Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Sci Rep ; 11(1): 6657, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758252

ABSTRACT

Restricted food intake, either from lack of food sources or endogenous fasting, during reproductive periods is a widespread phenomenon across the animal kingdom. Considering previous studies show the canonical upstream regulator of reproduction in vertebrates, the hypothalamic Gonadotropin-releasing hormone (Gnrh), is inhibited in some fasting animals, we sought to understand the neuroendocrine control of reproduction in fasted states. Here, we explore the roles of the midbrain neuropeptide, Gnrh2, in inducing reproduction via its pituitary prevalence, gonadotropin synthesis, gametogenesis, and reproductive outputs in the zebrafish model undergoing different feeding regimes. We discovered a fasting-induced four-fold increase in length and abundance of Gnrh2 neuronal projections to the pituitary and in close proximity to gonadotropes, whereas the hypothalamic Gnrh3 neurons are reduced by six-fold in length. Subsequently, we analyzed the functional roles of Gnrh2 by comparing reproductive parameters of a Gnrh2-depleted model, gnrh2-/-, to wild-type zebrafish undergoing different feeding conditions. We found that Gnrh2 depletion in fasted states compromises spawning success, with associated decreases in gonadotropin production, oogenesis, fecundity, and male courting behavior. Gnrh2 neurons do not compensate in other circumstances by which Gnrh3 is depleted, such as in gnrh3-/- zebrafish, implying that Gnrh2 acts to induce reproduction specifically in fasted zebrafish.


Subject(s)
Fasting/metabolism , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropins/biosynthesis , Neurons/metabolism , Oogenesis , Reproduction , Zebrafish/physiology , Animals , Animals, Genetically Modified , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Neurites/metabolism , Pituitary Gland/metabolism , Reproduction/physiology
2.
Peptides ; 138: 170504, 2021 04.
Article in English | MEDLINE | ID: mdl-33539873

ABSTRACT

Stress impairs the hypothalamic-pituitary-gonadal (HPG) axis, probably through its influence on the hypothalamic-pituitary-adrenal (= interrenals in the teleost, HPI) axis leading to reproductive failures. In this study, we investigated the response of hypothalamic neuropeptides, gonadotropin-inhibitory hormone (GnIH), a component of the HPG axis, and corticotropin-releasing hormone (CRH) a component of the HPI axis, to acute social defeat stress in the socially hierarchical male Nile tilapia (Oreochromis niloticus). Localization of GnIH cell bodies, GnIH neuronal processes, and numbers of GnIH cells in the brain during acute social defeat stress was studied using immunohistochemistry. Furthermore, mRNA levels of GnIH and CRH in the brain together with GnIH receptor, gpr147, and adrenocorticotropic hormone (ACTH) in the pituitary were quantified in control and socially defeated fish. Our results show, the number of GnIH-immunoreactive cell bodies and GnIH mRNA levels in the brain and the levels of gpr147 mRNA in the pituitary significantly increased in socially defeated fish. However, CRH and ACTH mRNA levels did not change during social defeat stress. Further, we found glucocorticoid type 2b receptor mRNA in laser captured immunostained GnIH cells. These results show that acute social defeat stress activates GnIH biosynthesis through glucocorticoid receptors type 2b signalling but does not change the CRH and ACTH mRNA expression in the tilapia, which could lead to temporary reproductive dysfunction.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/biosynthesis , Stress, Psychological/genetics , Tilapia/physiology , Adrenocorticotropic Hormone/biosynthesis , Adrenocorticotropic Hormone/physiology , Animals , Brain/physiology , Corticotropin-Releasing Hormone/physiology , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/physiology , Gonadotropins/biosynthesis , Gonadotropins/genetics , Male , Pituitary Gland/metabolism , Reproduction/genetics , Reproduction/physiology , Social Defeat , Tilapia/genetics
3.
Toxicol Sci ; 162(1): 43-52, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29149324

ABSTRACT

Pyrethroids are a class of widely used insecticides. Cypermethrin (CP) is one of most commonly used pyrethroid insecticides and its residue has been frequently detected in environmental media. Our recent animal study reported that early postnatal exposure to CP induced an increase in serum levels of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) as well as the expression of gonadotropin subunit genes (chorionic gonadotropin α [CGα], LHß and FSHß) in pituitary tissues. In this study, we further investigated the precise mechanism by which CP at concentrations of 1-100 nM affected the synthesis of gonadotropins using a murine pituitary gonadotropic cell line LßT2. We found that calcium (Ca2+)-dependent extracellular signal-regulated kinase 1/2 (ERK1/2) activity was required for CP-regulated transcription of CGαs, LHß and FSHß. We provided the novel evidence that CP caused both influx of extracellular Ca2+ through L-type voltage-gated calcium channels (VGCCs) and release of intracellular Ca2+ from endoplasmic reticulum (ER) via inhibition of Ca2+-ATPase. Our results showed that CP disrupted Ca2+ homeostasis via these two separate and independent pathways, thus resulting in the activation of protein kinase C /c-Raf/ERK1/2/immediate-early genes pathways and subsequent increase in the transcription of gonadotropin subunit genes. Our findings would have important implications for understanding the underlying mechanisms of the disrupting effects of some pyrethroids (such as CP) on the synthesis of pituitary gonadotropins.


Subject(s)
Calcium/metabolism , Gonadotropins/biosynthesis , Homeostasis/drug effects , MAP Kinase Signaling System/drug effects , Pesticide Residues/toxicity , Pituitary Gland/drug effects , Pyrethrins/toxicity , Animals , Calcium Signaling/drug effects , Cell Culture Techniques , Cell Line , Follicle Stimulating Hormone, beta Subunit/genetics , Gene Expression Regulation , Glycoprotein Hormones, alpha Subunit/genetics , Luteinizing Hormone, beta Subunit/genetics , Mice , Pituitary Gland/metabolism
4.
Int J Cancer ; 140(8): 1870-1880, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28120505

ABSTRACT

The role of progenitor/stem cells in pituitary tumorigenesis, resistance to pharmacological treatments and tumor recurrence is still unclear. This study investigated the presence of progenitor/stem cells in non-functioning pituitary tumors (NFPTs) and tested the efficacy of dopamine receptor type 2 (DRD2) and somatostatin receptor type 2 (SSTR2) agonists to inhibit in vitro proliferation. They found that 70% of 46 NFPTs formed spheres co-expressing stem cell markers, transcription factors (DAX1, SF1, ERG1) and gonadotropins. Analysis of tumor behavior showed that spheres formation was associated with tumor invasiveness (OR = 3,96; IC: 1.05-14.88, p = 0.036). The in vitro reduction of cell proliferation by DRD2 and SSTR2 agonists (31 ± 17% and 35 ± 13% inhibition, respectively, p < 0.01 vs. basal) occurring in about a half of NFPTs cells was conserved in the corresponding spheres. Accordingly, these drugs increased cyclin-dependent kinase inhibitor p27 and decreased cyclin D3 expression in spheres. In conclusion, they provided further evidence for the existence of cells with a progenitor/stem cells-like phenotype in the majority of NFPTs, particularly in those with invasive behavior, and demonstrated that the antiproliferative effects of dopaminergic and somatostatinergic drugs were maintained in progenitor/stem-like cells.


Subject(s)
Carcinogenesis/genetics , Neoplasm Recurrence, Local/drug therapy , Pituitary Neoplasms/drug therapy , Receptors, Dopamine D2/genetics , Receptors, Somatostatin/genetics , Adult , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Cyclin D3/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , DAX-1 Orphan Nuclear Receptor/biosynthesis , Dopamine Agents/administration & dosage , Drug Resistance, Neoplasm/genetics , ERG1 Potassium Channel/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Gonadotropins/biosynthesis , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , RNA Splicing Factors/biosynthesis , Receptors, Dopamine D2/agonists , Receptors, Somatostatin/agonists , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology
5.
Biochim Biophys Acta ; 1849(3): 328-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25591470

ABSTRACT

The genes encoding luteinizing hormone and follicle stimulating hormone are activated by gonadotropin-releasing hormone (GnRH), and we hypothesized that this involves GnRH-induction of various histone modifications. At basal conditions in an immature gonadotrope-derived cell line, the hormone-specific ß-subunit gene promoters are densely packed with histones, and contain low levels of H3K4 trimethylation (H3K4me3). GnRH both induces this modification and causes histone loss, creating a more active chromatin state. The H3K4me3 appears to be mediated by menin and possibly catalyzed by the menin-mixed-lineage leukemia (MLL) 1/2 methyl transferase complex, as inhibition of MLL recruitment or menin knockdown reduced gene expression and the levels of H3K4me3 on all three promoters. Menin recruitment to the ß-subunit gene promoters is increased by GnRH, possibly involving transcription factors such as estrogen receptor α and/or steroidogenic factor 1, with which menin interacts. Menin also interacts with ring finger protein 20, which ubiquitylates H2BK120 (H2BK120ub), which was reported to be a pre-requisite for H3K4me3 at various gene promoters. Although levels of H2BK120ub are increased by GnRH in the coding regions of these genes, levels at the promoters do not correlate with those of H3K4me3, nor with gene expression, suggesting that H3K4me3 is not coupled to H2BK120ub in transcriptional activation of these genes.


Subject(s)
Follicle Stimulating Hormone/biosynthesis , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropins/genetics , Luteinizing Hormone/biosynthesis , Proto-Oncogene Proteins/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Estrogen Receptor alpha/metabolism , Follicle Stimulating Hormone/genetics , Gene Expression Regulation, Developmental , Gonadotropin-Releasing Hormone/genetics , Gonadotropins/biosynthesis , Histone Demethylases/genetics , Histones/genetics , Luteinizing Hormone/genetics , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Transcriptional Activation
6.
J Reprod Dev ; 60(2): 115-27, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24492640

ABSTRACT

This study was performed to investigate the protective and therapeutic effects of resveratrol (RES) against CdCl2-induced toxicity in rat testes. Seven experimental groups of adult male rats were formulated as follows: A) controls+NS, B) control+vehicle (saline solution of hydroxypropyl cyclodextrin), C) RES treated, D) CdCl2+NS, E) CdCl2+vehicle, F) RES followed by CdCl2 and M) CdCl2 followed by RES. At the end of the protocol, serum levels of FSH, LH and testosterone were measured in all groups, and testicular levels of TBARS and superoxide dismutase (SOD) activity were measured. Epididymal semen analysis was performed, and testicular expression of Bcl-2, p53 and Bax was assessed by RT-PCR. Also, histopathological changes of the testes were examined microscopically. Administration of RES before or after cadmium chloride in rats improved semen parameters including count, motility, daily sperm production and morphology, increased serum concentrations of gonadotropins and testosterone, decreased testicular lipid peroxidation and increased SOD activity. RES not only attenuated cadmium chloride-induced testicular histopathology but was also able to protect against the onset of cadmium chloride testicular toxicity. Cadmium chloride downregulated the anti-apoptotic gene Bcl2 and upregulated the expression of pro-apoptotic genes p53 and Bax. Resveratrol protected against and partially reversed cadmium chloride testicular toxicity via upregulation of Bcl2 and downregulation of p53 and Bax gene expression. The antioxidant activity of RES protects against cadmium chloride testicular toxicity and partially reverses its effect via upregulation of BCl2 and downregulation of p53 and Bax expression.


Subject(s)
Cadmium Chloride/antagonists & inhibitors , Gonadotropins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Stilbenes/pharmacology , Testis/drug effects , Tumor Suppressor Protein p53/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Animals , Antioxidants/pharmacology , Cadmium Chloride/toxicity , Drug Interactions , Follicle Stimulating Hormone/blood , Gene Expression Regulation/drug effects , Gonadotropins/genetics , Histocytochemistry , Infertility, Male/blood , Infertility, Male/chemically induced , Infertility, Male/drug therapy , Luteinizing Hormone/blood , Male , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Rats, Wistar , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/blood , Testis/metabolism , Testosterone/blood , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics
7.
Reprod Biol Endocrinol ; 11: 68, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23866168

ABSTRACT

BACKGROUND: Injecting estradiol valerate (EV) to pre-pubertal or adult female rat results in effects similar to those observed in women with polycystic ovarian syndrome (PCOS). One of the mechanisms involved in PCOS development is the hyperactivity of the sympathetic nervous system. In EV-induced PCOS rats, the unilateral sectioning of the superior ovarian nerve (SON) restores ovulation of the innervated ovary. This suggests that, in addition to the sympathetic innervation, other neural mechanisms are involved in the development/maintenance of PCOS. The aims of present study were analyze if the vagus nerve is one of the neural pathways participating in PCOS development. METHODS: Ten-day old rats were injected with EV dissolved in corn oil. At 24-days of age sham-surgery, unilateral, or bilateral sectioning of the vagus nerve (vagotomy) was performed on these rats. The animals were sacrificed at 90-92 days of age, when they presented vaginal estrous preceded by a pro-estrus smear. RESULTS: In EV-induced PCOS rats, unilateral or bilateral vagotomy restored ovulation in both ovaries. Follicle-stimulating hormone (FSH) levels in PCOS rats with unilateral or bilateral vagotomy were lower than in control rats. CONCLUSIONS: This result suggests that in EV-induced PCOS rats the vagus nerve is a neural pathway participating in maintaining PCOS. The vagus nerve innervates the ovaries directly and indirectly through its synapsis in the celiac-superior-mesenteric ganglion, where the somas of neurons originating in the SON are located. Then, it is possible that vagotomy effects in EV-induced PCOS rats may be explained as a lack of communication between the central nervous system and the ovaries.


Subject(s)
Ovary/physiopathology , Ovulation/physiology , Polycystic Ovary Syndrome/physiopathology , Vagotomy , Animals , Estrous Cycle/physiology , Female , Gonadal Steroid Hormones/biosynthesis , Gonadotropins/biosynthesis , Neural Pathways/physiology , Ovary/pathology , Polycystic Ovary Syndrome/pathology , Rats , Steroids/biosynthesis , Vagus Nerve/physiology
8.
Mol Endocrinol ; 27(8): 1343-56, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23798575

ABSTRACT

Gonadotropin expression is precisely regulated within the hypothalamic-pituitary-gonadal axis through the complex interaction of neuropeptides, gonadal steroids. and both gonadal- and pituitary-derived peptides. In the anterior pituitary gland, the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) modulates gonadotropin biosynthesis and secretion, acting both alone and in conjunction with GnRH. Steroid hormone feedback also influences gonadotropin expression via both direct and indirect mechanisms. Evidence from nonpituitary tissues suggests that PACAP may be a target for gonadal steroid regulation. In the present study, we show that androgen markedly stimulates rat (r) PACAP promoter-reporter activity in the LßT2 mature mouse gonadotrope cell line. 5'-Serial deletion analysis of reporter constructs identifies 2 regions of androgen responsiveness located at (-915 to -818) and (-308 to -242) of the rPACAP promoter. Androgen receptor (AR) binds directly to DNA cis-elements in each of these regions in vitro. Site-directed mutagenesis of 3 conserved hormone response element half-sites straddling the (-308 to -242) region dramatically blunts androgen-dependent PACAP promoter activity and prevents AR binding at the mutated promoter element. Chromatin immunoprecipitation demonstrates that endogenous AR binds the homologous region on mouse chromatin in LßT2 cells in both the presence and absence of androgen. These data demonstrate that androgen stimulates PACAP gene expression in the pituitary gonadotrope via direct binding of AR to a specific cluster of evolutionarily conserved hormone response elements in the proximal rPACAP gene promoter. Thus, androgen regulation of pituitary PACAP expression may provide an additional layer of control over gonadotropin expression within the hypothalamic-pituitary-gonadal axis.


Subject(s)
Androgens/metabolism , Gonadotrophs/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Gland, Anterior/metabolism , Receptors, Androgen/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , Gonadotropins/biosynthesis , Gonadotropins/metabolism , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Pituitary Adenylate Cyclase-Activating Polypeptide/biosynthesis , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Gland, Anterior/enzymology , Promoter Regions, Genetic , Protein Binding/genetics , Rats , Regulatory Sequences, Nucleic Acid , Response Elements/genetics , Signal Transduction/genetics , Transcription, Genetic
9.
PLoS One ; 8(3): e59058, 2013.
Article in English | MEDLINE | ID: mdl-23536856

ABSTRACT

Successful mammalian reproduction depends on proper synthesis of the pituitary-derived glycoprotein hormones, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Several transcription factors cooperate to activate cell-specific and hormone-regulated expression of the gonadotropin beta subunits (Lhb and Fshb). Among these, NR5A1 (steroidogenic factor 1; SF1) has been shown to directly bind to the Lhb promoter, mediate basal and gonadotropin-releasing hormone (GnRH)-stimulated Lhb transcription, and possibly directly regulate Fshb expression. Recently, the closely-related NR5A2 was shown to activate the rat Lhb promoter in vitro. Here, we further characterized the role of NR5A2 in regulating gonadotropin synthesis. Ectopically expressed NR5A2 directly activated the murine Lhb promoter in a manner identical to that of NR5A1, whereas neither factor activated the murine Fshb promoter. In LßT2 gonadotrope-like cells, depletion of endogenous NR5A1 or NR5A2 impaired basal and GnRH-stimulated Lhb and Fshb transcription. To analyze the physiological role of NR5A2 in gonadotropes in vivo, we generated mice with a gonadotrope-specific deletion of Nr5a2. In contrast with our in vitro data, these mice had normal pituitary Lhb and Fshb expression and intact fertility. Together, our data establish that NR5A2 can act in a non-redundant manner to regulate Lhb and Fshb transcription in vitro, but is dispensable in vivo.


Subject(s)
Fertility/genetics , Follicle Stimulating Hormone, beta Subunit/genetics , Gene Expression Regulation , Gonadotrophs/metabolism , Gonadotropins/biosynthesis , Luteinizing Hormone, beta Subunit/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Animals , Cell Line , Female , Gene Expression Regulation/drug effects , Gene Order , Gonadotrophs/cytology , Gonadotropin-Releasing Hormone/pharmacology , Male , Mice , Mice, Knockout , Mice, Transgenic , Pituitary Gland/metabolism , Promoter Regions, Genetic , RNA Isoforms , RNA, Messenger , Receptors, Cytoplasmic and Nuclear/genetics , Response Elements , Transcriptional Activation
10.
Andrology ; 1(2): 206-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23413134

ABSTRACT

Recently, we reported large differences between rat strains in spermatogenesis recovery at 10 weeks after 5-Gy irradiation suggesting that there are interstrain as well as interspecies differences in testicular radiation response. To determine whether these interstrain differences in sensitivity might be a result of the particular dose and time-point chosen, we performed dose-response and time-course studies on sensitive Brown-Norway (BN) and more resistant spontaneously hypertensive rats (SHR) and Sprague-Dawley (SD) rats. Type A spermatogonia were observed in atrophic tubules at 10 weeks after irradiation in all strains indicating that tubular atrophy was caused by a block in their differentiation, but the doses to produce the block ranged from 4.0 Gy in BN to 10 Gy in SD rats. Although the numbers of type A spermatogonial were unaffected at doses below 6 Gy, higher doses reduced their number, indicating that stem cell killing also contributed to the failure of recovery. After 10 weeks, there was no further recovery and even a decline in spermatogonial differentiation in BN rats, but in SHR rats, sperm production returned to control levels by 20 weeks after 5.0 Gy and, after 7.5 Gy, differentiation resumed in 60% of tubules by 30 weeks. Suppression of testosterone and gonadotropins after irradiation restored production of differentiated cells in nearly all tubules in BN rats and in all tubules in SHR rats. Thus, the differences in recovery of spermatogenesis between strains were a result of both quantitative differences in their sensitivities to a radiation-induced, hormone-dependent block of spermatogonial differentiation and qualitative interstrain differences in the progression of post-irradiation recovery. The progression of recovery in SHR rats was similar to the prolonged delays in recovery of human spermatogenesis after cytotoxic agent exposure and thus may be a system for investigating a phenomenon also observed in men.


Subject(s)
Spermatogenesis/radiation effects , Spermatogonia/radiation effects , Testis/radiation effects , Animals , Cell Differentiation/radiation effects , Dose-Response Relationship, Radiation , Gonadotropins/biosynthesis , Gonadotropins/radiation effects , Male , Models, Animal , Rats/classification , Rats, Sprague-Dawley , Spermatogenesis/physiology , Spermatogonia/physiology , Stem Cells/radiation effects , Testis/physiology , Testosterone/biosynthesis , Testosterone/radiation effects
11.
Mol Endocrinol ; 27(2): 253-65, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23275456

ABSTRACT

GnRH is a potent hypothalamic regulator of gonadotropin hormones, LH and FSH, which are both expressed within the pituitary gonadotrope and are necessary for the stimulation of gametogenesis and steroidogenesis in the gonads. Differential regulation of LH and FSH, which is essential for reproductive fitness, is achieved, in part, through the varying of GnRH pulse frequency. However, the mechanism controlling the increase in FSH during the periods of low GnRH has not been elucidated. Here, we uncover another level of regulation by GnRH that contributes to differential expression of the gonadotropins and may play an important role for the generation of the secondary rise of FSH that stimulates folliculogenesis. GnRH stimulates LHß and FSHß subunit transcription via induction of the immediate early genes, Egr1 and c-Fos, respectively. Here, we determined that GnRH induces rapidly both Egr1 and c-Fos, but specifically decreases the rate of c-Fos degradation. In particular, GnRH modulates the rate of c-Fos protein turnover by inducing c-Fos phosphorylation through the ERK1/2 pathway. This extends the half-life of c-Fos, which is normally rapidly degraded. Confirming the role of phosphorylation in promoting increased protein activity, we show that a c-Fos mutant that cannot be phosphorylated by GnRH induces lower expression of the FHSß promoter than wild-type c-Fos. Our studies expand upon the role of GnRH in the regulation of gonadotropin gene expression by highlighting the role of c-Fos posttranslational modification that may cause higher levels of FSH during the time of low GnRH pulse frequency to stimulate follicular growth.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/biosynthesis , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Animals , Cell Line , Early Growth Response Protein 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Gene Expression Regulation , Luteinizing Hormone, beta Subunit/biosynthesis , Luteinizing Hormone, beta Subunit/genetics , Mice , Mutation , Ovarian Follicle/metabolism , Phosphorylation , Pituitary Gland/metabolism , Protein Processing, Post-Translational , Transcription, Genetic
12.
Mol Endocrinol ; 27(2): 224-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23211523

ABSTRACT

The regulation of gonadotropin synthesis by GnRH plays an essential role in the neuroendocrine control of reproduction. The known signaling mechanisms involved in gonadotropin synthesis have been expanding. For example, involvement of ß-catenin in LHß induction by GnRH has been discovered. We examined the role of ß-catenin in FSHß gene expression in LßT2 gonadotrope cells. GnRH caused a sustained increase in nuclear ß-catenin levels, which was significantly reduced by c-Jun N-terminal kinase (JNK) inhibition. Small interfering RNA-mediated knockdown of ß-catenin mRNA demonstrated that induction of FSHß mRNA by GnRH depended on ß-catenin and that regulation of FSHß by ß-catenin occurred independently of the JNK-c-jun pathway. ß-Catenin depletion had no impact on FSHß mRNA stability. In LßT2 cells transfected with FSHß promoter luciferase fusion constructs, GnRH responsiveness was conferred by the proximal promoter (-944/-1) and was markedly decreased by ß-catenin knockdown. However, none of the T-cell factor/lymphoid enhancer factor binding sites in that region were required for promoter activation by GnRH. Chromatin immunoprecipitation further corroborated the absence of direct interaction between ß-catenin and the 1.8-kb FSHß promoter. To elucidate the mechanism for the ß-catenin effect, we analyzed approximately 1 billion reads of next-generation RNA sequencing ß-catenin knockdown assays and selected the nuclear cofactor breast cancer metastasis-suppressor 1-like (Brms1L) as one candidate for further study. Subsequent experiments confirmed that Brms1L mRNA expression was decreased by ß-catenin knockdown as well as by JNK inhibition. Furthermore, knockdown of Brms1L significantly attenuated GnRH-induced FSHß expression. Thus, our findings indicate that the expression of Brms1L depends on ß-catenin activity and contributes to FSHß induction by GnRH.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotropin-Releasing Hormone/metabolism , Repressor Proteins/biosynthesis , beta Catenin/metabolism , Animals , Anthracenes/pharmacology , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Follicle Stimulating Hormone, beta Subunit/biosynthesis , Gene Expression , Gonadotropins/biosynthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , TCF Transcription Factors/metabolism , beta Catenin/genetics
13.
Annu Rev Neurosci ; 35: 133-51, 2012.
Article in English | MEDLINE | ID: mdl-22524786

ABSTRACT

In the course of evolution, social behavior has been a strikingly potent selective force in shaping brains to control action. Physiological, cellular, and molecular processes reflect this evolutionary force, particularly in the regulation of reproductive behavior and its neural circuitry. Typically, experimental analysis is directed at how the brain controls behavior, but the brain is also changed by behavior over evolution, during development, and through its ongoing function. Understanding how the brain is influenced by behavior offers unusual experimental challenges. General principles governing the social regulation of the brain are most evident in the control of reproductive behavior. This is most likely because reproduction is arguably the most important event in an animal's life and has been a powerful and essential selective force over evolution. Here I describe the mechanisms through which behavior changes the brain in the service of reproduction using a teleost fish model system.


Subject(s)
Brain/physiology , Sexual Behavior, Animal/physiology , Social Behavior , Social Control, Informal , Animals , Cichlids , Gene Expression Regulation/physiology , Gonadal Steroid Hormones/biosynthesis , Gonadotropins/biosynthesis , Models, Biological , Receptors, LHRH/biosynthesis , Spermatogenesis/genetics
14.
Endocrinology ; 153(3): 1421-34, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315445

ABSTRACT

The neuropeptide pituitary adenylate cyclase activating polypeptide (PACAP) is present in high concentrations within the hypothalamus, suggesting that it may be a hypophysiotropic factor, whereas pituitary expression suggests a paracrine function. PACAP stimulates gonadotropin secretion and enhances GnRH responsiveness. PACAP increases gonadotropin α-subunit (αGSU), lengthens LHß, but reduces FSHß mRNA levels in adult pituitary cell cultures in part by increasing follistatin. PACAP stimulates LH secretion in rats; however, acceptance of PACAP as a regulator of reproduction has been limited by a paucity of in vivo studies. We created a transgenic mouse model of pituitary PACAP overexpression using the αGSU subunit promoter. Real-time PCR was used to evaluate PACAP, follistatin, GnRH receptor, and the gonadotropin subunit mRNA in male transgenic and wild-type mice of various ages. Transgenic mice had greater than 1000-fold higher levels of pituitary PACAP mRNA; and immunocytochemistry, Western blot, and ELISA analyses confirmed high peptide levels. FSH, LH, and testosterone levels were significantly suppressed, and the timing of puberty was substantially delayed in PACAP transgenic mice in which gonadotropin subunit and GnRH receptor mRNA levels were reduced and pituitary follistatin expression was increased. Microarray analyses revealed 1229 of 45102 probes were significantly (P < 0.01) different in pituitaries from PACAP transgenic mice, of which 83 genes were at least 2-fold different. Genes involved in small molecule biochemistry, cancer, and reproductive system diseases were the top associated networks. The GnRH signaling pathway was the top canonical pathway affected by pituitary PACAP excess. These experiments provide the first evidence that PACAP affects gonadotropin expression and sexual maturation in vivo.


Subject(s)
Gene Expression Regulation , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Gland/metabolism , Animals , Follistatin/biosynthesis , Gonadotropins/biosynthesis , Luteinizing Hormone/metabolism , Male , Mice , Mice, Transgenic , Models, Biological , RNA, Messenger/metabolism , Rats , Receptors, LHRH/biosynthesis , Sexual Behavior, Animal , Signal Transduction , Testosterone/metabolism
15.
Mol Hum Reprod ; 17(12): 739-46, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21775412

ABSTRACT

DAX-1 [dosage-sensitive sex reversal-adrenal hypoplasia congenital (AHC) critical region on the X chromosome gene 1; NR0B1] is an orphan nuclear receptor that acts as a transcriptional repressor in adrenal/gonadal development, steroidogenesis and probably spermatogenesis. An alternatively spliced form called DAX-1A (NR0B1A) has been described in several tissues including the testis, and in vitro studies have shown an inhibitory effect on DAX-1 transcriptional function. We aimed to study the mRNA and protein expression of DAX-1 in testicular tissues of 65 men with primary spermatogenic failure [complete Sertoli cell only syndrome (SCOS), focal SCOS, maturation arrest and mixed atrophy] compared with 33 controls with normal spermatogenesis. As a novel finding, we observed intense immunostaining, not only in the nucleus of Sertoli cells, but also in pachytene spermatocytes and round spermatids. The quantitative mRNA expression of DAX-1 and DAX-1A was similar between cases and controls and was not associated with the levels of gonadotrophins and steroids. Moreover, DAX-I transcript expression level was ∼750-fold higher than DAX-1A, and there was a strong positive correlation between them (r = 0.52; P< 0.001). We conclude that, in addition to Sertoli cells, DAX-1/DAX-1A is expressed in germ cells from spermatogonia to round spermatids. Besides, the similar mRNA expression of DAX-I and DAX-IA in testicular tissues from cases and controls does not support the involvement of DAX-1 in the etiology of primary spermatogenic failure. Finally, the low level of expression of the alternative transcriptional variant DAX-1A would not support its putative inhibitory function in vivo.


Subject(s)
DAX-1 Orphan Nuclear Receptor/metabolism , Gene Expression , Protein Isoforms/metabolism , Reproduction/genetics , Sertoli Cell-Only Syndrome/genetics , Sertoli Cells/metabolism , Spermatogenesis , Adult , Alternative Splicing , Case-Control Studies , Chile , DAX-1 Orphan Nuclear Receptor/genetics , Gonadotropins/biosynthesis , Humans , Immunohistochemistry , Male , Protein Isoforms/genetics , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Sertoli Cells/pathology , Spermatids/cytology , Spermatids/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Steroids/biosynthesis
16.
Rev. chil. endocrinol. diabetes ; 4(2): 126-135, abr. 2011. tab, ilus
Article in Spanish | LILACS | ID: lil-640600

ABSTRACT

Pulsatile secretion of Gonadotropin-Releasing Hormone (GnRH) by the hipothalamus and its action on the pituitary gland is a complex process involving many pre and post natal events. For example, migration of GnRH neurons from the olfactory placode, GnRH release and signalling, normal anterior pituitary development and function are all needed to allow GnRH to stimulate pulsaltile pituitary secretion of follicle-stimulating hormone (FSH) and liteinizing hormone (LH). Hypogonadotropic hypogonadism can be the result of absent or inadequate GnRH secretion or action. Abnormalities in gonadotropin hormone release and function can arise from mutations in a variety of genes implicated in hypogonadotropic hypogonadism is continually growing. A given genotype at a single locus cannot reliably predict the phenotypic manifestations in any given member of affected families. Thus, the identification and characterization of these mutations are providing important information about the reproductive axis in humans and may result in improved treatment and counselling for patients with infertility.


Subject(s)
Humans , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone , Fertility , Follicle Stimulating Hormone , Gonadotropins/biosynthesis , Gonadotropins/genetics , Gonadotropins , Hypogonadism/genetics , Luteinizing Hormone , Mutation , Pituitary Gland , Reproduction , Kallmann Syndrome/genetics
17.
Brain Res ; 1364: 186-97, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20816944

ABSTRACT

Optimal fat mass is necessary for normal gonadotropin levels in adults, and both undernutrition and overnutrition suppress gonadotropins: thus, the gonadotropin response to relative adipose mass is biphasic. Adult obesity is associated with blunted luteinizing hormone (LH) pulse amplitude that is partially attributable to increased LH clearance rate. Testosterone appears to have a biphasic effect on gonadotropin production in females. Moderate elevations of testosterone appear to stimulate LH production at both the hypothalamic and pituitary level, while very high levels of testosterone suppress LH. Thus, obesity per se appears to suppress gonadotropin production, and moderate hyperandrogenemia in women appears to stimulate LH. The ordinary hypergonadotropic hyperandrogenism of obese women appears to be an exception to this model because it is usually due to polycystic ovary syndrome (PCOS), a condition in which intrinsic functional ovarian hyperandrogenism and excess adiposity share a common origin that involves insulin-resistant hyperinsulinemia. LH elevation seems to be secondary to hyperandrogenemia and is absent in the most obese cases. Overweight early pubertal girls have significant blunting of sleep-related LH production, which is the first hormonal change of puberty. The data are compatible with the possibility that excess adiposity may paradoxically subtly suppress hypothalamic-pituitary-gonadal function in early puberty although it is known to contribute to the early onset of puberty.


Subject(s)
Aging/physiology , Androgens/physiology , Gonadotropins/biosynthesis , Obesity/physiopathology , Puberty/physiology , Animals , Female , Humans , Luteinizing Hormone/physiology , Male , Neurosecretory Systems/physiology , Polycystic Ovary Syndrome/physiopathology , Sexual Maturation/physiology
18.
Endocrinology ; 151(10): 4787-800, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20702576

ABSTRACT

The Lin-11, Isl-1, and Mec-3 (LIM) homeodomain transcription factor Isl-1 has been reported to be involved in pituitary development in the early stages of mouse embryogenesis. Our recent studies have shown that Isl-1 is mainly located in the pituitary gonadotropes throughout pituitary development and persists to adulthood. We still do not know the physiological functions of Isl-1 expression and its related mechanisms in the pituitary gland. The aim of the present study was to examine the hypothesis that Isl-1 is involved in regulating pituitary gonadotropin hormone (FSH/LH) production by activating FSHß and LHß gene expressions. We have shown that Isl-1 activates FSHß and LHß subunit promoters and endogenous gene transcription in LßT2 cells. In addition, Isl-1 overexpression significantly increased FSH synthesis and secretion but not LH. The actions of Isl-1 were not observed when the homeodomain or LIM1 domains are mutated. This demonstrates that Isl-1 induction of FSHß and LHß is by both direct and indirect binding of Isl-1 to DNA sequences. Furthermore, Isl-1 expressional level was up-regulated in LßT2 cells after exposure to GnRH, activin, and leptin. However, RNA interference-induced knockdown of Isl-1 significantly reduced the effect of leptin but did not obviously influence the stimulating effects of GnRH and activin on LH and FSH production. In conclusion, the results demonstrate that the LIM-homeodomain transcription factor Isl-1 functions to increase FSHß/LHß gene transcription, and mediates the effects of leptin on gonadotropin synthesis.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotropins/biosynthesis , Homeodomain Proteins/physiology , Leptin/pharmacology , Luteinizing Hormone, beta Subunit/genetics , Animals , Base Sequence , Cells, Cultured , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression/drug effects , Gonadotrophs/drug effects , Gonadotrophs/metabolism , Gonadotropins/genetics , Gonadotropins/metabolism , Homeodomain Proteins/metabolism , Humans , LIM-Homeodomain Proteins , Leptin/metabolism , Luteinizing Hormone, beta Subunit/metabolism , Male , Mice , Molecular Sequence Data , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Transcription Factors , Up-Regulation/drug effects , Up-Regulation/genetics
19.
J Toxicol Sci ; 35(3): 365-73, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20519845

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) reduces the synthesis of pituitary gonadotropins in a fetal age-specific manner. The pituitary synthesis of gonadotropins is regulated by the hypothalamus and, thus, needs the differentiation and development of the hypothalamus requiring a number of factors including energy supply and neurotransmitters. To investigate the mechanism whereby TCDD reduces fetal gonadotropins, we carried out a comparative study on the metabolomes of the hypothalamus and pituitary using fetal and mature Wistar rats. Male fetuses at gestational day (GD)20 were removed from dams treated orally with TCDD (1 microg/kg) at GD15, and the metabolome profiles were analyzed by gas chromatography-mass spectrometry (GC-MS). The principal component analysis of GC-MS data revealed that TCDD caused a change in the profile of fetal metabolome more markedly in the hypothalamus than in the pituitary. In sharp contrast, TCDD did not cause any marked alteration in hypothalamic as well as pituitary metabolomes in male rats born of untreated dams and treated with TCDD at postnatal day 49. It was also demonstrated that a number of fetal hypothalamic components, including glutamine and gamma-aminobutyric acid, are reduced by TCDD. These results demonstrate a possibility that TCDD may reduce the metabolic activity of the hypothalamus in a fetus-specific fashion, resulting in the reduced synthesis of gonadotropins.


Subject(s)
Gonadotropins/biosynthesis , Hypothalamus/metabolism , Maternal Exposure/adverse effects , Maternal-Fetal Exchange , Metabolome/drug effects , Polychlorinated Dibenzodioxins/adverse effects , Animals , Depression, Chemical , Female , Gas Chromatography-Mass Spectrometry , Glutamine/metabolism , Male , Pituitary Gland/metabolism , Pregnancy , Rats , Rats, Wistar , gamma-Aminobutyric Acid/metabolism
20.
Singapore Med J ; 50(11): 1080-4, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19960164

ABSTRACT

INTRODUCTION: Autoimmune hypophysitis (AH) is a rare primary autoimmune inflammatory disorder involving the pituitary gland. METHODS: A retrospective analysis of the clinical features and outcome of patients diagnosed with AH between 1988 and 2006, was carried out. RESULTS: 15 patients (14 females and one male) with AH were identified. Three patients presented in the peripartum period. Headache, vomiting and visual field defects, suggestive of an expanding sellar mass, were the most common presenting symptoms (67 percent). The most common deficient hormone was adrenocorticotropic hormone (ACTH) (67 percent), followed by thyroid stimulating hormone (53 percent) and gonadotropins (40 percent). Imaging revealed a definite, enhancing sellar mass in 87 percent of the patients and stalk thickening in 33 percent of the patients. Three patients underwent surgery. On serial monitoring, the sellar mass regressed or disappeared spontaneously without any immunosuppressive treatment in the other ten patients with a definite sellar mass. CONCLUSION: We report a higher female to male ratio and a lower incidence of peripartum cases in our series. Symptoms of mass effect were the most common presentation, while ACTH was the most commonly-deficient hormone. Surgery was rarely needed, and most patients experienced a spontaneous resolution of the mass.


Subject(s)
Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Pituitary Diseases/diagnosis , Pituitary Diseases/therapy , Adrenocorticotropic Hormone/biosynthesis , Adult , Female , Gonadotropins/biosynthesis , Humans , Inflammation , Male , Middle Aged , Pituitary Gland/pathology , Retrospective Studies , Sex Factors , Thyrotropin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...