cows fromimmature <strong>and</strong> in vitro matured oocytes. RNA was reverse transcribed <strong>and</strong> cDNA was amplified with specific primerstargeted against TGF-~l <strong>and</strong> ~2. Both bovine <strong>and</strong> murine denuded oocytes (DO) expressed TGF-~l <strong>and</strong> ~2 mRNA atall stages of meiotic maturation. All somatic cells examined expressed mRNA <strong>for</strong> both TGF-~ transcripts. Thepossibility of CC contamination of DO .samples was excluded by screening <strong>for</strong> FSH receptor· mRNA expression byPCR. In order to determine whe<strong>the</strong>r DO secrete TGF-~ proteins, mural granulosa cells (MGC) from both species werecultured in <strong>the</strong> presence of ei<strong>the</strong>r DO or TGF-~ +/- a TGF-~ pan specific neutralising antibody. MGC were assessed <strong>for</strong>[3H]-thymidine incorporation as a marker of DNA syn<strong>the</strong>sis. eH] counts were increased significantly (P


T If41. DEVELOPMENT AND ANTRUM FORMATION IN MARSUPIAL PREANTRALFOLLICLES CULTURED IN A SERUM-FREE MEDIUMN M Richings, P D Temple-Smith & M B RenfreeDepartment ofZoology, University ofMelboume, Vic, 3010Culture of isolated preantral follicles is potentially an important source of mature eggs <strong>for</strong> research <strong>and</strong> assistedreproductive techniques, where an underst<strong>and</strong>ing of both <strong>the</strong> prepubertal <strong>and</strong> adult system is important. In <strong>the</strong> onlypublished report of follicle culture in a marsupial species [1], <strong>the</strong> culture system supported follicular growth but noantral development was observed. The aim of this study was to compare <strong>the</strong> growth <strong>and</strong> development of preantralfollicles from prepubertal <strong>and</strong> adult tammar wallabies, Macropus eugenii, in a serum-free culture system. Preantralfollicles were dissected from ovaries of 6 prepubertal <strong>and</strong> 5 adult tammars <strong>and</strong> cultured individually in drops ofmedium (Dulbecco's modified essential medium with various additives) at 37°C in a humidified gas environment of 6%CO 2in air <strong>for</strong> 4 days. Follicles were measured <strong>and</strong> observed each day. Growth rates were expressed in terms ofproportional increase in volume over <strong>the</strong> culture period <strong>and</strong> <strong>the</strong> follicles were assessed <strong>for</strong> signs of antrumdevelopment. Almost all follicles increased in volume (Prepubertal, 53/54; Adult, 54/55) <strong>and</strong> <strong>the</strong>re was no significantdifference in <strong>the</strong> average volume increase between groups (Prepubertal, 48%; Adult, 59%; p>0.05). There was,however, a significant difference between groups in <strong>the</strong> incidence of antrum <strong>for</strong>mation (Prepubertal, 10/54; Adult, 0/55;p1000 OSE cells were found to be BrdU positive after injections into <strong>the</strong> animals. In culture,however, <strong>the</strong> OSE showed increasing BrdU incorporation with 5%, 20% <strong>and</strong> over 50% labelled cells at 24,48 <strong>and</strong> 72hr. respectively. No o<strong>the</strong>r tissues within <strong>the</strong> ovary were found to have significant numbers of labelled cells. In a fewexamples <strong>the</strong> meso<strong>the</strong>lial lining of <strong>the</strong> ligament suspending ei<strong>the</strong>r <strong>the</strong> ovary or oviduct also had labelled cells. Theseresults indicate <strong>the</strong> considerable potential <strong>for</strong> proliferation of OSE cells in vitro. BrdU incorporation may be induced byan OSE-specific proliferative factor, or factors present in <strong>the</strong> culture medium. Alternatively <strong>the</strong> removal of <strong>the</strong> ovaryfrom <strong>the</strong> mouse may eliminate an inhibitory influence or be a wound-response to <strong>the</strong> act of dissection of <strong>the</strong> ovary.Whatever <strong>the</strong> reason, it remains to be determined if <strong>the</strong> cells of <strong>the</strong> OSE can proceed in vitro to enter <strong>and</strong> completemitotic division.44. IN VITRO REGULATION OF ACTIVIN / INIDBIN SUBUNITS, FOLLISTATIN ANDBAMBI IN THE 3 DAY OLD RAT TESTIS BY ACTIVIN, FOLLISTATIN AND FSHlTerri Meehan, 2Ann Drummond, IDavid de Kretser <strong>and</strong> lKate Lovel<strong>and</strong>1Monash Institute of<strong>Reproduction</strong> <strong>and</strong> <strong>Development</strong>, Monash University, <strong>and</strong> 2 Prince Henry's Institute ofMedicalResearch, Clayton, Victoria 3147Members of <strong>the</strong> trans<strong>for</strong>ming growth factor (TGF)P superfamily <strong>and</strong> <strong>the</strong>ir antagonists are produced by several celltesticular types during early postnatal development. We have observed a discrete switch from activin to follistatinexpression within gonocytes as <strong>the</strong>y trans<strong>for</strong>m into spermatogonia, <strong>and</strong> we have illustrated <strong>the</strong>ir functional effect onsomatic <strong>and</strong> germ cells (Meehan et al 2000. Dev BioI 220:225). This lead to <strong>the</strong> hypo<strong>the</strong>sis that <strong>the</strong>se proteins mayregulate <strong>the</strong>ir own expression <strong>and</strong> <strong>the</strong>reby modulate this stage of testicular differentiation. Using 24 hour cultures of 3day old rat testis fragments <strong>and</strong> real-time PCR analysis, expression levels of activin <strong>and</strong> inhibin subunits (a, pA <strong>and</strong>PB), follistatin <strong>and</strong> BAMBI mRNAs were measured in response to activin A, follistatin <strong>and</strong> FSH relative to mediaalone. BAMBI is a pseudoreceptor that antagonises signalling by activin, TGFp<strong>and</strong> some bone morphogenetic proteins,while both follistatin <strong>and</strong> inhibin can inhibit activin signalling. The inhibin a subunit rnRNA was increased 3-fold byFSH (to 280% of control, p


