Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 179(2): 195-203, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14596671

ABSTRACT

FoxO1 is a transcription factor implicated in a growing number of physiological processes, including apoptosis, cell cycle progression, and insulin signaling. Recent findings indicate that FSH and growth factors influence ovarian functions in part through regulation of FoxO1. The present study utilized immunohistochemical analysis to determine the ovarian localization and regulation of FoxO1 protein levels in neonatal rats, immature rats during gonadotropin-induced follicular development, ovulation, and luteinization, and in spontaneously developing ovarian cysts of aging rats. In postnatal rats, FoxO1 immunoreactivity was very faint in ovaries of 5- and 10-day-old females. In contrast, strong immunoreactivity was observed in granulosa cells of larger developing follicles at 25 days of age. To stimulate follicle development, immature female rats received equine chorionic gonadotropin (eCG) followed 52 h later by an ovulatory dose of human chorionic gonadotropin (hCG). Prior to gonadotropin treatment, moderate FoxO1 immunoreactivity was observed in granulosa cells of small follicles. Subsequently, treatment with eCG markedly decreased FoxO1 protein levels in granulosa cells of healthy antral and preovulatory follicles. Interestingly, FoxO1 staining was observed in cumulus and antral, but not mural granulosa cells of preovulatory follicles. Induction of ovulation and luteinization with hCG further decreased ovarian FoxO1 levels, with no staining evident in corpora lutea. At all time points, the most intensive FoxO1 staining was observed in granulosa cells of atretic follicles, with predominantly nuclear localization. Similarly, while FoxO1 levels were low in granulosa cells of preovulatory follicles in proestrous rats, FoxO1 staining was intense in granulosa cells of spontaneously developing cystic follicles in aged, acyclic females. Together, these findings indicate that FoxO1 is expressed in a regulated, cell-specific manner during ovarian follicular development, atresia and luteinization, suggesting roles in these physiological processes.


Subject(s)
DNA-Binding Proteins , Estrous Cycle/physiology , Nerve Tissue Proteins , Nuclear Proteins/analysis , Ovarian Follicle/physiology , Transcription Factors/analysis , Aging/physiology , Animals , Corpus Luteum/physiology , Female , Follicular Atresia/physiology , Forkhead Transcription Factors , Immunoblotting/methods , Immunohistochemistry/methods , Ovarian Follicle/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley
2.
Exp Biol Med (Maywood) ; 226(2): 127-32, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11446436

ABSTRACT

This study examined the effects of aging on LH surge magnitude, ovulation, and ovarian expression of tissue-type plasminogen activator (tPA), a protease implicated in follicular rupture. While mean LH levels and ovulation rates were similar in middle-aged cyclic and young groups, there was a significant correlation between peak LH levels and ovulation rates in individual rats, such that females with lower LH surges ovulated fewer ova. In a separate experiment, proestrous LH levels were characterized in young and middle-aged rats, followed by in situ hybridization analysis of ovarian tPA mRNA. In young proestrous rats, tPA expression was observed in thecal-interstitial cells and oocytes, but not granulosa cells, prior to the LH surge. After the LH surge, there was a marked increase in tPA mRNA levels in granulosa cells of preovulatory, but not smaller follicles, peaking at 0200 hr estrus. By 0500 hr estrus, ovarian tPA expression declined, and ovulation had occurred. In contrast, LH-induced follicular tPA mRNA levels were dramatically lower in middle-aged rats with attenuated LH surges, and persisting preovulatory follicles were common in ovaries of these females on estrus morning. These findings suggest that age-related declines in ovulatory function result in part from altered induction of ovarian tPA expression, likely due to decreased proestrous LH secretion.


Subject(s)
Aging/physiology , Luteinizing Hormone/metabolism , Ovary/physiology , Ovulation , Tissue Plasminogen Activator/biosynthesis , Animals , Female , Ovarian Follicle/physiology , Proestrus/physiology , RNA, Messenger/analysis , Rats , Rats, Long-Evans , Tissue Plasminogen Activator/genetics
3.
Proc Natl Acad Sci U S A ; 98(14): 7994-9, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11416163

ABSTRACT

Maintenance of female reproductive competence depends on the actions of several hormones and signaling factors. Recent reports suggest roles for bone morphogenetic proteins (BMPs) in early stages of folliculogenesis. A role for the type I BMP receptor BmprIB as a regulator of ovulation rates in sheep has been described recently, but little is known about the roles of BMP signaling pathways in other aspects of reproductive function. We report here that BMPRIB is essential for multiple aspects of female fertility. Mice deficient in BmprIB exhibit irregular estrous cycles and an impaired pseudopregnancy response. BmprIB mutants produce oocytes that can be fertilized in vitro, but defects in cumulus expansion prevent fertilization in vivo. This defect is associated with decreased levels of aromatase production in granulosa cells. Unexpectedly, levels of mRNA for cyclooxygenase 2, an enzyme required for cumulus expansion, are increased. BmprIB mutants also exhibit a failure in endometrial gland formation. The expression of BmprIB in uterine linings suggests that these defects are a direct consequence of loss of BMP signaling in this tissue. In summary, these studies demonstrate the importance of BMP signaling pathways for estrus cyclicity, estradiol biosynthesis, and cumulus cell expansion in vivo and reveal sites of action for BMP signaling pathways in reproductive tissues.


Subject(s)
Protein Serine-Threonine Kinases/physiology , Receptors, Growth Factor/physiology , Reproduction/physiology , Animals , Bone Morphogenetic Protein Receptors, Type I , Cyclooxygenase 2 , Female , Genitalia, Female/physiology , Isoenzymes/physiology , Mice , Prostaglandin-Endoperoxide Synthases/physiology , Signal Transduction/physiology
4.
Biol Reprod ; 64(4): 1056-62, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259250

ABSTRACT

This study examined the influences of aging and reduced ovarian follicular reserve on estrous cyclicity, estradiol (E(2)) production, and gonadotropin secretion. Young virgin and middle-aged (MA) retired breeder female rats were unilaterally ovariectomized (ULO) or sham operated (control). Unilateral ovariectomy of young rats reduced the ovarian follicular reserve by one-half, to a level similar to that found in MA controls. Unilateral ovariectomy of MA females reduced the follicular pool further, to one half of MA controls. The incidence of regular cyclicity was significantly lower in MA ULO females than in young controls, with intermediate cycle frequency in young ULO and MA controls. Among cyclic rats, the magnitude of the proestrous LH surge was highest in young controls, intermediate in young ULO rats and MA controls, and lowest in MA ULO females. Similarly, ovulation rates were highest in young controls, intermediate in young ULO rats and MA controls, and lowest in MA ULO females. While young ULO rats exhibited augmented secondary FSH surges on estrous morning, middle-aged ULO females displayed secondary FSH levels comparable to young controls. The effects of age and reduced follicle number on estrous cyclicity and gonadotropin secretion were not due to altered E(2) secretion, as preovulatory E(2) levels were similar among all groups. Thus, experimental reduction in the follicular reserve exerts acute effects on the preovulatory LH surge, ovulation rate, and estrous cyclicity in both young and MA rats. However, decreased follicle number increases FSH levels only in young rats, indicating aging-related alterations in the feedback regulation of FSH.


Subject(s)
Aging , Estrus , Gonadotropins, Pituitary/metabolism , Ovarian Follicle/physiology , Ovary/physiology , Ovulation , Animals , Estradiol/blood , Female , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Ovarian Follicle/anatomy & histology , Ovariectomy , Proestrus , Rats , Rats, Long-Evans
5.
J Endocrinol ; 168(2): 249-55, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11182762

ABSTRACT

Previous studies demonstrated inhibitory effects of nitric oxide (NO) and cGMP on ovarian steroidogenesis. This study examined the effects of NO on estrogen levels and cAMP accumulation from immature cultured rat granulosa cells. Granulosa cells were incubated with media alone (control), FSH or FSH plus increasing concentrations of the NO generator, (Z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA/NO). While FSH increased estrogen levels 15-fold compared with controls, DETA/NO inhibited FSH-stimulated aromatase activity in a dose-dependent manner. Time-course studies revealed that the inhibitory effects of DETA/NO on aromatase activity persisted throughout the 72 h culture period. Treatment with DETA/NO also inhibited the stimulatory effects of forskolin on estrogen production, indicating that NO can influence steroidogenesis by actions downstream of the FSH receptor. Incubation of cells with FSH plus DETA/NO increased cGMP accumulation over 100-fold, compared with cells treated with media or FSH alone. In this regard, a cGMP analog mimicked the inhibitory effects of NO on FSH- and forskolin-stimulated estrogen production, indicating a potential mechanism of NO action. NO also decreased FSH-stimulated (cAMP) accumulation from cultured cells, indicating an antagonistic effect of NO on the second messenger mediating FSH actions. These findings demonstrate that NO inhibits estrogen production from rat granulosa cells, potentially reflecting actions on the second messengers cGMP and cAMP.


Subject(s)
Cyclic AMP/metabolism , Estrogens/biosynthesis , Granulosa Cells/drug effects , Nitric Oxide/pharmacology , Vasodilator Agents/pharmacology , Animals , Aromatase/metabolism , Cell Culture Techniques , Colforsin/antagonists & inhibitors , Colforsin/pharmacology , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Female , Follicle Stimulating Hormone/antagonists & inhibitors , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Rats , Rats, Sprague-Dawley , Second Messenger Systems/physiology
6.
Proc Soc Exp Biol Med ; 224(4): 285-91, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964264

