Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Mais filtros

Medicinas Complementares
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(3)2022 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-35163194

RESUMO

Good sleep quality is essential for maintaining the body's attention during wakefulness, which is easily affected by external factors such as an ambient temperature. However, the mechanism by which an ambient temperature influences sleep-wake behaviors remains unclear. The dorsomedial hypothalamus (DMH) has been reported to be involved in thermoregulation. It also receives projection from the preoptic area, which is an important region for sleep and energy homeostasis and the suprachiasmatic nucleus-a main control area of the clock rhythm. Therefore, we hypothesized that the DMH plays an important role in the regulation of sleep related to ambient temperatures. In this study, we found that cold exposure (24/20/16/12 °C) increased wakefulness and decreased non-rapid eye movement (NREM) sleep, while warm exposure (32/36/40/44 °C) increased NREM sleep and decreased wakefulness compared to 28 °C conditions in wild-type mice. Then, using non-specific and specific apoptosis, we found that lesions of whole DMH neurons and DMH γ-aminobutyric acid (GABA)-ergic neurons induced by caspase-3 virus aggravated the fluctuation of core body temperature after warm exposure and attenuated the change in sleep-wake behaviors during cold and warm exposure. However, chemogenetic activation or inhibition of DMH GABAergic neurons did not affect the sleep-wake cycle. Collectively, our findings reveal an essential role of DMH GABAergic neurons in the regulation of sleep-wake behaviors elicited by a change in ambient temperature.


Assuntos
Neurônios GABAérgicos/metabolismo , Hipotálamo/metabolismo , Sono/fisiologia , Animais , Regulação da Temperatura Corporal/fisiologia , Temperatura Baixa , Núcleo Hipotalâmico Dorsomedial , Neurônios GABAérgicos/fisiologia , Temperatura Alta , Hipotálamo Médio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Qualidade do Sono , Sono REM , Temperatura , Vigília/fisiologia
2.
Mol Metab ; 6(3): 306-312, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28271037

RESUMO

OBJECTIVE: Central cholinergic neural circuits play a role in the regulation of feeding behavior. The dorsomedial hypothalamus (DMH) is considered the appetite-stimulating center and contains cholinergic neurons. Here, we study the role of DMH cholinergic neurons in the control of food intake. METHODS: To selectively stimulate DMH cholinergic neurons, we expressed stimulatory designer receptors exclusively activated by designer drugs (DREADDs) and channelrhodopsins in DMH cholinergic neurons by injection of adeno-associated virus (AAV) vectors into the DMH of choline acetyltransferase (ChAT)-IRES-Cre mice. We also generated transgenic mice expressing channelrhodopsins in cholinergic neurons with the Cre-LoxP technique. To delete the Chat gene exclusively in the DMH, we injected an AAV carrying a Cre recombinase transgene into the DMH of floxed ChAT mice. Food intake was measured with and without selective stimulation of DMH cholinergic neurons. RESULTS: Mice lacking the Chat gene in the DMH show reduced body weight as compared to control. Chemogenetic activation of DMH cholinergic neurons promotes food intake. This orexigenic effect is further supported by experiments of optogenetic stimulation of DMH cholinergic neurons. DMH cholinergic neurons innervate pro-opiomelanocortin neurons in the arcuate nucleus of the hypothalamus (ARC). Treatment with acetylcholine (ACh) enhances GABAergic inhibitory transmission to ARC POMC neurons that is blocked by the muscarinic receptor antagonist. Direct activation of cholinergic fibers in the ARC readily stimulates food intake that is also abolished by the muscarinic receptor antagonist. CONCLUSION: ACh released from DMH cholinergic neurons regulates food intake and body weight. This effect is mediated in part through regulation of ARC POMC neurons. Activation of muscarinic receptors on GABAergic axon terminals enhances inhibitory tone to ARC POMC neurons. Hence, this novel DMHACh â†’ ARCPOMC pathway plays an important role in the control of food intake and body weight.


Assuntos
Regulação do Apetite/fisiologia , Neurônios Colinérgicos/fisiologia , Hipotálamo Médio/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/metabolismo , Dependovirus/genética , Núcleo Hipotalâmico Dorsomedial/metabolismo , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Hipotálamo Médio/metabolismo , Integrases , Leptina/genética , Masculino , Camundongos , Camundongos Transgênicos , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
3.
J Neurosci ; 33(49): 19051-9, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24305803

RESUMO

Release of gonadotropin releasing hormone (GnRH) from the medial basal hypothalamus (MBH)/median eminence region (S-ME) is essential for normal reproductive function. GnRH release is profoundly regulated by the negative and positive feedback effects of ovarian estradiol (E2). Here we report that neuroestradiol, released in the S-ME, also directly influences GnRH release in ovariectomized female monkeys, in which the ovarian source of E2 is removed. We found that (1) brief infusion of E2 benzoate (EB) to the S-ME rapidly stimulated release of GnRH and E2 in the S-ME of ovariectomized monkeys, (2) electrical stimulation of the MBH resulted in GnRH release as well as E2 release, and (3) direct infusion of an aromatase inhibitor to the S-ME suppressed spontaneous GnRH release as well as the EB-induced release of GnRH and E2. These findings reveal the importance of neuroestradiol as a neurotransmitter in regulation of GnRH release. How circulating ovarian E2 interacts with hypothalamic neuroestrogens in the control of GnRH release remains to be investigated.


Assuntos
Estradiol/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Animais , Inibidores da Aromatase/farmacologia , Cromatografia Líquida de Alta Pressão , Estimulação Elétrica , Eletrodos Implantados , Estradiol/farmacologia , Feminino , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Letrozol , Macaca mulatta , Espectrometria de Massas , Eminência Mediana/efeitos dos fármacos , Eminência Mediana/metabolismo , Microdiálise , Nitrilas/farmacologia , Ovariectomia , Radioimunoensaio , Triazóis/farmacologia
4.
Curr Top Dev Biol ; 106: 49-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24290347

RESUMO

The medial hypothalamus is composed of nuclei of the tuberal hypothalamus, the paraventricular nucleus of the anterior hypothalamus, and the neurohypophysis. Its arrangement, around the third ventricle of the brain, above the adenohypophysis, and in direct contact with the vasculature, means that it serves as an interface with circulating systems, providing a key conduit through which the brain can sample, and control, peripheral body systems. Through these interfaces, and interactions with other parts of the brain, the medial hypothalamus centrally governs diverse homeostatic processes, including energy and fluid balance, stress responses, growth, and reproductive behaviors. Here, we summarize recent studies that reveal how the diverse cell types within the medial hypothalamus are assembled in an integrated manner to enable its later function. In particular, we discuss how the temporally protracted operation of signaling pathways and transcription factors governs the appearance and regionalization of the hypothalamic primordium from the prosencephalic territory, the specification and differentiation of progenitors into neurons in organized nuclei, and the establishment of interfaces. Through analyses of mouse, chick, and zebrafish, a picture emerges of an evolutionarily conserved and highly coordinated developmental program. Early indications suggest that deregulation of this program may underlie complex human pathological conditions and dysfunctional behaviors, including stress and eating disorders.


Assuntos
Hipotálamo Médio/fisiologia , Hipotálamo/fisiologia , Neuro-Hipófise/fisiologia , Transdução de Sinais/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Hipotálamo Médio/crescimento & desenvolvimento , Hipotálamo Médio/metabolismo , Modelos Biológicos , Neurogênese/genética , Neurogênese/fisiologia , Neuro-Hipófise/crescimento & desenvolvimento , Neuro-Hipófise/metabolismo , Transdução de Sinais/genética
5.
Endocrinology ; 154(9): 3130-40, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23751875

RESUMO

Chronic inflammation is involved in the pathogenesis of obesity and type 2 diabetes. Recently teasaponin, an extract from tea, has been shown to have antiinflammatory effects. We examined the effect of teasaponin on obesity, inflammation, glucose metabolism, and central leptin sensitivity in obese mice fed a high-fat (HF) diet for 16 weeks. Intraperitoneal injections of teasaponin (10 mg/kg, daily) for 21 days significantly decreased the food intake and body weight of HF diet-induced obese mice. Teasaponin treatment also reduced the protein levels of proinflammatory cytokines (TNF-α, IL-6, and/or IL-1ß) and nuclear factor-κB signaling (phosphorylated inhibitory-κB kinase and phosphorylated inhibitory-κBα) in adipose tissue and the liver. The antiinflammatory effects of teasaponin were associated with improved glycemic status in the treated animals, evidenced by improved glucose tolerance, homeostasis model assessment, and fasting plasma insulin. In the hypothalamus, teasaponin decreased both proinflammatory cytokines and inflammatory signaling in the mediobasal hypothalamus. Teasaponin treatment also enhanced the anorexigenic effect of central leptin administration, restored leptin phosphorylated signal transducer and activator of transcription-3 (p-STAT3) signaling in the arcuate nucleus, and increased hypothalamic expression of the anorexigenic peptide proopiomelanocortin. These results identify a potential novel application for teasaponin as an antiobesity and antiinflammatory agent.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Camellia sinensis/química , Hipotálamo Médio/efeitos dos fármacos , Leptina/metabolismo , Obesidade/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Chá/química , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Depressores do Apetite/administração & dosagem , Depressores do Apetite/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Hipotálamo Médio/imunologia , Hipotálamo Médio/metabolismo , Injeções Intraperitoneais , Resistência à Insulina , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Obesidade/etiologia , Obesidade/imunologia , Obesidade/metabolismo , Extratos Vegetais/administração & dosagem , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/metabolismo , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos
6.
Biol Reprod ; 87(6): 129, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23034157

RESUMO

Reproductive function is highly dynamic during postnatal developmental. Here, we performed molecular profiling of gene expression patterns in the hypothalamus of developing male and female rats to identify which genes are sexually dimorphic, to gain insight into a more complex network of hypothalamic genes, and to ascertain dynamic changes in their relationships with one another and with sex steroid hormones during development. Using a low-density PCR platform, we quantified mRNA levels in the preoptic area (POA) and medial basal hypothalamus (MBH), and assayed circulating estradiol, testosterone, and progesterone at six ages from birth through adulthood. Numerous genes underwent developmental change, particularly postnatal increases, decreases, or peaks/plateaus at puberty. Surprisingly, there were few sex differences; only Esr1, Kiss1, and Tac2 were dimorphic (higher in females). Cluster analysis of gene expression revealed sexually dimorphic correlations in the POA but not the MBH from P30 (Postnatal Day 30) to P60. Hormone measurements showed few sex differences in developmental profiles of estradiol; higher levels of progesterone in females only after P30; and a developmental pattern of testosterone with a nadir at P30 followed by a dramatic increase through P60 (males). Furthermore, bionetwork analysis revealed that hypothalamic gene expression profiles and their relationships to hormones undergo dynamic developmental changes that differ considerably from adults. These data underscore the importance of developmental stage in considering the effects of hormones on the regulation of neuroendocrine genes in the hypothalamus. Moreover, the finding that few neuroendocrine genes are sexually dimorphic highlights the need to consider postnatal development from a network approach that allows assessment of interactions and patterns of expression.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neurogênese , Neurocinina B/metabolismo , Maturidade Sexual , Animais , Animais Recém-Nascidos , Estradiol/sangue , Receptor alfa de Estrogênio/genética , Feminino , Perfilação da Expressão Gênica , Hipotálamo/crescimento & desenvolvimento , Hipotálamo Médio/crescimento & desenvolvimento , Hipotálamo Médio/metabolismo , Kisspeptinas/genética , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurocinina B/genética , Neurônios/metabolismo , Área Pré-Óptica/crescimento & desenvolvimento , Área Pré-Óptica/metabolismo , Progesterona/sangue , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Testosterona/sangue
7.
Science ; 334(6059): 1133-7, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22116886

RESUMO

Evolutionarily old and conserved homeostatic systems in the brain, including the hypothalamus, are organized into nuclear structures of heterogeneous and diverse neuron populations. To investigate whether such circuits can be functionally reconstituted by synaptic integration of similarly diverse populations of neurons, we generated physically chimeric hypothalami by microtransplanting small numbers of embryonic enhanced green fluorescent protein-expressing, leptin-responsive hypothalamic cells into hypothalami of postnatal leptin receptor-deficient (db/db) mice that develop morbid obesity. Donor neurons differentiated and integrated as four distinct hypothalamic neuron subtypes, formed functional excitatory and inhibitory synapses, partially restored leptin responsiveness, and ameliorated hyperglycemia and obesity in db/db mice. These experiments serve as a proof of concept that transplanted neurons can functionally reconstitute complex neuronal circuitry in the mammalian brain.


Assuntos
Hipotálamo Médio/citologia , Hipotálamo Médio/fisiopatologia , Hipotálamo/citologia , Leptina/metabolismo , Neurônios/fisiologia , Neurônios/transplante , Obesidade/fisiopatologia , Obesidade/terapia , Receptores para Leptina/metabolismo , Animais , Glicemia/análise , Peso Corporal , Forma Celular , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores , Glucose/administração & dosagem , Hipotálamo/metabolismo , Hipotálamo Médio/metabolismo , Potenciais Pós-Sinápticos Inibidores , Insulina/administração & dosagem , Insulina/sangue , Leptina/administração & dosagem , Potenciais da Membrana , Camundongos , Camundongos Obesos , Neurogênese , Neurônios/citologia , Obesidade/metabolismo , Transdução de Sinais , Transmissão Sináptica
8.
Am J Physiol Regul Integr Comp Physiol ; 295(5): R1446-54, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18753263

RESUMO

A limiting factor to the clinical management of diabetes is iatrogenic hypoglycemia. With multiple hypoglycemic episodes, the collective neuroendocrine response that restores euglycemia is impaired. In our animal model of recurrent hypoglycemia (RH), neuroendocrine deficits are accompanied by a decrease in medial hypothalamic activation. Here we tested the hypothesis that the medial hypothalamus may exhibit unique changes in the expression of regulatory proteins in response to RH. We report that expression of the immediate early gene FosB is increased in medial hypothalamic nuclei, anterior hypothalamus, and posterior paraventricular nucleus of the thalamus (THPVN) of the thalamus following RH. We identified the hypothalamic PVN, a key autonomic output site, among the regions expressing FosB. To identify the subtype(s) of neuronal populations that express FosB, we screened candidate neuropeptides of the PVN for coexpression using dual fluorescence immunohistochemistry. Among the neuropeptides analyzed [including oxytocin, vasopressin, thyrotropin-releasing hormone, and corticotropin-releasing factor (CRF)], FosB was only identified in CRF-positive neurons. Inhibitory gamma-aminobutyric acid-positive processes appear to impinge on these FosB-expressing neurons. Finally, we observed a significant decrease in the presynaptic marker synaptophysin within the PVN of RH-treated vs. saline-treated rats, suggesting that rapid alterations of synaptic morphology may occur in association with RH. Collectively, these data suggest that RH stress triggers cellular changes that support synaptic plasticity, in specific neuroanatomical sites, which may contribute to the development of hypoglycemia-associated autonomic failure.


Assuntos
Hipoglicemia/metabolismo , Hipotálamo/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Sinaptofisina/biossíntese , Hormônio Adrenocorticotrópico/metabolismo , Animais , Glicemia/metabolismo , Western Blotting , Epinefrina/metabolismo , Imunofluorescência , Glucagon/metabolismo , Hidrocortisona/metabolismo , Hipotálamo Médio/metabolismo , Imuno-Histoquímica , Masculino , Neurônios/fisiologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Wistar , Recidiva , Hormônio Liberador de Tireotropina/metabolismo , Vasopressinas/metabolismo , Ácido gama-Aminobutírico/metabolismo
9.
Ideggyogy Sz ; 60(3-4): 192-5, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17451067

RESUMO

Induction of Fos, a proto-oncogene c-fos protein product, was immunohistochemically examined in the rat hypothalamic neurons 3 h after a single (1 x120 min) or repeated (7x 120 min) immobilization (IMO) stress. The aim of the present study was to reveal a possible parallelism in the cell activation between the medial and lateral hypothalamic neurons, especially between the stress responsive neurons in the hypothalamic paraventricular nucleus (PVN) and hypocretin (Hcrt) synthesizing neurons, i.e. suspected stress active neurons of the lateral hypothalamus. After IMO, the animals were perfused and their brains processed with immunohistochemistry for Fos or Fos/Hcrt proteins. Acute IMO elicited extensive Fos expression in both the examined areas. Excessive Fos expression was mainly seen in the PVN, while Hcrt neurons failed to show a broad response (appr. 5%) to single IMO. Clear occurrence of Fos signal was also seen in both hypothalamic areas of IMO-habituated rats. However, in these animals, in both areas examined, the number of Fos neurons was considerably suppressed, including the PVN. These results indicate that IMO is able to evoke a concurrent activation of Fos in many medial and lateral hypothalamic neurons. However, the scanty response of Hcrt neurons to acute IMO does not allow to assort them to a distinct IMO stress-responsive neuronal phenotypes of the brain.


Assuntos
Hipotálamo/metabolismo , Imobilização , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Psicológico/metabolismo , Animais , Região Hipotalâmica Lateral/metabolismo , Hipotálamo Médio/metabolismo , Imuno-Histoquímica , Masculino , Neurotransmissores/metabolismo , Orexinas , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Brain Res ; 1070(1): 45-55, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16403465

RESUMO

Severe blood loss lowers arterial pressure through a central mechanism that is thought to include opioid neurons. In this study, we investigated whether hemorrhage activates proopiomelanocortin (POMC) neurons by measuring Fos immunoreactivity and POMC mRNA levels in the medial basal hypothalamus. Hemorrhage (2.2 ml/100 g body weight over 20 min) increased the number of Fos immunoreactive neurons throughout the rostral-caudal extent of the arcuate nucleus, the retrochiasmatic area and the peri-arcuate region lateral to the arcuate nucleus where POMC neurons are located. Double label immunohistochemistry revealed that hemorrhage increased Fos expression by beta-endorphin immunoreactive neurons significantly. The proportion of beta-endorphin immunoreactive neurons that expressed Fos immunoreactivity increased approximately four-fold, from 11.7+/-1.4% in sham-operated control animals to 42.0+/-5.2% in hemorrhaged animals. Hemorrhage also increased POMC mRNA levels in the medial basal hypothalamus significantly, consistent with the hypothesis that blood loss activates POMC neurons. To test whether activation of arcuate neurons contributes to the fall in arterial pressure evoked by hemorrhage, we inhibited neuronal activity in the caudal arcuate nucleus by microinjecting the local anesthetic lidocaine (2%; 0.1 or 0.3 microl) bilaterally 2 min before hemorrhage was initiated. Lidocaine injection inhibited hemorrhagic hypotension and bradycardia significantly although it did not influence arterial pressure or heart rate in non-hemorrhaged rats. These results demonstrate that hemorrhage activates POMC neurons and provide evidence that activation of neurons in the arcuate nucleus plays an important role in the hemodynamic response to hemorrhage.


Assuntos
Hemorragia/fisiopatologia , Hipotálamo/fisiopatologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Bradicardia/etiologia , Bradicardia/prevenção & controle , Hemorragia/complicações , Hemorragia/metabolismo , Hipotensão/etiologia , Hipotensão/prevenção & controle , Hipotálamo/metabolismo , Hipotálamo Médio/metabolismo , Imuno-Histoquímica , Injeções , Lidocaína/administração & dosagem , Lidocaína/farmacologia , Masculino , Pró-Opiomelanocortina/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , beta-Endorfina/metabolismo
11.
Brain Res ; 1070(1): 15-23, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16405927

RESUMO

Opioid peptides exert an inhibitory effect on hypothalamic gonadotropin releasing hormone (GnRH) secretion mainly by interacting with mu-opioid receptors. Although a direct role for opioids via delta-opioid receptors (DORs) has been suggested, the presence of these receptors on GnRH neurons has never been demonstrated. In the present study, we determined the distribution of DORs in the basal hypothalamus of rat with special focus on their relation to GnRH neurons. Double-labelling immunofluorescence and confocal microscopy revealed that DORs are exclusively present in a subpopulation of GnRH nerve terminals, with the highest density in the external layer of the median eminence. We then studied the functional characteristics of DORs in an immortalized GnRH-secreting neuronal cell line (GT1-1) known to endogenously express this receptor. Here, pertussis toxin pretreatment abolished the delta-agonist (DPDPE) inhibitory effect on cAMP accumulation. We also analyzed the type of G proteins involved in the signal transduced by the DOR and showed that GT1-1 cells express the inhibitory Go and Gi2 alpha-subunits. However, only Go was down-regulated under chronic DPDPE exposure. Finally, since DOR is expressed postnatally in brain, we compared GnRH neuronal cells immortalized at different developmental stages (the more mature GT1-1 and GT1-7 cells, versus the more immature GN11 cells), evidencing that only mature neurons express DOR. In conclusion, our study indicates that a direct control of opioids via delta-receptors occurs on GnRH neurons and validates the use of GT1 cells to further investigate the nature of the DOR present on GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptores Opioides delta/metabolismo , Animais , Linhagem Celular Transformada , Senescência Celular , AMP Cíclico/antagonistas & inibidores , Regulação para Baixo , D-Penicilina (2,5)-Encefalina/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Hipotálamo/citologia , Hipotálamo Médio/citologia , Hipotálamo Médio/metabolismo , Terminações Nervosas/metabolismo , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
12.
J Neuroendocrinol ; 17(10): 639-48, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16159376

RESUMO

A secondary surge of prolactin has been recently characterised on the afternoon of oestrus. Because the noradrenergic nucleus locus coeruleus participates in the genesis of the pro-oestrous and steroid-induced surges of prolactin, the aim of the present study was to investigate the importance of locus coeruleus norepinephrine in the generation of the prolactin surge of oestrus. For this purpose, we initially re-evaluated the profile of prolactin secretion during the oestrous cycle to verify whether this surge of prolactin was physiological and specific to the day of oestrus. Thereafter, the following were evaluated: (i) the effect of locus coeruleus lesion on the secondary surge of prolactin and on norepinephrine concentration in the medial preoptic area (MPOA), medial basal hypothalamus (MBH) and paraventricular nucleus (PVN) during the day of oestrus and (ii) locus coeruleus neurones activity during the same day by Fos immunoreactivity. Locus coeruleus lesion completely blocked the prolactin surge of oestrus in all rats studied and also significantly reduced norepinephrine concentration in the MPOA, MBH and PVN during the day of oestrus. The number of double-labelled tyrosine hydroxylase/Fos immunoreactive neurones in locus coeruleus was significantly higher at 14.00 h of oestrus, suggesting an increase in its activity preceding the prolactin surge that generally occurs at 15.00 h. Therefore, the increase in locus coeruleus activity on the afternoon of oestrus supports the data obtained with bilateral lesion of this nucleus, suggesting a stimulatory role of locus coeruleus norepinephrine in the genesis of the secondary surge of prolactin.


Assuntos
Estro/sangue , Hipotálamo/metabolismo , Locus Cerúleo/metabolismo , Norepinefrina/metabolismo , Prolactina/sangue , Animais , Estro/fisiologia , Feminino , Hipotálamo Médio/metabolismo , Imuno-Histoquímica , Locus Cerúleo/citologia , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Área Pré-Óptica/metabolismo , Prolactina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Tirosina 3-Mono-Oxigenase/metabolismo
13.
Endocrinology ; 145(4): 1794-801, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14670985

RESUMO

In vitro studies using immortalized GT1 cells suggest that hypothalamic astrocytes employ TGFbeta(1) to directly regulate the secretion of GnRH, the neurohormone that controls sexual maturation and adult reproductive function. However, whether such astrocyte-GnRH neuron signaling occurs in vivo is not clear. In the present study, we used in situ hybridization and immunohistochemistry to determine whether astrocytes and GnRH neurons express the molecular components necessary to set in motion communication processes involving TGFbeta(1) signaling. Double-labeling experiments showed that astrocytes in the male rat preoptic region (POA) expressed TGFbeta(1) mRNA and that GnRH perikarya were often found in close association with TGFbeta(1) mRNA-expressing cells. In addition, GnRH neuronal cell bodies in the POA expressed both type II TGFbeta receptors (TGFbeta-RII), which selectively bind TGFbeta, and Smad2/3, one of the primary transducers of TGFbeta signaling, suggesting that they are fully capable of responding directly to TGFbeta(1) stimulation. Consistent with this hypothesis, incubation of POA explants with TGFbeta(1) caused a significant, dose-dependent decrease in GnRH mRNA expression in individual neurons. This effect was observed within 1 h after TGFbeta(1)-treatment and was inhibited by addition of the soluble form of TGFbeta-RII to the incubation medium. In contrast, whereas both TGFbeta(1) and TGFbeta-RII mRNAs were abundantly expressed in both glial cells and capillaries in the median eminence, the projection field of GnRH neurons, TGFbeta-RII immunoreactivity was mainly restricted to the processes of tanycytes and did not colocalize with GnRH-immunoreactive fibers. This observation supports previous in vivo studies showing that TGFbeta(1) is unable to directly modulate decapeptide release from GnRH nerve terminals. Thus, astrocyte-derived TGFbeta(1) may directly influence GnRH expression and/or secretion in vivo by acting on the perikarya, but not the terminals, of GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Eminência Mediana/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Astrócitos/metabolismo , Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/citologia , Hipotálamo Médio/metabolismo , Masculino , Neurônios/metabolismo , Fenótipo , Área Pré-Óptica/metabolismo , Proteínas Serina-Treonina Quinases , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1
14.
Neurosignals ; 12(2): 95-102, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12876404

RESUMO

The neuroendocrine sequelae of acute or chronic superior cervical ganglionectomy in control or pituitary-grafted rats were studied by analyzing both plasma prolactin, growth hormone (GH) and ACTH levels, and taurine (TAU) content in the hypophysiotropic area of the hypothalamus or the median eminence. As expected, after either acute or chronic ganglionectomy, norepinephrine (NE) content decreased in the brain areas studied, although the values remained higher in hyperprolactinemic rats. TAU content was differentially modified by acute vs. chronic surgeries, thus indicating the possible existence of hypothalamic interactions between TAU and NE to regulate pituitary hormone secretion. Indeed, associated differential changes in plasma prolactin, GH and ACTH levels may be due to the observed TAU changes. As expected, pituitary grafting increased plasma prolactin, GH and ACTH levels, so that the presence of a pituitary graft differentially interferes with the effects of either surgery not only on TAU content but also on the plasma levels of the hormone studied. Globally, ongoing studies confirm the differential effects of acute and chronic superior cervical ganglionectomy on plasma prolactin, GH and ACTH levels, and provide new evidence about its effects on TAU content in the hypophysiotropic area of the hypothalamus and the median eminence that may partially explain the changes observed in the pituitary hormones studied.


Assuntos
Hiperprolactinemia/metabolismo , Hipotálamo/metabolismo , Gânglio Cervical Superior/cirurgia , Taurina/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Núcleo Hipotalâmico Anterior/metabolismo , Ganglionectomia , Hormônio do Crescimento/sangue , Hipotálamo Médio/metabolismo , Masculino , Eminência Mediana/metabolismo , Prolactina/sangue , Ratos , Ratos Wistar , Gânglio Cervical Superior/metabolismo
15.
Endocrinology ; 144(5): 2034-45, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12697712

RESUMO

Reproductive development in vertebrates is controlled by changes in hypothalamic GnRH neurons and their inputs from other neurons and glia. One factor involved in the regulation of the GnRH system is the neurotrophic factor, IGF-1. To better understand the regulation of GnRH neurons by hypothalamic IGF-1, we quantified levels of IGF-1 mRNA in hypothalamic and preoptic regions containing GnRH cells, studied the effects of IGF-1 on GnRH gene expression, and examined the neuroanatomical relationship between GnRH neurons and hypothalamic IGF-1 in neonatal, peripubertal, and reproductively mature mice. Our results indicated that IGF-1 mRNA levels in the preoptic area and anterior hypothalamus peaked at postnatal day (P) 5, decreased through P20, and then increased through peripubertal and adult development. Second, IGF-1 had stimulatory effects on GnRH gene expression in explanted preoptic area-anterior hypothalamuses of P5 and peripubertal mice, with results varying by sex and duration of treatment. In contrast, IGF-1 had no effect or even inhibited GnRH gene expression in adult P60 mice. Third, GnRH perikarya coexpressed IGF-1, and this increased throughout sexual maturation. Taken together, the results suggest that IGF-1 can modulate GnRH neurons, that the sensitivity of GnRH neurons to IGF-1 changes developmentally, and that GnRH cells coexpress IGF-1.


Assuntos
Envelhecimento/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Neurônios/metabolismo , Animais , Núcleo Hipotalâmico Anterior/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hipotálamo Médio/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Eminência Mediana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Área Pré-Óptica/metabolismo , RNA Mensageiro/metabolismo
16.
J Reprod Dev ; 49(1): 55-60, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14967949

RESUMO

The distribution and morphology of immunoreactive estrogen receptor alpha (ERalpha)- containing cells were examined in the cattle brain. The brains were collected from two cows and a male calf. One of the cows was sacrificed one week after parturition. ERalpha expressions in the preoptic area of the rostral forebrain and the medial basal hypothalamic area were evaluated immunohistochemically using a mouse monoclonal antibody. In all animals, the signals that indicate ERalpha were detected in the medial preoptic area, the level of the organum vasculosum of the lamina terminalis and the bed nucleus of the stria terminalis. In the caudal hypothalamic region, they were detected in the arcuate nucleus, the periventricular and ventromedial hypothalamic nuclei. ERalpha-containing cells were characterized by a dense nuclear reaction product. In all the subjects examined, ERalpha signals were clearly detected in the cytoplasm as well. The density of ERalpha expression was different among the three cattle; the number of ERalpha containing cells in the postpartum cow was much lower than that in the other cow and the male calf. The present findings suggest that ERalpha expression in the forebrain is influenced by the reproductive status in the cattle.


Assuntos
Hipotálamo Médio/metabolismo , Área Pré-Óptica/metabolismo , Receptores de Estrogênio/biossíntese , Animais , Peso Corporal , Encéfalo/metabolismo , Bovinos , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Receptor alfa de Estrogênio , Estrogênios/metabolismo , Feminino , Hipotálamo/metabolismo , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Estrutura Terciária de Proteína , Receptores de Estrogênio/metabolismo
17.
Endocrinology ; 143(12): 4683-92, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12446596

RESUMO

Leptin regulates food intake in adult mammals by stimulating hypothalamic anorexigenic pathways and inhibiting orexigenic ones. In developing rodents, fat stores are low, yet circulating leptin levels are high and do not appear to regulate food intake. We determined whether two appetite-related neuropeptides [neuropeptide Y (NPY) and proopiomelanocortin (POMC)] and food intake behavior are sensitive to leptin [3 mg/kg body weight (BW), ip] in neonates. We measured the effects of 1) acute leptin administration (3 mg/kg BW, ip, 3 h before testing) on food intake on postnatal day (PND) 5, 8, and 10; and 2) chronic leptin treatment (3 mg/kg BW, ip, daily PND3-PND10) on BW gain and fat pads weight on PND10. In addition to hypothalamic POMC and NPY expression, we determined the expression of suppressor of cytokine signaling-3, all subtypes of leptin receptors, and corticotropin-releasing factor receptor-2 mRNA in PND10 pups receiving either an acute (PND10) or a chronic (PND 3-10) leptin (3 mg/kg BW, ip) or vehicle treatment. Brains were removed 30 or 120 min after the last injection. Acute leptin administration did not affect food intake at any age tested. Chronic leptin treatment did not change BW but decreased fat pad weight significantly. In the arcuate nucleus (ARC), acute leptin increased SOCS-3 and POMC mRNA levels, but decreased NPY mRNA levels in the rostral part of ARC. Chronic leptin down-regulated all subtypes of leptin receptors mRNA and decreased NPY mRNA levels in the caudal ARC but had no further effect on POMC expression. Chronic leptin increased corticotropin-releasing factor receptor-2 mRNA levels in the ventromedial hypothalamus. We conclude that despite adult-like effects of leptin on POMC, NPY, and CRFR-2 expression in neonates, leptin does not regulate food intake during early development.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Apetite/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/metabolismo , Leptina/farmacologia , Neuropeptídeos/genética , Proteínas Repressoras , Fatores de Transcrição , Tecido Adiposo/crescimento & desenvolvimento , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Leptina/administração & dosagem , Neuropeptídeo Y/genética , Tamanho do Órgão/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Proteínas/genética , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores para Leptina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Aumento de Peso/efeitos dos fármacos
18.
Brain Res ; 895(1-2): 129-38, 2001 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-11259769

RESUMO

Using in vitro quantitative autoradiography and [3H]flunitrazepam we examined the rostrocaudal distribution of benzodiazepine binding sites in the human neonate/infant hypothalamus. The autoradiographic analysis shows the presence of a heterogeneous distribution throughout the rostrocaudal extent of this brain structure. High [3H]flunitrazepam binding corresponds primarily to the diagonal band of Broca and the preoptic region. The labelling in the preoptic region showed a rostrocaudal increase, contrasting in that with the other hypothalamic structures. Intermediate densities were present in the septohypothalamic, suprachiasmatic, periventricular and paraventricular nuclei as well as in the mammillary complex. Low binding was observed in the other hypothalamic structures. The benzodiazepine binding sites analyzed belong mostly to type II receptors. In an attempt to unravel possible differences related to age, we compared the autoradiographic distribution in three postnatal age ranges. The topographical distribution of these binding sites was almost identical in each period analyzed. We found, however, that benzodiazepine binding is generally low in the neonatal period and a tendency in increasing densities is observed during development. Taken together, these results provide evidence for a large distribution of benzodiazepine binding sites in neonate/infant hypothalamus, suggesting their implication in the development of this brain structure and the maintenance of its various functions.


Assuntos
Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Fatores Etários , Núcleo Hipotalâmico Anterior/citologia , Núcleo Hipotalâmico Anterior/crescimento & desenvolvimento , Núcleo Hipotalâmico Anterior/metabolismo , Ansiolíticos/farmacologia , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Feminino , Flunitrazepam/farmacocinética , Humanos , Hipotálamo/citologia , Hipotálamo Médio/citologia , Hipotálamo Médio/crescimento & desenvolvimento , Hipotálamo Médio/metabolismo , Hipotálamo Posterior/citologia , Hipotálamo Posterior/crescimento & desenvolvimento , Hipotálamo Posterior/metabolismo , Lactente , Recém-Nascido , Masculino , Neurônios/citologia , Ensaio Radioligante , Trítio/farmacocinética , Ácido gama-Aminobutírico/metabolismo
19.
J Neuroendocrinol ; 12(11): 1077-86, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11069123

RESUMO

During lactation, hypothalamic levels of neuropeptide Y (NPY) and agouti related protein (AGRP) mRNA are increased, while pro-opiomelanocortin (POMC) mRNA is decreased. Serum leptin levels are also decreased during lactation. These changes may underlie the large increases of both food and water intake that occur in concert with milk production. However, additional hypothalamic substances, such as the novel peptide, orexin, may be involved. In addition, in the presence of chronically suppressed levels of serum leptin, there may be a change in leptin receptor expression in the hypothalamus. The objectives of the present study were to determine if orexin and leptin receptor mRNA levels were changed during lactation. Rats were studied on dioestrus of the oestrous cycle or on day 10 postpartum (the lactating animals were suckling eight pups). Orexin mRNA levels in the lateral hypothalamus did not differ between dioestrus and lactation. There was a significant increase in leptin receptor mRNA levels in the supraoptic nucleus during lactation compared to dioestrus. Furthermore, leptin receptor protein, as determined by immunocytochemistry, was colocalized in virtually all vasopressin and oxytocin cells in the supraoptic nucleus. Lactating animals exhibited a decrease in leptin receptor mRNA in the ventromedial hypothalamic nucleus whereas no change was apparent in other hypothalamic areas compared to the dioestrus animals. These results demonstrate that changes in orexin do not appear to contribute to the increase in food intake during lactation. It is likely that the increases in NPY and ARGP, coupled with the decrease in POMC, are primarily responsible for sustaining the chronic hyperphagia of lactation. The changes observed in leptin receptor expression in the hypothalamus, along with the suppression of serum leptin levels, also suggest that the leptin signalling system may play a significant role in the regulation of food and water intake during lactation.


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Lactação/fisiologia , Neuropeptídeos/genética , Receptores de Superfície Celular , Proteína Relacionada com Agouti , Animais , Proteínas de Transporte/análise , Diestro , Ingestão de Alimentos , Feminino , Hipotálamo/química , Hipotálamo Médio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Neuropeptídeo Y/metabolismo , Receptores de Orexina , Orexinas , Ocitocina/análise , Pró-Opiomelanocortina/metabolismo , Proteínas/metabolismo , Sondas RNA , RNA Mensageiro/metabolismo , Ratos , Receptores Acoplados a Proteínas G , Receptores para Leptina , Receptores de Neuropeptídeos , Núcleo Supraóptico/química , Distribuição Tecidual , Vasopressinas/análise
20.
J Neuroendocrinol ; 12(9): 899-909, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10971815

RESUMO

Noradrenaline plays a key role in the initiation of ovulation in nonprimate species. A similar noradrenaline role in the primate has not been established experimentally. We utilized the ovariectomized-oestrogen-supplemented (OVX + E) rhesus macaque to examine the effects of intravenous (i.v.) infusion of oestradiol-17beta (E2) on the activity of the brain noradrenaline system. Experiment 1 established the induction of a preovulatory surge-like release of luteinizing hormone in OVX + E monkeys by i.v. infusion of E2 (OVX + E + E2). In experiment 2, a marked increase in hypothalamic microdialysate noradrenaline concentrations occurred after identical E2 infusion into the OVX + E monkeys that were used in experiment 1. In experiment 3, tyrosine hydroxylase (TH) mRNA expression in the locus coeruleus of the brainstem increased at various times after E2 infusion as determined by semiquantitative in situ hybridization. The amount of TH mRNA in OVX + E + E2 animals was higher (P < 0.05) than that in either the OVX + E or OVX monkeys; no difference was found in the latter two groups. Moreover, selected locus coeruleus sections from E2-infused monkeys were examined for the localization of oestrogen receptors (ER) by in situ hybridization. Both ER-alpha and ER-beta mRNAs were expressed in the locus coeruleus, although the expression was greater for ER-alpha than for ER-beta. We conclude that i.v. infusion of E2, which induces a preovulatory surge-like release of LH, stimulates brain noradrenaline activity; this enhanced activity likely involves an ER-mediated process and is reflected by hypothalamic noradrenaline release and locus coeruleus TH mRNA expression. The results support the concept that noradrenaline can influence the E2-stimulated ovulation in nonhuman primates and that the brainstem is one of the components in this neuroendocrine process.


Assuntos
Tronco Encefálico/enzimologia , Estradiol/farmacologia , Hipotálamo Médio/metabolismo , Norepinefrina/metabolismo , Ovariectomia , Tirosina 3-Mono-Oxigenase/genética , Animais , Estradiol/administração & dosagem , Feminino , Expressão Gênica/efeitos dos fármacos , Hipotálamo Médio/efeitos dos fármacos , Hibridização In Situ , Infusões Intravenosas , Locus Cerúleo/enzimologia , Hormônio Luteinizante/metabolismo , Macaca mulatta , RNA Mensageiro/análise , Receptores de Estrogênio/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA