Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Mol Sci ; 24(21)2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37958948

ABSTRACT

Human sexual and reproductive development is regulated by the hypothalamic-pituitary-gonadal (HPG) axis, which is primarily controlled by the gonadotropin-releasing hormone (GnRH) acting on its receptor (GnRHR). Dysregulation of the axis leads to conditions such as congenital hypogonadotropic hypogonadism (CHH) and delayed puberty. The pathophysiology of GnRHR makes it a potential target for treatments in several reproductive diseases and in congenital adrenal hyperplasia. GnRHR belongs to the G protein-coupled receptor family and its GnRH ligand, when bound, activates several complex and tissue-specific signaling pathways. In the pituitary gonadotrope cells, it triggers the G protein subunit dissociation and initiates a cascade of events that lead to the production and secretion of the luteinizing hormone (LH) and follicle-stimulating hormone (FSH) accompanied with the phospholipase C, inositol phosphate production, and protein kinase C activation. Pharmacologically, GnRHR can be modulated by synthetic analogues. Such analogues include the agonists, antagonists, and the pharmacoperones. The agonists stimulate the gonadotropin release and lead to receptor desensitization with prolonged use while the antagonists directly block the GnRHR and rapidly reduce the sex hormone production. Pharmacoperones include the most recent GnRHR therapeutic approaches that directly correct the misfolded GnRHRs, which are caused by genetic mutations and hold serious promise for CHH treatment. Understanding of the GnRHR's genomic and protein structure is crucial for the most appropriate assessing of the mutation impact. Such mutations in the GNRHR are linked to normosmic hypogonadotropic hypogonadism and lead to various clinical symptoms, including delayed puberty, infertility, and impaired sexual development. These mutations vary regarding their mode of inheritance and can be found in the homozygous, compound heterozygous, or in the digenic state. GnRHR expression extends beyond the pituitary gland, and is found in reproductive tissues such as ovaries, uterus, and prostate and non-reproductive tissues such as heart, muscles, liver and melanoma cells. This comprehensive review explores GnRHR's multifaceted role in human reproduction and its clinical implications for reproductive disorders.


Subject(s)
Hypogonadism , Klinefelter Syndrome , Puberty, Delayed , Female , Male , Humans , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Hypogonadism/drug therapy , Hypogonadism/genetics , Hypogonadism/metabolism , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Follicle Stimulating Hormone
2.
Cell Rep ; 42(6): 112543, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224016

ABSTRACT

Gonadotropes in the anterior pituitary gland are essential for fertility and provide a functional link between the brain and the gonads. To trigger ovulation, gonadotrope cells release massive amounts of luteinizing hormone (LH). The mechanism underlying this remains unclear. Here, we utilize a mouse model expressing a genetically encoded Ca2+ indicator exclusively in gonadotropes to dissect this mechanism in intact pituitaries. We demonstrate that female gonadotropes exclusively exhibit a state of hyperexcitability during the LH surge, resulting in spontaneous [Ca2+]i transients in these cells, which persist in the absence of any in vivo hormonal signals. L-type Ca2+ channels and transient receptor potential channel A1 (TRPA1) together with intracellular reactive oxygen species (ROS) levels ensure this state of hyperexcitability. Consistent with this, virus-assisted triple knockout of Trpa1 and L-type Ca2+ subunits in gonadotropes leads to vaginal closure in cycling females. Our data provide insight into molecular mechanisms required for ovulation and reproductive success in mammals.


Subject(s)
Gonadotrophs , Pituitary Gland, Anterior , Mice , Animals , Female , Luteinizing Hormone , Pituitary Gland , Ovulation , Mammals
3.
Biochem Pharmacol ; 212: 115553, 2023 06.
Article in English | MEDLINE | ID: mdl-37075816

ABSTRACT

Gonadotropin-Releasing Hormone (GnRH) is a decapeptide responsible for the control of the reproductive functions. It shows C- and N-terminal aminoacid modifications and two other distinct isoforms have been so far identified. The biological effects of GnRH are mediated by binding to high-affinity G-protein couple receptors (GnRHR), showing characteristic very short C tail. In mammals, including humans, GnRH-producing neurons originate in the embryonic nasal compartment and during early embryogenesis they undergo rapid migration towards the hypothalamus; the increasing knowledge of such mechanisms improved diagnostic and therapeutic approaches to infertility. The pharmacological use of GnRH, or its synthetic peptide and non-peptide agonists or antagonists, provides a valid tool for reproductive disorders and assisted reproduction technology (ART). The presence of GnRHR in several organs and tissues indicates additional functions of the peptide. The identification of a GnRH/GnRHR system in the human endometrium, ovary, and prostate has extended the functions of the peptide to the physiology and tumor transformation of such tissues. Likely, the activity of a GnRH/GnRHR system at the level of the hippocampus, as well as its decreased expression in mice brain aging, raised interest in its possible involvement in neurogenesis and neuronal functions. In conclusion, GnRH/GnRHR appears to be a fascinating biological system that exerts several possibly integrated pleiotropic actions in the complex control of reproductive functions, tumor growth, neurogenesis, and neuroprotection. This review aims to provide an overview of the physiology of GnRH and the pharmacological applications of its synthetic analogs in the management of reproductive and non-reproductive diseases.


Subject(s)
Gonadotropin-Releasing Hormone , Neoplasms , Male , Mice , Female , Animals , Humans , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Reproduction , Ovary/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Mammals/metabolism
4.
Endocrinology ; 164(4)2023 02 11.
Article in English | MEDLINE | ID: mdl-36791137

ABSTRACT

The 2 pituitary gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH), regulate the reproductive function in all vertebrates. While many studies have investigated the regulation of gonadotropin production and release by sex steroid feedback, its role on the regulation of gonadotrope cell number remains unclear. Using medaka as a model and an optimized protocol to restore physiological sex steroids levels following gonadectomy, we show that gonadal sex steroids not only decrease fshb transcript levels, but also Fsh cell number in both sexes. We then investigated the origin of Fsh cell hyperplasia induced by gonadectomy. In both sexes, bromodeoxyuridine incubation shows that this is achieved via Fsh cell mitosis. In situ hybridization reveals that new Fsh cells also originate from transdifferentiating Tsh cells in females, but not in males. Both phenomena are inhibited by sex steroid supplementation via feeding. In males (but not females), gonadectomy (without recovery with sex steroid supplementation) also reduces sox2 transcript levels and Sox2-immunopositive population size, suggesting that Sox2 progenitors may be recruited to produce new Fsh cells. Opposite to Fsh cells, gonadectomy decreases lhb levels in both sexes, and levels are not restored by sex steroid supplementation. In addition, the regulation of Lh cell number also seems to be sex dependent. Removal of gonadal sex steroids stimulates Lh cell mitosis in male (like Fsh cells) but not in females. To conclude, our study provides the first evidence on sexually dimorphic mechanisms used in the fish pituitary to remodel gonadotrope populations in response to sex steroids.


Subject(s)
Gonadotrophs , Oryzias , Female , Animals , Male , Cell Transdifferentiation , Hyperplasia , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/genetics , Pituitary Gland , Gonadal Steroid Hormones/pharmacology , Steroids , Mitosis
6.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34864945

ABSTRACT

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.


Subject(s)
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
7.
Endocrinology ; 161(7)2020 07 01.
Article in English | MEDLINE | ID: mdl-32270195

ABSTRACT

Activins are selective regulators of FSH production by pituitary gonadotrope cells. In a gonadotrope-like cell line, LßT2, activins stimulate FSH via the activin type IIA receptor (ACVR2A) and/or bone morphogenetic protein type II receptor (BMPR2). Consistent with these observations, FSH is greatly reduced, though still present, in global Acvr2a knockout mice. In contrast, FSH production is unaltered in gonadotrope-specific Bmpr2 knockout mice. In light of these results, we questioned whether an additional type II receptor might mediate the actions of activins or related TGF-ß ligands in gonadotropes. We focused on the activin type IIB receptor (ACVR2B), even though it does not mediate activin actions in LßT2 cells. Using a Cre-lox strategy, we ablated Acvr2a and/or Acvr2b in murine gonadotropes. The resulting conditional knockout (cKO) animals were compared with littermate controls. Acvr2a cKO (cKO-A) females were subfertile (~70% reduced litter size), cKO-A males were hypogonadal, and both sexes showed marked decreases in serum FSH levels compared with controls. Acvr2b cKO (cKO-B) females were subfertile (~20% reduced litter size), cKO-B males had a moderate decrease in testicular weight, but only males showed a significant decrease in serum FSH levels relative to controls. Simultaneous deletion of both Acvr2a and Acvr2b in gonadotropes led to profound hypogonadism and FSH deficiency in both sexes; females were acyclic and sterile. Collectively, these data demonstrate that ACVR2A and ACVR2B are the critical type II receptors through which activins or related TGF-ß ligands induce FSH production in mice in vivo.


Subject(s)
Activin Receptors, Type II/metabolism , Follicle Stimulating Hormone/biosynthesis , Activin Receptors, Type II/genetics , Animals , Female , Hypogonadism/genetics , Male , Mice , Mice, Knockout , Sex Characteristics
8.
J Endocrinol ; 244(1): 111-122, 2020 01.
Article in English | MEDLINE | ID: mdl-31585440

ABSTRACT

The progesterone receptor (PR, encoded by Pgr) plays essential roles in reproduction. Female mice lacking the PR are infertile, due to the loss of the protein's functions in the brain, ovary, and uterus. PR is also expressed in pituitary gonadotrope cells, but its specific role therein has not been assessed in vivo. We therefore generated gonadotrope-specific Pgr conditional knockout mice (cKO) using the Cre-LoxP system. Overall, both female and male cKO mice appeared phenotypically normal. cKO females displayed regular estrous cycles (vaginal cytology) and normal fertility (litter size and frequency). Reproductive organ weights were comparable between wild-type and cKO mice of both sexes, as were production and secretion of the gonadotropins, LH and FSH, with one exception. On the afternoon of proestrus, the amplitude of the LH surge was blunted in cKO females relative to controls. Contrary to predictions of earlier models, this did not appear to derive from impaired GnRH self-priming. Collectively, these data indicate that PR function in gonadotropes may be limited to regulation of LH surge amplitude in female mice via a currently unknown mechanism.


Subject(s)
Estrous Cycle/genetics , Gonadotrophs/metabolism , Luteinizing Hormone/metabolism , Receptors, Progesterone/deficiency , Animals , Female , Follicle Stimulating Hormone , Gonadotropins/metabolism , Male , Mice , Mice, Knockout
9.
Biol Reprod ; 101(4): 791-799, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31290547

ABSTRACT

The immortalized mouse gonadotrope cell lines alphaT3-1 and LbetaT2 cells have been a substitute model for primary gonadotropes. These cell lines have provided a homogeneous cell population, as compared to the dissociated anterior pituitaries, which contain a heterogeneous population of cells potentially responsive to estradiol-17beta (E2). Nonclassical actions of E2 assumed to occur through the plasma membrane estrogen receptor 1 (ESR1, also known as ERalpha). These actions have included inhibition of gonadotropin-releasing hormone (GnRH)-induced increases in intracellular calcium concentrations and phosphorylation of p44/42 mitogen-activated protein kinase (ERK-1/2) in ovine pituitaries including primary gonadotropes in vitro. The objective of the present experiment was to determine if alphaT3-1 and LbetaT2 are cell models with limitations to examine the nonclassical actions of E2 occurring in gonadotropes. Experiments were conducted to determine if the cells have ESR1 at the plasma membrane using biotinylation cell and isolation of surface protein and staining with a fluorescently labeled E2 conjugate. The alphaT3-1 cells contain ESR1 associated with but not enriched within lipid rafts of the plasma membrane and do not translocate to lipid rafts upon binding of E2. In contrast, LbetaT2 cells lack ESR1 associated with the plasma membrane. Pretreatment with E2 did not cause inhibition of GnRH-stimulated increases in intracellular concentrations of calcium for either cell type. Phosphorylation of ERK-1/2 was not stimulated by E2 in either cell type. Although these cells lines have been used extensively to study GnRH signaling, in vitro or in vivo effects of nonclassical actions of E2 cannot be replicated in either cell line.


Subject(s)
Estradiol/pharmacology , Gonadotrophs/drug effects , Animals , Calcium Signaling/drug effects , Cell Line, Transformed , Gonadotrophs/cytology , MAP Kinase Signaling System/drug effects , Mice , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
10.
Article in English | MEDLINE | ID: mdl-30858828

ABSTRACT

Since gonadotropin-inhibitory hormone (GnIH) was discovered in 2000 as the first hypothalamic neuropeptide that actively inhibits gonadotropin release, researches conducted for the last 18 years have demonstrated that GnIH acts as a pronounced negative regulator of reproduction. Inhibitory effect of GnIH on reproduction is mainly accomplished at hypothalamic-pituitary levels; gonadotropin-releasing hormone (GnRH) neurons and gonadotropes are major targets of GnIH action based on the morphological interaction with GnIH neuronal fibers and the distribution of GnIH receptor. Here, we review molecular studies mainly focusing on the signal transduction pathway of GnIH in target cells, GnRH neurons, and gonadotropes. The use of well-defined cellular model systems allows the mechanistic study of signaling pathway occurring in target cells by demonstrating the direct cause-and-effect relationship. The insights gained through studying molecular mechanism of GnIH action contribute to deeper understanding of the mechanism of how GnIH communicates with other neuronal signaling systems to control our reproductive function. Reproductive axis closely interacts with other endocrine systems, thus GnIH expression levels would be changed by adrenal and thyroid status. We also briefly review molecular studies investigating the regulatory mechanisms of GnIH expression to understand the role of GnIH as a mediator between adrenal, thyroid and gonadal axes.

11.
J Mol Endocrinol ; 62(2): 67-78, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30481159

ABSTRACT

Fertility is dependent on follicle-stimulating hormone (FSH), a product of gonadotrope cells of the anterior pituitary gland. Hypothalamic gonadotropin-releasing hormone (GnRH) and intra-pituitary activins are regarded as the primary drivers of FSH synthesis and secretion. Both stimulate expression of the FSH beta subunit gene (Fshb), although the underlying mechanisms of GnRH action are poorly described relative to those of the activins. There is currently no consensus on how GnRH regulates Fshb transcription, as results vary across species and between in vivo and in vitro approaches. One of the more fully developed models suggests that the murine Fshb promoter is tonically repressed by histone deacetylases (HDACs) and that GnRH relieves this repression, at least in immortalized murine gonadotrope-like cells (LßT2 and αT3-1). In contrast, we observed that the class I/II HDAC inhibitor trichostatin A (TSA) robustly inhibited basal, activin A-, and GnRH-induced Fshb mRNA expression in LßT2 cells and in primary murine pituitary cultures. Similar results were obtained with the class I specific HDAC inhibitor, entinostat, whereas two class II-specific inhibitors, MC1568 and TMP269, had no effects on Fshb expression. Collectively, these data suggest that class I HDACs are positive, not negative, regulators of Fshb expression in vitro and that, contrary to earlier reports, GnRH may not stimulate Fshb by inhibiting HDAC-mediated repression of the gene.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/metabolism , Gonadotrophs/metabolism , Histone Deacetylase Inhibitors/pharmacology , 17-Hydroxysteroid Dehydrogenases/metabolism , Activins/metabolism , Animals , Cell Line , Cells, Cultured , Forkhead Box Protein L2/metabolism , Gonadotrophs/drug effects , Hydroxamic Acids/pharmacology , Mice , Signal Transduction/drug effects , Smad Proteins/metabolism
12.
Front Mol Neurosci ; 11: 305, 2018.
Article in English | MEDLINE | ID: mdl-30214395

ABSTRACT

Based on pharmacological studies, corticotropin-releasing hormone (CRH) and its receptors play a leading role in the inhibition of the hypothalamic-pituitary-gonadal (HPG) axis during acute stress. To further study the effects of CRH receptor signaling on the HPG axis, we generated and/or employed male mice lacking CRH receptor type 1 (CRHR1) or type 2 (CRHR2) in gonadotropin-releasing hormone neurons, GABAergic neurons, or in all central neurons and glia. The deletion of CRHRs revealed a preserved decrease of plasma luteinizing hormone (LH) in response to either psychophysical or immunological stress. However, under basal conditions, central infusion of CRH into mice lacking CRHR1 in all central neurons and glia, or application of CRH to pituitary cultures from mice lacking CRHR2, failed to suppress LH release, unlike in controls. Our results, taken together with those of the earlier pharmacological studies, suggest that inhibition of the male HPG axis during acute stress is mediated by other factors along with CRH, and that CRH suppresses the HPG axis at the central and pituitary levels via CRHR1 and CRHR2, respectively.

