Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Endocrinology ; 163(2)2022 02 01.
Article En | MEDLINE | ID: mdl-34864945

Gonadotropin-releasing hormone (GnRH) regulates gonadal function via its stimulatory effects on gonadotropin production by pituitary gonadotrope cells. GnRH is released from the hypothalamus in pulses and GnRH pulse frequency differentially regulates follicle-stimulating hormone (FSH) and luteinizing hormone (LH) synthesis and secretion. The GnRH receptor (GnRHR) is a G protein-coupled receptor that canonically activates Gα q/11-dependent signaling on ligand binding. However, the receptor can also couple to Gα s and in vitro data suggest that toggling between different G proteins may contribute to GnRH pulse frequency decoding. For example, as we show here, knockdown of Gα s impairs GnRH-stimulated FSH synthesis at low- but not high-pulse frequency in a model gonadotrope-derived cell line. We next used a Cre-lox conditional knockout approach to interrogate the relative roles of Gα q/11 and Gα s proteins in gonadotrope function in mice. Gonadotrope-specific Gα q/11 knockouts exhibit hypogonadotropic hypogonadism and infertility, akin to the phenotypes seen in GnRH- or GnRHR-deficient mice. In contrast, under standard conditions, gonadotrope-specific Gα s knockouts produce gonadotropins at normal levels and are fertile. However, the LH surge amplitude is blunted in Gα s knockout females and postgonadectomy increases in FSH and LH are reduced both in males and females. These data suggest that GnRH may signal principally via Gα q/11 to stimulate gonadotropin production, but that Gα s plays important roles in gonadotrope function in vivo when GnRH secretion is enhanced.


Chromogranins/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Gonadotrophs/metabolism , Gonadotropins/metabolism , Animals , Castration , Cell Line , Chromogranins/genetics , Female , Fertility/genetics , Fertility/physiology , Follicle Stimulating Hormone, beta Subunit/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/physiology , Gonadotropins/genetics , HEK293 Cells , Humans , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LHRH/genetics , Receptors, LHRH/physiology , Sexual Maturation , Signal Transduction/physiology
2.
Cell Tissue Res ; 369(3): 567-578, 2017 09.
Article En | MEDLINE | ID: mdl-28451751

The natriuretic peptides, Atrial-, B-type and C-type natriuretric peptides (ANP, BNP, CNP), are regulators of many endocrine tissues and exert their effects predominantly through the activation of their specific guanylyl cyclase receptors (GC-A and GC-B) to generate cGMP. Whereas cGMP-independent signalling has been reported in response to natriuretic peptides, this is mediated via either the clearance receptor (Npr-C) or a renal-specific NPR-Bi isoform, which both lack intrinsic guanylyl cyclase activity. Here, we report evidence of GC-B-dependent cGMP-independent signalling in pituitary GH3 cells. Stimulation of GH3 cells with CNP resulted in a rapid and sustained enhancement of ERK1/2 phosphorylation (P-ERK1/2), an effect that was not mimicked by dibutryl-cGMP. Furthermore, CNP-stimulated P-ERK1/2 occurred at concentrations below that required for cGMP accumulation. The effect of CNP on P-ERK1/2 was sensitive to pharmacological blockade of MEK (U0126) and Src kinases (PP2). Silencing of the GC-B1 and GC-B2 splice variants of the GC-B receptor by using targeted short interfering RNAs completely blocked the CNP effects on P-ERK1/2. CNP failed to alter GH3 cell proliferation or cell cycle distribution but caused a concentration-dependent increase in the activity of the human glycoprotein α-subunit promoter (αGSU) in a MEK-dependent manner. Finally, CNP also activated the p38 and JNK MAPK pathways in GH3 cells. These findings reveal an additional mechanism of GC-B signalling and suggest additional biological roles for CNP in its target tissues.


Guanylate Cyclase/metabolism , MAP Kinase Signaling System/drug effects , Natriuretic Peptide, C-Type/pharmacology , Somatotrophs/metabolism , Animals , Cell Line , Cyclic GMP/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Receptors, Guanylate Cyclase-Coupled/metabolism , Somatotrophs/drug effects
3.
Mol Endocrinol ; 30(3): 348-60, 2016 Mar.
Article En | MEDLINE | ID: mdl-26835742

The pulsatile release of GnRH regulates the synthesis and secretion of pituitary FSH and LH. Two transcription factors, cAMP-response element-binding protein (CREB) and inducible cAMP early repressor (ICER), have been implicated in the regulation of rat Fshb gene expression. We previously showed that the protein kinase A pathway mediates GnRH-stimulated CREB activation. We hypothesized that CREB and ICER are activated by distinct signaling pathways in response to pulsatile GnRH to modulate Fshb gene expression, which is preferentially stimulated at low vs high pulse frequencies. In the LßT2 gonadotrope-derived cell line, GnRH stimulation increased ICER mRNA and protein. Blockade of ERK activation with mitogen-activated protein kinase kinase I/II (MEKI/II) inhibitors significantly attenuated GnRH induction of ICER mRNA and protein, whereas protein kinase C, calcium/calmodulin-dependent protein kinase II, and protein kinase A inhibitors had minimal effects. GnRH also stimulated ICER in primary mouse pituitary cultures, attenuated similarly by a MEKI/II inhibitor. In a perifusion paradigm, MEKI/II inhibition in LßT2 cells stimulated with pulsatile GnRH abrogated ICER induction at high GnRH pulse frequencies, with minimal effect at low frequencies. MEKI/II inhibition reduced GnRH stimulation of Fshb at high and low pulse frequencies, suggesting that the ERK pathway has additional effects on GnRH regulation of Fshb, beyond those mediated by ICER. Indeed, induction of the activating protein 1 proteins, cFos and cJun, positive modulators of Fshb transcription, by pulsatile GnRH was also abrogated by inhibition of the MEK/ERK signaling pathway. Collectively, these studies indicate that the signaling pathways mediating GnRH activation of CREB and ICER are distinct, contributing to the decoding of the pulsatile GnRH to regulate FSHß expression.


Cyclic AMP Response Element Modulator/genetics , Follicle Stimulating Hormone, beta Subunit/genetics , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element Modulator/metabolism , Follicle Stimulating Hormone, beta Subunit/metabolism , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Male , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Pituitary Gland/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism
4.
Endocrinology ; 156(6): 2313-22, 2015 Jun.
Article En | MEDLINE | ID: mdl-25856429

Puberty is a tightly regulated process that leads to reproductive capacity. Kiss1 neurons are crucial in this process by stimulating GnRH, yet how Kiss1 neurons are regulated remains unknown. Substance P (SP), an important neuropeptide in pain perception, induces gonadotropin release in adult mice in a kisspeptin-dependent manner. Here, we assessed whether SP, through binding to its receptor NK1R (neurokinin 1 receptor), participates in the timing of puberty onset and fertility in the mouse. We observed that 1) selective NK1R agonists induce gonadotropin release in prepubertal females; 2) the expression of Tac1 (encoding SP) and Tacr1 (NK1R) in the arcuate nucleus is maximal before puberty, suggesting increased SP tone; 3) repeated exposure to NK1R agonists prepubertally advances puberty onset; and 4) female Tac1(-/-) mice display delayed puberty; moreover, 5) SP deficiency leads to subfertility in females, showing fewer corpora lutea and antral follicles and leading to decreased litter size. Thus, our findings support a role for SP in the stimulation of gonadotropins before puberty, acting via Kiss1 neurons to stimulate GnRH release, and its involvement in the attainment of full reproductive capabilities in female mice.


Fertility/drug effects , Sexual Maturation/drug effects , Substance P/metabolism , Animals , Female , Fertility/genetics , Gonadotropins/metabolism , Mice , Peptide Fragments/pharmacology , Puberty/drug effects , Puberty/genetics , Receptors, Neurokinin-1/agonists , Receptors, Neurokinin-1/metabolism , Sexual Maturation/genetics , Substance P/analogs & derivatives , Substance P/genetics , Substance P/pharmacology
5.
Mol Cell Endocrinol ; 385(1-2): 28-35, 2014 Mar 25.
Article En | MEDLINE | ID: mdl-24056171

The pituitary gonadotropin hormones, FSH and LH, are essential for fertility. Containing an identical α-subunit (CGA), they are comprised of unique ß-subunits, FSHß and LHß, respectively. These two hormones are regulated by the hypothalamic decapeptide, GnRH, which is released in a pulsatile manner from GnRH neurons located in the hypothalamus. Varying frequencies of pulsatile GnRH stimulate distinct signaling pathways and transcriptional machinery after binding to the receptor, GnRHR, on the cell surface of anterior pituitary gonadotropes. This ligand-receptor binding and activation orchestrates the synthesis and release of FSH and LH, in synergy with other effectors of gonadotropin production, such as activin, inhibin and steroids. Current research efforts aim to discover the mechanisms responsible for the decoding of the GnRH pulse signal by the gonadotrope. Modulating the response to GnRH has the potential to lead to new therapies for patients with altered gonadotropin secretion, such as those with hypothalamic amenorrhea or polycystic ovarian syndrome.


Amenorrhea/metabolism , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Luteinizing Hormone/metabolism , Polycystic Ovary Syndrome/metabolism , Amenorrhea/pathology , Amenorrhea/therapy , Animals , Female , Gene Expression Regulation , Humans , Hypothalamus/pathology , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/therapy
6.
Cell Tissue Res ; 355(2): 425-36, 2014 Feb.
Article En | MEDLINE | ID: mdl-24352806

The guanylyl cyclases, GC-A and GC-B, are selective receptors for atrial and C-type natriuretic peptides (ANP and CNP, respectively). In the anterior pituitary, CNP and GC-B are major regulators of cGMP production in gonadotropes and yet mouse models of disrupted CNP and GC-B indicate a potential role in growth hormone secretion. In the current study, we investigate the molecular and pharmacological properties of the CNP/GC-B system in somatotrope lineage cells. Primary rat pituitary and GH3 somatolactotropes expressed functional GC-A and GC-B receptors that had similar EC50 properties in terms of cGMP production. Interestingly, GC-B signaling underwent rapid homologous desensitization in a protein phosphatase 2A (PP2A)-dependent manner. Chronic exposure to either CNP or ANP caused a significant down-regulation of both GC-A- and GC-B-dependent cGMP accumulation in a ligand-specific manner. However, this down-regulation was not accompanied by alterations in the sub-cellular localization of these receptors. Heterologous desensitization of GC-B signaling occurred in GH3 cells following exposure to either sphingosine-1-phosphate or thyrotrophin-releasing hormone (TRH). This heterologous desensitization was protein kinase C (PKC)-dependent, as pre-treatment with GF109203X prevented the effect of TRH on CNP/GC-B signaling. Collectively, these data indicate common and distinct properties of particulate guanylyl cyclase receptors in somatotropes and reveal that independent mechanisms of homologous and heterologous desensitization occur involving either PP2A or PKC. Guanylyl cyclase receptors thus represent potential novel therapeutic targets for treating growth-hormone-associated disorders.


Lactotrophs/enzymology , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction , Animals , Atrial Natriuretic Factor/pharmacology , Calcium Signaling/drug effects , Cell Line , Cyclic AMP/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Endocytosis/drug effects , Lactotrophs/drug effects , Ligands , Mice , Natriuretic Peptide, C-Type/pharmacology , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Sphingolipids/metabolism , Thyrotropin-Releasing Hormone/metabolism
7.
N Engl J Med ; 368(26): 2467-75, 2013 Jun 27.
Article En | MEDLINE | ID: mdl-23738509

BACKGROUND: The onset of puberty is first detected as an increase in pulsatile secretion of gonadotropin-releasing hormone (GnRH). Early activation of the hypothalamic-pituitary-gonadal axis results in central precocious puberty. The timing of pubertal development is driven in part by genetic factors, but only a few, rare molecular defects associated with central precocious puberty have been identified. METHODS: We performed whole-exome sequencing in 40 members of 15 families with central precocious puberty. Candidate variants were confirmed with Sanger sequencing. We also performed quantitative real-time polymerase-chain-reaction assays to determine levels of messenger RNA (mRNA) in the hypothalami of mice at different ages. RESULTS: We identified four novel heterozygous mutations in MKRN3, the gene encoding makorin RING-finger protein 3, in 5 of the 15 families; both sexes were affected. The mutations included three frameshift mutations, predicted to encode truncated proteins, and one missense mutation, predicted to disrupt protein function. MKRN3 is a paternally expressed, imprinted gene located in the Prader-Willi syndrome critical region (chromosome 15q11-q13). All affected persons inherited the mutations from their fathers, a finding that indicates perfect segregation with the mode of inheritance expected for an imprinted gene. Levels of Mkrn3 mRNA were high in the arcuate nucleus of prepubertal mice, decreased immediately before puberty, and remained low after puberty. CONCLUSIONS: Deficiency of MKRN3 causes central precocious puberty in humans. (Funded by the National Institutes of Health and others.).


Frameshift Mutation , Mutation, Missense , Puberty, Precocious/genetics , Ribonucleoproteins/genetics , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Child , Child, Preschool , Exome , Female , Genetic Association Studies , Heterozygote , Humans , Hypothalamus/metabolism , Male , Mice , Pedigree , RNA, Messenger/analysis , Ribonucleoproteins/deficiency , Sequence Analysis, DNA , Ubiquitin-Protein Ligases
8.
Mol Endocrinol ; 27(4): 606-18, 2013 Apr.
Article En | MEDLINE | ID: mdl-23393127

Expression of pituitary FSH and LH, under the control of pulsatile GnRH, is essential for fertility. cAMP response element-binding protein (CREB) has been implicated in the regulation of FSHß gene expression, but the molecular mechanisms by which pulsatile GnRH regulates CREB activation remain poorly understood. We hypothesized that CREB is activated by a distinct signaling pathway in response to pulsatile GnRH in a frequency-dependent manner to dictate the FSHß transcriptional response. GnRH stimulation of CREB phosphorylation (pCREB) in the gonadotrope-derived LßT2 cell line was attenuated by a protein kinase A (PKA) inhibitor, H89. A dominant negative PKA (DNPKA) reduced GnRH-stimulated pCREB and markedly decreased GnRH stimulation of FSHß mRNA and FSHßLUC activity, but had little effect on LHßLUC activity, indicating relative specificity of this pathway. In perifusion studies, FSHß mRNA levels and FSHßLUC activities were increased by pulsatile GnRH, with significantly greater increases at low compared with high pulse frequencies. DNPKA markedly reduced these GnRH-stimulated FSHß responses at both low and high pulse frequencies. Correlating with FSHß activation, both PKA activity and levels of pCREB were increased to a greater extent by low compared with high GnRH pulse frequencies, and the induction of pCREB was also attenuated by overexpression of DNPKA at both low and high pulse frequencies. Taken together, these data indicate that a PKA-mediated signaling pathway mediates GnRH activation of CREB at low-pulse frequencies, playing a significant role in the decoding of the hypothalamic GnRH signal to result in frequency-dependent FSHß activation.


Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotropin-Releasing Hormone/pharmacology , Transcription, Genetic/drug effects , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, Dominant , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Endocr Relat Cancer ; 19(4): 497-508, 2012 Aug.
Article En | MEDLINE | ID: mdl-22645228

C-type natriuretic peptide (CNP/Nppc) is expressed at high levels in the anterior pituitary of rats and mice and activates guanylyl cyclase B receptors (GC-B/Npr2) to regulate hormone secretion. Mutations in NPR2/Npr2 can cause achondroplasia, GH deficiency, and female infertility, yet the normal expression profile within the anterior pituitary remains to be established in humans. The current study examined the expression profile and transcriptional regulation of NPR2 and GC-B protein in normal human fetal pituitaries, normal adult pituitaries, and human pituitary adenomas using RT-PCR and immunohistochemistry. Transcriptional regulation of human NPR2 promoter constructs was characterized in anterior pituitary cell lines of gonadotroph, somatolactotroph, and corticotroph origin. NPR2 was detected in all human fetal and adult pituitary samples regardless of age or sex, as well as in all adenoma samples examined regardless of tumor origin. GC-B immunoreactivity was variable in normal pituitary, gonadotrophinomas, and somatotrophinomas. Maximal transcriptional regulation of the NPR2 promoter mapped to a region within -214 bp upstream of the start site in all anterior pituitary cell lines examined. Electrophoretic mobility shift assays revealed that this region contains Sp1/Sp3 response elements. These data are the first to show NPR2 expression in normal human fetal and adult pituitaries and adenomatous pituitary tissue and suggest a role for these receptors in both pituitary development and oncogenesis, introducing a new target to manipulate these processes in pituitary adenomas.


Adenoma/genetics , Fetus/metabolism , Natriuretic Peptide, C-Type/physiology , Pituitary Gland/metabolism , Pituitary Neoplasms/genetics , Receptors, Atrial Natriuretic Factor/genetics , Adenoma/metabolism , Adult , Aged , Animals , Cells, Cultured , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Natriuretic Peptide, C-Type/genetics , Natriuretic Peptide, C-Type/metabolism , Pituitary Gland/embryology , Pituitary Gland/pathology , Pituitary Neoplasms/metabolism , Pregnancy , Rats , Receptors, Atrial Natriuretic Factor/metabolism
10.
J Endocrinol ; 203(2): 215-29, 2009 Nov.
Article En | MEDLINE | ID: mdl-19666697

In the pituitary, C-type natriuretic peptide (CNP) has been implicated as a gonadotroph-specific factor, yet expression of the CNP gene (Nppc) and CNP activity in gonadotrophs is poorly defined. Here, we examine the molecular expression and putative function of a local gonadotroph natriuretic peptide system. Nppc, along with all three natriuretic peptide receptors (Npr1, Npr2 and Npr3), was expressed in both alphaT3-1 and LbetaT2 cells and primary mouse pituitary tissue, yet the genes for atrial-(ANP) and B-type natriuretic peptides (Nppa and Nppb) were much less abundant. Putative processing enzymes of CNP were also expressed in alphaT3-1 cells and primary mouse pituitaries. Transcriptional analyses revealed that the proximal 50 bp of the murine Nppc promoter were sufficient for GNRH responsiveness, in an apparent protein kinase C and calcium-dependent manner. Electrophoretic mobility shift assays showed Sp1/Sp3 proteins form major complexes within this region of the Nppc promoter. CNP protein was detectable in rat anterior pituitaries, and electron microscopy detected CNP immunoreactivity in secretory granules of gonadotroph cells. Pharmacological analyses of natriuretic peptide receptor activity clearly showed ANP and CNP are potent activators of cGMP production. However, functional studies failed to reveal a role for CNP in regulating cell proliferation or LH secretion. Surprisingly, CNP potently stimulated the human glycoprotein hormone alpha-subunit promoter in LbetaT2 cells but not in alphaT3-1 cells. Collectively, these findings support a role for CNP as the major natriuretic peptide of the anterior pituitary, and for gonadotroph cells as the major source of CNP expression and site of action.


Gonadotrophs/metabolism , Natriuretic Peptide, C-Type/physiology , Pituitary Gland/metabolism , Animals , Cells, Cultured , Cyclic GMP/metabolism , Female , Gene Expression Regulation , Glycoprotein Hormones, alpha Subunit/genetics , Luteinizing Hormone/metabolism , Male , Mice , Natriuretic Peptide, C-Type/analysis , Natriuretic Peptide, C-Type/genetics , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Atrial Natriuretic Factor/analysis , Receptors, Atrial Natriuretic Factor/genetics
...