Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neuroendocrinol ; 33(3): e12945, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33713519

RESUMO

Neurones in the arcuate nucleus co-expressing kisspeptin, neurokinin B (NKB) and dynorphin (KNDy) play a critical role in the control of gonadotrophin-releasing hormone (GnRH) and luteinising hormone (LH) secretion. In sheep, KNDy neurones mediate both steroid-negative- and -positive-feedback during pulsatile and preovulatory surge secretions of GnRH/LH, respectively. In addition, KNDy neurones receive glutamatergic inputs expressing vGlut2, a glutamate transporter that serves as a marker for those terminals, from both KNDy neurones and other populations of glutamatergic neurones. Previous work reported higher numbers of vGlut2-positive axonal inputs onto KNDy neurones during the LH surge than in luteal phase ewes. In the present study, we further examined the effects of the ovarian steroids progesterone (P) and oestradiol (E2 ) on glutamatergic inputs to KNDy neurones. Ovariectomised (OVX) ewes received either no further treatment (OVX) or steroid treatments that mimicked the luteal phase (low E2  + P), and early (low E2 ) or late follicular (high E2 ) phases of the oestrous cycle (n = 4 or 5 per group). Brain sections were processed for triple-label immunofluorescent detection of NKB/vGlut2/synaptophysin and analysed using confocal microscopy. We found higher numbers of vGlut2 inputs onto KNDy neurones in high E2 compared to the other three treatment groups. These results suggest that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase depend on increasing levels of E2 required for the preovulatory GnRH/surge. These synaptic changes likely contribute to the positive-feedback action of oestrogen on GnRH/LH secretion and thus the generation of the preovulatory surge in the sheep.


Assuntos
Dinorfinas/fisiologia , Estradiol/fisiologia , Fase Folicular/fisiologia , Glutamatos/fisiologia , Kisspeptinas/fisiologia , Neurocinina B/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Estradiol/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/sangue , Fase Luteal/efeitos dos fármacos , Hormônio Luteinizante/sangue , Ovariectomia , Ovinos , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
2.
Nat Rev Endocrinol ; 16(8): 407-420, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32427949

RESUMO

Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.


Assuntos
Metabolismo Energético/fisiologia , Kisspeptinas/fisiologia , Reprodução/fisiologia , Animais , Dinorfinas/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Homeostase , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Kisspeptinas/genética , Hormônio Luteinizante/fisiologia , Neurocinina B/fisiologia , Neurônios/fisiologia , Ovário/fisiologia , Puberdade/fisiologia
3.
J Neurosci ; 39(49): 9738-9747, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31645462

RESUMO

Fertility critically depends on the gonadotropin-releasing hormone (GnRH) pulse generator, a neural construct comprised of hypothalamic neurons coexpressing kisspeptin, neurokoinin-B and dynorphin. Here, using mathematical modeling and in vivo optogenetics we reveal for the first time how this neural construct initiates and sustains the appropriate ultradian frequency essential for reproduction. Prompted by mathematical modeling, we show experimentally using female estrous mice that robust pulsatile release of luteinizing hormone, a proxy for GnRH, emerges abruptly as we increase the basal activity of the neuronal network using continuous low-frequency optogenetic stimulation. Further increase in basal activity markedly increases pulse frequency and eventually leads to pulse termination. Additional model predictions that pulsatile dynamics emerge from nonlinear positive and negative feedback interactions mediated through neurokinin-B and dynorphin signaling respectively are confirmed neuropharmacologically. Our results shed light on the long-elusive GnRH pulse generator offering new horizons for reproductive health and wellbeing.SIGNIFICANCE STATEMENT The gonadotropin-releasing hormone (GnRH) pulse generator controls the pulsatile secretion of the gonadotropic hormones LH and FSH and is critical for fertility. The hypothalamic arcuate kisspeptin neurons are thought to represent the GnRH pulse generator, since their oscillatory activity is coincident with LH pulses in the blood; a proxy for GnRH pulses. However, the mechanisms underlying GnRH pulse generation remain elusive. We developed a mathematical model of the kisspeptin neuronal network and confirmed its predictions experimentally, showing how LH secretion is frequency-modulated as we increase the basal activity of the arcuate kisspeptin neurons in vivo using continuous optogenetic stimulation. Our model provides a quantitative framework for understanding the reproductive neuroendocrine system and opens new horizons for fertility regulation.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Animais , Dinorfinas/fisiologia , Ciclo Estral/fisiologia , Retroalimentação Fisiológica , Feminino , Kisspeptinas/fisiologia , Hormônio Luteinizante/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Teóricos , Neurocinina B/fisiologia , Neurônios/fisiologia , Optogenética , Gravidez , Reprodução/fisiologia , Ritmo Ultradiano/fisiologia
4.
Vitam Horm ; 111: 227-246, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421702