45. REGULATION OF SERTOLI CELL ACTIVIN AND INHffiIN SECRETION BYHORMONES, CYTOKINES AND THE SEMINIFEROUS EPITHELIAL CYCLEYoshinori Okuma, Anne O'Connor, Julie Muir, David Phillips, David de Kretser <strong>and</strong> Mark HedgerCentre <strong>for</strong> Molecular <strong>Reproduction</strong> <strong>and</strong> Endocrinology, Monash Institute of<strong>Reproduction</strong> <strong>and</strong> <strong>Development</strong>, Clayton,VictoriaActivin A <strong>and</strong> <strong>the</strong> inflammatory cytokines, interleukin-la (IL-la) <strong>and</strong> IL-6, are produced by <strong>the</strong> Sertoli cell <strong>and</strong> areimplicated in control of spermatogonial <strong>and</strong> spermatocyte development. Activin A also inhibits IL-l <strong>and</strong> IL-6production <strong>and</strong> action in several o<strong>the</strong>r systems, <strong>and</strong> <strong>the</strong>re<strong>for</strong>e is a potential negative regulator of testicularinflammation. However, <strong>the</strong>re have been few studies describing <strong>the</strong> production <strong>and</strong> regulation of activin A in <strong>the</strong> testis.Sertoli cells from immature (20 day-old) or adult rats were cultured <strong>for</strong> 48h in <strong>the</strong> presence of IL-la, IL-l~, IL-lreceptor antagonist (IL-lra), IL-6, <strong>the</strong> inflammatory mediator lipopolysaccharide (LPS from E. coli), testosterone<strong>and</strong>/or ovine FSH or dibutyryl cAMP (cAMP). Adult rat seminiferous tubules were stage-dissected <strong>and</strong> incubated withtest substances <strong>for</strong> 72h. Dimeric activin A <strong>and</strong> inhibin B in <strong>the</strong> culture media were measured by specific ELISAs. Inimmature <strong>and</strong> adult Sertoli cells, activin A secretion was stimulated by IL-l, <strong>and</strong> inhibited by FSH/cAMP. Conversely,inhibin B was stimulated by FSH/cAMP <strong>and</strong> inhibited by IL-l. LPS induced a large increase in activin A, which waspartially inhibited by IL-lra, <strong>and</strong> a decrease in inhibin B secretion by <strong>the</strong> Sertoli cells. In cultured seminiferous tubules,activin A secretion occurred across all stages, with a distinct peak of secretion at stage VIII. This secretion was almostcompletely blocked by IL-lra. In contrast to isolated Sertoli cells, activin A secretion by cultured seminiferous tubulesat stages VII-VIII was stimulated by cAMP. Inhibin B secretion was stimulated in all stages by cAMP, but not by IL-l.These data confrrm that Sertoli cell activin A production is positively regulated by endogenous <strong>and</strong> exogenous IL-l viaa cAMP-independent pathway, although o<strong>the</strong>r locally-produced cytokines also may be involved. Activin A isnegatively regulated by FSH/cAMP, but this arm of <strong>the</strong> regulation is modulated by specific germ cell associations.Inhibin B, on <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, is positively regulated by FSH/cAMP, <strong>and</strong> negatively regulated by IL-l. There isreciprocal control of activin A <strong>and</strong> inhibin B throughout <strong>the</strong> cycle of <strong>the</strong> seminiferous epi<strong>the</strong>lium by IL-l <strong>and</strong> FSH,which has important implications <strong>for</strong> control of spermatogonial development <strong>and</strong> <strong>the</strong> effects of inflammation on <strong>the</strong>pituitary-testicular axis.47. CONTRACEPTIVE EFFICACY OF A DEPOT ANDROGEN AND PROGESTINCOMBINATION IN MENL Turner, S Wishart, AJ Conway, PY Liu, E Forbes, RI McLachlan, D.I H<strong>and</strong>elsmanDepartment ofAndrology, Concord Hospital, ANZAC Research Institute, Sydney NSW 2139 & Prince Henry's InstituteofMedical Research, Monash Medical Centre, Clayton, Victoria 3168Hormonal male contraception was <strong>the</strong>oretically feasible <strong>for</strong> decades with <strong>for</strong>mal proof of concept demonstrated inl<strong>and</strong>mark WHO male contraceptive efficacy studies in early 1990s. Combined testosterone plus progestin treatment is<strong>the</strong> most promising strategy, but no contraceptive efficacy studies are reported. The superior efficacy plus lesserdem<strong>and</strong>s on compliance suggest depot regimens are ideal <strong>for</strong> hormonal male contraception. We completed <strong>the</strong> firststudy estimating <strong>the</strong> contraceptive efficacy of a depot hormonal regimen, an <strong>and</strong>rogen/progestin combination, in men.Fifty-five men in stable relationships seeking a change in contraceptive method were administered testosterone (four200 mg implants, 4 or 6 monthly) <strong>and</strong> a progestin (300 mg DMPA, 3 months). Once sperm output was suppressed«IM/mL, two consecutive months), men entered a 12 month efficacy period when all o<strong>the</strong>r contraception was ceased.Sperm output was monitored monthly. There were no pregnancies in 426 person-months (35.5 person-years) of efficacyexposure (upper 95% CL <strong>for</strong> contraceptive failure rate, 8% per annum). Mean sperm density fell rapidly (by 88% @1month <strong>and</strong> 98% @2 months) allowing nearly all men to enter efficacy within 3 months (50% @ 1 month, 83% @ 2months, 94% @ 3 months). Only 2/55 (3.6%) men were unable to enter efficacy due to insufficient suppression ofsperm output. A few men treated with T implants at 6 month intervals demonstrated <strong>and</strong>rogen deficiency symptoms&/or escape of spermatogenic suppression (predicted by suboptimal gonadotropin suppression) between months 5-6;men receiving T implants at 4 months intervals had no <strong>and</strong>rogen deficiency nor loss of gonadotropin <strong>and</strong> sperm outputsuppression. Recovery was slower than anticipated (median time 6-9 months to sperm density 20M/mL) presumablydue to DMPA kinetics. Discontinuations were <strong>for</strong> protocol (12), personal (10) <strong>and</strong> medical (5) reasons but <strong>the</strong>re wereno serious adverse effects related to drug exposure. We conclude that <strong>the</strong> first prototype depot <strong>and</strong>rogen/progestincombination regimen provides high contraceptive efficacy with satisfactory short-term safety but slow recovery ofspermatogenesis. Larger studies with purpose-developed depot combinations are required to clarify <strong>the</strong> overall safety<strong>and</strong> efficacy of <strong>the</strong> most promising approach to hormonal male contraception. This study was supported by CONRAD.46. CHARACTERISATION OF PLASMA INHmIN FORMS IN FERTILE ANDINFERTILE MEND.M. Robertson, T. Stephenson, R.I. McLachlanPrince Henry's Institute ofMedical Research, P.O. Box 5152, Clayton, 3168, VictoriaInhibin B is <strong>the</strong> major feedback regulator of FSH in <strong>the</strong> male. In vitro data suggest that precursor high mol wt <strong>for</strong>ms ofinhibin have reduced bioactivity. The aim of this study was to determine <strong>the</strong> levels of high mol wt <strong>for</strong>ms of inhibin B<strong>and</strong> inhibin a-subunit in plasma in fertile men <strong>and</strong> men with various <strong>for</strong>ms of infertility. Plasma from control fertilemen (n=l1), <strong>and</strong> men classified by testicular histology as hypospermatogenesis (HypoSG, n=4), Sertoli Cell Only(n=4), germ cell arrest (n=2) <strong>and</strong> Klinefelter's Syndrome (n=3), <strong>and</strong> following chemo<strong>the</strong>rapy (n=4) were fractionatedusing a combined immunoaffinity chromatography, preparative SDS-PAGE <strong>and</strong> electroelution procedure. Inhibin<strong>for</strong>ms were determined by ELISAs <strong>for</strong> inhibin B, total inhibin (all <strong>for</strong>ms of inhibin containing <strong>the</strong> aC fragment) <strong>and</strong>pro-aC. In fertile men <strong>and</strong> men with HypoSG, inhibin B was identified as mature (26-30k) <strong>and</strong> precursor (60k) <strong>for</strong>mswith similar proportions (29.1 % vs 25.2%, respectively) of <strong>the</strong> 60k <strong>for</strong>m in both groups. Inhibin B levels were too lowto be assessed in <strong>the</strong> o<strong>the</strong>r infertile groups. Precursor <strong>for</strong>ms of pro-aC (46k, pro-aN-aC) also showed no differencesbetween control (7.4%) <strong>and</strong> all infertile groups (7.4-11 %).To· establish if <strong>the</strong> Pro-aC ELISA was detecting <strong>the</strong> precursor <strong>for</strong>ms of inhibin B containing <strong>the</strong> pro- fragment, normalmale plasma was repeatedly immunoabsorbed with antiserum (INPRO) to <strong>the</strong> pro- region <strong>and</strong> <strong>the</strong> remainingimmunoabsorbed plasma <strong>the</strong>n immunoabsorbed with antiserum to <strong>the</strong> aC subunit. No inhibin B was detected in <strong>the</strong>INPRO-absorbed sample <strong>and</strong> <strong>the</strong> profile of immunoactivity as determined by Pro-aC <strong>and</strong> total inhibin ELISAs weresimilar. Similarly, <strong>the</strong> profiles of inhibin B <strong>and</strong> total inhibin in <strong>the</strong> a subunit-absorbed sample were identical. Thesedata indicate that <strong>the</strong> pro-aC ELISA is detecting all <strong>for</strong>ms of <strong>the</strong> monomeric aC subunit but not <strong>the</strong> precursor inhibinB <strong>for</strong>ms <strong>and</strong> that <strong>the</strong> inhibin B ELISA is detecting all dimeric inhibin B. The two ELISAs are thus separately detectingall <strong>the</strong> free a subunit <strong>and</strong> inhibin B in male plasma.It is concluded that a) <strong>the</strong> Pro-aC <strong>and</strong> inhibin B ELISAs provide an unambiguous assessment of <strong>the</strong> levels of <strong>the</strong>seinhibin <strong>for</strong>ms in <strong>the</strong> circulation <strong>and</strong> b) <strong>the</strong> proportion of precursor inhibin B <strong>for</strong>ms in plasma (25-30%) is unchanged inmen with sub-fertility.48. PRESENCE OF IMMUNE MODULATING MOLECULESIN BOAR SEMINALPLASMAS. O'Leary, D.T. Armstrong <strong>and</strong> S.A. RobertsonDept. OfObstetrics <strong>and</strong> Gynaecology, The University ofAdelaide <strong>and</strong> Reproductive Medicine Unit, The QueenElizabeth Hospital Woodville. South Australia.Factors in seminal plasma are recognised as potential modulators of reproductive success in <strong>the</strong> pig. Experiments inrodents <strong>and</strong> more recently in pigs indicate roles <strong>for</strong> specific proteins in seminal plasma in inducing cellular <strong>and</strong>molecular changes in <strong>the</strong> female reproductive tract. An active constituent of murine seminal plasma has been identifiedas <strong>the</strong> potent immune modulating cytokine trans<strong>for</strong>ming growth factor beta (TGF~)l. In vitro experiments suggest <strong>the</strong>response to TGF~ is fur<strong>the</strong>r influenced by interferon (IFN)y <strong>and</strong> lipoplysaccharide (LPS). To investigate whe<strong>the</strong>rimmune modulating molecules are present in pig semen, <strong>the</strong> content of TGF~}, TGF~2, IFNy <strong>and</strong> LPS in boar seminalplasma were measured. Semen was collected from 42 Large White boars of similar age <strong>and</strong> of known fertility, <strong>and</strong>seminal plasma was prepared by centrifugation. Commercial ELISA assays were used to measure TGF~}, TGF~2 (bothPromega) <strong>and</strong> IFNy (Endogen) <strong>and</strong> LPS was measured by Limulus Amebocyte Lysate assay (Bio Whittaker). TGF~l<strong>and</strong> TGF~2 were detected in all samples [median (range) = 185 (91-423) <strong>and</strong> 50.1 (26-101) ng/ml respectively], IFNywas detectable in only 2 samples (39 & 40 pg/ml) <strong>and</strong> endotoxin was present in 30 samples [125 (11-193) EU/ml].Variation over time <strong>and</strong> effect of frequency of collection on TGF~ content was also evaluated. Content of both iso<strong>for</strong>msvaried


49. INFLUENCE OF OXYTOCIN ON OXYTOCIN RECEPTOR AND 5 a-REDUCTASETRANSCRIPTION IN THE RAT PROSTATESteve Assinder, Mei Sun, Helen NicholsonArgo, Department ofAnatomy <strong>and</strong> Structural Biology, University ofOtago, New Zeal<strong>and</strong>.Dihydrotestosterone (DRT) is essential <strong>for</strong> normal function <strong>and</strong> growth of <strong>the</strong> prostate. DRT is produced by <strong>the</strong>reduction of testosterone, catalysed by 5 a-reductase. Two iso<strong>for</strong>ms of <strong>the</strong> 5 a-reductase enzyme exist. In <strong>the</strong> ratprostate type I is expressed in <strong>the</strong> epi<strong>the</strong>lium <strong>and</strong> type II in stromal cells. Oxytocin increases <strong>the</strong> activity of thisenzyme but it is unclear how. Nei<strong>the</strong>r expression nor distribution of oxytocin receptor have previously beendescribed in <strong>the</strong> rat prostate. This study aimed to determine <strong>the</strong> distribution of oxytocin receptor, <strong>and</strong> to investigateif oxytocin influences expression of its receptor <strong>and</strong> <strong>the</strong> iso<strong>for</strong>ms of 5 a-reductase. Adult male Wistar rats weretreated daily with ei<strong>the</strong>r 5 ~glKg oxytocin, 2 ~glKg of a specific oxytocin antagonist (OTA), both oxytocin <strong>and</strong>OTA, or saline (control) respectively <strong>for</strong> 3 days. On day 4 rats were euthanased by CO2 inhalation <strong>and</strong> <strong>the</strong> ventralprostate removed. Western blot <strong>and</strong> immunohistochemistry, employing a polyclonal antibody to <strong>the</strong> 3 rd intracellularloop of <strong>the</strong> oxytocin receptor, identified a specific peptide of -60 kDa <strong>and</strong> localised oxytocin receptor to both <strong>the</strong>stromal tissue <strong>and</strong> epi<strong>the</strong>lium. Total RNA was used in a semi-quantitative RT-PCR approach to estimate relativelevels of expression of 5 a- reductase type I <strong>and</strong> II, <strong>and</strong> oxytocin receptor with levels normalised against a B-actinamplicon as internal control. Oxytocin treatment significantly decreased (P


---------------------------------------------------~-~---~-~--53. IDENTIFICATION AND LOCALISATION OF A MESOTOCIN RECEPTOR IN THEPROSTATE OF THE BRUSHTAIL POSSUMHelen Nicholson!, Jo Fink!, Steve Assinderl, Bernie McLeod 2 , Laura ParrlIDepartment ofAnatomy & Structural Biology, University ofOtago, New Zeal<strong>and</strong>, 2AgResearch, Invermay AgriculturalCentre, 3New Zeal<strong>and</strong>, Howard Florey Institute, Melbourne.Oxytocin is present in <strong>the</strong> prostate of <strong>the</strong> eu<strong>the</strong>rian mammals <strong>and</strong> has been implicated in <strong>the</strong> regulation of prostatecontractility <strong>and</strong> growth [1]. The marsupial produces mesotocin, ra<strong>the</strong>r than oxytocin, <strong>and</strong> a mesotocin receptor hasbeen identified in <strong>the</strong> Tammar wallaby prostate [2]. The aim of this study was to identify a mesotocin receptor within<strong>the</strong> possum prostate <strong>and</strong> investigate <strong>the</strong> localisation of <strong>the</strong> receptor protein in <strong>the</strong> adult prostate in <strong>and</strong> out of <strong>the</strong>breeding season. Groups of possums (n=6-10), trapped alive in <strong>the</strong> wild, were killed at two monthly intervals during<strong>the</strong> year <strong>and</strong> prostate tissue collected. The prostates were divided into cranial <strong>and</strong> caudal portions <strong>and</strong> ei<strong>the</strong>r frozen orfixed in 10% <strong>for</strong>malin. Immunohistochemistry <strong>and</strong> western blot analysis were per<strong>for</strong>med using an antibody (020) raisedto <strong>the</strong> third intracellular loop of <strong>the</strong> sheep oxytocin receptor [3]. RT-PCR of total prostatic RNA using primersdesigned to <strong>the</strong> Tammar mesotocin receptor generated a 337bp amplicon. On sequencing this demonstrated. 92 %sequence homology with <strong>the</strong> Tammar receptor. Western blot analysis confirmed <strong>the</strong> presence of a single -60 kDaprotein. Immuno-reactive receptor protein was identified in all of <strong>the</strong> prostates examined <strong>and</strong> was localisedpredominantly to <strong>the</strong> epi<strong>the</strong>lial cells of <strong>the</strong> gl<strong>and</strong>ular acini of both <strong>the</strong> cranial <strong>and</strong> caudal regions of <strong>the</strong> prostate. Littlereceptor was detected in <strong>the</strong> stromal tissue surrounding <strong>the</strong> gl<strong>and</strong>s. Significant changes (P


