Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Endocrinology ; 165(8)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38923438

RESUMO

The neuroendocrine marker genes Ptprn and Ptprn2 encode protein tyrosine phosphatase receptors N and N2, 2 members of protein tyrosine phosphatase receptors void of enzymatic activity, and whose function and mechanism of action have not been elucidated. To explore the role(s) of Ptprn and Ptprn2 on the hypothalamic-pituitary-adrenal axis, we used mice in which both genes were knocked out (DKO). The focus in this study was on corticotrophs and melanotrophs from the anterior and intermediate lobes of the pituitary gland, respectively. In both sexes, DKO caused an increase in the expression of the corticotroph/melanotroph genes Pomc and Tbx19 and the melanotroph-specific gene Pax7. We also found in vivo and in vitro increased synthesis and release of beta-endorphin, alpha-melanocyte-stimulating hormone, and ACTH in DKO mice, which was associated with increased serum corticosterone levels and adrenal mass. DKO also increased the expression of other melanotroph-specific genes, but not corticotroph-specific genes. The dopaminergic pathway in the hypothalamus and dopaminergic receptors in melanotrophs were not affected in DKO mice. However, hyperplasia of the intermediate lobe was observed in DKO females and males, accompanied by increased proopiomelanocortin immunoreactivity per cell. These results indicate that protein tyrosine phosphatase receptor type N contributes to hypothalamic-pituitary-adrenal function by being involved in processes governing postnatal melanotroph development and Pomc expression.


Assuntos
Melanotrofos , Camundongos Knockout , Hipófise , Pró-Opiomelanocortina , Animais , Camundongos , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/genética , Feminino , Masculino , Hipófise/metabolismo , Melanotrofos/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Camundongos Endogâmicos C57BL
2.
Front Cell Neurosci ; 18: 1354095, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38633445

RESUMO

Vasoactive intestinal peptide (VIP) is an important component of the suprachiasmatic nucleus (SCN) which relays circadian information to neuronal populations, including GnRH neurons. Human and animal studies have shown an impact of disrupted daily rhythms (chronic shift work, temporal food restriction, clock gene disruption) on both male and female reproduction and fertility. To date, how VIP modulates GnRH neurons remains unknown. Calcium imaging and electrophysiology on primary GnRH neurons in explants and adult mouse brain slice, respectively, were used to address this question. We found VIP excites GnRH neurons via the VIP receptor, VPAC2. The downstream signaling pathway uses both Gs protein/adenylyl cyclase/protein kinase A (PKA) and phospholipase C/phosphatidylinositol 4,5-bisphosphate (PIP2) depletion. Furthermore, we identified a UCL2077-sensitive target, likely contributing to the slow afterhyperpolarization current (IAHP), as the PKA and PIP2 depletion target, and the KCa3.1 channel as a specific target. Thus, VIP/VPAC2 provides an example of Gs protein-coupled receptor-triggered excitation in GnRH neurons, modulating GnRH neurons likely via the slow IAHP. The possible identification of KCa3.1 in the GnRH neuron slow IAHP may provide a new therapeutical target for fertility treatments.

4.
Endocrinology ; 164(12)2023 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-37935042

RESUMO

Postnatal development of functional pituitary gonadotrophs is necessary for maturation of the hypothalamic-pituitary-gonadal axis, puberty, and reproduction. Here we examined the role of PI4-kinase A, which catalyzes the biosynthesis of PI4P in mouse reproduction by knocking out this enzyme in cells expressing the gonadotropin-releasing hormone (GnRH) receptor. Knockout (KO) mice were infertile, reflecting underdeveloped gonads and reproductive tracts and lack of puberty. The number and distribution of hypothalamic GnRH neurons and Gnrh1 expression in postnatal KOs were not affected, whereas Kiss1/kisspeptin expression was increased. KO of PI4-kinase A also did not alter embryonic establishment and neonatal development and function of the gonadotroph population. However, during the postnatal period, there was a progressive loss of expression of gonadotroph-specific genes, including Fshb, Lhb, and Gnrhr, accompanied by low gonadotropin synthesis. The postnatal gonadotroph population also progressively declined, reaching approximately one-third of that observed in controls at 3 months of age. In these residual gonadotrophs, GnRH-dependent calcium signaling and calcium-dependent membrane potential changes were lost, but intracellular administration of inositol-14,5-trisphosphate rescued this signaling. These results indicate a key role for PI4-kinase A in the postnatal development and maintenance of a functional gonadotroph population.