RESUMO

The organization of estrogenic signaling in the CNS is exceedingly complex. It is comprised of peripherally and centrally synthesized estrogens, and a plethora of types of estrogen receptor that can localize to both the nucleus and the plasma membrane. Moreover, CNS estrogen receptors can exist independent of aromatase (aka estrogen synthase) as well as oligomerize with it, along with a host of other membrane signaling proteins. This ability of CNS estrogen receptors to either to physically pair or exist separately enables locally produced estrogens to act on multiple spatial levels, with a high degree of gradated regulation and plasticity, signaling either in-phase or out-of phase with circulating estrogens. This complexity explains the numerous contradictory findings regarding sex-dependent pain processing and sexually dimorphic opioid antinociception. This review highlights the increasing awareness that estrogens are major endogenous arbiters of both opioid analgesic actions and the mechanisms used to achieve them. This behooves us to understand, and possibly intercede at, the points of intersection of estrogenic signaling and opioid functionality. Factors that integrate estrogenic actions at subcellular, synaptic, and CNS regional levels are likely to be prime drug targets for novel pharmacotherapies designed to modulate CNS estrogen-dependent opioid functionalities and possibly circumvent the current opioid epidemic.


Assuntos
Analgésicos Opioides/farmacologia , Estrogênios/fisiologia , Reprodução/fisiologia , Caracteres Sexuais , Analgesia , Animais , Aromatase , Encéfalo/fisiologia , Dinorfinas/fisiologia , Feminino , Humanos , Masculino , Sistemas Neurossecretores/fisiologia , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Receptores de Estrogênio/fisiologia , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/fisiologia , Transdução de Sinais/fisiologia
5.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-31297186

RESUMO

This review recounts the origins and development of the concept of the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator. It starts in the late 1960s when striking rhythmic episodes of luteinizing hormone secretion, as reflected by circulating concentrations of this gonadotropin, were first observed in monkeys and ends in the present day. It is currently an exciting time witnessing the application, primarily to the mouse, of contemporary neurobiological approaches to delineate the mechanisms whereby Kiss1/NKB/Dyn (KNDy) neurons in the arcuate nucleus of the hypothalamus generate and time the pulsatile output of kisspeptin from their terminals in the median eminence that in turn dictates intermittent GnRH release and entry of this decapeptide into the primary plexus of the hypophysial portal circulation. The review concludes with an examination of questions that remain to be addressed.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Kisspeptinas/fisiologia , Animais , Dinorfinas/fisiologia , Camundongos , Neurocinina B/fisiologia , Neurônios/fisiologia
6.
Reproduction ; 156(3): R83-R99, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29880718

RESUMO

Early work in ewes provided a wealth of information on the physiological regulation of pulsatile gonadotropin-releasing hormone (GnRH) secretion by internal and external inputs. Identification of the neural systems involved, however, was limited by the lack of information on neural mechanisms underlying generation of GnRH pulses. Over the last decade, considerable evidence supported the hypothesis that a group of neurons in the arcuate nucleus that contain kisspeptin, neurokinin B and dynorphin (KNDy neurons) are responsible for synchronizing secretion of GnRH during each pulse in ewes. In this review, we describe our current understanding of the neural systems mediating the actions of ovarian steroids and three external inputs on GnRH pulsatility in light of the hypothesis that KNDy neurons play a key role in GnRH pulse generation. In breeding season adults, estradiol (E2) and progesterone decrease GnRH pulse amplitude and frequency, respectively, by actions on KNDy neurons, with E2 decreasing kisspeptin and progesterone increasing dynorphin release onto GnRH neurons. In pre-pubertal lambs, E2 inhibits GnRH pulse frequency by decreasing kisspeptin and increasing dynorphin release, actions that wane as the lamb matures to allow increased pulsatile GnRH secretion at puberty. Less is known about mediators of undernutrition and stress, although some evidence implicates kisspeptin and dynorphin, respectively, in the inhibition of GnRH pulse frequency by these factors. During the anoestrus, inhibitory photoperiod acting via melatonin activates A15 dopaminergic neurons that innervate KNDy neurons; E2 increases dopamine release from these neurons to inhibit KNDy neurons and suppress the frequency of kisspeptin and GnRH release.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Homeostase/fisiologia , Ovinos/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Cruzamento , Dinorfinas/fisiologia , Estradiol/farmacologia , Ciclo Estral , Retroalimentação Fisiológica , Feminino , Kisspeptinas/fisiologia , Hormônio Luteinizante/metabolismo , Neurocinina B/fisiologia , Neurônios/fisiologia , Periodicidade , Progesterona/farmacologia , Estações do Ano , Maturidade Sexual/fisiologia
7.
Gynecol Endocrinol ; 34(11): 913-919, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29902942

RESUMO

Women during perimenopausal period experience a range of symptoms, which interfere with physical, sexual, and social life. About 65-75% of symptoms connected with postmenopausal period are vasomotor symptoms (VMS), such as hot flushes and night sweats. Hot flushes are subjective sensation of heat associated with cutaneous vasodilatation and drop in core temperature. It is suspected that VMS are strongly correlated with pulsatile oversecretion of gonadotropin-releasing hormone (GnRH) and subsequently luteinizing hormone (LH). Evidence has accumulated in parallel showing that lack of negative feedback of steroid hormones synthesized in ovary causes overactivation of hypertrophied kisspeptin/neurokinin B/dynorphin (KNDy) neurons, located in infundibular nucleus. Oversecretion of both kisspeptin (KISS1) and neurokinin B (NKB), as well as downregulation of dynorphin, plays dominant role in creation of GnRH pulses. This in turn causes VMS. Administration of senktide, highly potent and selective NK3R agonist, resulted in increase of serum LH concentration, induction of VMS, increase in heart rate, and skin temperature in postmenopausal women. These finding suggest that modulation of KNDy neurons may become new therapeutic approach in the treatment of VMS.


Assuntos
Fogachos/etiologia , Hipotálamo/fisiologia , Neurônios/fisiologia , Pós-Menopausa/fisiologia , Sistema Vasomotor/fisiologia , Dinorfinas/fisiologia , Retroalimentação Fisiológica , Feminino , Fogachos/tratamento farmacológico , Humanos , Kisspeptinas/fisiologia , Neurocinina B/fisiologia
8.
J Neuroendocrinol ; 29(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28475285

RESUMO

Pulsatile secretion of gonadotrophin-releasing hormone (GnRH)/luteinising hormone is indispensable for the onset of puberty and reproductive activities at adulthood in mammalian species. A cohort of neurones expressing three neuropeptides, namely kisspeptin, encoded by the Kiss1 gene, neurokinin B (NKB) and dynorphin A, localised in the hypothalamic arcuate nucleus (ARC), so-called KNDy neurones, comprises a putative intrinsic source of the GnRH pulse generator. Synchronous activity among KNDy neurones is considered to be required for pulsatile GnRH secretion. It has been reported that gap junctions play a key role in synchronising electrical activity in the central nervous system. Thus, we hypothesised that gap junctions are involved in the synchronised activities of KNDy neurones, which is induced by NKB-NK3R signalling. We determined the role of NKB-NK3R signalling in Ca2+ oscillation (an indicator of neuronal activities) of KNDy neurones and its synchronisation mechanism among KNDy neurones. Senktide, a selective agonist for NK3R, increased the frequency of Ca2+ oscillations in cultured Kiss1-GFP cells collected from the mediobasal hypothalamus of the foetal Kiss1-green fluorescent protein (GFP) mice. The senktide-induced Ca2+ oscillations were synchronised in the Kiss1-GFP and neighbouring glial cells. Confocal microscopy analysis of these cells, which have shown synchronised Ca2+ oscillations, revealed close contacts between Kiss1-GFP cells, as well as between Kiss1-GFP cells and glial cells. Dye coupling experiments suggest cell-to-cell communication through gap junctions between Kiss1-GFP cells and neighbouring glial cells. Connexin-26 and -37 mRNA were found in isolated ARC Kiss1 cells taken from adult female Kiss1-GFP transgenic mice. Furthermore, 18ß-glycyrrhetinic acids and mefloquine, which are gap junction inhibitors, attenuated senktide-induced Ca2+ oscillations in Kiss1-GFP cells. Taken together, these results suggest that NKB-NK3R signalling enhances synchronised activities among neighbouring KNDy neurones, and that both neurone-neurone and neurone-glia communications via gap junctions possibly contribute to synchronised activities among KNDy neurones.


Assuntos
Junções Comunicantes/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Fragmentos de Peptídeos/farmacologia , Substância P/análogos & derivados , Animais , Células Cultivadas , Conexinas/metabolismo , Dinorfinas/fisiologia , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacologia , Kisspeptinas/genética , Bulbo/metabolismo , Mefloquina/farmacologia , Camundongos Transgênicos , Neuroglia/metabolismo , Neurocinina B/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/antagonistas & inibidores , Substância P/antagonistas & inibidores , Substância P/farmacologia
9.
Neurobiol Aging ; 50: 30-38, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27842268

RESUMO

Pulsatile secretion of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) decreases during aging. Kisspeptin (encoded by Kiss1) neurons in the arcuate nucleus coexpress neurokinin B (Tac3) and dynorphin (Pdyn) and are critical for regulating the GnRH/LH pulse. We therefore examined kisspeptin neurons by histochemistry and pulsatile LH release in rats aged 2-3 (Young), 12-13 (Young-Middle), 19-22 (Late-Middle), and 24-26 (Old) months. Total LH concentrations, sampled for 3 hours, decreased in both sexes with aging. In females, numbers of Tac3 and Pdyn neurons were significantly reduced in all aging rats, and numbers of Kiss1 neurons were significantly reduced in Late-Middle and Old rats. In males, numbers of all 3 neuron-types were significantly decreased in all aging rats. GnRH agonist induced LH release in all animals; however, the increased LH concentration in all aging rats was less than that in Young rats. These results suggest that expression of each gene in kisspeptin neurons may be controlled individually during aging, and that reduction of their expression or change in pituitary responsiveness may cause attenuated pulsatile LH secretion.


