Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Hum Reprod ; 33(3): 531-538, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29425284

ABSTRACT

STUDY QUESTION: What is the prevalence of somatic chromosomal instability among women with idiopathic primary ovarian insufficiency (POI)? SUMMARY ANSWER: A subset of women with idiopathic POI may have functional impairment in DNA repair leading to chromosomal instability in their soma. WHAT IS KNOWN ALREADY: The formation and repair of DNA double-strand breaks during meiotic recombination are fundamental processes of gametogenesis. Oocytes with compromised DNA integrity are susceptible to apoptosis which could trigger premature ovarian aging and accelerated wastage of the human follicle reserve. Genomewide association studies, as well as whole exome sequencing, have implicated multiple genes involved in DNA damage repair. However, the prevalence of defective DNA damage repair in the soma of women with POI is unknown. STUDY DESIGN, SIZE, DURATION: In total, 46 women with POI and 15 family members were evaluated for excessive mitomycin-C (MMC)-induced chromosome breakage. Healthy fertile females (n = 20) and two lymphoblastoid cell lines served as negative and as positive controls, respectively. PARTICIPANTS/MATERIALS, SETTING, METHODS: We performed a pilot functional study utilizing MMC to assess chromosomal instability in the peripheral blood of participants. A high-resolution array comparative genomic hybridization (aCGH) was performed on 16 POI patients to identify copy number variations (CNVs) for a set of 341 targeted genes implicated in DNA repair. MAIN RESULTS AND THE ROLE OF CHANCE: Array CGH revealed three POI patients (3/16, 18.8%) with pathogenic CNVs. Excessive chromosomal breakage suggestive of a constitutional deficiency in DNA repair was detected in one POI patient with the 16p12.3 duplication. In two patients with negative chromosome breakage analysis, aCGH detected a Xq28 deletion comprising the Centrin EF-hand Protein 2 (CETN2) and HAUS Augmin Like Complex Subunit 7 (HAUS7) genes essential for meiotic DNA repair, and a duplication in the 3p22.2 region comprising a part of the ATPase domain of the MutL Homolog 1 (MLH1) gene. LIMITATIONS REASONS FOR CAUTION: Peripheral lymphocytes, used as a surrogate tissue to quantify induced chromosome damage, may not be representative of all the affected tissues. Another limitation pertains to the MMC assay which detects homologous repair pathway defects and does not test deficiencies in other DNA repair pathways. WIDER IMPLICATIONS OF THE FINDINGS: Our results provide evidence for functional impairment of DNA repair in idiopathic POI, which may predispose the patients to other DNA repair-related conditions such as accelerated aging and/or cancer susceptibility. STUDY FUNDING/COMPETING INTEREST(S): Funding was provided by the National Institute of Child Health and Human Development. There were no competing interests to declare.


Subject(s)
Chromosomal Instability , DNA Copy Number Variations , Ovary/metabolism , Primary Ovarian Insufficiency/genetics , Adult , Comparative Genomic Hybridization , Female , Genome-Wide Association Study , Humans , Mutation , Pilot Projects , Primary Ovarian Insufficiency/metabolism
2.
Mol Endocrinol ; 22(8): 1842-52, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18535249

ABSTRACT

Activation of the protein kinase A (PKA) signaling system is necessary for FSH-induced granulosa cell differentiation, but it is not known whether activation of PKA is sufficient to account for the complex pattern of gene expression that occurs during this process. We addressed this question by infecting granulosa cells with a lentiviral vector that directs the expression of a constitutively active mutant of PKA (PKA-CQR) and compared the cellular responses to PKA-CQR with cells stimulated by FSH. Expression of PKA-CQR in undifferentiated granulosa cells resulted in the induction of both estrogen and progesterone production in the absence of cAMP. The stimulatory effects of both PKA-CQR and FSH on estrogen and progesterone production were suppressed by the PKA inhibitor H-89 and were mimicked by PKA-selective cAMP agonists. mRNA levels for P450scc and 3beta-HSD were induced to a similar extent by FSH and PKA-CQR, whereas mRNA levels for P450arom and the LHr were induced to a greater extent by FSH. Microarray analysis of gene expression profiles revealed that the majority of genes appeared to be comparably regulated by FSH and PKA-CQR but that some genes appear to be induced to a greater extent by FSH than by PKA-CQR. These results indicate that the PKA signaling pathway is sufficient to account for the induction of most genes (as identified by microarray analysis), including those of the progesterone biosynthetic pathway during granulosa cell differentiation. However, optimal induction of aromatase, the LHr, and other genes by FSH appears to require activation of additional signaling pathways.


Subject(s)
Cell Differentiation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Granulosa Cells/enzymology , Animals , Caveolin 1/genetics , Cell Differentiation/genetics , Cyclic AMP/analogs & derivatives , Cyclic AMP/biosynthesis , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Down-Regulation/drug effects , Enzyme Activation/drug effects , Estradiol/biosynthesis , Female , Gene Expression Profiling , Genetic Vectors , Granulosa Cells/drug effects , Humans , Lentivirus/genetics , Mutant Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Progesterone/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
3.
Endocrinology ; 148(2): 726-34, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17095585

ABSTRACT

Granulosa cells express the closely related orphan nuclear receptors steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1). To determine whether SF-1 and LRH-1 have differential effects on steroid production, we compared the effects of overexpressing LRH-1 and SF-1 on estrogen and progesterone production by undifferentiated rat granulosa cells. Adenovirus mediated overexpression of LRH-1 or SF-1 had qualitatively similar effects. Neither LRH-1 nor SF-1 alone stimulated estrogen or progesterone production, but when combined with FSH and testosterone, each significantly augmented progesterone production and mRNAs for cholesterol side-chain cleavage enzyme and 3beta-hydroxysteroid dehydrogenase above that observed with FSH alone, with SF-1 being more effective than LRH-1. LRH-1 did not augment FSH-stimulated estrogen production, whereas SF-1 produced only a slight ( approximately 30%) augmentation of FSH-stimulated estrogen production. The stimulatory actions of both were reduced by overexpression of dosage-sensitive sex reversal, adrenal hypoplasia congenita, critical region on the X chromosome, gene 1. Expression of either LRH-1 or SF-1 together with constitutively active protein kinase B in the absence of FSH stimulated progesterone production and mRNAs for 3beta-hydroxysteroid dehydrogenase and cholesterol side-chain cleavage enzyme but did not stimulate estrogen production or mRNA for aromatase. These findings demonstrate that LRH-1 and SF-1 have qualitatively similar actions on FSH-stimulated estrogen and progesterone production, which would suggest that these factors may have overlapping actions in the regulation of steroidogenesis that accompanies granulosa cell differentiation.


Subject(s)
Granulosa Cells/metabolism , Homeodomain Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Steroids/biosynthesis , Transcription Factors/physiology , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Drug Interactions , Drug Synergism , Estrogens/biosynthesis , Female , Follicle Stimulating Hormone/pharmacology , Gene Transfer Techniques , Granulosa Cells/drug effects , Homeodomain Proteins/genetics , Humans , Mice , Progesterone/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , RNA, Messenger/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Steroidogenic Factor 1 , Time Factors , Transcription Factors/genetics
4.
Endocrinology ; 158(7): 2043-2051, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28460125

ABSTRACT

Protein kinase A (PKA) has recently been shown to mimic the actions of follicle-stimulating hormone (FSH) by activating signaling pathways that promote granulosa cell (GC) differentiation, such as phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK). We sought to elucidate the mechanism by which PKA, a Ser/Thr kinase, intersected the PI3K/AKT and MAPK/ERK pathways that are canonically activated by receptor tyrosine kinases (RTKs). Our results show that for both of these pathways, the RTK is active in the absence of FSH yet signaling down the pathways to commence transcriptional responses requires FSH-stimulated PKA activation. For both pathways, PKA initiates signaling by regulating the activity of a protein phosphatase (PP). For the PI3K/AKT pathway, PKA activates the Ser/Thr PP1 complexed with the insulinlike growth factor 1 receptor (IGF-1R) and insulin receptor substrate 1 (IRS1) to dephosphorylate Ser residues on IRS1, authorizing phosphorylation of IRS1 by the IGF-1R to activate PI3K. Treatment of GCs with FSH and exogenous IGF-1 initiates synergistic IRS1 Tyr phosphorylation and resulting gene activation. The mechanism by which PKA activates PI3K is conserved in preovulatory GCs, MCF7 breast cancer cells, and FRTL thyroid cells. For the MAPK/ERK pathway, PKA promotes inactivation of the MAPK phosphatase (MKP) dual specificity phosphatase (DUSP) MKP3/DUSP6 to permit MEK-phosphorylated ERK to accumulate downstream of the epidermal growth factor receptor. Thus, for the two central signaling pathways that regulate gene expression in GCs, FSH via PKA intersects canonical RTK-regulated signaling by modulating the activity of PPs.


Subject(s)
Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/physiology , Granulosa Cells/physiology , Protein-Tyrosine Kinases/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Female , Humans , Insulin Receptor Substrate Proteins/metabolism , MAP Kinase Signaling System/genetics , MCF-7 Cells , Phosphorylation , Rats , Signal Transduction/genetics
5.
JAMA Pediatr ; 171(9): 879-886, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28672284

ABSTRACT

Importance: Brain injury may interrupt menstrual patterns by altering hypothalamic-pituitary-ovarian axis function. Investigators have yet to evaluate the association of concussion with menstrual patterns in young women. Objective: To compare abnormal menstrual patterns in adolescent and young women after a sport-related concussion with those after sport-related orthopedic injuries to areas other than the head (nonhead). Design, Setting, and Participants: This prospective cohort study of adolescent and young women with a sport-related concussion (n = 68) or a nonhead sport-related orthopedic injury (n = 61) followed up participants for 120 days after injury. Patients aged 12 to 21 years who presented within 30 days after a sport-related injury to a concussion or sports medicine clinic at a single academic center were eligible. Menstrual patterns were assessed using a weekly text message link to an online survey inquiring about bleeding episodes each week. The first patient was enrolled on October 14, 2014, and follow-up was completed on January 24, 2016. Inclusion criteria required participants to be at least 2 years postmenarche, to report regular menses in the previous year, and to report no use of hormonal contraception. Exposures: Sport-related concussion or nonhead sport-related orthopedic injury. Main Outcomes and Measures: Abnormal menstrual patterns were defined by an intermenstrual interval of less than 21 days (short) or more than 35 days (long) or a bleeding duration of less than 3 days or more than 7 days. Results: A total of 1784 survey responses were completed of the 1888 text messages received by patients, yielding 487 menstrual patterns in 128 patients (mean [SD] age, 16.2 [2.0] years). Of the 68 patients who had a concussion, 16 (23.5%) experienced 2 or more abnormal menstrual patterns during the study period compared with 3 of 60 patients (5%) who had an orthopedic injury. Despite similar gynecologic age, body mass index, and type of sports participation between groups, the risk of 2 or more abnormal menstrual bleeding patterns after injury was significantly higher among patients with concussion than among those with an orthopedic injury (odds ratio, 5.85; 95% CI, 1.61-21.22). Conclusions and Relevance: Adolescent and young women may have increased risk of multiple abnormal menstrual patterns after concussion. Because abnormal menstrual patterns can have important health implications, monitoring menstrual patterns after concussion may be warranted in this population. Additional research is needed to elucidate the relationship between long-term consequences of concussion and the function of the hypothalamic-pituitary-ovarian axis.


Subject(s)
Athletic Injuries/complications , Brain Concussion/complications , Menstruation Disturbances/etiology , Adolescent , Child , Cohort Studies , Female , Follow-Up Studies , Humans , Menstrual Cycle , Menstruation Disturbances/epidemiology , Prospective Studies , Risk Factors , Sports , Young Adult
6.
Sci Rep ; 6: 28132, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27324437

ABSTRACT

Activation of protein kinase A (PKA) by follicle stimulating hormone (FSH) transduces the signal that drives differentiation of ovarian granulosa cells (GCs). An unresolved question is whether PKA is sufficient to initiate the complex program of GC responses to FSH. We compared signaling pathways and gene expression profiles of GCs stimulated with FSH or expressing PKA-CQR, a constitutively active mutant of PKA. Both FSH and PKA-CQR stimulated the phosphorylation of proteins known to be involved in GC differentiation including CREB, ß-catenin, AKT, p42/44 MAPK, GAB2, GSK-3ß, FOXO1, and YAP. In contrast, FSH stimulated the phosphorylation of p38 MAP kinase but PKA-CQR did not. Microarray analysis revealed that 85% of transcripts that were up-regulated by FSH were increased to a comparable extent by PKA-CQR and of the transcripts that were down-regulated by FSH, 76% were also down-regulated by PKA-CQR. Transcripts regulated similarly by FSH and PKA-CQR are involved in steroidogenesis and differentiation, while transcripts more robustly up-regulated by PKA-CQR are involved in ovulation. Thus, PKA, under the conditions of our experimental approach appears to function as a master upstream kinase that is sufficient to initiate the complex pattern of intracellular signaling pathway and gene expression profiles that accompany GC differentiation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone, Human/metabolism , Granulosa Cells/physiology , Ovary/cytology , Animals , Cell Differentiation , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , Humans , Mutation/genetics , Rats , Rats, Inbred Strains , Signal Transduction , Steroids/metabolism , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Mol Endocrinol ; 16(1): 184-99, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11773448

ABSTRACT

The mitochondrial phosphoprotein, the steroidogenic acute regulatory (StAR) protein, is an essential component in the regulation of steroid biosynthesis in adrenal and gonadal cells through cAMP-dependent pathways. In many cases transcriptional induction by cAMP is mediated through the interaction of a cAMP response-element binding protein (CREB) family member with a consensus cAMP response element (CRE; 5'-TGACGTCA-3') found in the promoter of target genes. The present investigation was carried out to determine whether a CRE-binding protein (CREB) family member [CREB/CRE modulator (CREM) family] was involved in the regulation of steroidogenesis and StAR protein expression. Transient expression of wild- type CREB in MA-10 mouse Leydig tumor cells further increased the levels of (Bu)2cAMP-induced progesterone synthesis, StAR promoter activity, StAR mRNA, and StAR protein. These responses were significantly inhibited by transfection with a dominant-negative CREB (A-CREB), or with a CREB mutant that cannot be phosphorylated (CREB-M1), the latter observation indicating the importance of phosphorylation of a CREB/CREM family member in steroidogenesis and StAR expression. The CREB/CREM-responsive region in the mouse StAR gene was located between -110 and -67 bp upstream of the transcriptional start site. An oligonucleotide probe (-96/-67 bp) containing three putative half-sites for 5'-canonical CRE sequences (TGAC) demonstrated the formation of protein-DNA complexes in EMSAs with recombinant CREB protein as well as with nuclear extracts from MA-10 or Y-1 mouse adrenal tumor cells. The predominant binding factor observed with EMSA was found to be the CREM protein as demonstrated using specific antibodies and RT-PCR analyses. The CRE elements identified within the -96/-67 bp region were tested for cAMP responsiveness by generating mutations in each of the CRE half-sites either alone or in combination. Although each of the CRE sites contribute in part to the CREM response, the CRE2 appears to be the most important site as determined by EMSA and by reporter gene analyses. Binding specificity was further assessed using specific antibodies to CREB/CREM family members, cold competitors, and mutations in the target sites that resulted in either supershift and/or inhibition of these complexes. We also demonstrate that the inducible cAMP early repressor markedly diminished the endogenous effects of CREM on cAMP-induced StAR promoter activity and on StAR mRNA expression. These are the first observations to provide evidence for the functional involvement of a CREB/CREM family member in the acute regulation of trophic hormone-stimulated steroidogenesis and StAR gene expression.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Phosphoproteins/metabolism , Repressor Proteins , Steroids/metabolism , 5' Flanking Region , Animals , Base Sequence , COS Cells , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP Response Element Modulator , Cyclic AMP Response Element-Binding Protein/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Leydig Cells , Male , Mice , Molecular Sequence Data , Phosphoproteins/drug effects , Phosphoproteins/genetics , Promoter Regions, Genetic , Response Elements/genetics , Transcription, Genetic
8.
Endocrinology ; 144(9): 3985-94, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12933673

ABSTRACT

Although FSH receptors are linked to the cAMP second messenger system, additional intracellular signaling pathways appear to be required for the induction of aromatase and the LH receptor during granulosa cell differentiation. We employed adenovirus vectors to modulate specific intracellular signaling systems in undifferentiated granulosa cells to identify the signaling pathway(s) that may be involved in the FSH-mediated induction of aromatase and the LH receptor. Expression of either the constitutively activated human LH receptor D578H or the constitutively active human G(s)alpha Q227L resulted in increased cAMP production without increasing aromatase activity or mRNA levels for the LH receptor. To explore the contributions of other pathways, we expressed the constitutively activated forms MAPK kinase (MEK) and protein kinase B (PKB). Neither MEK nor PKB alone increased estrogen or progesterone production by undifferentiated granulosa cells. Stimulation of granulosa cells by FSH in the presence of the constitutively active PKB, but not MEK, led to an amplification of FSH-induced aromatase and LH receptor mRNA levels, whereas a dominant negative PKB vector completely abolished the actions of FSH. The expression of the constitutively active PKB in combination with the constitutively active LH receptor D578H, the constitutively active G(s)alpha Q227L, or 8-bromo-cAMP led to an induction of aromatase as well as LH receptor mRNA comparable to that seen in cells stimulated with FSH alone. These results demonstrate that PKB is an essential component of the FSH-mediated granulosa cell differentiation and that both PKB and G(s)alpha signaling pathways are required.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Granulosa Cells/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Sulfonamides , Adenoviridae/genetics , Androstadienes/pharmacology , Aromatase/metabolism , Biomarkers , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/biosynthesis , Cyclic AMP/pharmacology , Enzyme Inhibitors/pharmacology , Estrogens/biosynthesis , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Granulosa Cells/drug effects , Humans , Isoquinolines/pharmacology , Progesterone/biosynthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Wortmannin
9.
Endocrinology ; 145(8): 3821-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15117876

ABSTRACT

FSH-stimulated granulosa cell differentiation is associated with the induction of the LH receptor (LHr) as well as induction of the estrogen and progesterone biosynthetic pathways. Although activation of the cAMP-protein kinase A pathway is sufficient to stimulate progesterone production, additional pathways are required for the induction of the LHr and p450 aromatase. The orphan nuclear receptor, liver receptor homolog-1 (LRH-1), is expressed in granulosa cells and has been shown to synergize with the cAMP signaling system to regulate the gonadal type II aromatase promoter in transient transfection assays. To determine whether LRH-1 can interact with the cAMP pathway in the induction of aromatase and the LHr, we examined the effects of an adenoviral vector that directs the expression of human LRH-1 (Ad-LRH-1) on FSH-stimulated granulosa cell differentiation. Infection of undifferentiated granulosa cells with LRH-1 alone had no effect on estrogen production, progesterone production, or the expression of the LHr. However, combination of FSH stimulation and Ad-LRH-1 infection led to significantly greater progesterone production and increases in mRNA for p450 side-chain cleavage and 3beta-hydroxysteroid dehydrogenase than granulosa cells stimulated by FSH alone. However, infection with Ad-LRH-1 did not stimulate estradiol production or increases in mRNA for p450 aromatase or the LHr above that seen with FSH treatment alone. Moreover, infection with Ad-LRH-1 was able to overcome H-89 inhibition of FSH-stimulated progesterone but not estrogen production. Collectively, these observations support a direct role for LRH-1 in the induction of the progesterone but not the estrogen biosynthetic pathway during granulosa cell differentiation.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Progesterone/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenoviridae/genetics , Animals , Cell Differentiation , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/physiology , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/physiology , Dose-Response Relationship, Drug , Estrogens/biosynthesis , Female , Granulosa Cells/metabolism , Humans , Rats , Receptors, Retinoic Acid/physiology , Repressor Proteins/physiology
10.
J Clin Endocrinol Metab ; 89(2): 860-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14764806

ABSTRACT

Androgens, in addition to serving as a substrate for estrogen biosynthesis, exert autocrine/paracrine actions on ovarian function. However, much of the information regarding the actions of androgens on the ovary has been obtained using rodents, and the extent to which these results can be extrapolated to higher primates is uncertain. The current study was initiated to determine the effects of dihydrotestosterone (DHT) and testosterone (T) on the responsiveness of the rhesus monkey ovary to exogenous FSH and LH in vivo. Rhesus monkeys whose spontaneous gonadotropin secretion was interrupted with a GnRH antagonist received s. . implants of either DHT or T for 5 d before and continuing throughout a 15-d i.v. infusion of human FSH and LH. Neither T nor DHT treatment synergized with FSH/LH to stimulate estrogen production or increases in ovarian weight. Rather, administration of DHT significantly reduced estrogen secretion and the augmentation of ovarian weight in response to exogenously administered FSH and LH. These results indicate that high concentrations of DHT are antagonistic to gonadotropin-stimulated ovarian function in primates.


Subject(s)
Androgens/pharmacology , Dihydrotestosterone/pharmacology , Estrogens/biosynthesis , Follicle Stimulating Hormone, Human/pharmacology , Hormone Antagonists/pharmacology , Luteinizing Hormone/pharmacology , Ovary/drug effects , Ovary/metabolism , Androstenedione/blood , Animals , Female , Humans , Macaca mulatta , Organ Size/drug effects , Osmolar Concentration , Ovary/anatomy & histology , Testosterone/pharmacology
11.
J Clin Endocrinol Metab ; 87(12): 5722-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466377

ABSTRACT

Although it is well established that IGF-I is able to amplify the actions of FSH and LH on ovarian cells in vitro, little information is available regarding the effects of IGF-I on ovarian function in vivo. To address this question, rhesus monkeys whose spontaneous gonadotropin secretion was interrupted with a GnRH antagonist received continuous iv infusions of saline, IGF-I (240 microg/kg.d), or IGF-I (240 microg/kg.d) plus human GH (hGH) (200 microg/kg.d) 7 d before and continuing throughout a 15-d iv infusion of hFSH and hLH during which serum LH concentrations were maintained at 7-10 mIU/ml and FSH concentrations were incrementally increased every 3 d from 7.5 to 17.5 mIU/ml. Serum estradiol concentrations in saline-treated control animals did not differ (P > 0.05) from animals treated with IGF-I + hGH. In contrast, serum estradiol levels in IGF-I-treated animals were significantly less (P < 0.05) than those of control or IGF-I + hGH-treated animals. Serum androstenedione levels did not differ among the three treatment groups. Analysis of follicular fluids on the final day of gonadotropin infusion indicated that intrafollicular IGF-I concentrations paralleled serum IGF-I concentrations in all treatment groups. Measurement of the ratio of IGF-I to IGF-binding protein-3 in follicular fluids indicated that there was not a disproportionate increase in I-binding protein-3 in animals infused with either IGF-I alone or IGF-I + hGH. Concentrations of GH in follicular fluids of IGF-I treated animals were less than control animals suggesting that the diminished responsiveness of ovaries to FSH in the IGF-I treatment group may have been due to reduced GH.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Hormones/biosynthesis , Insulin-Like Growth Factor I/pharmacology , Luteinizing Hormone/pharmacology , Ovary/drug effects , Ovary/metabolism , Animals , Female , Follicle Stimulating Hormone/blood , Growth Hormone/metabolism , Human Growth Hormone/blood , Human Growth Hormone/pharmacology , Infusions, Intravenous , Insulin-Like Growth Factor I/analysis , Luteinizing Hormone/blood , Macaca mulatta , Time Factors
12.
Reprod Biol Endocrinol ; 2: 31, 2004 Jun 16.
Article in English | MEDLINE | ID: mdl-15200680

ABSTRACT

During the follicular phase of the primate menstrual cycle, a single follicle usually matures to the preovulatory stage and releases its oocyte for fertilization and the potential establishment of pregnancy. In assisted reproductive technology procedures, it is desirable to override the natural process of follicle selection to produce many oocytes that are capable of being fertilized and undergoing normal embryo development. The goal of this chapter is to summarize the current views regarding the natural process of follicle selection in primates and to discuss how this process may be amplified to produce a greater number of oocytes.


Subject(s)
Ovarian Follicle/physiology , Selection, Genetic , Animals , Female , Humans , Pregnancy
13.
Mol Endocrinol ; 23(5): 649-61, 2009 May.
Article in English | MEDLINE | ID: mdl-19196834

ABSTRACT

The forkhead box transcription factor FOXO1 is highly expressed in granulosa cells of growing follicles but is down-regulated by FSH in culture or by LH-induced luteinization in vivo. To analyze the function of FOXO1, we infected rat and mouse granulosa cells with adenoviral vectors expressing two FOXO1 mutants: a gain-of-function mutant FOXOA3 that has two serine residues and one threonine residue mutated to alanines rendering this protein constitutively active and nuclear and FOXOA3-mutant DNA-binding domain (mDBD) in which the DBD is mutated. The infected cells were then treated with vehicle or FSH for specific time intervals. Infection of the granulosa cells was highly efficient, caused only minimal apoptosis, and maintained FOXO1 protein at levels of the endogenous protein observed in cells before exposure to FSH. RNA was prepared from control and adenoviral infected cells exposed to vehicle or FSH for 12 and 24 h. Affymetrix microarray and database analyses identified, and real time RT-PCR verified, that genes within the lipid, sterol, and steroidogenic biosynthetic pathways (Hmgcs1, Hmgcr, Mvk, Sqle, Lss, Cyp51, Tm7sf2, Dhcr24 and Star, Cyp11a1, and Cyp19), including two key transcriptional regulators Srebf1 and Srebf2 of cholesterol biosynthesis and steroidogenesis (Nr5a1, Nr5a2), were major targets induced by FSH and suppressed by FOXOA3 and FOXOA3-mDBD in the cultured granulosa cells. By contrast, FOXOA3 and FOXOA3-mDBD induced expression of Cyp27a1 mRNA that encodes an enzyme involved in cholesterol catabolism to oxysterols. The genes up-regulated by FSH in cultured granulosa cells were also induced in granulosa cells of preovulatory follicles and corpora lutea collected from immature mice primed with FSH (equine choriogonadotropin) and LH (human choriogonadotropin), respectively. Conversely, Foxo1 and Cyp27a1 mRNAs were reduced by these same treatments. Collectively, these data provide novel evidence that FOXO1 may play a key role in granulosa cells to modulate lipid and sterol biosynthesis, thereby preventing elevated steroidogenesis during early stages of follicle development.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Forkhead Transcription Factors/physiology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Lipid Metabolism/physiology , Steroids/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genetic Vectors , Horses , Humans , Lipid Metabolism/genetics , Lipids , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
14.
Endocrinology ; 150(11): 5036-45, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19819952

ABSTRACT

Progesterone secretion by the steroidogenic cells of the corpus luteum (CL) is essential for reproduction. Progesterone synthesis is under the control of LH, but the exact mechanism of this regulation is unknown. It is established that LH stimulates the LH receptor/choriogonadotropin receptor, a G-protein coupled receptor, to increase cAMP and activate cAMP-dependent protein kinase A (PKA). In the present study, we tested the hypothesis that cAMP/PKA-dependent regulation of the Wnt pathway components glycogen synthase kinase (GSK)-3beta and beta-catenin contributes to LH-dependent steroidogenesis in luteal cells. We observed that LH via a cAMP/PKA-dependent mechanism stimulated the phosphorylation of GSK3beta at N-terminal Ser9 causing its inactivation and resulted in the accumulation of beta-catenin. Overexpression of N-terminal truncated beta-catenin (Delta90 beta-catenin), which lacks the phosphorylation sites responsible for its destruction, significantly augmented LH-stimulated progesterone secretion. In contrast, overexpression of a constitutively active mutant of GSK3beta (GSK-S9A) reduced beta-catenin levels and inhibited LH-stimulated steroidogenesis. Chromatin immunoprecipitation assays demonstrated the association of beta-catenin with the proximal promoter of the StAR gene, a gene that expresses the steroidogenic acute regulatory protein, which is a cholesterol transport protein that controls a rate-limiting step in steroidogenesis. Collectively these data suggest that cAMP/PKA regulation of GSK3beta/beta-catenin signaling may contribute to the acute increase in progesterone production in response to LH.


Subject(s)
Corpus Luteum/metabolism , Cyclic AMP-Dependent Protein Kinases , Cyclic AMP/metabolism , Glycogen Synthase Kinase 3/metabolism , Progesterone/biosynthesis , Signal Transduction , beta Catenin/metabolism , Animals , Cattle , Corpus Luteum/enzymology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Luteinizing Hormone/metabolism , Phosphorylation , beta Catenin/genetics
15.
Endocrine ; 33(1): 21-31, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18401763

ABSTRACT

Activation of FSH and LH receptors in undifferentiated granulosa cells (i.e., no prior exposure to FSH) results in comparable induction of progesterone production, but activation of the LH receptor is less effective than FSH in inducing aromatase and the native LH receptor. Because the LH receptor can also activate the Galphaq signaling pathway, we investigated whether activation of this pathway could be responsible for these differences. Overexpression of Galphaq inhibited FSH induction of both the estradiol and progesterone biosynthetic pathways as well as mRNA levels for cholesterol side-chain cleavage enzyme (P450scc), 3beta-hydroxysteroid dehydrogenase (3beta-HSD), LH receptor (LHr), and P450aromatase (aromatase). This suppression was associated with a reduction (P < 0.05) in FSH-stimulated cAMP production. Lower cAMP levels were not due to reduced FSH receptor (FSHr) mRNA levels or reduced levels of Galphas. Phosphodiesterase (PDE) activity and regulator of G-protein signaling 2 (RGS2) mRNA levels were significantly (P < 0.05) increased by Galphaq, both of which could account for diminished cAMP levels. We conclude that Galphaq signaling pathway inhibits both estradiol and progesterone production comparably and thus activation of this pathway does not seem to account for differences between FSH and LH in the regulation of aromatase and the LH receptor.


Subject(s)
Cell Differentiation/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Granulosa Cells/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Estradiol/metabolism , Female , Follicle Stimulating Hormone/pharmacology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Progesterone/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Rats , Receptors, FSH/genetics , Receptors, FSH/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection
16.
Proc Natl Acad Sci U S A ; 103(33): 12435-40, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16895991

ABSTRACT

Estrogens profoundly influence the physiology and pathology of reproductive and other tissues. Consequently, emphasis has been placed on delineating the mechanisms underlying regulation of estrogen levels. Circulating levels of estradiol in women are controlled by follicle-stimulating hormone (FSH), which regulates transcription of the aromatase gene (CYP19A1) in ovarian granulosa cells. Previous studies have focused on two downstream effectors of the FSH signal, cAMP and the orphan nuclear receptor steroidogenic factor-1 (NR5A1). In this report, we present evidence for beta-catenin (CTNNB1) as an essential transcriptional regulator of CYP19A1. FSH induction of select steroidogenic enzyme mRNAs, including Cyp19a1, is enhanced by beta-catenin. Additionally, beta-catenin is present in transcription complexes assembled on the endogenous gonad-specific CYP19A1 promoter, as evidenced by chromatin immunoprecipitation assays. Transient expression and RNAi studies demonstrate that FSH- and cAMP-dependent regulation of this promoter is sensitive to alterations in the level of beta-catenin. The stimulatory effect of beta-catenin is mediated through functional interactions with steroidogenic factor-1 that involve four acidic residues within its ligand-binding domain, mutation of which attenuates FSH/cAMP-induced Cyp19a1 mRNA accumulation. Together, these data demonstrate that beta-catenin is essential for FSH/cAMP-regulated gene expression in the ovary, identifying a central and previously unappreciated role for beta-catenin in estrogen biosynthesis, and a potential broader role in other aspects of follicular maturation.


Subject(s)
Aromatase/metabolism , Cyclic AMP/metabolism , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Enzymologic , beta Catenin/metabolism , Animals , Aromatase/genetics , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Estradiol/blood , Female , Granulosa Cells/cytology , Granulosa Cells/physiology , Humans , Ovary/cytology , Ovary/physiology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Steroidogenic Factor 1
SELECTION OF CITATIONS
SEARCH DETAIL