Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 5(3): e9783, 2010 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-20339544

RESUMO

BACKGROUND: Diurnal variations in the incidence of events such as heart attack and stroke suggest a role for circadian rhythms in the etiology of cardiovascular disease. The aim of this study was to assess the influence of the suprachiasmatic nucleus (SCN) circadian clock on cardiovascular function. METHODOLOGY/PRINCIPAL FINDINGS: Heart rate (HR), blood pressure (BP) and locomotor activity (LA) were measured in circadian mutant (Vipr2(-/-)) mice and wild type littermates, using implanted radio-telemetry devices. Sleep and wakefulness were studied in similar mice implanted with electroencephalograph (EEG) electrodes. There was less diurnal variation in the frequency and duration of bouts of rest/activity and sleep/wake in Vipr2(-/-) mice than in wild type (WT) and short "ultradian" episodes of arousal were more prominent, especially in constant conditions (DD). Activity was an important determinant of circadian variation in BP and HR in animals of both genotypes; altered timing of episodes of activity and rest (as well as sleep and wakefulness) across the day accounted for most of the difference between Vipr2(-/-) mice and WT. However, there was also a modest circadian rhythm of resting HR and BP that was independent of LA. CONCLUSIONS/SIGNIFICANCE: If appropriate methods of analysis are used that take into account sleep and locomotor activity level, mice are a good model for understanding the contribution of circadian timing to cardiovascular function. Future studies of the influence of sleep and wakefulness on cardiovascular physiology may help to explain accumulating evidence linking disrupted sleep with cardiovascular disease in man.


Assuntos
Ritmo Circadiano , Frequência Cardíaca/fisiologia , Núcleo Supraquiasmático/metabolismo , Animais , Comportamento Animal , Pressão Sanguínea , Fenômenos Fisiológicos Cardiovasculares , Eletroencefalografia/métodos , Hemodinâmica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Movimento
2.
Stem Cells ; 27(10): 2539-51, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19650041

RESUMO

The controlled production of neurons in the postnatal dentate gyrus and throughout life is important for hippocampal learning and memory. The mechanisms underlying the necessary coupling of neuronal activity to neural stem/progenitor cell (NSPC) function remain poorly understood. Within the dentate subgranular stem cell niche, local interneurons appear to play an important part in this excitation-neurogenesis coupling via GABAergic transmission, which promotes neuronal differentiation and integration. Here we show that vasoactive intestinal polypeptide, a neuropeptide coreleased with GABA under specific firing conditions, is uniquely trophic for proliferating postnatal nestin-positive dentate NSPCs, mediated via the VPAC(2) receptor. We also show that VPAC(2) receptor activation shifts the fate of symmetrically dividing NSPCs toward a nestin-only phenotype, independent of the trophic effect. In contrast, selective VPAC(1) receptor activation shifts NSPC fate toward granule cell neurogenesis without any trophism. We confirm a trophic role for VPAC(2) receptors in vivo, showing reduced progeny survival and dentate neurogenesis in adult Vipr2(-/-) mice. We also show a specific reduction in type 2 nestin-positive precursors in vivo, consistent with a role for VPAC(2) in maintaining this cell population. This work provides the first evidence of differential fate modulation of neurogenesis by neurotransmitter receptor subtypes and extends the fate-determining effects of neurotransmitters to maintaining the nestin-positive pool of NSPCs. This differential receptor effect may support the independent pharmacological manipulation of precursor pool expansion and neurogenic instruction for therapeutic application in the treatment of cognitive deficits associated with a decline in neurogenesis.


Assuntos
Giro Denteado/crescimento & desenvolvimento , Neurônios/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Células-Tronco/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Giro Denteado/citologia , Feminino , Proteínas de Filamentos Intermediários/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fenótipo , Ratos , Ratos Wistar , Receptores Tipo II de Peptídeo Intestinal Vasoativo/agonistas , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/agonistas , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/farmacologia
3.
J Neurosci ; 27(16): 4351-8, 2007 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-17442819

RESUMO

The master clock driving mammalian circadian rhythms is located in the suprachiasmatic nuclei (SCN) of the hypothalamus and entrained by daily light/dark cycles. SCN lesions abolish circadian rhythms of behavior and result in a loss of synchronized circadian rhythms of clock gene expression in peripheral organs (e.g., the liver) and of hormone secretion (e.g., corticosterone). We examined rhythms of behavior, hepatic clock gene expression, and corticosterone secretion in VPAC2 receptor-null (Vipr2-/-) mice, which lack a functional SCN clock. Unexpectedly, although Vipr2-/- mice lacked robust circadian rhythms of wheel-running activity and corticosterone secretion, hepatic clock gene expression was strongly rhythmic, but advanced in phase compared with that in wild-type mice. The timing of food availability is thought to be an important entrainment signal for circadian clocks outside the SCN. Vipr2-/- mice consumed food significantly earlier in the 24 h cycle than wild-type mice, consistent with the observed timing of peripheral rhythms of circadian gene expression. When restricted to feeding only during the daytime (RF), mice develop rhythms of activity and of corticosterone secretion in anticipation of feeding time, thought to be driven by a food-entrainable circadian oscillator, located outside the SCN. Under RF, mice of both genotypes developed food-anticipatory rhythms of activity and corticosterone secretion, and hepatic gene expression rhythms also became synchronized to the RF stimulus. Thus, food intake is an effective zeitgeber capable of coordinating circadian rhythms of behavior, peripheral clock gene expression, and hormone secretion, even in the absence of a functional SCN clock.


Assuntos
Ritmo Circadiano/genética , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Fígado/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia , Animais , Corticosterona/metabolismo , Sinais (Psicologia) , Expressão Gênica , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Núcleo Supraquiasmático/fisiologia
4.
J Neurosci ; 26(35): 8955-64, 2006 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-16943551

RESUMO

A commonly occurring polymorphic variant of the human 5-hydroxytryptamine (5-HT) transporter (5-HTT) gene that increases 5-HTT expression has been associated with reduced anxiety levels in human volunteer and patient populations. However, it is not known whether this linkage between genotype and anxiety relates to variation in 5-HTT expression and consequent changes in 5-HT transmission. Here we test this hypothesis by measuring the neurochemical and behavioral characteristics of a mouse genetically engineered to overexpress the 5-HTT. Transgenic mice overexpressing the human 5-HTT (h5-HTT) were produced from a 500 kb yeast artificial chromosome construct. These transgenic mice showed the presence of h5-HTT mRNA in the midbrain raphe nuclei, as well as a twofold to threefold increase in 5-HTT binding sites in the raphe nuclei and a range of forebrain regions. The transgenic mice had reduced regional brain whole-tissue levels of 5-HT and, in microdialysis experiments, decreased brain extracellular 5-HT, which reversed on administration of the 5-HTT inhibitor paroxetine. Compared with wild-type mice, the transgenic mice exhibited a low-anxiety phenotype in plus maze and hyponeophagia tests. Furthermore, in the plus maze test, the low-anxiety phenotype of the transgenic mice was reversed by acute administration of paroxetine, suggesting a direct link between the behavior, 5-HTT overexpression, and low extracellular 5-HT. In toto, these findings demonstrate that associations between increased 5-HTT expression and anxiety can be modeled in mice and may be specifically mediated by decreases in 5-HT transmission.


Assuntos
Ansiedade/fisiopatologia , Ansiedade/psicologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Serotonina/metabolismo , Transmissão Sináptica , Animais , Ansiedade/genética , Ansiedade/metabolismo , Comportamento Animal , Sítios de Ligação , Encéfalo/metabolismo , Espaço Extracelular/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Fenótipo , RNA Mensageiro/metabolismo , Serotonina/biossíntese , Proteínas da Membrana Plasmática de Transporte de Serotonina/biossíntese
5.
Circ Res ; 97(3): 227-35, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-16002749

RESUMO

Heightened expression of the S100 calcium-binding protein, S100A4/Mts1, is observed in pulmonary vascular disease. Loss of serotonin (5-hydroxytryptamine [5-HT]) receptors or of the serotonin transporter (SERT) attenuates pulmonary hypertension in animals, and polymorphisms causing gain of SERT function are linked to clinical pulmonary vascular disease. Because 5-HT induces release of S100beta, we investigated the codependence of 5-HT receptors and SERT in regulating S100A4/Mts1 in human pulmonary artery smooth muscle cells (hPA-SMC). 5-HT elevated S100A4/Mts1 mRNA levels and increased S100A4/Mts1 protein in hPA-SMC lysates and culture media. S100A4/Mts1 in the culture media stimulated proliferation and migration of hPA-SMC in a manner dependent on the receptor for advanced glycation end products. Treatment with SB224289 (selective antagonist of 5-HT1B), fluoxetine (SERT inhibitor), SERT RNA-interference, and iproniazid (monoamine oxidase-A inhibitor), blocked 5-HT-induced S100A4/Mts1. 5-HT signaling mediated phosphorylation (p) of extracellular signal-regulated kinase 1/2 (pERK1/2), but pERK1/2 nuclear translocation depended on SERT, monoamine oxidase activity, and reactive oxygen species. Nuclear translocation of pERK1/2 was required for pGATA-4-mediated transcription of S100A4/Mts1. These data provide evidence for a mechanistic link between the 5-HT pathway and S100A4/Mts1 in pulmonary hypertension and explain how the 5-HT1B receptor and SERT are codependent in regulating S100A4/Mts1.


Assuntos
Hipertensão Pulmonar/etiologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptor 5-HT1B de Serotonina/fisiologia , Proteínas S100/genética , Transporte Ativo do Núcleo Celular , Movimento Celular , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição GATA4 , Produtos Finais de Glicação Avançada/farmacologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monoaminoxidase/fisiologia , Músculo Liso Vascular/citologia , Fosforilação , Artéria Pulmonar/citologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Serotonina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Serotonina , Transdução de Sinais , Fatores de Transcrição/metabolismo
6.
Eur J Neurosci ; 19(8): 2201-11, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15090046

RESUMO

Circadian rhythms in mammals depend on the properties of cells in the suprachiasmatic nucleus (SCN). The retino-recipient core of the mouse SCN is characterized by vasoactive intestinal peptide (VIP) neurons. Expression within the SCN of VPAC2, a VIP receptor, is required for circadian rhythmicity. Using transgenic mice with beta-galactosidase as a marker for VPAC2, we have phenotyped VPAC2-expressing cells within the SCN and investigated expression of the VPAC2 marker at sites previously shown to receive VIP-containing SCN efferents. In situ hybridization and immunohistochemistry demonstrated identical distributions for VPAC2 mRNA and beta-galactosidase and coexpression of the two signals in the SCN. Double-label confocal immunofluorescence identified beta-galactosidase in 32% of the VIP and 31% of the calretinin neurons in the SCN core. Of the arginine-vasopressin neurons that characterize the SCN shell, 45% expressed beta-galactosidase. In contrast, this marker was not apparent in astrocytes within the SCN core or shell. Cell bodies containing beta-galactosidase were detected at sites reportedly receiving VIP-containing SCN efferents, including the subparaventricular zone and lateral septum and the anteroventral periventricular, preoptic suprachiasmatic, medial preoptic and paraventricular hypothalamic nuclei. The detection of a marker for VPAC2 expression in the SCN in almost one-third of the VIP and calretinin core neurons and nearly half of the arginine-vasopressin shell neurons and also in cell bodies at sites receiving VIP-immunoreactive projections from the SCN indicates that VPAC2 may contribute to autoregulation and/or coupling within the SCN core and to the control of the SCN shell and sites distal to this nucleus.


Assuntos
Neurônios/metabolismo , Fenótipo , Receptores de Peptídeo Intestinal Vasoativo/biossíntese , Núcleo Supraquiasmático/metabolismo , Animais , Vias Eferentes/química , Vias Eferentes/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Neurônios/química , Receptores de Peptídeo Intestinal Vasoativo/análise , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo , Núcleo Supraquiasmático/química
7.
Endocrinology ; 145(3): 1203-10, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14617572

RESUMO

The neuropeptide vasoactive intestinal peptide (VIP) exerts its actions through two structurally related G protein-coupled receptors (VPAC(1) and VPAC(2)). Pituitary adenylate cyclase-activating polypeptide (PACAP) is also a potent agonist of VPAC(1) and VPAC(2) receptors as well as of a third, PACAP-specific receptor (PAC(1)). We report here the distribution of the VPAC(2) receptor in peripheral tissues of the mouse, determined by receptor autoradiography using [(125)I]VIP and the selective VPAC(2) receptor agonist [(125)I]Ro25-1553 in wild-type and VPAC(2) receptor-null mice. In addition, displacement experiments with the VPAC(2)-selective agonist Ro25-1553 and the VPAC(1)-selective agonist [K(15),R(16),L(27)]VIP(1-7)/GRF(8-27) were performed using the universal radioligand [(125)I]VIP. The VPAC(2) receptor is found predominantly in smooth muscle (in blood vessels and in the smooth muscle layers of the gastrointestinal and reproductive systems), the basal part of the mucosal epithelium in the colon, lung, the vasculature of the kidney, adrenal medulla, and retina. Unexpectedly, the receptor was also present in thyroid follicular cells and acinar cells of the pancreas, tissues that have not been found to express the receptor in other species, and in very large amounts in the lung. Our data suggest novel functions of the VPAC(2) receptor and additional potential therapeutic uses of drugs acting at the receptor (including the treatment of erectile dysfunction), but our results also indicate that caution should be exercised in using the mouse as an animal model for the evaluation of VIP analogs intended for diagnostic or therapeutic use in man.


Assuntos
Epitélio/metabolismo , Músculo Liso/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/análogos & derivados , Animais , Autorradiografia , Ligação Competitiva , Vasos Sanguíneos/metabolismo , Sistema Digestório/metabolismo , Sistema Endócrino/metabolismo , Sistema Linfático/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Ensaio Radioligante , Receptores de Peptídeo Intestinal Vasoativo/agonistas , Receptores Tipo II de Peptídeo Intestinal Vasoativo , Sistema Urogenital/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/farmacocinética , Peptídeo Intestinal Vasoativo/farmacologia
8.
Eur J Neurosci ; 17(2): 197-204, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12542655

RESUMO

Expression of coherent and rhythmic circadian (approximately 24 h) variation of behaviour, metabolism and other physiological processes in mammals is governed by a dominant biological clock located in the hypothalamic suprachiasmatic nuclei (SCN). Photic entrainment of the SCN circadian clock is mediated, in part, by vasoactive intestinal polypeptide (VIP) acting through the VPAC2 receptor. Here we used mice lacking the VPAC2 receptor (Vipr2-/-) to examine the contribution of this receptor to the electrophysiological actions of VIP on SCN neurons, and to the generation of SCN electrical firing rate rhythms SCN in vitro. Compared with wild-type controls, fewer SCN cells from Vipr2-/- mice responded to VIP and the VPAC2 receptor-selective agonist Ro 25-1553. By contrast, similar proportions of Vipr2-/- and wild-type SCN cells responded to gastrin-releasing peptide, arginine vasopressin or N-methyl-D-aspartate. Moreover, VIP-evoked responses from control SCN neurons were attenuated by the selective VPAC2 receptor antagonist PG 99-465. In firing rate rhythm experiments, the midday peak in activity observed in control SCN cells was lost in Vipr2-/- mice. The loss of electrical activity rhythm in Vipr2-/- mice was mimicked in control SCN slices by chronic treatment with PG 99-465. These results demonstrate that the VPAC2 receptor is necessary for the major part of the electrophysiological actions of VIP on SCN cells in vitro, and is of fundamental importance for the rhythmic and coherent expression of circadian rhythms governed by the SCN clock. These findings suggest a novel role of VPAC2 receptor signalling, and of cell-to-cell communication in general, in the maintenance of core clock function in mammals, impacting on the cellular physiology of SCN neurons.


Assuntos
Ritmo Circadiano/fisiologia , Neurônios/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/análogos & derivados , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Arginina Vasopressina/farmacologia , Eletrofisiologia , Peptídeo Liberador de Gastrina/farmacologia , Masculino , Camundongos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Peptídeos Cíclicos/farmacologia , Receptores de Peptídeo Intestinal Vasoativo/agonistas , Receptores de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Receptores de Peptídeo Intestinal Vasoativo/deficiência , Receptores Tipo II de Peptídeo Intestinal Vasoativo , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia
9.
Cell ; 109(4): 497-508, 2002 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-12086606

RESUMO

The neuropeptides pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are implicated in the photic entrainment of circadian rhythms in the suprachiasmatic nuclei (SCN). We now report that mice carrying a null mutation of the VPAC(2) receptor for VIP and PACAP (Vipr2(-/-)) are incapable of sustaining normal circadian rhythms of rest/activity behavior. These mice also fail to exhibit circadian expression of the core clock genes mPer1, mPer2, and mCry1 and the clock-controlled gene arginine vasopressin (AVP) in the SCN. Moreover, the mutants fail to show acute induction of mPer1 and mPer2 by nocturnal illumination. This study highlights the role of intercellular neuropeptidergic signaling in maintenance of circadian function within the SCN.


Assuntos
Ritmo Circadiano/genética , Proteínas de Drosophila , Proteínas do Olho , Regulação da Expressão Gênica/genética , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Células Fotorreceptoras de Invertebrados , Receptores de Peptídeo Intestinal Vasoativo/deficiência , Núcleo Supraquiasmático/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Fatores de Transcrição ARNTL , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Relógios Biológicos/genética , Proteínas de Ciclo Celular , Criptocromos , Flavoproteínas/genética , Flavoproteínas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Mutação/genética , Neurofisinas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peptídeo PHI/metabolismo , Proteínas Circadianas Period , Estimulação Luminosa , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo , Núcleo Supraquiasmático/fisiopatologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...