Assuntos
Envelhecimento/patologia , Envelhecimento/fisiologia , Dinorfinas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Animais , Dinorfinas/fisiologia , Feminino , Histocitoquímica , Hipotálamo/citologia , Hipotálamo/patologia , Kisspeptinas/fisiologia , Masculino , Menopausa/metabolismo , Menopausa/fisiologia , Neurocinina B/fisiologia , Neurônios/fisiologia , Fluxo Pulsátil , Ratos Wistar
10.
Peptides ; 76: 14-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26654796

RESUMO

Food intake and physical activity are regulated by multiple neuropeptides, including orexin and dynorphin (DYN). Orexin-A (OXA) is one of two orexin peptides with robust roles in regulation of food intake and spontaneous physical activity (SPA). DYN collectively refers to several peptides, some of which act through opioid receptors (opioid DYN) and some whose biological effects are not mediated by opioid receptors (non-opioid DYN). While opioid DYN is known to increase food intake, the effects of non-opioid DYN peptides on food intake and SPA are unknown. Neurons that co-express and release OXA and DYN are located within the lateral hypothalamus. Limited evidence suggests that OXA and opioid DYN peptides can interact to modulate some aspects of behaviors classically related to orexin peptide function. The paraventricular hypothalamic nucleus (PVN) is a brain area where OXA and DYN peptides might interact to modulate food intake and SPA. We demonstrate that injection of des-Tyr-dynorphin (DYN-A(2-17), a non opioid DYN peptide) into the PVN increases food intake and SPA in adult mice. Co-injection of DYN-A(2-17) and OXA in the PVN further increases food intake compared to DYN-A(2-17) or OXA alone. This is the first report describing the effects of non-opioid DYN-A(2-17) on food intake and SPA, and suggests that DYN-A(2-17) interacts with OXA in the PVN to modulate food intake. Our data suggest a novel function for non-opioid DYN-A(2-17) on food intake, supporting the concept that some behavioral effects of the orexin neurons result from combined actions of the orexin and DYN peptides.


Assuntos
Dinorfinas/fisiologia , Orexinas/metabolismo , Fragmentos de Peptídeos/fisiologia , Animais , Regulação do Apetite , Ingestão de Energia , Masculino , Camundongos Endogâmicos BALB C , Atividade Motora
11.
J Neuroendocrinol ; 28(12)2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-28328155

RESUMO

Puberty onset involves increased gonadotrophin-release (GnRH) release as a result of decreased sensitivity to oestrogen (E2 )-negative feedback. Because GnRH neurones lack E2 receptor α, this pathway must contain interneurones. One likely candidate is KNDy neurones (kisspeptin, neurokinin B, dynorphin). The overarching hypothesis of the present study was that the prepubertal hiatus in luteinising hormone (LH) release involves reduced kisspeptin and/or heightened dynorphin input. We first tested the specific hypothesis that E2 would reduce kisspeptin-immunopositive cell numbers and increase dynorphin-immunopositive cell numbers. We found that kisspeptin cell numbers were higher in ovariectomised (OVX) lambs than OVX lambs treated with E2 (OVX+ E2 ) or those left ovary-intact. Very few arcuate dynorphin cells were identified in any group. Next, we hypothesised that central blockade of κ-opioid receptor (KOR) would increase LH secretion at a prepubertal (6 months) but not postpubertal (10 months) age. Luteinising hormone pulse frequency and mean LH increased during infusion of a KOR antagonist, norbinaltorphimine, in OVX + E2 lambs at the prepubertal age but not in the same lambs at the postpubertal age. We next hypothesised that E2 would increase KOR expression in GnRH neurones or alter synaptic input to KNDy neurones in prepubertal ewes. Oestrogen treatment decreased the percentage of GnRH neurones coexpressing KOR (approximately 68%) compared to OVX alone (approximately 78%). No significant differences in synaptic contacts per cell between OVX and OVX + E2 groups were observed. Although these initial data are consistent with dynorphin inhibiting pulsatile LH release prepubertally, additional work will be necessary to define the source and mechanisms of this inhibition.


Assuntos
Dinorfinas/fisiologia , Estrogênios/fisiologia , Hormônio Luteinizante/metabolismo , Neurônios/metabolismo , Puberdade , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/administração & dosagem , Estrogênios/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ovariectomia , Receptores Opioides kappa/metabolismo , Carneiro Doméstico , Sinaptofisina/metabolismo
12.
Hum Reprod Update ; 20(4): 485-500, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24615662

RESUMO

BACKGROUND: The discovery of kisspeptin as key central regulator of GnRH secretion has led to a new level of understanding of the neuroendocrine regulation of human reproduction. The related discovery of the kisspeptin-neurokinin B-dynorphin (KNDy) pathway in the last decade has further strengthened our understanding of the modulation of GnRH secretion by endocrine, metabolic and environmental inputs. In this review, we summarize current understanding of the physiological roles of these novel neuropeptides, and discuss the clinical relevance of these discoveries and their potential translational applications. METHODS: A systematic literature search was performed using PUBMED for all English language articles up to January 2014. In addition, the reference lists of all relevant original research articles and reviews were examined. This review focuses mainly on published human studies but also draws on relevant animal data. RESULTS: Kisspeptin is a principal regulator of the secretion of gonadotrophins, and through this key role it is critical for the onset of puberty, the regulation of sex steroid-mediated feedback and the control of adult fertility. Although there is some sexual dimorphism, both neuroanatomically and functionally, these functions are apparent in both men and women. Kisspeptin acts upstream of GnRH and, following paracrine stimulatory and inhibitory inputs from neurokinin B and dynorphin (KNDy neuropeptides), signals directly to GnRH neurones to control pulsatile GnRH release. When administered to humans in different isoforms, routes and doses, kisspeptin robustly stimulates LH secretion and LH pulse frequency. Manipulation of the KNDy system is currently the focus of translational research with the possibility of future clinical application to regulate LH pulsatility, increasing gonadal sex steroid secretion in reproductive disorders characterized by decreased LH pulsatility, including hypothalamic amenorrhoea and hypogonadotropic hypogonadism. Conversely there may be scope to reduce the activity of the KNDy system to reduce LH secretion where hypersecretion of LH adds to the phenotype, such as in polycystic ovary syndrome. CONCLUSIONS: Kisspeptin is a recently discovered neuromodulator that controls GnRH secretion mediating endocrine and metabolic inputs to the regulation of human reproduction. Manipulation of kisspeptin signalling has the potential for novel therapies in patients with pathologically low or high LH pulsatility.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Infertilidade/etiologia , Kisspeptinas/fisiologia , Reprodução/fisiologia , Amenorreia/etiologia , Amenorreia/fisiopatologia , Animais , Dinorfinas/fisiologia , Retroalimentação Fisiológica/fisiologia , Feminino , Hormônios Esteroides Gonadais/fisiologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Humanos , Hipogonadismo/etiologia , Hipogonadismo/fisiopatologia , Infertilidade/fisiopatologia , Hormônio Luteinizante/metabolismo , Masculino , Neurocinina B/fisiologia , Neurônios/fisiologia , Caracteres Sexuais , Fatores Sexuais , Desenvolvimento Sexual/fisiologia , Transdução de Sinais/fisiologia
13.
Nihon Arukoru Yakubutsu Igakkai Zasshi ; 49(5): 227-37, 2014 Oct.
Artigo em Japonês | MEDLINE | ID: mdl-25651617

RESUMO

Nicotine (NIC) regulates various cellular functions acting on the nicotinic acetylcholine receptor (nAChR). And nAChR consists of ligand-gated cation channels with pentameric structure and composed of α and ß subunits. In the central nervous system, α 4 ß 2 and α 7 nAChRs are the most abundantly expressed as nAChR subtypes. There are several lines of evidence indicating that systemic administration of NIC elicits the release of endogenous opioids, such as, endorphins, enkephalins and dynorphins, in the brain. NIC exerts numerous acute effects, for example, antinociceptive effects and the activating effects of the hypothalamic-pituitary-adrenal (HPA) axis. In these effects, NIC-induced antinociception, but not HPA axis activation, was inhibited by opioid receptor antagonist, naloxone (NLX), and was also suppressed in morphine tolerated mice, indicating the participation of the endogenous opioid system in NIC-induced antinociception, but not HPA axis activation. Moreover, NIC-induced antinociception was antagonized by both α 4 ß 2 and α 7 nAChR antagonists, while NIC-induced HPA axis activation was antagonized by α 4 ß 2 nAChR antagonist, but not by α 7 nAChR antagonist. These results suggest that the endogenous opioid system may not be located on the downstream of α 4 ß 2 nAChR. On the other hand, NIC has substantial physical dependence liability. NLX elicits NIC withdrawal after repeated NIC administration evaluated by corticosterone increase as a withdrawal sign, and NLX-precipitated NIC withdrawal is inhibited by concomitant administration of other opioid receptor antagonist, naltrexone, indicating the participation of endogenous opioid system in the development of physical dependence on NIC. NLX-precipitated NIC withdrawal was also inhibited by concomitant administration of an α 7 nAChR antagonist, but not an α 4 ß 2 nAChR antagonist. Taken together, these findings suggest that the endogenous opioid system may be located on the downstream of α 7 nAChR and participates in the development of physical dependence on NIC.