57. SPERM SURFACE PROTEIN PH-20 EXPRESSION DURING SHEEPSPERMATOGENESISFu Yu, Helen D Nicholson, Robin McDonald!, Stuart A Meyers 2 , Grant W Montgomery3, John F Smith!, Jean SFlemingDept Anatomy & Structural Biology, University ojOtago School ofMedical Sciences, Dunedin, New Zeal<strong>and</strong>;IAgResearch, Ruakura Research Centre, Hamilton, New Zeal<strong>and</strong>; 2Dept Anatomy, Physiology & Cell Biology,University ofCali<strong>for</strong>nia, Davis, USA; 3Queensl<strong>and</strong> Inst Medical Research, Brisbane, Australia.The sperm surface protein PH-20 exhibits dual functions, having hyaluronidase activity that enables sperm to penetrate<strong>the</strong> cumulus <strong>and</strong> also being involved in zona pellucida binding. The aim of this study was to determine <strong>the</strong> site of ovinePH-20 expression during spermatogenesis in sheep testes. Fresh testis samples from foetal (n=3), new-born (n=4), 4month-old lambs (n=8), 5 month-old lambs (n=12), <strong>and</strong> 4-6 year-old rams (n=6) were collected <strong>and</strong> frozen <strong>for</strong> totalRNA extraction or fixed in 10% phosphate buffered <strong>for</strong>maldehyde <strong>for</strong> histological analysis. A fragment of ovine testisPH-20 cDNA (282 nt) was identified by reverse transcription <strong>and</strong> polymerase chain reaction (RT PCR) in testis RNA,but not gDNA extracts. The cDNA exhibited 77% identity to human PH-20 <strong>and</strong> 93% identity to red fox PH-20 eDNA<strong>and</strong> this sequence was submitted to GenBank (AFI74691). Ovine PH-20 expression was detected using RT-PCR <strong>and</strong>western blotting. The developmental stage of <strong>the</strong> 4-5 month ram testes was assessed histologically. Ovine PH-20protein was localised in <strong>the</strong> seminiferous tubules using immunohistochemistry with an anti equine PH-20 antibody.Study of PH-20 rnRNA expression using RT-PCR showed that PH-20 mRNA is only expressed in ram testis <strong>and</strong> not in<strong>the</strong> o<strong>the</strong>r sheep tissues including ovary, brain, liver, lung, heart, epididymis, spleen, or kidney. Western blotting (underreducing conditions) revealed a single 74 KDa immunoreactive b<strong>and</strong> in ram testis but not in liver tissue. PH-20 mRNA<strong>and</strong> protein were identified only in testes where spermatids were present in <strong>the</strong> seminiferous tubules <strong>and</strong> in 4-5 montholdram testes undergoing <strong>the</strong> first wave of spermatogenesis, but not in foetal, new-born or prepubertal testiculartissues. Immunohistochemistry <strong>for</strong> ovine PH-20 protein in adult ram testis confirmed <strong>the</strong> cellular localisation in <strong>the</strong>initial spermatogenic wave in developing rams. We conclude that ovine PH-20 is expressed post-meiotically in haploidround <strong>and</strong> elongate spermatids.58. OVARIAN FOLLICULAR DEVELOPMENT IS COMPROMISED IN TRANSGENIC(mREN-2)27 RATS AND ANGIOTENSIN II-INFUSED SPRAGUE DAWLEY RATST.E. de Gooyer*, D.J. KeUyt, R.E. Gilbertt <strong>and</strong> J.L. Wilkinson-Berka*Departments of*Physiology <strong>and</strong> tMedicine, St Vincent's Hospital, University ofMelbourne.Angiotensin II (ANG II) is implicated in ovulation, vascularisation <strong>and</strong> atresia, <strong>and</strong> may influence <strong>the</strong>se processes by alocal ovarian renin-angiotensin system (RAS), which we have previously localised in rat ovarian stroma <strong>and</strong> its bloodvessels. The aim of this study was to determine <strong>the</strong> role ofANG II in follicular development. Three groups of 12 weekold normally cycling, female rats were used: 1) untreated Sprague Dawley (SD), 2) SD infused with ANG II (145ng/kglmin <strong>for</strong> 12-14 days), <strong>and</strong> 3) hypertensive homozygous (HMZ) (rnRen-2)27 transgenic rats, which displayamplified extra-renal tissue renin <strong>and</strong> ANG II. Ovaries were collected at proestrus <strong>for</strong> histological analysis of folliculartype <strong>and</strong> number, immunolocalisation of renin <strong>and</strong> ANG II, in situ hybridisation <strong>for</strong> ATla receptor (ATla-R) mRNA<strong>and</strong> radioimmunoassay of ovarian renin content. Systolic blood pressure (SBP; tail cuff) was highest in HMZ Ren-2followed by SD+ANG II <strong>and</strong> SD. HMZ Ren-2 <strong>and</strong> SD+ANG II had more antral follicles than SD, but fewer largeantral <strong>and</strong> pre-ovulatory follicles. Litter size was reduced in HMZ Ren-2 compared to SD. Consistent with <strong>the</strong> highRAS of <strong>the</strong> HMZ Ren-2, ovarian renin content <strong>and</strong> renin <strong>and</strong> ANG II immunolabelling were increased <strong>and</strong> ATla-RSD+ANG IT exhibited a reduction in ovarian renin content <strong>and</strong> ATla-R rnRNA gene expression,rnRNA reduced.indicatin a ne ative feedback on <strong>the</strong> RAS.N = 4-6 rats er rou SD SD+ANG IISBP (mmHg) 125±1 199±12*Antral Follicles: 150-39011m (% Total) 37.5±4.5 53.5±6.3*Large Antral Follicles: 390-500/lm (%Total) 7.7±1.8 2.4±1.1 *Pre-ovulatory Follicles: >500llID (% Total) 17.1±3.6 3.8±1.5*Litter Size 13.7±O.7 NAOvarian Renin Content (mGU/g) 4.8±0.5 2.7±0.3*ATla-R mRNA (Relative 0 tical Densit ) 75.3±1.8 64.0+2.4*Values are mean ± sem. *p


61. ESTROGEN PROMOTES ANGIOGENESIS IN ERa-EXPRESSING HUMANMYOMETRIAL MICROVASCULAR ENDOTHELIAL CELLS BY UPREGULATINGVEGF RECEPTOR-2Caroline E Gargett, Marina Zaitseva, Kristina Bucak <strong>and</strong> Peter AW RogersCentre <strong>for</strong> Women's Health Research, Monash University Department ofObstetrics <strong>and</strong> Gynaecology, MonashMedical Centre, 246 Clayton Rd, Clayton, Vic. 3168Angiogenesis is <strong>the</strong> growth of new blood vessels from pre-existing vessels <strong>and</strong> involves proliferation of microvascularendo<strong>the</strong>lial cells (MEC). VEGF is a major promoter of angiogenesis <strong>and</strong> mediates angiogenic effects primarily throughinteraction with VEGF receptor-2 (VEGF-R2/KDR) present on <strong>the</strong> surface of MEC. Estrogen promotes bFGF-inducedangiogenesis, but its effect on VEGF-mediated angiogenesis is unknown. We have demonstrated that MEC derivedfrom human myometrium constitutively express estrogen receptor-~ (ER~), while ERa varies between subjects <strong>and</strong> isonly expressed in approximately 60% of MEC isolates (1). The aims of <strong>the</strong> present study were to determine whe<strong>the</strong>r (a)estrogen increases VEGF-R2 expression in ERa positive <strong>and</strong> ERa negative myometrial MEC, (b) ER mediates thiseffect <strong>and</strong> (c) estrogen promotes VEGF-induced MEC proliferation. Myometrial MEC were isolated fromhysterectomy tissue obtained from ovulating women, purified with DEA-l-coated Dynabeads, cultured <strong>and</strong> usedbetween passages 1-3 (purity >98% CD31+ cells) (1, 2). ERa <strong>and</strong> ER~ mRNA were determined by RT-PCR (3) <strong>and</strong>protein by flow cytometry or Western blotting. VEGF-R2 expression was measured by flow cytometry using ei<strong>the</strong>rVEGF-R2 antibody or biotin-rhVEGF 165 binding. MEC proliferation was determined by MTS bioassay (2). Estrogen(1 <strong>and</strong> 10 nM) significantly increased rhVEGF 165 binding <strong>and</strong> VEGF-R2 receptor expression in ERa positive (P


65. MECHANISMS UNDERLYING THE REDUCED SENSITIVITY TO PROLACTINNEGATIVE FEEDBACK DURING LACTATIONST Anderson!, JL Barclayt, DHL Kusters\ SP Tamt, PLaut, DR Grattan\ MJ Waters 1 ,2, JD Curlewis 11 School ofBiomedical Sciences <strong>and</strong> 2 Institute ofMolecular Biosciences, University ofQueensl<strong>and</strong>.3 Department ofAnatomy <strong>and</strong> Structural Biology, University ofOtago, Dunedin, New Zeal<strong>and</strong>.Prolactin negative feedback maintains normal, low prolactin concentrations in non-lactating individuals. Prolactinstimulates <strong>the</strong> tuberoinfundibular dopaminergic (TIDA) neurons of <strong>the</strong> hypothalamus to release dopamine whichsuppresses prolactin secretion from lactotrophs. During lactation, however, <strong>the</strong> sucking stimulus reduces <strong>the</strong> sensitivityof TIDA neurons to prolactin feedback <strong>and</strong> consequently high prolactin levels are observed. The mechanisms involvedin this loss of negative feedback are unknown, but may involve changes in prolactin signal transduction pathways.Activation of prolactin-receptors normally results in phosphorylation <strong>and</strong> nuclear translocation of Stat proteins,particularly StatS, so we investigatedStatS signalling in TIDA neurons of lactating rats. Lactating rats with or withoutpups (removed <strong>for</strong> 16h) were given a sc injection of ei<strong>the</strong>r oPRL (250J.lg) or saline vehicle (n=4/treatment), <strong>and</strong> after Ihwere killed <strong>and</strong> perfused. Hypothalamic sections were immunostained <strong>for</strong> tyrosine hydroxylase (Chemicon, MAB318)<strong>and</strong> StatS (Santa Cruz, Sc835) <strong>and</strong> confocal images of <strong>the</strong> dorsomedial arcuate nucleus were captured <strong>for</strong> quantitation.Stat5 intracellular distribution in TIDA neurons was determined by <strong>the</strong> ratio of Stat5 fluorescence intensity in <strong>the</strong>nucleus expressed as a proportion of that in <strong>the</strong> cytoplasm (N/C ratio). In lactating rats with pups removed <strong>for</strong> I6h,oPRL injection significantly (P


69. NEGATIVE REGULATION OF PROLACTIN-RECEPTOR SIGNALLING BY SOCSPROTEINS: A NEW MECHANISM IN LUTEOLYSIS?.J.D. Curlewis, M.J. Waters <strong>and</strong> S.T. AndersonSchool ofBiomedical Sciences, The University OfQueensl<strong>and</strong>, Qld 4072, AustraliaProlactin (PRL) <strong>and</strong> placental lactogen (PL) play essential roles in maintaining <strong>the</strong> rodent corpus luteum (CL) throughpregnancy by inducing transcription of some genes (e.g. ERa <strong>and</strong> ER~, LH-receptor, prolactin-receptor) <strong>and</strong> repressionof o<strong>the</strong>rs (e.g. 20a-hydroxysteroid dehydrogenase). These effects are mediated by <strong>the</strong> PRL-receptor, which signalsthrough several pathways although it is <strong>the</strong> JAK2/STAT 5 pathway that plays <strong>the</strong> central role in <strong>the</strong> CL. At <strong>the</strong> end ofpregnancy, luteolysis is induced by prostagl<strong>and</strong>in F2a (PGF2a), which reverses many of <strong>the</strong> effects of <strong>the</strong> PRLreceptorsignalling pathway on gene expression. Our recent research on day 19 pregnant rats has shown that one wayin which PGF2a could block PRL-induced gene expression is through inhibition of <strong>the</strong> JAK2/STAT 5 pathway.Following treatment of rats with cloprostenol, a PGF2a analogue, <strong>the</strong>re is rapid loss of active STAT 5 from ovariannuclear extracts <strong>and</strong> this effect persists <strong>for</strong> at least 8 h. Moreover, during this period, treatment with prolactin does notinduce activation of STAT 5, despite <strong>the</strong> continued presence of PRL-R. The recently discovered Suppressors OfCytokine Signalling (SOCS) have been shown to decrease cell sensitivity to cytokines, including PRL, <strong>and</strong> so wedetermined whe<strong>the</strong>r <strong>the</strong> negative regulation of PRL-receptor signalling by PGF2a could be due to upregulation ofSOCS proteins. In Nor<strong>the</strong>rn blots per<strong>for</strong>med on ovaries collected 0.5-16 h after cloprostenol, SOCS-l showed atransient increase at 0.5 h while SOCS-3 mRNA increased 3-fold at 0.5 h <strong>and</strong> remained elevated until 4 h afterinjection. Western blots confirmed that SOCS-3 protein was increased over <strong>the</strong> same period <strong>and</strong>, in ovaries collected 4h after cloprostenol, we confirmed by immunohistochemistry that SOCS-3 was localised predominantly within CLs. Inconclusion, induction of SOCS-3 <strong>and</strong> to a lesser extent SOCS-l by PGF2a may be an important element in <strong>the</strong>initiation ofluteolysis, via rapid suppression of luteotrophic support from PL.70. PARACRINE SIGNALLING WITHIN THE OVARIAN FOLLICLE: POTENTIALFOR TGF-p SUPERFAMILY MEMBERS TO INFLUENCE OOCYTE AND GRANULOSACELL INTERACTIONSRobert B. GilchristReproductive Medicine Unit, Department ofObstetrics <strong>and</strong> Gynaecology, The University ofAdelaide, TQEH,Woodville, South AustraliaSignalling between <strong>the</strong> germ cell <strong>and</strong> somatic cell compartments of <strong>the</strong> ovarian follicle is a critically important process<strong>for</strong> growth <strong>and</strong> differentiation of <strong>the</strong> oocyte <strong>and</strong> <strong>the</strong> granulosa cells. Communication occurs via paracrine <strong>and</strong> gapjunctionalsignalling between <strong>the</strong> two cell types, <strong>and</strong> indeed both <strong>for</strong>ms of communication are essential <strong>for</strong> normaloogenesis <strong>and</strong> folliculogenesis. Traditionally, research has been focused on one side of this communication axis, thatis, on <strong>the</strong> role of <strong>the</strong> granulosa cells in nurturing <strong>the</strong> growth <strong>and</strong> development of <strong>the</strong> oocyte. Recent studiesdemonstrate <strong>the</strong> importance of a bi-directional communication axis, <strong>and</strong> it is now becoming clear that <strong>the</strong> oocyte is apivotal regulator of ovarian function. Folliculogenesis fails in <strong>the</strong> absence of oocyte paracrine signalling, whe<strong>the</strong>rlacking due to genetic deficiencies or from experimental ablation of <strong>the</strong> oocyte. Oocytes regulate a broad range ofgranulosa cell functions by modulating fundamental control elements; <strong>for</strong> example, oocytes regulate expression of FSH<strong>and</strong> LH receptors, kit lig<strong>and</strong>, inhibin subunits <strong>and</strong> expression of extra-cellular matrix molecules. As such, oocytes notonly promote growth of granulosa cells <strong>and</strong> <strong>the</strong> follicle, but also regulate differentiation processes such assteroidogenesis <strong>and</strong> physical remodelling of <strong>the</strong> follicle. The primary recipients of oocyte paracrine signalling are <strong>the</strong>cumulus cells, which are <strong>the</strong> specialised granulosa cells immediately surrounding <strong>the</strong> oocyte. Cumulus cells have adistinct phenotype from <strong>the</strong> mural granulosa cells (MGC) lining <strong>the</strong> wall of <strong>the</strong> follicle, <strong>and</strong> in fact mairitenance of <strong>the</strong>cumulus cell phenotype is dependent on oocyte-secretions. In <strong>the</strong> absence of <strong>the</strong>se oocyte signals <strong>the</strong> cumulus cellswill spontaneously differentiate toward <strong>the</strong> MGC phenotype. Oocyte paracrine signalling <strong>the</strong>re<strong>for</strong>e establishes afunctional morphogenic gradient across <strong>the</strong> ovarian follicle. The exact identity of <strong>the</strong>se oocyte factors remains elusive,but members of <strong>the</strong> trans<strong>for</strong>ming growth factor-beta superfamily are <strong>the</strong> prime c<strong>and</strong>idates as some of <strong>the</strong>se growthfactors are able to mimic <strong>the</strong> effects of oocytes on granulosa cells in vitro. Two family members, which are exclusivelyexpressed in gametes, growth differentiation factor-9 (GDF-9) <strong>and</strong> bone-morphogenic factor-I5 (BMP-15), are criticalfactors as <strong>the</strong>ir absence leads to failed folliculogenesis <strong>and</strong> complete sterility. Interestingly <strong>the</strong>se molecules have moresubtle effects on fertility with high or low expression of <strong>the</strong>se oocyte factors leading to perturbations such as ovariancysts, anovulation, disrupted oocyte maturation <strong>and</strong> even increases in ovulation rate <strong>and</strong> litter size. There is now anemerging appreciation that not only does paracrine signalling from <strong>the</strong> oocyte allow <strong>the</strong> oocyte to control its ownmicroenvironment, but oocyte signalling also has profound effects on ovarian function, endocrine well being <strong>and</strong>fertility.71. EXPRESSION OF TGF-P SUPERFAMILY MEMBERS AND THEIR SIGNALLINGCOMPONENTRY BY THE RAT OVARYAnn E. Drummond, Mitzi Dyson, Minh-Tan Le, Jean-Francois Ethier <strong>and</strong> Jock K. FindlayPrince Henry's Institute ofMedical Research, PO Box 5152 Clayton, Victoria 3168, AustraliaAn increasing number of trans<strong>for</strong>ming growth factor-~ (TGF-~) superfamily members are being shown to exert effectson ovarian function. Most of <strong>the</strong>se factors, which include activin, <strong>the</strong> bone morphogenetic proteins (BMPs) <strong>and</strong> TGF-~,act via transmembrane serine <strong>and</strong> threonine kinase receptors which activate pertinent Smad proteins, complexes ofwhich translocate to <strong>the</strong> nucleus to mediate gene transcription. Despite underst<strong>and</strong>ing <strong>the</strong> molecular basis of <strong>the</strong>sesignalling pathways, little has been done to characterise <strong>the</strong>se pathways in <strong>the</strong> ovary. We've sought to elucidate whichelements of <strong>the</strong>se signalling pathways are present in ovarian follicles of <strong>the</strong> postnatal rat, as an initial step towardidentifying active pathways in follicle populations. Activin/BMP receptors (ActRIA, ActRIB, ActRIIA <strong>and</strong> ActRIIB),~glycan <strong>and</strong> Smad 1-8 mRNAs were expressed by <strong>the</strong> postnatal rat ovary. Activin receptor <strong>and</strong> Smad proteins werepresent in oocytes at all stages of follicular development; granulosa cells of primary-antral follicles <strong>and</strong> <strong>the</strong>ca cells.~glycan protein was present in oocytes, granulosa cells <strong>and</strong> <strong>the</strong>ca cells at all stages of folliculogenesis. The colocalisationof receptors <strong>and</strong> Smads supports <strong>the</strong> notion that activinrrGF~ <strong>and</strong> BMP signalling pathways may.befunctional in <strong>the</strong> cellular compartments of <strong>the</strong> follicle. We hypo<strong>the</strong>sise that follicle populations express elements of <strong>the</strong>TGF-~ superfamily (<strong>and</strong> <strong>the</strong>ir signalling pathways) in different combinations during development <strong>and</strong> thus it is <strong>the</strong>individual follicle 'blueprint' that determines <strong>the</strong> response to lig<strong>and</strong>s <strong>and</strong> ultimately <strong>the</strong> biological outcomes. Theseconcepts are currently under investigation in our laboratory.Supported by <strong>the</strong> NH&MRC of Australia (Regkey 983212)72. DIFFERENTIAL GENE EXPRESSION IN INTERLEUKIN-ll RECEPTOR aDEFICIENT MICE COMPARED TO WILD TYPE DURING DECIDUALISATIONChristine White 1 ,2, Lorraine Robb 3 <strong>and</strong> Lois Salamonsen 1 ,21Prince Henry's Institute ofMedical Research, 2Dept ofObstetrics & Gynaecology, Monash University,3Walter & Eliza Hall Institute ofMedical ResearchDifferentiation of endometrial stromal cells into decidual cells is essential <strong>for</strong> successful embryo implantation. Femaleinterleukin-l1 receptor a chain (IL-11Ra) null mice are infertile due to disrupted decidualisation, suggesting a criticalrole <strong>for</strong> IL-l1 <strong>and</strong> its target genes in implantation. The molecular mechanisms by which IL-11 acts through its receptorto mediate decidualisation are unknown, but it is hypo<strong>the</strong>sised that <strong>the</strong> expression of genes encoding o<strong>the</strong>r regulatorymolecules will be modified by IL-11 signalling. This study aims to identify genes regulated by IL-l1 duringdecidualisation in mouse uterus by DNA microarray analysis, <strong>and</strong> to examine <strong>the</strong> expression <strong>and</strong> functionalsignificance of <strong>the</strong>se genes during early pregnancy. Mice heterozygous <strong>for</strong> a targeted mutation of <strong>the</strong> IL-llRa gene(IL-IIRa+/-) were interbred to produce wild type (IL-I1Ra+/+) <strong>and</strong> null mutant (IL-IIRa-/-) mice. Decidualisation wasinduced in pseudopregnant (plug:::: day 0) IL-llRa+/+ <strong>and</strong> IL-llRa-/-littermates (n:::: 8/genotype) by oil injection into<strong>the</strong> uterine lumen on day 3. Whole uterus was removed at 0, 18, 24 or 48hrs after decidualisation (n :::: 2/group) <strong>and</strong>total RNA prepared by acid-phenol/chloro<strong>for</strong>m extraction. Reference RNA was isolated from wild type unstimulateduterus (n :::: 16). Total RNA (lOllg/sample) was reverse transcribed to fluorescent-labelled cDNA by indirect Cy3 orCy5 coupling. NIA 15K microarrays were probed with experimental (IL-llRa+/+ or IL-llRa-/-) <strong>and</strong> referenceCy3/Cy5-cDNA <strong>and</strong> scanned using an Axon GenePix 4000B microarray reader <strong>and</strong> GenePix Pro 3.0 software. Datawas analysed using R 1.4.1, including normalisation <strong>for</strong> labelling efficiencies <strong>and</strong> scanning properties of Cy3 <strong>and</strong> Cy5,print-tip <strong>and</strong> spatial effects. At 18 hours after decidualisation, 18 genes were found to be upregulated <strong>and</strong> 6 genesdownregulated >2-fold in IL-llRa-/- mice compared to wild type. These included expressed sequence tags (ESTs),genes with unknown function in <strong>the</strong> endometrium, <strong>and</strong> genes associated with transcription/chromatin, matrix/structuralproteins <strong>and</strong> angiogenesis. Data analyses <strong>for</strong> fur<strong>the</strong>r time points are underway. Genes identified as strongly regulatedby IL-l1 during decidualisation are likely to be important modulators of implantation, <strong>and</strong> will be fur<strong>the</strong>r verified <strong>and</strong>characterised. By elucidating <strong>the</strong> role of IL-11 regulated genes in murine decidualisation, <strong>the</strong>se studies may identifypotential new targets <strong>for</strong> <strong>the</strong> manipulation of human fertility.7879


73. MANIPULATION OF BOVINE OOCYTE-CUMULUS CELL GAP JUNCTIONALCOMMUNICATION DURING IN VITRO MATURATION BY MODULATING CELL­SPECIFIC cAMP LEVELSRE Thomas, JG Thompson, DT Armstrong, RB GilchristThe Reproductive Medicine Unit, Department ofObstetrics <strong>and</strong> Gynaecology, The University ofAdelaide, The QueenElizabeth Hospital, Woodville 5011 SA.Oocyte maturation is associated with <strong>the</strong> loss of oocyte-cumulus cell gap junctional communication, preventing entry ofmeiotic-modulating factors such as cAMP into <strong>the</strong> oocyte. Prolonging communication during in vitro maturation(IVM) may improve post-maturation outcomes, such as embryo development, through improved cytoplasmicmaturation. We have previously shown that oocyte <strong>and</strong> cumulus cell (CC) cAMP levels can be independently regulatedusing selective phosphodiesterase (PDE) inhibitors [1]. The objectives of this study were to examine <strong>the</strong> effects of celltype-specific PDE inhibitors on <strong>the</strong> maintenance of oocyte-CC gap junction communication <strong>and</strong> oocyte meioticprogression. Cumulus-oocyte complexes (COC) were aspirated from antral follicles of abattoir-derived ovaries.Oocyte-CC gap junction communication during oocyte maturation was measured using <strong>the</strong> fluorescent dye, calcein­AM. COCs were cultured in <strong>the</strong> presence of specific PDE inhibitors; milrinone (MR, oocyte PDE3 inhibitor) orrolipram (RP, CC PDE4 inhibitor), <strong>and</strong> were pulsed with calcein-AM to allow dye transfer between <strong>the</strong> two cell types.Following CC removal, fluorescence in denuded oocytes was measured by microphotometry <strong>and</strong> meiotic progressionassessed. In control COCs, dye transfer from CC to <strong>the</strong> oocyte fell progressively from 0 to 8h, after which oocyte-CCgap junction communication was completely lost. From 3-6h of maturation, loss of gap junction communication wassignificantly (P


77. INSEMINATION AND UTERINE LYMPHOCYTE TRAFFICKING IN MICEJohn J. Bromfield <strong>and</strong> Sarah A. RobertsonDepartment ofObstetrics <strong>and</strong> Gynaecology, University ofAdelaide, SA 5005Lymphocyte populations facilitating embryo implantation <strong>and</strong> early placental development have immunosuppressivephenotypes, similar to those mediating immune tolerance in o<strong>the</strong>r mucosal tissues. It is unclear how <strong>and</strong> when <strong>the</strong>sepopulations become activated, but <strong>the</strong>y have some similarities to those primed earlier in pregnancy at insemination.The mechanisms defining lymphocyte phenotype <strong>and</strong> homing patterns occur at activation, <strong>and</strong> lymphocytes primed inspecific tissues have a propensity to recruit back to those tissues [1]. Moreover, paternal <strong>and</strong> o<strong>the</strong>r antigens are sharedby semen <strong>and</strong> <strong>the</strong> conceptus. Thus we hypo<strong>the</strong>sise that mating comprises <strong>the</strong> 'priming' event <strong>for</strong> activation oflymphocytes required <strong>for</strong> embryo implantation [2]. The aim of this study was to investigate whe<strong>the</strong>r insemination leadsto <strong>the</strong> activation of lymphocytes that can be recruited preferentially into <strong>the</strong> implantation sites of pregnant mice. Weemployed cell tracking techniques, utilising [125l]-iodo deoxyuridine C 25 IdUR), to label lymphocytes recovered from<strong>the</strong> para-aortic lymph nodes (PALN) which drain <strong>the</strong> uterus, in virgin mice or 4 days after insemination. Labelled cellswere transferred to day 6 pregnant recipients via <strong>the</strong> lateral tail vein, <strong>and</strong> recruitment into a range of tissues wasassessed by y counting 24 hours after transfer. Tissue specific patterns of lymphocyte recruitment were identified, withpreferential recruitment of PALN lymphocytes into lymphoid <strong>and</strong> uterine tissues. PALN lymphocytes from mateddonors showed a pattern of recruitment into <strong>the</strong> uterus, preferentially into implantation tissues as opposed to interimplantationtissues (p=O.OI), which was not explained simply by <strong>the</strong> increased vascularity of this site. The distinctionwas not evident when PALN lymphocytes were recovered from virgin donors. PALN lymphocytes from virgin donorsalso showed a greater propensity to recruit back to <strong>the</strong> PALN <strong>and</strong> o<strong>the</strong>r lymphoid tissues (p


81. INTRAOVARIAN REGULATION OF FOLLICULOGENESIS: SIGNAL MOLECULESAND THEIR CELLULAR SOURCESDavid T ArmstrongReproductive Medicine Unit, Dept ofObstetrics <strong>and</strong> Gynaecology, University ofAdelaide, SAFolliculogenesis in mammals is a continuous <strong>and</strong> dynamic process that begins during embryonic development in utero<strong>and</strong> continues until <strong>the</strong> end of adult reproductive life. The overall process is regulated by a complex array of interactingchemical signals originating both within <strong>and</strong> external to <strong>the</strong> ovaries. The identities <strong>and</strong> relative importance of specificsignals change as follicle growth progresses through a series of morphologically distinct stages, beginning withinitiation of growth of <strong>the</strong> dormant primordial follicles, <strong>and</strong> ending in ei<strong>the</strong>r atresia or ovulation. The initiation offollicle growth occurs when a primordial follicle is activated by as yet unknown mechanisms <strong>and</strong> enters a pool ofgrowing follicles. Recent evidence has established <strong>the</strong> essential role of <strong>the</strong> oocyte as <strong>the</strong> driving <strong>for</strong>ce in promotingfollicle growth throughout pre-antral <strong>and</strong> early antral stages, mediated via recently identified members of <strong>the</strong> TGF~superfamily of growth factors, GDF9 <strong>and</strong> GDF9B, which have essential roles in both proliferation <strong>and</strong> differentiationof granulosa cells. The follicular somatic cells, including both granulosa <strong>and</strong> <strong>the</strong>cal cells, also contribute to regulationof follicle growth during <strong>the</strong>se stages, through secretions of several o<strong>the</strong>r peptide growth factors, in particular bFGF,IGF-I, TGF~, activin, <strong>and</strong> inhibin. Upon reaching antral stage of development, follicles acquire responsiveness tofollicle-stimulating hormone (FSH) <strong>and</strong> luteinizing hormone (LH), <strong>and</strong> <strong>the</strong>se gonadotrophins <strong>the</strong>n assume primaryimportance in regulation of growth to <strong>the</strong> pre-ovulatory stage, with growth factors continuing to play supportive <strong>and</strong>modulating roles. The actions of FSH <strong>and</strong> LH are mediated in part via <strong>and</strong>rogens <strong>and</strong> oestrogens secreted by <strong>the</strong> <strong>the</strong>cal<strong>and</strong> granulosa cells, respectively. With <strong>the</strong> approach of ovulation, migratory cells derived from <strong>the</strong> circulation,including macrophages <strong>and</strong> to a lesser extent o<strong>the</strong>r leukocyte subsets, infiltrate <strong>the</strong> ovarian stroma <strong>and</strong> <strong>the</strong>cal layer of<strong>the</strong> pre-ovulatory follicle, where <strong>the</strong>y play essential roles in <strong>the</strong> tissue remodelling that leads to rupture of <strong>the</strong> follicle<strong>and</strong> its trans<strong>for</strong>mation to a corpus luteum at ovulation. Their actions are mediated via pro-inflammatory moleculesincluding cytokines, chemokines <strong>and</strong> eicosanoids secreted by <strong>the</strong> macrophages <strong>and</strong> by <strong>the</strong> follicle cells <strong>the</strong>mselves.Recent evidence implicates signals initiated in <strong>the</strong> endometrium in response to semen exposure, in enhancing thisleukocytic infiltration of <strong>the</strong> ovary, <strong>and</strong> influencing <strong>the</strong>ir pattern of cytokine secretion, with important consequences inenhancing ovulation <strong>and</strong> luteinization.82. OESTROGEN TARGET SITES IN HUMAN TESTISPTK Saunders, TL Gaskell, JE Sierens, MR Millar, RM Sharpe <strong>and</strong> GA ScobieMRC Human Reproductive Sciences Unit, Edinburgh University Medical School, Edinburgh EH16 4SB, UKIt has been proposed that oestrogens play a role in regulating germ cell function in adulthood <strong>and</strong> during fetal life.Oestrogen action is mediated via high affinity intracellular receptors expressed in target tissues [1]. Two subtypes ofoestrogen receptor known as ERa (NR3A1) <strong>and</strong> ERp (NR3A2), have been cloned <strong>and</strong> hER~ variant iso<strong>for</strong>msidentified. In target cells <strong>the</strong>se receptors can exist as homo- or heterodimers [1]. We have used immunohistochemistryto examine <strong>the</strong> patterns of expression of ERs in human fetal <strong>and</strong> adult testis to determine <strong>the</strong> cellular targets <strong>for</strong>oestrogen action. In parallel studies we have prepared ER constructs <strong>and</strong> used <strong>the</strong>se to examine steroid binding to ERhomo- <strong>and</strong> heterodimers in transfected cells.In human testicular tissues (fetal or adult) we have never detected ERa mRNA or protein. With a polyclonal antibodywe detected ER~ protein in multiple testicular cell nuclei including those of Sertoli cells, Leydig cells <strong>and</strong> peritubularmyoid cells as well as some germ cells [2]. Additional studies using monoclonal antibodies capable of discriminatingbetween human wild type ER~ <strong>and</strong> <strong>the</strong> human ER~cx/~2 [3] splice variant revealed discrete patterns of expression of<strong>the</strong> subtypes. In adult testis immunoexpression of wtER~ was most intense in pachytene spermatocytes <strong>and</strong> roundspermatids, whilst low levels of expression were detected in Sertoli cells, spermatogonia, preleptotene, leptotene,zygotene <strong>and</strong> diplotene spermatocytes. Expression of ER~cx/P2 protein was highest in Sertoli cells <strong>and</strong> spermatogoniawith low/variable expression in preleptotene, pachytene <strong>and</strong> diplotene spermatocytes [4]. In <strong>the</strong> fetal testis wtER~ wasundetectable in gonocytes whereas <strong>the</strong>se cells expressed <strong>the</strong> highest levels of ER~cx/~2 compared with o<strong>the</strong>r testicularcell types. Both ERp1 <strong>and</strong> ER~cxl~2 were detected in some but not all Sertoli cells, peritubular cells <strong>and</strong> Leydig cells.Transfection studies with ERa <strong>and</strong> ER~ constructs confmned that wtER~ <strong>and</strong> ERa were activated following exposureto oestogenic lig<strong>and</strong>s but that <strong>the</strong> ER~cx/~2 variant did not bind oestrogens <strong>and</strong> was not capable of activating an EREcontainingreporter construct.The testicular cells most likely to be targets <strong>for</strong> oestrogens in adulthood are round spermatids in which levels ofexpression of wtERp1 are high. In contrast, expression of ERcxlP2, an iso<strong>for</strong>m that may act as a dominant negativeinhibitor of ER action, in adult Sertoli cells <strong>and</strong> spermatogonia, <strong>and</strong> in fetal gonocytes could prevent <strong>the</strong>se cellsresponding to oestrogens.1. Nilsson, S., et al., Phys Rev, 2001. 81: 1535. 2. Saunders, P.T.K., et al., Mol Hum Reprod, 2001. 7: 227. 3. Ogawa,S., et al., NAR, 1998.26: 3505.4. Saunders, P.T.K., et al., JCEM, 2002. 87: 2706.8483. MALE CONTRACEPTION - APPROACH TO HORMONAL METHODSRobert I McLachlanPrince Henry's Institute ofMedical Research, Melbourne VictoriaWorldwide, men particip~te actively in contraception through natural methods, condo~ usage <strong>and</strong> vasectomy but no new,acceptable, safe <strong>and</strong> reverSIble methods have become ~v~lable. Male hormonal contraception (MHC) is <strong>the</strong> most promising avenueof research. T~stosterone (T). treatment suppresses pItUItary LH <strong>and</strong> FSH secretion, <strong>the</strong> primary endocrine signals required <strong>for</strong>spermatoge~esI.s. Large mUltIc~ntre WH~-sponsored trials have shown that high doses of T reduce sperm counts to zero(azoospeI1~l1a) In -70% CaucaSIan men whIle over 90% have sperm counts < 1 million/m (severe oligospermia), a level at whichcontraceptIve efficacy appears adequate. ~ut <strong>the</strong> need <strong>for</strong> frequent injections <strong>and</strong> <strong>and</strong>rogenic side effects (acne, mood changes,redu~ed ~DL-cholesterol) dem<strong>and</strong> al~ernat~ve methods of T administration. Testosterone implants provide <strong>the</strong> prototype <strong>for</strong> morephySIOlogICal T treatm~nt .but pellet InsertI.o~extrusion limit <strong>the</strong>ir wide application. New long acting intramuscular preparations,such as T undecanoate III 011, are most promlSlng <strong>and</strong> may allow 8-12 weekly injection intervals.Co-admi~istration of .o<strong>the</strong>r gonadotropin-suppressing agents, such as GnRH antagonists or progestins, enhance spermatogenicsu~pressI~n .<strong>and</strong> permI.t <strong>the</strong> use of lower T doses. In MHC, progestin action is probably via gonadotropin suppression but directactIons WIthIn <strong>the</strong> testIS have not been excluded. A range of progestins have been used including levonorgestrel desogestrel <strong>and</strong>depot med:oxy-prog~sterone acetate, <strong>and</strong> studies have shown that -75% <strong>and</strong> -95% of men become azoospe~c or severelyohgospermIc, respectively. The only T plus progestin efficacy study to date has reported excellent contraceptive efficacy over a 1year ~xposure (Tu~er et al, ESA 2002, a~stract). The stage is set <strong>for</strong> pharmaceutical company involvement, sadly lacking in <strong>the</strong>past, In order t~ b~ng <strong>the</strong> first MHC regImen to market. About 5% of men fail to suppress adequately <strong>and</strong> underst<strong>and</strong>ing <strong>the</strong>reason(s) <strong>for</strong> <strong>the</strong>Ir faIlure to respond is important in gaining <strong>the</strong> widest acceptance of MHC.We have explored <strong>the</strong> MHC effects on sp~rmatogenesis in men using stereological techniques <strong>for</strong> germ cell quantification <strong>and</strong>showed that. type Apale--+B spermatogomal development <strong>and</strong> sperm release (spermiation) are <strong>the</strong> key sites of action. Thesperm~to~omal. defe~t probably results from FSH withdrawal but <strong>the</strong> subtypes affected <strong>and</strong> <strong>the</strong>ir mechanism of action are unknown.SpermIatIon faIlure IS a feature of both acute (accounting <strong>for</strong> dramatic falls in sperm output within 3 weeks) <strong>and</strong> chronic MHCtreatment. Both FSH <strong>and</strong> T regulate rodent spermiation but nothing is known in man. Cell-cell adhesion/communication <strong>and</strong>regulation of gene expression in later germ cell types can now be explored using laser capture microdissection <strong>and</strong> in vitro culture.F~om an endocrine regulatory viewpoint, MHC suppresses serum LH to 0.05). In ~ll treatment groups pnm?rdIal follicles were <strong>the</strong> most abundant follicle type, however primary <strong>and</strong>secondary f~lhcles were also observed m grafts from all treatment groups. This study confirms that xenograftingsup?orts folhcular development within wallaby pouch young ovarian tissue (1-2). Interestingly, follicle survival <strong>and</strong>folhcular development was sustained in all treatment groups, which indicates that <strong>the</strong> hormonal environment of <strong>the</strong>recipient may not playas crucial a role as previously believed. [1] Mattiske et al (2001) Proc Aust Mamm Soc 47:35.[2]Mattiske D et al (2002) <strong>Reproduction</strong> 123:143-153. [3]Cox S-L et al (2000) Fertil Steril74:366-371.85


85. DEVELOPMENT OF ANTRAL FOLLICLES IN RABBIT OVARIAN XENOGRAFTSIN IMMUNODEFICIENT MICEK. Muirhead l , S-L. Cox 2 <strong>and</strong> M. Holl<strong>and</strong> l .1Pest Animal Control Cooperative Research Center, CSIRO Sustainable Ecosystems, GPO Box 284, Canberra, ACT2615, 2Department ofPhysiology, Monash University, Clayton 3800The primordial follicle pool is established in mammalian ovaries in v~ry early ~ife. Fro~ ~is. time <strong>for</strong>ward <strong>the</strong>population of primordial follicles progressively decreases as a result of eI<strong>the</strong>r atre~Ia or th~ ImtlatlOn of g~owt~ <strong>and</strong>development. Very little is understood about <strong>the</strong> processes that regulate normal follIcle recrU1tme~t <strong>and</strong> .survIVal ~n <strong>the</strong>rabbit. This report describes an initial trial in <strong>the</strong> use of ovarian xenografting as a means to mvestlgate folhculardynamics in juvenile rabbits. Fresh ovarian tissue from NZ White rabbits aged 2, 4, 8 <strong>and</strong> 12 weeks old were graftedunder <strong>the</strong> renal capsule of ovariectomised NOD-SCID mice (n=35). Grafts were recovered 8 days, 2 weeks, 4 weeks or8 weeks later, assessed macroscopically <strong>and</strong> subjected to histological evaluation. All grafts were present <strong>and</strong> hadbecome vascularised at <strong>the</strong> time of autopsy. Grafts from two-week-old donor rabbits recovered 2 <strong>and</strong> 4 weeks aftergrafting did not contain any follicles. In contrast four-week-old donor rabbit tissue (grafted <strong>for</strong>.4 weeks) containe?healthy looking primordial <strong>and</strong> preantral follicles, resembling folliculogenesis normally observed m 8-week-old rabbItovaries. Similarly four-week-old donor tissue (grafted <strong>for</strong> 8 weeks) contained follicles at all stages of developmentincluding antral follicles, resembling 12-week-old normal rabbit ovaries. Whilst tissue from 8 week old donors (grafted<strong>for</strong> 4 weeks) displayed apparently normal primordial, preantral <strong>and</strong> antral follicles (diameters of approximatel~ 1rnn:),tissue grafted <strong>for</strong> only 8 days <strong>and</strong> 16 days contained primarily degenerating preantral follicles <strong>and</strong> healthy pnmordlalfollicles. Interestingly, corpora lutea like structures were detected in a number of grafts from donors aged 12 w~eksthough "ovulation" of normal oocytes remains unconfirmed. In general <strong>the</strong> dev~lopment a~d morpholo~y of folhcleswithin <strong>the</strong> xenografted rabbit ovarian tissue resembled <strong>the</strong> normal pattern of folhculogenesls. Thus ovarian xenograftsin NOD-SCID mice can serve as an experimental model <strong>for</strong> juvenile rabbit follicular growth <strong>and</strong> development. Weintend to use this technique in conjunction with an in vitro culture system we have developed <strong>for</strong> rabbit ovarian corticalexplants to investigate factors regulating folliculogenesis.86. ACUTE FSH WITHDRAWAL IMPAIRS SERTOLI AND GERM CELLDEVELOPMENT IN THE IMMATURE RATSarah J. Meachem, Jessica Ziolowski# <strong>and</strong> Kate L. Lovel<strong>and</strong>#Prince Henry's Institute ofMedical Research, Clayton, Victoria, 3168#Monash Institute of<strong>Reproduction</strong> <strong>and</strong> <strong>Development</strong>, Monash University, Clayton, Victoria, 3168Pituitary follicle stimulating hormone (FSH) is required <strong>for</strong> normal testicular developmen~, affecting both Serto.li ~dgerm cells (l) through its receptors on Sertoli cells, <strong>and</strong> <strong>the</strong>reby determining <strong>the</strong> po~ent1al <strong>for</strong> sperm productlO.n madulthood. The impact of FSH on in vitro Sertoli cell proliferation alters during maturatIOn (2), though <strong>the</strong> mech~ms~sunderpinning <strong>the</strong>se actions <strong>and</strong> <strong>the</strong>ir development are not understood. To establish an in ~ivo model <strong>for</strong> examm~tlOnof <strong>the</strong> role of FSH at discrete developmental stages, we assessed <strong>the</strong> impact of acute FSH wIthdrawal on <strong>the</strong> Sertoli <strong>and</strong>germ cell populations during early postnatal life. Sprague Dawley rat pups (n = 7/grou~) ~e.re ~assively immunisedagainst FSH using an ovine polyclonal antibody to rat FSH (10 mg/kg by subcutaneous daIly mJect~on) <strong>for</strong> 2 <strong>and</strong> ~ daysprior to death on days 3, 9 <strong>and</strong> 18 days postpartum. Control animals recei~ed a normal. sh~ep lmmunoglobulm ~10mg/kg). Testes were immersion fixed with Bouin's fluid <strong>and</strong> embedded in resm <strong>for</strong> det~rnun~tlOn of cell num~er us~ng<strong>the</strong> optical disector stereological technique, <strong>and</strong> paraffin <strong>for</strong> assessment of prolif~ratlOn <strong>and</strong> apopto~lS usmgimmunohistochemistry. Sertoli <strong>and</strong> germ cell numbers per testis were unchanged followmg 2 days of FSH WIthdrawalat day 3, 9, <strong>and</strong> 18 compared to controls. In response to 4 days of FSH withdrawal in day 9 rats, Sertoli cell an~ typeA/intermediate spermatogonial number were decreased to 73% (P


89. IMMUNOCONTRACEPTIVE EFFECT OF RECOMBINANT PORCINE ZONAPELLUCIDA C PROTEIN IN RABBITSE McLaughlin\ AJ Robinso~,B Inglis 2, J Swan 2 , M.K. Holl<strong>and</strong> 2 & PJ Kerr 21School ofEnvironmental & Life Sciences, University ofNewcastle, Callaghan NSW 2308& 2Pest Animal Control CRC, CSIRO Sustainable Ecosystems, GPO Box 284, Canberra, ACT2615.Despite <strong>the</strong> success of biocontrol methods of population control (such as calicivirus) rabbit populations within Australiaremain a significant problem particularly in temperate zones. An alternative is to induce sterility in <strong>the</strong> female rabbitpopulation via immunisation with a viral vector containing an ovarian antigen. <strong>Fertility</strong> trials using total porcine zonaepellucida as an immunogen have been shown to induce infertility in a number of diverse species. Here we describe <strong>the</strong>production of recombinant porcine Zona Pellucida C (pZPC) protein <strong>and</strong> its use as an immunocontraceptiveimmunogen in small scale fertility trials in domestic rabbits. Recombinant protein was produced by expression of <strong>the</strong>full length porcine ZPC cDNA in a vaccinia virustr7 system in a co-infected monkey kidney cell line (CVl) <strong>and</strong>purified using lectin affinity chromatography. Two adult male <strong>and</strong> ten adult female laboratory rabbits were inoculatedwith IOOllg recombinant pZPC in Freunds Complete adjuvant <strong>and</strong> boosted twice with 50llg in Incomplete Freundsadjuvant on Days 28 <strong>and</strong> 56. Sera were collected prior to immunization <strong>and</strong> following boosting (Days 14,44, 70). Fourmale rabbits of proven fertility were used to mate both immunized <strong>and</strong> untreated control rabbits (n=4). The immunizedrabbits were sacrificed 9 days post mating <strong>and</strong> <strong>the</strong> reproductive tracts examined <strong>for</strong> corpora lutea <strong>and</strong> implantation sacs.Ovaries from rabbits immunized with recombinant pZPC were fixed in Bouin's solution. Tissues were paraffinembedded <strong>and</strong> sectioned prior to staining with hematoxylin <strong>and</strong> eosin. By Day 70 sera from all animals containedantibody capable of detecting recombinant pZPC protein by Western blotting. Immunoperoxidase staining was detectedon <strong>the</strong> zona pellucida of early primary <strong>and</strong> more mature follicles when <strong>the</strong> antisera from immunized animals were usedto probe normal ovarian sections. At autopsy all ten immunized animals had visible corpora lutea but only two werepregnant with 10 <strong>and</strong> 2 viable implantations respectively. The remaining eight females displayed signs ofpseudopregnancy only. In contrast all control females (n=4) had normal sized litters (mean±sem. 7.25±1.3).Histological evaluation showed no difference in ovarian folliculogenesis between treated animals <strong>and</strong> age matchedcontrol specimens. Serum antibody titres measured using ELISA showed no difference between males <strong>and</strong> femalesimmunized with pZPC <strong>and</strong> no correlation with pregnancy. As antibody was not detected bound to <strong>the</strong> ZP in vivo <strong>the</strong>mechanism by which recombinant pZPC induced infertility remains to be elucidated.90. EFFECT OF ANGIOGENESIS INHIBITORS ON OESTROGEN-MEDIATEDENDOMETRIAL ENDOTHELIAL CELL PROLIFERATION IN THEOVARIECTOMISED MOUSEB Heryanto\KE Lipson 2 , <strong>and</strong> PAW Rogers!lCentre<strong>for</strong> Women's Health Research, Monash University Department ofObstetrics & Gynaecology, Monash MedicalCentre, 246 Clayton Rd, Clayton, Victoria 3168, Australia2SUGEN, Inc., 230 E. Gr<strong>and</strong>Ave, South San Francisco, CA,94080, USAIt has been suggested that endometrial angiogenesis in response to <strong>the</strong> sex steroids oestrogen (E) <strong>and</strong> progesterone (P)is mediated at a local level via compounds such as vascular endo<strong>the</strong>lial growth factor (VEGF), fibroblast growth factor(FGF), <strong>and</strong> platelet-derived growth factor (PDGF), acting through <strong>the</strong>ir respective tyrosine kinase receptors (RTK). Theaim of <strong>the</strong> present study was to use RTK angiogenic inhibitor compounds to determine if VEGF, FGF or PDGF playarole in mediating endometrial endo<strong>the</strong>lial cell (EC) proliferation following administration of E <strong>and</strong> P. Endometrial ECproliferation was induced in adult ovariectomized mice by ei<strong>the</strong>r E alone <strong>for</strong> 1 day (EI), or P with low dose E followedby P with high dose E (PE). Each angiogenesis inhibitor compound was injected daily <strong>for</strong> four days (lOOmg/Kg/day,s.c.) be<strong>for</strong>e endometrial tissue collection. We also evaluated <strong>the</strong> effect of VEGF antiserum (0.2ml i.p.) on ECproliferation at E1. All <strong>the</strong> angiogenic inhibitor compounds significantly reduced EC proliferation activity at El <strong>and</strong>PE. The greatest reduction in EC proliferative index was at EI in <strong>the</strong> group treated with a VEGF receptor inhibitor (2.5± 0.7% versus 27.9 ± 1.1 %, P


93. MAST CELL NUMBER AND SPATIAL LOCATION IN THE VAGINAL CUL-DE-SACOF THE BRUSHTAIL POSSUM AFTER ADMINISTRATION OF EXOGENOUSOESTRADIOLPM Mahonel, PR Hurse, BJ McLeod!,2, MA McConnele1Department ofAnatomy <strong>and</strong> Structural Biology; 3Department ofMicrobiology, University ofOtago, Dunedin, NewZeal<strong>and</strong>. 2AgResearch Invermay Agriculture Centre, Mosgiel, New Zeal<strong>and</strong>Mast cells are present in large numbers in <strong>the</strong> vaginal cul-de-sac, a component of <strong>the</strong> reproductive tract in femaleBrushtail possums. Their presence does not appear to be an immunological response to <strong>the</strong> presence of microflora, asestablished in a previous study of mast cell number <strong>and</strong> spatial location in cul-de-sac tissues (1). In this study mast cellnumbers <strong>and</strong> spatial location were determined in seasonally anoestrus possums <strong>and</strong> those treated with sesame oil oroestradiol 17~ implants <strong>for</strong> 6 days.Cul-de-sac tissue was collected aseptically <strong>for</strong> microbiological analysis, <strong>and</strong> <strong>the</strong> Fractionator <strong>and</strong> Optical Disectorstereological methods were used to quantify mast cell populations in tissue sections.In all groups (n=6/group), microflora populations were consistently low «4.0 x 10 4 organisms gram-I). Mean weight<strong>and</strong> volumes of epi<strong>the</strong>lial <strong>and</strong> connective tissues were significantly greater in oestradiol treated animals). Meannumbers of mast cells in epi<strong>the</strong>lial <strong>and</strong> connective tissues did not differ significantly amongst groups. Mean mast celldensity was <strong>the</strong>re<strong>for</strong>e significantly greater in <strong>the</strong> seasonally anoestrus <strong>and</strong> control animals than <strong>the</strong> oestradiol treated.Although previous emperical studies may have reported a decrease in mast cell number when circulating oestrogenlevels were elevated (2, 3) this study has highlighted <strong>the</strong> need <strong>for</strong> careful interpretation of both tissue volume <strong>and</strong> cellnumber (4).References(l)Mahoney et al., (2002) (in press). (2)Krishna et al., (1986) Journal of <strong>Reproduction</strong> <strong>and</strong> <strong>Fertility</strong> 32:1211-7.(3)Wilhelm et al., (2000) Endocrinology 141:1178-86. (4)Mayhew TM <strong>and</strong> Gundersen HIG (1996) Journal ofAnatomy 188:1-15.94. IMMUNOLOCALISATION OF INTERLEUKIN 11 AND ITS RECEPTOR INCYCLING ENDOMETRIUM AND IMPLANTATION SITES OF THE RHESUS MONKEYE. Dimitriadist, L. Robb 2 , v-x Liu 3, W. M. Carter 2 , H. M. Martin 2 , L. A. Salamonsen!Prince Henry's Institute ofMedical Research, Clayton, 3168 1 , The Walter And Eliza Hall Institute ofMedical Research,PO Royal Melbourne Hospital, Melbourni, Institute ofZoology, Beijing, 100080, China 3 .Interleukin 11 (IL-l1) signalling is essential <strong>for</strong> stromal cell decidualization <strong>and</strong> embryo implantation in <strong>the</strong> mouse 1<strong>and</strong> is involved in decidualization in <strong>the</strong> human 2 . IL-ll action is mediated via binding to its specific IL-l1 receptoralpha (IL-I1Ra). The aim was to examine <strong>the</strong> temporal <strong>and</strong> spatial location of IL-l1 <strong>and</strong> IL-I1Ra in cyclingendometrium <strong>and</strong> implantation sites of <strong>the</strong> Rhesus monkey. Immunohistochemistry was per<strong>for</strong>med on cycling monkeytissue from proliferative phase day 13 (one day be<strong>for</strong>e ovulation) <strong>and</strong> secretory phase days 19,24 <strong>and</strong> 29 (5, 10, <strong>and</strong> 15days respectively after ovulation) (n=4 per day), <strong>and</strong> from implantation sites between days 24 <strong>and</strong> 35 of pregnancy(n=8). Data was expressed as relative intensity units. IL-ll immunoreactivity was elevated in <strong>the</strong> secretory phase of<strong>the</strong> menstrual cycle compared to <strong>the</strong> proliferative phase. At day 24 of <strong>the</strong> cycle, both IL-ll (2.8±1.2, mean±SEM) <strong>and</strong>IL-llRa (3.4±0.8) staining were most intense in gl<strong>and</strong>ular epi<strong>the</strong>lium. Staining in <strong>the</strong> stroma <strong>for</strong> IL-ll was minimal(0.4±0.1) <strong>and</strong> moderate <strong>for</strong> IL-llRa (1.3±0.3). By day 29 of <strong>the</strong> cycle staining <strong>for</strong> IL-ll was higher in stromacompared to gl<strong>and</strong>ular epi<strong>the</strong>lium (3.4±0.8 vs 1.7±0.6, P