Assuntos
Gonadotrofos , Doenças da Hipófise , Camundongos , Animais , Gonadotrofos/metabolismo , Camundongos Knockout , Maturidade Sexual , Hipófise/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Doenças da Hipófise/metabolismo
5.
Front Endocrinol (Lausanne) ; 14: 1147554, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36950690

RESUMO

In vertebrates, gonadotropin-releasing hormone (GnRH)-secreting neurons control fertility by regulating gonadotrophs in the anterior pituitary. While it is known that acetylcholine (ACh) influences GnRH secretion, whether the effect is direct or indirect, and the specific ACh receptor (AChR) subtype(s) involved remain unclear. Here, we determined 1) whether ACh can modulate GnRH cellular activity and 2) a source of ACh afferents contacting GnRH neurons. Calcium imaging was used to assay GnRH neuronal activity. With GABAergic and glutamatergic transmission blocked, subtype-specific AChR agonists and antagonists were applied to identify direct regulation of GnRH neurons. ACh and nicotine caused a rise in calcium that declined gradually back to baseline after 5-6 min. This response was mimicked by an alpha3-specific agonist. In contrast, muscarine inhibited GnRH calcium oscillations, and blocking M2 and M4 together prevented this inhibition. Labeling for choline acetyltransferase (ChAT) and GnRH revealed ChAT fibers contacting GnRH neurons, primarily in the medial septum (MS), and in greater number in females than males. ChAT positive cells in the MS are known to express p75NGFRs. Labeling for p75NGFR, ChAT and GnRH indicated that ChAT fibers contacting GnRH cells originate from cholinergic cells within these same rostral areas. Together, these results indicate that cholinergic cells in septal areas can directly regulate GnRH neurons.


Assuntos
Acetilcolina , Hormônio Liberador de Gonadotropina , Animais , Feminino , Masculino , Acetilcolina/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Cálcio , Neurônios , Colinérgicos/farmacologia
6.
Sci Rep ; 13(1): 355, 2023 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-36611058

RESUMO

Simultaneous knockout of the neuroendocrine marker genes Ptprn and Ptprn2, which encode the protein tyrosine phosphatase receptors N and N2, causes infertility in female mice while males are fertile. To elucidate the mechanism of the sex-specific roles of Ptprn and Ptprn2 in mouse reproduction, we analyzed the effects of their double knockout (DKO) on the hypothalamic-pituitary-gonadal axis. In DKO females, delayed puberty and lack of ovulation were observed, complemented by changes in ovarian gene expression and steroidogenesis. In contrast, testicular gene expression, steroidogenesis, and reproductive organs development were not significantly affected in DKO males. However, in both sexes, pituitary luteinizing hormone (LH) beta gene expression and LH levels were reduced, as well as follicle-stimulating hormone beta gene and gonadotropin-releasing hormone (GnRH) gene, while the calcium-mobilizing and LH secretory actions of GnRH were preserved. Hypothalamic Gnrh1 and Kiss1 gene expression was also reduced in DKO females and males. In parallel, a significant decrease in the density of immunoreactive GnRH and kisspeptin fibers was detected in the hypothalamic arcuate nucleus of DKO females and males. The female-specific kisspeptin immunoreactivity in the rostral periventricular region of the third ventricle was also reduced in DKO females, but not in DKO males. These data indicate a critical role of Ptprn and Ptprn2 in kisspeptin-GnRH neuronal function and sexual dimorphism in the threshold levels of GnRH required to preserve reproductive functions.


Assuntos
Hormônio Liberador de Gonadotropina , Kisspeptinas , Masculino , Feminino , Camundongos , Animais , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Reprodução , Hipotálamo/metabolismo , Proteínas Tirosina Fosfatases/metabolismo
7.
Glia ; 71(2): 205-228, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36093576

RESUMO

The mammalian pituitary gland is a complex organ consisting of hormone-producing cells, anterior lobe folliculostellate cells (FSCs), posterior lobe pituicytes, vascular pericytes and endothelial cells, and Sox2-expressing stem cells. We present single-cell RNA sequencing and immunohistofluorescence analyses of pituitary cells of adult female rats with a focus on the transcriptomic profiles of nonhormonal cell types. Samples obtained from whole pituitaries and separated anterior and posterior lobe cells contained all expected pituitary resident cell types and lobe-specific vascular cell subpopulations. FSCs and pituicytes expressed S100B, ALDOC, EAAT1, ALDH1A1, and VIM genes and proteins, as well as other astroglial marker genes, some common and some cell type-specific. We also found that the SOX2 gene and protein were expressed in ~15% of pituitary cells, including FSCs, pituicytes, and a fraction of hormone-producing cells, arguing against its stem cell specificity. FSCs comprised two Sox2-expressing subclusters; FS1 contained more cells but lower genetic diversity, while FS2 contained proliferative cells, shared genes with hormone-producing cells, and expressed genes consistent with stem cell niche formation, regulation of cell proliferation and stem cell pluripotency, including the Hippo and Wnt pathways. FS1 cells were randomly distributed in the anterior and intermediate lobes, while FS2 cells were localized exclusively in the marginal zone between the anterior and intermediate lobes. These data indicate the identity of the FSCs as anterior pituitary-specific astroglia, with FS1 cells representing differentiated cells equipped for classical FSC roles and FS2 cells exhibiting additional stem cell-like features.


Assuntos
Adeno-Hipófise , Ratos , Feminino , Animais , Adeno-Hipófise/metabolismo , Astrócitos , Células Endoteliais , Células-Tronco , Hormônios/metabolismo , Mamíferos
8.
Curr Opin Pharmacol ; 67: 102316, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36347163

RESUMO

Gonadotropin-releasing hormone (GnRH) is the final output of the central nervous system that drives fertility. A characteristic of GnRH secretion is its pulsatility, which is driven by a pulse generator. Each GnRH pulse triggers a luteinizing hormone (LH) pulse. However, the puzzle has been to reconcile the synchronicity of GnRH neurons with the scattered hypothalamic distribution of their cell bodies. A leap toward understanding GnRH pulses was the discovery of kisspeptin neurons near the distal processes of GnRH neurons, which secrete kisspeptins, potent excitatory neuropeptides on GnRH neurons, and equipped with dual, but opposite, self-modulatory neuropeptides, neurokinin B and dynorphin. Over the last decade, this cell-to-cell communication has been dissected in animal models. Today the 50-year quest for the basic mechanism of GnRH pulse generation may be over, but questions about its physiological tuning remain. Here is an overview of recent basic research that frames translational research.


Assuntos
Núcleo Arqueado do Hipotálamo , Hormônio Liberador de Gonadotropina , Animais , Hormônio Liberador de Gonadotropina/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Neurocinina B/metabolismo , Kisspeptinas/metabolismo , Neurônios/fisiologia
9.
Curr Opin Pharmacol ; 66: 102274, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35994915

RESUMO

Pituitary gonadotrophs play a key role in reproductive functions by secreting luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The LH secretory activity of gonadotroph is controlled by hypothalamic gonadotropin-releasing hormone (GnRH) via GnRH receptors and is accompanied by only minor effects on high basal Lhb gene expression. The secretory profiles of GnRH and LH are highly synchronized, with the latter reflecting a depletion of prestored LH in secretory vesicles by regulated exocytosis. In contrast, FSH is predominantly released by constitutive exocytosis, and secretory activity reflects the kinetics of Fshb gene expression controlled by GnRH, activin, and inhibin. Here is a review of recent data to improve the understanding of multiple patterns of gonadotroph gene expression and hormone secretion.


Assuntos
Gonadotrofos , Ativinas/genética , Ativinas/metabolismo , Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/metabolismo , Expressão Gênica , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Inibinas/genética , Inibinas/metabolismo , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo
10.
eNeuro ; 9(2)2022.
Artigo em Inglês | MEDLINE | ID: mdl-35165199

RESUMO

Gonadotropin-releasing hormone (GnRH)-secreting neurons control fertility. The release of GnRH peptide regulates the synthesis and release of both luteinizing hormone (LH) and Follicle stimulation hormone (FSH) from the anterior pituitary. While it is known that dopamine regulates GnRH neurons, the specific dopamine receptor subtype(s) involved remain unclear. Previous studies in adult rodents have reported juxtaposition of fibers containing tyrosine hydroxylase (TH), a marker of catecholaminergic cells, onto GnRH neurons and that exogenous dopamine inhibits GnRH neurons postsynaptically through dopamine D1-like and/or D2-like receptors. Our microarray data from GnRH neurons revealed a high level of Drd4 transcripts [i.e., dopamine D4 receptor (D4R)]. Single-cell RT-PCR and immunocytochemistry confirmed GnRH cells express the Drd4 transcript and protein, respectively. Calcium imaging identified changes in GnRH neuronal activity during application of subtype-specific dopamine receptor agonists and antagonists when GABAergic and glutamatergic transmission was blocked. Dopamine, dopamine with D1/5R-specific or D2/3R-specific antagonists or D4R-specific agonists decreased the frequency of calcium oscillations. In contrast, D1/5R-specific agonists increased the frequency of calcium oscillations. The D4R-mediated inhibition was dependent on Gαi/o protein coupling, while the D1/5R-mediated excitation required Gαs protein coupling. Together, these results indicate that D4R plays an important role in the dopaminergic inhibition of GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina , Receptores de Dopamina D4 , Animais , Dopamina/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Neurônios/fisiologia , Receptores de Dopamina D4/metabolismo
11.
J Neuroendocrinol ; 34(5): e13073, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34939256

RESUMO

For about two decades, recordings of identified gonadotropin-releasing hormone (GnRH) neurons have provided a wealth of information on their properties. We describe areas of consensus and debate the intrinsic electrophysiologic properties of these cells, their response to fast synaptic and neuromodulatory input, Ca2+ imaging correlates of action potential firing, and signaling pathways regulating these aspects. How steroid feedback and development change these properties, functions of GnRH neuron subcompartments and local networks, as revealed by chemo- and optogenetic approaches, are also considered.


Assuntos
Estradiol , Hormônio Liberador de Gonadotropina , Potenciais de Ação/fisiologia , Estradiol/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Transdução de Sinais
12.
Endocrinology ; 162(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33564881

RESUMO

RFamide-related peptides (RFRPs, mammalian orthologs of gonadotropin-inhibitory hormone) convey circadian, seasonal, and social cues to the reproductive system. They regulate gonadotropin secretion by modulating gonadotropin-releasing hormone (GnRH) neurons via the RFRP receptor. Mice lacking this receptor are fertile but exhibit abnormal gonadotropin responses during metabolic challenges, such as acute fasting, when the normal drop in gonadotropin levels is delayed. Although it is known that these food intake signals to the reproductive circuit originate in the nucleus tractus solitarius (NTS) in the brainstem, the phenotype of the neurons conveying the signal remains unknown. Given that neuropeptide FF (NPFF), another RFamide peptide, resides in the NTS and can bind to the RFRP receptor, we hypothesized that NPFF may regulate GnRH neurons. To address this question, we used a combination of techniques: cell-attached electrophysiology on GnRH-driven green fluorescent protein-tagged neurons in acute brain slices; calcium imaging on cultured GnRH neurons; and immunostaining on adult brain tissue. We found (1) NPFF inhibits GnRH neuron excitability via the RFRP receptor and its canonical signaling pathway (Gi/o protein and G protein-coupled inwardly rectifying potassium channels), (2) NPFF-like fibers in the vicinity of GnRH neurons coexpress neuropeptide Y, (3) the majority of NPFF-like cell bodies in the NTS also coexpress neuropeptide Y, and (4) acute fasting increased NPFF-like immunoreactivity in the NTS. Together these data indicate that NPFF neurons within the NTS inhibit GnRH neurons, and thus reproduction, during fasting but prior to the energy deficit.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Receptores de Peptídeos/metabolismo , Animais , Tronco Encefálico/metabolismo , Jejum/metabolismo , Feminino , Glicoproteínas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/metabolismo , Oligopeptídeos/metabolismo
13.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33443156

RESUMO

Fertility relies upon pulsatile release of gonadotropin-releasing hormone (GnRH) that drives pulsatile luteinizing hormone secretion. Kisspeptin (KP) neurons in the arcuate nucleus are at the center of the GnRH pulse generation and the steroid feedback control of GnRH secretion. However, KP evokes a long-lasting response in GnRH neurons that is hard to reconcile with periodic GnRH activity required to drive GnRH pulses. Using calcium imaging, we show that 1) the tetrodotoxin-insensitive calcium response evoked by KP relies upon the ongoing activity of canonical transient receptor potential channels maintaining voltage-gated calcium channels in an activated state, 2) the duration of the calcium response is determined by the rate of resynthesis of phosphatidylinositol 4,5-bisphosphate (PIP2), and 3) nitric oxide terminates the calcium response by facilitating the resynthesis of PIP2 via the canonical pathway guanylyl cyclase/3',5'-cyclic guanosine monophosphate/protein kinase G. In addition, our data indicate that exposure to nitric oxide after KP facilitates the calcium response to a subsequent KP application. This effect was replicated using electrophysiology on GnRH neurons in acute brain slices. The interplay between KP and nitric oxide signaling provides a mechanism for modulation of the refractory period of GnRH neurons after KP exposure and places nitric oxide as an important component for tonic GnRH neuronal pulses.


Assuntos
Sinalização do Cálcio/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Feminino , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Óxido Nítrico/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/fisiologia , Cultura Primária de Células/métodos
14.
Front Cell Neurosci ; 12: 228, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30127721

RESUMO

One key signaling pathway known to influence neuronal migration involves the extracellular matrix protein Reelin. Typically, signaling of Reelin occurs via apolipoprotein E receptor 2 (ApoER2) and very low-density lipoprotein receptor (VLDLR), and the cytoplasmic adapter protein disabled 1 (Dab1). However, non-canonical Reelin signaling has been reported, though no receptors have yet been identified. Cariboni et al. (2005) indicated Dab1-independent Reelin signaling impacts gonadotropin releasing hormone-1 (GnRH) neuronal migration. GnRH cells are essential for reproduction. Prenatal migration of GnRH neurons from the nasal placode to the forebrain, juxtaposed to olfactory axons and olfactory ensheathing cells (OECs), has been well documented, and it is clear that alterations in migration of these cells can cause delayed or absent puberty. This study was initiated to delineate the non-canonical Reelin signaling pathways used by GnRH neurons. Chronic treatment of nasal explants with CR-50, an antibody known to interfere with Reelin homopolymerization and Dab1 phosphorylation, decreased the distance GnRH neurons and OECs migrated. Normal migration of these two cell types was observed when Reelin was co-applied with CR-50. Immunocytochemistry was performed to determine if OECs might transduce Reelin signals via the canonical pathway, and subsequently indirectly altering GnRH neuronal migration. We show that in mouse: (1) both OECs and GnRH cells express ApoER2, VLDLR and Dab1, and (2) GnRH neurons and OECs show a normal distribution in the brain of two mutant reeler lines. These results indicate that the canonical Reelin pathway is present in GnRH neurons and OECs, but that Reelin is not essential for development of these two systems in vivo.

15.
Int J Older People Nurs ; 13(3): e12200, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29770560

RESUMO

AIM: The aim of this integrative review of the literature was to evaluate and summarise current research about how nurses maintain and improve hospitalised older peoples' mobility levels. BACKGROUND: Older persons make up the majority of healthcare recipients, and they are at risk to experience significant decline in their mobility once hospitalised. This can result in longer hospitalisations or nursing home admissions. Currently, it is not well understood how nurses maintain and restore mobility of hospitalised older persons. DESIGN: An integrative literature review using key concepts related to hospitalised older people, mobility and nursing care was conducted. Whittemore and Khalf's five-stage methodological framework for integrative reviews was utilised. METHODS: Two reviewers screened 1640 resources from four computerised databases published in English during 2000-2017. Reviewers used the Mixed Methods Appraisal Tool (MMAT) and CASP quality appraisal tools to assess the thirteen included articles. RESULTS: The findings of this review reveal that little is known about how frequently nurses are mobilising, that many nurses perceive mobilising older patients to be physiotherapy's responsibility and that education about mobilisation can improve nurses' willingness to mobilise people. CONCLUSION: By investing in education and training programmes targeted for nurses, nurses can feel empowered in their ability to mobilise patients and are encouraged to take ownership of their patient's functional needs. In order to facilitate mobility, adequate staffing levels are necessary for transferring and ambulation, mobility assistive devices such as walkers and canes and environments with adequate space to mobilise. More research is needed to better understand and overcome barriers that nurses face in mobilised older people in acute care. IMPLICATIONS FOR PRACTICE: The nursing team can work together to prioritise mobilisation to assist in restoring and maintaining the function of hospitalised older people. Educators could review their mobility programmes to increase graduate nurses' confidence and self-efficacy in mobility assessments and thus prepare graduate nurses for the realities of practice.


Assuntos
Hospitalização , Limitação da Mobilidade , Papel do Profissional de Enfermagem , Recursos Humanos de Enfermagem Hospitalar , Caminhada , Humanos , Equipamentos Ortopédicos
16.
eNeuro ; 5(6)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30627649

RESUMO

The pulsatile release of gonadotropin-releasing hormone (GnRH) is a key feature of the hypothalamic-pituitary-gonadal axis. Kisspeptin neurons in the arcuate nucleus (ARC) trigger GnRH neuronal activity, but how GnRH neurons return to baseline electrical activity is unknown. Nociceptin/orphanin-FQ (OFQ) is an inhibitory neuromodulator. ARC proopiomelanocortin (POMC) neurons, known to receive inputs from ARC kisspeptin neurons, contact GnRH neurons and coexpress OFQ in the rat. In the present study, the effect of OFQ(1-13) on GnRH neurons was determined in the mouse. We identified transcripts for the OFQ receptor [opioid receptor like 1 (ORL1)] in GnRH neurons, and, using two-model systems (explants and slices), we found that OFQ exerted a potent inhibition on GnRH neurons, with or without excitatory inputs. We confirmed that the inhibition was mediated by ORL1 via Gi/o-protein coupling. The inhibition, occurring independently of levels of intracellular cyclic adenosine monophosphate, was sensitive to inwardly rectifying potassium channels. The only specific blocker of Gi/o-protein-coupled inwardly rectifying potassium (GIRK) channels, tertiapin-Q (TPNQ), was ineffective in the inhibition of OFQ. Two GIRK activators, one sharing the binding site of TPNQ and one active only on GIRK1-containing GIRK channels, failed to trigger an inhibition. In contrast, protein kinase C phosphorylation activation, known to inhibit GIRK2-mediated currents, prevented the OFQ inhibition. These results indicate a specific combination of GIRK subunits, GIRK2/3 in GnRH neurons. In vivo, double-labeled OFQ/POMC fibers were found in the vicinity of GnRH neurons, and OFQ fibers apposed GnRH neurons. Together, this study brings to light a potent neuromodulator of GnRH neurons.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Peptídeos Opioides/farmacologia , Vasodilatadores/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Venenos de Abelha/farmacologia , Bicuculina/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Antagonistas de Receptores de GABA-A/farmacologia , Hormônio Liberador de Gonadotropina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo , Valina/análogos & derivados , Valina/farmacologia , Receptor de Nociceptina , Nociceptina
17.
Artigo em Inglês | MEDLINE | ID: mdl-28790978

RESUMO

Fertility relies on the proper functioning of the hypothalamic-pituitary-gonadal axis. The hormonal cascade begins with hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH) into the hypophyseal portal system. In turn, the GnRH-activated gonadotrophs in the anterior pituitary release gonadotropins, which then act on the gonads to regulate gametogenesis and sex steroidogenesis. Finally, sex steroids close this axis by feeding back to the hypothalamus. Despite this seeming straightforwardness, the axis is orchestrated by a complex neuronal network in the central nervous system. For reproductive success, GnRH neurons, the final output of this network, must integrate and translate a wide range of cues, both environmental and physiological, to the gonadotrophs via pulsatile GnRH secretion. This secretory profile is critical for gonadotropic function, yet the mechanisms underlying these pulses remain unknown. Literature supports both intrinsically and extrinsically driven GnRH neuronal activity. However, the caveat of the techniques supporting either one of the two hypotheses is the gap between events recorded at a single-cell level and GnRH secretion measured at the population level. This review aims to compile data about GnRH neuronal activity focusing on the physiological output, GnRH secretion.

18.
Endocrinology ; 157(8): 3197-212, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27359210

RESUMO

GnRH neurons are regulated by hypothalamic kisspeptin neurons. Recently, galanin was identified in a subpopulation of kisspeptin neurons. Although the literature thoroughly describes kisspeptin activation of GnRH neurons, little is known about the effects of galanin on GnRH neurons. This study investigated whether galanin could alter kisspeptin signaling to GnRH neurons. GnRH cells maintained in explants, known to display spontaneous calcium oscillations, and a long-lasting calcium response to kisspeptin-10 (kp-10), were used. First, transcripts for galanin receptors (GalRs) were examined. Only GalR1 was found in GnRH neurons. A series of experiments was then performed to determine the action of galanin on kp-10 activated GnRH neurons. Applied after kp-10 activation, galanin 1-16 (Gal1-16) rapidly suppressed kp-10 activation. Applied with kp-10, Gal1-16 prevented kp-10 activation until its removal. To determine the mechanism by which galanin inhibited kp-10 activation of GnRH neurons, Gal1-16 and galanin were applied to spontaneously active GnRH neurons. Both inhibited GnRH neuronal activity, independent of GnRH neuronal inputs. This inhibition was mimicked by a GalR1 agonist but not by GalR2 or GalR2/3 agonists. Although Gal1-16 inhibition relied on Gi/o signaling, it was independent of cAMP levels but sensitive to blockers of G protein-coupled inwardly rectifying potassium channels. A newly developed bioassay for GnRH detection showed Gal1-16 decreased the kp-10-evoked GnRH secretion below detection threshold. Together, this study shows that galanin is a potent regulator of GnRH neurons, possibly acting as a physiological break to kisspeptin excitation.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/agonistas , Galanina/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos , Feminino , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/farmacologia , Camundongos , Gravidez
19.
Endocrinology ; 157(5): 1980-90, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26934298

RESUMO

Peripheral feedback of gonadal estrogen to the hypothalamus is critical for reproduction. Bisphenol A (BPA), an environmental pollutant with estrogenic actions, can disrupt this feedback and lead to infertility in both humans and animals. GnRH neurons are essential for reproduction, serving as an important link between brain, pituitary, and gonads. Because GnRH neurons express several receptors that bind estrogen, they are potential targets for endocrine disruptors. However, to date, direct effects of BPA on GnRH neurons have not been shown. This study investigated the effects of BPA on GnRH neuronal activity using an explant model in which large numbers of primary GnRH neurons are maintained and express many of the receptors found in vivo. Because oscillations in intracellular calcium have been shown to correlate with electrical activity in GnRH neurons, calcium imaging was used to assay the effects of BPA. Exposure to 50µM BPA significantly decreased GnRH calcium activity. Blockage of γ-aminobutyric acid ergic and glutamatergic input did not abrogate the inhibitory BPA effect, suggesting direct regulation of GnRH neurons by BPA. In addition to estrogen receptor-ß, single-cell RT-PCR analysis confirmed that GnRH neurons express G protein-coupled receptor 30 (G protein-coupled estrogen receptor 1) and estrogen-related receptor-γ, all potential targets for BPA. Perturbation studies of the signaling pathway revealed that the BPA-mediated inhibition of GnRH neuronal activity occurred independent of estrogen receptors, GPER, or estrogen-related receptor-γ, via a noncanonical pathway. These results provide the first evidence of a direct effect of BPA on GnRH neurons.


Assuntos
Compostos Benzidrílicos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Disruptores Endócrinos/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/efeitos dos fármacos , Fenóis/farmacologia , Animais , Cálcio/metabolismo , Estradiol/farmacologia , Camundongos , Neurônios/metabolismo , Receptores de Estrogênio/metabolismo , Reprodução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
J Neurosci ; 33(31): 12689-97, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23904605

RESUMO

Information processing by neurons has been traditionally envisioned to occur in discrete neuronal compartments. Specifically, dendrites receive and integrate synaptic inputs while axons initiate and conduct spikes to distal neuronal targets. We report here in mice, using morphological reconstructions and electrophysiology, that the gonadotropin-releasing hormone (GnRH) neurons that control mammalian fertility do not conform to this stereotype and instead possess a single projection structure that functions simultaneously as an axon and dendrite. Specifically, we show that the GnRH neuron projection to the median eminence to control pituitary hormone secretion possesses a spike initiation site and conducts action potentials while also exhibiting spines and synaptic appositions along its entire length. Classical axonal or dendritic markers are not detectable in the projection process. Activation of ionotropic glutamate and/or GABA receptors along the GnRH neuron projection is capable of depolarizing the membrane potential and initiating action potentials. In addition, focal glutamate application to the projection is able to regulate the width of propagating spikes. These data demonstrate that GnRH neurons elaborate a previously uncharacterized neuronal projection that functions simultaneously as an axon and dendrite. This structure, termed a "dendron," greatly expands the dynamic control of GnRH secretion into the pituitary portal system to regulate fertility.


Assuntos
Axônios/fisiologia , Dendritos/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Animais , Biotina/metabolismo , Vasos Sanguíneos/metabolismo , Channelrhodopsins , GABAérgicos/farmacologia , Galectina 1/metabolismo , Ácido Glutâmico/farmacologia , Hormônio Liberador de Gonadotropina/genética , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Masculino , Eminência Mediana/citologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Estimulação Luminosa , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...