Assuntos
Nicotina/farmacologia , Peptídeos Opioides/fisiologia , Tabagismo/genética , Receptor Nicotínico de Acetilcolina alfa7/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Analgésicos , Animais , Encéfalo/metabolismo , Dinorfinas/metabolismo , Dinorfinas/fisiologia , Endorfinas/metabolismo , Endorfinas/fisiologia , Encefalinas/metabolismo , Encefalinas/fisiologia , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Camundongos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nicotina/antagonistas & inibidores , Nociceptividade/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
14.
Front Neuroendocrinol ; 34(3): 211-27, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23872331

RESUMO

Despite affecting millions of individuals, the etiology of hot flushes remains unknown. Here we review the physiology of hot flushes, CNS pathways regulating heat-dissipation effectors, and effects of estrogen on thermoregulation in animal models. Based on the marked changes in hypothalamic kisspeptin, neurokinin B and dynorphin (KNDy) neurons in postmenopausal women, we hypothesize that KNDy neurons play a role in the mechanism of flushes. In the rat, KNDy neurons project to preoptic thermoregulatory areas that express the neurokinin 3 receptor (NK3R), the primary receptor for NKB. Furthermore, activation of NK3R in the median preoptic nucleus, part of the heat-defense pathway, reduces body temperature. Finally, ablation of KNDy neurons reduces cutaneous vasodilatation and partially blocks the effects of estrogen on thermoregulation. These data suggest that arcuate KNDy neurons relay estrogen signals to preoptic structures regulating heat-dissipation effectors, supporting the hypothesis that KNDy neurons participate in the generation of flushes.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Dinorfinas/fisiologia , Fogachos/fisiopatologia , Hipotálamo/metabolismo , Kisspeptinas/fisiologia , Neurocinina B/fisiologia , Animais , Regulação da Temperatura Corporal/efeitos dos fármacos , Estradiol/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Humanos , Hormônio Luteinizante/metabolismo , Modelos Biológicos , Neurônios/fisiologia , Ovariectomia , Pós-Menopausa/fisiologia , Área Pré-Óptica/metabolismo , Ratos , Receptores da Neurocinina-3/metabolismo , Transdução de Sinais , Pele/irrigação sanguínea , Cauda/irrigação sanguínea , Vasodilatação
15.
Addict Biol ; 18(4): 678-88, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22994904

RESUMO

The brain stress-response system is critically involved in the addiction process, stimulating drug consumption and the relapse to drug taking in abstinent addicts. At the same time, its functioning is affected by chronic drug exposure. Here, we have investigated the role of the endogenous opioid peptide dynorphin as a modulator of effects of long-term ethanol consumption on the brain stress-response system. Using the two-bottle choice paradigm, we demonstrate an enhanced ethanol preference in male dynorphin knockout mice. Exposure to mild foot shock increased ethanol consumption in wild-type control littermates, but not in dynorphin-deficient animals. Blood adrenocorticotropic hormone levels determined 5 minutes after the shock were not affected by the genotype. We also determined the neuronal reactivity after foot shock exposure using c-Fos immunoreactivity in limbic structures. This was strongly influenced by both genotype and chronic ethanol consumption. Long-term alcohol exposure elevated the foot shock-induced c-Fos expression in the basolateral amygdala in wild-type animals, but had the opposite effect in dynorphin-deficient mice. An altered c-Fos reactivity was also found in the periventricular nucleus, the thalamus and the hippocampus of dynorphin knockouts. Together these data suggest that dynorphin plays an important role in the modulation of the brain stress-response systems after chronic ethanol exposure.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Dinorfinas/fisiologia , Etanol/farmacologia , Sistema Límbico/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Fisiológico/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/metabolismo , Análise de Variância , Animais , Comportamento Aditivo/metabolismo , Dinorfinas/genética , Etanol/administração & dosagem , Feminino , Preferências Alimentares , Genótipo , Imunoquímica , Sistema Límbico/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout/genética , Reforço Psicológico , Autoadministração , Caracteres Sexuais , Estresse Fisiológico/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Tálamo/metabolismo
16.
J Neurosci ; 32(49): 17582-96, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23223282

RESUMO

Activation of the dynorphin/κ-opioid receptor (KOR) system by repeated stress exposure or agonist treatment produces place aversion, social avoidance, and reinstatement of extinguished cocaine place preference behaviors by stimulation of p38α MAPK, which subsequently causes the translocation of the serotonin transporter (SERT, SLC6A4) to the synaptic terminals of serotonergic neurons. In the present study we extend those findings by showing that stress-induced potentiation of cocaine conditioned place preference occurred by a similar mechanism. In addition, SERT knock-out mice did not show KOR-mediated aversion, and selective reexpression of SERT by lentiviral injection into the dorsal raphe restored the prodepressive effects of KOR activation. Kinetic analysis of several neurotransporters demonstrated that repeated swim stress exposure selectively increased the V(max) but not K(m) of SERT without affecting dopamine transport or the high-capacity, low-affinity transporters. Although the serotonergic neurons in the dorsal raphe project throughout the forebrain, a significant stress-induced increase in cell-surface SERT expression was only evident in the ventral striatum, and not in the dorsal striatum, hippocampus, prefrontal cortex, amygdala, or dorsal raphe. Stereotaxic microinjections of the long-lasting KOR antagonist norbinaltorphimine demonstrated that local KOR activation in the nucleus accumbens, but not dorsal raphe, mediated this stress-induced increase in ventral striatal surface SERT expression. Together, these results support the hypothesis that stress-induced activation of the dynorphin/KOR system produces a transient increase in serotonin transport locally in the ventral striatum that may underlie some of the adverse consequences of stress exposure, including the potentiation of the rewarding effects of cocaine.


Assuntos
Aprendizagem da Esquiva/fisiologia , Cocaína/farmacologia , Corpo Estriado/metabolismo , Dinorfinas/fisiologia , Recompensa , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Dinorfinas/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/fisiologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microinjeções/métodos , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacocinética , Nicotina/efeitos adversos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Núcleos da Rafe/efeitos dos fármacos , Núcleos da Rafe/metabolismo , Núcleos da Rafe/fisiologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/fisiologia , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Síndrome de Abstinência a Substâncias/metabolismo , Sinaptossomos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
17.
Neuropeptides ; 46(6): 383-94, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23062312

RESUMO

Chronic opiate usage, whether prescribed or illicit, has been associated with changes in bone mass and is a recognized risk factor for the development of osteoporosis; however, the mechanism behind this effect is unknown. Here we show that lack of dynorphin, an endogenous opioid, in mice (Dyn-/-), resulted in a significantly elevated cancellous bone volume associated with greater mineral apposition rate and increased resorption indices. A similar anabolic phenotype was evident in bone of mice lacking dynorphin's cognate receptor, the kappa opioid receptor. Lack of opioid receptor expression in primary osteoblastic cultures and no change in bone cell function after dynorphin agonist treatment in vitro indicates an indirect mode of action. Consistent with a hypothalamic action, central dynorphin signaling induces extracellular signal-regulated kinase (ERK) phosphorylation and c-fos activation of neurons in the arcuate nucleus of the hypothalamus (Arc). Importantly, this signaling also leads to an increase in Arc NPY mRNA expression, a change known to decrease bone formation. Further implicating NPY in the skeletal effects of dynorphin, Dyn-/-/NPY-/- double mutant mice showed comparable increases in bone formation to single mutant mice, suggesting that dynorphin acts upstream of NPY signaling to control bone formation. Thus the dynorphin system, acting via NPY, may represent a pathway by which higher processes including stress, reward/addiction and depression influence skeletal metabolism. Moreover, understanding of these unique interactions may enable modulation of the adverse effects of exogenous opioid treatment without directly affecting analgesic responses.


Assuntos
Osso e Ossos/fisiologia , Dinorfinas/fisiologia , Homeostase/fisiologia , Animais , Western Blotting , Composição Corporal/genética , Composição Corporal/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , DNA Complementar/biossíntese , DNA Complementar/isolamento & purificação , Dinorfinas/genética , Feminino , Homeostase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Neuropeptídeo Y/fisiologia , Osteoblastos/fisiologia , Gravidez , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Células Estromais/fisiologia , Tomografia Computadorizada por Raios X
18.
Endocr J ; 59(7): 631-40, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22641014

RESUMO

The role of Neurokinin B (NKB) and Dynorphin A (Dyn) in the regulation of the hypothalamic pituitary axis is an important area of recent investigation. These peptides are critical for the rhythmic release of GnRH, which subsequently stimulates the secretion of the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The present study utilized the gonadotroph cell line LßT2 and the somatolactotroph GH3 cell line to examine the possible role of these peptides in pituitary hormone secretion. The NKB receptor (NK3R) and the Dyn receptor (the κ-opiate receptor (KOR)) were both detected in LßT2 cells and GH3 cells. NKB, by itself, failed to increase gonadotropin LHß and FSHß promoter activities and did not modulate the effects of GnRH on gonadotropin promoter activity. In GH3 cells, NKB significantly increased TRH-induced PRL promoter activity although NKB alone did not have an effect on basal PRL promoter activity. Dyn had no effect on gonadotropin promoters alone or in combination with GnRH stimulation. PRL promoters stimulated by TRH were not significantly changed by Dyn. TRH-induced PRL promoter activity was further increased in the presence of higher concentrations of NKB, whereas Dyn did not have a significant effect on the PRL promoter even at a high concentration. In addition, TRH-induced ERK (Extracelluar signal-regulated kinase) activation was enhanced in the presence of NKB. Our current study demonstrated that NKB had a stimulatory effect on PRL expression in a PRL-producing cell, but had no effect on gonadotropin secretion from a gonadotroph cell line.


Assuntos
Dinorfinas/fisiologia , Gonadotrofos/metabolismo , Lactotrofos/metabolismo , Neurocinina B/fisiologia , Somatotrofos/metabolismo , Animais , Linhagem Celular , Dinorfinas/genética , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Gonadotrofos/efeitos dos fármacos , Gonadotropinas/genética , Gonadotropinas/metabolismo , Lactotrofos/efeitos dos fármacos , Neurocinina B/genética , Neurocinina B/metabolismo , Neurocinina B/farmacologia , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Prolactina/genética , Prolactina/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Receptores Opioides/genética , Receptores Opioides/metabolismo , Receptores de Taquicininas/genética , Receptores de Taquicininas/metabolismo , Somatotrofos/efeitos dos fármacos , Transfecção , Receptor de Nociceptina
19.
Endocrinology ; 153(1): 307-15, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22109887

RESUMO

Neurokinin B (NKB) and its receptor (neurokinin-3 receptor) are coexpressed with kisspeptin and dynorphin A (Dyn) within neurons of the hypothalamic arcuate nucleus, the suggested site of the GnRH pulse generator. It is thought that these neuropeptides interact to regulate gonadotropin secretion. Using the ovariectomized (OVX) and OVX 17ß-estradiol-replaced rat models, we have carried out a series of in vivo neuropharmacological and electrophysiological experiments to elucidate the hierarchy between the kisspeptin, NKB, and Dyn signaling systems. Rats were implanted with intracerebroventricular cannulae and cardiac catheters for frequent (every 5 min) automated serial blood sampling. Freely moving rats were bled for 6 h, with intracerebroventricular injections taking place after a 2-h control bleeding period. A further group of OVX rats was implanted with intra-arcuate electrodes for the recording of multiunit activity volleys, which coincide invariably with LH pulses. Intracerebroventricular administration of the selective neurokinin-3 receptor agonist, senktide (100-600 pmol), caused a dose-dependent suppression of LH pulses and multiunit activity volleys. The effects of senktide did not differ between OVX and 17ß-estradiol-replaced OVX animals. Pretreatment with a selective Dyn receptor (κ opioid receptor) antagonist, norbinaltorphimine (6.8 nmol), blocked the senktide-induced inhibition of pulsatile LH secretion. Intracerebroventricular injection of senktide did not affect the rise in LH concentrations after administration of kisspeptin (1 nmol), and neither did kisspeptin preclude the senktide-induced suppression of LH pulses. These data show that NKB suppresses the frequency of the GnRH pulse generator in a Dyn/κ opioid receptor-dependent fashion.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurocinina B/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Dinorfinas/fisiologia , Feminino , Kisspeptinas/farmacologia , Kisspeptinas/fisiologia , Hormônio Luteinizante/metabolismo , Antagonistas de Entorpecentes , Neurocinina B/fisiologia , Ovariectomia , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Kisspeptina-1 , Receptores da Neurocinina-3/agonistas , Receptores da Neurocinina-3/fisiologia , Receptores Opioides/fisiologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/fisiologia , Transdução de Sinais , Substância P/análogos & derivados , Substância P/farmacologia
20.
Cell Signal ; 24(5): 991-1001, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22200678

RESUMO

Apelin receptor (APJ) and kappa opioid receptor (KOR) are members of the family A of G protein-coupled receptors (GPCRs). These two receptors are involved in the central nervous system regulation of the cardiovascular system. Here, we explore the possibility of heterodimerization between APJ and KOR and investigate their novel signal transduction characteristics. Co-immunoprecipitation (Co-IP), co-localization and bioluminescence resonance energy transfer (BRET) assays confirmed the heterodimerization of APJ and KOR. In APJ and KOR stably transfected HEK293 cells, treatment with APJ ligand apelin-13 or KOR ligand dynorphinA (1-13) resulted in higher phosphorylation levels of extracellular-regulated kinases 1/2 (ERK1/2) compared to HEK293 cells transfected with either APJ or KOR alone. The siRNA knockdown of either APJ or KOR receptor in human umbilical vein endothelial cells (HUVECs) resulted in significant reduction of the apelin-13 induced ERK activation. Additionally both forskolin (FSK)-induced cAMP levels and cAMP response element reporter activities were significantly reduced, whereas the serum response element luciferase (SRE-luc) reporter activity was significantly upregulated. Moreover, the ERK phosphorylation and SRE-luc activity were abrogated by the protein kinase C (PKC) inhibitor. These results demonstrate for the first time that human APJ forms a heterodimer with KOR and leads to increased PKC and decreased protein kinase A activity leading to a significant increase in cell proliferation, which may translate to the regulation of diverse biological actions and offers the potential for the development of more selective and tissue specific drug therapies.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Sistema de Sinalização das MAP Quinases , Multimerização Proteica , Receptores Opioides kappa/metabolismo , Apelina , Proliferação de Células , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinorfinas/metabolismo , Dinorfinas/fisiologia , Ativação Enzimática , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA