Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-34502541

RESUMO

Inhibitor of DNA binding (Id) genes comprise a family of four helix-loop-helix (HLH) transcriptional inhibitors. Our earlier studies revealed a role for ID2 within the circadian system, contributing to input, output, and core clock function through its interaction with CLOCK and BMAL1. Here, we explore the contribution of ID4 to the circadian system using a targeted disruption of the Id4 gene. Attributes of the circadian clock were assessed by monitoring the locomotor activity of Id4-/- mice, and they revealed disturbances in its operation. Id4-mutant mice expressed a shorter circadian period length, attenuated phase shifts in responses to continuous and discrete photic cues, and an advanced phase angle of entrainment under a 12:12 light:dark cycle and under short and long photoperiods. To understand the basis for these properties, suprachiasmatic nucleus (SCN) and retinal structures were examined. Anatomical analysis reveals a smaller Id4-/- SCN in the width dimension, which is a finding consistent with its smaller brain. As a result of this feature, anterograde tracing in Id4-/- mice revealed retinal afferents innovate a disproportionally larger SCN area. The Id4-/- photic entrainment responses are unlikely to be due to an impaired function of the retinal pathways since Id4-/- retinal anatomy and function tested by pupillometry were similar to wild-type mice. Furthermore, these circadian characteristics are opposite to those exhibited by the Id2-/- mouse, suggesting an opposing influence of the ID4 protein within the circadian system; or, the absence of ID4 results in changes in the expression or activity of other members of the Id gene family. Expression analysis of the Id genes within the Id4-/- SCN revealed a time-of-day specific elevated Id1. It is plausible that the increased Id1 and/or absence of ID4 result in changes in interactions with bHLH canonical clock components or with targets upstream and/or downstream of the clock, thereby resulting in abnormal properties of the circadian clock and its entrainment.


Assuntos
Relógios Circadianos/genética , Proteínas Inibidoras de Diferenciação/genética , Proteínas Circadianas Period/genética , Fotoperíodo , Retina/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano , Expressão Gênica , Proteínas Inibidoras de Diferenciação/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Atividade Motora/fisiologia , Proteínas Circadianas Period/metabolismo , Retina/anatomia & histologia , Núcleo Supraquiasmático/anatomia & histologia
2.
FEBS Lett ; 594(17): 2734-2769, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32750151

RESUMO

The circadian system is composed of coupled endogenous oscillators that allow living beings, including humans, to anticipate and adapt to daily changes in their environment. In mammals, circadian clocks form a hierarchically organized network with a 'master clock' located in the suprachiasmatic nucleus of the hypothalamus, which ensures entrainment of subsidiary oscillators to environmental cycles. Robust rhythmicity of body clocks is indispensable for temporally coordinating organ functions, and the disruption or misalignment of circadian rhythms caused for instance by modern lifestyle is strongly associated with various widespread diseases. This review aims to provide a comprehensive overview of our current knowledge about the molecular architecture and system-level organization of mammalian circadian oscillators. Furthermore, we discuss the regulatory roles of peripheral clocks for cell and organ physiology and their implication in the temporal coordination of metabolism in human health and disease. Finally, we summarize methods for assessing circadian rhythmicity in humans.


Assuntos
Proteínas CLOCK/genética , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Doenças Metabólicas/genética , Núcleo Supraquiasmático/fisiologia , Animais , Proteínas CLOCK/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Humanos , Mamíferos , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Fotoperíodo , Transdução de Sinais , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/citologia
3.
Nat Neurosci ; 23(3): 456-467, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32066983

RESUMO

Mammalian circadian behaviors are orchestrated by the suprachiasmatic nucleus (SCN) in the ventral hypothalamus, but the number of SCN cell types and their functional roles remain unclear. We have used single-cell RNA-sequencing to identify the basic cell types in the mouse SCN and to characterize their circadian and light-induced gene expression patterns. We identified eight major cell types, with each type displaying a specific pattern of circadian gene expression. Five SCN neuronal subtypes, each with specific combinations of markers, differ in their spatial distribution, circadian rhythmicity and light responsiveness. Through a complete three-dimensional reconstruction of the mouse SCN at single-cell resolution, we obtained a standardized SCN atlas containing the spatial distribution of these subtypes and gene expression. Furthermore, we observed heterogeneous circadian gene expression between SCN neuron subtypes. Such a spatiotemporal pattern of gene regulation within the SCN may have an important function in the circadian pacemaker.


Assuntos
Expressão Gênica/fisiologia , Neurônios/fisiologia , Análise de Célula Única , Núcleo Supraquiasmático/fisiologia , Animais , Atlas como Assunto , Ritmo Circadiano/fisiologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Expressão Gênica/efeitos da radiação , Regulação da Expressão Gênica/fisiologia , Genômica , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/classificação , Estimulação Luminosa , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/citologia
4.
Endocrinology ; 157(9): 3439-51, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27429160

RESUMO

The suprachiasmatic nucleus (SCN) and arcuate nucleus (ARC) have reciprocal connections; catabolic metabolic information activates the ARC and inhibits SCN neuronal activity. Little is known about the influence of the SCN on the ARC. Here, we investigated whether the SCN modulated the sensitivity of the ARC to catabolic metabolic conditions. ARC neuronal activity, as determined by c-Fos immunoreactivity, was increased after a hypoglycemic stimulus by 2-deoxyglucose (2DG). The highest ARC neuronal activity after 2DG was found at the end of the light period (zeitgeber 11, ZT11) with a lower activity in the beginning of the light period (zeitgeber 2, ZT2), suggesting the involvement of the SCN. The higher activation of ARC neurons after 2DG at ZT11 was associated with higher 2DG induced blood glucose levels as compared with ZT2. Unilateral SCN-lesioned animals, gave a mainly ipsilateral activation of ARC neurons at the lesioned side, suggesting an inhibitory role of the SCN on ARC neurons. The 2DG-induced counterregulatory glucose response correlated with increased ARC neuronal activity and was significantly higher in unilateral SCN-lesioned animals. Finally, the ARC as site where 2DG may, at least partly, induce a counterregulatory response was confirmed by local microdialysis of 2DG. 2DG administration in the ARC produced a higher increase in circulating glucose compared with 2DG administration in surrounding areas such as the ventromedial nucleus of the hypothalamus (VMH). We conclude that the SCN uses neuronal pathways to the ARC to gate sensory metabolic information to the brain, regulating ARC glucose sensitivity and counterregulatory responses to hypoglycemic conditions.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Hipoglicemia/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/anatomia & histologia , Jejum/metabolismo , Masculino , Hormônios Estimuladores de Melanócitos/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Ratos Wistar , Núcleo Supraquiasmático/anatomia & histologia
5.
Neuroimage ; 117: 294-304, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26003859

RESUMO

Diurnal preference (morningness-eveningness) is known to be associated with several individual characteristics that are important in the fields of sociology, education, and psychiatry. Despite this importance, the anatomical correlates of individual differences in morningness-eveningness are unknown, and these were investigated in the present study. We used voxel-based morphometry and a questionnaire to determine individual morningness-eveningness and its association with brain structures in 432 healthy men and 344 healthy women (age, 20.7±1.8years). We demonstrated that morningness (less eveningness) was associated with (a) lower regional gray matter density (rGMD) in the precuneus and adjacent areas, (b) lower rGMD in the left posterior parietal cortex and adjacent areas, and (c) higher rGMD in the bilateral orbitofrontal cortex. Further, our exploratory analyses revealed that (d) higher rGMD in hypothalamic areas around the bilateral suprachiasmatic nuclei were associated with morningness. These findings demonstrate that variations in morningness-eveningness reflect the GM structures of focal regions across the cortex, and suggest a structural basis for individual morningness-eveningness and its association with a wide range of psychological variables distributed across different GM areas of the brain.


Assuntos
Ritmo Circadiano/fisiologia , Substância Cinzenta/anatomia & histologia , Lobo Parietal/anatomia & histologia , Córtex Pré-Frontal/anatomia & histologia , Núcleo Supraquiasmático/anatomia & histologia , Adulto , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Adulto Jovem
6.
J Chem Neuroanat ; 57-58: 42-53, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24727411

RESUMO

The suprachiasmatic nucleus (SCN), which is considered to be the master circadian clock in mammals, establishes biological rhythms of approximately 24 h that several organs exhibit. One aspect relevant to the study of the neurofunctional features of biological rhythmicity is the identification of communication pathways between the SCN and other brain areas. As a result, SCN efferent projections have been investigated in several species, including rodents and a few primates. The fibers originating from the two main intrinsic fiber subpopulations, one producing vasoactive intestinal peptide (VIP) and the other producing arginine vasopressin (AVP), exhibit morphological traits that distinguish them from fibers that originate from other brain areas. This distinction provides a parameter to study SCN efferent projections. In this study, we mapped VIP (VIP-ir) and AVP (AVP-ir) immunoreactive (ir) fibers and endings in the hypothalamus of the primate Sapajus apella via immunohistochemical and morphologic study. Regarding the fiber distribution pattern, AVP-ir and VIP-ir fibers were identified in regions of the tuberal hypothalamic area, retrochiasmatic area, lateral hypothalamic area, and anterior hypothalamic area. VIP-ir and AVP-ir fibers coexisted in several hypothalamic areas; however, AVP-ir fibers were predominant over VIP-ir fibers in the posterior hypothalamus and medial periventricular area. This distribution pattern and the receiving hypothalamic areas of the VIP-ir and AVP-ir fibers, which shared similar morphological features with those found in SCN, were similar to the patterns observed in diurnal and nocturnal animals. This finding supports the conservative nature of this feature among different species. Morphometric analysis of SCN intrinsic neurons indicated homogeneity in the size of VIP-ir neurons in the SCN ventral portion and heterogeneity in the size of two subpopulations of AVP-ir neurons in the SCN dorsal portion. The distribution of fibers and morphometric features of these neuronal populations are described and compared with those of other species in the present study.


Assuntos
Arginina Vasopressina/metabolismo , Vias Eferentes/anatomia & histologia , Hipotálamo/anatomia & histologia , Núcleo Supraquiasmático/anatomia & histologia , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Cebus , Vias Eferentes/metabolismo , Vias Eferentes/ultraestrutura , Hipotálamo/metabolismo , Hipotálamo/ultraestrutura , Imuno-Histoquímica , Masculino , Fibras Nervosas/metabolismo , Fibras Nervosas/fisiologia , Fibras Nervosas/ultraestrutura , Área Pré-Óptica/fisiologia , Área Pré-Óptica/ultraestrutura , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/ultraestrutura
7.
Neuroscience ; 266: 197-207, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24583038

RESUMO

The suprachiasmatic nucleus (SCN) is typically considered our autonomous clock synchronizing behavior with physiological parameters such as blood pressure (BP), just transmitting time independent of physiology. Yet several studies show that the SCN is involved in the etiology of hypertension. Here, we demonstrate that the SCN is incorporated in a neuronal feedback circuit arising from the nucleus tractus solitarius (NTS), modulating cardiovascular reactivity. Tracer injections into the SCN of male Wistar rats revealed retrogradely filled neurons in the caudal NTS, where BP information is integrated. These NTS projections to the SCN were shown to be glutamatergic and to terminate in the ventrolateral part of the SCN where light information also enters. BP elevations not only induced increased neuronal activity as measured by c-Fos in the NTS but also in the SCN. Lesioning the caudal NTS prevented this activation. The increase of SCN neuronal activity by hypertensive stimuli suggested involvement of the SCN in counteracting BP elevations. Examining this possibility we observed that elevation of BP, induced by α1-agonist infusion, was more than twice the magnitude in SCN-lesioned animals as compared to in controls, indicating indeed an active involvement of the SCN in short-term BP regulation. We propose that the SCN receives BP information directly from the NTS enabling it to react to hemodynamic perturbations, suggesting the SCN to be part of a homeostatic circuit adapting BP response. We discuss how these findings could explain why lifestyle conditions violating signals of the biological clock may, in the long-term, result in cardiovascular disease.


Assuntos
Pressão Sanguínea/fisiologia , Vias Neurais/fisiologia , Núcleo Solitário/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Retroalimentação , Imuno-Histoquímica , Masculino , Vias Neurais/anatomia & histologia , Ratos , Ratos Wistar , Núcleo Solitário/anatomia & histologia , Núcleo Supraquiasmático/anatomia & histologia
8.
Exp Neurol ; 243: 4-20, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22766204

RESUMO

The suprachiasmatic nucleus (SCN), site of the primary clock in the circadian rhythm system, has three major afferent connections. The most important consists of a retinohypothalamic projection through which photic information, received by classical rod/cone photoreceptors and intrinsically photoreceptive retinal ganglion cells, gains access to the clock. This information influences phase and period of circadian rhythms. The two other robust afferent projections are the median raphe serotonergic pathway and the geniculohypothalamic (GHT), NPY-containing pathway from the thalamic intergeniculate leaflet (IGL). Beyond this simple framework, the number of anatomical routes that could theoretically be involved in rhythm regulation is enormous, with the SCN projecting to 15 regions and being directly innervated by about 35. If multisynaptic afferents to the SCN are included, the number expands to approximately brain 85 areas providing input to the SCN. The IGL, a known contributor to circadian rhythm regulation, has a still greater level of complexity. This nucleus connects abundantly throughout the brain (to approximately 100 regions) by pathways that are largely bilateral and reciprocal. Few of these sites have been evaluated for their contributions to circadian rhythm regulation, although most have a theoretical possibility of doing so via the GHT. The anatomy of IGL connections suggests that one of its functions may be regulation of eye movements during sleep. Together, neural circuits of the SCN and IGL are complex and interconnected. As yet, few have been tested with respect to their involvement in rhythm regulation.


Assuntos
Ritmo Circadiano/fisiologia , Corpos Geniculados/anatomia & histologia , Corpos Geniculados/fisiologia , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/fisiologia , Animais , Corpos Geniculados/citologia , Humanos , Hipotálamo/anatomia & histologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Vias Neurais/anatomia & histologia , Vias Neurais/citologia , Vias Neurais/fisiologia , Fotoperíodo , Células Fotorreceptoras/citologia , Células Fotorreceptoras/fisiologia , Núcleo Supraquiasmático/citologia
9.
PLoS Comput Biol ; 8(3): e1002419, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22423219

RESUMO

In mammals, the suprachiasmatic nucleus (SCN) of the hypothalamus constitutes the central circadian pacemaker. The SCN receives light signals from the retina and controls peripheral circadian clocks (located in the cortex, the pineal gland, the liver, the kidney, the heart, etc.). This hierarchical organization of the circadian system ensures the proper timing of physiological processes. In each SCN neuron, interconnected transcriptional and translational feedback loops enable the circadian expression of the clock genes. Although all the neurons have the same genotype, the oscillations of individual cells are highly heterogeneous in dispersed cell culture: many cells present damped oscillations and the period of the oscillations varies from cell to cell. In addition, the neurotransmitters that ensure the intercellular coupling, and thereby the synchronization of the cellular rhythms, differ between the two main regions of the SCN. In this work, a mathematical model that accounts for this heterogeneous organization of the SCN is presented and used to study the implication of the SCN network topology on synchronization and entrainment properties. The results show that oscillations with larger amplitude can be obtained with scale-free networks, in contrast to random and local connections. Networks with the small-world property such as the scale-free networks used in this work can adapt faster to a delay or advance in the light/dark cycle (jet lag). Interestingly a certain level of cellular heterogeneity is not detrimental to synchronization performances, but on the contrary helps resynchronization after jet lag. When coupling two networks with different topologies that mimic the two regions of the SCN, efficient filtering of pulse-like perturbations in the entrainment pattern is observed. These results suggest that the complex and heterogeneous architecture of the SCN decreases the sensitivity of the network to short entrainment perturbations while, at the same time, improving its adaptation abilities to long term changes.


Assuntos
Potenciais de Ação/fisiologia , Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Modelos Neurológicos , Rede Nervosa/fisiologia , Neurônios/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Simulação por Computador , Humanos , Rede Nervosa/anatomia & histologia , Núcleo Supraquiasmático/anatomia & histologia , Transmissão Sináptica/fisiologia
10.
World Neurosurg ; 77(2): 342-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22079813

RESUMO

BACKGROUND: The anterolateral approach is one of the main routes for accessing suprachiasmatic lesions involving the anterior communicating artery (AComA) complex. Pterional (PT) craniotomy and its alternatives, including orbitozygomatic, orbitopterional, and mini-supraorbital craniotomies, have been developed as tailored frontotemporal craniotomies. One of the main differences between PT craniotomy and its alternatives is the removal of the orbital bone along with the sphenoid wing. However, which bone part is the most important to remove has not been discussed in relation to frontal lobe retraction. We have evaluated how the removal of the supraorbital bar versus the removal of the lateral orbital wall along with the sphenoid wing affects the relationship between the levels of frontal lobe retraction and area of exposure (AOE) in the suprachiasmatic region. METHODS: We performed three types of craniotomies: PT craniotomy, PT craniotomy with the removal of the supraorbital bar (PT-SO craniotomy), and PT craniotomy with the removal of the lateral orbital wall along with the sphenoid wing, i.e., the frontal process of the zygomatic bone and the orbital and cerebral faces of the greater sphenoid wing (PT-LO-SW craniotomy). For each craniotomy, the AOE around the suprachiasmatic region was measured at four different levels of frontal lobe retraction, namely, 5, 10, 15, and 20 mm, from the cranial base. RESULTS: At 5-mm retraction, PT-LO-SW craniotomy was the only craniotomy in which the AComA complex was visible. At 10-mm retraction, PT-LO-SW craniotomy afforded the greatest AOE among the three craniotomies, and the AOE was significantly greater than that of PT craniotomy (P = 0.025). At 15- and 20-mm retraction, there were no significant differences among the three craniotomies. CONCLUSIONS: Treatment of lesions in the suprachiasmatic region via an anterolateral route involving a frontotemporal craniotomy requires sufficient removal of the lateral orbital wall along with the greater sphenoid wing so that brain retraction is minimized.


Assuntos
Craniotomia/métodos , Osso Frontal/anatomia & histologia , Osso Frontal/cirurgia , Crânio/anatomia & histologia , Crânio/cirurgia , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/cirurgia , Osso Temporal/anatomia & histologia , Osso Temporal/cirurgia , Feminino , Humanos , Aneurisma Intracraniano/cirurgia , Pessoa de Meia-Idade , Artéria Cerebral Média/anatomia & histologia , Artéria Cerebral Média/cirurgia , Órbita/anatomia & histologia , Órbita/cirurgia , Base do Crânio/anatomia & histologia , Base do Crânio/cirurgia , Osso Esfenoide/anatomia & histologia , Osso Esfenoide/cirurgia , Instrumentos Cirúrgicos , Tomografia Computadorizada por Raios X , Zigoma/anatomia & histologia , Zigoma/cirurgia
11.
Brain Struct Funct ; 217(2): 549-76, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21826455

RESUMO

Lungfishes (dipnoans) are currently considered the closest living relatives of tetrapods. The organization of the cholinergic systems in the brain has been carefully analyzed in most vertebrate groups, and major shared characteristics have been described, although traits particular to each vertebrate class have also been found. In the present study, we provide the first detailed information on the distribution of cholinergic cell bodies and fibers in the central nervous system in two representative species of lungfishes, the African lungfish (Protopterus dolloi) and the Australian lungfish (Neoceratodus forsteri), as revealed by immunohistochemistry against the enzyme choline acetyltransferase (ChAT). Distinct groups of ChAT immunoreactive (ChAT-ir) cells were observed in the basal telencephalon, habenula, isthmic nucleus, laterodorsal tegmental nucleus, cranial nerve motor nuclei, and the motor column of the spinal cord, and these groups seem to be highly conserved among vertebrates. In lungfishes, the presence of a cholinergic cell group in the thalamus and the absence of ChAT-ir cells in the tectum are variable traits, unique to this group and appearing several times during evolution. Other characters were observed exclusively in Neoceratodus, such as the presence of cholinergic cells in the suprachiasmatic nucleus, the pretectal region and the superior raphe nucleus. Cholinergic fibers were found in the medial pallium, basal telencephalon, thalamus and prethalamus, optic tectum and interpeduncular nucleus. Comparison of these results with those from other classes of vertebrates, including a segmental analysis to correlate cell populations, reveals that the cholinergic systems in lungfishes largely resemble those of amphibians and other tetrapods.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Peixes/anatomia & histologia , Peixes/metabolismo , Receptores Colinérgicos/metabolismo , Animais , Calbindinas , Colina O-Acetiltransferase/metabolismo , Feminino , Masculino , Proteína G de Ligação ao Cálcio S100/metabolismo , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/metabolismo , Teto do Mesencéfalo/anatomia & histologia , Teto do Mesencéfalo/metabolismo , Telencéfalo/anatomia & histologia , Telencéfalo/metabolismo , Tálamo/anatomia & histologia , Tálamo/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Brain Res ; 1367: 146-61, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-20971082

RESUMO

The phases of many circadian rhythms differ between diurnal and nocturnal species. However, rhythms within the hypothalamic suprachiasmatic nucleus (SCN), which contains the central circadian pacemaker, are very similar, suggesting that the mechanisms underlying phase preference lie downstream of the SCN. Rhythms in Fos expression in the ventral subparaventricular zone (vSPVZ), a major target of the SCN, differ substantially between diurnal Nile grass rats and nocturnal lab rats, raising the possibility that the vSPVZ modulates the effects of SCN signals at its targets. To understand better how and where the SCN and vSPVZ communicate circadian signals within the grass rat brain, we mapped their projections using the anterograde tracer biotinylated dextran amine (BDA). Adult female grass rats received unilateral BDA injections directed at the SCN or vSPVZ and their brains were perfusion-fixed several days later. Immunohistochemistry revealed that the distribution patterns of SCN and vSPVZ efferents were very similar. Labeled fibers originating in each region were heavily concentrated in the medial preoptic area, paraventricular thalamic nucleus, the subparaventricular zone, and the hypothalamic paraventricular and dorsomedial nuclei. BDA-labeled fibers from the SCN and vSPVZ formed appositions with orexin neurons and gonadotropin-releasing hormone neurons, two cell populations whose rhythms in Fos expression track temporally reversed patterns of locomotor and reproductive behavior, respectively, in diurnal and nocturnal rodents. These data demonstrate that projections of the SCN and vSPVZ are highly conserved in diurnal and nocturnal rodents, and the vSPVZ projections may enable it to modulate the responsiveness of target cells to signals from the SCN.


Assuntos
Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleo Supraquiasmático/anatomia & histologia , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Mapeamento Encefálico , Dextranos/metabolismo , Lateralidade Funcional , Hormônio Liberador de Gonadotropina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fibras Nervosas/metabolismo , Fibras Nervosas/fisiologia , Vias Neurais/fisiologia , Neuropeptídeos/metabolismo , Proteínas Oncogênicas v-fos/metabolismo , Orexinas , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Núcleo Supraquiasmático/metabolismo
13.
Eur J Neurosci ; 32(12): 2143-51, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21143668

RESUMO

Circadian rhythms are an essential property of many living organisms, and arise from an internal pacemaker, or clock. In mammals, this clock resides in the suprachiasmatic nucleus (SCN) of the hypothalamus, and generates an intrinsic circadian rhythm that is transmitted to other parts of the CNS. We will review the evidence that basic adaptive functions of the circadian system rely on functional plasticity in the neuronal network organization, and involve a change in phase relation among oscillatory neurons. We will illustrate this for: (i) photic entrainment of the circadian clock to the light-dark cycle; and (ii) seasonal adaptation of the clock to changes in day length. Molecular studies have shown plasticity in the phase relation between the ventral and dorsal SCN during adjustment to a shifted environmental cycle. Seasonal adaptation relies predominantly on plasticity in the phase relation between the rostral and caudal SCN. Electrical activity is integrated in the SCN, and appears to reflect the sum of the differently phased molecular expression patterns. While both photic entrainment and seasonal adaptation arise from a redistribution of SCN oscillatory activity patterns, different neuronal coupling mechanisms are employed, which are reviewed in the present paper.


Assuntos
Adaptação Fisiológica , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Rede Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/fisiologia , Animais , Síndrome do Jet Lag , Rede Nervosa/anatomia & histologia , Neurotransmissores/metabolismo , Fotoperíodo , Estações do Ano
14.
J Comp Neurol ; 518(10): 1837-46, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20235090

RESUMO

We cloned the homologue of the clock gene Bmal from a cephalochordate, Branchiostoma lanceolatum (syn. amphioxus). Amphioxus possesses a single copy of this gene (amphiBmal) that encodes for a protein of 649 amino acids, which is quite similar to BMALs of other chordates. The gene is expressed by a restricted cell group in the anterior vesicle of the neural tube, and its expression site coincides with that of another clock gene, namely, amphiPer. The expression of amphiBmal shows a rhythmic fluctuation that persists under constant darkness and is, thus, circadian. Similar to the situation in craniates, the peak phases of the amphiBmal and amphiPer expression are offset by 12 hours. Based on these observations and the putative homology between the diencephalon of vertebrates and the anterior vesicle of lancelets, we suggest a homology between the suprachiasmatic nucleus of craniates and the amphiBmal/amphiPer-expressing cell group of amphioxus.


Assuntos
Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Relógios Biológicos/genética , Cordados não Vertebrados , Ritmo Circadiano/genética , Regulação da Expressão Gênica , Sistema Nervoso , Fatores de Transcrição ARNTL/classificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Evolução Biológica , Cordados não Vertebrados/anatomia & histologia , Cordados não Vertebrados/genética , Cordados não Vertebrados/metabolismo , Clonagem Molecular , Humanos , Dados de Sequência Molecular , Filogenia , Prosencéfalo/anatomia & histologia , Prosencéfalo/fisiologia , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/fisiologia
15.
Annu Rev Physiol ; 72: 551-77, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20148688

RESUMO

The suprachiasmatic nucleus (SCN) is the primary circadian pacemaker in mammals. Individual SCN neurons in dispersed culture can generate independent circadian oscillations of clock gene expression and neuronal firing. However, SCN rhythmicity depends on sufficient membrane depolarization and levels of intracellular calcium and cAMP. In the intact SCN, cellular oscillations are synchronized and reinforced by rhythmic synaptic input from other cells, resulting in a reproducible topographic pattern of distinct phases and amplitudes specified by SCN circuit organization. The SCN network synchronizes its component cellular oscillators, reinforces their oscillations, responds to light input by altering their phase distribution, increases their robustness to genetic perturbations, and enhances their precision. Thus, even though individual SCN neurons can be cell-autonomous circadian oscillators, neuronal network properties are integral to normal function of the SCN.


Assuntos
Ritmo Circadiano/fisiologia , Rede Nervosa/citologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Núcleo Supraquiasmático/química , Núcleo Supraquiasmático/fisiologia , Animais , Relógios Biológicos/fisiologia , Drosophila , Hormônios/fisiologia , Humanos , Luz , Rede Nervosa/anatomia & histologia , Fotoperíodo , Núcleo Supraquiasmático/anatomia & histologia
16.
Brain Res ; 1320: 34-46, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20096673

RESUMO

In this study, two circadian related centers, the suprachiasmatic nucleus (SCN) and the intergeniculate leaflet (IGL) were evaluated in respect to their cytoarchitecture, retinal afferents and chemical content of major cells and axon terminals in the rock cavy (Kerodon rupestris), a Brazilian rodent species. The rock cavy SCN is innervated in its ventral portion by terminals from the predominantly contralateral retina. It also contains vasopressin, vasoactive intestinal polypeptide and glutamic acid decarboxilase immunoreactive cell bodies and neuropeptide Y, serotonin and enkephalin immunopositive fibers and terminals and is marked by intense glial fibrillary acidic protein immunoreactivity. The IGL receives a predominantly contralateral retinal projection, contains neuropeptide Y and nitric oxide synthase-producing neurons and enkephalin immunopositive terminals and is characterized by dense GFAP immunoreactivity. This is the first report examining the neural circadian system in a crepuscular rodent species for which circadian properties have been described. The results are discussed comparing with what has been described for other species and in the context of the functional significance of these centers.


Assuntos
Corpos Geniculados/anatomia & histologia , Corpos Geniculados/metabolismo , Roedores/anatomia & histologia , Roedores/metabolismo , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Fotomicrografia , Retina/anatomia & histologia , Retina/metabolismo , Vias Visuais/anatomia & histologia , Vias Visuais/metabolismo
17.
J Neurosci Methods ; 187(1): 41-5, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20036282

RESUMO

New imaging technologies have increased our capabilities to resolve three-dimensional structures from microscopic samples. Laser-scanning confocal microscopy is particularly amenable to this task because it allows the researcher to optically section biological samples, creating three-dimensional image volumes. However, a number of problems arise when studying neural tissue samples. These include data set size, physical scanning restrictions, volume registration and display. To deal with these issues, we undertook large-scale confocal scanning microscopy in order to visualize neural networks spanning multiple tissue sections. We demonstrate a technique to create and visualize a three-dimensional digital reconstruction of the hypothalamic arginine vasopressin neuroendocrine system in the male mouse. The generated three-dimensional data included a volume of tissue that measures 4.35 mm x 2.6 mm x 1.4mm with a voxel resolution of 1.2 microm. The dataset matrix included 3508 x 2072 x 700 pixels and was a composite of 19,600 optical sections. Once reconstructed into a single volume, the data is suitable for interactive stereoscopic projection. Stereoscopic imaging provides greater insight and understanding of spatial relationships in neural tissues' inherently three-dimensional structure. This technique provides a model approach for the development of data sets that can provide new and informative volume rendered views of brain structures. This study affirms the value of stereoscopic volume-based visualization in neuroscience research and education, and the feasibility of creating large-scale high resolution interactive three-dimensional reconstructions of neural tissue from microscopic imagery.


Assuntos
Arginina Vasopressina/metabolismo , Hipotálamo/anatomia & histologia , Imageamento Tridimensional/métodos , Microscopia Confocal/métodos , Neurônios/citologia , Animais , Fluorescência , Hipotálamo/citologia , Hipotálamo/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos , Vias Neurais/anatomia & histologia , Vias Neurais/citologia , Vias Neurais/metabolismo , Neurônios/metabolismo , Sistemas Neurossecretores/anatomia & histologia , Sistemas Neurossecretores/citologia , Sistemas Neurossecretores/metabolismo , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo
18.
Eur J Neurosci ; 30(9): 1802-14, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19840112

RESUMO

The circadian clock located within the suprachiasmatic nuclei (SCN) of the hypothalamus responds to changes in the duration of day length, i.e. photoperiod. Recently, changes in phase relationships among the SCN cell subpopulations, especially between the rostral and caudal region, were implicated in the SCN photoperiodic modulation. To date, the effect of abrupt, rectangular, light-to-dark transitions have been studied while in nature organisms experience gradual dawn and twilight transitions. The aim of this study was to compare the effect of a long (18 h of light) and a short (6 h of light) photoperiod with twilight relative to that with rectangular light-to-dark transition on the daily profiles of Per1 and Per2 mRNA (in situ hybridization) and PER1 and PER2 protein (immunohistochemistry) levels within the rostral, middle and caudal regions of the mouse SCN. Under the short but not under the long photoperiod, Per1, Per2 and PER1, PER2 profiles were significantly phase-advanced under the twilight relative to rectangular light-to-dark transition in all SCN regions examined. Under the photoperiods with rectangular light-to-dark transition, Per1 and Per2 mRNA profiles in the caudal SCN were phase-advanced as compared with those in the rostral SCN. The phase differences between the SCN regions were reduced under the long, or completely abolished under the short, photoperiods with twilight. The data indicate that the twilight photoperiod provides stronger synchronization among the individual SCN cell subpopulations than the rectangular one, and the effect is more pronounced under the short than under the long photoperiod.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Proteínas Circadianas Period , Fotoperíodo , Núcleo Supraquiasmático , Animais , Escuridão , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/metabolismo
19.
Curr Biol ; 19(11): R460-2, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19515356

RESUMO

Uncoupling the oscillators in the dorsal and ventral subdivisions of the rat suprachiasmatic nucleus reveals which one of them regulates the circadian rhythm of rapid eye movement sleep.


Assuntos
Ritmo Circadiano , Núcleo Supraquiasmático/fisiologia , Animais , Fotoperíodo , Ratos , Sono REM , Núcleo Supraquiasmático/anatomia & histologia , Terminologia como Assunto
20.
Curr Biol ; 19(10): 848-52, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19375313

RESUMO

Sleep is consistently concentrated to a specific time of the day. Its timing and consolidation depend on the interplay between a homeostatic and a circadian process of sleep regulation [1-3]. Sleep propensity rises as a homeostatic response to increasing wake time, whereas a circadian clock determines the specific time when sleep will probably occur. This two-process regulation of sleep also determines which specific sleep stage will be manifested, and the circadian process governs tightly the manifestation of rapid eye movement sleep (REMS) [1, 4]. The role of the hypothalamic suprachiasmatic nucleus (SCN) in the circadian gating of sleep and wakefulness has been unequivocally established by lesion studies [5], but its role in the timing of specific sleep stages has remained unknown. Using a forced desynchrony paradigm that induces the stable dissociation of the ventrolateral (vl) and dorsomedial (dm) SCN, and a jetlag paradigm that induces desynchronization between these SCN subregions, we show that the SCN can time the occurrence of specific sleep stages. Specifically, the circadian regulation of REMS is associated with clock gene expression within the dmSCN. We provide the first neurophysiological model for the disruption of sleep architecture that may result from temporal challenges such as rotational-shift work and jetlag.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Sono REM/fisiologia , Núcleo Supraquiasmático , Animais , Comportamento Animal/fisiologia , Eletroencefalografia , Eletromiografia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Atividade Motora/fisiologia , Proteínas Circadianas Period , Fotoperíodo , Ratos , Ratos Wistar , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/fisiologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...