Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Neural Circuits ; 18: 1385908, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38590628

RESUMO

Animals need sleep, and the suprachiasmatic nucleus, the center of the circadian rhythm, plays an important role in determining the timing of sleep. The main input to the suprachiasmatic nucleus is the retinohypothalamic tract, with additional inputs from the intergeniculate leaflet pathway, the serotonergic afferent from the raphe, and other hypothalamic regions. Within the suprachiasmatic nucleus, two of the major subtypes are vasoactive intestinal polypeptide (VIP)-positive neurons and arginine-vasopressin (AVP)-positive neurons. VIP neurons are important for light entrainment and synchronization of suprachiasmatic nucleus neurons, whereas AVP neurons are important for circadian period determination. Output targets of the suprachiasmatic nucleus include the hypothalamus (subparaventricular zone, paraventricular hypothalamic nucleus, preoptic area, and medial hypothalamus), the thalamus (paraventricular thalamic nuclei), and lateral septum. The suprachiasmatic nucleus also sends information through several brain regions to the pineal gland. The olfactory bulb is thought to be able to generate a circadian rhythm without the suprachiasmatic nucleus. Some reports indicate that circadian rhythms of the olfactory bulb and olfactory cortex exist in the absence of the suprachiasmatic nucleus, but another report claims the influence of the suprachiasmatic nucleus. The regulation of circadian rhythms by sensory inputs other than light stimuli, including olfaction, has not been well studied and further progress is expected.


Assuntos
Hipotálamo , Núcleo Supraquiasmático , Animais , Núcleo Supraquiasmático/metabolismo , Hipotálamo/metabolismo , Ritmo Circadiano/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Sono , Arginina Vasopressina/metabolismo
2.
J Pharmacol Sci ; 153(4): 215-220, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37973219

RESUMO

OBJECTIVE: Imeglimin is a novel antidiabetic drug structurally related to metformin. Metformin has been shown to modulate the circadian clock in rat fibroblasts. Accordingly, in the present study, we aimed to determine whether imeglimin can impact the circadian oscillator in mouse embryonic fibroblasts (MEFs). METHODS: MEFs carrying a Bmal1-Emerald luciferase (Bmal1-ELuc) reporter were exposed to imeglimin (0.1 or 1 mM), metformin (0.1 or 1 mM), a nicotinamide phosphoribosyltransferase inhibitor FK866, and/or vehicle. Subsequently, Bmal1-ELuc expression and clock gene mRNA expression levels were measured at 10-min intervals for 55 h and 4-h intervals for 32 h, respectively. RESULTS: Imeglimin significantly prolonged the period (from 26.3 to 30.0 h at 0.1 mM) and dose-dependently increased the amplitude (9.6-fold at 1 mM) of the Bmal1-ELuc expression rhythm; however, metformin exhibited minimal effects on these parameters. Moreover, imeglimin notably impacted the rhythmic mRNA expression of clock genes (Bmal1, Per1, and Cry1). The concurrent addition of FK866 partly inhibited the effects of imeglimin on both Bmal1-ELuc expression and clock gene mRNA expression. CONCLUSION: Collectively, these results reveal that imeglimin profoundly affects the circadian clock in MEFs. Further studies are needed to evaluate whether imeglimin treatment could exert similar effects in vivo.


Assuntos
Relógios Circadianos , Metformina , Ratos , Camundongos , Animais , Relógios Circadianos/genética , Ritmo Circadiano , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Fibroblastos/metabolismo , RNA Mensageiro/metabolismo , Metformina/farmacologia
3.
Neuroscience ; 532: 87-102, 2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37778689

RESUMO

The thalamic reticular nucleus (TRN) is a thin sheet of GABAergic neurons surrounding the thalamus, and it regulates the activity of thalamic relay neurons. The TRN has been reported to be involved in sensory gating, attentional regulation, and some other functions. However, little is known about the contribution of the TRN to sequence learning. In the present study, we examined whether the TRN is involved in reward-based learning of action sequence with no eliciting stimuli (operant conditioning), by analyzing the performance of male and female Avp-Vgat-/- mice (Vgatflox/flox mice crossed to an Avp-Cre driver line) on tasks conducted in an operant box having three levers. Our histological and electrophysiological data demonstrated that in adult Avp-Vgat-/- mice, vesicular GABA transporter (VGAT) was absent in most TRN neurons and the GABAergic transmission from the TRN to the thalamus was largely suppressed. The performance on a task in which mice needed to press an active lever for food reward showed that simple operant learning of lever pressing and learning of win-stay and lose-shift strategies are not affected in Avp-Vgat-/- mice. In contrast, the performance on a task in which mice needed to press three levers in a correct order for food reward showed that learning of the order of lever pressing (action sequence learning) was impaired in Avp-Vgat-/- mice. These results suggest that the TRN plays an important role in action sequence learning.


Assuntos
Núcleos Talâmicos , Tálamo , Camundongos , Masculino , Feminino , Animais , Núcleos Talâmicos/fisiologia , Neurônios GABAérgicos/fisiologia , Aprendizagem/fisiologia , Condicionamento Operante
4.
Sci Rep ; 13(1): 16974, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37813987

RESUMO

Prokineticin 2 (Prok2) is a small protein expressed in a subpopulation of neurons in the suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals. Prok2 has been implicated as a candidate output molecule from the SCN to control multiple circadian rhythms. Genetic manipulation specific to Prok2-producing neurons would be a powerful approach to understanding their function. Here, we report the generation of Prok2-tTA knock-in mice expressing the tetracycline transactivator (tTA) specifically in Prok2 neurons and an application of these mice to in vivo recording of Ca2+ rhythms in these neurons. First, the specific and efficient expression of tTA in Prok2 neurons was verified by crossing the mice with EGFP reporter mice. Prok2-tTA mice were then used to express a fluorescent Ca2+ sensor protein to record the circadian Ca2+ rhythm in SCN Prok2 neurons in vivo. Ca2+ in these cells showed clear circadian rhythms in both light-dark and constant dark conditions, with their peaks around midday. Notably, the hours of high Ca2+ nearly coincided with the rest period of the behavioral rhythm. These observations fit well with the predicted function of Prok2 neurons as a candidate output pathway of the SCN by suppressing locomotor activity during both daytime and subjective daytime.


Assuntos
Cálcio , Neurônios do Núcleo Supraquiasmático , Camundongos , Animais , Cálcio/metabolismo , Núcleo Supraquiasmático/fisiologia , Ritmo Circadiano/fisiologia , Neurônios do Núcleo Supraquiasmático/metabolismo , Neurônios/metabolismo , Transativadores/metabolismo , Cálcio da Dieta/metabolismo , Mamíferos/metabolismo
5.
PLoS Biol ; 21(8): e3002281, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37643163

RESUMO

The central circadian clock of the suprachiasmatic nucleus (SCN) is a network consisting of various types of neurons and glial cells. Individual cells have the autonomous molecular machinery of a cellular clock, but their intrinsic periods vary considerably. Here, we show that arginine vasopressin (AVP) neurons set the ensemble period of the SCN network in vivo to control the circadian behavior rhythm. Artificial lengthening of cellular periods by deleting casein kinase 1 delta (CK1δ) in the whole SCN lengthened the free-running period of behavior rhythm to an extent similar to CK1δ deletion specific to AVP neurons. However, in SCN slices, PER2::LUC reporter rhythms of these mice only partially and transiently recapitulated the period lengthening, showing a dissociation between the SCN shell and core with a period instability in the shell. In contrast, in vivo calcium rhythms of both AVP and vasoactive intestinal peptide (VIP) neurons in the SCN of freely moving mice demonstrated stably lengthened periods similar to the behavioral rhythm upon AVP neuron-specific CK1δ deletion, without changing the phase relationships between each other. Furthermore, optogenetic activation of AVP neurons acutely induced calcium increase in VIP neurons in vivo. These results indicate that AVP neurons regulate other SCN neurons, such as VIP neurons, in vivo and thus act as a primary determinant of the SCN ensemble period.


Assuntos
Arginina Vasopressina , Cálcio , Animais , Camundongos , Neurônios , Núcleo Supraquiasmático , Neuroglia , Peptídeo Intestinal Vasoativo
6.
Neuron ; 111(14): 2119-2120, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37473722

RESUMO

In this issue of Neuron, Xie et al.1 highlight a role of cholecystokinin (CCK) neurons in the suprachiasmatic nucleus (SCN) central clock for tracking the onset of circadian activities, adapting circadian rhythms to long photoperiods, and regulating circadian phase resetting.


Assuntos
Relógios Circadianos , Ritmo Circadiano , Ritmo Circadiano/fisiologia , Fotoperíodo , Núcleo Supraquiasmático/fisiologia , Neurônios , Colecistocinina
7.
JCI Insight ; 8(7)2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36862514

RESUMO

Hypothalamic neurons regulate body homeostasis by sensing and integrating changes in the levels of key hormones and primary nutrients (amino acids, glucose, and lipids). However, the molecular mechanisms that enable hypothalamic neurons to detect primary nutrients remain elusive. Here, we identified l-type amino acid transporter 1 (LAT1) in hypothalamic leptin receptor-expressing (LepR-expressing) neurons as being important for systemic energy and bone homeostasis. We observed LAT1-dependent amino acid uptake in the hypothalamus, which was compromised in a mouse model of obesity and diabetes. Mice lacking LAT1 (encoded by solute carrier transporter 7a5, Slc7a5) in LepR-expressing neurons exhibited obesity-related phenotypes and higher bone mass. Slc7a5 deficiency caused sympathetic dysfunction and leptin insensitivity in LepR-expressing neurons before obesity onset. Importantly, restoring Slc7a5 expression selectively in LepR-expressing ventromedial hypothalamus neurons rescued energy and bone homeostasis in mice deficient for Slc7a5 in LepR-expressing cells. Mechanistic target of rapamycin complex-1 (mTORC1) was found to be a crucial mediator of LAT1-dependent regulation of energy and bone homeostasis. These results suggest that the LAT1/mTORC1 axis in LepR-expressing neurons controls energy and bone homeostasis by fine-tuning sympathetic outflow, thus providing in vivo evidence of the implications of amino acid sensing by hypothalamic neurons in body homeostasis.


Assuntos
Hipotálamo , Transportador 1 de Aminoácidos Neutros Grandes , Camundongos , Animais , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Neurônios/metabolismo , Homeostase/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo
8.
Proc Natl Acad Sci U S A ; 120(11): e2218209120, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36877841

RESUMO

Mammals exhibit circadian cycles of sleep and wakefulness under the control of the suprachiasmatic nucleus (SCN), such as the strong arousal phase-locked to the beginning of the dark phase in laboratory mice. Here, we demonstrate that salt-inducible kinase 3 (SIK3) deficiency in gamma-aminobutyric acid (GABA)-ergic neurons or neuromedin S (NMS)-producing neurons delayed the arousal peak phase and lengthened the behavioral circadian cycle under both 12-h light:12-h dark condition (LD) and constant dark condition (DD) without changing daily sleep amounts. In contrast, the induction of a gain-of-function mutant allele of Sik3 in GABAergic neurons exhibited advanced activity onset and a shorter circadian period. Loss of SIK3 in arginine vasopressin (AVP)-producing neurons lengthened the circadian cycle, but the arousal peak phase was similar to that in control mice. Heterozygous deficiency of histone deacetylase (HDAC) 4, a SIK3 substrate, shortened the circadian cycle, whereas mice with HDAC4 S245A, which is resistant to phosphorylation by SIK3, delayed the arousal peak phase. Phase-delayed core clock gene expressions were detected in the liver of mice lacking SIK3 in GABAergic neurons. These results suggest that the SIK3-HDAC4 pathway regulates the circadian period length and the timing of arousal through NMS-positive neurons in the SCN.


Assuntos
Nível de Alerta , Histona Desacetilases , Proteínas Serina-Treonina Quinases , Vigília , Animais , Camundongos , Alelos , Arginina Vasopressina , Proteínas Serina-Treonina Quinases/genética , Núcleo Supraquiasmático , Histona Desacetilases/genética
9.
Int J Mol Sci ; 24(2)2023 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-36675058

RESUMO

The circadian rhythm, which is necessary for reproduction, is controlled by clock genes. In the mouse uterus, the oscillation of the circadian clock gene has been observed. The transcription of the core clock gene period (Per) and cryptochrome (Cry) is activated by the heterodimer of the transcription factor circadian locomotor output cycles kaput (Clock) and brain and muscle Arnt-like protein-1 (Bmal1). By binding to E-box sequences in the promoters of Per1/2 and Cry1/2 genes, the CLOCK-BMAL1 heterodimer promotes the transcription of these genes. Per1/2 and Cry1/2 form a complex with the Clock/Bmal1 heterodimer and inactivate its transcriptional activities. Endometrial BMAL1 expression levels are lower in human recurrent-miscarriage sufferers. Additionally, it was shown that the presence of BMAL1-depleted decidual cells prevents trophoblast invasion, highlighting the importance of the endometrial clock throughout pregnancy. It is widely known that hormone synthesis is disturbed and sterility develops in Bmal1-deficient mice. Recently, we discovered that animals with uterus-specific Bmal1 loss also had poor placental development, and these mice also had intrauterine fetal death. Furthermore, it was shown that time-restricted feeding controlled the uterine clock's circadian rhythm. The uterine clock system may be a possibility for pregnancy complications, according to these results. We summarize the most recent research on the close connection between the circadian clock and reproduction in this review.


Assuntos
Fatores de Transcrição ARNTL , Proteínas CLOCK , Relógios Circadianos , Reprodução , Animais , Feminino , Humanos , Camundongos , Gravidez , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Criptocromos/genética , Criptocromos/metabolismo , Regulação da Expressão Gênica , Placenta/metabolismo , Reprodução/genética , Reprodução/fisiologia
10.
Front Cell Neurosci ; 16: 877131, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36274991

RESUMO

Astrocytes play key roles in supporting the central nervous system structure, regulating synaptic functions, and maintaining brain homeostasis. The number of astrocytes in the cerebrum has markedly increased through evolution. However, the manner by which astrocytes change their features during evolution remains unknown. Compared with the rodent brain, the brain of the ferret, a carnivorous animal, has a folded cerebral cortex and higher white to gray matter ratio, which are common features of the human brain. To further clarify the features of ferret astrocytes, we isolated astrocytes from ferret neonatal brains, cultured these cells, and compared their morphology, gene expression, calcium response, and proliferating ability with those of mouse astrocytes. The morphology of cultured ferret astrocytes differed from that of mouse astrocytes. Ferret astrocytes had longer and more branched processes, smaller cell bodies, and different calcium responses to glutamate, as well as had a greater ability to proliferate, compared to mouse astrocytes. RNA sequencing analysis revealed novel ferret astrocyte-specific genes, including several genes that were the same as those in humans. Astrocytes in the ferret brains had larger cell size, longer primary processes in larger numbers, and a higher proliferation rate compared to mouse astrocytes. Our study shows that cultured ferret astrocytes have different features from rodent astrocytes and similar features to human astrocytes, suggesting that they are useful in studying the roles of astrocytes in brain evolution and cognitive functions in higher animals.

11.
Front Endocrinol (Lausanne) ; 13: 956169, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992114

RESUMO

Disruptions to the circadian system alter reproductive capacity, particularly in females. Mice lacking the core circadian clock gene, Bmal1, are infertile and have evidence of neuroendocrine disruption including the absence of the preovulatory luteinizing hormone (LH) surge and enhanced responsiveness to exogenous kisspeptin. Here, we explore the role of Bmal1 in suprachiasmatic nucleus (SCN) neuron populations known to project to the neuroendocrine axis. We generated four mouse lines using Cre/Lox technology to create conditional deletion of Bmal1 in arginine vasopressin (Bmal1fl/fl:Avpcre ), vasoactive intestinal peptide (Bmal1fl/fl:Vipcre ), both (Bmal1fl/fl:Avpcre+Vipcre ), and neuromedin-s (Bmal1fl/fl:Nmscre ) neurons. We demonstrate that the loss of Bmal1 in these populations has substantial effects on home-cage circadian activity and temperature rhythms. Despite this, we found that female mice from these lines demonstrated normal estrus cycles, fecundity, kisspeptin responsiveness, and inducible LH surge. We found no evidence of reproductive disruption in constant darkness. Overall, our results indicate that while conditional Bmal1 knockout in AVP, VIP, or NMS neurons is sufficient to disrupted locomotor activity, this disruption is insufficient to recapitulate the neuroendocrine reproductive effects of the whole-body Bmal1 knockout.


Assuntos
Neurônios do Núcleo Supraquiasmático , Peptídeo Intestinal Vasoativo , Animais , Arginina Vasopressina/genética , Ritmo Circadiano/fisiologia , Feminino , Fertilidade , Kisspeptinas/genética , Hormônio Luteinizante , Camundongos , Núcleo Supraquiasmático/metabolismo , Neurônios do Núcleo Supraquiasmático/metabolismo
12.
Curr Biol ; 32(18): 3871-3885.e4, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-35907397

RESUMO

The sleep-wakefulness cycle is regulated by complicated neural networks that include many different populations of neurons throughout the brain. Arginine vasopressin neurons in the paraventricular nucleus of the hypothalamus (PVHAVP) regulate various physiological events and behaviors, such as body-fluid homeostasis, blood pressure, stress response, social interaction, and feeding. Changes in arousal level often accompany these PVHAVP-mediated adaptive responses. However, the contribution of PVHAVP neurons to sleep-wakefulness regulation has remained unknown. Here, we report the involvement of PVHAVP neurons in arousal promotion. Optogenetic stimulation of PVHAVP neurons rapidly induced transitions to wakefulness from both NREM and REM sleep. This arousal effect was dependent on AVP expression in these neurons. Similarly, chemogenetic activation of PVHAVP neurons increased wakefulness and reduced NREM and REM sleep, whereas chemogenetic inhibition of these neurons significantly reduced wakefulness and increased NREM sleep. We observed dense projections of PVHAVP neurons in the lateral hypothalamus with potential connections to orexin/hypocretin (LHOrx) neurons. Optogenetic stimulation of PVHAVP neuronal fibers in the LH immediately induced wakefulness, whereas blocking orexin receptors attenuated the arousal effect of PVHAVP neuronal activation drastically. Monosynaptic rabies-virus tracing revealed that PVHAVP neurons receive inputs from multiple brain regions involved in sleep-wakefulness regulation, as well as those involved in stress response and energy metabolism. Moreover, PVHAVP neurons mediated the arousal induced by novelty stress and a melanocortin receptor agonist melanotan-II. Thus, our data suggested that PVHAVP neurons promote wakefulness via LHOrx neurons in the basal sleep-wakefulness and some stressful conditions.


Assuntos
Região Hipotalâmica Lateral , Vigília , Arginina Vasopressina/metabolismo , Região Hipotalâmica Lateral/fisiologia , Hipotálamo/metabolismo , Neurônios/fisiologia , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Melanocortina/metabolismo , Sono/fisiologia , Vasopressinas/metabolismo , Vasopressinas/farmacologia , Vigília/fisiologia
13.
Mol Brain ; 15(1): 47, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35606816

RESUMO

Self-grooming plays an essential role in hygiene maintenance, thermoregulation, and stress response. However, the neural populations involved in self-grooming remain largely unknown. The paraventricular hypothalamic nucleus (PVH) has been implicated in the regulation of self-grooming. Arginine vasopressin-producing neurons are among the major neuronal populations in the PVH (PVHAVP), which play important roles in water homeostasis, blood pressure regulation, feeding, and stress response. Here, we report the critical role of PVHAVP neurons in the induction of self-grooming. Optogenetic activation of PVHAVP neurons immediately induced self-grooming in freely moving mice. Chemogenetic activation of these neurons also increased time spent self-grooming. In contrast, their chemogenetic inhibition significantly reduced naturally occurring self-grooming, suggesting that PVHAVP-induced grooming has physiological relevance. Notably, optogenetic activation of PVHAVP neurons triggered self-grooming over other adaptive behaviors, such as voracious feeding induced by fasting and social interaction with female mice. Thus, our study proposes the novel role of PVHAVP neurons in regulating self-grooming behavior and, consequently, hygiene maintenance and stress response. Furthermore, uncontrolled activation of these neurons may be potentially relevant to diseases characterized by compulsive behaviors and impaired social interaction, such as autism, obsessive-compulsive disorder, and anorexia nervosa.


Assuntos
Arginina Vasopressina , Núcleo Hipotalâmico Paraventricular , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Feminino , Asseio Animal , Camundongos , Neurônios/metabolismo , Optogenética , Núcleo Hipotalâmico Paraventricular/metabolismo
14.
Front Physiol ; 13: 895633, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35592033

RESUMO

The suprachiasmatic nucleus (SCN), the central circadian clock in mammals, is a neural network consisting of various types of GABAergic neurons, which can be differentiated by the co-expression of specific peptides such as vasoactive intestinal peptide (VIP) and arginine vasopressin (AVP). VIP has been considered as a critical factor for the circadian rhythmicity and synchronization of individual SCN neurons. However, the precise mechanisms of how VIP neurons regulate SCN circuits remain incompletely understood. Here, we generated Vip tTA knock-in mice that express tetracycline transactivator (tTA) specifically in VIP neurons by inserting tTA sequence at the start codon of Vip gene. The specific and efficient expression of tTA in VIP neurons was verified using EGFP reporter mice. In addition, combined with Avp-Cre mice, Vip tTA mice enabled us to simultaneously apply different genetic manipulations to VIP and AVP neurons in the SCN. Immunostaining showed that VIP is expressed at a slightly reduced level in heterozygous Vip tTA mice but is completely absent in homozygous mice. Consistently, homozygous Vip tTA mice showed impaired circadian behavioral rhythms similar to those of Vip knockout mice, such as attenuated rhythmicity and shortened circadian period. In contrast, heterozygous mice demonstrated normal circadian behavioral rhythms comparable to wild-type mice. These data suggest that Vip tTA mice are a valuable genetic tool to express exogenous genes specifically in VIP neurons in both normal and VIP-deficient mice, facilitating the study of VIP neuronal roles in the SCN neural network.

15.
Curr Dev Nutr ; 5(5): nzab064, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33981944

RESUMO

BACKGROUND: Skipping breakfast is associated with dysmenorrhea in young women. This suggests that the delay of food intake in the active phase impairs uterine functions by interfering with circadian rhythms. OBJECTIVES: To examine the relation between the delay of feeding and uterine circadian rhythms, we investigated the effects of the first meal occasion in the active phase on the uterine clock. METHODS: Zeitgeber time (ZT) was defined as ZT0 (08:45) with lights on and ZT12 (20:45) with lights off. Young female mice (8 wk of age) were divided into 3 groups: group I (ad libitum consumption), group II (time-restricted feeding during ZT12-16, initial 4 h of the active period), and group III (time-restricted feeding during ZT20-24, last 4 h of the active period, a breakfast-skipping model). After 2 wk of dietary restriction, mice in each group were killed at 4-h intervals and the expression profiles of uterine clock genes, Bmal1 (brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1), Per1 (period circadian clock 1), Per2, and Cry1 (cryptochrome 1), were examined. RESULTS: qPCR and western blot analyses demonstrated synchronized circadian clock gene expression within the uterus. Immunohistochemical analysis confirmed that BMAL1 protein expression was synchronized among the endometrium and myometrium. In groups I and II, mRNA expression of Bmal1 was elevated after ZT12 at the start of the active phase. In contrast, Bmal1 expression was elevated just after ZT20 in group III, showing that the uterine clock rhythm had shifted 8 h backward. The changes in BMAL1 protein expression were confirmed by western blot analysis. CONCLUSIONS: This study is the first to indicate that time-restricted feeding regulates a circadian rhythm of the uterine clock that is synchronized throughout the uterine body. These findings suggest that the uterine clock system is a new candidate to explain the etiology of breakfast skipping-induced uterine dysfunction.

16.
Mol Metab ; 49: 101202, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33676029

RESUMO

OBJECTIVE: Impaired circadian clocks can cause obesity, but their pathophysiological role in brown adipose tissue (BAT), a major tissue regulating energy metabolism, remains unclear. To address this issue, we investigated the effects of complete disruption of the BAT clock on thermogenesis and energy expenditure. METHODS: Mice with brown adipocyte-specific knockout of the core clock gene Bmal1 (BA-Bmal1 KO) were generated and analyzed. RESULTS: The BA-Bmal1 KO mice maintained normal core body temperatures by increasing shivering and locomotor activity despite the elevated expression of thermogenic uncoupling protein 1 in BAT. BA-Bmal1 KO disrupted 24 h rhythmicity of fatty acid utilization in BAT and mildly reduced both BAT thermogenesis and whole-body energy expenditure. The impact of BA-Bmal1 KO on the development of obesity became obvious when the mice were fed a high-fat diet. CONCLUSIONS: These results reveal the importance of the BAT clock for maintaining energy homeostasis and preventing obesity.


Assuntos
Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Adipócitos Marrons/metabolismo , Termogênese/genética , Termogênese/fisiologia , Tecido Adiposo Marrom/metabolismo , Animais , Peso Corporal , Ritmo Circadiano , Temperatura Baixa , Dieta Hiperlipídica , Metabolismo Energético , Ácidos Graxos , Homeostase , Masculino , Metaboloma , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Proteína Desacopladora 1/metabolismo
17.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33526663

RESUMO

The suprachiasmatic nucleus (SCN), the central circadian pacemaker in mammals, is a network structure composed of multiple types of γ-aminobutyric acid (GABA)-ergic neurons and glial cells. However, the roles of GABA-mediated signaling in the SCN network remain controversial. Here, we report noticeable impairment of the circadian rhythm in mice with a specific deletion of the vesicular GABA transporter in arginine vasopressin (AVP)-producing neurons. These mice showed disturbed diurnal rhythms of GABAA receptor-mediated synaptic transmission in SCN neurons and marked lengthening of the activity time in circadian behavioral rhythms due to the extended interval between morning and evening locomotor activities. Synchrony of molecular circadian oscillations among SCN neurons did not significantly change, whereas the phase relationships between SCN molecular clocks and circadian morning/evening locomotor activities were altered significantly, as revealed by PER2::LUC imaging of SCN explants and in vivo recording of intracellular Ca2+ in SCN AVP neurons. In contrast, daily neuronal activity in SCN neurons in vivo clearly showed a bimodal pattern that correlated with dissociated morning/evening locomotor activities. Therefore, GABAergic transmission from AVP neurons regulates the timing of SCN neuronal firing to temporally restrict circadian behavior to appropriate time windows in SCN molecular clocks.


Assuntos
Relógios Circadianos , Ritmo Circadiano , Neurônios/metabolismo , Núcleo Supraquiasmático/metabolismo , Vasopressinas/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Comportamento Animal , Cálcio/metabolismo , Relógios Circadianos/genética , Ritmo Circadiano/genética , Regulação da Expressão Gênica , Locomoção , Camundongos , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Fatores de Tempo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/deficiência , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
18.
Brain Nerve ; 72(11): 1143-1150, 2020 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-33191293

RESUMO

Circadian rhythms are oscillations with an approximately 24-h period and appear in most of the physiological events of our body. The suprachiasmatic nucleus (SCN) of the hypothalamus functions as the central circadian clock in mammals and entrains to the environmental light/dark (day/night) cycle. Here, I briefly review the molecular, cellular, and anatomical structures of the SCN, present findings of recent studies on the differential roles of multiple neuropeptides and neuropeptide-expressing neurons in the SCN, and discuss the mechanisms of the SCN network.


Assuntos
Relógios Circadianos , Neuropeptídeos , Animais , Ritmo Circadiano , Humanos , Hipotálamo , Núcleo Supraquiasmático
19.
Nutrients ; 12(5)2020 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-32370105

RESUMO

There are growing concerns that poor dietary behaviors at young ages will increase the future risk of chronic diseases in adulthood. We found that female college students who skipped breakfast had higher incidences of dysmenorrhea and irregular menstruation, suggesting that meal skipping affects ovarian and uterine functions. Since dysmenorrhea is more prevalent in those with a past history of dieting, we proposed a novel concept that inadequate dietary habits in adolescence become a trigger for the subsequent development of organic gynecologic diseases. Since inadequate feeding that was limited during the non-active phase impaired reproductive functions in post-adolescent female rats, we hypothesize that circadian rhythm disorders due to breakfast skipping disrupts the hypothalamic-pituitary-ovarian axis, impairs the reproductive rhythm, and leads to ovarian and uterine dysfunction. To explain how reproductive dysfunction is memorized from adolescence to adulthood, we hypothesize that the peripheral clock system also plays a critical role in the latent progression of reproductive diseases together with the central system, and propose naming this concept "adolescent dietary habit-induced obstetric and gynecologic disease (ADHOGD)". This theory will contribute to analyzing the etiologies of and developing prophylaxes for female reproductive diseases from novel aspects. In this article, we describe the precise outline of the above hypotheses with the supporting evidence in the literature.


Assuntos
Relógios Biológicos/fisiologia , Desjejum , Dismenorreia/etiologia , Comportamento Alimentar/fisiologia , Doenças dos Genitais Femininos/etiologia , Refeições , Distúrbios Menstruais/etiologia , Fenômenos Fisiológicos da Nutrição/fisiologia , Adolescente , Comportamento do Adolescente , Adulto , Feminino , Humanos , Sistema Hipotálamo-Hipofisário , Ovário , Reprodução , Adulto Jovem
20.
Neurosci Res ; 156: 24-31, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31560907

RESUMO

Circadian rhythms are oscillations with approximately 24-h period that appear in most of physiological events in our body. The suprachiasmatic nucleus (SCN) of the hypothalamus functions as the central circadian pacemaker in mammals and entrains to the environmental light/dark cycle. The SCN is a network structure composed of multiple types of γ-amino butyric acid (GABA)-ergic neurons and glial cells. Although individual SCN neurons have intracellular molecular machinery of circadian clock and the ability to oscillate cell-autonomously, interneuronal communications among these neurons are essential for the circadian pacemaking of the SCN. However, the mechanisms underlying the SCN network remain largely unknown. Here, I briefly review the molecular, cellular, and anatomical structures of the SCN and introduce recent studies aiming to understand the differential roles of multiple neuropeptides and neuropeptide-expressing neurons in the SCN network.


Assuntos
Relógios Circadianos , Neuropeptídeos , Animais , Ritmo Circadiano , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Núcleo Supraquiasmático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...