Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 234
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ann Neurol ; 91(2): 282-288, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34981555

RESUMO

Narcolepsy type 1 (NT1) is a chronic sleep disorder correlated with loss of hypocretin(orexin). In NT1 post-mortem brains, we observed 88% reduction in corticotropin-releasing hormone (CRH)-positive neurons in the paraventricular nucleus (PVN) and significantly less CRH-positive fibers in the median eminence, whereas CRH-neurons in the locus coeruleus and thalamus, and other PVN neuronal populations were spared: that is, vasopressin, oxytocin, tyrosine hydroxylase, and thyrotropin releasing hormone-expressing neurons. Other hypothalamic cell groups, that is, the suprachiasmatic, ventrolateral preoptic, infundibular, and supraoptic nuclei and nucleus basalis of Meynert, were unaffected. The surprising selective decrease in CRH-neurons provide novel targets for diagnostics and therapeutic interventions. ANN NEUROL 2022;91:282-288.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Hipotálamo/metabolismo , Hipotálamo/patologia , Narcolepsia/patologia , Neurônios/patologia , Idoso , Idoso de 80 Anos ou mais , Contagem de Células , Feminino , Humanos , Hipotálamo/diagnóstico por imagem , Imuno-Histoquímica , Locus Cerúleo/citologia , Locus Cerúleo/diagnóstico por imagem , Locus Cerúleo/metabolismo , Masculino , Eminência Mediana/citologia , Eminência Mediana/diagnóstico por imagem , Eminência Mediana/metabolismo , Pessoa de Meia-Idade , Narcolepsia/diagnóstico por imagem , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/diagnóstico por imagem , Núcleo Hipotalâmico Paraventricular/metabolismo
2.
Cell Rep ; 37(7): 109997, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788630

RESUMO

The anorexigenic effect of serotonergic compounds has largely been attributed to activation of serotonin 2C receptors (Htr2cs). Using mouse genetic models in which Htr2c can be selectively deleted or restored (in Htr2c-null mice), we investigate the role of Htr2c in forebrain Sim1 neurons. Unexpectedly, we find that Htr2c acts in these neurons to promote food intake and counteract the anorectic effect of serotonergic appetite suppressants. Furthermore, Htr2c marks a subset of Sim1 neurons in the paraventricular nucleus of the hypothalamus (PVH). Chemogenetic activation of these neurons in adult mice suppresses hunger, whereas their silencing promotes feeding. In support of an orexigenic role of PVH Htr2c, whole-cell patch-clamp experiments demonstrate that activation of Htr2c inhibits PVH neurons. Intriguingly, this inhibition is due to Gαi/o-dependent activation of ATP-sensitive K+ conductance, a mechanism of action not identified previously in the mammalian nervous system.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo , Animais , Anorexia , Depressores do Apetite/metabolismo , Depressores do Apetite/farmacologia , Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Fome/fisiologia , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Potássio/metabolismo , Receptor 5-HT2C de Serotonina/genética , Serotonina/metabolismo , Serotonina/farmacologia , Serotoninérgicos
3.
Genes Brain Behav ; 20(8): e12775, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34672092

RESUMO

The endocannabinoid system is an important regulator of the hormonal and behavioral stress responses, which critically involve corticotropin-releasing factor (CRF) and its receptors. While it has been shown that CRF and the cannabinoid type 1 (CB1) receptor are co-localized in several brain regions, the physiological relevance of this co-expression remains unclear. Using double in situ hybridization, we confirmed co-localization in the piriform cortex, the lateral hypothalamic area, the paraventricular nucleus, and the Barrington's nucleus, albeit at low levels. To study the behavioral and physiological implications of this co-expression, we generated a conditional knockout mouse line that selectively lacks the expression of CB1 receptors in CRF neurons. We found no effects on fear and anxiety-related behaviors under basal conditions nor after a traumatic experience. Additionally, plasma corticosterone levels were unaffected at baseline and after restraint stress. Only acoustic startle responses were significantly enhanced in male, but not female, knockout mice. Taken together, the consequences of depleting CB1 in CRF-positive neurons caused a confined hyperarousal phenotype in a sex-dependent manner. The current results suggest that the important interplay between the central endocannabinoid and CRF systems in regulating the organism's stress response is predominantly taking place at the level of CRF receptor-expressing neurons.


Assuntos
Receptor CB1 de Canabinoide/metabolismo , Reflexo de Sobressalto/genética , Estimulação Acústica , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Córtex Piriforme/citologia , Córtex Piriforme/metabolismo , Receptor CB1 de Canabinoide/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Sexo
4.
Dev Cell ; 56(9): 1326-1345.e6, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33887203

RESUMO

The interplay between hypothalamic neurons and microglia as they integrate stressors to regulate homeostasis is of growing interest. We asked if microglia in the embryonic hypothalamus were likewise stress responsive and, if so, whether their precocious activation perturbs nearby neural stem cell (NSC) programs. We performed single-cell transcriptomics to define embryonic hypothalamic microglia heterogeneity and identified four microglial subsets, including a subpopulation adjacent to NSCs that was responsive to gestational cold stress. Stress exposure elevated CCL3 and CCL4 secretion, but only in male brains, and ex vivo CCL4 treatment of hypothalamic NSCs altered proliferation and differentiation. Concomitantly, gestational stress decreased PVN oxytocin neurons only in male embryos, which was reversed by microglia depletion. Adult offspring exposed to gestational stress displayed altered social behaviors, which was likewise microglia dependent, but only in males. Collectively, immature hypothalamic microglia play an unappreciated role in translating maternal stressors to sexually dimorphic perturbation of neurodevelopmental programs.


Assuntos
Embrião de Mamíferos/citologia , Microglia/citologia , Células-Tronco Neurais/citologia , Estresse Fisiológico , Animais , Comportamento Animal , Contagem de Células , Diferenciação Celular/genética , Proliferação de Células/genética , Temperatura Baixa , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/citologia , Masculino , Camundongos , Microglia/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Oligodendroglia/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Caracteres Sexuais , Análise de Célula Única , Comportamento Social , Esferoides Celulares/citologia
5.
J Reprod Dev ; 65(2): 129-137, 2019 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-30662010

RESUMO

Hindbrain ependymocytes are postulated to have a glucose-sensing role in regulating gonadal functions. Previous studies have suggested that malnutrition-induced suppression of gonadotropin secretion is mediated by noradrenergic inputs from the A2 region in the solitary tract nucleus to the paraventricular nucleus (PVN), and by corticotropin-releasing hormone (CRH) release in the hypothalamus. However, no morphological evidence to indicate the neural pathway from the hindbrain ependymocytes to hypothalamic kisspeptin neurons, a center for reproductive function in mammals, currently exists. The present study aimed to examine the existence of a neuronal pathway from the hindbrain ependymocytes to kisspeptin neurons in the arcuate nucleus (ARC) and anteroventral periventricular nucleus (AVPV). To determine this, wheat-germ agglutinin (WGA), a trans-synaptic tracer, was injected into the fourth ventricle (4V) in heterozygous Kiss1-tandem dimer Tomato (tdTomato) rats, where kisspeptin neurons were visualized by tdTomato fluorescence. 48 h after the WGA injection, brain sections were taken from the forebrain, midbrain and hindbrain and subjected to double immunohistochemistry for WGA and dopamine ß-hydroxylase (DBH) or CRH. WGA immunoreactivities were found in vimentin-immunopositive ependymocytes of the 4V and the central canal (CC), but not in the third ventricle. The WGA immunoreactivities were detected in some tdTomato-expressing cells in the ARC and AVPV, DBH-immunopositive cells in the A1-A7 noradrenergic nuclei, and CRH-immunopositive cells in the PVN. These results suggest that the hindbrain ependymocytes have neuronal connections with the kisspeptin neurons, most probably via hindbrain noradrenergic and CRH neurons to relay low energetic signals for regulation of reproduction.


Assuntos
Epêndima , Hipotálamo , Kisspeptinas/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Rombencéfalo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Epêndima/citologia , Epêndima/efeitos dos fármacos , Epêndima/metabolismo , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/genética , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Ovariectomia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Transgênicos , Rombencéfalo/citologia , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/metabolismo , Aglutininas do Germe de Trigo/metabolismo
6.
BMC Res Notes ; 11(1): 852, 2018 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-30509318

RESUMO

OBJECTIVE: In this study, empathy is quantified using a novel social test. Empathy and prosocial behavior are linked to the expression of oxytocin in humans and rodent models. Specifically, prosocial behavior in prairie voles (Microtus ochrogaster) has been linked to the expression of oxytocin in the paraventricular nucleus of the hypothalamus. The animal's behavior was considered empathic if it spends significantly more time attempting to remove a loos fitting restraint (tether) from the stimulus animal than time in contact with a, simultaneously presented, non-social object similar to the tether. The behavioral data was cross-referenced with the number of neurons expressing oxytocin and arginine vasopressin, as well as the density of dopaminergic neurons (identified by the expression of tyrosine hydroxylase), in the paraventricular nucleus of the hypothalamus. These proteins influence empathic behavior in humans, non-human primates, rats, mice, and prairie voles. RESULTS: The consistency between neuroanatomical mechanisms linked to empathy, and the durations of time spent engaging in empathic contact, support the prediction that the empathic contact in this test is a distinct prosocial behavior, lacking prior behavioral training or the naturally occurring ethological relevance of other prosocial behaviors, and is a measure of empathy.


Assuntos
Arginina Vasopressina/genética , Arvicolinae/psicologia , Comportamento Animal/fisiologia , Comportamento Cooperativo , Empatia/fisiologia , Ocitocina/genética , Animais , Arginina Vasopressina/metabolismo , Arvicolinae/fisiologia , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Feminino , Expressão Gênica , Humanos , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Modelos Animais , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
7.
Nat Neurosci ; 21(5): 717-724, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29632359

RESUMO

'Sundowning' in dementia and Alzheimer's disease is characterized by early-evening agitation and aggression. While such periodicity suggests a circadian origin, whether the circadian clock directly regulates aggressive behavior is unknown. We demonstrate that a daily rhythm in aggression propensity in male mice is gated by GABAergic subparaventricular zone (SPZGABA) neurons, the major postsynaptic targets of the central circadian clock, the suprachiasmatic nucleus. Optogenetic mapping revealed that SPZGABA neurons receive input from vasoactive intestinal polypeptide suprachiasmatic nucleus neurons and innervate neurons in the ventrolateral part of the ventromedial hypothalamus (VMH), which is known to regulate aggression. Additionally, VMH-projecting dorsal SPZ neurons are more active during early day than early night, and acute chemogenetic inhibition of SPZGABA transmission phase-dependently increases aggression. Finally, SPZGABA-recipient central VMH neurons directly innervate ventrolateral VMH neurons, and activation of this intra-VMH circuit drove attack behavior. Altogether, we reveal a functional polysynaptic circuit by which the suprachiasmatic nucleus clock regulates aggression.


Assuntos
Agressão/fisiologia , Ritmo Circadiano/fisiologia , Hipotálamo/fisiologia , Vias Neurais/fisiologia , Animais , Mapeamento Encefálico , Corticosterona/sangue , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/citologia , Optogenética , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Ácido gama-Aminobutírico/fisiologia
8.
J Vet Sci ; 19(2): 172-178, 2018 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-29169227

RESUMO

It has been reported that Korean red ginseng (KRG), a valuable and important traditional medicine, has varied effects on the central nervous system, suggesting its activities are complicated. The paraventricular nucleus (PVN) neurons of the hypothalamus has a critical role in stress responses and hormone secretions. Although the action mechanisms of KRG on various cells and systems have been reported, the direct membrane effects of KRG on PVN neurons have not been fully described. In this study, the direct membrane effects of KRG on PVN neuronal activity were investigated by using a perforated patch-clamp in ICR mice. In gramicidin perforated patch-clamp mode, KRG extract (KRGE) induced repeatable depolarization followed by hyperpolarization of PVN neurons. The KRGE-induced responses were concentration- dependent and persisted in the presence of tetrodotoxin, a voltage sensitive Na+ channel blocker. The KRGE-induced responses were suppressed by 6-cyano-7-nitroquinoxaline-2,3-dione (10 µM), a non-N-methyl-D-aspartate (NMDA) glutamate receptor antagonist, but not by picrotoxin, a type A gamma-aminobutyric acid receptor antagonist. The results indicate that KRG activates non-NMDA glutamate receptors of PVN neurons in mice, suggesting that KRG may be a candidate for use in regulation of stress responses by controlling autonomic nervous system and hormone secretion.


Assuntos
Panax , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Extratos Vegetais/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Relação Dose-Resposta a Droga , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Núcleo Hipotalâmico Paraventricular/citologia , Técnicas de Patch-Clamp , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos
9.
Psychoneuroendocrinology ; 86: 73-77, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28917185

RESUMO

Early-life stress (ES) is a risk factor for metabolic disorders (e.g. obesity) with a notoriously higher prevalence in women compared to men. However, mechanisms underlying these effects remain elusive. The development of the hypothalamic feeding and metabolic regulatory circuits occurs mostly in the early sensitive postnatal phase in rodents and is tightly regulated by the metabolic hormones leptin and ghrelin. We have previously demonstrated that chronic ES reduces circulating leptin and alters adipose tissue metabolism early and later in life similarly in both sexes. However, it is unknown whether chronic ES might also affect developmental ghrelin and insulin levels, and if it induces changes in hypothalamic feeding circuits, possibly in a sex-dependent manner. We here show that chronic ES, in the form of exposure to limited nesting and bedding material from postnatal day (P)2 to P9 in mice, affects ghrelin levels differently, depending on the form of ghrelin (acylated vs desacylated), on age (P9 vs P14) and on sex, while insulin levels were similarly increased in both sexes after ES at P9. Even though ghrelin levels were more strongly affected in ES-exposed females, hypothalamic neuropeptide Y (NPY) and agouti-related peptide (AgRP) fiber density at P14 were similarly altered in both sexes by ES. In the paraventricular nucleus of the hypothalamus, both NPY and AgRP fiber density were increased, while in the arcuate nucleus of the hypothalamus, NPY was increased and AgRP unaltered. Additionally, the hypothalamic mRNA expression of ghrelin's receptor (i.e. growth hormone secretagogue receptor) was not affected by ES. Taken together, the specific alterations found in these important regulatory circuits after ES might contribute to an altered energy balance and feeding behavior in adulthood and thereby to an increased vulnerability to develop metabolic disorders.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Grelina/metabolismo , Neuropeptídeo Y/metabolismo , Tecido Adiposo/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Feminino , Grelina/genética , Grelina/farmacologia , Hipotálamo/metabolismo , Insulina/genética , Insulina/metabolismo , Insulina/farmacologia , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/farmacologia , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Fatores Sexuais , Estresse Psicológico/fisiopatologia
10.
Nature ; 543(7645): 385-390, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28273060

RESUMO

Bone has recently emerged as a pleiotropic endocrine organ that secretes at least two hormones, FGF23 and osteocalcin, which regulate kidney function and glucose homeostasis, respectively. These findings have raised the question of whether other bone-derived hormones exist and what their potential functions are. Here we identify, through molecular and genetic analyses in mice, lipocalin 2 (LCN2) as an osteoblast-enriched, secreted protein. Loss- and gain-of-function experiments in mice demonstrate that osteoblast-derived LCN2 maintains glucose homeostasis by inducing insulin secretion and improves glucose tolerance and insulin sensitivity. In addition, osteoblast-derived LCN2 inhibits food intake. LCN2 crosses the blood-brain barrier, binds to the melanocortin 4 receptor (MC4R) in the paraventricular and ventromedial neurons of the hypothalamus and activates an MC4R-dependent anorexigenic (appetite-suppressing) pathway. These results identify LCN2 as a bone-derived hormone with metabolic regulatory effects, which suppresses appetite in a MC4R-dependent manner, and show that the control of appetite is an endocrine function of bone.


Assuntos
Regulação do Apetite/fisiologia , Osso e Ossos/metabolismo , Lipocalina-2/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Osso e Ossos/citologia , AMP Cíclico/metabolismo , Ingestão de Alimentos/fisiologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Glucose/metabolismo , Homeostase , Hipotálamo/citologia , Hipotálamo/metabolismo , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Masculino , Camundongos , Neurônios/metabolismo , Obesidade/metabolismo , Osteoblastos/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Magreza/metabolismo
11.
Am J Physiol Heart Circ Physiol ; 312(4): H808-H817, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28159808

RESUMO

Orexin neurons, and activation of orexin receptors, are generally thought to be sympathoexcitatory; however, the functional connectivity between orexin neurons and a likely sympathetic target, the hypothalamic spinally projecting neurons (SPNs) in the paraventricular nucleus of the hypothalamus (PVN) has not been established. To test the hypothesis that orexin neurons project directly to SPNs in the PVN, channelrhodopsin-2 (ChR2) was selectively expressed in orexin neurons to enable photoactivation of ChR2-expressing fibers while examining evoked postsynaptic currents in SPNs in rat hypothalamic slices. Selective photoactivation of orexin fibers elicited short-latency postsynaptic currents in all SPNs tested (n = 34). These light-triggered responses were heterogeneous, with a majority being excitatory glutamatergic responses (59%) and a minority of inhibitory GABAergic (35%) and mixed glutamatergic and GABAergic currents (6%). Both glutamatergic and GABAergic responses were present in the presence of tetrodotoxin and 4-aminopyridine, suggesting a monosynaptic connection between orexin neurons and SPNs. In addition to generating postsynaptic responses, photostimulation facilitated action potential firing in SPNs (current clamp configuration). Glutamatergic, but not GABAergic, postsynaptic currents were diminished by application of the orexin receptor antagonist almorexant, indicating orexin release facilitates glutamatergic neurotransmission in this pathway. This work identifies a neuronal circuit by which orexin neurons likely exert sympathoexcitatory control of cardiovascular function.NEW & NOTEWORTHY This is the first study to establish, using innovative optogenetic approaches in a transgenic rat model, that there are robust heterogeneous projections from orexin neurons to paraventricular spinally projecting neurons, including excitatory glutamatergic and inhibitory GABAergic neurotransmission. Endogenous orexin release modulates glutamatergic, but not GABAergic, neurotransmission in these pathways.


Assuntos
Hipotálamo/citologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo , Acetamidas/farmacologia , Potenciais de Ação/fisiologia , Animais , Animais Geneticamente Modificados , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Ácido Glutâmico/metabolismo , Hipotálamo/diagnóstico por imagem , Isoquinolinas/farmacologia , Masculino , Vias Neurais/citologia , Vias Neurais/diagnóstico por imagem , Optogenética , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Núcleo Hipotalâmico Paraventricular/diagnóstico por imagem , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Medula Espinal/diagnóstico por imagem , Ácido gama-Aminobutírico/metabolismo
12.
Anat Rec (Hoboken) ; 300(7): 1307-1314, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27981779

RESUMO

Paraventricular nuclei (PVN) projections to the rostral ventrolateral medulla (RVLM)/C1 catecholaminergic neuron group constitute the pre-autonomic sympathetic center involved in the neural control of systemic cardiovascular function. However, the role of extra-hypothalamic and thalamic dopaminergic (DA) inputs in this circuit remains underexplored. Using retrograde neuroanatomical tracing and high contrast confocal imaging methods, we investigated the projections and morphology of the discrete thalamic DA neuron groups in the dorsal hypothalamic area and their contribution to the PVN-RVLM neural circuit. We found that DA neuron subgroups in the Zona Incerta (Zi; 60%) and Reuniens thalamic nuclei (Re; 40%) were labeled comparably to the PVN (85%) after a retrograde tracer was injected into the RVLM/C1 (P < 0.01 mean ± SEM). The Re/Zi DA neuron subgroups were characterized by angulated cell bodies, superiomedial and inferiomedial projections reaching the contralateral Re/Zi and ipsilateral PVN DA neurons respectively. Ultimately, we deduced that the DA projections of the Re/Zi to the PVN contribute to the PVN-RVLM/C1 neural circuit. As a result of these connections, the Re/Zi DA neuron groups may regulate preautonomic sympathetic events associated with the PVN-RVLM pathway. Anat Rec, 300:1307-1314, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Neurônios Dopaminérgicos/citologia , Bulbo/citologia , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Tálamo/citologia , Animais , Neurônios Dopaminérgicos/metabolismo , Masculino , Bulbo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Tálamo/metabolismo
13.
Front Neural Circuits ; 10: 92, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27932956

RESUMO

The arginine-vasopressin (AVP)-containing hypothalamic magnocellular neurosecretory neurons (VPMNNs) are known for their role in hydro-electrolytic balance control via their projections to the neurohypophysis. Recently, projections from these same neurons to hippocampus, habenula and other brain regions in which vasopressin infusion modulates contingent social and emotionally-affected behaviors, have been reported. Here, we present evidence that VPMNN collaterals also project to the amygdaloid complex, and establish synaptic connections with neurons in central amygdala (CeA). The density of AVP innervation in amygdala was substantially increased in adult rats that had experienced neonatal maternal separation (MS), consistent with our previous observations that MS enhances VPMNN number in the paraventricular (PVN) and supraoptic (SON) nuclei of the hypothalamus. In the CeA, V1a AVP receptor mRNA was only observed in GABAergic neurons, demonstrated by complete co-localization of V1a transcripts in neurons expressing Gad1 and Gad2 transcripts in CeA using the RNAscope method. V1b and V2 receptor mRNAs were not detected, using the same method. Water-deprivation (WD) for 24 h, which increased the metabolic activity of VPMNNs, also increased anxiety-like behavior measured using the elevated plus maze (EPM) test, and this effect was mimicked by bilateral microinfusion of AVP into the CeA. Anxious behavior induced by either WD or AVP infusion was reversed by CeA infusion of V1a antagonist. VPMNNs are thus a newly discovered source of CeA inhibitory circuit modulation, through which both early-life and adult stress coping signals are conveyed from the hypothalamus to the amygdala.


Assuntos
Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Ansiedade/metabolismo , Arginina Vasopressina/metabolismo , Núcleo Central da Amígdala , Glutamato Descarboxilase/metabolismo , Hipotálamo , Neurônios , Receptores de Vasopressinas/metabolismo , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/administração & dosagem , Ansiedade/induzido quimicamente , Comportamento Animal , Núcleo Central da Amígdala/citologia , Núcleo Central da Amígdala/metabolismo , Modelos Animais de Doenças , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Privação Materna , Neurônios/citologia , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Wistar , Núcleo Supraóptico/citologia , Núcleo Supraóptico/metabolismo , Privação de Água
14.
Biol Pharm Bull ; 39(2): 289-94, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26830488

