Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Biol Reprod ; 96(6): 1231-1243, 2017 Jun 01.
Article En | MEDLINE | ID: mdl-28520915

The orphan nuclear receptor, liver receptor homolog-1 (aka Nuclear receptor subfamily 5, Group A, Member 2 (Nr5a2)), is widely expressed in mammalian tissues, and its ovarian expression is restricted to granulosa cells of activated follicles. We employed the floxed Nr5a2 (Nr5a2f/f) mutant mouse line and two granulosa-specific Cre lines, Anti-Müllerian hormone receptor- 2 (Amhr2Cre) and transgenic cytochrome P450 family 19 subfamily A polypeptide 1 (tgCyp19Cre), to develop two tissue- and time-specific Nr5a2 depletion models: Nr5a2Amhr2-/- and Nr5a2Cyp19-/-. In the Nr5a2Cyp19-/- ovaries, Nr5a2 was depleted in mural granulosa, but not cumulus cells. We induced follicular development in mutant and wild-type (control, CON) mice with equine chorionic gonadotropin followed 44 h later treatment with human chorionic gonadotropin (hCG) to induce ovulation. Both Nr5a2Amhr2-/- and Nr5a2Cyp19-/- cumulus-oocyte complexes underwent a reduced degree of expansion in vitro relative to wild-type mice. We found downregulation of epiregulin (Ereg), amphiregulin (Areg), betacellulin (Btc) and tumor necrosis factor stimulated gene-6 (Tnfaip6) transcripts in Nr5a2Amhr2-/- and Nr5a2Cyp19-/- ovaries. Tnfaip6 protein abundance, by quantitative immunofluorescence, was likewise substantially reduced in the Nr5a2-depleted model. Transcript abundance for connexin 43 (Gja1) in granulosa cells was lower at 0 h and maximum at 8 h post-hCG in both Nr5a2Amhr2-/- and Nr5a2Cyp19-/- follicles, while Gja1 protein was not different prior to the ovulatory signal, but elevated at 8 h in Nr5a2Amhr2-/- and Nr5a2Cyp19-/- follicles. In both mutant genotypes, oocytes can mature in vivo and resulting embryos were capable of proceeding to blastocyst stagein vitro. We conclude that Nr5a2 is essential for cumulus expansion in granulosa cells throughout follicular development. The disruption of Nr5a2 in follicular somatic cells does not affect the capacity of the oocyte to be fertilized by intracytoplasmic sperm injection.


Cumulus Cells/physiology , Ovary/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Sperm Injections, Intracytoplasmic/methods , Animals , Connexin 43/genetics , Connexin 43/metabolism , Estrous Cycle , Female , Fertilization/physiology , Gene Deletion , Gene Expression Regulation/physiology , Male , Mice , Oocytes/physiology , Ovary/drug effects , Receptors, Cytoplasmic and Nuclear/genetics
2.
Biol Reprod ; 95(1): 13, 2016 07.
Article En | MEDLINE | ID: mdl-27281705

The mammalian target of rapamycin (Mtor) gene encodes a serine/threonine kinase that acts as a master regulator of processes as diverse as cell growth, protein synthesis, cytoskeleton reorganization, and cell survival. In the testis, physiological roles for Mtor have been proposed in perinatal Sertoli cell proliferation and blood-testis barrier (BTB) remodeling during spermatogenesis, but no in vivo studies of Mtor function have been reported. Here, we used a conditional knockout approach to target Mtor in Sertoli cells. The resulting Mtor(flox/flox); Amhr2(cre/+) mice were characterized by progressive, adult-onset testicular atrophy associated with disorganization of the seminiferous epithelium, loss of Sertoli cell polarity, increased germ cell apoptosis, premature release of germ cells, decreased epididymal sperm counts, increased sperm abnormalities, and infertility. Histopathologic analysis and quantification of the expression of stage-specific markers showed a specific loss of pachytene spermatocytes and spermatids. Although the BTB and the ectoplasmic specializations did not appear to be altered in Mtor(flox/flox);Amhr2(cre/+) mice, a dramatic redistribution of gap junction alpha-1 (GJA1) was detected in their Sertoli cells. Phosphorylation of GJA1 at Ser373, which is associated with its internalization, was increased in the testes of Mtor(flox/flox); Amhr2(cre/+) mice, as was the expression and phosphorylation of AKT, which phosphorylates GJA1 at this site. Together, these results indicate that Mtor expression in Sertoli cells is required for the maintenance of spermatogenesis and the progression of germ cell development through the pachytene spermatocyte stage. One mechanism of mTOR action may be to regulate gap junction dynamics by inhibiting AKT, thereby decreasing GJA1 phosphorylation and internalization. mTOR regulates gap junction alpha-1 protein distribution in Sertoli cells and is necessary for progression through the pachytene spermatocyte stage.


Connexin 43/metabolism , Gap Junctions/metabolism , Sertoli Cells/metabolism , Spermatogenesis/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Male , Mice , Mice, Knockout , Phosphorylation , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , Testis/metabolism
3.
Theriogenology ; 74(6): 912-21, 2010 Oct 01.
Article En | MEDLINE | ID: mdl-20546885

The objective of this study was to determine if pulsatile LH secretion was needed for ovarian follicular wave emergence and growth in the anestrous ewe. In Experiment 1, ewes were either large or small (10 x 0.47 or 5 x 0.47 cm, respectively; n = 5/group) sc implants releasing estradiol-17 beta for 10 d (Day 0 = day of implant insertion), to suppress pulsed LH secretion, but not FSH secretion. Five sham-operated control ewes received no implants. In Experiment 2, 12 ewes received large estradiol-releasing implants for 12 d (Day 0 = day of implant insertion); six were given GnRH (200 ng IV) every 4 h for the last 6 d that the implants were in place (to reinitiate pulsed LH secretion) whereas six Control ewes were given saline. Ovarian ultrasonography and blood sampling were done daily; blood samples were also taken every 12 min for 6 h on Days 5 and 9, and on Days 6 and 12 of the treatment period in Experiments 1 and 2, respectively. Treatment with estradiol blocked pulsatile LH secretion (P < 0.001). In Experiment 1, implant treatment halted follicular wave emergence between Days 2 and 10. In Experiment 2, follicular waves were suppressed during treatment with estradiol, but resumed following GnRH treatment. In both experiments, the range of peaks in serum FSH concentrations that preceded and triggered follicular wave emergence was almost the same as control ewes and those given estradiol implants alone or with GnRH; mean concentrations did not differ (P < 0.05). We concluded that some level of pulsatile LH secretion was required for the emergence of follicular waves that were triggered by peaks in serum FSH concentrations in the anestrous ewe.


Anestrus , Luteinizing Hormone/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/physiology , Sheep/metabolism , Sheep/physiology , Algorithms , Anestrus/blood , Anestrus/metabolism , Anestrus/physiology , Animals , Cell Size/drug effects , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Drug Implants , Estradiol/administration & dosage , Female , Follicle Stimulating Hormone/blood , Infusions, Subcutaneous , Luteinizing Hormone/blood , Ovarian Follicle/cytology , Ovulation/blood , Ovulation/drug effects , Pulsatile Flow , Sheep/blood
4.
Biol Reprod ; 83(1): 122-9, 2010 Jul.
Article En | MEDLINE | ID: mdl-20335640

Large antral follicles grow in waves in the ewe, and each wave is triggered by a peak in serum concentrations of FSH. The existence of follicular dominance in the ewe is unclear. The objective of experiment 1 was to determine if an endogenously driven follicular wave could emerge during the growth phase of a wave induced by injection of ovine FSH (oFSH). Cyclic ewes (n = 7) were given oFSH (two injections of 0.5 microg/kg; s.c.; 8 h apart) on 2 separate days equally spaced in the interval between the first two endogenously driven follicular waves of an estrous cycle. Injection of oFSH induced two follicular waves in the interval between the first two endogenously driven waves of the cycle. The second endogenously driven wave of the estrous cycle emerged in the midgrowth phase of a follicular wave induced by injection of oFSH and its day of emergence, and growth pattern did not differ from that of the equivalent wave in control ewes (emerging 5.4 +/- 0.2 and 4.8 +/- 0.5 days after ovulation, respectively; P > 0.05). Experiment 2 was designed to determine if emergence of follicular waves could be induced on a daily basis. Six anestrus ewes were given oFSH (two injections of 0.35 microg/kg; s.c.; 8 h apart) on each of 4 days, starting 24 h after the expected time of an endogenously driven FSH peak. Each injection of oFSH resulted in a discrete peak in serum FSH concentrations and the emergence of a new follicular wave. The present findings provide evidence for the lack of follicular dominance in the ewe.


Follicle Stimulating Hormone/blood , Ovarian Follicle/drug effects , Sheep/blood , Animals , Estradiol/blood , Female , Follicle Stimulating Hormone/administration & dosage
6.
Genes Dev ; 22(14): 1871-6, 2008 Jul 15.
Article En | MEDLINE | ID: mdl-18628394

Female fertility requires normal ovarian follicular growth and ovulation. The nuclear receptor liver receptor homolog 1 has been implicated in processes as diverse as bile acid metabolism, steroidogenesis, and cell proliferation. In the ovary, Lrh1 is expressed exclusively in granulosa and luteal cells. Using somatic targeted mutagenesis, we show that mice lacking Lrh1 in granulosa cells are sterile, due to anovulation. The preovulatory stimulus fails to elicit cumulus expansion, luteinization, and follicular rupture in these mice. Multiple defects, including severely reduced transactivation of the Lrh1 target gene, nitric oxide synthase 3, leads to increased intrafollicular estradiol levels in the absence of Lrh1. This further causes dysfunction of prostaglandin and hyaluronic acid cascades and interrupts cumulus expansion. Lack of Lrh1 also interferes with progesterone synthesis because of failure of normal expression of the Lrh1 targets, steroidogenic acute regulatory protein and cytochrome P450 side-chain cleavage. In addition, expression of extracellular matrix proteases essential for ovulation is compromised. These results demonstrate that Lrh1 is a regulator of multiple mechanisms essential for maturation of ovarian follicles and for ovulation. Lrh1 is therefore a key modulator of female fertility and a potential target for contraception.


Ovarian Follicle/physiology , Ovulation/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Chromatin Immunoprecipitation , Cytochrome P-450 Enzyme System/metabolism , Estradiol/metabolism , Female , Fertility/physiology , Granulosa Cells/metabolism , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Ovarian Follicle/cytology , Phenotype , Phosphoproteins/metabolism , Progesterone/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radioimmunoassay
7.
Biol Reprod ; 77(2): 252-62, 2007 Aug.
Article En | MEDLINE | ID: mdl-17429016

In a previous study, 10-day estradiol implant treatment truncated the FSH peaks that precede follicular waves in sheep, but subsequent ovine FSH (oFSH) injection reinitiated wave emergence. The present study's objectives were to examine the effects of a 20-day estradiol and progesterone treatment on FSH peaks, follicle waves, and responsiveness to oFSH injection. Also, different estradiol doses were given to see whether a model that differentially suppressed FSH peaks, LH pulses, or basal gonadotropin secretion could be produced in order to study effects of these changes on follicular dynamics. Mean estradiol concentrations were 11.8 +/- 0.4 pg/ml, FSH peaks were truncated, wave emergence was halted, and the number of small follicles (2-3 mm in diameter) was reduced (P < 0.05) in cyclic ewes given estradiol and progesterone implants (experiment 1). On Day 15 of treatment, oFSH injection failed to induce wave emergence. With three different estradiol implant sizes (experiment 2), estradiol concentrations were 5.2, 19.0, 27.5, and 34.8 (+/-4.6) pg/ml in control and treated ewes, respectively. All estradiol treatments truncated FSH peaks, except those that created the highest estradiol concentrations. Experiment 2-treated ewes had significantly reduced mean and basal FSH concentrations and LH pulse amplitude and frequency. We concluded that 20-day estradiol treatment truncated FSH peaks, blocking wave emergence, and reduced the small-follicle pool, rendering the ovary unresponsive to oFSH injection in terms of wave emergence. Varying the steroid treatment created differential FSH peak regulation compared with other gonadotropin secretory parameters. This provides a useful model for future studies of the endocrine regulation of ovine antral follicular dynamics.


Corpus Luteum/growth & development , Estradiol/administration & dosage , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Ovarian Follicle/growth & development , Sheep/physiology , Animals , Estradiol/blood , Female , Ovulation , Progesterone/blood
8.
Mol Endocrinol ; 21(5): 1014-27, 2007 May.
Article En | MEDLINE | ID: mdl-17341595

Oxysterol nuclear receptors liver X receptor (LXR)alpha and LXRbeta are known to regulate lipid homeostasis in cells exposed to high amounts of cholesterol and/or fatty acids. In order to elucidate the specific and redundant roles of the LXRs in the testis, we explored the reproductive phenotypes of mice deficient of LXRalpha, LXRbeta, and both, of which only the lxralpha;beta-/- mice are infertile by 5 months of age. We demonstrate that LXRalpha-deficient mice had lower levels of testicular testosterone that correlated with a higher apoptotic rate of the germ cells. LXRbeta-deficient mice showed increased lipid accumulation in the Sertoli cells and a lower proliferation rate of the germ cells. In lxralpha;beta-/- mice, fatty acid metabolism was affected through a decrease of srebp1c and increase in scd1 mRNA expression. The retinoid acid signaling pathway was also altered in lxralpha;beta-/- mice, with a higher accumulation of all-trans retinoid receptor alpha, all-trans retinoid receptor beta, and retinoic aldehyde dehydrogenase-2 mRNA. Combination of these alterations might explain the deleterious phenotype of infertility observed only in lxralpha;beta-/- mice, even though lipid homeostasis seemed to be first altered. Wild-type mice treated with a specific LXR agonist showed an increase of testosterone production involving both LXR isoforms. Altogether, these data identify new roles of each LXR, collaborating to maintain both integrity and functions of the testis.


DNA-Binding Proteins/physiology , Fertility/physiology , Infertility, Male/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Animals , DNA Primers , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Germ Cells , Lipids/physiology , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Orphan Nuclear Receptors , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Testis/cytology , Testis/physiology
9.
Genes Dev ; 21(3): 303-15, 2007 Feb 01.
Article En | MEDLINE | ID: mdl-17289919

The small heterodimer partner (SHP) is an atypical nuclear receptor known mainly for its role in bile acid homeostasis in the enterohepatic tract. We explore here the role of SHP in the testis. SHP is expressed in the interstitial compartment of the adult testes, which contain the Leydig cells. SHP there inhibits the expression of steroidogenic genes, on the one hand by inhibiting the expression of the nuclear receptors steroidogenic factor-1 and liver receptor homolog-1 (lrh-1), and on the other hand by directly repressing the transcriptional activity of LRH-1. Consequently, in SHP knockout mice, testicular testosterone synthesis is increased independently of the hypothalamus-pituitary axis. Independent of its action on androgen synthesis, SHP also determines the timing of germ cell differentiation by controlling testicular retinoic acid metabolism. Through the inhibition of the transcriptional activity of retinoic acid receptors, SHP controls the expression of stimulated by retinoic acid gene 8 (stra8) - a gene that is indispensable for germ cell meiosis and differentiation. Together, our data demonstrate new roles for SHP in testicular androgen and retinoic acid metabolism, making SHP a testicular gatekeeper of the timing of male sexual maturation.


Receptors, Cytoplasmic and Nuclear/physiology , Sexual Maturation , Testis/physiology , Animals , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/metabolism , Germ Cells/cytology , Gonadal Steroid Hormones/biosynthesis , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Steroidogenic Factor 1 , Testis/metabolism , Testosterone/blood , Transcription Factors/metabolism , Tretinoin/physiology
10.
Biol Reprod ; 75(4): 633-41, 2006 Oct.
Article En | MEDLINE | ID: mdl-16837645

Follicle waves are preceded by follicle-stimulating hormone (FSH) peaks in ewes. The purpose of the present study was to see whether estradiol implant treatment would block FSH peaks to create a model in which the effect of the timing and mode of FSH peaks could be studied by ovine FSH (oFSH) injection. In Experiment 1, 10 ewes received estradiol-17beta implants on Day 4 after ovulation (Day 0, day of ovulation); five ewes received large implants, and five ewes received small implants. Five control ewes received empty implants. In Experiment 2, 12 ewes received large implants on Day 4. On Day 9, six ewes received oFSH twice, 8 h apart (0.5 microg/kg; s.c.). Implants were left in place for 10 days in both experiments. In both studies, ovarian ultrasonography and blood sampling was done daily. In Experiment 1, estradiol concentrations were significantly higher in ewes with large implants (10.4 +/- 0.7 pg/ml) compared with controls (3.9 +/- 0.7 pg/ml) and ewes with small implants (5.4 +/- 0.7 pg/ml; P < 0.001). A significant reduction was found in mean FSH peak concentration (31%; P < 0.05) and FSH peak amplitude (45%; P < 0.05) in ewes with large implants compared with controls. Mean and basal FSH concentrations were unaffected by the large implants. The large implants halted follicle-wave emergence between Day 0 and 8 after implant insertion. The small follicle pool (2-3 mm in diameter) was unaffected by the large implants. When oFSH was injected into ewes with large implants, a follicle wave emerged 1.5 +/- 0.5 days after injection; however, in ewes given saline alone, a follicle wave emerged 4.8 +/- 0.8 days after injection (P < 0.01). We concluded that truncation of FSH peaks by estradiol implants prevented follicle-wave emergence, but injection of physiologic concentrations of oFSH reinitiated follicle-wave emergence.


Estradiol/pharmacology , Follicle Stimulating Hormone/administration & dosage , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Animals , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Estradiol/blood , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/pharmacology , Luteinizing Hormone/blood , Ovulation , Sheep, Domestic
11.
Theriogenology ; 66(4): 811-21, 2006 Sep 01.
Article En | MEDLINE | ID: mdl-16530258

In the ewe, a rise in circulating concentrations of FSH preceding follicular wave emergence begins in the presence of growing follicles from a previous wave. We hypothesized that prostaglandin F(2alpha) (PGF(2alpha)) given at the time of an endogenous FSH peak in cyclic ewes would result in synchronous ovulation of follicles from two consecutive waves, increasing ovulation rate. Twelve Western White Face (WWF) ewes received a single i.m. injection of PGF(2alpha) (15 mg/ewe) at the expected time of a peak in FSH secretion, from Days 9 to 12 after ovulation. The mean ovulation rate after PGF(2alpha) treatment (2.3+/-0.3) did not differ (P>0.05) from the pre-treatment ovulation rate (1.7+/-0.1). Five ewes ovulated follicles from follicular waves emerging before and after PGF(2alpha) injection (3.0+/-0.6 ovulations/ewe) and seven ewes ovulated follicles only from a wave(s) emerging before PGF(2alpha) treatment (2.0+/-0.3 ovulations/ewe; P>0.05). The mean interval from PGF(2alpha) to emergence of the next follicular wave (1.0+/-0.4 and 4.0+/-0.0 d, respectively; P<0.001) and the interval from PGF(2alpha) treatment to the next FSH peak (0 and 3.5+/-0.4d, respectively; P<0.05) differed between the two groups. Six ewes ovulated after the onset of behavioral estrus, with a mean ovulation rate of 1.7+/-0.2, and six ewes ovulated both before and after the onset of estrus (3.0+/-0.5 ovulations/ewe; P<0.05). None of the ovulations that occurred before estrus resulted in corpora lutea (CL) with a full life span. At 24h before ovulation, follicles ovulating before or after the onset of estrus differed in size (4.1+/-0.3 or 5.5+/-0.4mm, respectively; P<0.05) and had distinctive echotextural characteristics. In conclusion, the administration of PGF(2alpha) at the expected time of an FSH peak at mid-cycle in ewes may alter the endogenous rhythm of FSH secretion and was not consistently followed by ovulation of follicles from two follicular waves. In non-prolific WWF ewes, PGF(2alpha)-induced luteolysis disrupted the normal distribution of the source of ovulatory follicles and may be associated with untimely follicular rupture and luteal inadequacy.


Dinoprost/administration & dosage , Estrous Cycle/drug effects , Follicle Stimulating Hormone/blood , Ovary/drug effects , Sheep , Animals , Cell Size/drug effects , Dinoprost/pharmacology , Drug Administration Schedule , Estradiol/blood , Estrous Cycle/blood , Estrous Cycle/metabolism , Female , Luteolysis/blood , Luteolysis/drug effects , Luteolytic Agents/administration & dosage , Luteolytic Agents/pharmacology , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovary/metabolism , Sheep/blood , Sheep/physiology , Time Factors
12.
Biol Reprod ; 68(4): 1403-12, 2003 Apr.
Article En | MEDLINE | ID: mdl-12606440

Medroxyprogesterone acetate (MAP) from intravaginal sponges prolongs the lifespan of large ovarian follicles when administered after prostaglandin F2alpha (PGF2alpha)-induced luteolysis early in the luteal phase of ewes. The present study was designed to determine whether a PGF2alpha/MAP treatment applied at midcycle would alter the pattern of antral follicle growth and increase ovulation rate in nonprolific ewes. A single injection of PGF2alpha (15 mg, i.m.) was given, and an intravaginal MAP (60 mg) sponge was inserted for 6 days, on approximately Day 8 after ovulation, in 7 (experiment 1), 8 (experiment 2) or 11 (experiment 3) ultrasonographically monitored, cycling Western white-faced ewes; seven ewes (experiment 1) served as untreated controls. Blood samples were collected each day and also every 12 min for 6 h, halfway through the period of treatment with MAP (experiment 1), or every 4 h, from 1 day before to 1 day after sponging (experiment 2). Seventeen of 26 treated ewes (experiment 1, n = 6; experiment 2, n = 5; experiment 3, n = 6) ovulated 1 to 6 days after PGF2alpha, but this did not affect the emergence of ensuing follicular waves (experiments 1 and 2). These ovulations, confirmed by laparotomy and histological examinations of the ovaries (experiment 3), were not preceded by an increase in LH/FSH secretion and did not result in corpora lutea, as evidenced by transrectal ultrasonography and RIA of serum progesterone (experiments 1 and 2). Following the removal of MAP sponges, the mean ovulation rate was 3.1 +/- 0.4 in treated ewes and 2.0 +/- 0.3 in control ewes (experiment 1; P < 0.05). In experiments 1 and 2, the ovulation rate after treatment (3.1 +/- 0.4 and 2.8 +/- 0.4) was also greater than the pretreatment rate (1.9 +/- 0.3 and 1.9 +/- 0.1, respectively). Ovulations of follicles from two consecutive waves before ovulation were seen in five treated but only in two control ewes (experiment 1), and in seven ewes in experiment 2. There were no significant differences between the MAP-treated and control ewes in mean daily serum concentrations of FSH and estradiol, and no differences in the parameters of LH/FSH secretion, based on frequent blood sampling. Treatment of nonprolific Western white-faced ewes with PGF2alpha and MAP at midcycle changed follicular dynamics and increased ovulation rate by approximately 50%. These effects of MAP, in the absence of luteal progesterone, may not be mediated by changes in gonadotropin secretion.


Contraceptive Agents, Female/pharmacology , Dinoprost/pharmacology , Luteolysis , Medroxyprogesterone Acetate/pharmacology , Ovarian Follicle/physiology , Ovulation/drug effects , Sheep/physiology , Animals , Estradiol/blood , Estrus/physiology , Female , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Osmolar Concentration , Progesterone/blood , Sheep/blood
...