ABSTRACT

Reproductive aging in the female rat is associated with gradual declines in LH secretion and ovarian progesterone (P) production. This study examined whether the influences of aging on P levels reflect decreased ovarian responsiveness to gonadotropin stimulation, as opposed to changes in gonadotropin release. Young and middle-aged regularly cyclic female rats received sodium pentobarbital to block endogenous proestrous luteinizing hormone (LH) surges, followed by administration of various doses of human chorionic gonadotropin (hCG). Similar treatments were performed in middle-aged acyclic persistent-estrous (PE) females. Injection of hCG resulted in equivalent plasma hCG levels in each treatment group. At the lowest hCG dose tested, a significant rise in plasma P levels was observed in middle-aged cyclic rats, but not in young cyclic or middle-aged PE females. This unexpected finding may reflect accelerated follicular development in middle-aged cyclic females, as suggested by a previous study. At the intermediate dose, young and middle-aged cyclic but not PE rats displayed significantly increased P in response to hCG. At the highest dose tested, all three groups of rats displayed increased P levels after hCG stimulation. However, P concentrations were significantly lower in middle-aged PE than regularly cyclic females. Northern and slot blot hybridization analyses revealed that ovarian mRNA levels for cytochrome P450 side-chain cleavage, the rate-limiting enzyme in P synthesis, were markedly reduced in PE rats following hCG stimulation. These findings indicate that ovarian responsiveness to gonadotropin stimulation is impaired in middle-aged PE, but not regularly cyclic rats, and suggest influences of cycle status on the biochemical and molecular mechanisms regulating ovarian steroid production. Furthermore, these findings reveal that attenuated P production in middle-aged proestrous rats is due to attenuated preovulatory LH surges, rather than decreased ovarian sensitivity to LH.


Subject(s)
Aging/physiology , Chorionic Gonadotropin/pharmacology , Ovary/drug effects , Progesterone/blood , Animals , Body Weight/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Chorionic Gonadotropin/blood , Dose-Response Relationship, Drug , Estrus/drug effects , Female , Humans , Luteinizing Hormone/blood , Ovary/enzymology , Ovary/metabolism , Pentobarbital/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Long-Evans , Time Factors
7.
Biol Reprod ; 60(2): 365-73, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9916003

ABSTRACT

WT1 is a zinc finger protein with transcriptional repressor activity on several growth factor and growth factor receptor genes. In the ovary, a potential role for WT1 in the suppression of the development of immature follicles has been demonstrated. Here, gel retardation assays further showed that recombinant WT1 protein interacted with consensus DNA sequences in the inhibin-alpha gene promoter. We investigated the pattern of WT1 expression in a wide variety of species and also over the reproductive life span in rats. In chicken ovaries, Northern blot analysis revealed the presence of WT1 transcript in small healthy white follicles (1-5 mm in diameter) and its absence in small yellow (6-12 mm in diameter) or larger follicles (F1-F5). In pig and monkey ovaries, WT1 expression was limited to granulosa cells of preantral follicles, as shown by in situ hybridization analysis. In rats, Northern blot analyses demonstrated the presence of WT1 transcript in the ovaries of young (3-mo-old) and middle-aged (9-mo-old) rats on the proestrous day, with a decrease in old (12-mo-old) rats in persistent estrus. In situ hybridization analysis further suggested that the decrease in WT1 expression in aging ovaries was associated with fewer immature follicles. Thus, WT1 expression is restricted to immature follicles in diverse avian and mammalian species and over the reproductive life span in rats. These data demonstrated that WT1 is a marker for immature follicles and suggested a potential role of this transcriptional repressor in the slow growth of early follicles.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression , Inhibins , Ovarian Follicle/metabolism , RNA, Messenger/metabolism , Transcription Factors/genetics , Animals , Blotting, Northern , Chickens , DNA/metabolism , DNA-Binding Proteins/metabolism , Female , Granulosa Cells/chemistry , Humans , In Situ Hybridization , Macaca fascicularis , Peptides/genetics , Promoter Regions, Genetic , RNA, Messenger/analysis , Rats , Rats, Long-Evans , Species Specificity , Swine , Transcription Factors/metabolism , WT1 Proteins , Zinc Fingers
8.
Biol Reprod ; 59(1): 197-201, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9675012

ABSTRACT

The present study examined the effects of progesterone (P4) treatments on estrous cyclicity and the loss of ovarian follicles during aging. Young rats received repeated treatments with P4 or empty implants between 3.5 and 8 mo of age. At 8 mo, ovaries were obtained from some animals to determine the numbers of resting follicles, and estrous cycle patterns and hormone levels were determined from other groups of treated females. In contrast to the cyclic increases in P4, estradiol (E2), LH, and FSH in control animals, P4-implanted rats exhibited elevated serum P4 but low E2, LH, and FSH levels. After implant treatments, the follicular reserve was significantly (p < 0.05) larger in P4-treated females (2012 +/- 297 resting follicles per ovary, n = 5 rats per group) than in regularly cyclic control rats (713 +/- 226 follicles per ovary, n = 7). The effects of P4 implants on the follicular reserve were associated with a subsequently higher incidence of regular estrous cycles after P4 treatment. These results demonstrate that P4 prevents cyclic increases in E2 secretion and is associated with a conservation of the ovarian follicular reserve and the maintenance of regular estrous cycle patterns, indicating a protective effect of P4 on the age-related loss of ovarian follicles.


Subject(s)
Aging , Estrus/physiology , Ovarian Follicle/physiology , Progesterone/administration & dosage , Animals , Drug Implants , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Progesterone/blood , Rats
9.
Proc Soc Exp Biol Med ; 217(4): 455-60, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9521093

ABSTRACT

Reproductive aging in the female rat is associated with the gradual loss of regular ovulatory function, decreased fertility, and smaller litter sizes. In the present study, we assessed ovarian ovulatory responsiveness to exogenous gonadotropin stimulation in young and middle-aged cyclic females and in middle-aged acyclic persistent-estrous (PE) rats. The ovulatory response to human chorionic gonadotropin (hCG) was dose-dependent in both young and middle-aged cyclic rats, with the percentages of rats ovulating and the numbers of ova shed per rat increasing with the dose of hCG administered. At the highest dose tested (10 mIU hCG/g bw), the range in ovulation rates among middle-aged cyclic rats (0-18 ova shed/rat) was greater than that in young animals (12-18 ova/rat). However, there were no statistically significant differences in either the percentages of females ovulating or in the mean ovulation rates between young and middle-aged cyclic groups. In contrast to the normal ovulatory responses observed in most middle-aged cyclic animals, response to hCG was markedly impaired in PE females of the same age. Middle-aged PE rats consistently failed to ovulate in response to a dose of hCG (10 mIU/g bw), which elicited high ovulation rates in young rats. At an even higher dose (20 mIU/g bw), only minimal ovulatory responses were observed (1.8 +/- 0.8 ova/rat; 80% of rats ovulating). Thus, most middle-aged regularly cyclic females maintain a similar ovulatory responsiveness to hCG as young rats, suggesting that decreased ovulation rates during aging may be related to attenuated preovulatory LH surges. However, impaired ovulatory responses were observed in middle-aged PE females, indicating altered ovarian function in acyclic animals, which may contribute to their anovulatory state.


Subject(s)
Aging/physiology , Chorionic Gonadotropin/pharmacology , Estrus , Ovulation/physiology , Animals , Chorionic Gonadotropin/blood , Dose-Response Relationship, Drug , Female , Luteinizing Hormone/blood , Ovary/drug effects , Ovary/physiology , Pentobarbital/pharmacology , Proestrus , Rats
10.
Biol Reprod ; 57(5): 990-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9369162

ABSTRACT

The stimulatory effects of gonadotropins on antral and preovulatory follicles are well known, but conflicting results have been reported regarding the gonadotropin responsiveness and dependency of preantral follicles. Taking advantage of the relatively uniform development of the first wave of follicles in the postnatal rat ovary, we evaluated the role of endogenous and exogenous gonadotropins on preantral follicle development. Reduction of the high levels of gonadotropins present in juvenile rats by either hypophysectomy (at Day 15) or GnRH antagonist treatment (starting from Day 11 of age) resulted in decreased ovarian weight at Day 19 of age that was associated with a reduced number of developing follicles and increased atresia of remaining follicles. In contrast, treatment with FSHctp (a long-acting FSH agonist) in intact (Days 5-19 of age), hypophysectomized (Days 15-19), or GnRH antagonist-treated (Days 11-19) animals resulted in increased ovarian weight and follicle development as determined histologically and by inhibin-alpha expression. A dose-dependent stimulatory effect of hCG on ovarian weight was seen when animals were cotreated with FSHctp and the GnRH antagonist. At low doses of hCG, augmentation of antral follicle formation occurred, whereas higher doses of hCG led to morphological signs of luteinization. These findings demonstrate the important role of endogenous gonadotropins in preantral follicle development and indicate that preantral follicles are highly responsive to exogenous gonadotropins.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Ovarian Follicle/growth & development , Animals , Animals, Newborn , Blotting, Western , Chorionic Gonadotropin/metabolism , Female , Hypophysectomy , Inhibins/immunology , Inhibins/metabolism , Luteinizing Hormone/metabolism , Organ Size/drug effects , Ovarian Follicle/drug effects , Ovary/drug effects , Ovary/growth & development , Rats , Rats, Sprague-Dawley , Receptors, LHRH/antagonists & inhibitors
11.
J Clin Endocrinol Metab ; 80(12): 3412-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8530574

ABSTRACT

Chronic GnRH agonist therapy lowers androgens and decreases androgen-dependent hair shaft diameter, but the resulting induction of hypoestrogenemia has limited its usefulness as a single agent. Estrogen- and progestin-containing oral contraceptives also reduce circulating androgen levels and are commonly used empirically for the treatment of hirsutism, but have not been evaluated in a blinded randomized controlled fashion. The present study is the first double masked trial to evaluate the combination use of a GnRH agonist and an estrogen-containing oral contraceptive and tests our hypothesis that these could synergistically reduce androgen levels and suppress hormone-dependent hair growth while avoiding the symptoms and risks of agonist-induced hypoestrogenemia. We enrolled 64 women in a 24-week blinded randomized controlled trial to compare placebo, nafarelin (NAF; 400 micrograms, intranasal spray, twice daily), norethindrone (1 mg), and ethinyl estradiol (NOR 1/35; 0.035 mg, daily, for 3 of 4 weeks), or combined use of NAF and NOR 1/35 for 24 weeks. At baseline and every 8 weeks, we measured gonadotropins, estrogens, androgens, and hair growth. Bone density was assessed by dual energy x-ray adsorptiometry, and hot flashes were measured objectively. Baseline total testosterone (T), free T, percent free T, and sex hormone-binding globulin-binding capacity were similar among groups. With treatment, significant reductions (P = 0.01) in total T were seen with combination and NAF only therapy. Significant increases (P < 0.001) in the sex hormone-binding globulin-binding capacity were seen in women given NOR 1/35 alone or in combination with NAF. Free T levels decreased to approximately half of baseline levels with combination treatment (17.9 to 6.4 nmol/L; P < 0.001) and NOR 1/35 alone (20.8 to 10.2 nmol/L; P < 0.001). There was a significant decrease in hair shaft diameter after combination therapy (P < 0.05) that was not seen with either agent alone. Combination therapy also prevented the hot flashes and bone loss that occurred with agonist alone. In summary, our results demonstrate that combination GnRH agonist and low dose oral contraceptive therapy is more effective than either agent alone in the treatment of hirsutism and avoids the hypoestrogenic complications that occur with agonist only therapy.


Subject(s)
Contraceptives, Oral/administration & dosage , Ethinyl Estradiol/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Hirsutism/drug therapy , Nafarelin/therapeutic use , Norethindrone/therapeutic use , Adult , Contraceptives, Oral/therapeutic use , Dose-Response Relationship, Drug , Drug Combinations , Female , Hair/drug effects , Hair/pathology , Hirsutism/pathology , Humans , Middle Aged , Treatment Outcome
12.
Endocrinology ; 136(12): 5533-9, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7588305

ABSTRACT

Superoxide dismutases (SOD) modulate oxygen free radical metabolism and influence second messenger signaling in a variety of cell types. We have investigated the influence and possible mechanisms of action of SOD on aromatase activity in cultured rat granulosa cells. Although treatment of granulosa cells with FSH (0.3-30 ng/ml) resulted in a dose-dependent stimulation of estrogen levels, cotreatment of cells with SOD (10(-6) M) significantly attenuated estrogen production at the highest doses of FSH. The effects of SOD were dose dependent between 10(-7)-10(-5) M, with increasing amounts of SOD causing decreasing concentrations of estrogen. Cotreatment of cells with catalase (1500 U/ml) failed to prevent the inhibitory influence of SOD on estrogen production, indicating that the effects of SOD were not due to accumulation of hydrogen peroxide. Although incubation with either forskolin or (Bu)2cAMP alone stimulated estrogen production from granulosa cells, cotreatment with SOD significantly attenuated estrogen levels, indicating that SOD can inhibit aromatase activity at one or more post-FSH receptor sites. Treatment of cells with SOD, FSH, or forskolin resulted in small, but significant, increase in cGMP concentrations. In contrast, cotreatment of cells with FSH plus SOD as well as forskolin plus SOD had a marked synergistic effect on cGMP content, increasing cGMP levels over 100-fold. Incubation of granulosa cells with (Bu)2cGMP (2 mM) significantly decreased FSH-induced estrogen levels in a dose-dependent manner (0.25-2 mM). In addition, (Bu)2cGMP attenuated both forskolin- and (Bu)2cAMP-induced estrogen production. In contrast to the effects of (Bu)2cGMP and SOD on estradiol levels, these agents had no significant effect on progesterone production by cultured granulosa cells. These results demonstrate attenuated induction of aromatase activity by FSH in cultured rat granulosa cells cotreated with SOD, suggesting a potential modulatory role of this antioxidant on granulosa cell functions. The findings that SOD and activators of the cAMP-dependent signaling pathway synergistically increase the levels of the second messenger cGMP and that (Bu)2cGMP treatment attenuates FSH-, forskolin-, and cAMP-induced aromatase activity suggest a potential mechanism of SOD action and demonstrate the antagonist action of cGMP on cAMP-mediated estrogen production.


Subject(s)
Cyclic GMP/physiology , Estrogens/biosynthesis , Granulosa Cells/metabolism , Superoxide Dismutase/pharmacology , Animals , Bucladesine/pharmacology , Catalase/pharmacology , Cells, Cultured , Colforsin/pharmacology , Cyclic GMP/analysis , Female , Follicle Stimulating Hormone/pharmacology , Nitric Oxide/metabolism , Progesterone/biosynthesis , Rats , Rats, Sprague-Dawley
13.
Fertil Steril ; 63(2): 273-6, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7843430

ABSTRACT

OBJECTIVE: To evaluate follicular FSH and LH requirements during suppression of endogenous gonadotropins with the GnRH antagonist Nal-Glu and whether LH-like activity could be supplied by administering subcutaneous hCG. DESIGN: Randomized clinical trial. PARTICIPANTS: Thirty-two normally cycling females in the late follicular phase (dominant follicle mean diameter > or = 14 mm). INTERVENTION: Twelve normal women were randomized to receive 150 IU IM FSH with or without 75 IU SC hCG; 11 subjects were randomized to receive 225 IU FSH with or without 50 IU SC hCG; 9 women received 150 or 225 IU IM hMG. Subjects returned the next day for repeat blood sample and ultrasound. RESULTS: Continued follicular maturation, as evidenced by rising E2 levels, correlated with serum immunoactive and bioactive FSH levels and was unrelated to bioactive LH-hCG. Two hundred twenty-five international units of exogenous FSH consistently supported follicular maturation. There was a similar increase in mean follicular diameter in women with an E2 rise versus those with a plateau or fall. In subjects receiving SC mini-dose hCG, serum bioactive LH-hCG levels were increased significantly and were similar to levels before Nal-Glu. CONCLUSIONS: During administration of a GnRH-a, the maturing follicle appears to require only FSH support. In markedly hypogonadotropic women, mini-dose hCG may be a more practical alternative to recombinant LH to promote normal follicle maturation.


Subject(s)
Follicle Stimulating Hormone/administration & dosage , Luteinizing Hormone/administration & dosage , Adult , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/blood , Chorionic Gonadotropin/therapeutic use , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Luteinizing Hormone/blood , Luteinizing Hormone/therapeutic use , Menotropins/therapeutic use
15.
Endocrinology ; 133(6): 2875-80, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8243314

ABSTRACT

Ovulation and subsequent luteal tissue formation are preceded by midcycle surges of both LH and FSH. Although LH has been widely known as the luteinizing hormone, a potential role for FSH in the luteinization process is possible. Our earlier studies using recombinant FSH (rcFSH) without LH contamination have shown that treatment with a surge dose of rcFSH induces ovulation of mature follicles in hypophysectomized rats. The present studies examined further whether FSH alone is sufficient to induce normal corpus luteum formation. Immature hypophysectomized rats were implanted with an estrogen pellet (10 mg diethylstilbestrol). Two days later, a minipump releasing 4 IU rcFSH/day was placed to induce follicular growth. Forty-eight hours after FSH treatment, both DES pellet and FSH minipump were removed, and rats were injected with a single sc dose of 40 IU rcFSH, 5 micrograms hCG, or saline. For some animals, oviducts were excised the following day to determine the number of ovulated oocytes. The remaining animals received, 2 days later, sc injections of 125 micrograms ovine PRL twice daily for 3 days to maintain luteal function. All rats that received a surge dose of rcFSH or hCG ovulated similar numbers of oocytes, whereas none of the control animals did. Ovaries and blood samples were obtained 5 days after the gonadotropin surge. rcFSH and hCG significantly increased ovarian weight to 73.9 +/- 4.8 and 94.7 +/- 5.6 mg, respectively, compared to 10.0 +/- 0.5 mg in controls. Serum progesterone levels were increased by 192- and 102-fold in rcFSH- and hCG-treated animals, respectively, compared with those in the saline-treated rats. rcFSH and hCG also induced a marked elevation of ovarian [125I]hCG binding (4.2 +/- 0.2 and 3.7 +/- 0.1 ng/mg ovary, respectively), whereas ovaries from control animals exhibited low binding (0.6 +/- 0.1 ng/mg ovary). These gonadotropin-induced increases in [125I]hCG binding were associated with similar elevations in the levels of three LH receptor transcripts of 2.5, 4.2, and 7.0 kilobases. Also, levels of the ovarian cholesterol side-chain cleavage enzyme (CYP 11A) mRNA (2 kilobases) were low in control animals, but increased 20.5- and 14.3-fold after surge doses of rcFSH and hCG, respectively. Accompanied by biochemical signs of luteinization, morphological features typical of luteinized ovaries were found in both rcFSH and hCG groups, showing the formation of large polyhedral lutein cells and small spindle-shaped lutein cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Corpus Luteum/growth & development , Follicle Stimulating Hormone/pharmacology , Ovarian Follicle/drug effects , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Chorionic Gonadotropin/metabolism , Chorionic Gonadotropin/pharmacology , Corpus Luteum/metabolism , Enzyme Induction/drug effects , Female , Hypophysectomy , Organ Size/drug effects , Ovarian Follicle/physiology , Ovary/drug effects , Progesterone/blood , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, LH/genetics , Recombinant Proteins
16.
Endocrinology ; 131(6): 2514-20, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1446593

ABSTRACT

The induction of granulosa cell differentiation and follicle maturation is dependent upon the stimulatory actions of FSH. Our recent studies used recombinant DNA technology to fuse the carboxyl-terminal peptide (CTP) of hCG beta-subunit to the carboxyl-terminus of the FSH beta-subunit. The resulting FSH analog has identical in vitro receptor-binding and biological activities as wild-type FSH (WT-FSH), but an increased circulating half-life. The present studies examined further the ability of FSH with one (FSH-CTP1) or two (FSH-CTP2) appended CTPs to promote granulosa cell differentiation and follicle ovulatory potential. WT-FSH, FSH-CTP1, and FSH-CTP2 were produced from Chinese hamster ovary cells transfected with the common alpha-subunit and respective beta-subunit. Hormone concentrations were quantitated by RIA, and relative levels confirmed by radioligand receptor assay. Both FSH-CTP1 and FSH-CTP2 retained full FSH receptor-binding activity, but did not bind LH receptors. To compare in vivo bioactivity, immature estrogen-primed female rats received ip injections of FSH or the agonists at 0 and 24 h. At 48 h, substantial stimulation (up to 2.5-fold) of ovarian weight was induced by 1.0 and 3.0 IU/day FSH-CTP1 or FSH-CTP2, whereas a higher dose (10 IU/day) of WT-FSH was required for an 1.8-fold stimulation. Although the in vivo potencies of FSH-CTP1 and FSH-CTP2 were similar, FSH-CTPs were about 10-fold more potent than WT-FSH in inducing granulosa cell aromatase activity and LH receptors. We further reduced the frequency of hormone administration. Increasing doses (1-10 IU) of a single ip injection of FSH-CTP1 resulted in dose-dependent increases in granulosa cell aromatase activity and LH receptor content 48 h later. Although a single injection (10 IU) of WT-FSH had no effect, the same total dose of WT-FSH administered as four 2.5-IU injections 12 h apart was effective. To test the ovulatory potential of ovarian follicles, rats received a single injection of FSH-CTP1, followed 52 h later by 5 IU hCG to induce ovulation. Although hCG did not induce ovulation in females receiving a single dose (10 IU) of WT-FSH, 20 +/- 2 and 43 +/- 5 ovulated ova/rat were found in animals primed with 3 and 10 IU FSH-CTP1, respectively. Because twice daily injections of WT-FSH (2.5 IU/injection) also increased the ovulatory potential of the ovary, the enhanced effectiveness of FSH-CTP1 appears to be related to its increased circulating half-life.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Follicle Stimulating Hormone/analogs & derivatives , Follicle Stimulating Hormone/pharmacology , Ovarian Follicle/physiology , Ovulation/drug effects , Animals , Aromatase/metabolism , Cell Differentiation/drug effects , Female , Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone, beta Subunit , Granulosa Cells/cytology , Granulosa Cells/enzymology , Organ Size/drug effects , Ovarian Follicle/drug effects , Ovary/anatomy & histology , Ovary/drug effects , Ovary/physiology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, FSH/metabolism , Receptors, LH/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
17.
Trends Endocrinol Metab ; 3(5): 164-70, 1992 Jul.
Article in English | MEDLINE | ID: mdl-18407096

ABSTRACT

The anterior pituitary hormones, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), act upon the ovary and testis via occupancy of specific cell membrane receptors, resulting in increased cAMP production, steroidogenesis, and expression of differentiation-related genes. Recent cloning of the cDNAs for LH and FSH receptors allows the analysis of mRNA levels for these receptors in gonadal tissues. This review summarizes progress in elucidating the molecular basis of LH and FSH receptor gene regulation in the ovary and testis during different physiologic states.

18.
Biol Reprod ; 46(6): 1016-20, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1391300

ABSTRACT

On the basis of earlier observations of changing testicular LH receptor levels during postnatal development of the rat, we analyzed the levels of LH receptor mRNA transcripts in testes of rats at ages 5-70 days. Extracted testis RNA, prepared for Northern blotting, was hybridized with a specific LH receptor cRNA probe derived from subcloned cDNA corresponding to the extracellular domain of the receptor. Six LH receptor mRNA transcripts with molecular sizes of 7.8, 7.0, 4.2, 2.5, 1.8, and 1.2 kb were identified. Of these, the 1.2- and the 1.8-kb mRNA transcripts presumably code for truncated forms of LH receptor. At 5 days, only the 1.8- and the 4.2-kb mRNA transcripts were observed. Additional 7.0- and 1.2-kb transcripts appeared at 10 and 15 days, respectively. From the age of 25 days through adulthood, all six mRNA transcripts were observed. Densitometric analyses revealed that the amounts of the 7.0- and 1.8-kb mRNA transcripts correlated well with LH receptor levels, while the 4.2-kb transcript showed high levels earlier in life with poor correlation to LH receptor number. Because the 1.8 kb receptor transcript lacked transmembrane domains, the present results suggest the 7.0-kb LH receptor transcript as the likely candidate to encode the functional receptor. These data provide the basis for future analyses of the molecular regulation of LH receptor expression.


Subject(s)
Gene Expression Regulation , RNA, Messenger/analysis , Receptors, LH/genetics , Testis/growth & development , Transcription, Genetic , Animals , Male , Organ Size , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Receptors, LH/metabolism , Testis/metabolism
19.
Proc Natl Acad Sci U S A ; 89(10): 4304-8, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1374895

ABSTRACT

Follitropin (FSH) is a pituitary glycoprotein hormone that is essential for the development of ovarian follicles and testicular seminiferous tubules. FSH is used clinically to stimulate follicular maturation for in vitro fertilization and treatment of anovulatory women. One issue regarding the clinical use of FSH is its short half-life in the circulation. To address this point, we constructed chimeric genes containing the sequence encoding the C-terminal peptide of the chorionic gonadotropin beta subunit (CG beta) fused to the translated sequence of the human FSH beta subunit (FSH beta). This region of CG beta is important for maintaining the prolonged plasma half-life of human CG dimer. The presence of the C-terminal peptide sequence did not significantly affect assembly of FSH beta with the alpha subunit or secretion of the dimer. In vitro receptor binding and steroidogenic activity of dimer bearing the FSH beta-C-terminal peptide chimera were the same as wild-type FSH. However, both the in vivo potency and half-life in circulation of the dimer bearing either one or two C-terminal peptide units were enhanced. Dimers containing FSH beta-CG beta chimeras could serve as potent FSH agonists for clinical use, and the present strategy may have wide applications for enhancing the in vivo half-life of diverse proteins.


Subject(s)
Chorionic Gonadotropin/genetics , Chorionic Gonadotropin/metabolism , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Animals , Aromatase/metabolism , Biological Assay , CHO Cells , Cells, Cultured , Chimera , Chorionic Gonadotropin/pharmacology , Chorionic Gonadotropin, beta Subunit, Human , Cricetinae , Drug Design , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Half-Life , Male , Metabolic Clearance Rate , Organ Size/drug effects , Peptide Fragments/pharmacology , Rats , Receptors, FSH/metabolism , Recombinant Fusion Proteins/pharmacology , Restriction Mapping , Testis/metabolism , Transfection
20.
Fertil Steril ; 57(3): 495-500, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1531463

ABSTRACT

OBJECTIVE: To examine the possible impact of abnormal adrenal steroidogenesis on the ovarian dysfunction seen in polycystic ovarian disease (PCOD). DESIGN: Prospective analysis of blood sampling monthly for 6 months, then three times weekly for 90 days. SETTING: Tertiary institutional outpatient care. PARTICIPANTS: Six anovulatory women with a diagnosis of PCOD. INTERVENTION: Six-month suppression with gonadotropin-releasing hormone agonist (GnRH-a) followed by suppression with dexamethasone (DEX) for 90 days. MAIN OUTCOME MEASURES: Serum levels of testosterone (T), androstenedione (A), dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), cortisol, estradiol (E2), progesterone (P), follicle-stimulating hormone (FSH), luteinizing hormone (LH) and bioactive LH. RESULTS: Gonadotropin-releasing hormone agonist administration suppressed greater than 60% of the circulating levels of T and A, suggesting an ovarian origin. Minimal changes of DHEA, DHEAS, and cortisol were seen. With the addition of DEX, there was greater than 90% suppression of the total circulating A, T, DHEA, DHEAS, and cortisol, supporting the adrenal origin of the non-GnRH-a suppressible androgens. Excessive ovarian T and A secretion returned during the 90-day recovery study period in spite of rises of FSH concentrations that changed the ratio of FSH to LH in all subjects. Four of the six women failed to ovulate. In comparison of the women who did and did not ovulate during recovery, no differences in absolute levels or changes in concentrations of steroids or gonadotropins could be detected. CONCLUSIONS: Using sequential and simultaneous administration of GnRH-a and DEX, we were able to delineate the contributions of the ovaries and adrenals to the abnormal steroidogenesis seen in PCOD. Despite prolonged suppression of ovarian and then adrenal steroidogenesis, ovarian dysfunction, evidenced by abnormal androgen production, returned with cessation of agonist administration.


Subject(s)
Adrenal Glands/metabolism , Dexamethasone/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Polycystic Ovary Syndrome/drug therapy , Triptorelin Pamoate/analogs & derivatives , Adrenal Glands/drug effects , Adult , Androstenedione/blood , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/blood , Dehydroepiandrosterone Sulfate , Delayed-Action Preparations , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Hydrocortisone/blood , Luteinizing Hormone/blood , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Progesterone/blood , Prospective Studies , Testosterone/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...