RESUMO

Sickness behavior is a series of behavioral and psychological changes that develop in those stricken with cancers and inflammatory diseases. The etiological mechanism of sickness behavior is not known in detail, and consequently there are no established standard therapies. Kamikihito (KKT), a Kampo (traditional Japanese herbal) medicine composed of 14 herbs, has been used clinically to treat psychiatric dysfunction. Previously, we found that KKT ameliorated sickness behavior in mice inoculated with murine colon 26 adenocarcinoma cells. In this study, we examined the effects of KKT on bacterial endotoxin lipopolysaccharide (LPS)-induced sickness behavior in mice. The administration of LPS caused the emotional aspects of sickness behavior, such as loss of object exploration, social interaction deficit, and depressive-like behavior. LPS also induced mRNA expression for cyclooxygenase (COX)-2, interleukin (IL)-1ß and IL-6, and increased the number of c-Fos immunopositive cells in the hypothalamus and amygdala. KKT ameliorated the behavioral changes and reversed the increases in c-Fos immunopositive cells in the two brain regions, but did not influence the mRNA expression. These results suggest that KKT ameliorates sickness behavior via the suppression of neural activation without anti-inflammatory effects, and that KKT has the potential to treat sickness behavior.


Assuntos
Comportamento Animal/efeitos dos fármacos , Núcleo Central da Amígdala/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Lipopolissacarídeos/toxicidade , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Animais , Núcleo Central da Amígdala/citologia , Comportamento de Doença/efeitos dos fármacos , Comportamento de Doença/fisiologia , Masculino , Camundongos , Núcleo Hipotalâmico Paraventricular/citologia
15.
Nutr Neurosci ; 19(10): 467-474, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25087680

RESUMO

OBJECTIVES: Genistein is a plant-derived estrogenic isoflavone commonly found in dietary and therapeutic supplements, due to its potential health benefits. Growth hormone-releasing hormone (GHRH) and somatostatin (SS) are neurosecretory peptides synthesized in neurons of the hypothalamus and regulate the growth hormone secretion. Early reports indicate that estrogens have highly involved in the regulation of GHRH and SS secretions. Since little is known about the potential effects of genistein on GHRH and SS neurons, we exposed rats to genistein. METHODS: Genistein were administered to adult rats in dose of 30 mg/kg, for 3 weeks. The estradiol-dipropionate treatment was used as the adequate controls to genistein. Using applied stereology on histological sections of hypothalamus, we obtained the quantitative information on arcuate (Arc) and periventricular (Pe) nucleus volume and volume density of GHRH neurons and SS neurons. Image analyses were used to obtain GHRH and SS contents in the median eminence (ME). RESULTS: Administration of estradiol-dipropionate caused the increase of Arc and Pe nucleus volume, SS neuron volume density, GHRH and SS staining intensity in the ME, when compared with control. Genistein treatment increased: Arc nucleus volume and the volume density of GHRH neurons (by 26%) and SS neurons (1.5 fold), accompanied by higher GHRH and SS staining intensity in the ME, when compared to the orhidectomized group. DISCUSSION: These results suggest that genistein has a significant effect on hypothalamic region, involved in the regulation of somatotropic system function, and could contribute to the understanding of genistein as substance that alter the hormonal balance.


Assuntos
Genisteína/farmacologia , Hormônio Liberador de Hormônio do Crescimento/agonistas , Hipotálamo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fitoestrógenos/farmacologia , Somatostatina/agonistas , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Tamanho Celular/efeitos dos fármacos , Suplementos Nutricionais/efeitos adversos , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/administração & dosagem , Estrogênios/efeitos adversos , Estrogênios/farmacologia , Genisteína/administração & dosagem , Genisteína/efeitos adversos , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Injeções Subcutâneas , Masculino , Eminência Mediana/citologia , Eminência Mediana/efeitos dos fármacos , Eminência Mediana/crescimento & desenvolvimento , Eminência Mediana/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Orquiectomia , Tamanho do Órgão/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Núcleo Hipotalâmico Paraventricular/metabolismo , Fitoestrógenos/administração & dosagem , Fitoestrógenos/efeitos adversos , Ratos Wistar , Somatostatina/metabolismo , Técnicas Estereotáxicas
16.
Nature ; 525(7570): 519-22, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26375004

RESUMO

It is commonly assumed, but has rarely been demonstrated, that sex differences in behaviour arise from sexual dimorphism in the underlying neural circuits. Parental care is a complex stereotypic behaviour towards offspring that is shared by numerous species. Mice display profound sex differences in offspring-directed behaviours. At their first encounter, virgin females behave maternally towards alien pups while males will usually ignore the pups or attack them. Here we show that tyrosine hydroxylase (TH)-expressing neurons in the anteroventral periventricular nucleus (AVPV) of the mouse hypothalamus are more numerous in mothers than in virgin females and males, and govern parental behaviours in a sex-specific manner. In females, ablating the AVPV TH(+) neurons impairs maternal behaviour whereas optogenetic stimulation or increased TH expression in these cells enhance maternal care. In males, however, this same neuronal cluster has no effect on parental care but rather suppresses inter-male aggression. Furthermore, optogenetic activation or increased TH expression in the AVPV TH(+) neurons of female mice increases circulating oxytocin, whereas their ablation reduces oxytocin levels. Finally, we show that AVPV TH(+) neurons relay a monosynaptic input to oxytocin-expressing neurons in the paraventricular nucleus. Our findings uncover a previously unknown role for this neuronal population in the control of maternal care and oxytocin secretion, and provide evidence for a causal relationship between sexual dimorphism in the adult brain and sex differences in parental behaviour.


Assuntos
Hipotálamo/citologia , Hipotálamo/fisiologia , Comportamento Materno/fisiologia , Ocitocina/metabolismo , Caracteres Sexuais , Agressão , Animais , Núcleo Hipotalâmico Anterior/citologia , Núcleo Hipotalâmico Anterior/enzimologia , Núcleo Hipotalâmico Anterior/fisiologia , Neurônios Dopaminérgicos/enzimologia , Neurônios Dopaminérgicos/metabolismo , Feminino , Hipotálamo/enzimologia , Masculino , Camundongos , Ocitocina/sangue , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/enzimologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Período Pós-Parto , Sinapses/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
17.
Endocrinology ; 156(12): 4769-80, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26402844

RESUMO

Transgenic mice, including lines targeting corticotropin-releasing factor (CRF or CRH), have been extensively employed to study stress neurobiology. These powerful tools are poised to revolutionize our understanding of the localization and connectivity of CRH-expressing neurons, and the crucial roles of CRH in normal and pathological conditions. Accurate interpretation of studies using cell type-specific transgenic mice vitally depends on congruence between expression of the endogenous peptide and reporter. If reporter expression does not faithfully reproduce native gene expression, then effects of manipulating unintentionally targeted cells may be misattributed. Here, we studied CRH and reporter expression patterns in 3 adult transgenic mice: Crh-IRES-Cre;Ai14 (tdTomato mouse), Crfp3.0CreGFP, and Crh-GFP BAC. We employed the CRH antiserum generated by Vale after validating its specificity using CRH-null mice. We focused the analyses on stress-salient regions, including hypothalamus, amygdala, bed nucleus of the stria terminalis, and hippocampus. Expression patterns of endogenous CRH were consistent among wild-type and transgenic mice. In tdTomato mice, most CRH-expressing neurons coexpressed the reporter, yet the reporter identified a few non-CRH-expressing pyramidal-like cells in hippocampal CA1 and CA3. In Crfp3.0CreGFP mice, coexpression of CRH and the reporter was found in central amygdala and, less commonly, in other evaluated regions. In Crh-GFP BAC mice, the large majority of neurons expressed either CRH or reporter, with little overlap. These data highlight significant diversity in concordant expression of reporter and endogenous CRH among 3 available transgenic mice. These findings should be instrumental in interpreting important scientific findings emerging from the use of these potent neurobiological tools.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/genética , Genes Reporter/genética , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Proteínas de Fluorescência Verde , Hipocampo/metabolismo , Hipotálamo/metabolismo , Sítios Internos de Entrada Ribossomal , Masculino , Camundongos , Camundongos Transgênicos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleos Septais/metabolismo , Transcriptoma
18.
J Neurosci ; 35(29): 10440-50, 2015 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-26203139

RESUMO

The hypothalamus is critical for feeding and body weight regulation. Prevailing studies focus on hypothalamic neurons that are defined by selectively expressing transcription factors or neuropeptides including those expressing proopiomelanocortin (POMC) and agouti-related peptides (AgRP). The Cre expression driven by the pancreas-duodenum homeobox 1 promoter is abundant in several hypothalamic nuclei but not in AgRP or POMC neurons. Using this line, we generated mice with disruption of GABA release from a major subset of non-POMC, non-AgRP GABAergic neurons in the hypothalamus. These mice exhibited a reduction in postweaning feeding and growth, and disrupted hyperphagic responses to NPY. Disruption of GABA release severely diminished GABAergic input to the paraventricular hypothalamic nucleus (PVH). Furthermore, disruption of GABA-A receptor function in the PVH also reduced postweaning feeding and blunted NPY-induced hyperphagia. Given the limited knowledge on postweaning feeding, our results are significant in identifying GABA release from a major subset of less appreciated hypothalamic neurons as a key mediator for postweaning feeding and NPY hyperphagia, and the PVH as one major downstream site that contributes significantly to the GABA action. Significance statement: Prevalent studies on feeding in the hypothalamus focus on well characterized, selective groups neurons [e.g., proopiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons], and as a result, the role of the majority of other hypothalamic neurons is largely neglected. Here, we demonstrated an important role for GABAergic projections from non-POMC non-AgRP neurons to the paraventricular hypothalamic nucleus in promoting postweaning (mainly nocturnal) feeding and mediating NPY-induced hyperphagia. Thus, these results signify an importance to study those yet to be defined hypothalamic neurons in the regulation of energy balance and reveal a neural basis for postweaning (nocturnal) feeding and NPY-mediated hyperphagia.


Assuntos
Comportamento Alimentar/fisiologia , Neurônios GABAérgicos/fisiologia , Hiperfagia/fisiopatologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Proteína Relacionada com Agouti/biossíntese , Animais , Hipotálamo/citologia , Hipotálamo/fisiologia , Hibridização In Situ , Camundongos , Camundongos Mutantes , Neuropeptídeo Y/metabolismo , Técnicas de Cultura de Órgãos , Núcleo Hipotalâmico Paraventricular/citologia , Técnicas de Patch-Clamp , Pró-Opiomelanocortina/biossíntese
19.
Endocrinology ; 156(7): 2713-23, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25942072

RESUMO

Fasting down-regulates the hypothalamus-pituitary-thyroid (HPT) axis activity through a reduction of TRH synthesis in neurons of the parvocellular paraventricular nucleus of the hypothalamus (PVN). These TRH neurons project to the median eminence (ME), where TRH terminals are close to the cytoplasmic extensions of ß2 tanycytes. Tanycytes express pyroglutamyl peptidase II (PPII), the TRH-degrading ectoenzyme that controls the amount of TRH that reaches the anterior pituitary. We tested the hypothesis that regulation of ME PPII activity is another mechanism by which fasting affects the activity of the HPT axis. Semiquantitative in situ hybridization histochemistry data indicated that PPII and deiodinase 2 mRNA levels increased in tanycytes after 48 hours of fasting. This increase was transitory, followed by an increase of PPII activity in the ME, and a partial reversion of the reduction in PVN pro-TRH mRNA levels and the number of TRH neurons detected by immunohistochemistry. In fed animals, adrenalectomy and corticosterone treatment did not change ME PPII activity 72 hours later. Methimazole-induced hypothyroidism produced a profound drop in tanycytes PPII mRNA levels, which was reverted by 3 days of treatment with T4. The activity of thyroliberinase, the serum isoform of PPII, was increased at most fasting time points studied. We conclude that delayed increases in both the ME PPII as well as the thyroliberinase activities in fasted male rats may facilitate the maintenance of the deep down-regulation of the HPT axis function, despite a partial reactivation of TRH expression in the PVN.


Assuntos
Aminopeptidases/genética , Células Ependimogliais/enzimologia , Jejum/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/metabolismo , Eminência Mediana/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , RNA Mensageiro/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Adrenalectomia , Aminopeptidases/efeitos dos fármacos , Aminopeptidases/metabolismo , Animais , Antitireóideos/farmacologia , Corticosterona/farmacologia , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotireoidismo , Iodeto Peroxidase/genética , Masculino , Metimazol/farmacologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Precursores de Proteínas/genética , Ácido Pirrolidonocarboxílico/metabolismo , RNA Mensageiro/efeitos dos fármacos , Ratos , Hormônio Liberador de Tireotropina/efeitos dos fármacos , Hormônio Liberador de Tireotropina/genética , Tiroxina/farmacologia , Iodotironina Desiodinase Tipo II
20.
Bull Exp Biol Med ; 156(6): 736-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24824683

RESUMO

Patterns of expression of TRPM7, the major cellular magnesium transporters in neurons of the hypothalamic region and hippocampus, were studied immunohistochemically. Multidirectional nature and different levels of the expression of the above antigen were revealed during modeled magnesium deficiency with regard to structural and functional features of neuron organization in the hypothalamic paraventricular and supraoptic nuclei as well as hippocampal field CA1 and CA3. Changes in the structural characteristics of neurons in the studied areas (absolute and relative indicators) and TRPM7 expression patterns were quantitatively analyzed considering the data on the role of the studied antigen in magnesium homeostasis, cell damage, and compensation.


Assuntos
Hipocampo/metabolismo , Hipotálamo/metabolismo , Deficiência de Magnésio/metabolismo , Neurônios/metabolismo , Canais de Cátion TRPM/biossíntese , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/metabolismo , Transporte de Íons , Magnésio , Masculino , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Núcleo Supraóptico/citologia , Núcleo Supraóptico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA