Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 801
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cereb Cortex ; 31(9): 3975-3985, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34037726

RESUMO

Musical training is thought to be related to improved language skills, for example, understanding speech in background noise. Although studies have found that musicians and nonmusicians differed in morphology of bilateral arcuate fasciculus (AF), none has associated such white matter features with speech-in-noise (SIN) perception. Here, we tested both SIN and the diffusivity of bilateral AF segments in musicians and nonmusicians using diffusion tensor imaging. Compared with nonmusicians, musicians had higher fractional anisotropy (FA) in the right direct AF and lower radial diffusivity in the left anterior AF, which correlated with SIN performance. The FA-based laterality index showed stronger right lateralization of the direct AF and stronger left lateralization of the posterior AF in musicians than nonmusicians, with the posterior AF laterality predicting SIN accuracy. Furthermore, hemodynamic activity in right superior temporal gyrus obtained during a SIN task played a full mediation role in explaining the contribution of the right direct AF diffusivity on SIN performance, which therefore links training-related white matter plasticity, brain hemodynamics, and speech perception ability. Our findings provide direct evidence that differential microstructural plasticity of bilateral AF segments may serve as a neural foundation of the cross-domain transfer effect of musical experience to speech perception amid competing noise.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Núcleo Arqueado do Hipotálamo/ultraestrutura , Percepção Auditiva/fisiologia , Música/psicologia , Ruído , Percepção da Fala/fisiologia , Anisotropia , Núcleo Arqueado do Hipotálamo/diagnóstico por imagem , Circulação Cerebrovascular , Imagem de Tensor de Difusão , Feminino , Lateralidade Funcional , Humanos , Masculino , Plasticidade Neuronal/fisiologia , Lobo Temporal/irrigação sanguínea , Lobo Temporal/fisiologia , Substância Branca/fisiologia , Adulto Jovem
2.
J Neurosci ; 40(16): 3203-3216, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32209609

RESUMO

Giving birth triggers a wide repertoire of physiological and behavioral changes in the mother to enable her to feed and care for her offspring. These changes require coordination and are often orchestrated from the CNS, through as of yet poorly understood mechanisms. A neuronal population with a central role in puerperal changes is the tuberoinfundibular dopamine (TIDA) neurons that control release of the pituitary hormone, prolactin, which triggers key maternal adaptations, including lactation and maternal care. Here, we used Ca2+ imaging on mice from both sexes and whole-cell recordings on female mouse TIDA neurons in vitro to examine whether they adapt their cellular and network activity according to reproductive state. In the high-prolactin state of lactation, TIDA neurons shift to faster membrane potential oscillations, a reconfiguration that reverses upon weaning. During the estrous cycle, however, which includes a brief, but pronounced, prolactin peak, oscillation frequency remains stable. An increase in the hyperpolarization-activated mixed cation current, Ih, possibly through unmasking as dopamine release drops during nursing, may partially explain the reconfiguration of TIDA rhythms. These findings identify a reversible plasticity in hypothalamic network activity that can serve to adapt the dam for motherhood.SIGNIFICANCE STATEMENT Motherhood requires profound behavioral and physiological adaptations to enable caring for offspring, but the underlying CNS changes are poorly understood. Here, we show that, during lactation, neuroendocrine dopamine neurons, the "TIDA" cells that control prolactin secretion, reorganize their trademark oscillations to discharge in faster frequencies. Unlike previous studies, which typically have focused on structural and transcriptional changes during pregnancy and lactation, we demonstrate a functional switch in activity and one that, distinct from previously described puerperal modifications, reverses fully on weaning. We further provide evidence that a specific conductance (Ih) contributes to the altered network rhythm. These findings identify a new facet of maternal brain plasticity at the level of membrane properties and consequent ensemble activity.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Neurônios Dopaminérgicos/fisiologia , Lactação/fisiologia , Rede Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Potenciais de Ação/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos
3.
Neuroendocrinology ; 111(1-2): 45-69, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32028278

RESUMO

OBJECTIVE: We examined whether pituitary adenylate cyclase-activating polypeptide (PACAP) excites proopiomelanocortin (POMC) neurons via PAC1 receptor mediation and transient receptor potential cation (TRPC) channel activation. METHODS: Electrophysiological recordings were done in slices from both intact male and ovariectomized (OVX) female PACAP-Cre mice and eGFP-POMC mice. RESULTS: In recordings from POMC neurons in eGFP-POMC mice, PACAP induced a robust inward current and increase in conductance in voltage clamp, and a depolarization and increase in firing in current clamp. These postsynaptic actions were abolished by inhibitors of the PAC1 receptor, TRPC channels, phospholipase C, phosphatidylinositol-3-kinase, and protein kinase C. Estradiol augmented the PACAP-induced inward current, depolarization, and increased firing, which was abrogated by estrogen receptor (ER) antagonists. In optogenetic recordings from POMC neurons in PACAP-Cre mice, high-frequency photostimulation induced inward currents, depolarizations, and increased firing that were significantly enhanced by Gq-coupled membrane ER signaling in an ER antagonist-sensitive manner. Importantly, the PACAP-induced excitation of POMC neurons was notably reduced in obese, high-fat (HFD)-fed males. In vivo experiments revealed that intra-arcuate nucleus (ARC) PACAP as well as chemogenetic and optogenetic stimulation of ventromedial nucleus (VMN) PACAP neurons produced a significant decrease in energy intake accompanied by an increase in energy expenditure, effects blunted by HFD in males and partially potentiated by estradiol in OVX females. CONCLUSIONS: These findings reveal that the PACAP-induced activation of PAC1 receptor and TRPC5 channels at VMN PACAP/ARC POMC synapses is potentiated by estradiol and attenuated under conditions of diet-induced obesity/insulin resistance. As such, they advance our understanding of how PACAP regulates the homeostatic energy balance circuitry under normal and pathophysiological circumstances.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Metabolismo Energético/fisiologia , Neurônios/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Pró-Opiomelanocortina , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Fenômenos Eletrofisiológicos , Metabolismo Energético/efeitos dos fármacos , Feminino , Cobaias , Homeostase , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/efeitos dos fármacos
4.
Neuroendocrinology ; 110(11-12): 1010-1027, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31935735

RESUMO

INTRODUCTION: Synchronous and pulsatile neural activation of kisspeptin neurons in the arcuate nucleus (ARN) are important components of the gonadotropin-releasing hormone pulse generator, the final common pathway for central regulation of mammalian reproduction. However, whether ARN kisspeptin neurons can intrinsically generate self-sustained synchronous oscillations from the early neonatal period and how they are regulated remain unclear. OBJECTIVE: This study aimed to examine the endogenous rhythmicity of ARN kisspeptin neurons and its neural regulation using a neonatal organotypic slice culture model. METHODS: We monitored calcium (Ca2+) dynamics in real-time from individual ARN kisspeptin neurons in neonatal organotypic explant cultures of Kiss1-IRES-Cre mice transduced with genetically encoded Ca2+ indicators. Pharmacological approaches were employed to determine the regulations of kisspeptin neuron-specific Ca2+ oscillations. A chemogenetic approach was utilized to assess the contribution of ARN kisspeptin neurons to the population dynamics. RESULTS: ARN kisspeptin neurons in neonatal organotypic cultures exhibited a robust synchronized Ca2+ oscillation with a period of approximately 3 min. Kisspeptin neuron-specific Ca2+ oscillations were dependent on voltage-gated sodium channels and regulated by endoplasmic reticulum-dependent Ca2+ homeostasis. Chemogenetic inhibition of kisspeptin neurons abolished synchronous Ca2+ oscillations, but the autocrine actions of the neuropeptides were marginally effective. Finally, neonatal ARN kisspeptin neurons were regulated by N-methyl-D-aspartate and gamma-aminobutyric acid receptor-mediated neurotransmission. CONCLUSION: These data demonstrate that ARN kisspeptin neurons in organotypic cultures can generate synchronized and self-sustained Ca2+ oscillations. These oscillations controlled by multiple regulators within the ARN are a novel ultradian rhythm generator that is active during the early neonatal period.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Sinalização do Cálcio/fisiologia , Kisspeptinas , Neurônios/fisiologia , Ritmo Ultradiano/fisiologia , Animais , Animais Recém-Nascidos , Camundongos , Camundongos Transgênicos
5.
Neuroendocrinology ; 110(7-8): 582-594, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31484184

RESUMO

Obesity and anorexia result in dysregulation of the hypothalamic-pituitary-gonadal axis, negatively impacting reproduction. Ghrelin, secreted from the stomach, potentially mediates negative energy states and neuroendocrine control of reproduction by acting through the growth hormone secretagogue receptor (GHSR). GHSR is expressed in hypothalamic arcuate (ARC) Kisspeptin/Neurokinin B (Tac2)/Dynorphin (KNDy) neurons. Ghrelin is known to inhibit the M-current produced by KCNQ channels in other ARC neurons. In addition, we have shown 17ß-estradiol (E2) increases Ghsr expression in KNDy neurons 6-fold and increases the M-current in NPY neurons. We hypothesize that E2 increases GHSR expression in KNDy neurons to increase ghrelin sensitivity during negative energy states. Furthermore, we suspect ghrelin targets the M-current in KNDy neurons to control reproduction and energy homeostasis. We utilized ovariectomized Tac2-EGFP adult female mice, pretreated with estradiol benzoate (EB) or oil vehicle and performed whole-cell-patch-clamp recordings to elicit the M-current in KNDy neurons using standard activation protocols in voltage-clamp. Using the selective KCNQ channel blocker XE-991 (40 µM) to target the M-current, oil- and EB-treated mice showed a decrease in the maximum peak current by 75.7 ± 13.8 pA (n = 10) and 68.0 ± 14.7 pA (n = 11), respectively. To determine the actions of ghrelin on the M-current, ghrelin was perfused (100 nM) in oil- and EB-treated mice resulting in the suppression of the maximum peak current by 58.5 ± 15.8 pA (n = 9) and 59.2 ± 11.9 pA (n = 9), respectively. KNDy neurons appeared more sensitive to ghrelin when pretreated with EB, revealing that ARC KNDy neurons are more sensitive to ghrelin during states of high E2.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Estradiol/farmacologia , Grelina/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Relação Dose-Resposta a Droga , Feminino , Grelina/metabolismo , Potenciais da Membrana/genética , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ovariectomia , Técnicas de Patch-Clamp , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Taquicininas/genética , Taquicininas/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-32380163

RESUMO

Oxyntomodulin (OXM) is a proglucagon-derived peptide that suppresses hunger in humans. There are some differences in its food intake-inhibitory effects among species. The central mechanisms are unclear and it is unknown if OXM is more efficacious in a gallinaceous species that has not undergone as much selection for growth as the chicken. The objective was thus to determine the effects of OXM on food and water intake and hypothalamic physiology in Japanese quail. At 7 days post-hatch, 6-h-fasted quail were injected intracerebroventricularly (ICV) or intraperitoneally (IP) with 0.32, 0.65, or 1.3 nmol of OXM. All doses decreased food intake for 180 min post-ICV injection. On a cumulative basis, water intake was not affected until 120 min, with the lowest and highest doses decreasing water intake after ICV injection. The two highest doses were anorexigenic when administered via the IP route, whereas all doses were anti-dipsogenic starting at 30 min post-injection. In hypothalamic samples collected at 1-h post-ICV injection, there was an increase in c-Fos immunoreactivity, an indicator of recent neuronal activation, in the arcuate nucleus (ARC) and dorsomedial nucleus (DMN) of the hypothalamus in OXM-injected individuals. Results suggest that quail are more sensitive than chickens to the satiety-inducing effects of OXM. The central mechanism is likely mediated through a pathway in the ARC that is conserved among species, and through activation of the DMN, an effect that is unique to quail. Such knowledge is critical for facilitating the development of novel, side effect-free anti-eating strategies to promote weight-loss in obesity.


Assuntos
Apetite/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Coturnix/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Oxintomodulina/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Distribuição Aleatória
7.
Annu Rev Physiol ; 78: 207-21, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26863324

RESUMO

Although it has been known for more than a century that the brain controls overall energy balance and adiposity by regulating feeding behavior and energy expenditure, the roles for individual brain regions and neuronal subtypes were not fully understood until recently. This area of research is active, and as such our understanding of the central regulation of energy balance is continually being refined as new details emerge. Much of what we now know stems from the discoveries of leptin and the hypothalamic melanocortin system. Hypothalamic circuits play a crucial role in the control of feeding and energy expenditure, and within the hypothalamus, the arcuate nucleus (ARC) functions as a gateway for hormonal signals of energy balance, such as leptin. It is also well established that the ARC is a primary residence for hypothalamic melanocortinergic neurons. The paraventricular hypothalamic nucleus (PVH) receives direct melanocortin input, along with other integrated signals that affect energy balance, and mediates the majority of hypothalamic output to control both feeding and energy expenditure. Herein, we review in detail the structure and function of the ARC-PVH circuit in mediating leptin signaling and in regulating energy balance.


Assuntos
Metabolismo Energético/fisiologia , Leptina/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Alimentos/fisiologia , Humanos , Neurônios/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo
8.
J Neurosci ; 38(5): 1061-1072, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29114074

RESUMO

Estradiol feedback regulates gonadotropin-releasing hormone (GnRH) neurons and subsequent luteinizing hormone (LH) release. Estradiol acts via estrogen receptor α (ERα)-expressing afferents of GnRH neurons, including kisspeptin neurons in the anteroventral periventricular (AVPV) and arcuate nuclei, providing homeostatic feedback on episodic GnRH/LH release as well as positive feedback to control ovulation. Ionotropic glutamate receptors are important for estradiol feedback, but it is not known where they fit in the circuitry. Estradiol-negative feedback decreased glutamatergic transmission to AVPV and increased it to arcuate kisspeptin neurons; positive feedback had the opposite effect. Deletion of ERα in kisspeptin cells decreased glutamate transmission to AVPV neurons and markedly increased it to arcuate kisspeptin neurons, which also exhibited increased spontaneous firing rate. KERKO mice had increased LH pulse frequency, indicating loss of negative feedback. These observations indicate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and neuroendocrine output by estradiol.SIGNIFICANCE STATEMENT The brain regulates fertility through gonadotropin-releasing hormone (GnRH) neurons. Ovarian estradiol regulates the pattern of GnRH (negative feedback) and initiates a surge of release that triggers ovulation (positive feedback). GnRH neurons do not express the estrogen receptor needed for feedback (estrogen receptor α [ERα]); kisspeptin neurons in the arcuate and anteroventral periventricular nuclei are postulated to mediate negative and positive feedback, respectively. Here we extend the network through which feedback is mediated by demonstrating that glutamatergic transmission to these kisspeptin populations is differentially regulated during the reproductive cycle and by estradiol. Electrophysiological and in vivo hormone profile experiments on kisspeptin-specific ERα knock-out mice demonstrate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and for neuroendocrine output.


Assuntos
Estradiol/farmacologia , Glutamatos/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Receptores de Estrogênio/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Dinorfinas/farmacologia , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/fisiologia , Camundongos , Núcleos da Linha Média do Tálamo/fisiologia , Neurônios/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Proestro/fisiologia , Receptores Ionotrópicos de Glutamato/efeitos dos fármacos , Receptores Ionotrópicos de Glutamato/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Receptor ERRalfa Relacionado ao Estrogênio
9.
J Physiol ; 597(6): 1757-1775, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30628058

RESUMO

KEY POINTS: Intracerebroventricular insulin increased sympathetic nerve activity (SNA) and baroreflex control of SNA and heart rate more dramatically in obese male rats; in obese females, the responses were abolished. In obese males, the enhanced lumbar SNA (LSNA) responses were associated with reduced tonic inhibition of LSNA by neuropeptide Y (NPY) in the PVN. However, PVN NPY injection decreased LSNA similarly in obesity prone/obesity resistant/control rats. Collectively, these results suggest that NPY inputs were decreased. In obese females, NPY inhibition in the PVN was maintained. Moreover, NPY neurons in the arcuate nucleus became resistant to the inhibitory effects of insulin. A high-fat diet did not alter arcuate NPY neuronal InsR expression in males or females. Obesity-induced 'selective sensitization' of the brain to the sympathoexcitatory effects of insulin and leptin may contribute to elevated basal SNA, and therefore hypertension development, in males with obesity. These data may explain in part why obesity increases SNA less in women compared to men. ABSTRACT: Obesity increases sympathetic nerve activity (SNA) in men but not women; however, the mechanisms are unknown. We investigated whether intracerebroventricular insulin infusion increases SNA more in obese male than female rats and if sex differences are mediated by changes in tonic inhibition of SNA by neuropeptide Y (NPY) in the paraventricular nucleus (PVN). When consuming a high-fat diet, obesity prone (OP) rats accrued excess fat, whereas obesity resistant (OR) rats maintained adiposity as in rats eating a control (CON) diet. Insulin increased lumbar SNA (LSNA) similarly in CON/OR males and females under urethane anaesthesia. The LSNA response was magnified in OP males but abolished in OP females. In males, blockade of PVN NPY Y1 receptors with BIBO3304 increased LSNA in CON/OR rats but not OP rats. Yet, PVN nanoinjections of NPY decreased LSNA similarly between groups. Thus, tonic PVN NPY inhibition of LSNA may be lost in obese males as a result of a decrease in NPY inputs. By contrast, in females, PVN BIBO3304 increased LSNA similarly in OP, OR and CON rats. After insulin, PVN BIBO3304 failed to increase LSNA in CON/OR females but increased LSNA in OP females, suggesting that with obesity NPY neurons become resistant to the inhibitory effects of insulin. These sex differences were not associated with changes in arcuate NPY neuronal insulin receptor expression. Collectively, these data reveal a marked sex difference in the impact of obesity on the sympathoexcitatory actions of insulin and implicate sexually dimorphic changes in NPY inhibition of SNA in the PVN as one mechanism.