14.
Mol Cell Endocrinol ; 463: 97-105, 2018 03 05.
Article in English | MEDLINE | ID: mdl-28392410

ABSTRACT

The role of protein kinase C (PKC) isoforms (PKCs) in GnRH-stimulated MAPK [ERK1/2, JNK1/2 and p38) phosphorylation was examined in gonadotrope derived cells. GnRH induced a protracted activation of ERK1/2 and a slower and more transient activation of JNK1/2 and p38MAPK. Gonadotropes express conventional PKCα and PKCßII, novel PKCδ, PKCε and PKCθ, and atypical PKC-ι/λ. The use of green fluorescent protein (GFP)-PKCs constructs revealed that GnRH induced rapid translocation of PKCα and PKCßII to the plasma membrane, followed by their redistribution to the cytosol. PKCδ and PKCε localized to the cytoplasm and Golgi, followed by the rapid redistribution by GnRH of PKCδ to the perinuclear zone and of PKCε to the plasma membrane. The use of dominant negatives for PKCs and peptide inhibitors for the receptors for activated C kinase (RACKs) has revealed differential role for PKCα, PKCßII, PKCδ and PKCε in ERK1/2, JNK1/2 and p38MAPK phosphorylation in a ligand-and cell context-dependent manner. The paradoxical findings that PKCs activated by GnRH and PMA play a differential role in MAPKs phosphorylation may be explained by persistent vs. transient redistribution of selected PKCs or redistribution of a given PKC to the perinuclear zone vs. the plasma membrane. Thus, we have identified the PKCs involved in GnRH stimulated MAPKs phosphorylation in gonadotrope derived cells. Once activated, the MAPKs will mediate the transcription of the gonadotropin subunits and GnRH receptor genes.


Subject(s)
Gonadotrophs/cytology , Gonadotrophs/enzymology , Gonadotropin-Releasing Hormone/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Animals , Enzyme Activation/drug effects , Humans , Isoenzymes/metabolism , Mice , Phosphorylation/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
15.
Biochem Biophys Res Commun ; 491(1): 53-58, 2017 09 09.
Article in English | MEDLINE | ID: mdl-28705737

ABSTRACT

Leptin, a peptide hormone produced by adipocytes, is recognized as one of the signals involved in the onset of reproductive activity. The leptin receptor has been found in hypothalamic neurons and pituitary gonadotropes, suggesting that the hormone may act at both sites to stimulate the secretion of GnRH and consequently, FSH and LH. In response to a stimulus such as a hypothalamic secretagogue, gonadotropes respond with changes in electrical activity, intracellular Ca2+ and hormone release. The main aim of this report was to investigate whether leptin promotes a change in the electrical and secretory activities of bovine gonadotropes. After 48 h of treatment with leptin (10 nM) significant changes in the action potential properties were observed in gonadotropes, which included an increase in amplitude, time-to-pike and post-hyperpolarization, as well as a decrease in firing threshold. Likewise, leptin induced a significant (∼1.3-fold) up-regulation of voltage-gated Na+ channel current density, and a selective increase (∼2.1-fold) in Ca2+ current density through high voltage-activated channels. Consistent with this, leptin enhanced GnRH-induced secretion of LH measured by ELISA. We suggest that leptin enhances membrane expression of voltage-gated Na+ and Ca2+ channels, which results in a modulation of the action potential properties and an increase in hormone release from gonadotropes.


Subject(s)
Action Potentials/physiology , Endocrine Cells/physiology , Gonadotropin-Releasing Hormone/metabolism , Leptin/metabolism , Luteinizing Hormone/metabolism , Membrane Potentials/physiology , Animals , Cattle , Cells, Cultured , Male
16.
Article in English | MEDLINE | ID: mdl-28626446

ABSTRACT

We have previously described a signaling complex (signalosome) associated with the GnRH receptor (GnRHR). We now report that GnRH induces bleb formation in the gonadotrope-derived LßT2 cells. The blebs appear within ~2 min at a turnover rate of ~2-3 blebs/min and last for at least 90 min. Formation of the blebs requires active ERK1/2 and RhoA-ROCK but not active c-Src. Although the following ligands stimulate ERK1/2 in LßT2 cells: EGF > GnRH > PMA > cyclic adenosine monophosphate (cAMP), they produced little or no effect on bleb formation as compared to the robust effect of GnRH (GnRH > PMA > cAMP > EGF), indicating that ERK1/2 is required but not sufficient for bleb formation possibly due to compartmentalization. Members of the above mentioned signalosome are recruited to the blebs, some during bleb formation (GnRHR, c-Src, ERK1/2, focal adhesion kinase, paxillin, and tubulin), and some during bleb retraction (vinculin), while F-actin decorates the blebs during retraction. Fluorescence intensity measurements for the above proteins across the cells showed higher intensity in the blebs vs. intracellular area. Moreover, GnRH induces blebs in primary cultures of rat pituitary cells and isolated mouse gonadotropes in an ERK1/2-dependent manner. The novel signalosome-bleb pathway suggests that as with the signalosome, the blebs are apparently involved in cell migration. Hence, we have extended the potential candidates which are involved in the blebs life cycle in general and for the GnRHR in particular.

17.
Cell Calcium ; 67: 138-147, 2017 11.
Article in English | MEDLINE | ID: mdl-28522036

ABSTRACT

Transient receptor potential (TRP) channels play important functional roles in the signal transduction machinery of hormone-secreting cells and have recently been implicated in reproductive physiology. While expression studies have demonstrated TRP channel expression at all levels of the hypothalamic-pituitary-gonadal (hpg) axis, functional details about TRP channel action at the level of the individual cells controlling reproduction are just beginning to emerge. Canonical TRP (TRPC) channels are prominently expressed in the reproductive center of the neuroendocrine brain, i.e. in kisspeptin and gonadotropin-releasing hormone (GnRH) neurons. Kisspeptin neurons are depolarized by leptin via activation of TRPC channels and kisspeptin depolarizes GnRH neurons through TRPC4 activation. Recent studies have functionally identified TRPC channels also in gonadotrope cells in the anterior pituitary gland, which secrete gonadotropins in response to GnRH and thus regulate gonadal function. TRP channel expression in these cells exhibits remarkable plasticity and depends on the hormonal status of the animal. Subsequent functional analyses have demonstrated that TRPC5 in gonadotropes contributes to depolarization of the plasma membrane upon GnRH stimulation and increases the intracellular Ca2+ concentration via its own Ca2+ permeability and via the activation of voltage-gated Ca2+ channels. However, conditional gene targeting experiments will be needed to unambiguously dissect the physiological role of TRPC channels in the different cell types of the reproductive axis in vivo.


Subject(s)
Calcium/metabolism , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/metabolism , Reproduction/genetics , TRPC Cation Channels/genetics , Animals , Gene Expression Regulation , Gonadotrophs/cytology , Gonadotropin-Releasing Hormone/genetics , Gonads/cytology , Gonads/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Leptin/genetics , Leptin/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , TRPC Cation Channels/metabolism
18.
J Endocrinol ; 233(2): 159-174, 2017 05.
Article in English | MEDLINE | ID: mdl-28330973

ABSTRACT

Kisspeptin and neurokinin B (NKB) are neuropeptides co-expressed in the mammalian hypothalamus and coordinately control GnRH signaling. We have found that Nkb and kisspeptin neurons are distinct in the teleost, striped bass (STB) and capitalized on this phenomenon to study the mode of action of Nkb and its related neuropeptide-F (Nkf), both of which are encoded by the tac3 gene. In vitro brain slices and in vivo administration studies revealed that Nkb/f consistently downregulated kiss2, whereas antagonist (AntD) administration restored this effect. Overall, a minor effect was noted on gnrh1 expression, whereas Gnrh1 content in the pituitaries was reduced after Nkb/f treatment and increased with AntD. Concomitantly, immunostaining demonstrated that hypothalamic Nkb neurons border and densely innervate the largest kiss2 neuronal population in the hypothalamus, which also coexpresses Nkb receptor. No expression of Nkb receptor or Nkb neuronal projections was detected near/in Gnrh1 soma in the preoptic area. At the level of the pituitary, however, the picture was more complex: both Nkb/f and AntD upregulated lhb and fshb expression and Lh secretion in vivo Together with the stimulatory effect of Nkb/f on Lh/Fsh secretion from pituitary cells, in vitro, this may indicate an additional independent action of Nkb/f within the pituitary, in which the hypothalamic pathway is more dominant. The current study demonstrates that Nkb/f utilizes multiple pathways to regulate reproduction in the STB and that in the brain, Nkb mainly acts as a negative modulator of kiss2 to regulate the release of Gnrh1.


Subject(s)
Bass/metabolism , Gene Expression Regulation/physiology , Kisspeptins/metabolism , Neurokinin B/physiology , Reproduction/physiology , Animals , Cloning, Molecular , DNA, Complementary/metabolism , Kisspeptins/antagonists & inhibitors , Kisspeptins/genetics , Male , Neurokinin B/genetics , Pituitary Gland/metabolism
19.
Mol Cell Endocrinol ; 439: 141-154, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27810601

ABSTRACT

We examined the role of PKCs and Ca2+ in GnRH-stimulated p38MAPK phosphorylation in the gonadotrope derived αT3-1 and LßT2 cell lines. GnRH induced a slow and rapid increase in p38MAPK phosphorylation in αT3-1 and LßT2 cells respectively, while PMA gave a slow response. The use of dominant negatives for PKCs and peptide inhibitors for the receptors for activated C kinase (RACKs), has revealed differential role for PKCα, PKCßII, PKCδ and PKCε in p38MAPK phosphorylation in a ligand-and cell context-dependent manner. The paradoxical findings that PKCs activated by GnRH and PMA play a differential role in p38MAPK phosphorylation may be explained by differential localization of the PKCs. Basal, GnRH- and PMA- stimulation of p38MAPK phosphorylation in αT3-1 cells is mediated by Ca2+ influx via voltage-gated Ca2+ channels and Ca2+ mobilization, while in the differentiated LßT2 gonadotrope cells it is mediated only by Ca2+ mobilization. p38MAPK resides in the cell membrane and is relocated to the nucleus by GnRH (∼5 min). Thus, we have identified the PKCs and the Ca2+ pools involved in GnRH stimulated p38MAPK phosphorylation.


Subject(s)
Calcium/metabolism , Gonadotrophs/drug effects , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Phorbol Esters/pharmacology , Protein Kinase C/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Calcimycin/pharmacology , Cell Line, Transformed , Ionomycin/pharmacology , Isoenzymes/metabolism , Models, Biological , Peptides/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Time Factors , src-Family Kinases/metabolism
20.
Neuroendocrinology ; 105(4): 357-371, 2017.
Article in English | MEDLINE | ID: mdl-27871073

ABSTRACT

BACKGROUND: Kisspeptins are important regulators of the development and function of the hypothalamic-pituitary-gonadal axis. However, the importance of kisspeptin at the pituitary level is unclear. METHODS: We examined the expression profile of kisspeptin in the mouse pituitary during development and in adulthood using RT-PCR, quantitative PCR and immunohistochemistry. RESULTS: Kiss1 mRNA was detected in both embryonic and postnatal pituitaries. Kisspeptin-immunoreactive (+) cells were detected from embryonic day (E) 13.5 throughout adulthood, being localized to the rostroventral portion in the anterior pituitary (AP) in embryos, and also to the dorsocaudal AP postnatally. A large proportion of kisspeptin+ cells were double-labeled with gonadotrope markers including Foxl2, SF-1, and LHß, and the percentage of LHß+ cells in kisspeptin+ cells increased during development. No kisspeptin+ cells were positive for the proliferating cell marker MCM7 (minichromosome maintenance protein 7), but a few kisspeptin+ cells co-expressed the stem/progenitor cell marker Sox2. Kisspeptin expression was similar between sexes and between agonadal SF-1 knockout embryos and wild-type littermates. Kiss1 mRNA levels were not significantly different between sexes or during early postnatal development, but levels in females increased when puberty began and were significantly higher than in males at postpubertal ages. CONCLUSIONS: These results suggest that kisspeptin is expressed in gonadotrope precursors during gonadotrope differentiation, and that kisspeptin expression begins soon after the initiation of αGSU production and is extinguished soon after the initiation of LH production. Furthermore, pituitary kisspeptin expression may be regulated in a gonad-independent manner during development, but may be associated with gonadotrope function in adulthood.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Gonadotrophs/metabolism , Hypothalamus , Kisspeptins/metabolism , Pituitary Gland , Age Factors , Animals , Animals, Newborn , Embryo, Mammalian , Female , Hypothalamus/embryology , Hypothalamus/growth & development , Hypothalamus/metabolism , Kisspeptins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Pituitary Gland/embryology , Pituitary Gland/growth & development , Pituitary Gland/metabolism , RNA, Messenger/metabolism , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Thyrotropin, beta Subunit/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL