27.01.2015 Views

&. u - the Society for Reproductive Biology

&. u - the Society for Reproductive Biology

&. u - the Society for Reproductive Biology

SHOW MORE
SHOW LESS

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

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

&. u


The Australian <strong>Society</strong> <strong>for</strong> <strong>Reproductive</strong> <strong>Biology</strong><br />

Twentieth<br />

Newcastle University<br />

29-31 August 1988<br />

Programme and Abstracts of Papers<br />

Copyright Australian <strong>Society</strong> <strong>for</strong> <strong>Reproductive</strong> <strong>Biology</strong>, 1988<br />

ISSN 0705-6044<br />

CONTENTS<br />

Personnel<br />

Acknowledgements.<br />

Programme Guide<br />

Programme<br />

Author Index.<br />

Abstracts.<br />

iii<br />

iv<br />

v<br />

vi-xx<br />

xxi-xxvii<br />

1-108


AUSTRALIAN SOCIETY FOR REPRODUCTIVE BIOLOGY<br />

August, 1988<br />

Chairman<br />

Secretary<br />

Treasurer<br />

Committee Members<br />

OFFICE BEARERS<br />

Professor B. Setchell<br />

Dr G. Evans<br />

Dr L.A. Hinds<br />

Dr B.M. Bindon<br />

Dr I.J. Clarke<br />

Dr L. Martin<br />

Dr C.D. Nancarrow<br />

Dr J.N. Shelton<br />

Postgraduate Student<br />

Representative<br />

Mr H.J. Jabbour<br />

PROGRAMME COMMITTEE<br />

Chairman<br />

Committee Members<br />

Ass:x::. P.r:ofessar G.M. Stale<br />

Dr D. Handelsman<br />

Dr B. Miller<br />

Dr R. Scaramuzzi<br />

Dr C. Tsonis<br />

LOCAL ORGANISING COMMITTEE<br />

Dr J.R. Rodger<br />

Mr J. Clulow<br />

Dr R. Jones<br />

Convenor<br />

Dr R.Murdoch<br />

Dr T. Roberts<br />

iii


• ~~':!,<br />

'U 'O:s: ~~~S!<br />

~. ~ ~ rt ~ 0 CD<br />

° i3 CD ::r' 0 f-'. 0<br />

CD f-"M g ~ CD 0<br />

Ho<br />

o ~~. mmrt~ ~. til<br />

f-'. III • f-'. "< rt Ul ....<br />

o~ tJ' CD ::r' ...<br />

'U ~ f-'. ~ CD ....<br />

M ~ ~Ort Ul<br />

o o~ ~MUl en 6<br />

oCD Ho rt "< rto:S: Ho I:"il rt 0 ~<br />

a ~~ ~~ ~ rt<br />

~.•• gmrt OJ .. ~ H<br />

~ U>~ g t< g; g ~<br />

'lil 0 ~OJm " ~ 0 ~"<br />

rt ~." n~. "0 11l, ""'"rt g<br />

°:"t o :; rt:'l " !'l 0; '" ~ ,,'" rtg H<br />

0'U OP.CD a a 1-3 tJ>I 0 en<br />

5 ~£ ~gg I~~ ~S~~~;rHo I:"il<br />

~ a .... U> 0'" 0"" 0 ~<br />

1l- 'lil rt ~ 15~C:~~[il~g;d~"~ (Xlf-" ~<br />

H .. U> ~ U> '" .. t< n a ~ '"<br />

M l:l CDM ...• nZ::i1-3t:i (j)~Hen CDIQ bil<br />

m ~ n·~ ['l""~t: 8 iii"U> :"t~o ~<br />

,:, ~. 0 g m ~ ~ "' .. ~ ~ _ .. 15 ~ ::;;::: '"<br />

• ~ ~p.~ 0 '" (:J ~ d U> U> .... U> U> "'~ 0<br />

III t-t t:i.... q en H H f-'.<br />

O§M~<br />

a<br />

CJ)H en ~M t:1<br />

{J) ::s CD CD > t-


AUSTRALIAN SOCIETY OF REPRODUCTIVE BIOLOGY<br />

PROGRAMME 1988<br />

Session 2 (Poster) : DEVELOPMENTAL BIOLOGY<br />

chairman: Professor R. Wales<br />

Time: 1100 - 1230 Venue: Function Room 3<br />

REGISTRATION: The registration desk will be located at <strong>the</strong> top of<br />

<strong>the</strong> Main Entrance Stairs (middle floor) of <strong>the</strong> Town Hall and will<br />

be open from 1530-1800 on Sunday, 28 August and 0830-1200 on<br />

Monday, 29 August.<br />

The Goding Lecture will be presented in <strong>the</strong> Concert Hall and oral<br />

sessi~ns will be in Function Room 2. Posters will be displayed in<br />

Funct~on ~oom 3. All poster presentations will be in Function<br />

Room 3 except <strong>for</strong> Session 12 where <strong>the</strong> presentation will be in<br />

Function Room 2.<br />

5<br />

6<br />

S.A. McKay and A. Lopata<br />

The h.lock t:o po.lyspeI111.:ic fert:.:i.l.:izat:.:ion .:in mouse oocyt:es<br />

P.L. Kaye<br />

Ion.:ic requ.:ire.ment:s of g.lyc.:ine t:ransport: .:in mouse emh.zyos<br />

7 E. De Luca and A. Lopata<br />

Session 1 (Oral) FERTILITY<br />

Chairman: Dr G. Evans<br />

Time: 0930 - 1030<br />

Monday 29 August<br />

Venue: Function Room 2<br />

1 P.A. Hamilton, 1.0. Killeen and J. Reeve<br />

2<br />

Effect:s of met:hod of freez.:ing ram semen and dose of PHSG on<br />

fert:.:i.l.:it:y of ewes .:inse.m.:inat:ed .int:o t:he ut:erus<br />

J.F. Smith, J.N. Clarke, D.L. Johnson and L.T. McGowan<br />

D.:ifference het:ween t:wo genet:.:ic .l.:ines of Romney ewes,<br />

se.lect:ed <strong>for</strong> .increased fecund.:it:y, .:in t:he.:ir ovu.lat:ory<br />

response t:o PNSG<br />

Effect: of an .:insu.l.:in/t:ransferr.:in/se.len.:it:e supp.le.ment: on DNA<br />

synt:he~ ..is .:in mouse emh.zyos cu.lt:ured .:in v.:it:ro<br />

8 I. Bruck and J.H. Hyland<br />

Ana.lys.:is of t:he cont:r.:ihut:.:ions of t:he Emdben-./t:feyerhoff and<br />

pent:ose-phosphat:e pat:hways t:o g.lucose met:aho.l.ism hy s.1:ng.le<br />

equ.:ine e.m.b.zyos .:in cu.lt:ure<br />

9 A. Szell and J.N. Shelton<br />

10<br />

Osmot:.:ic response of sheep and cat:t:.le emhiyos t:o peI111eat:.:ing<br />

c.zyoprot:ect:ant:s<br />

C. Herr, N. Holt, and K.C. Reed<br />

Effect: of sucrose and ca.lc.:ium .:in t:he sp.l.it:t:.ing med.:ium on<br />

surv.:iva.l of quart:ered ov.ine moru.lae<br />

11 M.B. Harvey and P.L. Kaye<br />

3<br />

A.J. Ritar, P.O. Ball, P.J. O'May, T.M.<br />

and N. Murray<br />

Black, R.B. Jackson<br />

AnahoLic effect:s of .I."nsu.l.:in<br />

med.:iat:ed v.:ia .I."nsu.l.:in recept:ors<br />

on mouse h.last:ocyst:s are<br />

Superova.lat:.1:on response and e.m.b.zyo recove.zy from cashmere<br />

and angora does aft:er t:reat:ment: w.:it:h FSH (fo.l.lt:roph.:inJ<br />

12<br />

J.L.Clarkson and C.D. Nancarrow<br />

4<br />

J.G.E. Thompson and G.W.<br />

Asher<br />

Superova.lat:.:ion and ova recovery .:in faI111ed fa.l.low deer (Dama<br />

damaJ<br />

13<br />

Compar.:ison of t:rophob.last:.:ic ves.:ic.le and ov.:ine b.last:ocyst:<br />

secret:ed prot:e.:ins and ana.lys.:is of ves.:ic.le met:abo.l.ic<br />

act:.:iv.:it:y.:in v.:it:ro<br />

A..Lopata, P.M.<br />

Summers and J. P. Hearn<br />

vi<br />

Cont:ro.l of t:he ovar.:ian cyc.le .in maI111oset: monkeys <strong>for</strong> .:in<br />

v.:i t:ro and .:in v.I.'vo fert:.:i.l.:izat:.I.·on and t:he t:ransfer of<br />

vii


viii<br />

ix<br />

14<br />

cu.ltured embryos to synchron.ized rec.ip.ients<br />

M.J. Sinosich, S. Lanzendorf, M.D. Bonifacio, D.M. Saunders<br />

and G.D. Hodgen<br />

IJl1Illunof.luorescent stud.ies of pregnancy-assoc.iated e.lastase<br />

.inh.ib.itor (PAEI) express.ion by act.i.rrated gametes and<br />

pre.i.mp.lantat..ion hamster embryos<br />

15 Y.C. Smart, L. Adamson, R.A. Wilcox and T.K. Roberts<br />

Re.lease of qrowth factor(s) by embryo der..ived p.late.let<br />

act.:ivat.:ing factor (EPAF)<br />

III<br />

16 L. Adamson, J.D. Stanger, Y.C. Smart and T.K. Roberts<br />

PAF-Pretreat:ment ..impa.1.'rs embryon..ic deve.lopment<br />

21<br />

22<br />

23<br />

P.Pollanen and B.P. Setchell<br />

The pe.I7lleab.i..l..ity to qamma-q.loba.l.i.n of <strong>the</strong> b.lood vesse.ls of<br />

<strong>the</strong> rat test.i.s around <strong>the</strong> t..ime of puberty<br />

A.J. Tilbrook, D.B. Galloway, A.H. Williams, R.C.<br />

Oppenheim, W.J. Thiel and I.J. Clarke<br />

Treatment w..ith a GnRH agon.ist de.lays reproduct..ive<br />

deve.lopment .1.'n ram .lambs<br />

C.J. Carati, K.E. Creed and E.J. Keogh<br />

Hechan..isms of pen..i.le erect.1.'on .i.n<br />

<strong>the</strong> doq<br />

17 P.A. Batt, A.W.M. Cameron, D. Sakkas and A.O. Trounson<br />

In v.:itro cu.lture of goat<br />

embryos<br />

SESSION 4<br />

Chairman:<br />

(Poster) PITUITARY HORMONES, INHIBIN AND OVARY<br />

Dr J.K. Findlay<br />

Time: 1530 - 1700 Venue : Function Room 3<br />

Session 3 (Oral) : MALE REPRODUCTIVE FUNCTION<br />

Chairman: Professor D. de Kretser<br />

Time: 1330 - 1500 Venue Function Room 2<br />

24 R. Bathgate, C. Sernia and R.T. Gemmell<br />

Ident..if.i.cat.i.on by HP.LC and ..immunocytochem.i.stry of<br />

hypotha.lam.:ic oxytoc.1.·n and mesotoc.1.'j] .:in <strong>the</strong> brashta.1.·.l possum<br />

25 P.R. Lewis, M.B. Renfree and R.V. Short<br />

18<br />

Qihan Dong and D.J. Handelsman<br />

Va.l.1.'dat.:ion of methodo.loqy <strong>for</strong> study of pu.lsat..i.le .LH<br />

secret.:ion ..in <strong>the</strong> rat: cannu.lat.i.on route, samp.l.:inq<br />

.:intens.1.'tyand durat.:ion<br />

26<br />

The re.lat.1.·onsh.1."p of breastfeed.ing patterns to <strong>the</strong> .lenqth of<br />

.lactat..iona.l amenorrhoea and ovar.ian .i.nact..iv.1.·ty post partum<br />

Qi Fa Wang, P.G. Farnworth, H.G. Burger and J.K. Findlay<br />

19<br />

A.E. Drummond, G.P. Risbridger and D.M. de Kretser<br />

The ro.le of .Leyd..iq ce.l.ls ..in <strong>the</strong> requ.lat.i.on of .:inh.1.·b..in<br />

product.1.'on<br />

27<br />

Inh.1.·b.i.tory effect of pure 31 kDa Bov.ine .1.·nh..ib..in on GnRH­<br />

..induced up-requ.lat.1.·on of GnRH b.i.nd..inq s..ites ..in cv.ltured rat<br />

anter..ior p..itu.1.'tary ce.l.ls<br />

Zhang Zhiwen, J.K. Findlay, R.S. Carson, A.C. Herington and<br />

H.G. Burger<br />

20<br />

J.A. Spaliviero, D.M. Robertson, E. Kidston, P.F. Hall and<br />

D.J. Handelsman<br />

H..igh.ly po.lar..ized secret.1.·on of .i.nh.:ib..in by ra t Serto.l.i. ce.l.ls<br />

..in tw..in-chamber cu.lture system<br />

28<br />

Trans<strong>for</strong>m.1.·nq growth factor B enhances basa.l and FSH<br />

st..imu.lated .inh..ib..in product..ion by rat: qranu.losa ce.l.ls ..in<br />

v.i.tro<br />

B.M. Bindon, T. 0' Shea, K. Miyamoto, M.A. Hillard, L.R..<br />

Piper, R.D. Ne<strong>the</strong>ry and G. Uphill


viii<br />

ix<br />

cu.ltured embryos to synchron.ized rec.fp.fents<br />

21<br />

P.Pollanen and B.P. Setchell<br />

14<br />

M.J. Sinosich, S. Lanzendorf, M.D. Bonifacio, D.M. Saunders<br />

and G.D. Hodgen<br />

The permeab.f.l.ity to gamma-g.lobu.l.fn of <strong>the</strong> b.lood vesse.ls of<br />

<strong>the</strong> rat test.fs around <strong>the</strong> t.fme of puberty<br />

15<br />

Immunof.luorescent stud.fes of pregnancy-assoc.fated e.lastase<br />

.fnh.fb.ftor (PAEI) express.fon by act.fvated gametes and<br />

pre.fmp.lantat.fon hamster embryos<br />

Y.C. Smart, L. Adamson, R.A. Wilcox and T.K. Roberts<br />

22<br />

A.J. Tilbrook, D.B. Galloway, A.H. Williams, R.C.<br />

Oppenheim, W.J. Thiel and I.J. Clarke<br />

Treatment w.fth a GnRH agon.fst de.lays reproduct.ive<br />

deve.lopment .in ram .lambs<br />

Re.lease of growth factor(s) by embryo der.fved p.lat:e.let<br />

act:ivat.fng factor (EPAF)<br />

,~l<br />

23<br />

C.J. Carati, K.E. Creed and E.J. Keogh<br />

Hechan.fsms of pen.f.le erect.fon .fn <strong>the</strong> dog<br />

16 L. Adamson, J.D. Stanger, Y.C. Smart and T.K. Roberts<br />

PAF-Pretreat:ment .fmpa.irs embryon.fc deve.lopment<br />

17 P.A. Batt, A.W.M. Cameron, D. Sakkas and A.O. Trounson<br />

In v.ftro cu.lture of goat emb.ryos<br />

SESSION 4<br />

Chairman:<br />

(Poster) PITUITARY HORMONES, INHIBIN AND OVARY<br />

Dr J.K. Findlay<br />

Time: 1530 - 1700 Venue Function Room 3<br />

Session 3 (Oral) : MALE REPRODUCTIVE FUNCTION<br />

Chairman: Professor D. de Kretser<br />

Time: 1330 - 1500 Venue Function Room 2<br />

24 R. Bathgate, C. Sernia and R.T. Gemmell<br />

Ident.if.fcat.ion by HP.LC and .fmmunocytochem.istry of<br />

hypotha.lam.fc oxytoc.fn and mesotoc.in .in <strong>the</strong> brushta.i.l possum<br />

25 P.R. Lewis, M.B. Renfree and R.V. Short<br />

18 Qihan Dong and D.J. Handelsman<br />

Va.l.fdat.fon of methodo.logy <strong>for</strong> study of pu.lsat.f.le .LH<br />

secret.fon .fn <strong>the</strong> rat: cannu.lat.fon route/ samp.l.fng<br />

.fntens.fty and durat.fon 26<br />

The re.latJ:onsh.ip of breastfeed.ing patterns to <strong>the</strong> .length of<br />

.lactat.iona.l amenorrhoea and ovar.ian .inact.iv.ity post partum<br />

Qi Fa Wang, P.G. Farnworth, H.G. Burger and J.K. Findlay<br />

19 A.E. Drummond, G.P. Risbridger and D.M. de Kretser<br />

The ro.le of .Leyd.Ig ce.l.ls .fn <strong>the</strong> regu.lat.fonof .fnh.fb.fn<br />

product.ion<br />

Inh.fbJ.·tory effect of pure 31 kDa BovJ.·ne .inh.fb.fn on GnRH­<br />

.fnduced up-regu.lat.ion of GnRB bJ.·nd.fng s.ftes .fn cu.ltured rat<br />

anter.<strong>for</strong> p.ftu.ftary ce.l.ls<br />

27 Zhang Zhiwen, J.K. Findlay, R.S. Carson, A.C. Herington and<br />

H.G. Burger<br />

20 J.A. Spaliviero, D.M. Robertson, E. Kidston, P.F. Hall and<br />

D.J. Handelsman<br />

H.fgh.ly po.lar.fzed secret.ion of .fnh.fb.fn by rat Serto.l.f ce.l.ls<br />

.fn tw.fn-chamber cu.lture system<br />

28<br />

Trans<strong>for</strong>m.fng growth factor B enhances basa.l and FSH<br />

stJ.·mu.lated J.·nh.fb.fn product.ion by rat: granu.losa ce.l.ls .in<br />

v.itro<br />

B.M. Bindon, T. O'Shea, K. Miyamoto, M.A. Hillard, L.R..<br />

Piper, R.D. Ne<strong>the</strong>ry and G. Uphill


29<br />

Superovu.lat:.ion .in pubert:a.l he.ifers .immun.ized aqa.inst: ov.ine<br />

.inh.ib.in pur.if.ied by monoc.lona.l ant:.ibody aff.in.it:y<br />

chromat:oqraphy<br />

J.K. Finqlay, I.J. Clarke, H. Quigg, S. Katsahambas, P.<br />

Juhola, M. de Blaslis and B. Doughton<br />

Tuesday 30 August<br />

Session 5 (Oral>: SP~RM AND THE MALE TRACT<br />

Chairman: Professor R.C. Jones<br />

Time: 0830 - 1000 Venue: Function Room 2<br />

Inh.ib.in .in t:he sheep ovar.ian cyc.le<br />

30 R.J. Rogers, S.J. Stuchbery and J.K. Findlay<br />

Lack of express.ion of .inh.ib.in qenes .in ov.ine corpora .lut:ea<br />

38<br />

Yulu Tang, <strong>the</strong> late D.E. Brooks, A.M. Snoswell and B.P.<br />

Setchell<br />

Ga.lact:osy.lt:ransferase act:.iv.it:y on sperm surface and .in<br />

ep.id.idyma.l p.lasma of severa.l ma.mma.lJ:an spec.ies<br />

31<br />

T. O'Shea, B.M. Bindon, J.K. Findlay, M.A. Hillard, L.R.<br />

Piper and K. Miyamoto<br />

Immun.iza t:.ion of ewes w.i t:h .inh.ib.in prepara t:.ions of<br />

.increas.inq pur.it:y<br />

32 R.A. Parr, I.F. Davis and M.L. Phillips<br />

Inf.luence of shear.inq st:ress on oest:rus and ovu.lat:.ion<br />

33 L.A. Fitzpatrick, T.J. Mullins and G. Fordyce<br />

Pred.ict:.inq fo.l.l.ic.le popu.lat:J:ons .in Bos .ind.icus cows from<br />

surface count:s of oVi1r.ies exam.ined aft:er ovar.iect:omy and by<br />

endoscopy<br />

34 J.D. O'Shea,. R.J. Rogers and M.J. D'Occhio<br />

Ce.l.lu.lar compos.it:.ion of t:he cyc.l.ica.l corpus .lut:eUJ11 of t:he<br />

cow<br />

39 De Yi Liu and H.W. Gordon Baker<br />

Corre.lat:.ion bet:ween spe.I7/1 charact:er.ist:.ics,<br />

hUJ11an .in v.it:ro fert:.i.l.izat:.ion<br />

40 De Yi Liu and H.W. Gordon Baker<br />

zona b.ind.inq and<br />

Corre.lat:.ion bet:ween human sperm morpho.loqy, acrosomes,<br />

acros.in and fert:.i.l.izat:.ion .in v.it:ro<br />

41 F.C. Molinia, P.A. Howe and M.A. Swan<br />

A t:echn.ique of s.imu.lt:aneous.ly est:.imat:.inq <strong>for</strong>ward ve.loc.it:y,<br />

beat: frequency, ,. Live and <strong>the</strong> nat:ure of f.laqe.l.lar<br />

wavefo.I7lls .in spe.I7/1at:ozoa<br />

42 A.J. Conway, L.M. Boylan, M. Wood and D.J. Handelsman<br />

Sem.inlJ.l p.lasma ret:.ino.l-b.ind.ing prot:ein as an .index of hUJ11an<br />

Sert:o.l.i ce.l.l funct:.ion<br />

35 E-M.A. Bugledich, L.A. Hinds and P.A. Janssens<br />

Seasona.l fact:ors .inf.luence t:he t:.im.inq of f.irst: preqnancy .in<br />

t:he t:ammar<br />

43<br />

\.<br />

D.A.Taggart and P.D. Temple-Smith<br />

Effect:s of mu.lt:.ip.le mat:J:ng on ep.id.idyma.l spe.I1ll d.ist:r.ibut:.ion<br />

.in t:he brow.n marsup.ia.l mouse, A. st:uart:.i.i<br />

36 M.B. Renfree, G. Shaw and T.P. Fletcher<br />

Superovu.lat:.ion ..in a macropod marsup.ia.l, Hacropus euqen.i.i<br />

37 M. Sathanandan, S.K. Walker, I.J. Clarke and C.D. Mat<strong>the</strong>ws<br />

The effect: of qonadot:roph.in re.leas.inq hO.I7/1one aqon.ist: on<br />

sheep p.it:u.it:ary and ovar.ian funct:.ion<br />

xi


Session 6 (Oral):<br />

Chairman: Dr C. O'Neill<br />

DEVELOPMENTAL BIOLOGY<br />

Session 8 (Poster):<br />

Chairman:<br />

Dr L.A. Hinds<br />

UTERUS AND PREGNANCY<br />

Time: 1030 - 1200 Venue: Function Room 2 Time: 1400 - 1530 Venue: Function Room 3<br />

44 J.M. Shaw and A.O. Trounson 50 T.P. Fletcher and D.R. Blandon<br />

U.ltrarap.:id freez.:ing of 2-ce.l.l mouse embryos<br />

45 L.J. Wilton and L. Diotallevi<br />

U.ltra-rap.:id cryopreservat.:ion of 8 ce.1.l mouse embryos w.:ith a<br />

punctured zona pe.l.luc.:ida<br />

46 G. ,Somers and L.J. wilton<br />

A.l.locat.:ion of ce.l.ls to .:inner ce.l.l mass and trophectoderm .:in<br />

mouse embryos b.:iops.:ied at <strong>the</strong> 4-ce.l.l stage<br />

47 J.P. Ryan, N.R. Spinks, C. O'Neill and R. G. Wales<br />

51<br />

52<br />

The ant.igest:agens RU486 and .3K2,g,g are not hound by <strong>the</strong><br />

uter.ine progesterone receptor of <strong>the</strong> tammar wa.l.laby"<br />

Hacropus euqen.:i.i<br />

R.A.<br />

Cherny and J.K. Findlay<br />

Prote.in secret.ion patterns of separated ov.ine endometr.ia.l<br />

ce.l.ls cu.lt::ured .in dua.l env.ironment: chambers<br />

N.R. Spinks and C. O'Neill<br />

The effects of a spec.if.icp.late.let act.ivat.ing factor (PAF)<br />

recept:or ant:agon.:ist (SRI 63441) on some aspects of materna.l<br />

phys.io.logy<br />

Illlp'.lantat.:ion potent.:ia.l and foet:a.l v.:iab.:i.l.:ity of embryos<br />

cu.ltured .:in <strong>the</strong> presence of p.lat:e.1et actJ:vat.ing factor<br />

53<br />

R.J. Fairclough, T.M. Lau, L.G. Moore, A.J. Peterson and<br />

W.B. Watkins<br />

48<br />

49<br />

D. Sakkas, P. Batt, A. Cameron and A.O. Trounson<br />

In v.itro deve.lopment of goat: pre.:imp.lantat.:ion embryos .:in<br />

cocu.lture w.:ith ov.iduct ep.:i<strong>the</strong>.l.ia.l ce.l.ls<br />

S.K. Walker, R.F. Seamark, P. Quinn, G.M. Warnes, R.J.<br />

Ashman, D.H. Smith and P. Ancell<br />

Pregnancy .inh.ibits oestradJ.·o.l .:induced re.lease of uter.:ine<br />

prostag.1and.in Fpand oxytoc.in-neurophys.in .:in <strong>the</strong> ewe<br />

54 B. Williams and R.N. Murdoch<br />

Resp.:irato.ry propert:J.·es of <strong>the</strong> mouse ut:erJ.·ne endomet:r.:ium<br />

durJ.·ng ear.ly post:-J.mp.lant:at:.lon pregnancy<br />

V.:iab.:i.l.ity of pronuc.lear embryos of sheep after cu.lture .in<br />

v.itro <strong>for</strong> one" three or fJ:ve days 55 C.S. Pow and L. Martin<br />

DJ.·str.ihutJ.·on of oestrogen receptors J.·n <strong>the</strong> fe.ma.le<br />

reproduct.ive t:ract of <strong>the</strong> f.lyJ.·ng fox Pt:eropus scapu.latus<br />

Session 7:<br />

Chairman:<br />

JAMES GODING MEMORIAL LECTURE<br />

Time: 1200 - 1300 Venue: Concert Hall<br />

Professor B. P. Setchell<br />

56 P.A. Towers and L. Martin<br />

Effect:s of gonadotrophJ.·ns on fo.l.l.ic.le deve.lopment dur.:ing<br />

pregnancy J.·n t:he f.ly.ing fox Pteropus scapu.latus<br />

Professor F.TY. Dazer: ESTABLISHNENT OF PREGNANCY IN SHEEP AND<br />

PIGS<br />

57 R.G. Alders and J.N. Shelton<br />

Ov.ine uter.ine .lymphat.ics: J.·n v.ivo passage of IndJ.·a J.·nk<br />

from ut:er.:ine subserosa" myomet:rJ.·u.m and .lumen<br />

xii


58 F. Abdi and I. Pollard<br />

The effect of pregnancy on <strong>the</strong> rate of <strong>the</strong><br />

b.iot:ransfo.r.mat.ion of caffe.ine<br />

Wednesday 31 August<br />

Session 11


69<br />

R. Vishwanath, I.G. White,P.D. Brown-Woodman and J.R.<br />

Mercer<br />

Ant.ifert.i.l.ity act"iv1."ty of gossypo.l .in ma.le rats and<br />

manganese status of d.iet<br />

78<br />

.M.J. D'Occhio and D.R. Gif<strong>for</strong>d<br />

P.itu.itary and ovar.ian responses of post-partum acyc.l.ic beef<br />

cows to cont.inuous .lonq-t:entl treatment w.ith GnRH' and a GnRH<br />

aqon.ist<br />

70<br />

J.L. Zupp and B.P. Setchell<br />

79<br />

W.M.C. Maxwell, S.K. Walker, D.H.<br />

Smith and H.R. Wilson<br />

Pro.lonqed effect of subfert.i.le ma.les .in reduc.inq numbers of<br />

fetuses 1."n no.rma.l fema.le rats<br />

Effect of GnRH' on fert.i.l.ityof Ker.ino ewes .insem.inated w.ith<br />

frozen semen<br />

71<br />

B.C. Jefferies, B.P. Setchell and W.M.C. Maxwell<br />

Fert.i.l.ity of ewes mated to Ner.ino rams from ·d.ifferent<br />

sources<br />

72 J.L. Reeve<br />

Durat.ion of proqestaqen pr.im.inq on <strong>the</strong> response of Border<br />

Le.icester X Jlfer.ino ewes to <strong>the</strong> 'Ram Effect /'<br />

73 S.R.D. Su<strong>the</strong>rland and K.W. Entwistle<br />

80<br />

D.O.<br />

D.H.<br />

Kleemann, S.K. Walker, J.R.W. Walkley, R.W. Ponzoni,<br />

Smith and R.F. Seamark<br />

Factors .inf.luenc.ing .lamb surv1."va.l 1."n a h.iqh fecundJ."ty<br />

Booroo.la X South Austra.l.ian· Ner.ino f.lock<br />

81 A.W.N. Cameron and P.A. Batt<br />

PNSG may d1."rect.ly st1.Jnu.late ova.lat.ion .in goats<br />

82 B.M. Bindon, J.K. Findlay, L.R. Piper and M.A. Hillard<br />

EffJ."cacy of me.laton.in .imp.lants (Regu.lJ."n)<br />

cows<br />

.in Bos .ind.icus<br />

Ova.lato.ry potency' of ASRB-bFSH-1 .in sheep and catt.le<br />

74 C.R. Earl, R.H. Male and E.A. Dunstan<br />

Synerg.ist.ic effects of me.laton.in and .immun.isat.ion aga.inst<br />

androstenedJ."one .in ma.iden BIt x JIf ewes<br />

75 K.P. Croker, M.A. Johns and L.D. Staples<br />

Session 13 (Poster): MALE 'R.EPRODUCTIVE FUNCTION<br />

Chairman: Dr E.J. Keogh<br />

Time: 1030 - 1230 Venue: Function Room 3<br />

Use of Requ.l.in me.laton.in .imp.lant:s 1."n conjunct1."on w.ith<br />

teas.inq of Jlfer.ino ewe f.locks jo.ined .in spr.ing<br />

76 P.J. Wright, A.H. Williams and LJ. Clarke<br />

Phe effects of nutr.ient restr.ict.ion and of .lamb remova.l on<br />

ovar.ian cyc.l.ic.ity and on <strong>the</strong> .inh.ib1."tory effects of<br />

oestrad.io.l on p.lasma concentrat.ions of LH and FSH 1."n postpartum<br />

ewes<br />

83<br />

84<br />

S. Wibullaksanakul and D.J. Handelsman<br />

Gonadotroph.in-re.leas.ing hOntlone re.lease from med.io-basa.l<br />

hypotha.lamus .in vIt:ro~ .va.lJ."dat:.ion of met:.'Jodo.loqy<br />

D.J. Handelsman and R. Lazarus<br />

Kat:hemat:.ica.l mode.l and camput:er s1.Jnu.lat:.ion of pu.lsat:.i.le<br />

hOI11lonesecret:.ion w.it:h app.l.icat:.ion t:o LH<br />

77 G. Fordyce, T.J. Mullins, C.A.J. Ridd and K.W. Entwistle<br />

Phe ro.le of <strong>the</strong> p.itu.itary .in post-partum nutr1."t.iona.l<br />

anoestrus .in cows<br />

85 S.S. Raychoudhury, M.G.· Irving and A.W. Blackshaw<br />

Phe effect:s of Serto.l.i ce.l.l matr1x/ fet:a.l ca.lf serum and<br />

Serto.l1." ce.l.ls on JH-t:hym.id.ine 1."ncorporat:ed .int:o DNA of<br />

cu.lt:ured myo.i-d ce.l.ls<br />

xvi<br />

xvii


86 G.F. Gonzales, J. Muir, G.P. Risbridger, and D.M. de'Xretser<br />

Effects of seroton.in on .inh.ib,in and testosterone product.ion<br />

by'adu1t rat' sem.in.iferous tubu1es and, Leyd.ig ce11s .in v.itro<br />

94 L.K.-P. Leung and J.M. Cummins<br />

Norpho1ogy of .immature speDTIatozoa of <strong>the</strong> Ch.inese pango.l.in<br />

(Nan.i$ pentadacty.la : Phol.idota)<br />

87 J.R. Ford, I.W. Purvis and G.B. Martin<br />

88<br />

.Effect of <strong>the</strong> Booroo1a F gene on p1asma FSH .in ent.ire,<br />

c.ryptorch.id and castrate riflJl/1ambs<br />

R.T. Gemmell and C. Sernia<br />

Immunocytoche.m.ica1 1ocat.ion of oxytoc.in and mesotoc.in<br />

W'.ith.in <strong>the</strong> hypotha1iH11us and reproduct.ive tract of <strong>the</strong> ma1e<br />

marsup.ia1 badd.icoot<br />

95.<br />

96<br />

M.. Lin, R.C.Jones and A.W. Blackshaw<br />

phe cyc1e of <strong>the</strong> sem.in.iferous ep.i<strong>the</strong>l.ium of <strong>the</strong> Japanese<br />

qua.i1, Coturn.ix coturn.ix<br />

R. Vishwanath, I.G. White and S.A. Matlin<br />

89 C.R. Earl, E.A. Dunstan and M. Schleuniger<br />

Phe effect of day 1ength treatment on <strong>the</strong> reproduct.ive<br />

perfoDTIance of Border .Le.:lcester rams 97<br />

Effect of racem.ic gossypol and .its .isomers on surv.iva.l of<br />

riH11 speDTI and on react.ivat.ion of speDTI models<br />

5. Sujarit, G. Chaturapanich, M. Lin, R.C. Jones, B.P.<br />

Setchell and G.M. Stone<br />

Does rete test.isf.lu.id conta.in a<br />

secretagogue<br />

98 S. Sujarit, R.C. Jones, M. Lin, B.P. Setchell and G.M.<br />

Stone<br />

Session 14 (Poster): SPERM AND THE MALE TRACT<br />

Chairman: Dr H.G. Baker<br />

Time: 1300 - 1400 Venue: Function ROOm 3<br />

90 D.P. Windsor and I.G. White<br />

Regulat.J:on of <strong>the</strong> .J."n.it.ia.l segment of <strong>the</strong> ep.id.idym.is<br />

99 S.J. Gatie, A.W. Blackshaw and T.D. Glover<br />

Phe effects of b.ilatera1 castrat.ion and ethy.lene d.imethane<br />

su1phonate (EDS) on ep.id.idyDlt!l1 funct.J.'on (speDTIatozoal<br />

maturat.ion) .in gu.inea-p.ig<br />

91<br />

Effect of phosphat.idy1ser.ine on ca1c.ium uptake of co1d<br />

shocked boar and riH11 speDTIatozoa<br />

P.A. Howe and M.A.<br />

Swan<br />

100 J. Clulow and R.C. Jones<br />

F1u.id and so.lute f1uxes .in <strong>the</strong> gen.ita.l ducts of <strong>the</strong> ma1e<br />

Japanese qua.i1<br />

92<br />

Protect.ive effect of phosphat.idy1cho1.ine aga.inst co1d shock<br />

.in goat sper:t1/iltozoa<br />

H.N. Jabbour, G. Evans and N.W. Moore<br />

101<br />

G. Chaturapanich and R.C. Jones<br />

Or.ig.in of 1um.ina.l prote.J.'ns .in <strong>the</strong> ep.id.idym.is of <strong>the</strong> tammar,<br />

Nacropus euqen.i.i<br />

Fert.i1.isat.ion .in superovu1ated ,ewesfo11oW'.ing .lnsem.inat.ion<br />

W'.ith d.ifferent doses of fresh or frozen-thawed semen<br />

93 G.M.. O'Bri~n, J. Clulow and R.C. Jones<br />

Separat.ion of human speDTI <strong>for</strong> e1ementa1 ana1ys.is<br />

102<br />

P.O. Temple-Smith and G.J. Southwick<br />

Problems .in screen.ing azoospeDTI.ic pat.J.'ents <strong>for</strong> correct.ive<br />

m.icrosurge.z::y<br />

xix


Session 15


Crane, L.R. . 63<br />

Creed, K.E 23<br />

Crocker, K. P. . .........•....•.....••...•..•..••..••.........•• 75<br />

Cummins, J .M..............•..•....•••.•....•..•.•.....•...•••• 94<br />

Cummins, J.T•...••..•...•.......•.•••.•.•...•..........••.••. 104<br />

Czapala-Peeters, C. . ....••••..•.•.•..•.....'. . . . . . . . . • . • . . . • . .. 66<br />

D'Occhio, M.J. . .•.......•...•.••.••••.•..........•..•.•.•.. 34,78<br />

Davis, I.F•.•.............•..•....•...••........••............ 32<br />

de Blasiis, M. ......•.....•..•••..•......•.••....••.•.....•.•• 29<br />

de Kretser, D. M. ...•.••....•..•...••••...•..•...•••...•.••• 19, 86<br />

de Luca, E.......•.....•..•.............•.•.•.••........ ,••..... 7<br />

Diotallevi, L. . '................................... 45<br />

Dong, Q••••••••••••••••••••••••••••••••••••••••••••••••••••••• 18<br />

Doughton, B. . .....................•.................•.......•. 29<br />

Drake, .B .L. ..•..•..•...••...........•..•..•..•.......•.....••. 64<br />

Drummond, A. E. . ...........•........•.............•..........,.. 19<br />

Dunstan, E.A•..........................•.•........•.••....• ,74.,89<br />

Earl, C.R 74,89<br />

Entwistle, K. W. • 73,77<br />

Evans, G•........................•.•.........•...•.....•...... 92<br />

Evans, J .R. . ............•..... '. . . . . . . . . • . • . . . . . . . . . . . . . . . . . . .. 67<br />

Fairclough, R.J. ........................................•. 53,108<br />

Farnworth, P .G. . ...................•...•.................. 26,103<br />

Findlay, J.K......................•.......• 27,29,30,31,51,82,103<br />

Fitzpatrick, L.A...................•....•.......•............. 33<br />

Fletcher, T.P....................•••..••.•.............. 36,50,59<br />

Ford, J .R. . ..........•.....•..•........•...................•.. 87<br />

Fordyce, G. . ...•.........•........•.•.•..•..........•....'.. 33, 77<br />

Fry, R.C•........'........•.........••..... ' 106<br />

Galloway, D.B......................••..•.....•........•...•... 22<br />

Garcia, J. . .....................•..••....•...••......•.•...•.• 62<br />

Gatie" S. J • •..•••........••.••..•.••..••.•.•....•.•.•••••.•••• 99<br />

Gemmell, R.T. •.............•.....•.•..•.................... 62,88<br />

Gif<strong>for</strong>d, D.R....•.....•'....•'..• '..•....•..•.•...•••........••.••. 78<br />

Glover, T.D......•.........• '•......•.•.•.•..••...•..••..•.•... 99<br />

Gogolin-Ewens, K...........•............•.•............•••.•.• 60<br />

Gonzales, G.F. • ...•.....•...•.......••..•••..•.•.......•.....• 86<br />

Hall, p.F.....•.........•.......•...•••.•.•....•....••.......• 20<br />

Hamilton, P.A.............•.•.•..••..•....•........••.........• 1<br />

Handelsman, D.J. . ......•........••..••••...••...•. 18,20,42,83,84<br />

Harvey, M.B...........•.•.•...•....•••..............•..•....•• 11<br />

Hearn, J.P. • •.•.....•..•.....••.•.•.•••..•......•..••........• 13<br />

Henniawati .•....................•......•.........•........... 107<br />

Herr, C. . .....••..........••.••.•...•••...•.•......••...•....• 10<br />

Herrington, A. C • . .....•.•..•.....•..•••.•••.....•........•.,...' 27<br />

Hillard, M.A. . .•...., ,. 28,31,82<br />

Hinds, L.A. ••...........•.....•.•.•.•..•...•........••..... 35,59<br />

Hodgen, G.D..•.....•......•.•.•.....••..•..•.........•.•.•. 14,65<br />

Holt, N. • •••.•••••,••.••••••••••••••••••••••••••••••••.•••.••••• 10<br />

Howe, P.A.•.........................•..••..•.........•..... 41,91<br />

Howse, A.M. .. ........•.....•...•...•.................••....... 106<br />

Hyland, J .R. . ...........•••.•.......•...•..•..............•.... 8<br />

Irving, M.G...•........................•...................... 85<br />

Jabour, R. N• ••.••..•....•...•••..••.••..•..•••...•....••..•••. 92<br />

Jackson, R.B••...............................................•. 3<br />

Janssens, P.A 35<br />

Jefferies, B. C. . •..........•......•......•......•............. 71<br />

Johns, M.A. . ........•...............................•.....•... 75<br />

Johnson, D.L....................••.........'..•..•......•....•.. 2<br />

Johnson, K.L.....•.....•......•.....•.•.•.......•...•.'•...•.• 106<br />

Jones, R.C...•.........•.....•..•...•........... 93,95,97,100,101<br />

Juhola, P. . ..••.•..•••...•......•.....•.•.......•.•...•...•.•• ' 29<br />

Katsahambas, S. .....••............••.•.....•.•..•...........•. 29<br />

Kaye, P.L.......•.•,.•.........••................•.....•..... 6,11<br />

Keogh, E.J. . ••................•.....•....•...•........•... ~ . •. 23<br />

Kidston, E•..........••...•..........•...•.................... 20<br />

Killeen" I.D.••..•.........••......••....•............•..•..... 1<br />

King" N.J. C. ....•...............•...••..•.........•...•....... 64<br />

Kleeman, D.O....................•...•.......................•. 80<br />

Lam, S-Y. • •••....•.•.•........•..••.•...••••.••.•••••.•.•.•... 67<br />

Lanzendorf, S. . . • . . . • . • • . • • . • . • • . • . . • . • . . . • • . . . . . . . . . . . • . . . . ... 14<br />

Lau, T.M.•.•........................•...•••...........•....... 53<br />

xxii<br />

xxiii


Lazarus, R. • ..•..•.....•..••••....•••.••••..•...•.•.•.•.•.•••'. 84<br />

Lee, J. • ..••••..•....•......••...•.•••.••..••.•.••...•.••.••••• 65<br />

Lee, C.S.. . . 60<br />

Leung, L.K•- P • . .•.........•.....•••.•••..••..••......•...•...• 94<br />

Lewis, P.R. • .•.•.••.•....•...••.••.•...•••.•••...•...•••••••.• 25<br />

Lin, M. •...•..••....••.....•.••.....••••..••.•••.••..••• 95, 97 , 98<br />

39,40<br />

Liu, D.Y•••••••••••••••••••••••••••••••••••••••••••••••••••<br />

Lopata, A. .••.•....•.•...••....•..•.•.....••••.•.•..••.... 5, 7 , 13<br />

Male, R.H. . •....•....•.•...•••.••••..••••.•..•.••..•..•.•..... 74<br />

Martin, G.B. . ...•......•...•..•.•.••••..••••••••...•••.•.•..•. 87<br />

. L ••.••.•••..••••••••••••••••••••••••.••••••.••• 55,56,63<br />

Mart~n, •<br />

Matlin, S .A. . ••.•..•.........•....••...••........•..•.....•••• 96<br />

Mat<strong>the</strong>ws, C. D. . •.............•..•.•••••.•..••....•..........•. 37<br />

Maxwell, W.M.C. . .•.............••.••....•.•.....••.•.•.•.•• 71,79<br />

Maxwell, L.E......................••..•.•.•.•.••••••........•• 64<br />

McGowan, L.T•......•..............•.•.•.•...•...•...•.•...•.... 2<br />

McKay, S.A....•..................•.•.••......•..••....•.•.••••. 5<br />

Mercer, J.E..............•.....•...•..•.......•.•...•.•...••. 105<br />

Mercer, J .R. . ...........•........•.•..•..................•..•. 69<br />

Mercer, W.R. . ...........................• 60<br />

~ .•............•.•...<br />

Miyamoto, K. . ...........•.•... 28,31<br />

~ ....•................•......<br />

Molinia, F.C. . .............................•....•.....•....... 41<br />

Moore, L.G. . ....•........•...•...•..•....•.•........•••..••.•. 53<br />

92<br />

Moore, N.W. . '.........•....••..••...••.....•••••<br />

Muir, J .••••.......•..........•.....•...•......•....••..••.... 86<br />

Mullins, T.J.................•...••...•.••..•.•....•.....•. 33,77<br />

Murdoch, 'R.N...............•.......•....•••.•.•...•..•....•..• 54<br />

Murray, N.•..••...........•...•..•.•..••.•...•......••...•.••.• 3<br />

Nancarrow, C. D. . ...•......•.•....•..•..•.•..•......••.••.•...• 12<br />

Ne<strong>the</strong>ry, R.D........•...•...•.......•....••.•.•.••.•.....••.•. 28<br />

Nunan, K. • •.•.•.....•....•.••..•....•....•......••.••..••••'... 6 2<br />

0, W-S ...•....•..•.•............•...•.......•........••.•••...<br />

O'Brien, G.M.<br />

. ••........•....•....•....••..•......•..•..•..•.•<br />

O'May, P.J•...... ,; ..•.........•.••....•..•..•.....•.•........••<br />

O'Neill, C....•...••.....•.......•••................•••••.• 47,52<br />

O'Shea, J.D• ..•.............•...•.••••••.•..•..••...•..••.•..• 34<br />

68<br />

93<br />

3<br />

O'Shea, T. ...•........•......••••.•.•.....••.......•....•.. 28, 31<br />

Oppenheim, R.C........•.••..••..•.....•.••........•........•.. 22<br />

Parr, R.A. . .•....••.•••........••••••....•••...•..•........ 32,62<br />

pepperell, R.J....•...•.•...........•......................... 67<br />

Peterson, A.J. . ....••...•.•.•.....•.••....•••....•..•••.••..•. ·53<br />

Phillips, D.J. ..•......••.....••....••.••.......•.•..•....... 104<br />

Phillips, M.L•........•....••.•....••......•.•.•.•...•..•..... 32<br />

Piper, L.R. . ..•••.•..•..••......•..••...•.••..•...•..••. 28,31,82<br />

pollanen, P. •..••.•...•.•.••......•••...•••.....••.•..••...••• 21<br />

pollard, r. . 58<br />

Ponzoni, R.W•.........•..•..•.............••......••.......••• 80<br />

Pow, C.S....•..•....•.•......•....•.••....•..•....•......••.•• 55<br />

Purvis, r.w 87<br />

Quigg, H. . •..........••.•..••......••.....•.......•...••...•.. 29<br />

Quinn, P. . ..•......•....•....•.•....•...•.•.............••...• 49<br />

Raychoudhury, S. S. ......•........•......•.••..........••...... 85<br />

Raymond, S. . .........•.........•........•..................•.. 66<br />

Reed, K. C • ......•............•••..........•..................• 10<br />

Reeve, J. ...............•............•..•........•.....•.... 1 , 72<br />

Renfree, M.B.....•.....................•••........... 25,36,59,68<br />

Restall, B.J..................•.....•...•......•.........•.•. 107<br />

Ridd, C.A.J.....•..•.........•................................ 77<br />

Risbridger, G.P..•..•..........•..•........................ 19.,86<br />

Ritar, A.J. ...........•........•....•..••.............•...•..•• 3<br />

Roberts, T.K..•...•......•.•.......•.............•....•..•. 15,16<br />

Robertson, D.M.•.. ~ ...•.....•..........•...••......•.....••... 20<br />

Rodger, J. C. ............•...•.....••..........•.......•...•.•• 64<br />

Rogers, R.J••.....·......•..........•.................•..... 30,34<br />

Ryan, J. P • ..••....••....•.......•..•.............•...••....•.• 47<br />

Sakkas, D. ............•............•.........•............. 17, 48<br />

Sathanandan, M. .............•.....•....•.............•.....••• 37<br />

Saunders, D.M..•...................•..............•........... 14<br />

Scaramuzzi, R.J.......•.•...•••....•.........•.•.•....••..•.. 107<br />

Schleuniger, M. . .....••..•.....•......................•.•.•... 89<br />

Seamark, R.F..•......•.................••.....•.•.•..•...... 49,80<br />

Sernia, C••.•..............•......••.•..•.........••.•..... 62,88<br />

xxiv<br />

xxv


Setchell, B.P.•..•........•.•••...••••••.....•. 21,38,70,71,97,98<br />

Shaw, G.•.......•..•.•....•...•.•..•.••••.•..........••.. 36,59,68<br />

Shaw, J .M. . ..••................•.•.••••••..........••••.•..•.• 44<br />

Shelton, J.N. •...........•.•••..•..••..•....•......•...•••. .9,57<br />

Short, R.V. •.....•...•.•....••...•..••••••.•.•....•••..•••• 25,68<br />

Sinosich, M.J. . ......•..•...••••.•..•••••.•....•... ~ .•.•••. 14,65<br />

Smart, Y.C..•..•..........••.....•.•.•.•••.••.....•••... 15,16,66<br />

Smith, D. H. ....•..•.•....•..•••••.••••••••.••.....•.•... 490' 79 , 80<br />

Smith, J .K. • •...•.•..•••..••.•.••....•.••.••..•.••.....•.•.••.• 1<br />

Snoswell; A.M. . .•..•.......••••••.•••.••••.••.•......••••..••• 38<br />

Somers, G.....................•............•.•••......•..••••. 46<br />

Southwick, G.J. • ..............•........•.•...••..•......••.... 102<br />

Spaliviero, J .A. . .........•.••...•.•.•...............•.....••• 20<br />

Spinks, N. R. •...•..........•......•...••..........•.•..•... 47,52<br />

Stanger, J. D. ..............•.....•...................•..•.•.•• 16<br />

Staples, L.D. . ...•..................•.........•.....•••..•.••• 75<br />

Stone, G.M...........•............•.•....•..........•...... 97,98<br />

Stuchbery, S.J. . ..•........••.......•.........•.............•• 30<br />

Sujarit, S........................•.•.••.......•..........• 97,98<br />

Summers, P.M. . ............•...............•..........•.......• 13<br />

Su<strong>the</strong>rland, S.R.D...........• ~ .......•....•................... 73<br />

Swan, M.A. . .•....•..................•........•......••..... ·41,91<br />

Szell, A.............•...............•.......•..............••. 9<br />

Taggart, D.A. . ..•..................•...••......•..•...•••..••. 43<br />

Tang, Y.•........................•...•.......••.....•......••. 38<br />

Temple-Smith, P.D.•..••.•••..••.•.••••.•••.••.•.•••••.••.• 43,102<br />

Thiel, W.J. . ............•.•.......••..•••.••..••.....•..•••••• 22<br />

Thomas, W.G. . .•.........•..••......••..•.•..•..........•...•.• 62<br />

Thompson, J .G.E...................•••....•...•..•...... ,; .•.•..• 4<br />

Tilbrook, A.J...•......•...•.......••.•..•................•.•. 22<br />

Towers, P.A......•...............•.......•...•....•....•••.•.• 56<br />

Trounson, A.a••......•.....•.......•...•.••....•••.• 17,44,48,108<br />

Tyndale-Biscoe, C.H......•..•.......•••.••.•.....•••..•....•.• 59<br />

Uphill, G.....••..... ~ 28<br />

Vishwanath, R. •......•.••........••.....•...........•.•...• 69,96<br />

Wales, R.G...........•....•...•..•.......•.....•••.•....•. '•... 47<br />

Walker, S .K. • .•.•.•..•.••.......••.....•.••.•.......•...• 37,79,80<br />

Walkley, J .R.W. . ....•...•..........•••.••........•..•......... 80<br />

Wang, Q•F • • .••...••.....••••,.............................. 26,103<br />

Warnes, G.M•.••.•..•.......•...•......•...•.........•......... 49<br />

Watkins, W.B.••...••....•••.•.....•.•.••.......•....•.•....... 53<br />

White, I.G. •.••.....••...•..•.....•.•.•.........•...•... 69, 90 , 96<br />

Wibullaksanakul, S. ..•........•.............•...•...•......•.. 83<br />

Wilcox, R.A. . ••........•...••.•.•....•..•...•..•....•.......•. 15<br />

Williams, R.F•.•....•..•............•..••.•.••.......•.•..••.. 65<br />

Williams, A.H......•.•........•.•..•..•.•....•..••......... 22,76<br />

Williams, B. •...•.......•.............•...•.....•...•.•....•.. 54<br />

Wilson, H.R. • •.....•.......•.....••.....•...•.........••...... 79<br />

Wilton, L.J·•...•••............•..••....•..............•.... 45,46<br />

Windsor, D.P....•.•.......•..'..............•....••..........•. 90<br />

Wolf, J.P•...•....•.....................•..••....•............. 65<br />

Wood, M•.•.....•...•.•.•.•............•.•......•.............. 42<br />

Wooding, P. . ••...•...•...........••.....•....•.. ,;............. 60<br />

Wright, P.J.....•..•...•.•............•..•••................•. 76<br />

Zhang, Z. . .•.•..................•........•..................•. 27<br />

Zupp, J .L...•..........•.........•...•...............•.••..•.. 70<br />

xxv;<br />

xxvii


,;;:as<br />

EFFECTS OF METHOD OF FREEZING RAM SEMEN AND DOSE OF PMSG<br />

ON FERTILITY OF EWES INSEMINATED INTO THE UTERUS.<br />

P.A. HAMILTON1, 1.0. Killeen2 and J. Reeve3<br />

1. Elders Breeding Services, Tongala, Vic.<br />

2. Heriot Agvet, 9 Edina Road, Ferntree Gully, Vic. 3156.<br />

3. Agricultural Research Institute, Ru<strong>the</strong>rglen,Vic.<br />

The effect on fertility of inseminating ewes with semen frozen in straws or<br />

pellets and of dose of PMSG was examined in 4 trials. The Border Leicester X<br />

Merino (Trials I, II and IV and Merino Trial III) ewes were treated with<br />

progestagen intravaginal sponges (Repromap, Upjohn), PMSG (Pregnecol, Heriot<br />

Agvet or Folligon, Intervet) at sponge withdrawal, <strong>the</strong>n 56-64 h later inseminated<br />

intra-uterine, using glass pipettes and a laparoscope.<br />

For each trial semen was collected from three rams and pooled be<strong>for</strong>e frezzing<br />

in pellets or 0.25ml straws(l). A tris based diluent was used <strong>for</strong> <strong>the</strong> pellets(1)<br />

which contained 200xl06 motile spermatozoa in a final dilution of 1 in 20. TThe<br />

diluent <strong>for</strong> straw freezing contained 8.5gm trisodium citrate, 6gm fructose, 44g<br />

skim .milk powder, 100mi egg yolk, 60ml glycerol made up to 1000ml with<br />

distilled water. Each straw contained 60xl0b motile spermatozoa in a final<br />

dilution of 1 in 20. Three ewes were inseminated with each pellet and one straw<br />

was used per ewe. Pregnancy rates were determined by real time ultrasound and<br />

presented in Table 1.<br />

TABLE 1 Effect of method of freezing and dose of PMSG on fertility of ewes.<br />

Main % pregnant (no. of ewes) Total<br />

effect Trial I Trial II Trial III Trial IV<br />

Freeze Straw 60(55) 47(53) 65(124) 60(106) 59(338)<br />

Method Pellet 78(54) 49(59) 59( 119) 59(113) 60(345)<br />

Dose 0 53(17) 53(19) 53(36) 30(37) 45(109)<br />

PMSG 200 64(22) 39(18) 58(72) 53(15 ) 56(127)<br />

i.u. 400 76(21) 39(23) 63(71 ) 61(69) 61 (127)<br />

600 62(53) 62(53)<br />

800 85(21) 76(21) 70(64) 73(15) 74(121)<br />

1200 71(14) 40(15) 75(16) 62(45)<br />

1600 57( 14) 38(16) 71(14) 57(44)<br />

The method of freezing <strong>the</strong> semen had no effect on fertility. Increasing <strong>the</strong> dose<br />

of PMSG, at least to 800 iou., increased fertility except in Trial II in which <strong>the</strong><br />

PMSG had been frozen and thawed after reconstitution, reducing potency.<br />

(1) Evans,G. and Maxwell, W.M.C. Salamon's Artificial Insemination of Sheep<br />

and Goats. Butterworth's, Sydney (1987).


2 3<br />

DIFFERENCE BETWEEN TWO GENETIC LINES OF ROMNEY EWES, SELECTED FOR<br />

INCREASED FECUNDITY, IN THEIR OVULATORY RESPONSE TO PMSG<br />

J.F. Smith, J.N. Clarke, D.L.<br />

Johnson and L.T. McGowan<br />

MAFTech North, Ruakura Agricultural Centre, P.B., Hamilton, N.Z.<br />

The ovarian response to exogenous gonodotrophin can provide a<br />

useful indirect measure of <strong>the</strong> endocrine differences between genetic<br />

groups (I, 2). This technique was used as part of a study into <strong>the</strong><br />

endocrinological and physiological basis <strong>for</strong> differences in fecundity<br />

of two flocks of Romney sheep selected <strong>for</strong> high and low incidence of<br />

multiple births (3,4). Ewes were allocated to 4 groups on <strong>the</strong> basis of<br />

flock, ~election line, age and liveweight. All ewes were treated with<br />

CIDRs <strong>for</strong> 14 d and joined with vasectomised rams. Pre-treatment<br />

ovulation rate was recorded by laparoscopy 7 d later, 16 dafter CIDR<br />

removal ewes received ei<strong>the</strong>r 0, 375, 750, or 1500 iu PMSG (Folligon®<br />

Intervet International B.V. Boxmeer-Holland; Batch 376551 and 7 d later<br />

post-treatment ovulation rate was recorded. The pre-treatment<br />

ovulation rates <strong>for</strong> <strong>the</strong> Ruakura Fertility flock were high = 1.47±0.06,<br />

control = 1.02±0.04, and low = 1.03±0.04, while <strong>for</strong> <strong>the</strong> Hight flock<br />

<strong>the</strong>y were high = 2.04±0.07, and control = 1.64±0.05. The posttreatment<br />

ovulation rates are presented in table 1.<br />

Ovulation rate after PMSG<br />

(mean±s. e .m.)<br />

Flock Line n Dose of PMSG (iu)<br />

0 375 750 1500<br />

Ruakura High 22 1.79±0.12 2.00±0.17 2.09±0.14 3.l9±O.23<br />

Control 19 1. 32±0.13 1. 21±0. 09 1. 7l±0 .18 2.20±0.35<br />

Low 17 1.06±0.06 1. 33±0.11 1.63±0.23 1. 94±0 .46<br />

Hight High 25 2.04±0.12 2.16±0. 11 3.28±0.37 5.48±0.87<br />

Control 31 1. 48±0. 09 1.71±0.O9 2.47±0.22 2.97±O.33<br />

The response of <strong>the</strong> Hight high line differed from that of <strong>the</strong><br />

o<strong>the</strong>r lines by having a greater slope which suggests a higher ovarian<br />

sensitivity to gonodotrophin. This corroborates a similar suggestion<br />

(5) based on data from prepuberal lambs from this flock.<br />

(1) Bindon, B.M. , Chang, T.S. , Turner, H.N. (1971) . A.J.A.R. 11.:809.<br />

(2) Smith, J.F. (1976). Proc.. NZ Soc. Anim. Prod. 1§.:247.<br />

(3 ) Clarke, J.N. (1972). Proc. NZ Soc. Anim. Prod. 32:99.<br />

(4) Hight, G.K. (1969). Ann. Rep. Res. Div. MAF 74.<br />

(5) Bodin, L. et al (1986). Proc. Endoc. Soc. Aust. 29: Supl 2. E9.<br />

SUPEROVULATION RESPONSE AND EMBRYO RECOVERY FROM CASHMERE<br />

AND ANGORA DOES AFTER TREATMENT WITH FSH (FOLLTROPIN)<br />

A.J. Ritar, P.D. Ball, P.J. O'May, T.M. Black,<br />

R.B. Jackson and N. Murray<br />

Department of Agriculture, Launceston South, Tasmania<br />

superovulation in goats may be induced by numerous regimes of<br />

gonadotro~hin adminis~ration. We examined <strong>the</strong> ovulation dose-response<br />

of goats ~n <strong>the</strong> breed~ng season to a highly purified preparation of<br />

FSH containing very low levels of LH contamination.<br />

Mature Cashmere and Angora females (mean live weight + sem 37.3 + 0.72<br />

and 33.4 ± 1.80kg) were continuously fed a high-energy diet (450g oats,<br />

1100g hay; 13.1 MJ ME/day) and had CIDRs (AHI Plastic Moulding Co, NZ)<br />

removed on <strong>the</strong> afternoon of <strong>the</strong> 18th day after insertion. FSH<br />

(Folltropin, donated by Biocene Int Pty Ltd, Melb) was administered<br />

7r~m 5h be 0re CIDR removal and continued twice-daily <strong>for</strong> seven<br />

7<br />

~nJect~ons ~n a decreasing regime (20, 20, 15, 15, 10, 10 and 10% of<br />

total dose) to give total FSH doses of 3, 6, 9 and 12 mg. Testosteronetreated<br />

we<strong>the</strong>rs maintained a continuing male presence. Does were in<br />

oestrus from 20-65h and were naturally mated at 39 and 47h after CIDR<br />

removal. Embryos were surgically collected from females under halothane<br />

anaes<strong>the</strong>sia 8 and 9 days after CIDR removal. Ovulation points (corpora<br />

luteal were counted at this time.<br />

Table 1.<br />

Total FSH<br />

Dose (mg)<br />

3<br />

6<br />

9<br />

12<br />

Totals & Means<br />

Superovulation rate of Cashmere and Angora does<br />

treated with Folltropin<br />

Ovulations/does treated (mean)<br />

Cashmere Angora Total<br />

25/4 ( 6.3)<br />

56/3 '(18.7)<br />

144/5 (28.8)<br />

118/4 (29.5)<br />

343/16(21.4)<br />

21/4 5.3)<br />

38/4 9.5)<br />

17/3 5.7)<br />

72/4 (18.0)<br />

148/15 ( 9.9)<br />

46/8 ( 5.8)<br />

94/7 (13.4)<br />

161/8 (20.1)<br />

190/8 (23.8)<br />

491/31(15.8)<br />

Results are presented in Table 1. Folltropin appeared to be an<br />

effective gonadotrophin <strong>for</strong> <strong>the</strong> induction of superovulation in goats.<br />

There was an overall linear response in <strong>the</strong> ovulation rate to an<br />

increase i~ <strong>the</strong> FSH dose (P


4<br />

SUPEROVULATION AND OVA RECOVERY IN FARMED FALLOW DEER (Dama dama)<br />

J.G.E. Thompson and G.W. Asher<br />

MAFTech North, Ruakura Agricultural Centre, P.B., Hamilton, N.Z.<br />

Fallow deer are highly seasonal breeders. Does are monovular and<br />

can exhibit regular oestrous cycles of 21-23 days duration from April<br />

to August on New Zealand farms (1).<br />

Superovulation of 36 mature fallow does was attempted in May 1987<br />

by gonadotrophin administration following intravaginal CIDR (12%<br />

progesterone, Type-S, ARI) insertion <strong>for</strong> 14 days. Three treatments<br />

were applied: (A) 1000 iu PMSG (Pregnecol, Heriot Agencies),<br />

administered as a single i.m. dose 48 hr be<strong>for</strong>e CIDR withdrawal; (B)<br />

20 mg FSH (Folltropin, Vetripharm) administered i.m. in a decreasing<br />

dose regimen, twice daily <strong>for</strong> 4 days, <strong>the</strong> last dose coinciding with<br />

CIDR withdrawal; (C) 750 iu PMSG + 14 mg FSH; PMSG administered as <strong>for</strong><br />

(A) and FSH administered as <strong>for</strong> (B). Immediately after CIDR withdrawal<br />

does were joined with crayon harnessed fertile bucks. Onset of oestrus<br />

was recorded by frequent observation over four days following CIDR<br />

withdrawal. Ova recovery (OR) was per<strong>for</strong>med after 6-8 days from CIDR<br />

removal by uterine flush under surgical conditions. Numbers of corpora<br />

1utea (CL) and total stimulation points (TS; including cystic and<br />

1uteinised follicles) were also recorded.<br />

Group n CL TS* O~ %REC %FEC<br />

A 12 9.2±2.5 16.8±2.0 a 3.7±1.l a 40.0 70.0<br />

B 12 6.3±2.9 7.0±3.1 b 1.1±0.5 b 17.7 84.6<br />

C 12 11.2±3.3 20.4±3.0 a 1.9±O.5a ,b 17.2 52.2<br />

* differing superscripts indicate significant differences (p


6<br />

IONIC REQUIREMENTS OF GLYCINE TRANSPORT<br />

IN MOUSE EMBRYOS<br />

Peter L. Kaye,<br />

Department of Physiology & Pharmacology, University of Queensland,<br />

St.Lucia, QLD 4067<br />

At <strong>the</strong> 2-cell stage mouse embryos possess a specific highly<br />

concentrative glycine uptake system which is kinetically dependent on<br />

2Na+ and resembles <strong>the</strong> gly-system of red cells. After development<br />

to <strong>the</strong> blastocyst stage glycine transport is less specific, with<br />

characteristics of o<strong>the</strong>r systems, however <strong>the</strong> kinetic dependence ,on<br />

2Na+ remains. It has been proposed that <strong>the</strong> energy <strong>for</strong> concentrat7 ve<br />

uptake ,was derived from <strong>the</strong> [Na+] gradien~, which presu~ably ~el~es<br />

on Na+/K+-ATPase activity. However <strong>the</strong>re ~s no such gra~~ent ~n 2­<br />

cell embryos and significant ATPase does not appear unt~l <strong>the</strong> morula<br />

stage. In order to fur<strong>the</strong>r characterise <strong>the</strong> ionic requirements, ~he<br />

role of ATPase and that of <strong>the</strong> Na+ ,H+-exchanger, we have stud~ed<br />

glycine uptake in embryos of both stages.<br />

Two-cell embryos and blastocysts were collected from<br />

superovulated Quackenbush mice 48 or 9~h respectivelY,after hCG.<br />

Uptake of 3H-glycine over 10 min was determ~ned. The requ~rement <strong>for</strong><br />

CI- was determined by replacement with S04= or acetate. Bo~h<br />

substitutions inhibited uptake by 90% in 2-cell embryos and 80% ~n<br />

blastocysts.The role of Na+ transport was invest~ga~e~ using ~he<br />

Na+ ,K+exchange blocker, amiloride and <strong>the</strong> ATPase ~nh~b~tor ouaba~n.<br />

Nei<strong>the</strong>r was effective on 2-cell embryos.<br />

The ionic requirements of uptake by 2-cell embryos were<br />

fur<strong>the</strong>r investigated by removing K+ from <strong>the</strong> medium an~ measuri~g<br />

uptake rate at increasing intervals. This was also done w~~h ouaba~n<br />

in <strong>the</strong> absence of K+, since it has been reported that K+ b~nds close<br />

to <strong>the</strong> ouabain binding site of Na+/K+-ATPase. Uptake was constant<br />

when assayed at intervals up to 3h of incubation; omission of K+ had<br />

little effect and ouabain reduced uptake by about 30%.<br />

These results show that during development <strong>the</strong> major neutral<br />

amino-acid transport system maintains its requirement <strong>for</strong> CI-. The<br />

gly-system in 2-cell embryos is only marginally dependent on,<strong>the</strong>,K+<br />

gradient, <strong>the</strong> Na+/K+-ATPase and <strong>the</strong> Na+ ,K+-e~changer to susta~n 7ts<br />

concentrative activity. This suggests that ~n <strong>the</strong>se embryos glyc~ne<br />

uptake is independent of <strong>the</strong> Na+ gradient as might have been<br />

predicted from <strong>the</strong> absence of such a gradient at this stage, and<br />

possibly Na+ flux••<br />

Funded by NHMRC.<br />

EFFECT OF AN INSULIN/TRANSFERRIN/SELENITE SUPPLEMENT ON DNA<br />

SYNTHESIS IN MOUSE EMBRYOS CULTURED IN VITRO.<br />

E. De Luca, A. Lopata<br />

University of Melbourne, Dept. Obstetrics and Gynaecology,<br />

Women's Hospital, 132 Grattan st., Carlton, Vic., 3053<br />

7<br />

Royal<br />

It is generally accepted that growth factors are important <strong>for</strong><br />

<strong>the</strong> development of mammalian cells in vitro. The aim of <strong>the</strong> present<br />

study was to determine whe<strong>the</strong>r insulin/transferrin/selenite (ITS)<br />

would influence cell proliferation of preimplantation mouse embryos<br />

in culture. This was determined by assaying <strong>for</strong> <strong>the</strong> incorporation<br />

of 3H-thymidine into DNA.<br />

Preliminary experiments were per<strong>for</strong>med to examine <strong>the</strong> optimal<br />

conditions <strong>for</strong> measuring 3H-thymidine incorporation into DNA. Two<br />

cell embryos were obtained from superovulated CBAxBalb/C (F1)<br />

female mice, 6-8 weeks old. The embryos were cultured to <strong>the</strong><br />

hatching blastocyst stage in serum-free minimum essential medium<br />

(~-MEM) and <strong>the</strong>n examined <strong>for</strong> DNA syn<strong>the</strong>sis. This was determined by<br />

incubati,ng <strong>the</strong> embryos in 5 J,JCi/ml 3H-thymidine (specific<br />

activity, 5 Ci/mmol) <strong>for</strong> 4 h under standard culture conditions.<br />

Embryos were transferred to a multi-well "Bio-Dot" apparatus<br />

(BIO-RAD) which contained a nitrocellulose sheet. The embryos were<br />

treated with or without 2% 80S and/or 0.3M/3M NaOH. After washing<br />

with 1M ammonium acetate <strong>the</strong> DNA bound to <strong>the</strong> nitrocellulose paper<br />

was determined by liquid scintillation counting. These experiments<br />

indicated that <strong>the</strong> optimum conditions <strong>for</strong> 3H-thymidine<br />

incorporation into DNA involved treatment with 2% SDS followed by<br />

a.3M NaOH. This method proved to be more sensitive than <strong>the</strong> TCA<br />

precipitation method (1, 2) used by us previously.<br />

Using this Bio-Dot method <strong>for</strong> <strong>the</strong> assay of 3H-thymidine<br />

incorporation into DNA a dose response curve <strong>for</strong> ITS was<br />

determined. Embryos were incubated with ~-MEM containing 0-10%<br />

ITS. At <strong>the</strong> hatching blastocyst stage <strong>the</strong>y were assayed <strong>for</strong><br />

3H-thymidine incorporation as described above. The results showed<br />

that at a concentration of 5% (this corresponds to 25~g/ml insulin,<br />

25~g/ml transferrin and 25ng/ml selenite), ITS stimulated<br />

3H-thymidine incorporation to a maximum of 184% of control(ie.<br />

291±46 cpm/embryo compared to 534±53 cpm!embryo, mean±S.E.M, n=4).<br />

Following <strong>the</strong> observation that ITS increased DNA syn<strong>the</strong>sis of<br />

hatching blastocysts, preliminary studies have indicated that <strong>the</strong><br />

individual components of ITS were ineffective and that <strong>the</strong> growth<br />

promoting effect required <strong>the</strong> presence of both insulin and<br />

transferrin. Future studies will examine <strong>the</strong> stage of embryonic<br />

development at which insulin and transferrin exert an influence on<br />

DNA syn<strong>the</strong>sis.<br />

(1) Fi scher, B.<br />

(2) De Luca, E.,<br />

(1987) J. Reprod. Fert., 79, 115-123<br />

Lopata, A. (1988) Proc. ANZSCB, I, 5


8<br />

ANALYSIS OF THE CONTRIBUTIONS OF THE EMBDEN-MEYERHOFF AND<br />

PENTOSE-PHOSPHATE PATHWAYS TO GLUCOSE METABOLISM BY SINGLE<br />

EQUINE EMBRYOS IN CULTURE<br />

I.Bruck and J.H.Hyland<br />

Department of Veterinary Clinical Sciences,<br />

University of Melbourne<br />

Werribee, Vic.<br />

The mare has <strong>the</strong> lowest reproductive efficiency among <strong>the</strong><br />

domestic animals. Seasonality, strict uniparity and a gestation<br />

period of 11 months account <strong>for</strong> part of <strong>the</strong> problem.<br />

Embryonic loss during <strong>the</strong> first 20 days of gestation (up to 45 %)<br />

fur<strong>the</strong>r contributes to poor reproductive per<strong>for</strong>mance.<br />

To study <strong>the</strong> reasons <strong>for</strong> <strong>the</strong> high incidence of early embryonic<br />

loss in <strong>the</strong> mare, our aim was to develop a culture system to<br />

assess embryo viability, based on measurements of metabolic<br />

activity. The contributions of <strong>the</strong> Embden-Meyerhoff (EMP) and <strong>the</strong><br />

Pentose-Phosphate Pathways (PPP) to <strong>the</strong> turnover of glucose<br />

were measured separately.<br />

Equine embryos were collected on Day 7 (n=6) or Day 9 (n=9) after<br />

ovulation from 10 Standardbred mares using a non-surgical<br />

transcervical flushing technique.<br />

After determination of diameter and visual quality each embryo<br />

underwent 2 x 3 h cultures in <strong>the</strong> inner well of a sealed two-well<br />

system at 37 C. The culture medium (60 or 120 ul) contained<br />

ei<strong>the</strong>r (U-14C)- or (S-3H)-glucose. l4C-labelled carbon dioxide<br />

(from activities of <strong>the</strong> PPP) or tritiated water (from activities<br />

of <strong>the</strong> EMP) released by <strong>the</strong> embryo were trapped in 1.0 mol<br />

sodium hydroxide in <strong>the</strong> outer well.<br />

Metabolism of radioactively-labelled glucose was considered to<br />

have occurred if <strong>the</strong> counts produced by <strong>the</strong> embryo were<br />

significantly greater than <strong>the</strong> mean counts of <strong>the</strong> controls<br />

(p


10<br />

EFFECT OF SUCROSE AND CALCIlM IN THE SPLITTING Iv1EDIlM ON<br />

SURVIVAL OF QUARTERED OVINE MJRULAE<br />

C. Herr,l N. Holt,2 an d K.C. Reed l<br />

lAdvanced Breeding Technology Pty. Limited<br />

2Riverina Artificial Breeders Pty. Limited<br />

Dissection of blastocyst stage embryos is more<br />

successful than dissection of morulae. In morulae, cells<br />

are tightly associated; when dissected (split) many cells<br />

are damaged. In contrast, blastocysts have a fluid filled<br />

coe I e so f ewe r c e I I s come inc0 n t act wit h <strong>the</strong> s p lit tin g<br />

instrument. Commercial application of embryo splitting<br />

technology requires that both morula and blastocyst stage<br />

embryos yield acceptable results, i.e. <strong>the</strong>re is a<br />

significant increase in <strong>the</strong> number of offspring produced<br />

from a given number of embryos. This study attempted to<br />

improve <strong>the</strong> survival rate of quartered morula stage ovine<br />

embryos.<br />

The effect of <strong>the</strong> presence or absence of 500 roM<br />

s u c r a s e (S) or 2. 2 1 mM c a I c i urn (Ca + + ) in <strong>the</strong> s p lit ting<br />

me d i urn was i n v est i gat e d . Em b r y a s we r e su r g i c a I I Y<br />

collected from <strong>the</strong> uterus of superovulated and<br />

I a para s cop i c a I I y art i f i cia I Iyins eminated ewe s • Mo r u I a e<br />

we reofun i <strong>for</strong>m qua lit Y, and werea r bit r a r i Iy a I I 0 cat e d<br />

<strong>the</strong> treatments. After microsurgical dissection, embryos<br />

were cultured in bicarbonated medium at 39 0 C under a 5%<br />

C02 atmosphere. Forty eight hours later, embryos were<br />

evaluated <strong>for</strong> development to <strong>the</strong> expanded blastocyst<br />

stage. Results are summarised in <strong>the</strong> Table 1.<br />

In vitro development of quartered embryos dissected under<br />

different media conditions.<br />

Trea tmen t<br />

Ca++ S<br />

+<br />

+ +<br />

+<br />

Total quarters (Unsplit) Percentage (Unsplit)<br />

per treatment (control) Developing (control)<br />

40<br />

24<br />

40<br />

24<br />

6<br />

2<br />

3<br />

2<br />

2.5<br />

87.5<br />

42.5<br />

0.0<br />

100<br />

100<br />

67<br />

100<br />

Mo r u I a e s p lit wit h out C a + + ten dedt a 10 s e<br />

b I as tome res, wh i c h f a i led tor e ass a cia t e ins u b seq u e n t<br />

cui t u r e . Na a t t emp twasmadeta i n d u cereass a cia t ion<br />

because, in field use, embryos are transferred directly to<br />

recipients. Splitting in standard conditions (without S,<br />

wit h Ca + +) res u I ted in. <strong>the</strong> I y sis a f rna n y b I as tome res.<br />

Mo r u 1a e s p lit inS and Ca + + can t a i n i n g me d i urn had no<br />

o b s e r v a b Iely 5 i s 0 fbi as tome res, and par tit ion e din t a<br />

quarters without loss of blastomeres.<br />

11<br />

ANABOLIC EFFECTS OF INSULIN ON MOUSE BLASTOCYSTS<br />

ARE MEDIATED VIA INSULIN RECEPTORS<br />

Mark B. Harvey and Peter L. Kaye<br />

Department of Physiology and Pharmacology,<br />

University of Queensland, St. Lucia, 4067.<br />

The addition of hormones to <strong>the</strong> culture medium <strong>for</strong> preimplantation<br />

embryos affects metabolism and improves developmental rate. In<br />

particular, insulin doubles <strong>the</strong> protein syn<strong>the</strong>tic rate in compacted<br />

embryos (1) and stimulates cellular division. The specific mediation of<br />

insulin's effect on embryos was investigated at varying concentrations<br />

of insulin and utilising an anti-insulin receptor antiserum.<br />

Two-cell embryos collected from superovulated Quackenbush mice were<br />

cultured in BMOC2 medium containing varying concentrations of insulin<br />

<strong>for</strong> 48h be<strong>for</strong>e blastocysts were transferred to fresh droplets of <strong>the</strong><br />

same medium plus 6p.M 3H-Ieucine (1Ci/L) and assay of protein syn<strong>the</strong>sis<br />

over 2h. Blastocysts from cultures with 0.17pM insulin were no more<br />

active than those derived from culture in control medium, but culture<br />

with 0.43-1~7pM insulin stimulated protein syn<strong>the</strong>sis by 25-75% with an<br />

EC~o=0.5pM (P


12<br />

COMPARISON OF TROPHOBLASTIC VESICLE AND OVINE BLASTOCYST SECRETED<br />

PROTEINS AND ANAYLSIS OF VESICLE METABOLIC ACTIVITY IN VITRO<br />

Clarkson, J .L. and Nancarrow, C.D. CSIRO Division of Animal<br />

production, P.O. Box 239, Blacktown, N.S.W. Australia, 2148.<br />

The aim of this study was to compare <strong>the</strong> proteins secreted by<br />

trophoblastic vesicles (TV's) with those secreted by day-17 ovine<br />

blastocysts cultured in vitro, and to estimate <strong>the</strong> metabolic activity<br />

of TV's during a 13 dayculture period.<br />

TV's were produced by cutting <strong>the</strong> trophoblast from day-17 ovine<br />

blastocysts into 2 rom sections. Each section was placed in 200 ~l B-2<br />

medium and incubated at 37°C in a humidified, 5% 02, 5 C02, 90% N2<br />

atmosphere. In Experiment I, <strong>the</strong> proteins secreted from TV'S and<br />

intact blastocysts cultured in vitro were collected, washed and<br />

concentrated using ultrafiltration:-Th"ey were <strong>the</strong>n separated using a<br />

Pharmacia FPLC with S-12 and S-6 gel filtration columns linked in<br />

series and monitored at 280 nm. Experiment II was identical to<br />

Experiment I except 1/10 <strong>the</strong> normal concentration of amino acids was<br />

used and 16.5 ~Ci/ml [3 H]-amino acid mixture was added to <strong>the</strong> medium.<br />

Additionally, <strong>the</strong> proteins were separated with a Pharmacia Mono-Q<br />

anion-exchange column using a linear 0 to 0.25 M NaCl gradient in 0.01<br />

M Tris-HCl (pH 8.2). One ml fractions were collected of which half was<br />

added to scintillant and <strong>the</strong> presence of labelled proteins determined<br />

on a Rack Beta counter. In Experiment III, 20 TV's were assigned to<br />

one of four treatments (trt). 0.125 ~Ci [methyl-3 H]-thymidine was<br />

added to each well in trt's I, II, III and IVan days 1, 4, 7 and 10<br />

respectively. After 72 hr of culture with <strong>the</strong> label, <strong>the</strong> TV'S were<br />

washed and placed in scintillation fluid and <strong>the</strong> thymidine<br />

incorporation determined.<br />

For statistical analysis, <strong>the</strong> morphological<br />

state of <strong>the</strong> vesicles were given <strong>the</strong> following values: fragmented<br />

tissue = 1, healthy, nonvesicle tissue = 2, TV-1 (multi-cell walled<br />

small vesicles) = 3, and TV-2 (single-cell walled, large vesicles) = 4.<br />

A vesicle which changed to a different stage during <strong>the</strong> label period<br />

was given a positive or negative value equal to <strong>the</strong> number of stages it<br />

progressed or regressed to. .<br />

In Experiment I, <strong>the</strong> gel filtration profile of <strong>the</strong> blastocyst<br />

secreted proteins consisted of nine peaks which corresponded to<br />

molecular weights (MW) of > 120, 103, 82, 54, 32, 8, 3, 2 and < 1<br />

kilodaltons (kD). The TV protein profile consisted of eleven peaks<br />

which corresponded to MW's of > 120, 103, 39, 28, 18, 10, 6, 2 and<br />

3 < 1 kD. In Experiment II, eight labelled protein peaks were recorded<br />

from <strong>the</strong> blastocyst extract in contrast to five labelled protein peaks<br />

recorded from <strong>the</strong> TV extract. In Experiment III, <strong>the</strong> mean % thymidine<br />

incorporation was 4.0, 1.2, 2.8 and 2.9 in trt' s I, II, III and IV<br />

respectively. The % incorporation was significantly less (p < .05) in<br />

trt II than in <strong>the</strong> o<strong>the</strong>r treatments. There was also significantly less<br />

(p < .05) progressive morphological change in trt II with mean changes<br />

of 0.8, -0.2, 0.0 and 0.4 in <strong>the</strong> respective treatments.<br />

We conclude that TV's produced several protein compounds similar to<br />

those produced by intact blastocysts cultured in vitro. Additionally,<br />

<strong>the</strong> TV's remain metaboli'cally active throughout a 13 day culture<br />

period.<br />

13<br />

CONTROL OF THE OVARIAN CYCLE IN MARMOSET MONKEYS FOR IN VITRO AND<br />

IN VIVO FERTI LIZATIoN AND THE TRANSFER OF CULTURED EMBRYOS TO<br />

SYNCHRONIZED RECIPIENTS<br />

A. Lopata,l P.M. Summers2 and J.P. Hearn2<br />

Department of Obstetrics and Gynaecology, University of Melbourne<br />

and Royal Women's Hospital, Melbourne, Victoria.<br />

2 Institute of Zoology, London, United Kingdom.<br />

It has recently been reported that an analogue of prostaglandin F2 alpha<br />

(cloprostenol, Estrumate, ICI, U.K.) caused a prompt shut-down of luteal function in <strong>the</strong><br />

marmoset monkey (1) and that this agent could be used <strong>for</strong> initiating a new cycle and<br />

subsequently inducing controlled ovulation in this primate species (2). We have applied<br />

<strong>the</strong>se procedures <strong>for</strong> timing <strong>the</strong> recovery of follicular and tubal oocytes<strong>for</strong> in vitro<br />

fertilization (IVF) and embryo culture. In addition we have evaluated <strong>the</strong> use of<br />

cloprostenol <strong>for</strong> synchronizing <strong>the</strong> reproductive cycles of oocyte donors with that of embryo<br />

reci pi ents.<br />

The marmoset monkeys used in <strong>the</strong> present studies were proven breeders. In each<br />

treatment protocol a new reproductive cycle was initiated by injecting 0.5 ug cloprostenol<br />

between days 10 and 24 of pregnancy. In 40 animals <strong>the</strong> timing of ooycte collection was<br />

based on <strong>the</strong> administration of 75 IU hCG at <strong>the</strong> following intervals after cloprostenol: at<br />

9 am on <strong>the</strong> eighth day (protocol I, n=2o), at 9 am on <strong>the</strong> seventh day (protocol 2, n=6), at<br />

5 pm on <strong>the</strong> seventh day (protocol 3, n=14). Animals that were to be used as embryo<br />

recipients were treated simultaneously and in <strong>the</strong> same way as <strong>the</strong> oocyte donors. At 24<br />

hours after hCG oocytes were obtained by aspirating ovarian follicles, or flushing <strong>the</strong><br />

Fallopian tubes, under general anaes<strong>the</strong>sia. Semen was obtained by electroejaculating<br />

fertile males under general anaes<strong>the</strong>sia. The sperm used <strong>for</strong> in vitro insemination were<br />

prepared by a swim up procedure. Insemination and embryo culture was per<strong>for</strong>med in minimum<br />

essentia1 medi um supplemented with 10% human cord serum. Embryos were transferred<br />

surgically at <strong>the</strong> 4 to 6 cell stages to <strong>the</strong> uterine lumen of synchronized recipients.<br />

In treatment protocol 1 9/20 (45%) marmosets had ovulated be<strong>for</strong>e oocyte collection,<br />

none had ovulated in protocol 2, and 2/14 (14%) in protocol 3. There was no significant<br />

difference in <strong>the</strong> mean !lumber of follicles aspirated and oocytes recovered between <strong>the</strong><br />

protocols. The fertilization rate of morphologically mature eggs was 64% (21/33) and 40%<br />

(6/15) <strong>for</strong> eggs assessed as being immature. Of <strong>the</strong> 21 mature eggs that ferti lized 20<br />

produced cleaving embryos. Five embryos were transferred to recipients at 4 to 6-cell<br />

stages and of <strong>the</strong> 15 'that remained in culture six developed to <strong>the</strong> morula stage and three<br />

progressed to advanced blastocyst stages. Two synchroni zed reci pi ents recei ved two embryos<br />

each and one of <strong>the</strong> marmosets gave birth to twins. A third recipient -eceived a single<br />

embryo and gave birth to a male infant. Two additional recipients received single embryos<br />

that were cultured in vitro but were derived from oocytes fertilized in <strong>the</strong> oviducts. Both<br />

recipients became pregnant but only one delivered a normal infant.<br />

This is <strong>the</strong> first report of successful IVF and embryo transfer in <strong>the</strong> marmoset monkey.<br />

Moreover we have shown that pharmaco1ogi ca1 agents can be used to accurately synchroni ze<br />

<strong>the</strong> reproductive cycles of oocyte donors and embryo recipients following IVF in a nonhuman<br />

primate.<br />

(1) Summers, P.M., Wennik, C.J. and Hodges, J.K. J. Reprod. Fertil. (1985). U.:133-138.<br />

(2) Hodges, J.K., Cottingham, P.G., Summers, P.M. and Yingnan, L. Fertil. Steril. (1987).<br />

~:299-305.


14.<br />

IMMrJIDFIOORFSCENr SlUDIES OF F'R.B3NANCY-ASOOCIATED ELASTASE INHIBI'IOR<br />

(PAEI) EXPRESSION BY ACrIVATED GAMETES AND PREIMPLANI'ATION HAMSTER<br />

EMBRYOS.<br />

M.J. Sinosich, S. Ianzendorf l , M.D. Bonifacio, D.M. saunders, G.D.<br />

Hoogen<br />

Department of Obstetrics and Gynaecology, Royal North Shore<br />

Hospital, st. Leonards NSW, Australia.<br />

~e Jones Institute <strong>for</strong> <strong>Reproductive</strong> Medicine, Department of<br />

Obstetrics and Gynaecology, Eastern Virginia Medical School,<br />

Norfolk, Virginia 23507. (SFON: R. Stillman)<br />

Human pregnancy-associated plasma protein-A (PAPP-A), a large (Mr<br />

820kd) heparin-binding proteoglycan, specifically and potently<br />

inhibited .granulocyte elastase activity. Wheras irranunological and<br />

functional analogs of human PAPP-A were readily demonstrated in<br />

pregnant high-order non-hLmlan primates, functional analogs have been<br />

demonstrated in non-primate mammals with hemochorial placentation.<br />

A large hep:lrin-binding elastase inhibitor was isolated fran term<br />

guinea pig placentae and injected into N Z white rabbits to prepare<br />

antisera <strong>for</strong> indirect irranunofluorescent studies on rna.ture oocytes<br />

and embryos obtained fran superovulated golden hamsters.<br />

Pronuclear, two-cell, four-cell arrl blastocyst embryos were<br />

surgically removed at 1, 2, 3 and 4 days after rna.ting and fixed in<br />

phosp:lte buffered <strong>for</strong>rna.lin <strong>for</strong> 60 minutes. No fluorescence was<br />

Elanonstrated on intact oocytes, whereas in vivo derived<br />

preimplantation embryos were strongly fluorescent. Fur<strong>the</strong>rmore, in<br />

vitro microinjection (with or without sperm) irrluced PAEI expression<br />

within 4 h. '!hese' findings suggest; 1) hLnnan PAPP-A is a more<br />

recently evolved <strong>for</strong>m of marmnalian PAEI. 2) sperm nuclei and gene<br />

replication are not required <strong>for</strong> expression ,of PAEI, 3) oocyte<br />

activation irrluces PAEI expression, 4) PAEI rna.y act as a barrier<br />

against maternal phagocytic-proteolytic defenses, and 5) PAEI may<br />

prove a useful target antigen <strong>for</strong> immunological contraception,<br />

having high specificity <strong>for</strong> activated gametes or <strong>the</strong>ir early<br />

embryonic derivatives.<br />

15<br />

RELEASE OF GROWTH FACTOR(S) BY EMBRYO DERIVED PLATELET<br />

ACTIVATING FACTOR (EPAF)<br />

Y.C. Smart, L.A. Adamson, R. A. Wilcox and T.K. Roberts<br />

Faculty of Medicine and Dept. of Biological Sciences, University of Newcastle<br />

Preimplantation pregnancy in <strong>the</strong> mouse is associated with a transient state<br />

of thrombocytopenia induced by <strong>the</strong> release from <strong>the</strong> embryo of a platelet<br />

activating factor (EPAF) (1,2). In vivo and in vitro studies have implicated EPAF<br />

in a role which acts to initiate implantation and maintain foetal survival (3,4).<br />

The mechanism by which EPAF triggers <strong>the</strong> events required <strong>for</strong> successful<br />

pregnancy is not known.<br />

This paper reports <strong>the</strong> results of an in vitro experiment which measures <strong>the</strong><br />

effect of a molecule(s), released by <strong>the</strong> action of mouse EPAF on human platelets,<br />

on Balb/c 3T3 fibroblast cells. Day 5 embryo culture medium (ECM) with predetermined<br />

thrombocytopenic activity in mice in vivo was incubated with washed<br />

human platelet rich plasma (PRP) in microtitre wells at 37oC/5%C02 <strong>for</strong> 4<br />

hours. Jhe wells were washed to remove ECM and platelets. Quiescent 3T3 cells<br />

(2 x 10 ) were seeded into <strong>the</strong> EPAF/PRP treated wells. The cells were cu Itured<br />

at 37 o C/5%C0 2 <strong>for</strong> 8 days. Each day <strong>the</strong> 3T3 cells were scored morphologically,<br />

trypsinised and <strong>the</strong> cell concentration determined. Wells pre-treated with <strong>the</strong><br />

syn<strong>the</strong>tic phospholipid platelet activating factor (PAF) and PRP served as <strong>the</strong><br />

positive control while wells containing ECM alone, control medium (CM) and PRP,<br />

and PRP alone were <strong>the</strong> negative controls.<br />

Results of replicate experiments showed that EPAF/PRP and PAF/PRP<br />

treated wells supported continued proliferation of 313 cells with rapid growth<br />

from Days 3-7, peaking on Days 6 and 7 of culture. 313 cells seeded into wells<br />

pre-treated with ECM alone, PRP alone and CM/PRP proliferated poorly. Results<br />

of a typical experiment are shown in Figure I. We conclude that EPAF and PAF<br />

activates platelets to release growth factor(s) which binds to <strong>the</strong> tissue culture<br />

wells and acts mitogenically on <strong>the</strong> 3T3 fibroblast cells. It is likely that in vivo,<br />

EPAF activates platelets and/or oviduct cells to release growth factors important<br />

in successful pregnancy.<br />

Figure I. Effects of EPAF and ~<br />

PAF on 3T3 cells<br />

100<br />

80<br />

60<br />

40<br />

20<br />

-0- EeM<br />

... ,CM-PRP<br />

.. CM-PRP<br />

+ PAF-PRP<br />

.. PRP<br />

o .J--:¢;+::::=~~==8===&--.--,<br />

o 4 6 10<br />

DAYS IN CULTURE<br />

(I) O'Neill, C. (1985) J. Reprod. Fert. 73:559.<br />

(2) Roberts, T.K. et 01 (1987) Fert. Sterif. 47:848.<br />

(3) Adamson, L.M. et al (1987) Am. J. Reprod. Immunol. Microbiol. 13: 117.<br />

(4) O'Neill, C. (1985). J. Reprod. Fert. 73:567.


16 17<br />

PAP-PRETREATMENT IMPAIRS EMBRYONIC DEVELOPMENT<br />

ADAMSON LMi' STANGER JDj SMART YC<br />

and ROBERTS TK.<br />

Department of Biological Sciences, Department of Surgical<br />

Sciences and Lingard Hospital, Newcastle, NSW.<br />

The way in which <strong>the</strong> mammalian embryo signals its presence to <strong>the</strong><br />

maternal system remains an enigma to a large extent. Recently however,<br />

work in this area has established <strong>the</strong> production of a factor by <strong>the</strong><br />

pre-implantation mouse and human embryo with platelet activating<br />

activity(1,2). We call this factor, embryo-derived platelet activating<br />

factor (EPAF), because of its biological resemblance to <strong>the</strong><br />

phospholipid platelet activating factor, PAF-ace<strong>the</strong>r (3,4). The result<br />

of EPAF production during <strong>the</strong> first 5 days post-mating in mice is <strong>the</strong><br />

induction of a mild thrombocytopenia, early pregnancy associated<br />

thrombocytopenia (EPAT), in <strong>the</strong> maternal system.<br />

We have previously demons trated that, in <strong>the</strong> mouse, pre-mating<br />

treatment with semi-syn<strong>the</strong>tic PAF-ace<strong>the</strong>r has two obvious effects.<br />

Firstly, <strong>the</strong> treatment inhibits <strong>the</strong> onset of early pregnancy associated<br />

thrombocytopenia, presumably by inducing platelet desensitization to<br />

PAF-ace<strong>the</strong>r and to EPAF. Secondly, examining <strong>the</strong>se animals on day 10<br />

post-mating reveals that PAF-pretreated animals have significantly<br />

fewer implantation sites than <strong>the</strong> PBS-pretreated controls (4).<br />

We have extended this study to determine <strong>the</strong> effects of<br />

PAF-pretreatment on pre-implantation embryo development. QS female<br />

mice were given single injections of sub-threshold levels of<br />

PAF-ace<strong>the</strong>r or PBS on three consecutive days immediately prior to<br />

mating. Oviducts and uteri were removed from <strong>the</strong>se animals on ei<strong>the</strong>r<br />

day 1, 2, 3 or 4, and <strong>the</strong> embryos flushed out to record both embryo<br />

number and developmental stage. Results illustrated that eventhough<br />

PAF-pretreatment had no effect on embryo development during days 1, 2<br />

and 3, by day 4 post-mating, PAF pre-treated animals had fewer embryos<br />

and <strong>the</strong>ir development was impaired (Table 1). We suggest that<br />

PAF-pretreatment affects embryonic development, perhaps by inducing<br />

uterine asynchrony resulting in a utero-toxic effect on <strong>the</strong> implanting<br />

embryos. If this be <strong>the</strong> case, EPAF may be an initial embryonic signal<br />

to <strong>the</strong> maternal system <strong>for</strong> uterine receptivity.<br />

TABLE 1: EMBRYONIC NUMBER AND DEVELOPMENT DURING<br />

PREIMPLANTATION STAGE IN PAF AND PBS TREATED MICE.<br />

DAY<br />

MEAN EMBRYO NUMBER % EMBRYO DEVELOPMENT TO<br />

(No. Animals)<br />

EXPECTED STAGE<br />

PAF<br />

PBS<br />

PAF<br />

PBS<br />

1 21+3(9) 29+3(9) 98.5<br />

93.6<br />

2 2ft"5(7) 30+6(8) 77.0<br />

80.7<br />

3 22+6(10) 18+2(7) 74.1<br />

78.6<br />

4 9±2(15) 19±2(13)* 30.6<br />

81.1<br />

* P


18 19<br />

VAI.JDATICN OF MEIHXXJ[(X;Y FeR Sl'ODY OF PULSATILE IH SECRRl'ICN IN '.!HE<br />

RAT: CANNUIATICN RaJrE, SAMPLIN::; INI'ENSITY AND DURATICN<br />

Qillan Ibng, David J Handelsrran<br />

Department of Medicine, University of Sydney<br />

Pituitary-gonadal function is regulated by <strong>the</strong> hypothalamus through<br />

episodic GnRH release into pituitary portal blcxxi which induces<br />

pulsatile LH secretion. The experimental study of pulsatile LH<br />

secretion as a non-invasive index of reproductive neuroendocrine<br />

function of <strong>the</strong> hypothalamus has been validated in large a.niJnal<br />

species however <strong>the</strong>re has been little systematic study of <strong>the</strong><br />

requirem:mts <strong>for</strong> valid LH pulse studies in <strong>the</strong> rat which species has<br />

econanic and ethical advantages as well as having <strong>the</strong> rrost well<br />

studied reproductive neuroendocrine system. In particular <strong>the</strong>re is no<br />

co~sensus about which vessel to cannulate (carotid artery vs jugular<br />

vell) or about standards of sampling intensity and duration re:;ruired<br />

to obtain stable and valid estiJrates of pulsatile LH secretion.<br />

There<strong>for</strong>e we have examined (a) <strong>the</strong> short-tenn effect of unilateral<br />

carotid artery ligation on pulsatile LH secretion and (b) compared<br />

para:rreters of pulsatile LH secretion (mean, max:iJnurn, minimum, pulse<br />

frequency and interpulse interval, pulse amplitude, pulse length)<br />

estiJrated fran an intensive sampling regime (ql0 min, 6hr) with (i)<br />

<strong>the</strong> first 1,2,3,4 and 5hr at q10 min and (ii) with less intensive<br />

sampling (q20 min, 6hr). Castrate mature male rats (n=18) underwent<br />

serial blcxxi sampling (0.25 rnl, q10 min, 6 hr) via right external<br />

jugular vein cannula while freely rrobile and under minimal stress<br />

conditions and with volumetric blcxxi replacerrent by thrice-washed<br />

donor rat erythrocytes resuspended in a charcoal-extracted plasma<br />

P70te~ substitut~. Half <strong>the</strong> rats ·also unde:rwent left carotid artery<br />

1J.gatJ..on at <strong>the</strong> tJ.n'le of venous cannulation. Pulse study samples were<br />

assayed toge<strong>the</strong>r in duplicate and pulses analysed by an objective<br />

method (PULSAR) optimized <strong>for</strong> pulsatile LH studies in <strong>the</strong> rat. .<br />

(g10 min) 1 hr 2 hr 3 hr 4 hr 5 hr 6 hr<br />

Mean LH(ng/rnl) 4.0+0.6 3.7+0.5 3.5+0.4 3.5+0.4 3.5+0.4 3.4+0.5<br />

Peaks (/6hr) *16.0+1.0 *12.7+0.8 11.8+0.8 11.2+0.7 10.9+0.6 10.4+0.5<br />

Amplit (ng/rnl) 3.9+0.7 3.8+0.6 3.7+0.6 3.6+0.6 3.6+0.5 3.6+0.6<br />

Mean LH (ng/rnl)<br />

Peaks (/6hr )<br />

Amplitude (ng/rnl)<br />

(Mean+SEM,<br />

Carotid ligation<br />

q20 min/6 hr ql0 min/6 hr ql0 min/6 hr<br />

3.5+0.3 3.4+0.5 *1.8+0.3<br />

*4.9+0.3 10.4+0.5 *6.2+1.2<br />

3.6+0.4 3.6+0.6 2.7+0.4<br />

* P0.05 repeatedmeasures.ANOVA).<br />

~r intensity sampling regime (q20 min) gave marked<br />

underestJ..mates of para:rreters of. pulsatile LH secretion canpared with<br />

qlO min sampling within a 6 hr pulse study. we conclude that <strong>for</strong> valid<br />

estiJrates of pulsatile LH secretion in physiological studies of <strong>the</strong><br />

castrate mature male rat (i) carotid artery cannulation is undesirable<br />

and (ii) sampling intensity of at least q10 min and durations of at<br />

least 3 hours is required.<br />

THE ROLE OF LEYDIG CELLS IN THE REGULATION OF INHIBIN PRODUCTION<br />

A.E. Drummond, G.P. Risbridger, D.M. de Kretser<br />

Department ofAnatomy, Monash University, Melbourne, Victoria.<br />

It is established that <strong>the</strong> Sertoli cell is <strong>the</strong> source of inhibin in <strong>the</strong> testis and<br />

<strong>the</strong> production of this hormone is controlled by FSH (1-2). The role of testosterone<br />

in regulating inhibin syn<strong>the</strong>sis is equivocal of Leydig cell function (3). The aim of<br />

this study was to determine <strong>the</strong> effect of acute stimulation of Leydig cell function by<br />

<strong>the</strong> administration of hCG/LH (human chorionic gonadotrophin/Luteinizing hormone),<br />

or depletion of Leydig cells by implementing <strong>the</strong> Leydig cell specific cytotoxin ethane<br />

dimethane sulphonate.<br />

Adult male rats received a single injection of saline or 100 IU hCG (Pregnyl,<br />

Organon). Serum was collected at time intervals up to 5 days after hCG<br />

administration. A second group of animals received EDS, which destroys Leydig cells,<br />

75mg/kg body weight, or <strong>the</strong> vehicle dimethyl sulphoxide (DMSO) in a single<br />

intraperitoneal injection. Serum was collected 1, 2 and 4 weeks after injection. A<br />

third group of animals received combined EDS/hCG treatment. Four days after EDS<br />

injection a single subcutaneous injection of 100 IU hCG was administered. Serum was<br />

collected 6 hours after hCG injection. Serum samples were assayed <strong>for</strong> inhibin using<br />

a specific double antibody radioimmunoassay (4).<br />

Serum inhibin levels were significantly elevated within 6 hours of hCG injection<br />

peaking at 24 hours and did not return to control levels within <strong>the</strong> 5 day period of<br />

<strong>the</strong> study suggesting a stimulatory effect of Leydig cell products on inhibin<br />

production. Serum inhibin levels were significantly elevated 2 and 4 weeks after EDS<br />

injection. The destruction of Leydig cells by EDS blocked <strong>the</strong> rise in serum inhibin<br />

levels after hCG injection (Table 1). Paradoxically, after EDS injection alone inhibin<br />

levels were unchanged until 2 and 4 weeks later when <strong>the</strong>y were significantly elevated<br />

suggesting that an inhibitory influence of <strong>the</strong> Leydigs on inhibin secretion had been<br />

removed by EDS treatment.<br />

These data suggest an effect of Leydig cell products in regulating inhibin<br />

production. The role of testosterone and o<strong>the</strong>r factors in this phenomenon remains to<br />

be elucidated.<br />

Table 1.<br />

Saline<br />

hCG (6h)<br />

DMSO<br />

EDS (4 days)<br />

EDS/hCG (6h)<br />

Serum inhibin and testosterone concentrations after hCG, EDS or<br />

combined treatment.<br />

Inhibin (Ujml)<br />

2.77 ± 0.43<br />

4.84 ± 0.24*<br />

2.64 ± 0.78<br />

3.03 ± 0.55<br />

2.89 ± 0.64<br />

*p


HIGHLY roIARIZED SECREl'ICN OF INHIBIN BY RAT SERIOLI CEUS IN<br />

'lWllH::IWffiER CULTURE SYSTEM:<br />

Jermifer A Spaliviero~ *David M Robertson, Elsa Kidston,<br />

Peter F Hall, David J Handelsman<br />

Deparl:m9nt of Medicine, University of Sydney, Sydney and<br />

*DeparbUent of Anatcmy, Monash University, Melbourne<br />

20<br />

Sertoli cells are <strong>the</strong> princir.al source of circulating inhibin in<br />

<strong>the</strong> male however <strong>the</strong> ho:t:IOClnal regulation of inhibin secretion and its<br />

route of entry into <strong>the</strong> blood-stream remain unclear. The central role<br />

of Sertoli cells in spenratogenesis includes secretion of n'UllErotis<br />

peptides and <strong>the</strong> maintenance of <strong>the</strong> blood-testis barrier which<br />

secludes ,developing genninal cells within <strong>the</strong> diffusion-tight<br />

adluminal canpartrnent away fran <strong>the</strong> extracellular fluid. The<br />

distinctly polar aspects of sertoli cells in-situ pennits secretion<br />

via <strong>the</strong> basal surface into <strong>the</strong> testicular interstitial and<br />

extracellular fluid and/or via <strong>the</strong> apical surface into <strong>the</strong> adluminal<br />

carnpartment and <strong>the</strong>reby into <strong>the</strong> rete testis fluids and seminal<br />

plasrra. There<strong>for</strong>e using a twin chamber culture system, which :mimics<br />

<strong>the</strong> polarised state of sertoli cells in vivo (1), we have investigated<br />

<strong>the</strong> vectorial secretion of inhibin and its ho:t:IOClnal regulation.<br />

Bertoli cells isolated f:rom 2l-day old rats were cultured in a twin<br />

c~r system using fully defined medium (supplerrented Eagles MEM) in<br />

trlp1J.cate wells under basal conditions or stinmlated with oFSH<br />

(250ng/ml), testosterone ([TESJ ,1000nM), insulin ([INSJ 5ug/ml)<br />

retinoic acid ([RAJ ,500nM) or pai:rw:i.se cernbinations of FSH with INS'<br />

TES or RA or all 4 stimuli (FIRT). Rat inhibin and transferrin ~<br />

media (24 hr, days 5-7) were measured by specific RIA.<br />

UPPER CHAMBER RAT INHIBIN Wlml)<br />

BASAL FSH TES INS RA FSH/INS FSH/TES<br />

1.4 5.3 1.0 3.1 1.1 5.9 7.9<br />

(0.1) (0.1) (0) (0.1) (0.3) (0.5) (0.5)<br />

Mean (SEM), * p


22<br />

TREATMENT WITH A GnRH AGONIST DELAYS REPRODUCTIVE<br />

DEVELOPMENT IN RAM LAMBS<br />

A. J. Tilbrookl, D.E. Galloway2, A.H. Williams!, R.C. Oppenheim 3 ,<br />

W.J. Thie1 3 and LJ. Clarke 4<br />

1 Animal Research Institute, Dept. of Agric. & Rural Affairs, Werribee, 3030.<br />

2 Veterinary Clinical Centre, Univ. Melb., Werribee, 3030,<br />

3 Victorian College of Pharmacy, Parkville, 3052,<br />

4 Medical Research Centre, Prince Henry's Hospital, St. Kilda, 3004.<br />

Prolonged continuous infusion of adult Soay rams with an agonist of<br />

gonadotrophin rp.leasing hormone (GnRH) initially stimulated <strong>the</strong> secretion of<br />

luteinizing hormone (LH) and testosterone (T), followed by a suppression in <strong>the</strong><br />

secretion of LH, T and follicle stimulating hormone (FSH)(l). There was also a<br />

small decrease in testicular diameter and disappearance of <strong>the</strong> sexual flush (1) but<br />

sexual behaviour was not studied. The aim of our study was to determine if<br />

development of sexual behaviour and testicular size could be delayed in<br />

prepubertal rams by continuous administration of a GnRH agonist, and if this<br />

treatment would influence <strong>the</strong>ir growth.<br />

Preliminary work (Tilbrook et al., unpublished) demonstrated that a GnRH<br />

agonist ({D_trpli_Pro 9 N-ethyl amide}GnRH) was effective in inhibiting LH and FSH<br />

secretion in adult we<strong>the</strong>rs when administered using s.c. mini-osmotic pumps<br />

(Alzetminipumps) or prototype slow-release pellets (pellets).<br />

In March, rams (aged 20-28 weeks) weighing 30.7±O.7 kg (mean±SD) were<br />

ei<strong>the</strong>r untreated (n=10; entire), treated with 50llg of <strong>the</strong> GnRH agonist/day using<br />

minipumps <strong>for</strong> 16 weeks (n=lO), given a pellet containing 100 Ilg of <strong>the</strong> GnRH<br />

agonist every 4 weeks <strong>for</strong> 16 weeks (n=lO) or surgically castrated (n=15; castrate).<br />

Rams that mounted and/or ejaculated during .15 minutes of individual exposure to 4<br />

oestrous ewes were defined as sexually active. Testicular volume (ml) was<br />

estimated using calibrated beads, and liveweight (kg) was also measured.<br />

Table 1. Effect of GnRH agonist on <strong>the</strong> testicular volume (TV) and liveweight<br />

(LWT) oframs and <strong>the</strong> proportion oframs sexually active (SA)<br />

Be<strong>for</strong>e treatment End of treatment 8 weeks after treatment<br />

Group TV" LWT* SA TV" LWT* SA TV" LWT*<br />

Entire 45±8 3l±1.0 9/lO x 118±12 ax 44±1.Ox lO/lO x 158±1 IX 51±1.0 xa<br />

Castrate 32±O.5 0/15 Y 41±1.0a 0/15 Y 45±1.0 Yc<br />

Minipump 45±7 29±OA 2/lO Y 68±lO b 40±1.0 zb 7/l0 x 91±1 JY 41±1.0 z<br />

Pellet 39±6 31±0.5 3/10 Y 82±lO cy 42±1.oya 7/lO x 92±5 Y 49±2.0 b<br />

"means±standard errors; x.y,Z differ at p


24<br />

IDENTIFICATION BY HPLC AND IMMUNOCYTOCHEMISTRY OF<br />

HYPOTHALAMIC OXYTOCIN AND MESOTOCIN IN THE<br />

BRUSHTAIL POSSUM<br />

R. Bathgate!, C. Sernia! and R.T. Gemmel1 2<br />

tDepartment of Physiology and Pharmacology, 2Department of Anatomy,<br />

University of Queensland, St. Lucia, 4067<br />

All of <strong>the</strong> nine species of Australian marsupials investigated to<br />

date appear to syn<strong>the</strong>sise mesotocin, which is typical of non-mammalian<br />

tetrapods, instead of oxytocin (1,2). Since much of <strong>the</strong> reported data<br />

are based on bioassay of pituitary extracts and are qualitative only,<br />

we decided to examine <strong>the</strong> hypothalamus of one of <strong>the</strong>se species, <strong>the</strong><br />

brushtail possum, using high pressure liquid chromatography (HPLC) and<br />

immunocytochemistry.<br />

Fresh hypothalamic tissue was homogenized in 20 vols (w:v) of<br />

1 molll HCI containing 15% TFA, 5% <strong>for</strong>mic acid and 1% NaCI. The<br />

homogenate was centrifuged and <strong>the</strong> peptides in <strong>the</strong> supernatant<br />

extracted on C-18 cartridges and eluted with 0.1% C-18 HPLC column<br />

(LKB UltroPac) using 0.1% TFA in a 10-35% ACN gradient and" assayed <strong>for</strong><br />

mesotocin and oxytocin by specific radioimmunoassays. The major<br />

peptide was mesotocin, found at 9 to 12 ng per hypothalamus. An<br />

additional peak with <strong>the</strong> relative mobility of oxytocin was also<br />

detected but at <strong>the</strong> lower quantity of 1.5 to 2.5 ng per hypothalamus.<br />

Serial sections of para<strong>for</strong>maldehyde-fixed hypothalami were<br />

i~munostained <strong>for</strong> ei<strong>the</strong>r oxytocin or mesotocin using a biotinstreptavidin-peroxide<br />

system (Amersham). Heavy mesotocin staining was<br />

found in <strong>the</strong> Paraventricular and Supraoptic nuclei with only light<br />

oxytocin staining in <strong>the</strong> same cells.<br />

These results show that at least one Australian marsupial<br />

expresses both mesotocin and oxytocin genes; a situation which has<br />

also been reported <strong>for</strong> <strong>the</strong> opossum, D. virginiana (3).<br />

(1) Chauvet, M.T., et. a1. (1981) FEBS Lett. 129: 120-122.<br />

(2) Hurpet, D., et. a1. (1982) Int. J. Pept. Prot. Res. 19: 366-371.<br />

(3) Chauvet, J., et. a1. (1984) BBRe 123: 306-311.<br />

25<br />

THE RELATIONSHIP OF BREASTFEEDING PATTERNS TO THE· LENGTH OF<br />

LACTATIONAL AMENORRHOEA AND OVARIAN INACTIVITY POST PARTUM<br />

P.R. Lewis, M.B. Renfree, R.V. Short<br />

Depts. of Physiology and Anatomy, Monash University, Melbourne, 3168<br />

The duration and frequency of breastfeeding are major factors<br />

contributing to <strong>the</strong> length of lactational amenorrhoea. Starting in<br />

1984, a study was conducted at Monash University to provide additional<br />

in<strong>for</strong>mation on <strong>the</strong> influence of <strong>the</strong>se and o<strong>the</strong>r factors on <strong>the</strong> duration<br />

of lactational amenorrhoea. A total of 146 breastfeeding women were<br />

recruited in <strong>the</strong>ir first month post partum. They kept records of all<br />

feeds given to <strong>the</strong> baby during two 24 hour periods each week and<br />

collected saliva samples 2-3 times per week <strong>for</strong> progesterone assay to<br />

monitor corpus luteum function following ovulation. A subset of women<br />

also collected urine twice per week and this was analysed <strong>for</strong> total<br />

oestrogens and pregnanediol.<br />

Menstruati.on resumed within 1 year post partum in 70% of <strong>the</strong> women<br />

(shortest amenorrhoea: 1 month); <strong>for</strong> <strong>the</strong> remaining 30%, amenorrhoea<br />

lasted from 13 to 22 months. Three women became pregnant in <strong>the</strong>ir<br />

second year of lactation without experiencing a menstrual bleed. First<br />

post partum ovulations occurred from less than 1 month (1%) to 22<br />

months (1%) after birth; median time to first ovulation was 10.5<br />

months. Initial menstrual cycles were irregular in length and<br />

frequently anovulatory; women who first ovulated early in lactation<br />

often had luteal phases characterised by short durations or low<br />

progesterone concentrations. Of women with amenorrhoea lasting less<br />

than 3 months, 30% ovulated be<strong>for</strong>e <strong>the</strong>ir first menstruation. However,<br />

70% of women experiencing <strong>the</strong>ir first menstruation after 12 months<br />

ovulated be<strong>for</strong>e that menstruation.<br />

Breastfeeding patterns varied greatly between women both with<br />

respect to frequency and total time spent feeding during <strong>the</strong> 24 hour<br />

recording period. During <strong>the</strong> first 1-2 months of lactation <strong>the</strong>re were<br />

no differences in <strong>the</strong>se parameters between groups of women with<br />

different lengths of amenorrhoea. The use of dummies and introduction<br />

of supplements, however, were signiyicantly different between groups;<br />

women who menstruated early used dummies more frequently ~nd introduced<br />

supplements earli.er and more rapidly.<br />

As yet, no specific <strong>for</strong>mula can be given to predict <strong>the</strong> length of a<br />

woman's lactational amenorrhoea from her breastfeeding pattern alone.<br />

Total duration of feeding may prqve to be lI.lore important than<br />

frequency, hut. an interacti.on between factors may explain <strong>the</strong> wide<br />

variation in breastfeeding patterns seen between women with similar<br />

periods of amenorrhoea.


2~<br />

INHIBITORY EFFECT OF PURE 31 kDa BOVINE INHIBIN ON GnRH-INDUCED<br />

UP-REGULATION OF GnRH BINDING SITES IN CULTURED RAT ANTERIOR<br />

PITUITARY CELLS<br />

Wang Qi Fa, P.G. Farnworth, H.G. Burger and J.K.<br />

Findlay<br />

Medical Research Centre, Prince Henry's Hospital Campus of<br />

The Monash Medical Centre, St Kilda Road, Melbourne, Victoria, 3004<br />

GnRH modifies gonadotroph functions, having both acute effects<br />

(minutes) on hormone release and delayed effects (hours) on receptor<br />

up-regulation and gonadotrophin syn<strong>the</strong>sis. It is well documented that<br />

inhibin, a gonadal glycoprotein hormone, suppresses <strong>the</strong> acute effects<br />

of GnRH both in vivo and in vitro (1). However, whe<strong>the</strong>r inhibin also<br />

modulate~ delayed effects of GnRH remains to be determined. The<br />

present study was designed to investigate <strong>the</strong> effect of inhibin on<br />

GnRH-induced up-regulation of GnRH binding sites in cultured rat<br />

anterior pituitary cells.<br />

Pituitary cells from adult male Sprague-Dawley rats were dispersed<br />

and cultured <strong>for</strong> 2 days, after which <strong>the</strong> media were replaced and <strong>the</strong><br />

cells were <strong>the</strong>n exposed to stimuli, with or without test substances,<br />

<strong>for</strong> 10 h. Upon <strong>the</strong> completion of incubation, <strong>the</strong> cells were removed<br />

and <strong>the</strong> binding of GnRH was determined using iodinated Buserelin as<br />

tracer (2). Exposure to GnRH (10 nM) resulted in a 90% increase in<br />

specific binding sites <strong>for</strong> GnRH. Inhibin (0.01 to 10 U/ml) suppressed<br />

GnRH-induced up-regulation of GnRH binding sites in a dose-dependent<br />

manner with an IC of 0.13 U/ml (5.5 pM). Unlike inhibin, nei<strong>the</strong>r<br />

Trans<strong>for</strong>ming Grow~R Factor-~ (0.4-400 pM) nor Mullerian Inhibitory<br />

Substance (0.1-100 nM), two inhibin-related peptides, had any<br />

detectable effect on <strong>the</strong> GnRH-stimulated increase in GnRH binding<br />

sites; suggesting that <strong>the</strong> effect was specific to inhibin. In<br />

addition, inhibin (10 U/ml) significantly inhibited <strong>the</strong> up-r~gulation<br />

of GnRH binding sites induced by <strong>the</strong> calcium ionophore A23187 (0.1<br />

~M), indicating that this effect 2f inhibin can occur, at least in<br />

part, at a stage subsequent to Ca + mobilization. Finally, inhibin at<br />

a concentration up to 300 Ulml did not compete with iodinated GnRH-A<br />

<strong>for</strong> GnRH binding sites.<br />

We conclude from this study that inhibin at low concentrations<br />

modulates delayed effects of GnRH on its own receptors and that<br />

inhibin might <strong>the</strong>re<strong>for</strong>e participate in <strong>the</strong> physiological regulation of<br />

GnRH receptors.<br />

(1) Findlay, J. Fertil. Steril. 46(5):770-783, 1986.<br />

(2) Loumaye, E. &Catt, K.J. Science 215:983-985, 1983.<br />

27<br />

TRANSFORMING GROWTH FACTOR ~ ENHANCES BASAL AND FSH STIMULATED<br />

INHIBIN PRODUCTION BY RAT GRANULOSA CELLS IN VITRO<br />

Zhang Zhiwen, J.K. Findlay, R.S. Carson, A.C. Herington<br />

and H.G. Burger<br />

Medical Research Centre, Prince Henry's Hospital Campus,<br />

Monash Medical Centre, St Kilda Road, Melbourne, Victoria, 3004<br />

Trans<strong>for</strong>ming growth factor ~ (TGF-~), a member of <strong>the</strong> inhibin gene<br />

family, is produced by ovarian <strong>the</strong>cal tissue (1), and has a paracrine<br />

influence on granulosa cell steroidogenesis (2) and EGF receptor<br />

number (3). We investigated <strong>the</strong> effects of TGF-~ on inhibin<br />

production by granulosa cells from diethylstilbestrol-treated,<br />

immature rats.<br />

Granulosa cell primary cultures and inhibin radioimmunoassay of<br />

conditioned media were per<strong>for</strong>med as described previously (4). TGF~<br />

(0.01-3 ng/ml) caused a dose-dependent increase in both basal and<br />

FSH-stimulated inhibin production by rat granulosa cells in culture.<br />

The TGF~ dose-response curve in <strong>the</strong> absence of FSH was approximately<br />

parallel to that in <strong>the</strong> presence of ei<strong>the</strong>r a minimally effective d~se<br />

(1 ng/ml) or a maximally effective dose (30 ng/ml) of FSH, suggestlng<br />

an additive effect of <strong>the</strong>se two agents on inhibin production. There<br />

was also a suggestion of an increased sensitivity of granulosa cell<br />

inhibin production to FSH when <strong>the</strong> cells were coincubated with TGF~.<br />

The time course study showed that similar to FSH, <strong>the</strong> stimulatory<br />

effect of TGF~ on basal and FSH-stimulated inhibin production was<br />

evident on day 1 and was maximal by day 4. In addition, EGF reduced<br />

FSH-stimulated inhtbin production with an I~O value of 1.3 ng/ml.<br />

Coincubation of cells with EGF and 1 ng TGF~ml enhanced greatly <strong>the</strong><br />

inhibitory action of EGF on FSH-induced inhibin production (ID 50<br />

< 0.1<br />

ng/ml).<br />

It is concluded that: (a) TGF~ directly stimulates inhibin<br />

production by rat granulosa cells and <strong>the</strong> combined effect with FSH was<br />

largely additive, (b) <strong>the</strong> inhibitory effect of EGF on FSH-induced<br />

inhibin production was enhanced by TGF~, (c) individual members of <strong>the</strong><br />

TGF~/inhibin gene family regulate ovarian function, not only by direct<br />

action on follicle cells but also indirectly in influencing <strong>the</strong><br />

production rate of o<strong>the</strong>r members of that family.<br />

(1) Skinner, M.K., Keski-Oja, J., Osteen, K.G. and Moses, H.L. (1987)<br />

Endocrinology 121:786-792.<br />

(2) Dodson, W.C. and Schomberg, D.W. (1987) Endocrinology 120:512-516.<br />

(3) Knecht, M., Feng, P. and Catt, K. (1987) Endocrinology<br />

120:1243-1249.<br />

(4) Zhiwen, Z., Carson, R.S., Herington, A.C., Lee, V.W.K. and<br />

Burger, H.G. (1987) Endocrinology 120:1633-1638.


28<br />

SUPEROVULATION IN PUBERTAL HEIFERS IMMUNIZED AGAINST OVINE INHIBIN<br />

PURIFIED BY MONOCLONAL ANTIBODY AFFINITY CHROMATOGRAPHY<br />

1, 2 3 1<br />

B.M. Bindon, T~OIShea, K. MiYimoto, M.A. Hillard,<br />

L.R. Piper, R.D. Ne<strong>the</strong>ry and G. Uphill<br />

2 CSIRO Division of Animal Production, Armidale<br />

De~artment of Physiology, University of New England, Armidale<br />

Gunma University School of Medicine, Gunma 371, Japan<br />

Increased ovulation rate has been observed in cattle immunized<br />

against ovine follicular fluid proteins (1). To explore this fur<strong>the</strong>r,<br />

crossbred beef heifers aged 10 months and weighing an average of 216 kg<br />

were immunized against ei<strong>the</strong>r ovine serum albumin (Group C; n=8) or<br />

ovine .inhibin (Group I, n=8) purified from follicular fluid using a<br />

monoclonal antibody against bovine inhibin (2). This procedure<br />

resulted in an approximate 300-fold enrichment of <strong>the</strong> inhibin. The<br />

animals were injected at subcutaneous and intramuscular sites with<br />

approximately 350 JIg protein (<strong>for</strong> each immunogen) mixed with Freund's<br />

complete adjuvant on Days 1, 25 and 65 of <strong>the</strong> experiment. Only one<br />

heifer had shown oestrus when <strong>the</strong> experiment began. Laparoscopic<br />

ovarian examinations were made on Days 57 and 87. On Day 57, three<br />

heifers from Group C had ovulated, all with one ovulation while four<br />

from Group I had ovulated, with ovulation rates of 3, 2, 1 and 16.<br />

Ovarian data on Day 87, which is 22 days after <strong>the</strong> third<br />

immunization are shown in Table 1. Inhibin-immunized heifers had<br />

substantially higher ovulation rates and follicular development.<br />

Table 1. Ovulation and follicle development of heifers immunized<br />

against ovine serum albumin (Group C) or ovine inhibin (G~oup I)<br />

Mean + S.E.<br />

Live-<br />

Group weight No. No. with Follicle % INH<br />

(n=8) (kg) ovulated >1 ovulation Ov. rate score* binding<br />

C 258+14 4 0 1.0+0 10.3+ 1.9 0<br />

I 260+7 7 7 11.6+3.9 44.3~14.7 19.8<br />

(range-2-32)<br />

* Sum of follicle diameters (mm) (1 cm follicle = score 10)<br />

Heifers from Group I showed measurable plasma binding of <strong>the</strong><br />

iodinated inhibin preparation from Day 38 and reached peak binding (13%<br />

to 27% binding with 100 pI of 1/500 plasma dilution) by Day 87. Plasma<br />

from Group C heifers did not show detectable binding of inhibin.<br />

These results confirm <strong>the</strong> importance of inhibin as a feed-back<br />

regulator of ovarian function in <strong>the</strong> bovine and promise useful<br />

techniques <strong>for</strong> superovulation in this species.<br />

(1) Cummins, L.3., O'Shea, T. and Bindon, B.M. (1986). Proc. Aust.<br />

Soc. Reprod. BioI. 18: 39.<br />

(2) Miyamoto, K., Hasegawa, Y., Fukuda, M. and Igarashi, M. (1986).<br />

Biochem. Biophys. Res. Com. 136: 1103-1109.<br />

29<br />

INHIBIN IN THE<br />

SHEEP OVARIAN CYCLE<br />

J.K. Findlay, I.J. Clarke, H. Quigg, S. Katsahambas, P. Juhola,<br />

M. de Blasiis and B. Doughton<br />

Medical Research Centre, Prince Henry's Hospital Melbourne,<br />

Victoria, 3004.<br />

Inhibin is an ovarian glycoprotein hormone consisting of 2<br />

dissimilar, disulphide-linked subunits, termed ~ and S, which inhibits<br />

<strong>the</strong> production and/or secretion of pituitary gonadotrophins,<br />

preferentially FSH. According to <strong>the</strong> inhibin hypo<strong>the</strong>sis, <strong>the</strong>re should<br />

be (a) higher concentrations of inhibin in follicular fluid and<br />

ovarian venous plasma than in peripheral plasma, and (b) a reciprocal<br />

relationship between peripheral concentrations of inhibin and FSH<br />

during <strong>the</strong> ovarian cycle. We examined <strong>the</strong>se hypo<strong>the</strong>ses in <strong>the</strong> ewe<br />

using a radioimmunoassay developed recently <strong>for</strong> ovine inhibin (1).<br />

Inhibin concentrations are expressed as nl equiv. ovine follicular<br />

fluid standard/ml plasma or follicular fluid.<br />

To examine inhibin release from <strong>the</strong> ovary, 6 Corriedale ewes on<br />

days 12-13'of <strong>the</strong> cycle were subjected to midventral laparotomy under<br />

general anaes<strong>the</strong>sia. Paired jugular and ovarian venous blood samples<br />

were taken immediately be<strong>for</strong>e and 10 min after cauterizing <strong>the</strong> visible<br />

follicles on one ovary. Where possible, follicular fluid was also<br />

harvested be<strong>for</strong>e ablation of follicles. Be<strong>for</strong>e cautery, <strong>the</strong> mean +<br />

s.d. (n) plasma inhibin levels were 130 + 72 (5) in <strong>the</strong> ovarian veIn<br />

and 39 + 9 (6) in <strong>the</strong> jugular vein, a ratio of 3.37 + 1.90 (5). The<br />

follicular fluid concentration was 936000 + 371000 (9 follicles),<br />

approximately 7400-fold higher than ovarian venous plasma. After<br />

cautery, inhibin in ovarian venous plasma fell to 65 + 20% (44-95%<br />

range) of control, with little or no change in jugular concentrations<br />

(97 ~ 10%; 83-114% range).<br />

To examine <strong>the</strong> relationship between peripheral inhibin and FSH, 4<br />

Corriedale ewes (days 8,-12) were treated with prostaglandin and bled<br />

via <strong>the</strong> jugular vein twice daily across <strong>the</strong> ensuing 2 episodes of<br />

estrus and daily during <strong>the</strong> luteal phase (days 3-15). Plasma samples<br />

were assayed <strong>for</strong> LH, FSH and inhibin. During <strong>the</strong> luteal phase of <strong>the</strong><br />

cycle of all 4 ewes, 'inhibin concentrations varied in waves lasting<br />

several days and <strong>the</strong>re was an inverse relationship between inhibin and<br />

FSH, with <strong>the</strong> onset of a rise in FSH preceding that of inhibi~ by 1-2<br />

days. At <strong>the</strong> onset of luteolysis, FSH and inhibin levels decreased<br />

toge<strong>the</strong>r. During <strong>the</strong> follicular phase nadir in FSH, 3/4 ewe~ had<br />

increasing inhibin concentrations which were terminated by tre LH<br />

surge. The second FSH peak followed this decrease in inhibin. The<br />

fourth ewe did not exhibit a rise in inhibin prior to <strong>the</strong> LH surge.<br />

We conclude that (a) <strong>the</strong> ovarian vein is a major route of inhibin<br />

secretion by large follicles, and (b) inhibin and FSH are inversely<br />

related during <strong>the</strong> ovarian cycle except during <strong>the</strong> follicular phase<br />

when we hypo<strong>the</strong>size that LH ra<strong>the</strong>r than FSH stimulates inhibin<br />

production.<br />

(1) Findlay, J.K., Quigg, H., Juhola, P., Katsahambas, S., Clarke,<br />

I.J., Dough ton , B. and Robertson, D.M. (1988) Proc. 70th Ann.<br />

Meet. Endocr. Soc. USA, Abstr.488.


30<br />

LACK OF EXPRESSION OF INHIBIN GENES IN OVINE CORPORA LUTEA<br />

R~J.<br />

Rodgers, S.J. Stuchbery and J.K. Findlay<br />

Medical Research Centre, Prince Henry's Hospital, St Kilda Road,<br />

Melbourne, Victoria, 3004.<br />

Pituitary FSH secretion is regulated by <strong>the</strong> gonadal hormone<br />

inhibin, a glycoprotein composed of two subunits, «and ~) each <strong>the</strong><br />

product of a separate gene. It was originally postulated that in<br />

females inhibin would be produced by ovarian follicles, however, <strong>the</strong><br />

recent detection of inhibin mRNA in human corpora lutea (1) and o<strong>the</strong>r<br />

more circumstantial evidence in <strong>the</strong> sheep (2) suggest that ovine<br />

corpora lutea (CL) may also produce inhibin.<br />

To investigate this possibility Nor<strong>the</strong>rn RNA blotting was per<strong>for</strong>med<br />

on RNA extracted from two pools of ovine follicles «4 mm and >6 mm in<br />

diameter) and CL from both cyclic (n=7) and pregnant (n=6; foetal<br />

crown-rump length 90 to 270 em) animals killed at an abattoir. Total<br />

RNA (25 ~g) was subjected to electrophoresis through agarose/<br />

<strong>for</strong>maldehyde gels and <strong>the</strong>n electroblotted onto Biodyne A membrane.<br />

Prehybridizations and hybridizations were carried out at 60 0 C in<br />

4xSSC, 10 x Denhart's solution, 0.5% SDS, 1 mM EOTA and 100 ~g salmon<br />

sperm DNA/ml <strong>for</strong> eDNA probes and at 60 0 C in 50% <strong>for</strong>mamid, 5xSSPE, 5 x<br />

Denhart's splution, 0.1% 50S and 100 ~g DNA/ml <strong>for</strong> RNA probes l (3). The<br />

cDNA probes were bovine ~ inhibin (720 base SphI-SmaI fragment; 4),<br />

and bovine cholesterol side chain cleavage (SCC) cytochrome P-4S0 ·(890<br />

base EcoRI-Pvu II fragment; 5) as a positive control. An RNA probe<br />

was made from pGEM containing an insert of bovine ~A inhibin (320 base<br />

PstI - RsaI; 4). Stringency washes were in O.lxSSC at 4SoC <strong>for</strong><br />

cytochrome P-450 scc<br />

' 50 C <strong>for</strong> ~ inhibin and 60 0 C <strong>for</strong> ~ inhibin.<br />

Both ~ and ~ inhibin mRNA were readily detected in <strong>the</strong> pool of<br />

follicles < 4 mm in diameter but not in <strong>the</strong> pool of larger follicles<br />

and not in any of <strong>the</strong> CL. Cytochrome P-450 mRNA was readily<br />

detected in <strong>the</strong> pool of large follicles andsrg all of <strong>the</strong> CL. It is<br />

not known why inhibin mRNA was undetectable in <strong>the</strong> pool of larger<br />

follicles but it is highly likely that <strong>the</strong>se follicles were partially<br />

atretic as cytochrome P-450 mRNA levels were high. However it is<br />

clear that ovine CL do not ~~~ress inhibin genes, consistent with<br />

bovine CL (6) but not with human CL (1). Interestingly ovarian<br />

inhibin gene expression in CL of <strong>the</strong>se three species appears to be<br />

linked to expression of cytochrome P-450 aromatase activity.<br />

(1) Davis, S.R. et al. (1987) J. Endocr. 115:R21-R23.<br />

(2) Tsonis, e.G. et al. (1988) J. Endocr. 116:R3-R5.<br />

(3) Manniatis, T. et al. "Molecular Cloning", Cold Spring Harbor<br />

Laboratory. 1982.<br />

(4) Forage, R.G. et al. (1986) Proc. Natl. Acad. Sci. USA<br />

83:3091-3095.<br />

(5) John, M.E. et al. (1984) Proc. Natl. Acad. Sci. 81:5628-5632.<br />

(6) Rodgers, R.J. et al. (1987) Proc. Aust. Soc. Reprod. BioI. 19:117.<br />

31<br />

IMMUNIZATION OF EWES WITH INHIBIN PREPARATIONS OF INCREASING PURITY<br />

T. O'Shea 1 , B.M. Bindon 2 , J.K. Findlay3, ~.A. Hillard 1 , L.R. Piper 2<br />

and K. Miyamoto<br />

~Department of Physiology, University of New E~gland, Armidale,<br />

CSIRO, Division of Animal Production, Armi~ale, Medical Research<br />

Centre, Prince Henry's Hospital, Melbourne, Department of Obstetrics<br />

and Gynecology, Gunma University School of Medicine, Gunma 371, Japan.<br />

Immunization of ewes with an inhibin-enriched fraction from<br />

bovine follicular fluid (bFF) results in an increased ovulation rate<br />

(OR). To confirm that <strong>the</strong> effects (1) were due to inhibin, ewes were<br />

immunized with inhibin preparations (from bFF) of increasing purity.<br />

bPPI (enriched 20-fold) was prepared as preViously described<br />

(1). bMPI (enriched 200-fold) was prepared using a monoclonal<br />

antibody agAinst bovine inhibin (2). bMPI 2<br />

(enriched 1000-fold) was<br />

prepared from bMPI 1<br />

using antibodies raised in ewes immunized with<br />

steer serum. Groups (n=20) of Merino ewes were immunized (using<br />

Freund's adjuvant) with bovine serum albumin (bsa; 2.25 mg protein on<br />

Days 0, 30 and 60), bPPI (2.25 mg on Days 0, 30 and 60), bMPI 1<br />

(0.2<br />

mg on Days 0 and 30), or bMPI 2<br />

(0.1 mg Days 0 and 30). Laparoscopic<br />

ovarian examinations were made on Days -14, 21, 51, 79 and 105.<br />

Means ± sem of OR data are presented in Table 1. Immunization<br />

with <strong>the</strong> purer preparations increased OR after two injections, and<br />

resulted in a transient increase (P < 0.01) in plasma FSH at this<br />

time (bsa, 1.4 ± 0.1 ng/ml plasma; bPPI, 1.7 ± 0.1; bMPI 1<br />

, 2.0 ± 0.2;<br />

bMPI 2<br />

2.3 ± 0.3). Inhibin antibody titres were greater with <strong>the</strong><br />

purer preparations (Day 40; bPPI, 2 ± 0.5% specific binding at 1:500<br />

dilution; bMPI 1<br />

, 19 ± 3%; bMPI 2<br />

, 20 ± 4%). Inhibin antibody titre<br />

was correlated with OR (Day 40; N= 77, r = 0.44, P < 0.01).<br />

Table 1<br />

Ovulation rate following immunization (means ± s. e.m.)<br />

Day<br />

Group 21 51 79 105<br />

bsa 1.7±0.1 1.6 ± 0.1 1.6 ± 0.2 1.1 ± 0.1<br />

bPPI 1.7 ± 0.2 2.2 ± 0.6 2.0 ± 0.5 1.2 ± 0.1<br />

bMPI 2.1 ± 0.3 3.8 ± 0.6 n 3.1 ± 0.9 2.3 ± 0.5*<br />

bMPI 1<br />

2<br />

1.8 ± 0.1 5.2±1.0 n 2.5 ± 0.4· 2.2 ± 0.6<br />

• P < 0.05, .* P < 0.01.<br />

Significantly different from bsa group<br />

These data are consistent with <strong>the</strong> postulate that <strong>the</strong> results<br />

with crude preparations (1) were due to <strong>the</strong>ir inhibin content.<br />

(1) Cummins, L.J. et al., J. Reprod. Fert. (1986),11: 365-372.<br />

(2) Miyamoto, K. et al., Biochem. Biophys. Res. Commun. (1986),<br />

.13Q.: 1103-1109.


32<br />

INFLUENCE OF SHEARING STRESS ON OESTRUS AND OVULAnON<br />

RA.Parr, LF.Davis, and M.L.Phillips*<br />

Animal Research Institute, Werribee, 3030 and *Dookie Agricultural College,<br />

Dookie,3647.<br />

Shearing is associated with severe stress (1) and can delay <strong>the</strong> onset of<br />

oestrus in ewes shorn just prior to mating (2). In <strong>the</strong> study reported here, we<br />

investigated <strong>the</strong> effect of shearing on <strong>the</strong> incidence of behavioural oestrus and<br />

ovulation rate in Merino ewes.<br />

Mature Merino ewes, (n=212) were given intravaginal sponges (1ntervet)<br />

<strong>for</strong> 12 days during September. Sponges were withdrawn on <strong>the</strong> day of shearing<br />

and all ewes were injected (im) with 400 iu of PMSG (Heriot,Aust.). Ewes had<br />

been randomly allocated to ei<strong>the</strong>r a shorn or an unshorn group. Unshorn ewes<br />

were crutched and were joined toge<strong>the</strong>r with shorn ewes and 5% fertile rams.<br />

During <strong>the</strong> expected period of mating, a group of 50 shorn ewes were isolated<br />

with rams <strong>for</strong> a 6h period. Mated ewes and a sample of 30 unmated, shorn<br />

ewes were endoscoped one week after shearing and, 2 days later, were given a<br />

prostaglandin (1C1,Aust.) injection (im) and 400 iu PMSG. At this stage, shorn<br />

and unshorn ewes were separately joined with 5% fertile rams. All ewes were<br />

endoscoped one week later.<br />

Table L Number and (percentage) of shorn and unshorn ewes detected in<br />

oestrus and mean (+/- sem) ovulation rates (OR) of ewes at fIrst and second<br />

oestrus.<br />

GROUP 1st OESTRUS 2nd OESTRUS<br />

n (%) OR n (%) OR<br />

SHORN 5 a (4.7) . L9(0.18t 76 b (71.7) 2.3(0.21)<br />

UNSHORN . 76b (71.7) 2.1(0.29) 71 b (67.0) 2.8(0.21)<br />

~~ted ewes plus a sample of thirty unmated ewes.<br />

a, ; differ at P2 mm recovered and classified as<br />

above. Correlation coefficients between surface and dissected small<br />

medium and large follicles were 0.77, 0.05 and 0.58 respectively. '<br />

Data from ano<strong>the</strong>r previously reported experiment (2) ill ustrates <strong>the</strong><br />

use of <strong>the</strong> three methods to assess ovarian follicle populations (Table<br />

1) . Endoscopy was carri ed out when <strong>the</strong> group's average postpartum<br />

interval was 81 days (s.d. = 14).<br />

TABLE 1. Follicle populations at 80 days postpartum in ovaries from<br />

cows fed three pr.epartu!J1 diets assessed by endoscopy (E) (n=19), or<br />

surface (OS) and d1ssect10n (00) counts (n=33) following ovariectomy.<br />

Prepartum Small Medium Large<br />

Diet E OS 00 E OS 00 E OS 00<br />

Control 6.3 8b 13.5 8 20.0 1.6 1.9 8.3 8 1.3 0.7 0.4<br />

Supp. 1 9.4 b 19.5 8b 28.5 1.5 1.9 9.5 8 0.6 0.6 0.7<br />

Supp. 2 5.4 8 28.8 b 38.5 1.2 1.2 16.2 b 0.8 0.4 0.4<br />

Values within columns with different superscripts differ (P


34 35<br />

CELLULAR COMPOSITION OF THE CYCLICAL CORPUS LUTEUM OF THE COW<br />

J.D. O'Shea 1 , R.J. Rodgers 2 and M.J. D'Occhi0 3 .<br />

2 1School of Veterinary Science, University of Melbourne, Victoria.<br />

Medical Research Centre, Prince Henry's Hospital, Melbourne,<br />

3 Victoria.<br />

C.S.I.R.O., Division of Tropical Animal Production, Rockhampton,<br />

Queensland.<br />

It is important to know <strong>the</strong> numbers of large and small luteal<br />

cells in <strong>the</strong> bovine corpus luteum (CL) to assess claims (1,2) that<br />

granulosa-derived luteal cells disappear from <strong>the</strong> CL during pregnancy.<br />

These ~laims, based on immunocytochemistry on enzymatically-dispersed<br />

populations of luteal cells, depend in part on <strong>the</strong> assumption that<br />

dispersed populations accurately represent <strong>the</strong> populations in intact<br />

tissue. This study investigates this assumption.<br />

Luteal tissue from 6 CL obtained on Day 12 of <strong>the</strong> oestrous cycle<br />

was studied by ultrastructural morphometry (3). Data (mean ~ s.d.)<br />

are shown below.<br />

Volume density (%6<br />

No. cells/CL x 10 10 3 )<br />

Cell volume (~m x<br />

Large luteal<br />

cells<br />

40.2 + 7.0<br />

51.5 + 15.4<br />

29.6 + 6.3<br />

Small luteal<br />

cells<br />

27.7 +<br />

392.4 +<br />

2.7 +<br />

6.3<br />

135.1<br />

0.4<br />

The CL studied weighed 3.8 + 0.8 g. Large and small luteal cells g<br />

combined occupied 67.9% of <strong>the</strong> luteal tissue. Of a total of 1.5 x 10<br />

cells of all cell types/CL, large luteal cells comprised 3.5% and<br />

small luteal cells 26.7%, a ratio of 1:7.6. Mean volumes of <strong>the</strong> large<br />

and small luteal cells would convert, in spherical <strong>for</strong>m, to diameters<br />

of 34 ~m and 17 ~m respectively.<br />

These estimates of cell diameter agree closely with measurements<br />

from dispersed populations (4). However, numbers of luteal cells/g<br />

luteal tissue as estimated here are 10-fold higher than numbers<br />

counted after tissue dispersion (2) <strong>for</strong> large luteal cells, and 5-fold<br />

higher <strong>for</strong> small luteal cells. We conclude that substantial and<br />

selective losses of luteal cells may occur during enzymatic<br />

dispersion.<br />

(1) Alila, H.W. and Hansel, W. (1984) Biol. Reprod. 31:1015-1025.<br />

(2) Hansel, \1. et al. (1987) Aust. J. Biol. Sci. 40:331-347.<br />

(3) O'Shea, J.D. et al. (1986) J. Reprod. Fert. 76:685-691.<br />

(4) Ursely, J. and Leymarie, P. (1979) J. Endocr. 83:303-310.<br />

SEASONAL FACTORS INFLUENCE THE TIMING OF FIRST PREGNANCY IN<br />

THE TAMMAR<br />

E-M.A.<br />

Bugledich, L.A.Hinds* and P.A.Janssens<br />

Department of Zoology, Australian National University,<br />

Canberra, ACT and * CSIRO Div. Wildlife and Ecology, PO Box<br />

84 Lyneham ACT 2602.<br />

Tammar wallabies emerge from <strong>the</strong> pouch in October at<br />

approximately 10 months of age and females will enter<br />

oestrus and conceive at this time. However <strong>the</strong> resulting<br />

embryo enters diapause at <strong>the</strong> blastocyst stage and remains<br />

in quiescence as is normal in adult females at this time of<br />

year. Following <strong>the</strong> summer solstice reactivation occurs and<br />

birth follows at <strong>the</strong> end of January (1). The aim of this<br />

study was to determine <strong>the</strong> potential of females born late in<br />

<strong>the</strong> reproductive season to produce young in <strong>the</strong> following<br />

year.<br />

Females from two different groups have been checked <strong>for</strong><br />

reproductive status at <strong>for</strong>tnightly intervals from early in<br />

pouch life. Group 1 contained 6 females born in late<br />

January-early February 1987. Permanent pouch exit occurred<br />

when <strong>the</strong> young were 35-38 weeks old (1150-1600 g body<br />

weight). They produced young in February or early March 1988<br />

when <strong>the</strong>y were 53-57 weeks of age (2800-3400 g body weight) .<br />

Group 2 contained 3 females born June 2-4 1987. Permanent<br />

pouch exit was at 34 weeks (600-900g body weight) and young<br />

were produced in early May 1988 when <strong>the</strong>y were 48 weeks of<br />

age (2250-2380 g body weight) .<br />

Thus <strong>the</strong> first blastocyst of female juveniles need not<br />

enter diapause if conception occurs during a period of<br />

decreasing daylength. This study fur<strong>the</strong>r shows that female<br />

tammars less than 2kg body weight can conceive and<br />

subsequently undergo a successful pregnancy.<br />

(1) Tyndale-Biscoe, C.H. and Hawkins J. (1977) In<br />

"Reproduction and 'evolution" ed. J.H. Calaby and C.H.<br />

Tyndale-Biscoe, pp. 245-252. Canberra: Australian Academy<br />

of Science.


36<br />

SUPEROVULATION IN A MACROPODID MARSUPIAL, MACROPUS EUGENII<br />

M.B. Renfree, G.<br />

Shaw and T.P. Fletcher<br />

Department of Anatomy, Monash University, Victoria, 3168<br />

Macropodid marsupials are monovular, so a technique <strong>for</strong> inducing<br />

multiple ovulations to provide cleaving eggs and blastocysts <strong>for</strong><br />

experimental embryological techniques is increasingly needed. Whilst<br />

it is easy to induce multiple follicular development using exogenous<br />

gonadotrophin, ovulation usually fails and <strong>the</strong> ovary remains<br />

hyperstimulated.<br />

In a series of trials in 1979 and 1986-1988, we have tested a<br />

,variety of stimulatory treatments during <strong>the</strong> follicular phase of<br />

animals whose cycle had been initiated by removal of pouch young (RPY).<br />

In 1979, 12 animals were treated with 5 to 125 III PMS (Folligon,<br />

Intervet) on day 23 RPY. Those receiving 5 IU had no stimulation,<br />

whilst 25 III animals were slightly stimulated, with one animal having 4<br />

new CL's. The 6 animals receiving 50 IU PMS or more had grossly<br />

enlarged ovaries with multiple >7 rom diameter follicles at laparotomy<br />

on day 30, but no new CL's and all died within <strong>the</strong> next month. A<br />

fur<strong>the</strong>r 9 animals receiving ei<strong>the</strong>r 50 IU or 75 IU PMS on day 23 and<br />

100 III anti.-PMS (courtesy Dr B.M. Bindon, CSIRO Armidale) on d~y 26<br />

and 5 J.1g LH-RH on day 27 had more controlled follicular stimulation.<br />

One had 3 newCL's and 2 eggs were recovered, while 4 o<strong>the</strong>rs had 2<br />

CL's. In animals treated with 50 IU PMS on day 23, anti-PMS on day 26<br />

and ei<strong>the</strong>r 5 I1g LH-RH (HRF, Ayerst) or 50 IU HCG (Primogonyl,<br />

Organon), 3/4 mated on day 27 and on day 30 at autopsy one RCG treated<br />

animal had 4 uterine blastocysts of 40 cells.<br />

In 1986-88, we measured plasma luteinising hormone (LH) ,<br />

progesterone (P) and oestradiol 173 (E2B) to fur<strong>the</strong>r monitor response.<br />

Eleven animals treated with 50, 25 or 15 IU PMS at day 22, 100 1 anti­<br />

PMS at day 25, and 5 I1g LHRH or 50 IU HCG at day 27 led us to adopt<br />

ei<strong>the</strong>r 15 or 26 IU as <strong>the</strong> optimum dose <strong>for</strong> follicle stimulation<br />

although no ovulation resulted. All animals receiving 50 IU PMS had<br />

grossly elevated concentrations of E2 3 (80-100 pg/ml) while P & LH<br />

concentrations were normal <strong>for</strong> that stage of <strong>the</strong> cycle whereas those<br />

receiving 15 or 25 IU had levels similar to those ~t oestrus (15<br />

pg/ml). In 1988 8 animals were treated with 15 IU PMS on days 22 & 23<br />

100 111 anti-PMS on day 25, and 50 l1g LH-RH at 09.00, 13.00 and 18.00 h<br />

ei<strong>the</strong>r on day 27 (n=4) or day 26 (n=4). Four and 3 blastocysts were<br />

recovered from 2/4 animals in <strong>the</strong> last treatment group.<br />

We conclude that it is possible to superovulate macropodid<br />

marsupials, but that results may be highly variable, and that timing is<br />

critical. Fur<strong>the</strong>r refinements of <strong>the</strong> last treatment groups should<br />

allow us to develop a more reliable method.<br />

-<br />

37<br />

THE EFFECT OF GONADOTROPHIN RELEASING HORMONE AGONJST ON<br />

SHEEP prrUrrARY AND OVARIAN FUNCTION<br />

M. sathanandan, S.K. Walker*, LJ. Clarke**, C.D. Mat<strong>the</strong>ws<br />

Def>C!l!ment of Obstetrics and Gynaeoclogy, University ~ Adelaide<br />

and Department of Agri.cuJt.ure, Rcsedale, 5350 and MRC, Pr:ince Henry's<br />

Hcspital, Melbourne, 3003.<br />

Variation in response to gonadotrophin treatment is a limitation to embryo<br />

collection in <strong>the</strong> sheep. GnRH agonists (GnRHa) have aided synchronous<br />

follicular development in <strong>the</strong> human and this phenomenon may be applied to<br />

domestic species. The aims of this study were (a) to determine <strong>the</strong> time<br />

required <strong>for</strong> pituitary down regulation fallowing GnRHa and (b) to assess <strong>the</strong><br />

effect of PMSG on ovulatory response fallowing GnRHa. Four prC9'esterone<br />

treated Merino ewes were administered Leuprolideacetate (GnRHa) (Abbott;<br />

1mg/ewEV'day) <strong>for</strong> 14 days. GnRH Ohtervet; 50ug iv/ewe) challenge tests were<br />

per<strong>for</strong>med at day 0, day 7 and day 14.<br />

Table:<br />

Peak LH and FSH concentrations (ng/m}) fallowing GnRH challenge<br />

prior to (day 0) and during GnRHa treatment (day 7 and 14). Mean<br />

basalleve1s (n=3) in paren<strong>the</strong>sis.<br />

Sheep Day 0 Day 7 Day 14<br />

no. LH FSH LH FSH LH FSH<br />

1 107.3<br />

(1.0)<br />

9.2<br />

(9.0)<br />

2.8<br />

(1.3)<br />

9.7<br />

(2.5)<br />

2.8<br />

(1.7)<br />

15.9<br />

(9.3)<br />

2 205.0<br />

(2.0)<br />

24.6<br />

(13.4)<br />

4.3<br />

(3.3)<br />

3.6<br />

(3.0)<br />

2.0<br />

(1.4)<br />

15.8<br />

(13.9)<br />

3 113.1 26.6 4.5 10.3 2.1 16.3<br />

(1.4) (11.0) (1.6) (8.8) (1.6) (14.0)<br />

4 45.2<br />

(1.7)<br />

22.4<br />

(14.9)<br />

2.4<br />

(1.4)<br />

10.0<br />

(8.6)<br />

2.1<br />

(1.5)<br />

18.7<br />

(16.3)<br />

Peak LH and FSH responses to GnRH (peak/base1ine ratio) were significantly<br />

rerluced from day 7 compared with Day O. Baseline FSH was elevated at day 14<br />

compared with day 7.<br />

Ovarian response (ei<strong>the</strong>r number of corpora lutea (CL) or CL + large unruptured<br />

follicles) was assessed by Japarcscopy at two dcses of P MSG (1200ID and 24001U)<br />

following 14 days GnRHa treatment (n=12/group). Contro1s received similar<br />

PMSG dcses without GnRHa. GnRHa reduced <strong>the</strong> number of CL (P


38<br />

GALACTOSYLTRANSFERASE ACTIVITY ON SPERM SURFACE AND<br />

IN EPIDIDYMAL PLASMA OF SEVERAL MAMMALIAN SPECIES<br />

Y. Tang, <strong>the</strong> late D. E..Brooks, A.M. Snoswell and B. P. Setchell<br />

Department of Animal Sciences, Waite Agricultural Research Institute,<br />

Glen Osmond, SA 5064<br />

Galactosyltransferase (GalTase) activity is present on mouse sperm surface (1) and in<br />

epididymal plas~a of mouse and rat (1, 2). It has been reported that in mouse <strong>the</strong> sperm<br />

surfa~e GalTase IS <strong>the</strong> sperm receptor <strong>for</strong> <strong>the</strong> zona pellucida (ZP) in sperm-zona binding,<br />

and hIgh sperm surface GalTase activity is related with preferential fertilization ability (1).<br />

Sperm-zona binding is not strictly species specific, ego mouse sperm can bind with rat ZP<br />

(3). There<strong>for</strong>e, it is interesting to see if GalTase can be found on sperm surface of o<strong>the</strong>r<br />

species. The GalTase activity in caudal epididymal plasma (CEP) has also been examined,<br />

because of <strong>the</strong> absoption of soluble epididymal proteins by epididymal sperm.<br />

C~P and sperm v:ere coll~c~ed from rats, rabbits, rams and boars ei<strong>the</strong>r by<br />

punctunng or by cannulatmg <strong>the</strong> epIdidymal duct. Mouse sperm were collected by mincing<br />

<strong>the</strong> tissue (1). The enzyme assay procedures are essentially <strong>the</strong> same asreported (1, 2).<br />

Sperm surface GalTase activity (pmole substrate transferred I 1.5 hr. ) in mouse rat<br />

sheep and pig , ,<br />

Sperm Number xlO-6 0.4 0.8 1.6 3.2<br />

Mouse (1 Group of 24) 11.7 28.5 62.7<br />

Rat (2 Groups of 2) 12.3±2.8 21.9±1.9 33.4±1.3<br />

Ram n=3 10.9±3.5 18.9±10.5 48.2±20.9<br />

Boar n=3 2.2±0.8 6.2±3.7 10.6±1.8<br />

CEP. GalTase activity (pmole substrate transferred I ug protein· 20 min. ) in mouse, rat,<br />

rabblt, sheep and pig<br />

Mouse<br />

n=3(groups)<br />

12.3±4.1<br />

Rat<br />

n=8<br />

5.5±1.7<br />

Rabbit<br />

n=4<br />

9.9±3.9<br />

Ram<br />

n=8<br />

7.1±2.3<br />

Boar<br />

n=8<br />

9.4±2.1<br />

The results show that GalTase is present on sperm surface and in caudal<br />

epididymal plasma in several mammalian species. The enzyme activity was lower·in<br />

plg sperm (p


4[1<br />

41<br />

CORRELATION BETWEEN HU}ffiN SPEID1 MORPHOLOGY, ACROSOMES,<br />

ACROSIN AND FERTILIZATION IN VITRO<br />

De Yi Liu and H W Gordon Baker<br />

Department of Obstetrics & Gynecology, University of<br />

Melbourne and <strong>Reproductive</strong> <strong>Biology</strong> Unit, Royal Women's<br />

Hospital, Melbourne, VIC 3053<br />

The human sperm acrosome reaction is important <strong>for</strong> zona<br />

binding and penetration of human oocytes both in vivo and in<br />

vitro. Acrosin, a neutral proteinase, is one of <strong>the</strong> most<br />

important acrosomal enzymes. In order to determine whe<strong>the</strong>r <strong>the</strong><br />

proportion of sperm with normal intact acrosomes, and acrosin<br />

levels are related to fertility, sperm samples remaining after<br />

preparation <strong>for</strong> in vitro fertilization (IVF) in 119 treatments<br />

were studied and related to <strong>the</strong> proportion of oocytes which<br />

fertilized in IVF. Sperm normal intact acrosomes in semen and<br />

in insemination medium were examined by staining with<br />

fluorecein isothiocyanate conjugated pisum sativum agglutinin<br />

(FITC-PSA) according to <strong>the</strong> method described previously. 1,2<br />

Acrosin activity of sperm in semen was determined by a<br />

spectrophotometric method. 3 Sperm concentration in semen and<br />

in insemination medium, motility and motility index, viability<br />

(% live) and normal morphology in semen were also examined by<br />

standard methods.<br />

The results showed that acrosin levels were only<br />

correlated with motility index (p < 0.05) and proportion of<br />

sperm with normal intact acrosomes (p < 0.05). The percentage<br />

of sperm with normal intact acrosomes in semen was strongly<br />

correlated with motility (p < 0.001), motility index (p<br />

< 0.001), viability (p


SEMINAL<br />

42<br />

PlASMA RErINOLr-BINDING PROI'EIN AS AN INDEX OF<br />

HUMAN SERIOLI CELL FUN:TIOO<br />

Ann J Conway, Lyn 11 Boylan, *Margaret Wocx:i, David J Handelsrran<br />

DE:;partment of l1edicine, University of Sydney and Andrclogy Unit &<br />

*DE:;partment of Biochemistry, Royal Prince Alfred Hospital, Sydney<br />

Sertoli cells syn<strong>the</strong>size vitamin A-binding proteins in-vivo and<br />

<strong>the</strong>se ITB.y be involved in <strong>the</strong> essential requirement <strong>for</strong> vitamin A by<br />

spenre.togenesis. There<strong>for</strong>e VJe sought to determine whe<strong>the</strong>r hUITB.n<br />

seminal plaSITB. retinol-binding protein (REP) could represent a direct,<br />

non-invasive ITB.rker of Sertoli cell secretory function in-vivo<br />

comparable with seminal plasma transferrin (TRFN) which is prinlarily<br />

of testicular origin and <strong>the</strong> levels of which are correlated with rate<br />

of spenre.togenesis. There<strong>for</strong>e VJe have studied <strong>the</strong> relationship betvveen<br />

seminal and blocx:i plaSITB. levels of REP and TRFN in 61 men aged 25-49<br />

yr undergoing semen analysis as spenn donors or during infertility<br />

investigations covering a wide range of sperm output and quality from<br />

nonre.l to various degrees of abnonre.l spenre.togenesis (range spenn<br />

density 0-458 MlOO, ootility 0-70%). REP and TRFN were measured by an<br />

imnunoturbidometric assay in spenn-free seminal plasma and concurrent<br />

blocx:i samples.<br />

Number<br />

REP (ug/ml)<br />

(ug/ejac)<br />

TRFN (ug/ml<br />

(ug/ejac)<br />

Number<br />

REP (ug/ml)<br />

(ug/ejac)<br />

TRFN (ug/OO<br />

(ug/ejac)<br />

SPERM DENSITY<br />

10M/ml<br />

20 41<br />

21+6 21+5<br />

84+28 80+25<br />

34+5 * 54+6<br />

118+18 * 178+20<br />

MarILE SPERl'1<br />

10M/ml<br />

22 39<br />

20+6 21+6<br />

83+25 80+26<br />

33+4 55+7<br />

118+17 181+21<br />

(Mean+SEM,<br />

MOI'ILITY<br />

50%<br />

36 25<br />

26+7 12+3<br />

105+31 47+12<br />

50+7 44+4<br />

162+20 1 53+23<br />

PLASMA FSH<br />

nUll<br />

17 20<br />

24+10 14+4<br />

81+32 62+23<br />

58+13 38+5<br />

191+35 129+18<br />

*p 10IU/l<br />

29 6<br />

18+6 23+10<br />

67+22 96+52<br />

49+8 45+7<br />

162+23 161+30<br />

Similar lack of significant differences in REP VJere also observed<br />

<strong>for</strong> total and total nntile sperm output and plaSITB. testosterone<br />

levels. REP and TRFN were correlated in blocx:i (r=O. 285, p=0. 026 ) and<br />

(weakly) in seminal (r=0.200, p=0.122) plaSITB. hOVJever <strong>the</strong>re was no<br />

correlation between blocx:i and seminal plaSITB. levels of TRFN (r=0.054)<br />

or REP (r=O. 006). We conclude that seminal, but not blocx:i, plaSITB. TRFN<br />

is correlated with spenre.togenic output but not sperm rrotility or<br />

testicular endocrine function whereas nei<strong>the</strong>r seminal nor blood plaSITB.<br />

REP are correlated with testicular exocrine, endocrine or sperm<br />

function. This indicates that seminal plaSITB. REP is unlikely to be a<br />

useful ITB.rker of human Sertoli cell function in-vivo.<br />

43<br />

EFFECTS OF MULTIPLE M..ATING ON EPIDIDYMAL SPERM DISTRIBUTION<br />

IN THE BROWN MARSUPIAL MOUSE, ASTUARTII<br />

D.A Taggart & P.D. Temple-Smith<br />

Department of Anatomy, Monash University, Melbourne, Victoria.<br />

The cauda epididymidis in Amechinus stuartii (brown mars~pial mouse) is<br />

characterized by an extremely variable epi<strong>the</strong>lial height ~sso~iat.ed.wIth. a n~rrow a~d<br />

irregular lumen(l) suggesting that sperm stora~e capa~Ity IS limIted. m thIs sp~cles<br />

(2,3). This study examined <strong>the</strong> effects of multIple matmg to test thIS hypo<strong>the</strong>sIs by<br />

assessing sperm distribution in <strong>the</strong> epididymis.<br />

Males were hemicastrated in July be<strong>for</strong>e <strong>the</strong> mating season and sperm<br />

concentrations in various segments of <strong>the</strong> epididymis were determined in groups of<br />

animals given ei<strong>the</strong>r no access (controls) or restricted access to females (caug~t J~ly,<br />

mated three times in August), be<strong>for</strong>e complete castration in August and <strong>the</strong> estImatIOn<br />

of sperm concentrations in <strong>the</strong> rema.ining ep~didymis. T~ese result~ were compared<br />

with a group caught and mated three tImes durmg <strong>the</strong> breedmg season m August.<br />

Epididymal sperm numbers reached a maximum in July (approx. 2 million), with<br />

peak concentrations of s~e:m i~ <strong>the</strong> distal 7aput a~d distal corpus segments. Sperm<br />

distribution along <strong>the</strong> epIdIdymIS changed m relatIOn to <strong>the</strong> date <strong>the</strong> sample~ were<br />

collected. Very few sperm remained in <strong>the</strong> epididymis, even in cont.rol groups, ill late<br />

August (approx. 200,000), indicating a significant loss of sp.erm vIa ~permatorr~ea.<br />

The mean sperm content in <strong>the</strong> caput and proximal corpus regIOns comb~ed, <strong>the</strong> dI~tal<br />

corpus region and <strong>the</strong> caudal region of each epididymidis in August pnor t.o matmg<br />

totalled 494,000 ± 263,000, 717,000 ± 214,000 and 417,000 ± 123,000 respectIvely.<br />

than 300,000 sperm were delivered (per epididymidis) with each matir:g (e~timated by<br />

determining <strong>the</strong> total epididymal sperm number be<strong>for</strong>e and after matmgs m August),<br />

suggesting that epididymal structure may be influencing sperm release.<br />

This study provides important in<strong>for</strong>mation on <strong>the</strong> effects of mating. on ep~didymal<br />

sperm distribution in A. stuartii and suggests that spermatozoa from thIS specIes must<br />

have a remarkabIv high success in <strong>the</strong> female in terms of <strong>the</strong> absolute number and <strong>the</strong><br />

proportion of spermatozoa inseminated.<br />

(1)<br />

(2)<br />

(3)<br />

Taggart, D.A & Temple-Smith, P.D. (1985) Proc. 17th Ann. Conf. Aust. Soc.<br />

Reprod. BioI. p101, Adelaide, Sth. Australia.<br />

Bed<strong>for</strong>d et al (1984). Proc. R. Soc. Lond. B221,221-233. .<br />

Taggart, D.A & Temple-Smith, p.o. (1988). Cell Tiss. Res. (submItted).<br />

Less


44<br />

ULTRARAPID FREEZING OF 2-CELL MOUSE EMBRYOS<br />

Shaw 3.M.<br />

and Trounson A.a.<br />

Centre <strong>for</strong> Early Human Development, Monash Medical Centre, Monash<br />

University, Clayton Vic. 3168.<br />

The ultrarapid embryo freezing technique developed by Trounson et<br />

at. (1,2) <strong>for</strong> early cleavage stages, gave promising results with<br />

2-cell mouse embryos (up to 53% blastocysts and 26% fetuses), but<br />

<strong>the</strong>re was scope <strong>for</strong> improvement.<br />

They used cryoprotectant solutions containing up to 4 M Dimethylsulfoxide<br />

(DMSO), and 0.5 or 0.25 M sucrose in Dulbecco's phosphate<br />

buffered saline with 4 mg/ml bovine serum albumin (BSA). Embryos were<br />

exposed to this solution in a dish <strong>for</strong> 1 minute, <strong>the</strong>n loaded into<br />

plastic insemination straws containing 3 beads of cryoprotectant<br />

solution. Two minutes later <strong>the</strong> straw was plunged directly into<br />

liquid nitrogen.<br />

We have modified this technique and are acheiving excellent<br />

results in vitro and following transfer of intact embryos to<br />

pseudopregnant recipients (Table 1). The development of <strong>the</strong> ultrarapidly<br />

frozen and thawed embryos in vitro is not statistically less<br />

than <strong>the</strong>ir non-frozen controls in <strong>the</strong> 4.5 M DMSO group.<br />

TABLE 1.<br />

Development in vivo and in vitro of slow cooled, and<br />

ultrarapidly frozen 2-cell embryos.<br />

BLASTOCYSTS/TOTAL N. FETUSES/N. TRANSFERRED<br />

Frozen-thawed Solution contr. Frozen-thawed Control<br />

N (%) N (%) N (%) N (%)<br />

slow cool 43/60 (71 )** 46/50 (92) 92/130 (71) 62/84 (74)<br />

3.0M UR. 242/280 (86)* 151/163 (92) 106/157 (68) 124/179 (69)<br />

4.5M UR. 257/280 (92) 143/150 (95) 48/79 (61) 29/47 (62)<br />

* Lower than own control P


2.4<br />

46<br />

ALLOCATION OF CELLS TO INNER CELL MASS AND TROPHECTODERM IN<br />

MOUSE EMBRYOS BIOPSIED AT THE 4-CELL STAGE.<br />

Gino Somers and Leeanda Wilton.<br />

Centre <strong>for</strong> Early Human Development, Monash Medical Centre, Clayton.<br />

It has been proposed that pre-natal diagnosis could be carried out<br />

in <strong>the</strong> pre-implantation embryo by biopsy of a single cell. However, it<br />

has recently been shown that such embryos have a significantly reduced<br />

implantation rate (1). In <strong>the</strong> developing embryo, <strong>the</strong> first<br />

differentiative step occurs at <strong>the</strong> transition from morula to blastocyst,<br />

with <strong>the</strong> emergence of <strong>the</strong> inner cell mass (ICM) and trophectodermal (TE)<br />

lineages. As <strong>the</strong> ICM gives rise to all tissues of <strong>the</strong> foetus proper, it<br />

is critical that <strong>the</strong> number of cells in this lineage is sufficient <strong>for</strong><br />

successful implantation and development. Consequently, we have examined<br />

<strong>the</strong> possibility of a disturbed ICM:TE ratio being responsible <strong>for</strong> <strong>the</strong><br />

decreased implantation rate of mouse embryos biopsied at <strong>the</strong> 4-cell<br />

stage.<br />

Four-cell embryos were flushed from F1 (CBA x C57) mice which had<br />

been superovulated with sIU PMS followed 48 hours later by 5IU hCG. The<br />

zona pellucida was removed by brief exposure to aCid-Tyrode's solution<br />

(pH 2.5) and <strong>the</strong> embryos cultured in Ca-H-/~1g-H--free medium 2 U12) <strong>for</strong><br />

60-90 minutes. Gentle pipetting with a flame-polished micropipette<br />

dislodged one blastomere and produced 3/4 embryos. Controls included<br />

untreated embryos and embryos in which one cell was dislodged and<br />

immediately re-aggregated. After 42 hours in culture (96 hours post-hCG)<br />

in medium' 16 (M16) in 5% CO, embryos had reached <strong>the</strong> early blastocyst<br />

stage and were fixed using -<strong>the</strong> fluorochromes propidium iodide and<br />

bisbenzimide to stain <strong>the</strong> TE or IC~l respectively (2). The number of<br />

cells allocated to each lineage was counted.<br />

EMBRYO NUMBER CELL N~ffiER(~mAN ± S.D.)<br />

TREATMENT OF EMBRYOS TOTAL ICH TE RATIO ICH:TE<br />

UNTREATED 37 28.9+4.7 11.9+2.6 17.0+3.1 0.70<br />

RE-AGGREGATED 37 26.9+4.0 1l.0+2.2 15.9+2.9 0.69<br />

BIOPSIED (3/4) 35 20.3±4.3 7.3±2.0 13.0±3.3 0.56*<br />

*Significantly different to re-aggregated embryos, P


48<br />

IN VITRO DEVELOPMENT OF GOAT PREIMPLANTAnON EMBRVOS IN<br />

COCULTURE WITH OVIDUCT EPITHELIAL CELLS<br />

D. SAKI1:ment of o~etrics and Gynaecology, University of Adelaide, Box 498<br />

GPO, Adelaide, and Turret&.-ld RESearch Centre, Department of AJTiculture,<br />

R03eda1=, SA 5350.<br />

Pronuc1-ar embrycs are required <strong>for</strong> studies on <strong>the</strong> microinjection of <strong>for</strong>eign<br />

gens; and on embryogenesis. A previou:3limitation to such studiES in sheer; was<br />

<strong>the</strong> availability of a culture system which \tHS able to support <strong>the</strong> development<br />

of embryos of this age. In our laboratory, h.igh rate of development of<br />

pronuclear embryos (50-80 % blastocysts) are now regularly obtained in<br />

syn<strong>the</strong>tic oviductuuid medium (SOFM) (1) without soma::ic cell support. This<br />

atBtract prESents in<strong>for</strong>mation on <strong>the</strong> viability of embryos after 1, 3 or 5 days of<br />

culture.<br />

Pronucleac embrycs ~',1ere cultursJ in SOul droplets of mediu m under<br />

paraffin oil in an atmosphere of 5 % CO), 5 % 02 and 90 % N 2<br />

. He::tt inactivated<br />

hum3.n serum (20 % concentration) 1';3.3- <strong>the</strong> preferred protein source and <strong>the</strong><br />

NaHC 03 concentration of 25m M was par-ely rebJIace:1 \o,'ith Hepes at<br />

concentrations of 12.5-17.5mM. 'T'empe..rature of <strong>the</strong> incub3.tor was 38°C,<br />

humidity 93 % and pH of medium was calailatei.l to be 7.47±0.01l.<br />

Viability after culture was as..sessed by transfer of embryos to synchron.i~2d<br />

c.::ci;Jients which were slaught:~red on day 13 of pre;Jnancy and <strong>the</strong> nll11 :.:2c-:Jc<br />

,;::1Dngated conC89b~ses deter mined.<br />

C3.t>le 1.<br />

Effect of cult:uce in SOFM on <strong>the</strong> viability of oronuclJ":;"I.r'".:,:'n;xy-:J;;.<br />

;::; ay; ill culture<br />

Control 1 3 5<br />

,0. embryo transferred 36 42 51 36<br />

Jo. recovered 35 40 51 35*<br />

10. elon'jatej 34 39 48 19<br />

(% ) (97.1) (97.5) (94.1) (54.3)<br />

* ill elongated concer>tusES obtained from ern~ry:::B which had<br />

com menced blastulation at <strong>the</strong> time of transfec (19/22 v. 0/13).<br />

Th,:; results indi':::ate that SOF M can be used <strong>for</strong> <strong>the</strong> cllltJre of pr-onuclear<br />

e :nrXy03 f:x 3t least 3 days without jeopanlizinj 'liability. Bmbryos which had<br />

not com mencei blastul3.tion at <strong>the</strong> time of transfer (day-5 'jroup) f;ill.e::1 to<br />

de-Jelop beyond <strong>the</strong> Jila.:,""tocy.-3t stage possibly due to an asynchrony between <strong>the</strong><br />

developmental age of <strong>the</strong> embryo and <strong>the</strong> stage of <strong>the</strong> recipient's repro::1uctive<br />

cycle. Despite this, <strong>the</strong> culture .system de3cribec1 provides a simple and easy<br />

means of asse'""~g emtlryo 'Viability.<br />

(1) Tervit, H.R., Whittingham, D.G. and Rowson, L.E.A. (1972).<br />

,J. Repro::1. FeLt. 30: 493-497.<br />

Supported by A['1 LROC.


50<br />

THE ANTIGESTAGENS RU486 AND ZK299 ARE NOT BOUND BY THE UTERINE<br />

PROGESTERONE RECEPTOR OF THE TAMMAR WALLABY, MACROPUS EUGENII<br />

51<br />

PROTEIN SECRETION PATTERNS OF SEPARATED OVINE ENDOMETRIAL CELLS<br />

CULTURED IN DUAL ENVIRONMENT CHAMBERS<br />

~__Uetcher and D.R. Blandon R.A. Chernx and J.K. Findlay<br />

Department of Anatomy, Monash University, Victoria, 3168 Hedical Research Centre, 110nash l'kdical Centre, Prillce !h=llry'<br />

Hospital Campus, Melbourne, Victoria, ]004.<br />

H[4813 (Mifr:pristone) and ZK299 are structurally rel.ated syn<strong>the</strong>Lic<br />

st.eroids which have anti-progest.erone and anti-glucocorticoid<br />

properties. R[486 binds to <strong>the</strong> uterine progesterone receptor of rats,<br />

rabbits, dogs, monkeys and humans with an affinit.y eCjual to, or greater<br />

than, progesterone (1,2). We have devc-doped an a~,SHY to measure <strong>the</strong><br />

proge~d.erone receptOl" in cytosol preparations from <strong>the</strong> endometrium of<br />

<strong>the</strong> talluuar, using [3H]-ORG 2058 as <strong>the</strong> specific competitor. Binding<br />

was measun'd by incubat.ing different. concentrations of Lhe test<br />

compounds with <strong>the</strong> cytosol preparation and comparing <strong>the</strong> effect on<br />

tutul binding in <strong>the</strong> absence of competit.or. The assay has been<br />

validated using rat and rabhit uterine cytosoL preparations which gave<br />

values consistent. \oJith published recept.or concentrations at similar<br />

stages of <strong>the</strong> reproductive cycle.<br />

Table 1. The effect of competitor concentration on h:inding of (3H]-ORG<br />

2058 to tammar endometrium progesterone ['("ceptors<br />

Prog<br />

I 3<br />

(nM/I)<br />

10 100<br />

Hal n 37 22 10 25<br />

ftahbi t 49 26 11 4 27<br />

TalJUnar n 72 72 51 38<br />

ORG 2058 (nM/l)<br />

1 3 10 100<br />

14 9 8<br />

15 7 :3<br />

30 L7 l5<br />

RU 486 (oM/I) ZK 299 (nM/I)<br />

10 lOO 1000 10 100 1000<br />

40 1 B<br />

68 2 t3<br />

80 T~ 100<br />

Both progesterone and ORG 205H displaced <strong>the</strong> [3H]-OHC; 2058 from<br />

tammar cytosol preparations, but nei<strong>the</strong>r RU486 or ZK299 displaced label<br />

at concenlr


52<br />

THE EFFECTS OF A SPECIFIC PLATELET ACTIVATING FACTOR (PAP) RECEPTOR<br />

ANTAGONIST (SRI 63441) ON SOME ASPECTS OF MATERNAL PHYSIOLOGY<br />

N. R. Spinks and C. O'Neill<br />

Human Reproduction Unit, Royal North Shore Hospital of Sydney, St.<br />

Leonards. NSW, 2065<br />

To test <strong>the</strong> influence of embryo-derived PAF on <strong>the</strong> earliest stages<br />

of implantation, eight week old Quackenbush strain mice were induced to<br />

ovulate with 3 i.u. pregnant mare serum gonadotrophin and human<br />

chorionic gonadotrophin, given 48 h apart, and mated with fertile<br />

males. Animals were injected. intraperitoneally, with 40ug SRI 63-441<br />

(Sandoz. USA) in 200ul Dulbecco's phosphate buffered saline (PBS) or<br />

200ul PBS alone.at 1600 h on day 1 (day of vaginal plug) and 0900 h on<br />

days 2-4 of pregnancy. On day 5, animals were injected intravenously<br />

with O.15ml of a 1% solution of pontamine sky blue. The uteri were<br />

examined. 15 minutes later, <strong>for</strong> <strong>the</strong> presence of blue bands. indicating<br />

<strong>the</strong> site of embryo implantation.<br />

The mean number of implantation sites per uterine horn in <strong>the</strong> SRI<br />

63-441 treated animals (2.17 ~0.69) was significantly (P


54<br />

RESPIRATORY PROPERTIES OF THE MOUSE UTERINE ENDOMETRIUM<br />

DURING EARLY POST-IMPLANTATION PREGNANCY<br />

Barbara Williams and R.N. Murdoch<br />

Department of Biological Sciences, University of Newcastle, NSW 2308<br />

A transient decline in <strong>the</strong> respiratory activity of mouse uterine<br />

endometrial tissue on day 6 of pregnancy was a finding stemming from<br />

recent investigations (1). Although similar responses were earlier<br />

described in <strong>the</strong> rat (2,3), very little work has since been undertaken<br />

to explain <strong>the</strong> regulation of this phenomenon or its importance to <strong>the</strong><br />

maintenance of decidual tissue <strong>for</strong> <strong>the</strong> continued support of embryonic<br />

development. In view of this, <strong>the</strong> present study was initiated to<br />

fur<strong>the</strong>r - investigate <strong>the</strong> respiratory properties of <strong>the</strong> early<br />

post-implantation uterine endometrium.<br />

The O 2<br />

consumption of OS mouse endometrial tissue, measured by<br />

Warburg manometric techniques and expressed in relation to its DNA<br />

content on days 5, 6 and 7 of pregnancy and pseudopregnancy (day 1 :<br />

day of vaginal plug), transiently decreased on day 6 only when <strong>the</strong><br />

uterus had been adequately stimulated to mount a decidual cell<br />

reaction. This was achieved naturally by implanting bIastocysts<br />

during pregnancy, and artificially by <strong>the</strong> introduction of 30 ul of<br />

peanut oil into <strong>the</strong> uterine lumen on <strong>the</strong> afternoon of day 4 of<br />

pseudopregnancy. Adenosine (50-500uM) had no significant effect on<br />

<strong>the</strong> respiratory pattern suggesting that <strong>the</strong> response was not due to a<br />

competition <strong>for</strong> a limited supply of cofactors common <strong>for</strong> both<br />

glycolysis and oxidative phosphorylation. In addition, <strong>the</strong> response<br />

was not effectively altered by supplementing incubation media with<br />

ei<strong>the</strong>r glucose (5mM) or glutamine (1 mM), indicating that endogenous<br />

s~bstrates support respiration at <strong>the</strong>se times. Ho~t.ver, <strong>the</strong> tissue<br />

dlsplayed 14<br />

some capacity to oxidise exogenous [U- C] glucose and<br />

evolved CO 2<br />

in a pattern consistent with that of 0 uptake.<br />

Respiratory quotients suggested that utilizable &ndogenOUs<br />

carbohydrates were rapidly depleted during incubation in vitro and<br />

that, subsequently, lipids were probably utilized as respiratory<br />

fuels. Experiments conducted with rotenone implicated mitochondrial<br />

processes as being responsible <strong>for</strong> <strong>the</strong> 0 consumption of <strong>the</strong> tissue.<br />

However, no significant changes were dete€ted in uterine levels of S ­<br />

hydroxybutyrate to account <strong>for</strong> <strong>the</strong> lIballooninglt of mitochondria that<br />

has been suggested to also occur transiently on day 6 of pregnancy<br />

(4) •<br />

In conclusion, <strong>the</strong> results indicate that <strong>the</strong> transient decline in<br />

respiratory activity of <strong>the</strong> endometrium on day 6 of pregnancy is a<br />

function of decidual cells and involves alterations which are<br />

ultimately exerted at <strong>the</strong> mitochondrial level. Fur<strong>the</strong>r work is<br />

required to elucidate <strong>the</strong> regulatory mechanisms concerned and to<br />

establish <strong>the</strong> extent to which <strong>the</strong> phenomenon is important <strong>for</strong> <strong>the</strong><br />

maintenance of decidualized tissue.<br />

(1) Murdoch, R.N. (1987) Teratology 35, 169-176.<br />

(2) Saldarini, R.J. &Yochim, J.M. (1967) Endocrinology 80, 453-466.<br />

(3) Surani, M.A.H. &Heald, P.J. (1971) Acta Endocrinol.156, 16-24.<br />

(4) Jollie, W.P. & Benscombe, S.A. (1965) Amer.J.Anat. 116, 217-236.<br />

55<br />

DISTRIBUTION OF OESTROGEN RECEPTORS IN THE FEMALE REPRODUCTIVE<br />

TRACT OF THE FLYING FOX PTEROPUS SCAPULATUS<br />

Craig S. Pow and Len Martin<br />

Department of Physiology and Pharmacology, University of<br />

Queensland, St Lucia, Qld 4067<br />

Flying foxes are monovulatory seasonal breeders. Both ovaries<br />

function and are thought to ovulate alternately in successive seasons.<br />

Both horns of <strong>the</strong> uterus function, but preimplantation endometrial<br />

development is limited to <strong>the</strong> cranial tip of <strong>the</strong> horn adjacent to <strong>the</strong><br />

single preovulatory follicle and subsequent corpus iuteum (1). We have<br />

proposed that localised growth is due in part, to preferential<br />

delivery of sex hormones from ovary to ipsilateral uterus via countercurrent<br />

transfer between <strong>the</strong> ovarian vein and coiled ovarian artery,<br />

<strong>the</strong> major blood supply to <strong>the</strong> cranial tip of <strong>the</strong> uterus (2). However,<br />

endometrial growth may also be limited by a restricted uterine<br />

distribution of hormone receptors. A peroxidase anti-peroxidase (PAP)<br />

immunocytochemistry kit (Abbott) was used to explore this possibility.<br />

Two bats· were used: one untreated, one given 5 pg of oestradiol<br />

(Ez) subcutaneously 24 h be<strong>for</strong>e autopsy. Tissue was dissected out,<br />

frozen in liquid Nz, and cryostat sections (10 pm) fixed in 3.7%<br />

<strong>for</strong>maldehyde-PBS (15 min), cold methanol (5 min), and acetone (3 min).<br />

After treatment with blocking reagent (goat serum, 15 min), <strong>the</strong>y were<br />

incubated (30 min) with rat monoclonal antibody H222, which was<br />

raised against human oestrogen receptor (ER), but cross-reacts with<br />

many mammalian ERs (3). ER positive MeF7 breast cancer cells were<br />

treated similarly. As negative controls, MCF7 cells and bat sections<br />

were incubated with normal rat antibody. Goat anti-rat antibody was<br />

used to link primary antibody and PAP complex. Diaminobenzidine was<br />

used as chromogen with haemotoxylin counterstain.<br />

Negative control sections and cells did not stain. In<br />

preparations incubated with H222, endometrial gland and luminal<br />

epi<strong>the</strong>lial nuclei stained intensely throughout each uterine horn.<br />

Stromal and myometrial nuclei also stained, as did those of MCr7<br />

cells. Intensity of staining of individual nuclei varied, but <strong>the</strong>re<br />

was no difference in <strong>the</strong> degree of nuclear staining between Ez primed<br />

and unprimed horns, and no cytoplasmic staining. Overall <strong>the</strong> pattern<br />

is similar to that obtained with H222 in primate uterus (4).<br />

Significantly, cells of <strong>the</strong> vaginal epi<strong>the</strong>lium, which is unresponsive<br />

to oestrogens in Pteropus (5), did not stain.<br />

Since staining occurred along <strong>the</strong> length of each horn, it appears<br />

that limitation of endometrial development is not dictated by receptor<br />

distribution.<br />

(1) Marshall, A.J. (1949) Proc. Linn. Soc. Lond. 161: 26.<br />

(2) Pow, C., &Martin, L. (1987) Proc. ASRB 19: 1~<br />

(3) Greene, G.L., et ai. (1984) J. Ster. Biochem. 20: 51.<br />

(4) McClelland, M.C., et ai. (1984) Endocrinol. 114: 20oi.<br />

(5) Martin, L., et ai. (1987) Aust. Mammal. 10: 115.


56<br />

EFFECTS OF GONADOTROPHINS ON FOLLICLE DEVELOPMENT DURING<br />

PREGNANCY IN THE FLYING FOX PTEROPUS SCAPULATUS<br />

Philip A.<br />

Towers* and Len Martin<br />

Department of Physiology and Pharmacology, University<br />

of Queensland, St Lucia, Qld, 4067<br />

Flying foxes are monotocous seasonal breeders. Both ovaries<br />

function. Ovulation, alleged to alternate from side to side, is not a<br />

simple reflex response to copulation. The 4-6 week breeding season<br />

involves repetitive copulation which continues well into pregnancy.<br />

Over this period preimplantation endometrial growth of <strong>the</strong> duplex<br />

uterus is restricted to <strong>the</strong> tip ipsilateral to <strong>the</strong> single preovulatory<br />

follicle or corpus luteum (CL). Consistent with this, peripheral<br />

oestradiol and progesterone levels remain low until mid-pregnancy when<br />

placental secretion predominates (1). Supplementary ovulations/CLs do<br />

not occur. There are no au<strong>the</strong>ntic records of twin births. Never<strong>the</strong>less<br />

our small series of specimens contains two cases of superfoetation in<br />

which a foetus was found in each horn and appeared to have been<br />

conceived -2 months apart. We <strong>the</strong>re<strong>for</strong>e used gonadotrophins (GNs) to<br />

determine if ovulation could be induced in pregnant Pteropus.<br />

Six mid-pregnant (March) P. scapulatus were used: 4 received 4 iu<br />

pFSH (Calbiochem, USA) s.c. daily <strong>for</strong> 4 consecutive days; 2 were <strong>the</strong>n<br />

killed on d4 while 2 received 250 iu hCG (Intervet, NSW) s.c. on d4<br />

and were killed on d7. As controls, 2 received saline s.c. on d1, and<br />

were killed on days 1 and 7. At autopsy ovaries were fixed, embedded,<br />

sectioned and examined <strong>for</strong> follicular development.<br />

In control animals, ovaries ipsilateral to <strong>the</strong> foetus contained 1<br />

CL and small preantral follicles; contralateral ovaries contained<br />

preantral/small atretic antral follicles. All ovaries contralateral to<br />

<strong>the</strong> foetus, in GN treated bats contained large (>800 ~m) healthy<br />

antral follicles but none were preovulatory, luteinized or had<br />

ovulated. Each ovary ipsilateral to <strong>the</strong> foetus in <strong>the</strong>se animals<br />

contained 1 CL, but none showed any follicular growth.<br />

The failure of GNs to induce ovulation in ovaries contralateral<br />

to <strong>the</strong> foetus contrasts with non-pregnant non-breeding season<br />

P. scapulatus in which <strong>the</strong>y induce multiple ovulations by d4 (2).<br />

This suggests that high circulating levels of placental hormones may<br />

inhibit ovulation. The failure to induce any follicle development in<br />

ovaries ipsilateral to <strong>the</strong> foetus could result from <strong>the</strong> same<br />

inhibitory factors reaching <strong>the</strong> ovary at much higher concentrations<br />

because of preferential transport from uterus to ipsilateral ovary by<br />

counter current transfer. In Pteropus <strong>the</strong> uterine vein runs cranially<br />

fusing with <strong>the</strong> ovarian vein to <strong>for</strong>m a common sinus which completely<br />

encloses <strong>the</strong> ovarian artery <strong>for</strong> much of its path (3). The preferential<br />

inhibition of <strong>the</strong> ipsilateral ovary may reflect mechanisms involved in<br />

<strong>the</strong> alleged alternation of ovulation from season to season.<br />

(1) Towers, P.A., &Martin, L. (1984) Proc. ASRB 16: 9.<br />

(2) Towers, P.A., & Martin, L. (1985) Proc. ASRB 17: 115.<br />

(3) Pow, C., & Martin, L.(1987) Proc. ASRB 19: 100.<br />

*Present address Riverina-Murray Inst. Higher Educat., Wagga, NSW 2650<br />

57<br />

OVINE UTERINE LYMPHATICS: IN VIVO PASSAGE OF INDIA INK FROM<br />

UTERINE SUBSEROSA. MYOMETRIUM AND LUMEN.<br />

R.G. Alders and J.N. Shelton<br />

Department of Immunology, The John Curtin School of Medical Research,<br />

Australian National University, Canberra, ACT.<br />

The results of histological investigations into <strong>the</strong> location of<br />

lymphatic vessels within <strong>the</strong> ovine uterus have b~en equivocal. T~e<br />

work has been conducted in post mortem speclmens and has pald<br />

particular attention to <strong>the</strong> distribution of vessels within <strong>the</strong> various<br />

layers of <strong>the</strong> uterus. However, post mortem injection of dye provides<br />

little indication of <strong>the</strong> ability of uterine lumen contents to enter <strong>the</strong><br />

peripheral lymphoid apparatus in vivo. Given that immunologi:al<br />

responses within lymph nodes are usually dictated by <strong>the</strong> materlal<br />

carried to <strong>the</strong> node by afferent lymphatic vessels and that several<br />

sites which are recognised as immunologically privileged have been<br />

found to have poor or absent lymphatic drainage, it was decided to<br />

investigate <strong>the</strong> ability of a <strong>for</strong>eign substance placed within <strong>the</strong><br />

various layers of <strong>the</strong> uterus to be carried to <strong>the</strong> regional lymph nodes<br />

in vivo.<br />

The' uterus was exposed via a ventral midline laparotomy following<br />

general anaes<strong>the</strong>sia in both pregnant and nonpregnant merino ewes.<br />

Subserosal injections of 80 ~l of sterile india ink (II) were made<br />

through a 30 gauge needle at 6 sites along both uterine horns.<br />

Subserosal injections were made by inserting <strong>the</strong> needle just beneath<br />

<strong>the</strong> outer serosal covering. Lumenal and myometrial injections were<br />

made with <strong>the</strong> aid of a blunt 30 gauge needle inserted into ei<strong>the</strong>r an<br />

interplacentomal or placentomal region of <strong>the</strong> gravid horn. Lumboaortic<br />

and medial iliac lymph nodes (DLN) were collected at necropsy<br />

and fixed in 10 % neutral buffered <strong>for</strong>malin. Specimens were mounted in<br />

paraffin blocks and 10 ~m sections stained with Giemsa.<br />

In <strong>the</strong> majority of cases <strong>the</strong> II injections were made subsequent to<br />

cannulation of a peripheral lymph vessel dr~ining <strong>the</strong> uterus. Cells in<br />

<strong>the</strong> lymph collected post-operatively were examined under <strong>the</strong> light<br />

microscope after cytocentrifuge smears were stained with a modified<br />

Wright-Giemsa stain (Diff Quik; Lab-aids, Narrabeen, N.S.~I.).<br />

Subserosal injection of II resulted in rapid transport (0: 10<br />

mi nutes) to <strong>the</strong>, DLN in pregnant and nonpregnant ewes. Free and<br />

phagocytosed II was evident within <strong>the</strong> subcapsular sinus and medulla of<br />

DLN. The passage of II from deposits within <strong>the</strong> myometrium was less<br />

rapid. In constrast, when II was injected into <strong>the</strong> uterine lumen of<br />

pregnant and nonpregnant ewes no II was evident within DLN up to 2<br />

weeks after deposition. II within <strong>the</strong> uterine lumen was found both<br />

free and within mononuclear cells.<br />

In ewes of various reproductive states <strong>the</strong> DLN displayed an<br />

eosinophilic infiltrate and occasional mast cells within <strong>the</strong> medulla.<br />

A dramatic eosinophilic infiltrate was observed at <strong>the</strong> site of <strong>the</strong> II<br />

deposit in <strong>the</strong> myometrium.<br />

These results indicate that <strong>the</strong>re is effectively a barrier<br />

preventing transport of II from <strong>the</strong> uterine lumen to <strong>the</strong> DLN. This<br />

lack of lymphatic drainage may <strong>for</strong>m part of <strong>the</strong> immunological<br />

mechanisms involved in <strong>the</strong> maintenance of pregnancy.


58 59<br />

The effect of pregnancy on <strong>the</strong> rate of <strong>the</strong><br />

biotrans<strong>for</strong>mation of caffeine<br />

Fatmida Abdi and Irina Pollard<br />

School of Biological Sciences, Macquarie University N.S.W. 2109<br />

Caffeine, a methylxanthine, is consumed in coffee, tea, some cola<br />

soft drinks or in analgesics and o<strong>the</strong>r over-<strong>the</strong>-counter drugs. Perhaps<br />

most importantly, caffeine is consumed by children in chocolate and<br />

cocoa drinks. Until recently caffeine was considered a mild stimulant<br />

and harmless if taken in amounts not exceeding <strong>the</strong> equivalent of 6<br />

cups of coffee per day. Recently, however, adverse effic~s of low<br />

level caffeine intakes has been demonstrated on <strong>the</strong> fetus ' .<br />

The concentration of caffeine and its major metabolites in <strong>the</strong><br />

blood 'and tissues of <strong>the</strong> pregnant rat was measured by HPLC and<br />

compared to <strong>the</strong> non-pregnant rat. It was demonstrated that in <strong>the</strong><br />

pregnant rat <strong>the</strong> biotrans<strong>for</strong>mation and elimination of caffeine was<br />

significantly (P


60<br />

STUDIES OF BINUCLEATE CELLS IN THE PLACENTA OF GOAT, COW AND DEER USING<br />

THE MONOCLONAL ANTIBODY SBU-3<br />

C.S. Lee, K. Gogolin~Ewens, W.R. Mercer, #P. Wooding and M.R. Brandon.<br />

Department of Veterinary Preclinical Sciences, University of Melbourne<br />

and #Department of Cell <strong>Biology</strong>, Agricultural and Food Research Council,<br />

Babraham, Cambridge, England.<br />

The monoclonal antibody, SBU-3, raised against sheep trophoblast<br />

microvilli, which recognises a population of binucleate cells and <strong>the</strong><br />

syncytium in <strong>the</strong> sheep placenta (1) also recognises <strong>the</strong> binucleate<br />

cells in o<strong>the</strong>r ruminant placentae. The present studies investigate<br />

<strong>the</strong> distribution of <strong>the</strong> binucleate cells and <strong>the</strong> subcellular localization<br />

of <strong>the</strong> SBU-3-positive antigen in <strong>the</strong> placentae of <strong>the</strong> goat, cow<br />

and deer.<br />

Ti ssues of p1acentomes and interp1acentoma1 areas were obtained<br />

from goat, cow and deer at different stages of pregnancy. Tissues <strong>for</strong><br />

immunohistochemical studies were fixed in 95% cold alcohol and <strong>the</strong> sections<br />

prepared were stained with <strong>the</strong> indirect immunoperoxidase technique.<br />

All tissues <strong>for</strong> electron microscope immunocytochemical studies<br />

were fixed in 2% glutaraldehyde and ultrathin sections reacted sequentially<br />

with SBU-3 and colloidal gold labelled goat anti-mouse antibody.<br />

In all <strong>the</strong> specimens (goat, cow and deer) studied, <strong>the</strong> appearance<br />

of SBU-3-positive binucleate cells was associated with <strong>the</strong> <strong>for</strong>mation of<br />

chorionic villi-The number of SBU-3-positive binucleate cells increased<br />

dramatically with advancement of pregnancy and <strong>the</strong> most striking<br />

feature was <strong>the</strong> high concentration of SBU-3-positive binucleate<br />

cells at <strong>the</strong> tips of <strong>the</strong> chorionic villi. In <strong>the</strong> goat, <strong>the</strong> syncytium<br />

covering <strong>the</strong> caruncular septa.was stained positively. However, in <strong>the</strong><br />

cow and <strong>the</strong> deer trinucleate cells were found at random in <strong>the</strong> epi<strong>the</strong>lium<br />

covering <strong>the</strong> caruncular septa. Electron microscopy revealed that<br />

gold particles were local ized in <strong>the</strong> granules and <strong>the</strong> Golgi apparatus<br />

of <strong>the</strong> binucleate cells and in <strong>the</strong> granules of <strong>the</strong> syncytia.<br />

It is concluded that <strong>the</strong> syncytia in <strong>the</strong> goat placenta are derived<br />

from <strong>the</strong> SBU-3-positive binucleate cells whereas in <strong>the</strong> cow and deer no<br />

syncytium is <strong>for</strong>med and granule transfer seems to be <strong>the</strong> primary function<br />

of binucleate cell migration. The histological classification of<br />

<strong>the</strong> goat placenta is syndesmochorial while that of <strong>the</strong> cow and deer is<br />

epi<strong>the</strong>liochorial.<br />

(1) Lee, C.S., Gogolin-Ewens, K., White, T.R. and Brandon, M.R. (1985)<br />

Journal of Anatomy ~, 565-576.<br />

OCYTOCIN RECEPTORS<br />

61<br />

IN THE UTERUS OF THE BRUSHTAIL POSSUM<br />

C. Sernia 1 , J. Garci~l, W.G. Thomas 1 and R.T. Gemmel1 2<br />

lDepartment of Physiology and Pharmacology, 2Department of Anatomy,<br />

University of Queensland, St. Lucia, 4067.<br />

The role of pituitary oxytocin (OT) in marsupial parturition is<br />

uncertain. However, evidence that <strong>the</strong> gravid uterus of <strong>the</strong> tammar<br />

(H. eugenii) is sensitive to exogenous OT (1) suggests <strong>the</strong> presence<br />

of uterine OT receptors in marsupials.<br />

Receptors were prepared from uteri of ovariectomized, oestrogentreated<br />

(30 ug/kg) possums using a published procedure. (2) For<br />

comparison, rat and sheep uterine tissue was similarly prepared.<br />

Receptors were measured by radioreceptor assay using a 3H-oyxtocin<br />

tracer and PEG precipitation of receptor-bound OT. Receptor number<br />

(Ro) and affinity (Kd) were estimated from Scatchard plots of <strong>the</strong><br />

data. Receptor specificity was characterized by inhibition<br />

experiments with related peptides (dOT=des-amino-oxytocin, M=<br />

mesotocin, AVP=Arg-vasopressin, LVP=Lys-vasopressin, IT=isotocin, VT=<br />

vasotocin), an oxytocin-selective agonist (TGO= [Thr 4 , Gly7]­<br />

oxytocin) an antidiuretic antagonist (PIA= [d(CH2)~, D-Phe 2 , Ile 4 ,<br />

Ala 9 -NH]- AVP) and unrelated peptides (AII= angiotensin II, NT=<br />

neurotensin). Table 1 shows that <strong>the</strong> Kd of <strong>the</strong> possum OT receptor is<br />

similar to that of <strong>the</strong> rat and sheep. Under our experimental<br />

conditions <strong>the</strong> Ro was lowest in <strong>the</strong> possum.<br />

Table 1.<br />

Affinity and Concentration of OT Receptors.<br />

SHEEP RAT POSSUM<br />

Kd (nmol/l) 2.5 ± 0.5 (n=5) 2.5 ± 0.5 (n=5) 3.8 ± 0.4 (n=8)<br />

Ro (fmol/mg prot) 590 ± 40 250 ± 30 200 ± 60<br />

Data shown as mean+/-s.e.m. n=number of estimates.<br />

Table 2. Inhibition (%) of OT binding by analogues.<br />

OT VT TGO AVP M dOT LVP IT PIA AII NT<br />

SHEEP 100 120 96 96 94 85 64 55


A RELATIONSHIP BETWEEN NUTRITION AND PROGESTERONE AT PARTURITION AND<br />

THE' IMPLICATIONS FOR LAMB SURVIVAL<br />

62<br />

63<br />

IN VIVO MYOMETRIAL ACTIVITY IN PREGNANT RATS DURING THE<br />

PERI-IMPLANTATION PERIOD<br />

M.A. Brockhus, 1 K. Nunan 2 and Ron A. Parr 3 Linda R~ Crane and Len Martin<br />

1 P~storal Research Ins titute, Hamilton, Victoria<br />

3Dookie Agricultural College, Dookie, Victoria<br />

Animal Research Institute, Werribee, Victoria<br />

Two essential conditions <strong>for</strong> <strong>the</strong> survival of lambs are a sufficient<br />

milk supply and a strong mo<strong>the</strong>r-offspring bond. Both conditions are<br />

hormonally controlled and probably linked. Progesterone has been<br />

shown to effect lactogenesis (1), as well as <strong>the</strong> ewes receptivity to<br />

<strong>the</strong> lamb (2). Nutrition in late pregnancy can alter progesterone<br />

patterns and delay lactogenesis (3). This study aimed to test <strong>the</strong><br />

effect of nutrition on progesterone concentrations, ewe behaviour and<br />

lactation in Merinos at parturition.<br />

Twenty-two Merino. ewes were divided into two groups and kept on<br />

diets of ei<strong>the</strong>r ad libitum hay (LOW) or 800g lupins/day plus ad lib<br />

hay (HIGH) between days 120 of gestation and term. Blood samples<br />

were collected regularly, stored, <strong>the</strong>n analyzed <strong>for</strong> progesterone using<br />

a radioimmunoassay. Continuous observations about parturition gave<br />

records of times of lambing, grooming, lamb standing, ewe nuzzling,<br />

lamb suckling and ewe feeding. At 48 hrs post-partum milk volume was<br />

measured using a standard technique (4). See Table 1 <strong>for</strong> results.<br />

Table 1 Plasma progesterone levels (ng/ml), milk vo1um~s at 48hrs (ml)<br />

and behaviour of ewes at lambing on two planes of nutrition.<br />

HIGH<br />

LOW<br />

Progesterone conc.(ng/ml) @ -48 hrs 5.12 ± 1.09 10.67~2.01 -24 2.96 ± 0.79 6.43 ± 1.68 *<br />

o 0.84 ± 0.16 0.77 ± 0.21 NS<br />

+2 0.23 ± 0.02 0.33 ± 0.09 NS<br />

+5 0.23 ± 0.04 0.28 ± 0.09 NS<br />

+10 0.03 ± 0.03 0.18 ± 0.01 NS<br />

Milk volume (mls)<br />

73.3 ± 5.4 34.7 ± 3.8 **<br />

Time from lamb born to ewe<br />

allowing to suckle (min)<br />

32 ± 8 59 ± 12 NS<br />

Time from lamb born to ewe<br />

recommencing feeding (min) 124 ± 24 39 ± 10<br />

Values are means + SEM: n=ll. * P < 0.05 ** P < 0.01 (ANOVA)<br />

**<br />

The slower decline of progesterone in <strong>the</strong> ~OW ewes supports <strong>the</strong><br />

view (3) that undernutrition affects progesterone secretion and/or<br />

metabolism, and could contribute to <strong>the</strong> lower milk volume (p


64<br />

INDUCTION OF MHC ANTIGEN EXPRESSION ON MHC-NEGATIVE PRIMARY<br />

AND SECONDARY TROPHOBLAST GIANT CELLS<br />

KinQ<br />

N.J.C.*, Rodger, J.C.t, Maxwell, L.E.§ and Drake, B.L.·<br />

*Dept. of Anatomy, University of Sydney N.S.W. 2006.<br />

tDept. of Biological Sciences, University of Newcastle, N.S.W.<br />

§John Curtin School, A.N.U., Canberra, A.C.T.<br />

·Dept. of Cell <strong>Biology</strong>, University of Texas, Dallas, Texas.<br />

Absence or low expression of surface major histocompatibility<br />

complex (MHC) antigens on semi-allogeneic trophoblast cells during<br />

implantation is a possible strategy <strong>for</strong> evading <strong>the</strong> maternal<br />

cellular immune response, as MHC expression by target cells is<br />

required <strong>for</strong> recognition by immune cytotoxic T (Tc) cells.<br />

Cells of <strong>the</strong> preimplantation murine blastocyst and established<br />

placenta express MHC antigens, but we have found, using immunogold<br />

labelling and electron microscopy, nei<strong>the</strong>r primary (1°) nor<br />

secondary (2°) (CBAxC57BLl6) F 1 trophoblast giant cells (TGC) outgrown<br />

in vitro from 3.5 day post coital (pc) blastocysts, and 7.5 day<br />

pc ectoplacental cones, respectively, express detectable quantities<br />

of paternal class I (Kk) MHC antigens. Treatment of 2° TGC <strong>for</strong> 24­<br />

40h with recombinant gamma-interferon (y-IFN) (Boehringer Ingelheim),<br />

.produced significant paternal class I MHC antigen expression<br />

but none was detectable on 1° TGC after ex, ~ or y-IFN treatment.<br />

This suggests that during <strong>the</strong> early period of implantation; trophoblast<br />

cell surface MHC is nei<strong>the</strong>r expressed nor inducible.<br />

Concurrently, we have found infection of some embryonic cell<br />

types with <strong>the</strong> flavivirus West Nile (WNV), causes an IFNindependent<br />

increase in class I and sometimes class II MHC antigen<br />

expression. There<strong>for</strong>e, in an attempt to induce MHC antigens, we<br />

infected 1 0 TGC monolayers with purified WNV. This induced de<br />

novo paternal class I MHC antigen expression within 16h. Induction<br />

is unlikely to be due to secreted WNV-induced IFN's or o<strong>the</strong>r<br />

factors, as it occurred in <strong>the</strong> presence of high concentrations of<br />

anti-exlB IFN antibodies, and was not induced by virus-inactivated<br />

supernatants from MHC-induced 1° TGC cultures. Fur<strong>the</strong>rmore,<br />

attempts to induce MHC expression with poly I:C or recombinant<br />

tumor necrosis factor-ex in 1° TGC cultures failed.<br />

Thus inhibition of constituent MHC expression on 1° TGC during<br />

early implantation and <strong>the</strong> associated block to MHC induction is not<br />

absolute. As such, manipulation of this phenomenon may<br />

conclusively establish <strong>the</strong> significance of lack of MHC expression<br />

in trophoblast cells of <strong>the</strong> implanting semi-allogeneic embryo.<br />

65<br />

DEVELOPMENr OF A CYNCM)LGUS mDEL 'IO SIUDY ffiffiNANCY-ASs::>CIATED<br />

PLASMA PROl'EIN-A (PAPP-A)<br />

;;1-._ Lee, J.P. WOlf l , R.F. Williams l , G.D. Hoogen l and M.J. Sinosich<br />

Reproc1uctive Biochemistry and Immunology, Department of Obstetrics<br />

and Gynaecology, Royal North Shore Hospital, St. Leonards NSW 2065<br />

lpregnancy Research, The Jones Institute <strong>for</strong> <strong>Reproductive</strong> Medicine,<br />

Eastern Virginia f.1edical School, Norfolk, Va. USA.<br />

Pregnancy-associated plasma protein-A (PAPP-A) is a large<br />

he:r;:arin-binding proteoglycan ",ith o(-electrophoretic mobility. In<br />

<strong>the</strong> human, PAPP-A is uniquely distributed within <strong>the</strong> reproductive<br />

tract, wherE' clinical and functicnal studies suggest a vital role<br />

fOt this antigen in human reproduction. However to test this<br />

hypo<strong>the</strong>sis an animal mooel was required. Criteria <strong>for</strong> selection of<br />

a model, included reversible binding to heparin, reversible<br />

interaction with ilnrnobilized zinc ions, molecular size (Mr) of<br />

820kd and secreted into maternal cirCUlation by a haemochorrially<br />

implanted, placenta. The cynanolgus monkey proved a most suitable<br />

model, and thus monkey (m) PAPP-A was purified fran cynanolgus<br />

placental tissue and pregnancy serum. The antigen was injected<br />

into NZ male rabbits and <strong>the</strong> resultant polyclonal artiserum used to<br />

develop mPAPP-A radioimmunoasay. Distinct immunological<br />

differences were apparent between cynanolgus and hmnan PAPP-A, with<br />

<strong>the</strong> latter analog containing more recently evolved antigenic<br />

determinants or epitoIA=s. P.s <strong>for</strong> t.he human anaJog, mPAPP-A was not<br />

detected in blood of normal non-pregnant adults but was readily<br />

measured in seminaJ plasma preovulatory follicular fluid and<br />

ooternal blood. After initial detection at about 40 days gestation,<br />

cirCUlating PAPP-A increased E'i


67<br />

IMMUNOLOGICAL ASSESSMENT OF PATIENTS WITH RECURRENT PREGNANCY<br />

LOSS AND UNEXPLAINED INFERTILITY.<br />

S. Raymond, C Czapla-Peters and Y.C Smart.<br />

Faculty of Medicine, University of Newcastle.<br />

Recent studies have described immunological factors as important<br />

in <strong>the</strong> aetiology of recurrent abortion and possibly unexplained<br />

infertility.<br />

To evaluate <strong>the</strong> importance of <strong>the</strong>se factors 3 groups of patients:<br />

were assessed:<br />

a) 17 couples with proven fertility (two term deliveries<br />

of normal hirthweight).<br />

b) 30 couples with obstetrical histories of unexplained<br />

recurrent pregnancy loss (URPL) divided into primary aborters (~3<br />

miscarriages and no live children) and secondary aborters<br />

(abortions after having children or stillbirths).<br />

c) 17 Couples with unexplained infertility (UI).<br />

Maternal sera were collected from all patients and screened <strong>for</strong><br />

<strong>the</strong> presence of antipaternal lymphocytotoxic activity, auto<br />

antibodies, immune complexes (IC) and immunoglobulins (IG).<br />

Results (table 1) showed that 4 of <strong>the</strong> 17 normal couples showed<br />

antipaternal lymphocytotoxic activity. None of <strong>the</strong> 14 primary<br />

aborters or <strong>the</strong> 17 patients with unexplained infertility showed<br />

any antipaternal lymphocytotoxic ~ctivitx whereas 3 of <strong>the</strong> 19<br />

secondary aborters did.<br />

TABLE 1 Immunological Assessment<br />

Antipaternal Auto<br />

n Antibody Pos Antibodies<br />

'1IC<br />

IIG<br />

FACTORS AFFECTING FETAL LOSS IN INDUCTION OF OVULATION WITH<br />

GONADOTROPHINS:<br />

INCREASED ABORTION RATES RELATED TO HORMONAL PROFILES IN CONCEPTUAL<br />

CYCLES<br />

suk-Yee Lam, H.W. Gordon Baker, James H. Evans, and Roger J. Pepperell<br />

University of Melbourne, Department of Obstetrics & Gynaecology,<br />

Royal Women's Hospital, 3053, Victoria, Australia<br />

The aim of this study is to compare <strong>the</strong> fetal wastage in<br />

gonadotrophin-induced pregnancies with that of spontaneous pre~nanc~es<br />

be<strong>for</strong>e and after treatment and to analyse <strong>the</strong> risk factors pred1spos1ng<br />

to such fetal loss. All <strong>the</strong> women treated with gonadotropin <strong>for</strong><br />

induction of ovulation at <strong>the</strong> Royal Women's Hospital between 1963 and<br />

1985 were studied. Among 230 women, 36 first-trimester abortions<br />

(9.7%), 16 second-trimester abortions (4.3%), 11 ectopic pregnancies<br />

(2.9%) and 10 stillbirths (2.7%) occurred in 373 conceptual cycles<br />

after gonadotropin induction of ovulation. Fetal loss (25 of 46<br />

pregnancies, 54.3%) was higher in spontaneous pregnancies prior to<br />

<strong>the</strong>rapy (p


68<br />

SEXUAL DIFFERENTIATION IN WALLABY POUCH YOUNG TREATED WITH<br />

STEROIDS<br />

G. Shawl, M. B. Renfreel , R. V. Shortl , 2 and Wai-Sum ()3<br />

Departments of lAnatomy and 2physiology, Monash University, Clayton,<br />

Vic., and 3Department of Anatomy, University of Hong Kong.<br />

In marsupials much of gonadal differentiation occurs post natally,<br />

so <strong>the</strong> young are readily accessible <strong>for</strong> experimental studies of <strong>the</strong><br />

hormonal control of sexual differentiation (1,2). We have investigated<br />

how gonadal steroids can influence development of <strong>the</strong> reproductive<br />

system in tammar wallabies by daily administration of estradiol<br />

benzoate (E2) (n=4) to male neonates, and testosterone propionate (T)<br />

(n=7) or oil (n=6) to female pouch young from <strong>the</strong> day of birth until<br />

autopsy' on day 25. After dissection, <strong>the</strong> bodies were fixed and<br />

serially sectioned.<br />

In control females <strong>the</strong> ovaries were located in <strong>the</strong> abdomen, and<br />

were well developed with <strong>the</strong> germ cells concentrated in <strong>the</strong> cortical<br />

area. The Mullerian ducts (MD) were large, whilst <strong>the</strong> Wolffian ducts<br />

(WD) were small and undifferentiated. A well developed pouch and four<br />

mammary gland buds were present. The gubernaculum and processus<br />

vaginalis terminated just inside <strong>the</strong> abdominal wall.<br />

Testosterone treatment did not affect ovarian histology or<br />

position, although it stimulated growth of both WD and MD. Pouch,<br />

mammary glands and gubernaculum were all unaffected.<br />

In control males <strong>the</strong> testes had well developed seminiferous cords,<br />

and Leydig cells. The WD were well developed, but <strong>the</strong> MD were<br />

regressing. The processus vaginalis and gubernaculum passed into <strong>the</strong><br />

centre of <strong>the</strong> well developed scrotum. The testes had completed <strong>the</strong>ir<br />

trans-abdominal migration and were situated in <strong>the</strong> inguinal canal.<br />

Testicular histology in <strong>the</strong>· E2 treated males was abnormal, with<br />

reduced tubule diameter, necrotic germ cells, few, small Leydig cells,<br />

and enlarged cortical areas containing germ cells. Although E2<br />

treatment did not affect WD size, it prevented MD regression presumably<br />

by inhibiting ei<strong>the</strong>r Mullerian Inhibiting Substance (MIS) secretion by<br />

<strong>the</strong> testis, or its action. The processus vaginalis terminated in <strong>the</strong><br />

inguinal region at <strong>the</strong> base of <strong>the</strong> scrotum, and <strong>the</strong> testes were located<br />

high in <strong>the</strong> abdomen, at a similar level to that of females.<br />

Thus, in <strong>the</strong> tammar wallaby, WD and MD development are affected by<br />

exogenous steroids as <strong>the</strong>y are in eu<strong>the</strong>rian mammals, but scrotum, pouch<br />

and mammary glands are unaffected by <strong>the</strong>se hormones, supporting our<br />

previous suggestion that differentiation of <strong>the</strong>se structures is<br />

independent of gonadal hormones (2). The prevention of both MD<br />

regression and testicular descent fur<strong>the</strong>r supports <strong>the</strong> hypo<strong>the</strong>sis (3)<br />

that MIS may mediate testicular trans-abdominal migration.<br />

(1) Renfree, M.B., Shaw, G. & Short, R.V. (1987) in: Genetic Markers<br />

of SeK Differentiation. pp 27-41. Eds F.P. Haseltine, M.E. McClure &<br />

E.H. Goldberg (Plenum Press).<br />

(2) 0, W.-S., Short, R.V., Renfree, M.B. & Shaw, G. (1988) Nature<br />

(Lond.) 331:716-717.<br />

(3) Hutson, J.M. (1985) The Lancet, Aug 24th, pp 419-421.<br />

69<br />

ANTIFERTILITY ACTIVITY OF GOSSYPOL IN MALE RATS AND<br />

MANGANESE STATUS OF DIET<br />

R. Vishwanath, I.G. White, P.O. Brown-Woodman,1 and J.R. Mercer 2<br />

Department of Veterinary Physiology and 2Animal Husbandry, University<br />

1 of Sydney, N.S.W.<br />

Cumberland College of Health Sciences, Lidcombe, 2141, N.S.W.<br />

There is good evidence that gossypol chelates with manganese (Mn)<br />

and it has been suggested that <strong>the</strong> antifertil ity activity of gossypol<br />

might be due to induction of Mn deficiency (1) which allegedly causes<br />

degeneration of <strong>the</strong> testes of rats with loss of spermatids and<br />

spermatozoa (2). This experiment was designed to investigate Mn<br />

deficiency in male rats and to check if excess Mn in <strong>the</strong> diet would<br />

mitigate <strong>the</strong> antifertility effects of gossypol.<br />

Th i rty male rats were randomi zed into six groups and fed one of<br />

three diets with or without gossypol in a factorial design. The diets<br />

were Mn deficient, normal and Mn excess. Gossypol in 2% carboxymethylcellulose<br />

vehicle was given orally (20 mg/kg body weight) each day.<br />

On day 57 two female rats were caged with each male to test <strong>the</strong>ir<br />

fertility and on day 64 <strong>the</strong> males were killed.<br />

The mean body weight of control rats did not change significantly,<br />

but rats given gossypol lost about 10% of <strong>the</strong>ir body weight. Nei<strong>the</strong>r Mn<br />

nor gossypol significantly affected <strong>the</strong> weights of <strong>the</strong> testes,<br />

epididymides, adrenals or seminal vesicles. However, <strong>the</strong>re was a<br />

significant decrease in weights of <strong>the</strong> coagulating glands of control rats<br />

on <strong>the</strong> Mn deficient diet, and of all three groups receiving gossypol.<br />

Fewer sperm coul d be recovered from <strong>the</strong> epi di dymi des of rats<br />

receiving gossypol and most of <strong>the</strong>se sperm were immotile and<br />

morphologically abnormal with a large proportion of detached heads.<br />

Gossypol greatly reduced <strong>the</strong> oxygen uptake of epididymal sperm from rats<br />

on all three diets, and sperm from control rats on <strong>the</strong> Mn deficient diet<br />

had a lower oxygen uptake than those on normal or Mn excess diets.<br />

Gossypol rendered all male rats compeletly infertile, irrespective<br />

of <strong>the</strong> level of Mn in <strong>the</strong> diet. The fertil ity of control rats on a Mn<br />

defi ci ent di et was not affected although <strong>the</strong> Mn concentrati on of <strong>the</strong>i r<br />

livers was reduced.<br />

(1) Vishwanath, R. and White, LG. (1987) New Horizons in Sperm Cell<br />

Research. Ed. H. Mohri. Japan Sci. Soc. Press. 431-437.<br />

(2) Boyer, P.O., Shaw, J.H. and Phillips, p.H. (1942) J. Biol. Chern.,<br />

143:417-425.


70 71<br />

PROLONGED EFFECT OF SUBFERTlLE MALES IN REDUCING NUMBERS OF FETUSES IN<br />

NORMAL FEMALE RATS.<br />

J.L.Zupp and B.P.Setchell.<br />

Department of Animal Sciences, Waite Agricultural Reasearch Institute,<br />

University of Adelaide, South Australia.<br />

When normal female rats were mated to males just be<strong>for</strong>e or after<br />

a period of infertility produced by local heating of <strong>the</strong> testes, <strong>the</strong>re<br />

was an increase in embryonic mortality and partial fertilization<br />

failure, even when <strong>the</strong> percentage of females becoming pregnant was no<br />

different from control. This was reflected in a decrease in <strong>the</strong><br />

fetus/corpus luteum (F/CL) ratio between 7 and 14 days after <strong>the</strong> end<br />

of a 7 'day mating period (1). In <strong>the</strong>se earlier experiments, it<br />

appeared that this effect continued <strong>for</strong> a considerable time after<br />

fertility was regained, but because adequate controls were not<br />

available <strong>for</strong> this aspect of <strong>the</strong> study, fur<strong>the</strong>r experiments have been<br />

undertaken to see how long <strong>the</strong> effect on F/CL ratio persisted.<br />

Groups of 6 adult male rats (Porton strain in Exp.1, 3 and 4 and<br />

Hooded Wistar in Exp 2) were anaes<strong>the</strong>tized with pentobarbitone sodium<br />

(50mg/kg intraperitoneally) and <strong>the</strong>ir testes and scrotum immersed in a<br />

water bath at ei<strong>the</strong>r 43 or 33 0 C <strong>for</strong> 30 min. At various times after <strong>the</strong><br />

heating, each male was caged with 6 normal females <strong>for</strong> 7 days. The<br />

females were <strong>the</strong>n removed and autopsied after a fur<strong>the</strong>r 7 days, when<br />

<strong>the</strong> number of fetuses and corpora lutea were.90unted. There were·<br />

significant differences in <strong>the</strong> F/CL ratio in 2 experiIilents <strong>for</strong> <strong>the</strong><br />

Il,ating period beginning 60 days after heating, associated with a<br />

reduced percentage of feo.ales pregnant j<strong>the</strong>re was also a significant<br />

difference in one experiment, but not in ano<strong>the</strong>r in <strong>the</strong> F/CL ratio 90<br />

days after heating, and no difference in 2 experiolents at 114 days.<br />

Exp.No Days after %females F/CL ratio in<br />

heating pregnant<br />

pregnant females<br />

Control Heated Control Heated males<br />

90-97<br />

50 42 0.80 0.58**<br />

114-121<br />

91 75 0.71 0.86<br />

126-133<br />

51 43 0.54 0.62<br />

2 114-121 83 94 0.79 0.79<br />

3 60-67<br />

58 13* 0.64 0.34-<br />

90-97<br />

63 70 0.67 0.76<br />

4 -7-0<br />

53 81 0.68 0.69<br />

60-67<br />

54 29* 0.66 0.51.<br />

.: P


72<br />

DURATION OF PROOESTAGEN PRIMING ON THE RESPONSE OF<br />

BORDER. LEICESTER. X MERINO EWES TO THE 'RAM EFFECT'<br />

J.L. Reeve<br />

Department of Agriculture & Rural Affairs<br />

Ru<strong>the</strong>rglen Research Institute, Ru<strong>the</strong>rglen, Victoria.<br />

Short tenn use of intravaginal progestagen sponges prior to <strong>the</strong><br />

introduction of rams to ewes in late Spring has been shown to<br />

enhance <strong>the</strong> response to <strong>the</strong> 'rarn effect' (1),(2),(3). A high<br />

proportion of primed ewes show oestrus at <strong>the</strong> 'rarn induced'<br />

ovulation, when a majority of twins are conceived. 'Short' cycles<br />

are eliminated and almost no responding ewes return innnediately to<br />

anoestrus. In <strong>the</strong> initial experiment (1) progestagen sponges were<br />

used <strong>for</strong> 4,6 or 10 days. The 6 day treatment was superior to <strong>the</strong> 4<br />

,day in ewes conceiving at <strong>the</strong> ' rarn induced' cycle, and overall<br />

fertility and fecundity. The 10 day treatment resulted in <strong>the</strong><br />

highest proportion of ewes lIIating at <strong>the</strong> 1st cycle but with fewer<br />

ewes conceiving with lower fecundity than <strong>the</strong> 6 day. This study<br />

examined <strong>the</strong> sensitivity of response to <strong>the</strong> duration of progestagen<br />

priming between 5 and 9 days.<br />

On 28 OCtober 1987 415 1.5 y.o. Border Leicester x Merino ewes were<br />

treated with sponges (Repromap-60mg) and allocated at random to 5<br />

equal groups. At intervals of 5,6,7,8,9 days <strong>the</strong> sponges were<br />

withdrawn from a group of ewes which were <strong>the</strong>n joined to 8% Poll<br />

Dorset rams <strong>for</strong> 42 days. Mating was recorded daily and foetuses<br />

were counted and aged by real time ultrasonography 13 weeks after<br />

joining. In <strong>the</strong> table below, 1st CyCle refers to mating and<br />

conceptions 2-3 days after joining.<br />

Ewes<br />

Foetuses<br />

Conceiving Mean days Conceived Mean days Foetuses<br />

Prog. cycle post cycle post per ewe<br />

days n 1 2or3 joining 1 2or3 joining joined<br />

-------------------------------------------------------------------<br />

5 85 49 29 9.25 a 63 31 8.4 a 1.107 a<br />

6 83 51 19 6.9 a 71 21 6.0 b 1.108 a<br />

7 82 61 14 5.6 b 78 15 5.1 b 1.134 a<br />

8 82 63 13 5.0 b 77 15 4.9 b 1.135 a<br />

9 83 64 9 4.0 b 75 9 3.7 b 1.012 a<br />

-------------------------------------------------------------------<br />

Longer duration of progestagen priming increased <strong>the</strong> proportion of<br />

ewes conceiving at <strong>the</strong> 1st cycle and concentrated oestrus behaviour<br />

earlier in <strong>the</strong> post joining period. There was no overall effect on<br />

foetuses produced per ewe joined.<br />

The response to duration of progestagen priming has low, sensitivity<br />

around an optimum of 7 or 8 days but it should be longer than 5<br />

days.<br />

(1) Reeve, J.L. and'Charnley, W.A. (1982). Proc. ASRB, 14:89.<br />

(2) Reeve, J.L. and Charnley, W.A. (1983). Proc. ASRB, 15:71.<br />

(3) Reeve, J.L. and Charnley, W.A. (1984). Proc. ASAP, ,15:161-164<br />

Figure 1. Mean melatonin 1000<br />

concentration in three pM<br />

cows with four Regulin 500<br />

implants. Shaded area is<br />

endogenous night~time range.<br />

(assay limit 112 pM)<br />

73<br />

EFFICACY OF MELATONIN IMPLANTS (REGULlN) IN BOS INDICUS COWS.<br />

S R P<br />

Su<strong>the</strong>rland and K.W. Entwistle<br />

Graduate School of Tropical veterinary Science, James Cook University<br />

Townsville, Queensland 4811.<br />

Reproduction in Bos taurus cattle is affected by season and can be<br />

modified by manipulation of <strong>the</strong> photoperiod environment (1). There<strong>for</strong>e<br />

it may be possible to use melatonin to modify reproduction in cattle.<br />

The efficacy of Regulin implants (~egulin Ltd, Australia) to increase<br />

circulating melatonin levels in Bos indiclls cattle was tested using 16<br />

multiparous cows (mean liveweight 421± 7.7(SE) kg) kept on native<br />

pasture. Each cow was treated with ei<strong>the</strong>r 0, 1, 2 or 4 Regulin<br />

implants (s . c.) at <strong>the</strong> base of one or both ears. All cows :were'bled<br />

from <strong>the</strong> tail at 10pm and lOam at 0, 1, and 2 weeks, and at lOam, 3,<br />

4, and 5 weeks after treatment. Night-time samples were taken by<br />

torchlight. Be<strong>for</strong>e treatment melatonin was detectable (assay limit 40<br />

pM) in <strong>the</strong> tail plasma of 12 cows at 10pm (95.2± 12.5 pM) and 4 cows<br />

at lOam (50.2± 3.1 pM). One to tive weeks after treatment melatonin<br />

was undetectable (assay limit 112 pM) in <strong>the</strong> tail plasma of most<br />

animals in both day and night-time samples. At five weeks after<br />

treatment jugular samples were taken from <strong>the</strong> cows without implants at<br />

10pm, and from all <strong>the</strong> cows at lOam. In untreated cows <strong>the</strong> mean<br />

jugular' concentration of melatonin at night-time was 426± 86 pM<br />

(range 312 - 645). The night-time range reported <strong>for</strong> Jersey heifers is<br />

190 - 1410 pM(2). Melatonin was undetectable «112 pM) in <strong>the</strong> day-time<br />

jugular plasma of untreated cows and those with one implant. Melatonin<br />

was detectable in <strong>the</strong> jugular plasma of three cows with two implants<br />

(271± 97 pM) and all of <strong>the</strong> cows with four implants. One cow with four<br />

implants had a very high concentration of melatonin (2645 pM)<br />

suggesting abnormal liver function. The mean jugular concentration in<br />

<strong>the</strong> o<strong>the</strong>r three cows with four implants was 227± 21 pM, about half <strong>the</strong><br />

mean endogenous night-time concentration. These three cows were<br />

sampled <strong>for</strong> a fur<strong>the</strong>r 13 weeks and during this period melatonin<br />

concentrations were extremely variable, but were frequently within <strong>the</strong><br />

endogenous night-time range up to 16 weeks after treatment (Figure 1) .<br />

6 8 10 12 14 16<br />

weeks after 1reatment<br />

conclude that' four subcutaneous Regulin implants elevate plasma<br />

We<br />

melatonin in cattle and may continue to deliver <strong>for</strong> up to 16 weeks. As<br />

in sheep(3) melatonin released from <strong>the</strong> implants or from <strong>the</strong> pineal is<br />

more readily detected in <strong>the</strong> jugular vein than in peripheral vessels.<br />

(1) Hansen, P.J. and Hauser, E.R. (1984). Theriogenology 22:1-14<br />

(2) Martin, T.C., Cunningham, N.F. and Saba, N. (1983). J. Endocr.<br />

.9.6.:189-196<br />

(3) Howse, A., Kennaway, D., Carbone, F., Staples, L. and Williams, A.<br />

(1987). Proc. ASRB ~:20<br />

The authors acknowledge Dr L.D. Staples (Regulin Ltd) <strong>for</strong> supply of<br />

<strong>the</strong> implants and Dr D. Kennaway <strong>for</strong> melatonin assays.<br />

18


74<br />

SYNERGISTIC EFFECTS OF MELATONIN AND IMMUNISATION<br />

AGAINST ANDROSTENEDIONE IN MAIDEN BL X MEWES<br />

C.R. Earl, S: McPhee, R.H. Male and E.A. Dunstan<br />

S.A. Dept. of Agriculture. Struan and Kybybolite Research Centres<br />

Immunisation against androstenedione, Fecundin (Glaxo Aust.<br />

Ltd.) and melatonin treatment, Regulin, (Genelink Aust. Ltd.) improve<br />

<strong>the</strong> reproductive per<strong>for</strong>mance of crossbred ewes mated in summer. A<br />

recent study by Dunstan et al (1) indicated that those two products<br />

could act synergistically to improve lambing percentages but <strong>the</strong><br />

mechanism of action has not been identified.<br />

In this study conducted at Struan, S.A. groups of maiden 1 1/2<br />

year, old BL x M ewes received ei<strong>the</strong>r no treatment (control n=63) a<br />

Regulin implant at 4 weeks be<strong>for</strong>e mating (Regulin n=62) injections'of<br />

Fecundin at 6 and 2 weeks (Fecundin n=64) or combined treatment with<br />

both Regulin and Fecundin. All groups were grazed toge<strong>the</strong>r and were<br />

joined with 3% untreated Dorset rams on 4/1/88. Foetal percentages<br />

were determined by mid pregnancy sonagraphy.<br />

Table 1 - Fertility fecundity and distribution of litter size of<br />

maiden ewes after no treatment, treatment with Regulin,<br />

Fecundin or treatment with both.<br />

Foetuses Per No. of ewes with<br />

Group Ewe Ewe Preg. 0 1 2<br />

Control n=63 1.11 a 1.37 a 12 32a 19a 0<br />

Regulin 62 1.23 a 1.41 a 8 32a 22a 0<br />

Fecundin 64 1.30 b 1.77 b 17 12b 34b 1<br />

Combined 63 1. 56 b 1.81 b 9 15b 34b 5<br />

a~b within<br />

tables.<br />

columns. P


76<br />

THE EFFECTS OF NUTRIENT RESTRICTION AND OF LAMB REMOVAL ON OVARIAN<br />

CYCLICITY AND ON THE INHIBITORY EFFECTS OF OESTRADIOL ON PlASMA<br />

CONCENTRATIONS OF LH, AND FSH IN POST-PARTUM EWES<br />

P.J<br />

Wright, A.H. Williams and I.J. Clarke<br />

Department of Veterinary Clinical Sciences, University of Melbourne,<br />

Werribee 3030, Victoria, Australia<br />

Corriedale ewes, 4-7 days post partum (PP) and non-post-partum<br />

cyclic (C) ewes, were ovariectomized (OVX) during <strong>the</strong> ovulatory<br />

season and received oestradiol (E) implants s.c. or remained<br />

untreated. Ewes <strong>the</strong>n received i) above-maintenance (M) nutrition,<br />

ii) below maintenance (R) nutrition, or iii) R nutrition and lambs<br />

rem9ved within 2 days of parturition (RW). Blood samples (3/ewe) <strong>for</strong><br />

<strong>the</strong> determination of plasma concentrations of LH and FSH were taken<br />

over <strong>the</strong> period 18-24 days post partum or 12-18 days after<br />

ovariectomy (C ewes). OVary-intact ewes also received treatments M<br />

(n-20), R (n-26)" and RW (n-2l). Blood samples (2/week) were taken<br />

from <strong>the</strong>se ewes (from 7 to 60 days PP) <strong>for</strong> plasma progesterone<br />

determination to assess <strong>the</strong> onset of ovarian cyclicity.<br />

The plasma hormone concentrations (ng/ml) (mean (s.e.m.)) in<br />

OVX ewes are shown below.<br />

Treatment -==LH~ .-...F""SH ... _<br />

group OVX OVX+E OVX OVX+E<br />

n cone sig n cone sig conc sig cone sig<br />

(a) values with no letters common are significantly different (P


78<br />

P:t'rUITARY AND OVARIAN BESPOHSES OF POS':r-PAR'.rtJM ACYCL:tC :aBEl' COWS TO<br />

CON'.rDlUOUS~ TREA'l'MENT WITH GnRH AND A GnRH AGONIST<br />

M.J. D'Occhio and D.R. Gif<strong>for</strong>d t<br />

CSIRO, Division of Tropical Animal Production, Rockhampton, Qld<br />

t SA Department of Agriculture, Turretfield Research Centre,<br />

Rosedale, SA<br />

Gonadotrophic hormone releasing hormone (GnRH) can be used to<br />

promote ovarian activity and ovulation in acyclic post-partum (PP)<br />

cows when given ei<strong>the</strong>r by continuous infusion over 2-5 days or bolus<br />

injection. Corpora lutea (CL) arising from GnRH-induced ovulations PP<br />

tend to have a short life span, as do CL of <strong>the</strong> first spontaneous<br />

ovulation PP. The present study examined whe<strong>the</strong>r sustained CL<br />

function could be achieved with continuous long-term GnRH <strong>the</strong>rapy.<br />

Chronic GnRH could ei<strong>the</strong>r ensure a normal life span of <strong>the</strong> first<br />

induced CL by maintaining elevated LH or, alternatively, induce a<br />

second ovulation with normal CL function. Multiparous PP (17.1 i: 0.6<br />

days) acyclic beef cows received continuous GnRH or buserelin ([D-Ser<br />

(tBu) 6, Pro 9 NEt] GnRH) from s.c. osmotic minipumps(2ML4) designed to<br />

remain active <strong>for</strong> 28 days. Plasma LH, FSH and progesterone (P 4<br />

) were<br />

determined by RIA and P4 concentrations ) 1 ng/ml were taken as<br />

indicative of active CL tissue. Results <strong>for</strong> P4 and CL are shown in<br />

<strong>the</strong> Table.<br />

P4<br />

'Treatment n ) 1 ng/ml<br />

corpus luteum<br />

short-lived<br />

extended<br />

control 5 0 0 0<br />

200 ng GnRH/kg BWjh 5 4 4 0<br />

400 ng GnRH/kg BWjh 4 4 3 1<br />

5.5 ng buserelin/kg BW/h 5 4 3 1<br />

11.0 ng buserelin/kg BWjh 5 3 2 1<br />

Plasma LH concentrations, but not FSH, were elevated at least to<br />

day 10 in all cows receiving GnRH or buserelin. The outstanding<br />

features of <strong>the</strong> P 4 profiles in 15/19 treated cows that responded were<br />

<strong>the</strong> synchrony, both within and across groups, in P4 ) 1 ng/ml around<br />

day 6, and <strong>the</strong> fact that most CL were short-lived (4-6 days). Only 3<br />

cows showed evidence of extended CL function. The results suggest<br />

that endogenous LH secretion in PP cows following GnRH-induced<br />

ovulation is not limiting to CL function,and that o<strong>the</strong>r factors cause<br />

early demise of <strong>the</strong> first CL. The most likely explanation is that<br />

normal CL development appears to reqUire pre-exposure to P • In this<br />

4<br />

regard, it is not clear why cows failed to show a second ovulation<br />

around day 10 that was followed by normal CL function; however, it is<br />

considered that this was due to <strong>the</strong> doses of GnRH and buserelln used,<br />

ra<strong>the</strong>r than physiological constraints within <strong>the</strong> cows.<br />

Stu'ay supported in part by <strong>the</strong> Australian Meat and Livestock<br />

Research and Development Corporation; GnRH was supplied by Peptide<br />

Technology Limited, NSW; and buserelin by ~oechst AG, W Germany.<br />

79<br />

EFFECT OF GnRH ON FERTILITY OF MERINO EWES INSEMINATED WITH<br />

FROZEN SEMEN<br />

W.M.C. Maxwell l , S.K. Walker 1, D.H. Smith 1 and H.R. Wilson 2<br />

Department of Agriculture, S.A., and<br />

Collinsville Breeding Research Centre, S.A.<br />

Tight synchrony of <strong>the</strong> time of ovulation may improve fertility<br />

following AI of ewes. For this purpose, non-superovulated Merino ewes<br />

have been treated with GnRH 36h after sponge removal (SR) (1). This<br />

study examined <strong>the</strong> effect of GnRH treatment and time of insemination on<br />

fertility of ewes inseminated with frozen semen.<br />

Ewes treated with progestagen sponges (Intervet [Aust.] Pty. Ltd.) <strong>for</strong><br />

12 days and an injection of 400 Lu. PMSG (Intervet [Aust.] Pty. Ltd.)<br />

at SR, received ei<strong>the</strong>r no GnRH or 40ug syn<strong>the</strong>tic GnRH (Intervet [Aust.]<br />

Pty. Ltd.) 36h after SR. Semen was collected from three rams (A, B, C)<br />

and frozen-stored in pellet <strong>for</strong>m. The ewes were inseminated by<br />

laparoscOpy (0.04ml semen containing 40 million motile spermatozol-)<br />

ei<strong>the</strong>r be<strong>for</strong>e (51-54h) or after (69-72h after SR [2]) <strong>the</strong> expected time<br />

of ovulation. Pregnancy and number of foetuses were determined by<br />

ultrasound 50 days after insemination.<br />

Percentage ewes pregnant and foetuses present <strong>for</strong> rams A, B, C were<br />

42.2 52.3,46.0 (p


Supported by <strong>the</strong> Australian Wool Research and Development Fund.<br />

80<br />

FACTORS INFLUENCING LAMB SURVIVAL IN A HIGH FECUNDITY<br />

BOOROOLA X SOUTH AUSTRALIAN MERINO FLOCK<br />

D.O. Kleemann,1 S.K. walker 1 , J.R.W~ wa~k~ey2, R.W. Ponzoni 2 ,<br />

D.H. Smith 1 and R.F. Seamark 3<br />

1Turretfie~d Research centre, Department of Agricu~ture, Roseda~e, SA<br />

5350, 2Department of AgricUlture, Ade~aide, SA 5000 and 3Department of<br />

Obstetrics and Gynaecology, University of Adelaide, Adelaide SA 5000<br />

<strong>Reproductive</strong> potential of <strong>the</strong> South Australian (SA) Merino can be<br />

improved substantially by incorporation of <strong>the</strong> Booroola Merino high<br />

fecundity gene (F). However lamb survival is poor. Factors<br />

associated with high lamb mortality need to be identified so that<br />

appropriate strategies to reduce ~osses can be investigated.<br />

Lamb surviva~ (birth to weaning) and birthweight data were<br />

collected from a mixed-aged 1/2 Booroola x 1/2 SA Merino flock at Cape<br />

Borda, Kangaroo Island, SA, in June-July of years 1981 to 1986. Ewes<br />

were managed as in norma~ commercial practice. The effects of year,<br />

ewe age (2-year-o~d vs older) and birthweight (0.5 kg categories<br />

between 1.5 and 6.0 kg) on lamb surviva~ were arta~ysed within singleborn<br />

and mUltiple-born categories, using <strong>the</strong> procedure GLM in SAS.<br />

Least-squares means (+ s.e.) <strong>for</strong> birthweight and survival, of ~ambs<br />

born in litter sizes ~f 1-4 from a 1/2 Booroola x 1/2 SA Merino flock<br />

Litter size<br />

Lambs born<br />

Birthweight (kg)<br />

Survival (%)<br />

580<br />

4.13 + 0.04<br />

70.8 :; 1.6<br />

Twin<br />

784<br />

3.28 + 0.03<br />

53. 6 ~ 1.4<br />

Triplet<br />

321<br />

2.80 + 0.05<br />

35.9 "+ 2.2<br />

Quadruplet<br />

52<br />

2.59.+ 0.15<br />

15.9"+ 5.9<br />

There was a decline in survival rate as litter size increased,<br />

<strong>the</strong> reduction being paralleled by a reduction in birthweight (Table<br />

1). In <strong>the</strong> analyses of both single and multiple-born categories<br />

birthweight was <strong>the</strong> dominant factor inf~uencing survival (P


82<br />

OVULATORY POTENCY OF ASRB-bFSH-l IN SHEEP AND<br />

CATTLE<br />

B.M. Bindon,l J.K. Findlay,2 L.R. Piper 1 and M.A. Hi11ard1<br />

1 (On behalf of ASRB Pituitary Hormone Committee)<br />

2 CSIRO Division of Animal Production, Armida1e, NSW<br />

Medical Research Centre, Prince Henry's Hospital, Melbourne, VIC<br />

Biological studies of pituitary hormones in vivo in domestic<br />

livestock are restricted in Australia by <strong>the</strong> unavailability of suitable<br />

reference preparations. Some years ago AMLRDC, AWC and NH&MRC funds<br />

were provided to ASRB to begin <strong>the</strong> local purification of pituitary<br />

hormones <strong>for</strong> use by <strong>the</strong> <strong>Society</strong>'s members. The present<br />

report deals with one of <strong>the</strong> preparations from this project.<br />

ASRB-bFSH-1 was compared with commercial porcine FSH ("FSH-P"<br />

Burns Biotec) in sheep and cattle. The latter has approximately fou;<br />

times <strong>the</strong> FSH radioreceptor assay potency of ASRB-bFSH-1. Merino ewes<br />

(8 per dose) were synchronized with progestagen sponges, <strong>the</strong>n given FSH<br />

as intramuscular injections twice daily <strong>for</strong> four days beginning on Day<br />

11 after sponge insertion. Total FSH dose was given in decreasing<br />

doses in <strong>the</strong> ratio 4:3:2:1 over four days and <strong>the</strong> sponges removed on<br />

<strong>the</strong> morning of <strong>the</strong> third day. A similar protocol was followed in<br />

cattle (three-year-old crossbred heifers, 300-350 kg !1veweight; 5-7<br />

cows per dose), using two injections of prostaglandin analogue <strong>for</strong><br />

synchronization. In both species ovulation rates were assessed by<br />

laparoscopy 5-7 days after oestrus. The results in Table 1 show that<br />

in sheep ASRB-bFSH-1 has less than one-eighth <strong>the</strong> ovulatory potency of<br />

FSH-P. In cattle, however, ASRB-bFSH-1 at a dose of 200 mg produced<br />

useful superovulation with no evidence of ovarian over-stimulation,<br />

suggesting a potency of about one-third that of FSH-P. ASRB-bFSH-1,<br />

with


84 85<br />

MN:IlIEWd'ICAL MDEL AND a:MIOl.'m SlMIJLATICfi OF PUI:SATILE JI:lMIilE<br />

SOCRErr'ICfi wr:m APPLICATICfi 'It) III<br />

David J Handelsnan., *Ross lazarus<br />

Depart:.rrent of Medicine, University of Sydney and *Bundoora Extended<br />

care Centre, Bundoora, Victoria<br />

Episodic secretion of hypothalamic GnRH regulates pituital:y-gonadal<br />

function via <strong>the</strong> pulsatile secretion of LH. Despite its importance,<br />

mmy physiological aspects of pulsatile LH secretion remain<br />

technically difficult to study arpirically. Various objective,<br />

canputer-based pulse detection algorithmns have been applied to serial<br />

LH :rreaSuretrents in blood as a non-invasive index of neuroendocrine<br />

activity of <strong>the</strong> hypothalamus. Ccmparisons and validation of<br />

algorithrnns is limited to a few intensive human LH pulse series and<br />

ert1irical optimizing against false positive rates as detennined fran<br />

signal-free noise series however <strong>the</strong> :i..nlp3.ct of <strong>the</strong>se reccmrendations<br />

on <strong>the</strong> false negative rates has not been studied SYSte:natically.<br />

There<strong>for</strong>e we have developed a mathamtical node! of pulsatile honrone<br />

secretion with specific reference to LH and inplemented this in a<br />

versatile canputer simulation package (SimPulse). The nodel is based<br />

on pulses created f:ran a point source (pituitaJ::y) of LH secretion with<br />

subsequent continuous decrementing by netabolic clearance in a<br />

biexponential phanracokineticnodel using available ert1irical data.<br />

Pulse Contour has adjustable shape and pulses are spaced at defined<br />

ti.lTe intervals with net honrone level being <strong>the</strong> sum of all pulses<br />

above a set threshold. All pararreters are user-definable and 4 key<br />

variables - interpulse interval, secretory burst duration, secretory<br />

rate, metabolic· clearance rate - may have user-defined gaussian<br />

distribution of known nean and variance. Runs generate an output file<br />

suitable <strong>for</strong> passing to pulse detection prog:r;ams and record files with<br />

<strong>the</strong> actual pulse pararreters f:ran <strong>the</strong> simulation run to canpare with<br />

output of pulse detection programs. A second nodule (SimError)<br />

introduces stochastic variations due to sarrpling-time and assay<br />

precision with definable statistical distributions and selectable<br />

pararreters. This program will be useful <strong>for</strong> m:x:lelling of physiological<br />

and pathological aspects of pulsatile LH secretion especially those at<br />

present inaccessible to ert1irical estimation (burst durations and<br />

pituitaJ::y secretory rates) as well as pulse detection strategies<br />

including sarrpling schemata and canputer algorithrnns .<br />

NO STOCHASllC VAIltATlON IN PNWolEreRS<br />

1.0.,------ ---, 1.0,----_- -.<br />

TlI.lE<br />

0.8<br />

STOCHAS!1C VNIlAnON ~ _ SAllE IGN·p~<br />

TlI.lE<br />

THE EFFECTS OF SERTOLI CELL lATRIX, FETAL CALF SERUI AND SERTOLI<br />

CELLS ON 3H-THYMIDINE INCORPORATED INTO DNA OF CULTURED MYOID CELL8<br />

cpmfug DNA<br />

X on I XF on I 1 F on I S on I<br />

2351<br />

* 17. LSD values<br />

8.8. Raychoudhuryl, I.G. Irving 1 and A.V. Blackshaw2<br />

lSchool of Science, Griffith University, Nathan, Qld. 2De~artment of<br />

Physiology k Pharmacology, University of Queensland, St. LUCIa, Qld.<br />

Ve have reported radiolabel incorporation in~o myoid cell. P!ote~n<br />

syn<strong>the</strong>sis and secretion, GAG production, secretIon and deposlt.lOn In<br />

<strong>the</strong> extracellular matrix (1). This study extends <strong>the</strong> work to Include<br />

aH-thymidine incorporated into DNA.<br />

8ertoli cells (S) and myoid cells (I) were prepared from 20-22 day<br />

old rat testes and cultured (2). There were six treatment com~inations<br />

of I S Sertoli cell matrix (X) and fetal calf serum (F), VIZ; I, S,<br />

SI, XI, 'IF and IMF. All culture types were labelled with aH-thymidine<br />

(1 JjCijml) <strong>for</strong> 96 h at 37° C and treated with (BU) 2 cAMP , (0.5 roM) or<br />

FUT CFSH 25 ngjmlj insulin, 5 Jjgjml; retinol, 0.35 JjM; testo~terone,<br />

0.7 Jjl). 'The extent of DNA syn<strong>the</strong>sis in <strong>the</strong>se culture combina~lons w~s<br />

measured by both scintillation counting and autoradlographlc<br />

examination.<br />

The incorporation of 3H- thymidine into cultures was low in all<br />

preparations except those containing feta~ calf serum. . In <strong>the</strong>se<br />

cultures <strong>the</strong> presence of Bertoli cell matnx had a relatlVely small<br />

inhibitory effect on <strong>the</strong> incorporation of label.<br />

The effect of culture conditions on <strong>the</strong> uptake of [3HJ thymidine by<br />

myoid cells.<br />

29555 497 34722<br />

701<br />

944*<br />

7. labelled nuclei<br />

X on 1 XF on M I F on I S on I<br />

16.9<br />

58.9 14.7 69.0 10.9<br />

1.9*<br />

The hormone mix (FUT) stimulated an~ cAMP inhib~ted ~ncorporation<br />

under most culture conditions, although In <strong>the</strong> Sertoll-myold co-cu~ture<br />

both FIRT and cAIP reduced <strong>the</strong> already low level of nuclear labelhng.<br />

Ve conclude that DNA syn<strong>the</strong>sis in myoid cells is slightly stimulated<br />

by Sertoli cell extracellular matrix. The effect is minor compared to<br />

<strong>the</strong> response to fetal calf serum, which clearly s~ppli~s necessary<br />

growth factors. T~e inhib~to!yeffect of cAMP conhrms ItS.accepted<br />

role of promoting dIfferentIatIon, whereas <strong>the</strong> hormonal cocktaIl (FIRT)<br />

facilitates cell replication.<br />

(1) Raychoudhury, S.S., Ir~ing, I.G. &Blackshaw, A.V. (1987)<br />

Proc.Aust.Soc.Reprod.Blol. 19:84. .<br />

(2) Skinner, I.K. &Fritz, I.B. (1985) Proc.Natl.Acad.Scl. 82:114-118.


86<br />

EFFEcrs OF SEROTONIN ON INHIBIN AND TESTOSTERONE PRODUCTION BY<br />

ADULT RAT SEMINIFEROUS TUBULES AND LEYDIG CELLS IN VITRO<br />

G.F. Gonzales*, J. Muir, G.P. Risbridger and D.M. de Kretser<br />

Department ofAnatomy, Monash University, Melbourne, 3168.<br />

Hyperserotoninemia has been shown to be deleterious to spermatogenesis without<br />

affecting serum testosterone levels in men (1) and male rats (2). Howeversome<br />

authors have claimed that serotonin alters spermatogenesis, secondary to an inhibition<br />

oftestosterone production (3,4). The aim of <strong>the</strong> present study was to determine <strong>the</strong> in<br />

vitro effect of serotonin on both testosterone production by Percoll-purified Leydig<br />

cells and inhibin production by isolated seminiferous tubules (ST) from normal adult<br />

rats.<br />

Leydig cells were isolated from <strong>the</strong> testes of adult rats and purified on discontinuous<br />

gradients of Percoll (Pharmacia, Uppsala, Sweden). 50,000 cells were <strong>the</strong>n cultured in<br />

plastic multiwell dishes containing 250 J.lI Dulbeceo modified Eagle medium: F12 ± 2.5 IU<br />

hCG at 32·C <strong>for</strong> 20 h. Serotonin was added in <strong>the</strong> dose range 10-1000 ng/well. 5 em<br />

segments of ST were dissected free of interstitial tissue and cultured in 1 rnl DMEM<br />

with <strong>the</strong> following additions: serotonin (Sigma) (1-1000 ng/rnl) ± ei<strong>the</strong>r FSH (500<br />

ng/rnl), dbcAMP (10 J.lg/ml) 0:' insulin (Sigma) (20 ng/rnl).<br />

Serotonin.has no effect on basal testosterone production, but an increase in <strong>the</strong> hCGinduced<br />

T production was observed with 10 ng serotonin (p


88<br />

IMMUNOCYTOCHEMICAL LOCATION OF OXYTOCIN AND MESOTOCIN WITHIN THE<br />

HYPOTHALAt4US AND REPRODUCTIVE TRACT OF THE MALE MARSUPIAL BANDICOOT<br />

R.T. Gemmell l and C. Sernia 2<br />

lDepartment fAt 2D t f<br />

o na.omy,. epar ment 0 Physiology and Pharmacology,<br />

Un1vers1ty of Queensland, Brisbane.<br />

T~e structure o~ <strong>the</strong> ~eurohypophyseal peptide hormones isotocin,<br />

mesotoc1 nand OXytOC1 n var1 es by ei<strong>the</strong>r one or two ami no aci ds.<br />

Isotocin hc:s been identified in all bony fishes, mesotocin in all <strong>the</strong><br />

non-mammal1an tetrapods, namely birds, reptiles amphibians and lung<br />

fishes nd oxytocin in <strong>the</strong> placental mammals.O) In <strong>the</strong> marsupials<br />

mesotoc;n, but no~ oxytocin, has been identified in nine species of<br />

Austral;an marsup1al. In American marsupials, both oxytocin and<br />

meso~oc1n have been reported in two species and oxytocin alone in one<br />

spec; es. If one acce~ts <strong>the</strong> o~thodo~ evol uti onary pathway of <strong>the</strong>se<br />

pept1 de hormones as be1 ng from 1S0toc1 n through mesotoci n to oxytoci n<br />

<strong>the</strong>n t~e p~es~nce of mes~toci n wi thi.n <strong>the</strong> Austral i an marsupi a1s, and<br />

oxytoc;n w1t~1n <strong>the</strong> A!"encan marsup1als, <strong>the</strong> more primitive <strong>for</strong>m of<br />

marsup1al, 1S puzz11ng.(2) In this study we examined <strong>the</strong><br />

neurohypophyseal hormones of <strong>the</strong> bandi coot (1. macrourus) an<br />

Australian marsupial not examined previously.<br />

'<br />

Oxytocin, known to be produced by <strong>the</strong> hypothalamus has now been<br />

obser~ed within <strong>the</strong> .ovary.. ~he testis and <strong>the</strong> adrenal gland.(3) The<br />

loca~10n of oxytoc1n w1th1n <strong>the</strong> reproductive tract of <strong>the</strong> male<br />

band;coot ~as also ~xamined. Tissue was fixed in para<strong>for</strong>maldehyde and<br />

se~t1on~ 1mmunos~a;ned b~ a biotin-streptavidin-peroxidase system,<br />

us;ng h1 gh l~ spec1 f1 c l'abb1 t an.ti -oxytoci n and sheep anti -mesotoci n as<br />

pnmary ant1 sera. Immunoreact1Ve oxytoci n cells were demonstrated in<br />

<strong>the</strong> paraventri cul ar and supra opti c nucl ei of <strong>the</strong> bandi coot<br />

hypoth~lamus. These cells did not stain with <strong>the</strong> mesotocin antibody.<br />

OxytoC1n but not mesotocin was present in <strong>the</strong> Leydig cells and in <strong>the</strong><br />

spermatids of <strong>the</strong> bandicoo~. The bandicoot prostate has two segments,<br />

~he dorsal and v~ntral reg10ns. Oxytocin and mesotocin were present in<br />

l.he.ventral port1on of <strong>the</strong> prostate, but were not present in <strong>the</strong> dorsal<br />

reg1on.<br />

In conclusion, <strong>the</strong>se results show that both oxytocin and mesotocin<br />

are prese~t in Australian marsupials. Fur<strong>the</strong>rmore, <strong>the</strong>se peptides are<br />

not restncted to <strong>the</strong> hypothalamus but are also found in <strong>the</strong> testes<br />

~ oxytoci n1 and prostate (.oxytoci nand mesotoci n) where <strong>the</strong>y coul d be<br />

1nvolved 1n spermatogenes1s, contractility of <strong>the</strong> seminiferous tubules<br />

and in sperm transport in <strong>the</strong> female reproductive tract.<br />

(1) Acher, R. (1980). Proc. Roy. Soc. Lond. Ser B. 210, 21-43.<br />

(2) Chauvet, J., Roui11e, Y., Chauvet, M.T. and Acher, R. (1987). Gen.<br />

Compo Endocr. 67, 399-408.<br />

(3) Nicho~son,.H.D., Swann, R.W., Bur<strong>for</strong>d,.G.D., Wa<strong>the</strong>s, D.C., Porter,<br />

D.G. and P1ckenng·, B.T. (984), Reg. Pept1des 8, 141-146.<br />

89.<br />

THE EFFECT OF DAY LENGTH TREATMENT ON THE<br />

REPRODUCTIVE PERFORMANCE OF BORDER LEICESTER RAMS<br />

C.R. Earl, E.A. Dunstan and M.<br />

Schleuniger<br />

Dept. Ag. S.A. Struan « Kybybolite<br />

Daylength is recognised as <strong>the</strong> principal environmental cue<br />

determining <strong>the</strong> seasonal breeding activity of rams (1,2). In<br />

Australia <strong>the</strong> majority of rams are used <strong>for</strong> mating during long days<br />

at a time when testosterone levels and mating activity are low (3}.<br />

Schanbacher (4) has reported that exposure of rams to short days<br />

(L:D, 8:16) significantly improved <strong>the</strong> reproductive per<strong>for</strong>mance of<br />

rams when singly mated.<br />

In our experiment <strong>the</strong> reproductive per<strong>for</strong>mance of rams exposed to<br />

long (L:D, 16:8) or short (L:D, 10:14) days <strong>for</strong> 12 weeks prior to<br />

mating was compared.<br />

Ten 1 1/2 year old Border Leicester rams were allocated to two<br />

equal groups on <strong>the</strong> basis of liveweight on 16/9/85. Testicular<br />

diameter and liveweight were recorded <strong>for</strong>tnightly prior to mating on<br />

12/12/85. Each ram was individually mated to 40 merino ewes and<br />

reproductive per<strong>for</strong>mance determined by <strong>the</strong> number of foetuses present<br />

and mean lambing day.<br />

Results<br />

~eweight. testicular diameter. foetuses per ewe pregnant. and<br />

meaning mating day <strong>for</strong> rams exposed to long and short day<br />

len ths.<br />

T est<br />

1 Foetuses<br />

c<br />

u<br />

per ewe<br />

1<br />

40 Wt.<br />

a r 5.0 (kg)<br />

60<br />

Mean Lambing<br />

day<br />

0<br />

1 Long day 1.18 143<br />

a<br />

II<br />

e Short day 1.12 143<br />

t 20<br />

e N.S. N.S.<br />

r<br />

( 4.0,....---1._---1._......._ ...._.1-.......<br />

em) 16i9 30/9 15/1027/10 11/11 28/11 12/12<br />

Date<br />

Although testicular diameter was significantly increased by<br />

exposure to short day length no improvement in <strong>the</strong> reproduct.ive<br />

per<strong>for</strong>mance of short day length treated rams was observed. The<br />

proportion of rams to ewes (2 1/2%) is comparable with that used in<br />

commercial practice with maiden rams. It is <strong>the</strong>re<strong>for</strong>e unlikely that<br />

treatment of rams with short day length or with products that mimic<br />

short day length is likely to improve <strong>the</strong>ir reproductive per<strong>for</strong>mance.<br />

(1) Yeates N.T.H. 1949. J. Ag. Sci. 39:1<br />

(2) Ortavant R., Mauleon P., Thibault C. 1964.<br />

Ann. N.Y. Acad Sci. 117:157<br />

(3) D'Occhio H.J., Brooks D.E., 1983 Aust. J. Exp. Anim. Husb. 23:248<br />

(4) Schanbacher B.D. 1979 J. Anim. Sci. 49:927


90 91<br />

EFFECT OF PHOSPHATIDYLSERINE ON CALCIUM UPTAKE OF COLD SHOCKED<br />

BOAR AND RAM SPERMATOZOA<br />

D.P. Windsor and I.G. White<br />

Department of Veterinary Physiology, Univer.sity of Sydney, NSW 2006<br />

It has long been recognised that phospholipids play a role in<br />

protecting mammalian sperm from cold shock. Phosphatidylcholine (PC) has<br />

bee~ shown to protect ram (1) and boar (2) sperm, whil e phosphati dyl­<br />

~er1ne (PS) has been shown to reduce acrosomal damage and motility loss<br />

1n col d shocked boar sperm (3). The aim of this study was to fur<strong>the</strong>r<br />

assess <strong>the</strong> cryoprotective effect of PS on boar sperm by measuring uptake<br />

of radioactive calcium and to test its efficacy in protecting ram sperm.<br />

Boar sperm were flushed from cauda epididymides and ram sperm were<br />

collected by ~lectroej8culation: PS (4 mg/ml) was added 25 minutes prior<br />

to cO]d shock1ng at 0 C. Calc1um uptake was determined at 5, 10, 20 and<br />

40 ffilnutes (2). Data were subjected to a split plot analysis of<br />

variance.<br />

TABLE 1:<br />

Species<br />

Boar<br />

(n=8)<br />

Ram<br />

(n=7)<br />

Ca uptake of sperm (nmoles/10 9 sperm, mean + S.E.)<br />

Control 25+9ab 42+15ab 66+25b<br />

Colq shock 117+8c 129+8c 135+6c<br />

Control+PS 5+1 a 7+2 a 9+2 a<br />

Cold shock+PS 62+21b 66+20b 57+18b<br />

Control 21+7a 33+11a 46+13a<br />

Col d shock 101+21c 125+17b 144+16b<br />

Control+PS 35+16ab 49+19a 54+21a<br />

Cold shock+PS 71+9bc 101+12b 133+25b<br />

Different subscripts denote differences (p < 0.05) within <strong>the</strong><br />

and species.<br />

40min<br />

86+28b<br />

128+7c<br />

7+1 a<br />

30+10a<br />

42+13a<br />

120+10b<br />

52+18a<br />

126+25b<br />

same column<br />

These data confi rm <strong>the</strong> observati on (3) that PS protects boar sperm<br />

from col~ shock. Calcium uptake .(indicating plasma membrane disruption)<br />

was cons1stently lower (p


92 93<br />

FERTILISATION IN SUPEROVULATED EWES FOLLOWING INSEMINATION<br />

WITH DIFFERENT DOSES OF FRESH OR FROZEN-THAWED SEMEN<br />

H.N. Jabbour, G.<br />

Evans and N.W. Moore<br />

Department of Animal Husbandry, The University of Sydney, N.S.W. 2006<br />

Intrauterine insemination is necessary to achieve satisfactory<br />

fertilisation in superovulated ewes. The following study was<br />

designed to examine fertilisation in superovulated ewes inseminated<br />

with various doses of fresh or frozen-thawed spermatozoa.<br />

TABLE 1. Proportion of ova fertilised (no. fertilised/no. recovered)<br />

following insemination with different doses of fresh or frozen-thawed<br />

semen.<br />

100<br />

50<br />

25<br />

12.5<br />

6.2<br />

3.1<br />

SEPARATION OF HUMAN SPERM FOR ELEMENTAL ANALYSIS<br />

G.H. O'Brien a ,<br />

J. Clulow and R.C. Jones<br />

Department of Biological Sciences, University of Newcastle, NSW 2308<br />

O'Brien, Clulow and Jones (1) reported <strong>the</strong> elemental composition<br />

of sperm that were pipetted onto electron microscopy grids and airdried,<br />

as described by Chandler and Battersby (2). The concentrations<br />

obtained in that study suggested serious inadequacies in <strong>the</strong><br />

technique, especially contamination of sperm by extracellular fluid.<br />

We now describe a more appropriate technique developed in this<br />

laboratory, and initial observations obtained with its application..<br />

Semen collected from 3 Merino rams was pooled and <strong>the</strong>n<br />

inseminated ei<strong>the</strong>r fresh after dilution with phosphate buffered<br />

saline (PBS) or after frozen storage in glycerol-egg yolk diluent.<br />

A <strong>for</strong>mvar-coated nickel slot grid was placed in <strong>the</strong> bottom of a<br />

Forty-nine mature Merino ewes were treated with intravaginal<br />

0.5 mlmicrotestube and covered with a layer of di-n-butyl phthalate.<br />

progestagen pessaries (Repromap, Upjohn) <strong>for</strong> 12 days and 1200 IU PMSG<br />

An aliquot of semen was placed on <strong>the</strong> oil and centrifuged <strong>for</strong> 0.5­<br />

(Folligon, Intervet) administered 48 h be<strong>for</strong>e pessary withdrawal<br />

4 min in an Eppendorf centrifuge <strong>the</strong>n <strong>the</strong> grid was rinsed in isopentane<br />

(PW). Intrauterine inseminations were per<strong>for</strong>med by laparoscopy<br />

and freeze-dried at 0.5 x 10- 3 Pa overnight. The nuclei of 5<br />

at 44 h after PW. A total dose of 100X106 , 50x106 , 25x106 , 12.5xl06,<br />

sperm from each of 4 ejaculates (collected from fertile donors) were<br />

6.2xl0 6 or 3.1x10 6 of ei<strong>the</strong>r fresh or frozen-thawed motile<br />

analysed by energy dispersive spectroscopy (X-ray probe microanalysis)<br />

spermatozoa in a total volume of 0.1 ml PBS was deposited in <strong>the</strong><br />

in a scanning electron microscope.<br />

uterine horns. Ova were recovered and ovaries inspected at<br />

Mean concentrations of Na, Mg, P, S, CI, K and Ca are listed in<br />

laparotomy under general anaes<strong>the</strong>sia 92 h after PW. Cleaved and<br />

<strong>the</strong> Table below. Presented with <strong>the</strong>m are results (<strong>for</strong> sperm in semen)<br />

pronucleate ova were classified as fertilised.<br />

from O'Brien et al. (1) converted to <strong>the</strong> same units (mmol.kgrl dry wt)<br />

The mean ovulation rate was 11.9 ± 0.9 and <strong>the</strong> overall recovery<br />

to allow direct comparison.<br />

of ova (no. recovered/no. of ovulation points) was 56.5%: Multiple<br />

X 2 analysis was per<strong>for</strong>med and <strong>the</strong> data are presented in Table 1. Technique used Elemental concentrations in sperm nuclei<br />

to prepare sperm<br />

(mmol.kgrl drywt)<br />

Dose of spermatozoa (X10 6 )<br />

Fresh semen<br />

Frozen-thawed semen<br />

12/15<br />

14/15<br />

27/33<br />

31/34<br />

17/25<br />

14/19<br />

13/22<br />

25/27<br />

14/15<br />

14/32<br />

23/23<br />

21/31<br />

The overall fertilisation rate was 77.3%. Overall, <strong>the</strong>re was no·<br />

effect of <strong>the</strong> type of semen used on fertilisation, but <strong>the</strong>re was an<br />

interaction between <strong>the</strong> type of semen and <strong>the</strong> dose of spermatozoa<br />

inseminated (P


94 95<br />

MORPHOLOGY OF IMMATURE SPERMATOZOA OF THE CHINESE PANGOLIN<br />

(MANIS P.ENTADACTYLA : PHOLIDOTA)<br />

Luke K.-P. Leung and J.M.<br />

Cummins<br />

PIVET Medical Centre, 166 Cambridge Street, Leederville, WA 6007.<br />

Pangolins are testicond eu<strong>the</strong>rian mammals which <strong>for</strong> many years<br />

were classified along with <strong>the</strong> Edentata because of superficial<br />

resemblances to Armadillos and lack of teeth. This affinity is now<br />

discounted, and <strong>the</strong>ir phylogenetic placement is uncertain (1).<br />

Ballowitz (2) first described by light microscopy <strong>the</strong> spermatozoon of<br />

a species of this Order (M. longicaudata - a frozen specimen from W.<br />

Africa), and considered it to be similar to that of monotremes,<br />

reptiles or birds. No subsequent work on <strong>the</strong> sperm morphology appears<br />

to have been published. We were interested in fur<strong>the</strong>r evaluating<br />

Ballowitz's observations using electron microscopy. Material from a<br />

small male M. pentadactyla Was fixed using 2.5% glutaraldehyde in<br />

cacodylate buffer (pH 7.4). Representative areas of <strong>the</strong> reproductive<br />

tract were excised, osmicated and embedded in Araldite follOWing<br />

dehydration in an ethanol-acetone series. Ultrathin sections were<br />

studied by TEM.<br />

The animal was not fully reproductively active, and only a few<br />

seminiferous tubules exhibited elongated spermatids. Immature<br />

spermatozoa were found in <strong>the</strong> ductuli efferentes, but <strong>the</strong> rest of <strong>the</strong><br />

tract was devoid of gametes. Never<strong>the</strong>less, <strong>the</strong> spermatozoa showed<br />

striking similarities with Ballowitz's descriptions <strong>for</strong> M.<br />

longicaudata. The sperm head, unlike that of any o<strong>the</strong>r eu<strong>the</strong>rian<br />

mammal described, is not flattened, but has a pointed, elongated<br />

nucleus (10-12 urn long) with a round cross-section (0.8-1.2 um<br />

diameter). The chromatin in many of <strong>the</strong> spermatozoa of this specimen<br />

was irregularly condensed, a feature also noted by Ballowitz. The<br />

flagellum, midpiece and axoneme are round in cross-section, and <strong>the</strong>re<br />

is a spiral fibrous sheath underlying <strong>the</strong> principal piece<br />

plasmalemma. The axoneme con<strong>for</strong>ms to <strong>the</strong> common 9 outer doublet + 2<br />

central microtubule pattern with 9 accessory coarse fibres extending<br />

along <strong>the</strong> bulk of <strong>the</strong> flagellum. The acrosome (1.7-2.2 um long) has a<br />

largely homogenous matriX, sits cap-like over <strong>the</strong> rostral fifth of<br />

<strong>the</strong> nucleus, with no substantial subaerosomal material, and does not<br />

appear to have a well-defined equatorial segment (ES). Lack of this<br />

feature distinguishes M. pentadactyla from o<strong>the</strong>r eu<strong>the</strong>ria with<br />

flattened sperm heads, where <strong>the</strong> presence of an ES is possibly<br />

related to common features of fertilization (3).<br />

The sperm morphology of this group thus clearly differs from<br />

<strong>the</strong> rest of eu<strong>the</strong>rian mammals. Fur<strong>the</strong>r work is needed to see whe<strong>the</strong>r<br />

<strong>the</strong> ostensible resemblance to <strong>the</strong> sauropsid sperm of monotremes is o.f<br />

taxonomic significance.<br />

(1) Nowak, R.M., Paradiso, J.L. (1983). In, "Walker's Mammals of <strong>the</strong><br />

World" 4th Edn, pp 470-472. Johns Hopkins Press, Baltimore.<br />

(2) Ballowitz, E.(1907). Zeits. f. Wiss.enschaft. Zool. 86:619-625.<br />

(3) Bed<strong>for</strong>d, J.M. (1982). In C.R. Austin and R.V. Short (eds)<br />

"Reproduction in Mammals" 2nd Ed, Vol 1, pp 128-163. C.U.P.<br />

THE CYCLE ot THE SEMINIFEROUS EPITHELIUM OF THE JAPANESE<br />

QUAIL, COTURNIX COTURNIX<br />

M.Lin"', R.C.Jones'" and A.W.Blaekshaw 2<br />

'" Department of Biological Sciences. University of Newcastle. NSW<br />

2 Department of Physiology & Pharmacology. University of<br />

Queensland. Qld.<br />

It is extremely difficult to identify <strong>the</strong> various cellular<br />

associations which constitute <strong>the</strong> cycle of <strong>the</strong> seminiferous<br />

epi<strong>the</strong>lium of many birds with <strong>the</strong> routine histological methods<br />

used <strong>for</strong> mammals and this problem has lead to <strong>the</strong> publication of<br />

some unusual combinations of cellular associations (1). The<br />

problem in <strong>the</strong> bird is that an individual cellular association<br />

covers a very small area of <strong>the</strong> wall of a seminiferous tubule so<br />

that several different adjacent associations overlap and it is<br />

not possible to clearly distinguish <strong>the</strong> germ cells in one<br />

association from <strong>the</strong> germ cells in an adjacent association.<br />

This studY determined <strong>the</strong> cellular associations in <strong>the</strong><br />

Japanese quail using thin epoxy sections of single seminiferous<br />

tubules. Tubules were isolated using a hypodermic syringe and<br />

needle to flush <strong>the</strong>m apart with 0.1 M phosphate buffer.<br />

Subsequently <strong>the</strong>y were fixed in glutaraldehyde and embedded in<br />

Spurr's resin. The cytoplasm of individual Sertoli cells was<br />

traced in 1-2 pm sections stained with toluidine blue. .<br />

Germ cells were classified into 12 steps of spermatId<br />

development. 3 types of spermatogonia, primary spermatocytes in<br />

meiotic phases and secondary spermatocytes. Table 1 shows <strong>the</strong> 10<br />

cellular associations of <strong>the</strong> cycle of <strong>the</strong> seminiferous<br />

epi<strong>the</strong>lium based on <strong>the</strong> morphological changes of <strong>the</strong> acrosome and<br />

nuclei of <strong>the</strong> developing spermatids. There is some uncertainty<br />

about <strong>the</strong> relationship between spermatogonia which can only be<br />

resolved using a radiolabelled tracer.<br />

.r---."..-.·--I-·-.--,r----~ u _.-~'-...------u<br />

11' I 11 I 11 U 12 B 12 I U • H I B<br />

1---1-·-·..··-,-1",·....,·,-···1·..····,,",·-1--.-....---'11---,.11--- 1 1---1<br />

11 I 2 I 3 I 4 B 5 I 6 J 7 I B I 9 1 10 I<br />

I'_'__'_I_'__-II,_"""·_,,~,,,,_~ '_""'·" I-"_""__·_I_"_·"'.._·........---·1----·1---·-1<br />

BPI PIP 1 PIP I P I Dp I Dk I M-An I I I 1<br />

1-"'-'1·-.."".-..-,,,0.-,,••·-.,,·--·1..,,,,-,,"'-'--1-,-----..1,,·,,-,----11·-,--·-·-.----11---1-----1<br />

UB 1 BIB n LIZ 1 PIP 1 PIP I P I<br />

'1----8----1-.-...-,"'1-·-'--1----......----11·---·1---1----1 1<br />

UAp I Ap I Ap I Ap I Ap n Ap I BIB I BIB 1<br />

DAd 1 Ad I Ad I Ad 1 Ad 1 Ad lAd Ap I Ad Api Ad Api Ad A~I<br />

I I H II I I II I IV I V VI I VI I VI I IH IX I X<br />

=-=~.::SYM:BOLEt;=cA'd;::'''·~d';;:)~==t~~~A·-~~7.~~~~~=~~;ti~~:::.A~.~ie ty~~ A<br />

spermatogonia; B. type B spermatogonia; L. leptotene primary<br />

spermatocytes; Z, zygotene primary spermatocytes; P. pachytene<br />

primary spermatocytes; Dp. diplotene primary spermat0cytes; Ok.<br />

diakinesis of primary spermatocyt~s; M. metaphase primary<br />

spermatocytes; An. anaphase primary spermatocytes; 1-12. step 1­<br />

step 12 spermatids; I-X, stages of spermatogenesis.<br />

The<br />

duration<br />

birds.<br />

results provide a<br />

of spermatogenesis<br />

(1) Yamamoto.S; Tamate.H &<br />

Res.1B, 27-37.<br />

basis <strong>for</strong> fur<strong>the</strong>r studies of <strong>the</strong><br />

and renewal of spermatogonia in<br />

Itikawa,O (1967)<br />

Tohoku Agric.


96 97<br />

EFFECT OF RACEMIC GOSSYPOL AND ITS ISOMERS ON SURVIVAL<br />

OF RAM SPERM AND ON REACTIVATION OF SPERM MODELS<br />

R. Vishwanath. I.G. White and S.A. Matlin 1<br />

1 Department of Veterinary Physiology. University of Sydney. NSW. 2006<br />

Chemistry Department. City University. Northampton Sq .• London. ECIV OHB<br />

Gossypol is an optically active molecule and preliminary studies<br />

indicate that <strong>the</strong> (-) isomer renders male rats infertile when given<br />

orally. but <strong>the</strong> (+) isomer is ineffective (1). The aim of this study was<br />

to investigate <strong>the</strong> stereospecificity of <strong>the</strong> action of gossypol on sperm<br />

function in vitro.<br />

Ram sperm were washed free of seminal pl asma by dil uting with<br />

Krebs Ri nger phosphate gl ucose. centrifugi ng. removi ng <strong>the</strong> supernatant<br />

and resuspendi ng <strong>the</strong> sperm pellet. Demembranated sperm models were<br />

prepared by extracti ng <strong>the</strong> washed sperm in Triton-X medi urn and were<br />

reactivated with ATP (2). Motil ity of sperm was scored on a scal e of 1<br />

to 4, and reactivati on of <strong>the</strong> sperm model s as <strong>the</strong> percentage movi ng.<br />

Oxygen uptake of sperm was determined with a Clark electrode and glucose<br />

and lactic acid concentration of <strong>the</strong> medium by enzymatic assays.<br />

Sperm motil ity decreased 1i nearl y with i ncreasi ng concentrations<br />

of racemic (i.e. .:!:.) gossypol or its isomers; 50 IlM of <strong>the</strong> (+) or (-)<br />

isomer or 100 IlM of <strong>the</strong> racemer depressed motility within 10 seconds.<br />

Sperm'motility ceased after 30 minutes exposure to 50 IlM of each of <strong>the</strong><br />

compounds but weak motility persisted <strong>for</strong> 60 minutes in <strong>the</strong> presence of<br />

25 IlM of <strong>the</strong> (+) or (-) isomers. A strong uncoupling (i.e. increase) of<br />

sperm oxygen uptake was evident at low (25 and 50 IlM) concentrations of<br />

racemic gossypol or its isomers, but higher concentrations ([50 and 200<br />

IlM) of racemic gossypol reduced oxygen uptake. At 50 IlM <strong>the</strong> (+) or (-)<br />

isomers decreased glucose utilization of sperm and 100 IlM of each of <strong>the</strong><br />

compounds completely inhibited glucose utilization and lactic acid<br />

accumulation.<br />

Demembranated sperm models were more resistant to gossypol or its<br />

isomers; 25 or 50 IlM had no effect on reactivation of sperm models which<br />

immediately became motile on addition of ATP. At 100 IlM. <strong>the</strong> %<br />

reactivation (mean + S.E .• n=5) was 66.6 + 8.9 <strong>for</strong> <strong>the</strong> racemer. 53.6 +<br />

7.1 <strong>for</strong> <strong>the</strong> (+) and-zero <strong>for</strong> <strong>the</strong> (-) isomer compared to 65.5 + 11.3 <strong>for</strong><br />

<strong>the</strong> control; 200 IlM of <strong>the</strong> compounds inhibited reactivation completely.<br />

Overall, <strong>the</strong> in vitro spermicidal activities of racemic gossypol<br />

and its isomers were not remarkably different. This suggests a stereospecific<br />

in vivo interaction of gossypol at <strong>the</strong> testicular target site<br />

leading to disruption of spermatogenesis.<br />

(1) Wang. N.G., Zhou, L.F .• Guan, M.H. and Lei, H.P. (1987). J.<br />

Ethno~harmacOl.• 20:21-24.<br />

(2) -hite.I.G. and Voglmayr. J.K. (1986). J. Reprod. Fert.• 34:183-193.<br />

Perfusion fluid<br />

DRTF<br />

sRTF<br />

DOES .RETE TESTIS FLUID CONTAIN A SECRETAGOGUE 7<br />

S. SUj~rit1, G. ChaturaP1nich1, H, Lin 1 , !.c. Jones 1 ,<br />

B.P. Setchell ,.and G.M. Stone<br />

1 Department of Biological Sciences, University of Newcastle, NSY 2308<br />

2 Department of Animal Physiology, Yaite Agricultural Research<br />

Institute, University of Adelaide, SA 5064<br />

3 Department of Veterinary Physiology, University of Sydney, NSY2006<br />

Last year we described a method of perfusing <strong>the</strong> initial segment of<br />

<strong>the</strong> epididymis of <strong>the</strong> rat (1). It involved anaes<strong>the</strong>tising rats with<br />

Inactin (Byk Gulden Konstanz, 'West Germany), cannulating region 1A (2)<br />

with polyethylene tubing and perfusing at 0.5 ~l/min.<br />

The present study examined <strong>the</strong> ef£ects of perfusing <strong>the</strong> initial<br />

segment <strong>for</strong> 7.5 h with native rete testis fluid (nRTF) and modifications<br />

of nRTF as weH as a syn<strong>the</strong>tic medium (sRTF) based on <strong>the</strong> inorganic<br />

compositions ofnRTF(3). None of <strong>the</strong> treatments affected <strong>the</strong> structure<br />

of <strong>the</strong> duct epi<strong>the</strong>lium. However., Table 1 shows that protein secretion<br />

was higher when nRTF or a high molecular weight (M'IJ) fraction<br />

(concentrated 3-fold) of nRTF was used to perfuse <strong>the</strong> duct than<br />

modifications- of nRTF which damaged or removed <strong>the</strong> protein, or <strong>the</strong> sRTF<br />

was used. The addition of bovine serum albumin (BSA) to sRTF enhanced<br />

protein secretion into <strong>the</strong> lumen, but it was not as great as with nRTF<br />

or <strong>the</strong> high MY fraction of nRTF.<br />

PAGE of <strong>the</strong> perfusates indicated that <strong>the</strong> composition of <strong>the</strong><br />

perfusion fluid did not effect <strong>the</strong> type of protein secreted.<br />

TABLE 1. Rate of protein secretion into <strong>the</strong> initial segment of <strong>the</strong> rat<br />

epididymis during perfusion <strong>for</strong> 7.5 h wi th nRTF, sRTF or various<br />

modi~ications of <strong>the</strong> fluids. (MEAN ± SE)<br />

Unmodified<br />

Steroid extracted<br />

High MY fraction<br />

Low MY fraction<br />

Trypsinized<br />

Protein-free<br />

1.3 ~g/~l BSA<br />

No. of<br />

animals<br />

It is concluded that nRTF may contain a secretagogue.<br />

7<br />

4<br />

5<br />

3<br />

3<br />

8<br />

6<br />

Rate of protein secretion<br />

(ng/min)<br />

93 ± 22<br />

99 ± 45<br />

145 ± 36<br />

19 ± 3<br />

41 ± 18<br />

25 ± 4<br />

45 ± 21<br />

(1) Sujarit, S. et ~l (1987). Proc. 19 tho Ann. Meet. Aust. Soc. Reprod.<br />

BioI., Sydney, August, p. 90 (Abstract).<br />

(2) Reid, B.L. & Cleland, K.Y. (1957). Aust. J. Zool. ~, 223-246.<br />

(3) Setchell, B.P. et al (1969). J. Physiol. 22, 73-85.


Table 1.<br />

Untreated (n=7)<br />

EDL (n=5)<br />

98<br />

REGULATION OF THE INITIAL SEGMENT OF THE EPIDIDYMIS<br />

S. Sujarit 1 , R.C. Jones 1 , M. Lin 1 , B.P. Setchel1 2 and G.M. Stone 3<br />

1 Department of Biological Sciences, University of Newcastle, NSY 2308<br />

2 Department of Animal Physiology, Yaite Agricultural Research<br />

Institute, University of Adelaide, SA 5064<br />

3 Department of Veterinary Physiology, University of Sydney, NSY 2006<br />

The initial segment (IS) of <strong>the</strong> mammalian epididymis has a<br />

characteristic ultrastructure and <strong>the</strong> epi<strong>the</strong>lium regresses following<br />

efferent duct ligation (EDL). Nicander et al (1) showed that regression<br />

occurs within 6 hours of efferent dUlc~ligation, we developed a<br />

microperfusion technique to study <strong>the</strong> effects in vivo of modifying <strong>the</strong><br />

composi tion of <strong>the</strong> luminal fluids of <strong>the</strong> initialsegment (2).<br />

Mature Yistar rats were anaes<strong>the</strong>tised with Inactin(Byk Gulden<br />

Pharm., Y. Germany) and prepared <strong>for</strong> microperfusion. The duct was<br />

flushed free of sperm wi th a syn<strong>the</strong>tic medium (sRTF) with <strong>the</strong> same<br />

inorganic composi tion as native ram rete testis fluid (nRTF) , and <strong>the</strong><br />

duct was perfused <strong>for</strong> at least 6 hours at 0.5 lli/min with ei<strong>the</strong>r sRTF or<br />

nRTF. The perfusate was analysed quantitively <strong>for</strong> protein and after<br />

perfusion samples of <strong>the</strong> duct were fixed and epoxy sections were<br />

prepared <strong>for</strong> light microscopy.<br />

. It was found that <strong>the</strong> structure of <strong>the</strong> duct epi<strong>the</strong>lium was much <strong>the</strong><br />

same after 7.5 h perfusion wi th nRTF and sRTF .as in unperfused duc ts<br />

whilst 7.5 h after EDL some of <strong>the</strong> structural signs of regression<br />

described by Nicander et al (1) were present. In order to distinguish<br />

<strong>the</strong> effect of EDL from <strong>the</strong> effect of withdrawing <strong>the</strong> luminal fluids from<br />

<strong>the</strong> lumen of <strong>the</strong> IS we examined <strong>the</strong> effect of EDL just prior to<br />

perfusing <strong>the</strong> IS with nRTF (Table i). The reduction in protein<br />

secretion following EDL indicates that <strong>the</strong> main effect of EDL on <strong>the</strong><br />

initial segment· may not be due to <strong>the</strong> withdrawal of <strong>the</strong> luminal fluids.<br />

Protein secreted into <strong>the</strong> IS perfused with nRTF (ng/min).<br />

Perfusion time (h)<br />

1.5-3.5 3.5-5.5 5.5-7.5<br />

76+1. 9<br />

70+0.6<br />

105+5.9<br />

68+0.2<br />

97+2.6<br />

31+0.7<br />

(1) Nicander, L. et al (1983) Int. J. Androl. 6, 91-103.<br />

(2) Sujarit, S. et al (1987) Proc. 19th Ann. Meet. ASRB, Sydney,<br />

August 24-26.<br />

99<br />

THE EFFECTS OF BILATERAL CASTRATION AND ETHYLENE DIMETHANE<br />

SULPHONATE (EDS) ON EPIDIDYMAL FUNCTION (SPERMATOZOAL<br />

MATURATION) IN GUINEA-PIG<br />

S.J. Gatie 1 , A.W. Blackshaw 1 and T.D. Glover 2<br />

lDepartment of Physiology and Pharmacology, University of<br />

Queensland, St Lucia, Qld and 233 Lyddon Tee, Leeds,<br />

LS2 9JT, West Yorkshire, England<br />

Epididymal function is androgell dependent (1) and <strong>the</strong> secretory<br />

functionof <strong>the</strong> rat epididymal epi<strong>the</strong>lial cells is inhibited by<br />

castration. After EDS treatment in <strong>the</strong> rat, testosterone is decreased<br />

to <strong>the</strong> castrated level (2) and <strong>the</strong> present studies examine <strong>the</strong> effects<br />

of androgen withdrawal and replacement on spermatozoal maturation<br />

(rouleaux <strong>for</strong>mation) in <strong>the</strong> guinea-pig.<br />

Five groups were used in <strong>the</strong> bilateral castration study with<br />

five guinea-pigs in each group: group 1 - castrated <strong>for</strong> 1 week + 10 mg<br />

(SIC) of testosterone propionate (TP) daily: group 2 - castrated <strong>for</strong> 1<br />

week without TP: group 3 castrated <strong>for</strong> 2 weeks + TP: group 4­<br />

castrated <strong>for</strong> 2 weeks without TP and group 5 - sham operated control.<br />

Three groups were used in <strong>the</strong> EDS study (five animals in each. group) ,<br />

group 1 - received one dose of 75 mg EDS (liP): group 2 -received 2<br />

doses of EDS two weeks apart and group 3 - received vehicle only. At<br />

<strong>the</strong> end of each period of <strong>the</strong> two studies, epididymides and also<br />

testes in <strong>the</strong> EDS treatment, were removed and processed <strong>for</strong><br />

histological study. Glycoproteins in groups 2 and 3 of <strong>the</strong> EDS study<br />

<strong>for</strong> epididymal tissues and smears <strong>for</strong> epididymal spermatozoa were<br />

examined using lectins. Protease activity of . acrosomes in smears of<br />

spermatozoa was studied using gelatine coated slides.<br />

In castrated animals: without testosterone (TP) replacement, <strong>the</strong><br />

epididymal epi<strong>the</strong>lium was thin and atrophic with nuclear degeneration.<br />

Stereocilia were absent from <strong>the</strong> luminal surface. Spermatozoa,<br />

contained in <strong>the</strong> terminal segment, were totally separated from<br />

rouleaux; some were decapitated and <strong>the</strong> concentration was reduced.<br />

Epi<strong>the</strong>lial and spermatozoal changes were prevented in animals<br />

recovering TP. EDS treatment demonstrated almost <strong>the</strong> same histological<br />

and spermatozoal pictures as in <strong>the</strong> castrated group. The testes<br />

showed massive damage to <strong>the</strong> seminiferous tubuleS with only a few<br />

degenerated germ cells present. Lectin binding to glycoproteins was<br />

reduced markedly in <strong>the</strong> epididymal tissue and spermatozoa after EDS<br />

treatment. Protease activity in <strong>the</strong> acrosomes was almost abolished in<br />

<strong>the</strong> seminal smears collected from EDS treated guinea-pigs.<br />

The present observations confirm <strong>the</strong> view (1) that androgens<br />

control epididymal function in spermatozoarmaturation.<br />

(1) Moore, H.D.M., & Bed<strong>for</strong>d, J.M. (1979) Anat. Rec. 193: 293-312.<br />

(2) Morris, I.D., Phillips, D.M., & Bardin, C.W. (1986) Endocrinology<br />

118: 709-7.19.


--~-------------------------------------------------------------------<br />

Fluid Na Cl K Protein<br />

lJlIh nmoles/h nmoles/h nmoles/h lJg/h<br />

Testicular output 221.9 40,000 38,000 2,800 355.2<br />

Reabsorption in <strong>the</strong>:<br />

Ductuli efferentes 205.7 30,OOOU 26,0000 2000 365.7<br />

Ductus epididYJllidis 0.4 87U 54D 12D 0.6<br />

Ductus deferens 0.4 63U 480 60 ..,.0.8<br />

The data in Table 1 indicate that most of <strong>the</strong> fluid and solutes<br />

produced by <strong>the</strong> testis are reabsorbed in <strong>the</strong> ductuli efferentes.<br />

Subsequent rates of reabsorption in <strong>the</strong> more distal ducts are very<br />

low. NaCl is <strong>the</strong> principal solute produced by <strong>the</strong> testis, and <strong>the</strong><br />

principal solute of <strong>the</strong> reabsorbate. from <strong>the</strong> ductuli efferentes.<br />

Reabsorp~ion of Na is against <strong>the</strong> electrochemical gradient in all<br />

regions of <strong>the</strong> ducts and may indicate that water and solute<br />

reabsorption is secondary to active transport of sodium.<br />

100<br />

FLUID AND SOLlfl'E FLUXES IN THE GENITAL DUCTS OF THE<br />

MALE JAPANESE QUAIL<br />

J. Clulow and R.C. Jones<br />

Dept. of Biological Sciences, University of Newcastle, N.S.W., 2308.<br />

The extratesticular genital ducts of <strong>the</strong> Japanese quail are<br />

involved in <strong>the</strong> transport, concentration, maturation and transient<br />

storage of spermatozoa released from <strong>the</strong> testis (1). The<br />

transepi<strong>the</strong>lial transport of fluid and electrolytes is, presumably, of<br />

fundamental importance in <strong>the</strong>se processes through <strong>the</strong> reabsorption of<br />

fluid and <strong>the</strong> modification of <strong>the</strong> luminal milieu of spermatozoa, but<br />

has not been well characterized in any avian species. Consequently,<br />

testicular output and transmural fluxes were studied in <strong>the</strong> quail.<br />

Net fluid fluxes were determined by stereological procedures (2).<br />

Solute concentrations were determined by X-ray microanalysis <strong>for</strong><br />

elements (3) and by <strong>the</strong> Goomassie procedure <strong>for</strong> protein (4).<br />

Transmural solute fluxes were calculated from data <strong>for</strong> solute<br />

concentrations and net fluid fluxes.<br />

Table 1 Estimates of testicular output of fluid and solutes and rates<br />

of reabsorption (positive values) and secretion (negative values) in<br />

<strong>the</strong> extratesticular ducts. U indicates transport against, 0 transport<br />

down <strong>the</strong> electrochemical gradient.<br />

It is concluded that <strong>the</strong> active reabsorption of water and solutes<br />

in <strong>the</strong> ductuli efferentes concentrates spermatozoa in <strong>the</strong> ductus<br />

epididymidis and ductus deferens, which <strong>the</strong>n require only low rates of'<br />

absorption of inorganic electrolytes and secretion of protein to<br />

complete sperm maturation and to store spermatozoa.<br />

(1) Clulow, J. and Jones, R.C. (1982) J. Reprod. Fert. 64, 259-266.<br />

(2) Clulow, J. and Jones, R.C. (1988) J. AnaL In press~<br />

(3) Clulow, J. and Jones, R.C. (1986) Proc. 9th Aus. E.M. Conf.p24.<br />

(4) Brad<strong>for</strong>d, M.M. (1976) Analyt. Biochem. 72, 248-254.<br />

101<br />

ORIGIN OF LUMINAL PROTEINS IN THE EPIDIDYMIS OF·THE TAHHAR,<br />

~<br />

Q. Chaturapanich and R.C. Jones<br />

MACROPUS<br />

Department of Biological Sciences, University of Newcastle, NSW 2308<br />

We have studied <strong>the</strong> origins of <strong>the</strong> proteins in <strong>the</strong> epididymis of<br />

<strong>the</strong> tammar by comparing PAGE patterns of micropuncture samples of<br />

luminal fluids (LF) from <strong>the</strong> epididymis and rete testis (1), perfusates<br />

after microperfusion of <strong>the</strong> duct (2), blood plasma and spermatozoa, and<br />

autoradiographs (AR) of ~ubation media following protein syn<strong>the</strong>sis in<br />

vitro in <strong>the</strong> presence of S-methionine.<br />

--Table 1 shows <strong>the</strong> occurrence of proteins which were i!l <strong>the</strong><br />

epididymal fluids, but were not present in blood plasma or rete testis<br />

fluid. The autoradiographs revealed that <strong>the</strong>re were a·lso la,belled<br />

protein bands, with apparent melecular weights of 43, 37, 34.5, 27 and<br />

14.4 which migrated to <strong>the</strong>same'position on <strong>the</strong> gels as blood proteins.<br />

PAGE' of spermatozoa showed that .<strong>the</strong>y lost 3 bands (corresponding to MW's<br />

47, 29.4 and 28) and gainedonoe (corresponding to aMWof 30.4) during<br />

passage through <strong>the</strong> epididymis.<br />

Table 1. Protein secretion by <strong>the</strong> epididymis of <strong>the</strong> tammar determined by<br />

different methods.<br />

A\l1<br />

Caput &corpus<br />

LF<br />

Perfusate<br />

AR<br />

90.675.76356.339.8 32.3 31.3 30 21.418.717.412.812.110.9<br />

epididymis<br />

Cauda epididymis<br />

LF + +<br />

Perfusate<br />

AR + + +<br />

+ + + + + + + + +<br />

+ + + + $<br />

* + + + + + + +<br />

+ +<br />

+<br />

+ +<br />

'l' secreted by caput only if secreted by corpus only<br />

It is concluded from <strong>the</strong> in vitro incubation studifes hthad t tt he<br />

hi' t"h same in all regions 0 t e uc us<br />

pai~~~nm~:iSProt:~ns:::\~S t~e l.;rot:ins have <strong>the</strong> same MW as proteins in<br />

~iood,yaddi~io~aldproJeinsr:~edth~;ethSeh~:nct~~ ~~~ t~~ii~:.li~:v~~t~:~;:s~<br />

may be sYb nt es ze dand St~Cat some proteins which are syn<strong>the</strong>si~ed and<br />

it cane cone l u e . C" f gels of<br />

secreted in vitro are not secret~~ ~~divci:tO~s th~~pa::bsso;an~es in <strong>the</strong><br />

i~:i~:i i~f1sa:: t:ro~~~1;t~~~ec~~n1 <strong>the</strong> ~~:ep::t:irnosterie;e~~~~e~~~~~~:<br />

<strong>the</strong> MW of some of <strong>the</strong> secrete pro e ns. be art of <strong>the</strong><br />

sperm do not appear in <strong>the</strong> ePhidiod:mt;se~l~Smath~od~~~ epiihelium (3).<br />

cytoplasmic droplet which is p:g Yt . n of ~lJ 30 binds to spermatozoa.<br />

It is suggested that <strong>the</strong> secrete pro e1.<br />

Jones R.C. (1987) J. Reprod. Fert. 80: 193-199.<br />

~~~ Chatu~apanich, G. and R.C. Jones (1987) Proc. 19th Ann. Conf.<br />

ASRB Sydney, p.75. . (1984) C 11 Tiss<br />

(3) Jones: R.C., Hinds, L.A. and Tyndale-Blscoe, C.H. e.·<br />

Res. 237: 525-535.<br />

+<br />

+<br />

+<br />

+


102<br />

PROBLEMS IN SCREENING AZOOSPERMIC PATIENTS FOR CORRECTIVE<br />

MICROSURGERY<br />

P.D. Temple-Smith & GJ. Southwick<br />

Department ofAnatomy, Monash University, Clayton, Victoria 3168<br />

Azoospermia associated with normal FSH levels and testicular volumes is<br />

diagnosed as obstructive azoospermia and scrotal exploration with corrective surgery is<br />

usually <strong>the</strong> only option available to reinstate fertility. This paper discusses<br />

difficulties in preoperative diagnosis of efferent duct agenesis or germ cell arrest in .<br />

azoospermic patients. Failure to identify <strong>the</strong>se causes of azoospermia results in<br />

unnecessary and unsuccessful surgery in a small number of azoospermic patients.<br />

In each patient, azoospermia was confirmed by semen analysis. Semen fructose<br />

and serum FSH, LH and testosterone levels were measured preoperatively. The<br />

presence of vas and epididymis, and an estimation of testicular size, were determined<br />

by preoperative clinical examination. Spermatogenic activity of <strong>the</strong> testis was<br />

assessed from testicular biopsies and testicular volumes were confirmed from testicular<br />

measurements taken during scrotal exploration.<br />

Corrective surgery was not possible in 16 of 139 patients recommended <strong>for</strong><br />

scrotal exploration and epididymovasostomy during <strong>the</strong> period 1980 to 1987. In 12<br />

patients microdissection showed that <strong>the</strong> epididymis was completely disconnected from<br />

<strong>the</strong> testis and in <strong>the</strong> remaining group of four azoospermia resulted from spermatogenic<br />

arrest at .<strong>the</strong> primary spermatocyte stage (2) and germ cell aplasia (2). In <strong>the</strong> twelve<br />

patients with testiculo-epididymal discontinuity, five had normal testicular activity,<br />

five had spermatogenic arrest and <strong>the</strong> remaining two had germ cell aplasia. In all<br />

patients <strong>the</strong> epididymis showed no signs of obstruction, and hormone· levels and<br />

testicular size were within <strong>the</strong> normal range, except <strong>for</strong> two patients with germ cell<br />

aplasia who had raised FSH levels (>7IU/I).<br />

As with germ cell aplasia (1) failure to identify patients with bilateral agenesis<br />

of <strong>the</strong> efferent ducts of germ cell arrest using standard preoperative clinical and<br />

laboratory assessment procedures results in unnecessary surgery. FSH levels and<br />

testicular volumes are a guide <strong>for</strong> patients with germ cell aplasia but <strong>the</strong>se parameters<br />

were not able to distinguish patients with germ cell arrest or disconnection of <strong>the</strong><br />

epididymis from <strong>the</strong> testis. Fur<strong>the</strong>r studies are needed to develop. simple diagnostic<br />

tests which can be used to identify preoperatively <strong>the</strong>se causes ofazoospermia.<br />

(1) Jequier,A.M., Ansell, l.D. & Bullimore, N.J. (1984). Brit. J. Urol, 56: 537-539.<br />

MECHANISM OF ACTION OF INH1BIN ON GnRH-INDUCED RELEASE OF FSH AND LH<br />

103<br />

Wang Qi Fa, P.G. Farnworth, H.G. Burger and J.K. Findlay<br />

Medical Rese~rch Centre, Prince Henry's Hospital Campus of<br />

The Monash Medical Centre, 5t Kilda Road, Melbourne, Victoria, 3004.<br />

Inhibin, a gonadal glycoprotein hormone, suppresses GnRH-mediated<br />

FSH and LH secretion by a mechanism that ma.y·be common to both<br />

gonadotrophins (1), in addition to inhibiting FSH biosyn<strong>the</strong>sis. We<br />

previously observed that inhibin treatment decreased <strong>the</strong> number of<br />

GnRH binding sites in cultured pituitary cells with no change in<br />

receptor affinity (2). In <strong>the</strong> present study, we have used various<br />

secretagogues, which stimulate gonadotrophin release by acting beyond<br />

<strong>the</strong>GnRH receptor, to fur<strong>the</strong>r characterize <strong>the</strong> anti-GnRH action of<br />

inhibin.<br />

Primary cultures of anterior pituitary cells were prepared using<br />

tissue from adult male Sprague-Dawley rats. On day 2 of culture, t.h~'<br />

cells were treated with pure 31 kDa bovine inhibin (0.1-3 U/ml) or <strong>the</strong><br />

protein syn<strong>the</strong>sis inhibitor, cyc:J,oheximid:e (0.01-100 llM), <strong>for</strong>.


104<br />

INDUCTION OF LH SURGES BY OESTROGEN IN OVARIECTOMIZED<br />

(OVX) EWES REQUIRES A LARGE AMPLITUDE PULSE OF GnRH<br />

OR A RAPID VOLLEY OF PULSES.<br />

D.!. Phillips!, IT. Cummins 2 and 1.1 Clarke 3 .<br />

lSchool of Agriculture & Forestry, University ofMelbourne, Parkville, 3052.<br />

2St. Vincent's Hospital, Fitzroy, 3065.<br />

3Medical Research Centre, Prince Henry's Hospital, Melbourne, 3004.<br />

We have reported previously a modest 2-3 fold rise in plasma LH levels in<br />

OVX-hypothalamo-pituitary disconnected (HPD) ewes receiving hourly GnRH<br />

pulses and 17-19 hours after an oestradiol benzoate (OB) injection (1). Thus<br />

!he 10-20 fold rise in pl~sma LH concentrations seen in hypothalamo-pituitary<br />

mtact (HPI) OVX ewes gIVen OB reflects more than just a change in pituitary<br />

responsiveness to GnRH. In this study we tested whe<strong>the</strong>r <strong>the</strong> LH surge may be<br />

initiated by a large pulse of GnRH or a rapid volley ofpulses.<br />

Six OYX-HPD ewes were given 250 ng pulses of GnRH (peninsula<br />

Laboratories, U.S.A.) each hour and 50 ~g OB (Lm.) at t=O hours. Blood samples<br />

were taken each 15 minutes <strong>for</strong> 15 hours beginning at t=16 hours. Two GnRH<br />

pulse patterns were employed, with 3 ewes per treatment, after which 3 of <strong>the</strong><br />

6 ewes received <strong>the</strong> reverse treatment <strong>the</strong> following week. Treatment A<br />

involved a bolus injection of 2.25 ~g GnRH at t=16 hours, <strong>the</strong>n half-hourly 250<br />

ng pulses from t=1630-3100 hours. Treatment B involved a volley of 500 ng<br />

GnRH pulses each 10 minutes from t=1600-1630, a 500 ng pulse at 1645 hours<br />

and <strong>the</strong>n half-hourly 250 ng pulses from 1700-3100 hours. In HPI-OYX ewes<br />

(n=lO) plasma LH concentrations were measured in half-hourly samples taken<br />

across OB-induced LH surges. The area under LH vs time curves were<br />

computed in all ewes across <strong>the</strong> LH surge (Table 1). There were no significant<br />

differences in <strong>the</strong> responses of <strong>the</strong> 3 groups.<br />

Table 1. Plasma LH responses (mean +/- s.e.m.) in OVX-HPI and GnRH-pulsed<br />

OYX-HPD ewes given 50 ~g OB.<br />

Group<br />

OVX-HPD; Treatment A (bolus)<br />

OVX-HPD; Treatment B (volley)<br />

OVX-HPI<br />

In HPX-OVX ewes, both treatments were able to release an eqUivalent<br />

amount of LH to that obtained in OVX-HPI ewes given OB, with GnRH pulse<br />

patterns similar to those measured during OB-induced LH surges in HPI ewes<br />

(2). This suggests that initiation of <strong>the</strong> positive-feedback event requires a<br />

large amplitude GnRH pulse or a rapid volley of pulses.<br />

(1) Clarke, 1.1 & Cummins, IT. (1984) Neuroendocrinology 3..2.: 267-274.<br />

(2) Clarke, 1.1 & Cummins, IT. (1985) Endocrinology ill: 2376-2383.<br />

N<br />

5<br />

4<br />

10<br />

Response; Area under<br />

curve (n&/ml h)<br />

259 +/- 55<br />

250 +/- 24<br />

346 +/- 37<br />

105<br />

REGULATION OF PITUITARY LEVELS OF MESSENGER RNA (mRNA) FOR THE<br />

SUBUNITS OF FOLLICLE STIMULATING HORMONE (FSH) AND LUTEINIZING<br />

HORMONE (LH) BY GONADOTROPHIN-RELEASING HORMONE (GnRH) IN THE EWE<br />

Julie E. Mercer and lain J. Clarke<br />

Medical Research Centre, Prince Henry's Hospital,<br />

Melbourne, Australia 3004<br />

Secretion of LH and FSH often shows divergent patterns. In<br />

hypothalamo~pituitary disconnected (HPD) ewes LH secretion falls<br />

rapidly, while FSH secretion continues <strong>for</strong> a number of days following<br />

GnRH withdrawal. Similarly, in HPD ewes treated wi th a constant<br />

infusion or small pulses (25 ng) of GnRH, LH secretion diminishes much<br />

more rapidly than does FSH (1,2). To determine <strong>the</strong> extent to which<br />

<strong>the</strong>se secretory patterns reflect ongoing gene transcription we have<br />

moni tored mRNA levels <strong>for</strong> LHt3, FSH/3 and <strong>the</strong> cdmmon a. subunit in <strong>the</strong>se<br />

circumstances.<br />

Pituitary glands were collected from OVD/HPD ewes which received a) no<br />

fur<strong>the</strong>r treatment,b) 250 Rg pulses of GnRH each 2h, c) 25 ng pulses<br />

of GnRH, d) cons taRt infusion of 250 ng/h of GuRH, e) one week of 250<br />

ngl2 h GnRH pulses and withdrawal of GnRH <strong>for</strong> 30 h. Total RNA was<br />

prepared from individual pituitaries and analyzed on Nor<strong>the</strong>rn blots.<br />

Table 1. Relative pituitary mRNA levels <strong>for</strong> FSHI3, LH/3 and a.-subunit<br />

GROUP<br />

OVX/HPD 6<br />

OVX/HPD+GnRH (250 ng/2 h) 8<br />

OVX/HPD+GnRH (25 ng/h) 4<br />

OVX/HPD+GnRH 3<br />

(constant infusion, 250 ng/h)<br />

OVX/HPD+GnRH (pump off 30 h) 4<br />

FSHI3 LH(3 a.<br />

0.30+0.07 0.19++0.0q 0.42+0.13<br />

2.06+0.38 5.46+1.17 4.69+1.47<br />

3.63+0.64 3.55+0.95 3.77+0.33<br />

1. 36+0.16 1.43+0.21 2.15+0.45<br />

2.90+0.46 3.89+0.54 2.50+0.26<br />

GnRH (250 ng pulses) in <strong>the</strong> OVX/HPD ewe significantly elevated mRNA<br />

levels <strong>for</strong> all gonadotrophin subunits. 25 ng pulses fur<strong>the</strong>r increased<br />

FSH/3 mRNA levels over those seen with 250 ng GnRH while levels of LH(3<br />

and a..mRNA did not fur<strong>the</strong>r change. Withdrawal of GnRH <strong>for</strong> 30 h does<br />

not change FSHj3 or LH(3 mRNA levels. In contrast, administrati.on of a<br />

constant infusion of GnRH (250 ng/h) lowers mRNA levels <strong>for</strong> both<br />

subun~ts compared with pulsatile administration.<br />

These data show that secretion of <strong>the</strong> gonadotrophins does not<br />

necessarily reflect cellular mRNA levels, in that LH(3 mRNA levels<br />

remain elevated in <strong>the</strong> face of treatments which reduce or abolish<br />

secretion. High amplitude pulses of GnRH are required <strong>for</strong> maintained<br />

secretion of LB, whereas lower doses are sufficient to maintain LH(3<br />

mRNA levels. For FSB, secretion appears to be more closely coupled to<br />

cellular FSH/3 mRNA levels, as evidenced by both <strong>the</strong> pump-off and<br />

constant infusion studies.<br />

1. Clarke lJ &Cummins JT. J. Endocrinol. 113:413-418, 1987.<br />

2. Clarke et al. J. Endocrinol. 111:43-49, 1986.<br />

n


PMSG (iu)<br />

oFF (rnl) 0 7.5 10 15 20<br />

0 0 8A a 18.2 b 39.6~ 64Ad<br />

1.5 6A a 8.6 a 23.6 39.8 b<br />

% inhibition 24 53 40 38<br />

a,b,c,d, differ at P


108<br />

INVOLVEMENT OF PROSTAGLANDINS IN PREMATURE LUTEOLYSIS<br />

IN THE SUPEROVULATED GOAT<br />

R.M.. Battye, A.W.N. Cameron,<br />

Regression<br />

Fig. 1<br />

R.J.<br />

Fairclough,l A.O Trounson<br />

C 1<br />

E.H.D. Monash Medical Centre, Clayton, Vic.,<br />

Animal Research Institute, Werribee, Vic.<br />

of corpora lutea within 6 days of superovulation in goats<br />

results in low rates of embryo recovery (1). The cause of early luteal<br />

regression in <strong>the</strong> goat has not been elucidated. However, luteal<br />

regression in <strong>the</strong> naturally cycling goat is characterised by surges in <strong>the</strong><br />

concentration of 13, 14-d1hydro-15-keto-postaglandin F2~(PGFM) (2). The<br />

administration of <strong>the</strong> PGF syn<strong>the</strong>tase inhibitor. indomethacin, suppressed<br />

PGFM peaks and prolonged <strong>the</strong> oestrous cycle. This study investigated<br />

whe<strong>the</strong>r prostaglandins were <strong>the</strong> cause of prematurely regressing corpora<br />

lutea in <strong>the</strong> superovulated doe.<br />

Ten does were superovulated in June 1987, by treatment with<br />

intravaginal progestagen impregnated sponges (Repromop, Upjohn, 60 mg<br />

medroxy progesterone) <strong>for</strong> 16 days and 1200 iu PMSG (Pregnacol Heriot J,<br />

Agencies). administered 1m, 2 days be<strong>for</strong>e sponge removal. Six does were ~<br />

injected twice daily with <strong>the</strong> prostaglandin syn<strong>the</strong>tase inhibitoX-f~nixlJl ~<br />

meglumine (Finadyne, Schering) at 2.2 mg~·7rom·-·Day3-7 of <strong>the</strong><br />

synchronized" cycle. Four does served h "untreated controls. Jugular<br />

blood samples were collected from 2 days be<strong>for</strong>e sponge removal to Day 7.<br />

On Days 4 and 5 blood was sampled hourly between 0900 and 1700h.<br />

Laparoscopy was conducted on Day 7 to determine <strong>the</strong> incidence of corpus<br />

luteum regression.<br />

Significant peaks of PGFM were detected in <strong>the</strong> 4 untreated does on<br />

days 4 or 5 or both but only in 2 of <strong>the</strong> treated females on <strong>the</strong>se days<br />

(P

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

Saved successfully!

Ooh no, something went wrong!