-AUTHOR INDEXAuthorAbstract No.Aitken, RJ 5,9,54,56 Duckworth, J 28Aloe, J 78 Dyson, M 35, 71Amman,G 63Anderson, ST 65,69 Ecroyd, H 54,88Armstrong, DT 40,48,73,81 Eriksson, BM 19Ashman, RJ 14 Ethier, J-F 71Aspden, WJ 24,37 Evans, G 19,21,22Asquith, K 54Assinder, S 49,53 Fenwick, MA 59Findlay, IK 35,62,71,92Baker, MA 9 Fink, J 53Barclay, JL 65 Fisher, PI 17Bathgate, R 19 Fleming, JS 27,57Beagley, KW 88 Forbes, E 47Blewitt, M 2Breier, B 15 Galloway, SM 80Britt,K 62,63 Gargett, CE 61Brockway, A 20 Garry, PG 42Bromfield, JJ 77 Gaskell, TL 82Bucak, K 61 Gibbs, GM 8Bunn, SJ 42 Gilbert, RE 58Byers, S 38 Gilchrist, RB 39,40,70,73Grattan, DR 65Cakouros, D 68 Green, DPL 36Carter, WM 94 Gregan, S 80Cetica, P 39 Gromoll, E 6Chong, S 2 Grupen, CG 14Chrousos, GP 66Clarke, HG 38 Hallett, A 29Clarke, II 43 Hamilton, HM 16Clyne, CD 64 H<strong>and</strong>elsman, DJ 47Collett, RA 13 Hanrahan, SP 80Conway, AJ 47 Harding, R 95Cook, S 78 Hartwich, KM 16Cotton, LM 8 Harvey, AJ 12,13Cowan, P 28 Hedger, M 45,51Cox, S-L 84,85 Herlihy, A 55Crone, J 63 Herrid, M 7Cui, X 28 Heryanto, B 90Curlewis, JD 65,69 Hessian, PA 36Hickox, D 55Daish, T 68 Hill, JR 17Davies, CJ 17 Holl<strong>and</strong>, MK 1,85,89Davis, GH 80 Hollinshead, FK 21,22de Gooyer, TE 58 Hope, SA 38de Kretser, DM 8,44,45,51,55,87 Hospers, M 95Dekker, GA 75 Hurst, PR 27,42,59,93Dent, L 29Dharmarajan, AM 67 Inglis, B 89Dimitriadis, E 94 Ingman, WV 50D'Occhio, MJ 24,37 Irving-Rodgers, HF 37,60Dorstyn, L 68Druker, R 2 Jasper, MJ 26Drummond, AE 35,44,62,71 Jenkin, G 8492 98


Jones, D 28 Montgomery, GW 57 Sawyer, H 80 Thomas, RE 73Jones, M 62 Morrison, JR 8 Schlafer, DH 17 Thompson, JG 12,13,39,73Jones, RC 56,88 Morrissey, MP 40 Schlatt, S 32 Tilbrook, AJ 43Juengel, JL 80 Moss, TJM 95 Scobie, GA 82 Torres, CAA 18Muir, J 45,51 Scobie, S 28 Tremellen, KP 33, 75Kakar, MA 23 Muirhead, K 85 Scott, CJ 43 Trigg, TE 24Kaye, PL 11 Murata, Y 63 Sebire, K 8,55 Trounson, A 30Keelan, J 15 Sferruzzi-Perri, A 29 Tumer,L 47Kelly,DJ 58 Newcombe, N 88 Sharkey, DJ 75Kelly, JM 16 Newnham, JP 95 Sharpe, RM 82 Waddell, BJ 74Kerr, PJ 89 Nicholson, HD 49,53,57 Shaw, G 25,96 Wade,MA 56Kind, KL 12, 13 Nie,G 92 Shaw, J 20,84 Waldhauser, F 63Kleemann, DO 16,23 Nieschlag, E 6 Short, RV 87 Walker, SK 16,23Knee, RA 88 Nitsos, I 95 Sidhu, KS 10 Waters, MJ 65,69Kovacic, A 64 Nogoy,N 52 Sierens, IE 82 Wells, D 15Kovacs, J 63 Nogueira, ET 18 Simoni, M 6 Weston, GC 91Krupa,M 60 Norman, RJ 26 Simpson, ER 62,63,64 White, C 72Krutskikh, A 9 NottIe, MB 14 Smith, JF 52,57 Whitelaw, E 2Kumar, S 68 Smith, JT 74 Wijayanti, GE 25Kusters, DHL 65 O'Brien, JK 21,22 Snow,M 84 Wilkinson-Berka, JL 58O'Bryan, MK 8,55 Speed, CJ 64 Williams, PJ 4Lau,P 65 Ochsenktihn, R 51 Stanton, JL 36 Wilson, T 80Le, M-T 71 O'Connor, A 45 Stephenson, T 46 Wishart, S 47Lee,R 15,52 Okuma, Y 45 Sun,M 49Lepore, D 78 O'Leary, S 48 Sutton, M 39 Yu,F 57Li,A 76 O'Neill, C 76 Swan, J 89Lipson, E 90 Zaitseva, M 61Liu, PY 47 Parry,L 53 Tam, SP 65 Zhang, Z 87Liu, Y-X 94 Paya, K 63 Tan, OL 27 Zhou, J 64Lorimer, MF 23 Peterson, J 15 Temple-Smith, PD 41 Ziolowski, J 86Lovel<strong>and</strong>, BE 31 Peura, TT 16 Thomas, P 78Lovel<strong>and</strong>, KL 44,86,87 Phillips, D 45Luetjens, CM 6 Preis, J 2Luu,K 92Quinn, MCJMacmillan, K 2036Maddocks, S 23 Rakyan, V 2Mahoney, PM 93 Rath,D 22Mancio, AB 18 Ravelich, S 15Markham, K 11 Renfree, MB 25,41,87,96Martin, GB 18 Richings, NM 41Martin, HM 94 Ritter, LJ 26,40Martin, L 3 Robb,L 72,94Mate, KE 10 Roberts, CT 95Mattiske, D 84 Robertson, DM 46Maxwell, WMC 19,21,22 Robertson, SA 29,34,48,50,75,77McConnell, MA 93 Robinson, AJ 89McDonald, R 57 Rodger, JC 10McFarlane,. J 7 Rodgers, RJ 37,38,60McIlfatrick, SM 14 Rogers, PAW 61, 90, 91McLachlan, RI 46,47,83 Roman, SD 56McLaughlin, E 89 Ross, S 78McLeod, BJ 53,93 Rudiger, SR 16,23McNair, L 96 Ryan, NK 26McNatty, KP 80Meachem, SJ 86 Salamonsen, LA 72,92,94Meehan, T 44, 87 Santiago, LL 18Meyers, SA 57 Saunders, PTK 82Millar, MR 82 Sawyer, DE 594 95


start time8:30am9:00am9:30am10:00am10:30am11:00am11:30amnoon12:30pm1:00pm1:30pm2:00pm2:30pmSunday22ndESAESAlSRB PROGRAM AT A GLANCEMonday23rdESAJapan/Australiaplenary,Hall 0, start8:45amServierAwardInvsetigatorScientistAwardHall 0SRBFoundersLectureHall 0SRBOrals #1&2Rooms 2 &10, start8:45amOrals #3&4Rooms 2 &10Lunch & Poster viewing* Posterdiscussion#1,2,3,4&5until 3:45includesafternoonteamorning teaSRBSymposiumRoom 2Symposium,Hall 0* FreeCommun.Rooms 11 &2HarrisonLectureHall 0Lunch & Poster viewing* Posterdiscussion#6,7,8,9,10 & 11until 3:45includesafternoonteamorning teaSRBSymposiumHall 0Wednesday25thESAJointADS/ESAplenary, HallB/C*SRBSymposiumRoom 2* ESAIADSSymposiumHall B/C* ESASymposiumRoom 1morning teaafternoon teaSRBSRBSymposium,Hall 0Lunch & Poster viewingposter Orals # 9 &viewing 10--..,...--,-__--l Rooms 1 & 2ESAIADSPlenaryHall B/Cstart time3:00pm3:30pm4:00pm4:30pm5:00pm5:30pm6:00pm6:30pm7:00pm7:30pm8:00pmSunday22ndESASRBSymposiumRoom 2Welcome FunctionExhibition FoyerWomen in EndocrinologyRiverview 3 (starts 7pm)Monday23rdESA* FreeCommun.(rooms 4 &10) <strong>and</strong> Jointreproductivesession (HallD)ESAAGM,Hall 0, starts5:45pm,SRBSRBafternoonteaOral #5Room 2<strong>and</strong>JointReproductive sessionHall 0SRB StudentMeeting,AdelaideClubStudent BBQ, AdelaideUniversity ClubGold Coast RoomTuesday24thESA* FreeCommun.(rooms 11 &10 <strong>and</strong> HallD)SRBSRBafternoonteaSRB JuniorScientistAwardFinalistsRoom 1Taft Lecture, SRB AGM,Hall 0 Room 1Conference DinnerStam<strong>for</strong>d Gr<strong>and</strong> Gelenelg,catch tram at 7pmWednesday25thESAESASymposium,Room 1ESAIADSsymposium,Hall B/CSRBESAlSRBlate breakingscience,Room 296 97

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!