Assuntos
Insulina/farmacologia , Inibição Neural , Neuropeptídeo Y/farmacologia , Obesidade/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/fisiologia , Arginina/análogos & derivados , Arginina/farmacologia , Barorreflexo , Feminino , Insulina/metabolismo , Masculino , Neuropeptídeo Y/metabolismo , Obesidade/fisiopatologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Fatores Sexuais , Sistema Nervoso Simpático/fisiologia
10.
J Physiol ; 597(6): 1605-1625, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30618146

RESUMO

KEY POINTS: Excitatory glutamate neurons are sparse in the rostral hypothalamic arcuate nucleus (ARC), the subregion that has received the most attention in the past. In striking contrast, excitatory neurons are far more common (by a factor of 10) in the caudal ARC, an area which has received relatively little attention. These glutamate cells may play a negative role in energy balance and food intake. They can show an increase in phosphorylated Stat-3 in the presence of leptin, are electrically excited by the anorectic neuromodulator cholecystokinin, and inhibited by orexigenic neuromodulators neuropeptide Y, met-enkephalin, dynorphin and the catecholamine dopamine. The neurons project local axonal connections that excite other ARC neurons including proopiomelanocortin neurons that can play an important role in obesity. These data are consistent with models suggesting that the ARC glutamatergic neurons may play both a rapid and a slower role in acting as anorectic neurons in CNS control of food intake and energy homeostasis. ABSTRACT: Here we interrogate a unique class of excitatory neurons in the hypothalamic arcuate nucleus (ARC) that utilizes glutamate as a fast neurotransmitter using mice expressing GFP under control of the vesicular glutamate transporter 2 (vGluT2) promoter. These neurons show a unique distribution, synaptic characterization, cellular physiology and response to neuropeptides involved in energy homeostasis. Although apparently not previously appreciated, the caudal ARC showed a far greater density of vGluT2 cells than the rostral ARC, as seen in transgenic vGluT2-GFP mice and mRNA analysis. After food deprivation, leptin induced an increase in phosphorylated Stat-3 in vGluT2-positive neurons, indicating a response to hormonal cues of energy state. Based on whole-cell recording electrophysiology in brain slices, vGluT2 neurons were spontaneously active with a spike frequency around 2 Hz. vGluT2 cells were responsive to a number of neuropeptides related to energy homeostasis; they were excited by the anorectic peptide cholecystokinin, but inhibited by orexigenic neuropeptide Y, dynorphin and met-enkephalin, consistent with an anorexic role in energy homeostasis. Dopamine, associated with the hedonic aspect of enhancing food intake, inhibited vGluT2 neurons. Optogenetic excitation of vGluT2 cells evoked EPSCs in neighbouring neurons, indicating local synaptic excitation of other ARC neurons. Microdrop excitation of ARC glutamate cells in brain slices rapidly increased excitatory synaptic activity in anorexigenic proopiomelanocortin neurons. Together these data support the perspective that vGluT2 cells may be more prevalent in the ARC than previously appreciated, and play predominantly an anorectic role in energy metabolism.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Alimentos , Metabolismo Energético , Potenciais Pós-Sinápticos Excitadores , Neurônios/metabolismo , Potenciais de Ação , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Colecistocinina/farmacologia , Dopamina/farmacologia , Dinorfinas/farmacologia , Encefalina Metionina/farmacologia , Ácido Glutâmico/metabolismo , Homeostase , Leptina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neuropeptídeo Y/farmacologia , Pró-Opiomelanocortina/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
11.
Arch Toxicol ; 93(2): 547-558, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30377736

RESUMO

Triclosan (TCS), as a broad spectrum antibacterial agent, is commonly utilized in personal care and household products. Maternal urinary TCS level has been associated with changes in birth weight of infants. We in the present study investigated whether exposure of mice to 8 mg/kg TCS from gestational day (GD) 6 to GD14 alters prenatal and postnatal growth and development, and metabolic phenotypes in male and female offspring (TCS-offspring). Compared with control offspring, body weight in postnatal day (PND) 1 male or female TCS-offspring was reduced, but body weight gain was faster within postnatal 5 days. PND30 and PND60 TCS-offspring showed overweight with increases in visceral fat and adipocyte size. PND60 TCS-offspring displayed delayed glucose clearance and insulin resistance. PND30 TCS-offspring showed an increase in food intake without the changes in the oxygen consumption and respiratory exchange ratio (RER). The expression levels of proopiomelanocortin (POMC), α-melanocyte-stimulating hormone (α-MSH) and single-minded 1 (SIM1) in hypothalamus arcuate nucleus (ARC) and paraventricular nucleus (PVN), respectively, were significantly reduced in PND30 TCS-offspring compared to controls. The hypermethylation of CpG sites at the POMC promoter was observed in PND30 TCS-offspring, while the concentration of serum leptin was elevated and the level of STAT3 phosphorylation in ARC had no significant difference from control. This study demonstrates that TCS exposure during early/mid-gestation through the hypermethylation of the POMC promoter reduces the expression of anorexigenic neuropeptides to cause the postnatal hyperphagic obesity, leading to metabolic syndrome in adulthood.


Assuntos
Hiperfagia/complicações , Obesidade/etiologia , Efeitos Tardios da Exposição Pré-Natal , Pró-Opiomelanocortina/genética , Triclosan/toxicidade , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/fisiologia , Metilação de DNA/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hiperfagia/induzido quimicamente , Hiperfagia/genética , Resistência à Insulina , Masculino , Obesidade/genética , Gravidez , Pró-Opiomelanocortina/metabolismo , Regiões Promotoras Genéticas , Aumento de Peso/efeitos dos fármacos , alfa-MSH/biossíntese
12.
J Obstet Gynaecol Res ; 45(12): 2318-2329, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31608564

RESUMO

Ovulation is an essential phenomenon for reproduction in mammalian females along with follicular growth. It is well established that gonadal function is controlled by the neuroendocrine system called the hypothalamus-pituitary-gonadal (HPG) axis. Gonadotropin-releasing hormone (GnRH) neurons, localized in the hypothalamus, had been considered to be the head in governing the HPG axis for a long time until the discovery of kisspeptin. In females, induction of ovulation and folliculogenesis has been linked to a surge mode and pulse mode of GnRH releases, respectively. The mechanisms of how the two modes of GnRH are differently regulated had long remained elusive. The discovery of kisspeptin neurons, distributed in two hypothalamic nuclei, such as the arcuate nucleus in the caudal hypothalamus and preoptic area or the anteroventral periventricular nucleus in the rostral hypothalamic regions, and analyses of the detailed functions of kisspeptin neurons have led marked progress on the understanding of different mechanisms regulating GnRH surges (ovulation) and GnRH pulses (folliculogenesis). The present review will focus on the role of kisspeptin neurons as the GnRH surge generator, including the sexual differentiation of the surge generation system and factors that regulate the surge generator. Comparative aspects between mammalian species are especially focused on.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Hormônio Liberador de Gonadotropina/sangue , Hipotálamo Anterior/fisiologia , Kisspeptinas/fisiologia , Animais , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Hormônio Luteinizante/sangue , Camundongos , Ovulação , Ratos , Diferenciação Sexual , Ácido gama-Aminobutírico/fisiologia
13.
J Physiol ; 596(5): 885-899, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29214635

RESUMO

KEY POINTS: Neurons in the hypothalamus of the brain which secrete the peptide kisspeptin are important regulators of reproduction, and normal reproductive development. Electrical activity, in the form of action potentials, or spikes, leads to secretion of peptides and neurotransmitters, influencing the activity of downstream neurons; in kisspeptin neurons, this activity is highly irregular, but the mechanism of this is not known. In this study, we show that irregularity depends on the presence of a particular type of potassium ion channel in the membrane, which opens transiently in response to electrical excitation. The results contribute to understanding how kisspeptin neurons generate and time their membrane potential spikes, and how reliable this process is. Improved understanding of the activity of kisspeptin neurons, and how it shapes their secretion of peptides, is expected to lead to better treatment for reproductive dysfunction and disorders of reproductive development. ABSTRACT: Kisspeptin neurons in the hypothalamus are critically involved in reproductive function, via their effect on GnRH neuron activity and consequent gonadotropin release. Kisspeptin neurons show an intrinsic irregularity of firing, but the mechanism of this remains unclear. To address this, we carried out targeted whole-cell patch-clamp recordings of kisspeptin neurons in the arcuate nucleus (Kiss1Arc ), in brain slices isolated from adult male Kiss-Cre:tdTomato mice. Cells fired irregularly in response to constant current stimuli, with a wide range of spike time variability, and prominent subthreshold voltage fluctuations. In voltage clamp, both a persistent sodium (NaP) current and a fast transient (A-type) potassium current were apparent, activating at potentials just below the threshold for spiking. These currents have also previously been described in irregular-spiking cortical interneurons, in which the A-type current, mediated by Kv4 channels, interacts with NaP current to generate complex dynamics of the membrane potential, and irregular firing. In Kiss1Arc neurons, A-type current was blocked by phrixotoxin, a specific blocker of Kv4.2/4.3 channels, and consistent expression of Kv4.2 transcripts was detected by single-cell RT-PCR. In addition, firing irregularity was correlated to the density of A-type current in the membrane. Using conductance injection, we demonstrated that adding Kv4-like potassium conductance (gKv4 ) to a cell produces a striking increase in firing irregularity, and excitability is reduced, while subtracting gKv4 has the opposite effects. Thus, we propose that Kv4 interacting dynamically with NaP is a key determinant of the irregular firing behaviour of Kiss1Arc neurons, shaping their physiological function in gonadotropin release.


Assuntos
Potenciais de Ação , Núcleo Arqueado do Hipotálamo/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Canais de Potássio Shal/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia
14.
Reproduction ; 156(4): 283-297, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30305241

RESUMO

The objective of this study was to examine the effect of nutrition during the first 18 weeks of life on the physiological and transcriptional functionality of the hypothalamic (arcuate nucleus region), anterior pituitary and testes in Holstein­Friesian bull calves. Holstein­Friesian bull calves with a mean (±S.D.) age and bodyweight of 19 (±8.2) days and 47.5 (±5.3) kg, respectively, were assigned to either a HIGH (n = 10) or LOW (n = 10) plane of nutrition, to achieve an overall target growth rate of 1.2 or 0.5 kg/day, respectively. At 126 ± 1.1 days of age, all calves were euthanised. Animal performance (weekly) and systemic concentrations of metabolic (monthly) and reproductive hormones (fortnightly) were assessed. Testicular histology, targeted gene and protein expression of the arcuate nucleus region, anterior pituitary and testes were also assessed using qPCR and immunohistochemistry, respectively. The expression of candidate genes in testicular tissue from post pubertal 19-month-old Holstein­Friesian bulls (n = 10) was compared to that of the 18-week-old calves. Metabolite and metabolic hormone profiles generally reflected the improved metabolic status of the calves on the HIGH (P< 0.001). Calves offered a HIGH plane of nutrition were heavier at slaughter (P < 0.001), had larger testes (P < 0.001), larger seminiferous tubule diameter (P < 0.001), more mature spermatogenic cells (P < 0.001) and more Sertoli cells (P < 0.05) in accordance with both morphological and transcriptional data. Overall, testicular gene expression profiles suggested a more mature stage of development in HIGH compared with LOW and were more closely aligned to that of mature bulls. Ghrelin receptor was the only differentially expressed gene between LOW and HIGH calves in either the anterior pituitary (P < 0.05) or arcuate nucleus region of the hypothalamus (P < 0.10) and was upregulated in LOW for both tissues. This study indicates that an enhanced plane of nutrition during early calfhood favourably alters the biochemical regulation of the hypothalamus­anterior pituitary­testicular axis, advancing testicular development and hastening spermatogenesis.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Hormônios/fisiologia , Estado Nutricional , Adeno-Hipófise/fisiologia , Testículo/crescimento & desenvolvimento , Animais , Bovinos , Masculino , Testículo/metabolismo
15.
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
16.
Curr Hypertens Rep ; 20(3): 25, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29556733

RESUMO

PURPOSE OF REVIEW: Here, we review the current understanding of the functional neuroanatomy of neurons expressing Agouti-related peptide (AgRP) and the angiotensin 1A receptor (AT1A) within the arcuate nucleus (ARC) in the control of energy balance. RECENT FINDINGS: The development and maintenance of obesity involves suppression of resting metabolic rate (RMR). RMR control is integrated via AgRP and proopiomelanocortin neurons within the ARC. Their projections to other hypothalamic and extrahypothalamic nuclei contribute to RMR control, though relatively little is known about the contributions of individual projections and the neurotransmitters involved. Recent studies highlight a role for AT1A, localized to AgRP neurons, but the specific function of AT1A within these cells remains unclear. AT1A functions within AgRP neurons to control RMR, but additional work is required to clarify its role within subpopulations of AgRP neurons projecting to distinct second-order nuclei, and the molecular mediators of its signaling within these cells.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Angiotensinas/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Metabolismo Energético/fisiologia , Receptor Tipo 1 de Angiotensina/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Humanos , Hipertensão/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Transdução de Sinais/fisiologia
17.
Bioessays ; 38(4): 316-24, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26898524

RESUMO

AgRP and POMC neurons are two key cell types that regulate feeding in response to hormones and nutrients. Recently, it was discovered that these neurons are also rapidly modulated by the mere sight and smell of food. This rapid sensory regulation "resets" the activity of AgRP and POMC neurons before a single bite of food has been consumed. This surprising and counterintuitive discovery challenges longstanding assumptions about the function and regulation of these cells. Here we review these recent findings and discuss their implications for our understanding of feeding behavior. We propose several alternative hypotheses for how these new observations might be integrated into a revised model of the feeding circuit, and also highlight some of the key questions that remain to be answered.


Assuntos
Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Retroalimentação Fisiológica , Fome/fisiologia , Neurônios/fisiologia , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Antecipação Psicológica , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/fisiologia , Peso Corporal/fisiologia , Regulação da Expressão Gênica , Grelina/genética , Grelina/metabolismo , Homeostase/fisiologia , Humanos , Hipotálamo/citologia , Hipotálamo/fisiologia , Leptina/genética , Leptina/metabolismo , Neurônios/citologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Saciação/fisiologia
18.
J Neurochem ; 143(6): 697-707, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28984906

RESUMO

It has been well-known that hypothalamic orexigenic neuropeptides, orexin-A, and melanin-concentrating hormone (MCH), play important roles in regulation of gastric function. However, what neural pathway mediated by the two neuropeptides affects the gastric function remains unknown. In this study, by way of nucleic stimulation and extracellular recording of single unit electrophysiological properties, we found that electrically stimulating the lateral hypothalamic area (LH) or microinjection of orexin-A into the arcuate nucleus (ARC) excited most gastric distension-responsive neurons in the nuclei and enhanced the gastric function including motility, emptying, and acid secretion of conscious rats. The results indicated that LH-ARC orexin-A-ergic projections may exist and the orexin-A in the ARC affected afferent and efferent signal transmission between ARC and stomach. As expected, combination of retrograde tracing and immunohistochemistry showed that some orexin-A-ergic neurons projected from the LH to the ARC. In addition, microinjection of MCH and its receptor antagonist PMC-3881-PI into the ARC affected the role of orexin-A in the ARC, indicating a possible involvement of the MCH pathway in the orexin-A role. Our findings suggest that there was an orexin-A-ergic pathway between LH and ARC which participated in transmitting information between the central nuclei and the gastrointestinal tract and in regulating the gastric function of rats.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Região Hipotalâmica Lateral/citologia , Vias Neurais/citologia , Vias Neurais/metabolismo , Estômago/inervação , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Região Hipotalâmica Lateral/fisiologia , Masculino , Orexinas/metabolismo , Ratos , Ratos Wistar , Estômago/fisiologia
19.
EMBO J ; 32(6): 844-57, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23403926

RESUMO

pRb is frequently inactivated in tumours by mutations or phosphorylation. Here, we investigated whether pRb plays a role in obesity. The Arcuate nucleus (ARC) in hypothalamus contains antagonizing POMC and AGRP/NPY neurons for negative and positive energy balance, respectively. Various aspects of ARC neurons are affected in high-fat diet (HFD)-induced obesity mouse model. Using this model, we show that HFD, as well as pharmacological activation of AMPK, induces pRb phosphorylation and E2F target gene de-repression in ARC neurons. Some affected neurons express POMC; and deleting Rb1 in POMC neurons induces E2F target gene de-repression, cell-cycle re-entry, apoptosis, and a hyperphagia-obesity-diabetes syndrome. These defects can be corrected by combined deletion of E2f1. In contrast, deleting Rb1 in the antagonizing AGRP/NPY neurons shows no effects. Thus, pRb-E2F1 is an obesity suppression mechanism in ARC POMC neurons and HFD-AMPK inhibits this mechanism by phosphorylating pRb in this location.


Assuntos
Dieta Hiperlipídica , Gorduras na Dieta/farmacologia , Hipotálamo , Obesidade/genética , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/fisiologia , Adenilato Quinase/metabolismo , Adenilato Quinase/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F1/fisiologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotálamo/patologia , Peso Corporal Ideal/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Obesidade/metabolismo , Obesidade/patologia , Fosforilação/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo
20.
Horm Behav ; 89: 98-103, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28063803

RESUMO

In the female rat, sexual receptivity (lordosis) can be facilitated by sequential activation of estrogen receptor (ER) α and G protein-coupled estrogen receptor 1 (GPER) by estradiol. In the estradiol benzoate (EB) primed ovariectomized (OVX) rat, EB initially binds to ERα in the plasma membrane that complexes with and transactivates metabotropic glutamate receptor 1a to activate ß-endorphin neurons in the arcuate nucleus of the hypothalamus (ARH) that project to the medial preoptic nucleus (MPN). This activates MPN µ-opioid receptors (MOP), inhibiting lordosis. Infusion of non-esterified 17ß-estradiol into the ARH rapidly reduces MPN MOP activation and facilitates lordosis via GPER. Tamoxifen (TAM) and ICI 182,780 (ICI) are selective estrogen receptor modulators that activate GPER. Therefore, we tested the hypothesis that TAM and ICI rapidly facilitate lordosis via activation of GPER in the ARH. Our first experiment demonstrated that injection of TAM intraperitoneal, or ICI into the lateral ventricle, deactivated MPN MOP and facilitated lordosis in EB-primed rats. We then tested whether TAM and ICI were acting rapidly through a GPER dependent pathway in the ARH. In EB-primed rats, ARH infusion of either TAM or ICI facilitated lordosis and reduced MPN MOP activation within 30min compared to controls. These effects were blocked by pretreatment with the GPER antagonist, G15. Our findings demonstrate that TAM and ICI deactivate MPN MOP and facilitate lordosis in a GPER dependent manner. Thus, TAM and ICI may activate GPER in the CNS to produce estrogenic actions in neural circuits that modulate physiology and behavior.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Estradiol/análogos & derivados , Lordose/tratamento farmacológico , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Tamoxifeno/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Estradiol/farmacologia , Feminino , Fulvestranto , Masculino , Ratos , Ratos Long-Evans , Estimulação Química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA