Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 883
Filtrar
1.
PLoS Comput Biol ; 17(12): e1009698, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34919546

RESUMO

We propose a stochastic distributed delay model together with a Markov random field prior and a measurement model for bioluminescence-reporting to analyse spatio-temporal gene expression in intact networks of cells. The model describes the oscillating time evolution of molecular mRNA counts through a negative transcriptional-translational feedback loop encoded in a chemical Langevin equation with a probabilistic delay distribution. The model is extended spatially by means of a multiplicative random effects model with a first order Markov random field prior distribution. Our methodology effectively separates intrinsic molecular noise, measurement noise, and extrinsic noise and phenotypic variation driving cell heterogeneity, while being amenable to parameter identification and inference. Based on the single-cell model we propose a novel computational stability analysis that allows us to infer two key characteristics, namely the robustness of the oscillations, i.e. whether the reaction network exhibits sustained or damped oscillations, and the profile of the regulation, i.e. whether the inhibition occurs over time in a more distributed versus a more direct manner, which affects the cells' ability to phase-shift to new schedules. We show how insight into the spatio-temporal characteristics of the circadian feedback loop in the suprachiasmatic nucleus (SCN) can be gained by applying the methodology to bioluminescence-reported expression of the circadian core clock gene Cry1 across mouse SCN tissue. We find that while (almost) all SCN neurons exhibit robust cell-autonomous oscillations, the parameters that are associated with the regulatory transcription profile give rise to a spatial division of the tissue between the central region whose oscillations are resilient to perturbation in the sense that they maintain a high degree of synchronicity, and the dorsal region which appears to phase shift in a more diversified way as a response to large perturbations and thus could be more amenable to entrainment.


Assuntos
Relógios Circadianos/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano , Regulação da Expressão Gênica/genética , Modelos Biológicos , Transcrição Gênica/genética , Animais , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Criptocromos/genética , Criptocromos/metabolismo , Camundongos , Fenótipo , Análise de Célula Única , Processos Estocásticos , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo
2.
EMBO J ; 40(20): e108614, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34487375

RESUMO

Circadian rhythms in mammals are governed by the hypothalamic suprachiasmatic nucleus (SCN), in which 20,000 clock cells are connected together into a powerful time-keeping network. In the absence of network-level cellular interactions, the SCN fails as a clock. The topology and specific roles of its distinct cell populations (nodes) that direct network functions are, however, not understood. To characterise its component cells and network structure, we conducted single-cell sequencing of SCN organotypic slices and identified eleven distinct neuronal sub-populations across circadian day and night. We defined neuropeptidergic signalling axes between these nodes, and built neuropeptide-specific network topologies. This revealed their temporal plasticity, being up-regulated in circadian day. Through intersectional genetics and real-time imaging, we interrogated the contribution of the Prok2-ProkR2 neuropeptidergic axis to network-wide time-keeping. We showed that Prok2-ProkR2 signalling acts as a key regulator of SCN period and rhythmicity and contributes to defining the network-level properties that underpin robust circadian co-ordination. These results highlight the diverse and distinct contributions of neuropeptide-modulated communication of temporal information across the SCN.


Assuntos
Relógios Circadianos/genética , Ritmo Circadiano/genética , Hormônios Gastrointestinais/genética , Neuropeptídeos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Núcleo Supraquiasmático/metabolismo , Transcriptoma , Animais , Peptídeo Liberador de Gastrina/genética , Peptídeo Liberador de Gastrina/metabolismo , Hormônios Gastrointestinais/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Receptores da Bombesina/genética , Receptores da Bombesina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Transdução de Sinais , Análise de Célula Única , Núcleo Supraquiasmático/citologia , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/metabolismo , Vasopressinas/genética , Vasopressinas/metabolismo
3.
Neuron ; 109(20): 3268-3282.e6, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34416169

RESUMO

The suprachiasmatic nucleus (SCN) is the master circadian pacemaker in mammals and is entrained by environmental light. However, the molecular basis of the response of the SCN to light is not fully understood. We used RNA/chromatin immunoprecipitation/single-nucleus sequencing with circadian behavioral assays to identify mouse SCN cell types and explore their responses to light. We identified three peptidergic cell types that responded to light in the SCN: arginine vasopressin (AVP), vasoactive intestinal peptide (VIP), and cholecystokinin (CCK). In each cell type, light-responsive subgroups were enriched for expression of neuronal Per-Arnt-Sim (PAS) domain protein 4 (NPAS4) target genes. Further, mice lacking Npas4 had a longer circadian period under constant conditions, a damped phase response curve to light, and reduced light-induced gene expression in the SCN. Our data indicate that NPAS4 is necessary for normal transcriptional responses to light in the SCN and critical for photic phase-shifting of circadian behavior.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Ritmo Circadiano/genética , Luz , Neurônios/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Arginina Vasopressina/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Colecistocinina/metabolismo , Imunoprecipitação da Cromatina , Ritmo Circadiano/efeitos da radiação , Perfilação da Expressão Gênica , Camundongos , Camundongos Knockout , Neurônios/efeitos da radiação , Análise de Sequência de RNA , Análise de Célula Única , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/efeitos da radiação , Peptídeo Intestinal Vasoativo/metabolismo
4.
J Neurophysiol ; 126(2): 637-652, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34259044

RESUMO

Synaptic and extrasynaptic GABAA receptor (GABAAR)-mediated neurotransmission is a critical component of the suprachiasmatic nucleus (SCN) neuronal network. However, the properties of the GABAA tonic current (Itonic) and its origin remain unexplored. Spontaneous GABAA postsynaptic currents (sGPSCs) and Itonic were recorded from SCN neurons with the whole cell voltage-clamp technique at different times of the day. GABAAR antagonists (bicuculline, gabazine, and picrotoxin) inhibited sGPSC and induced an outward shift of the holding current, which defined the Itonic amplitude. The sGPSC frequency, synaptic charge transfer, and Itonic amplitude all demonstrated significant diurnal rhythms, with peaks in the middle of the day [zeitgeber time (ZT)7-8] and nadirs at night (ZT19-20). The Itonic amplitude increased proportionally with the sGPSC frequency and synaptic charge transfer during the day and required action potential-mediated GABA release, which was confirmed by TTX application. The activation of presynaptic GABAB receptors by baclofen did not significantly alter the Itonic of neurons with low-frequency sGPSC. The equilibrium potential (Eq) for Itonic was similar to the Eq for chloride and GABAA receptor-activated currents. Itonic showed outward rectification at membrane potentials over the range of -70 to -10 mV and then was linear at voltages greater than -10 mV. GABAAR containing α4-, α5-, and δ-subunits were expressed in SCN, and their contribution to Itonic was confirmed by application of the GABAAR agonist 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol (THIP) and the GABAAR inverse agonist 11,12,13,13a-tetrahydro-7-methoxy-9-oxo-9H-imidazo[1,5-a]pyrrolo[2,1-c][1,4]benzodiazepine-1-carboxylic acid ethyl ester (L655,708). Thus, the Itonic was mediated by extrasynaptic GABAARs activated predominantly by GABA diffusing out of GABAergic synapses.NEW & NOTEWORTHY A tonic current (Itonic) mediated by GABAA receptors (GABAARs) containing α4-, α5- and δ-subunits was observed in the suprachiasmatic nucleus. The Itonic amplitude strongly depended on the action potential-mediated synaptic release of GABA. The equilibrium potential for Itonic corresponds to that for GABAA currents. The frequency of GABAA postsynaptic currents and Itonic amplitude increased during the day, with peak in the middle of the day, and then gradually declined with a nadir at night, thus showing a diurnal rhythm.


Assuntos
Ritmo Circadiano , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Núcleo Supraquiasmático/fisiologia , Potenciais Sinápticos , Animais , Agonistas de Receptores de GABA-A/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo
5.
Nature ; 592(7856): 763-767, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33762728

RESUMO

Systemic insulin sensitivity shows a diurnal rhythm with a peak upon waking1,2. The molecular mechanism that underlies this temporal pattern is unclear. Here we show that the nuclear receptors REV-ERB-α and REV-ERB-ß (referred to here as 'REV-ERB') in the GABAergic (γ-aminobutyric acid-producing) neurons in the suprachiasmatic nucleus (SCN) (SCNGABA neurons) control the diurnal rhythm of insulin-mediated suppression of hepatic glucose production in mice, without affecting diurnal eating or locomotor behaviours during regular light-dark cycles. REV-ERB regulates the rhythmic expression of genes that are involved in neurotransmission in the SCN, and modulates the oscillatory firing activity of SCNGABA neurons. Chemogenetic stimulation of SCNGABA neurons at waking leads to glucose intolerance, whereas restoration of the temporal pattern of either SCNGABA neuron firing or REV-ERB expression rescues the time-dependent glucose metabolic phenotype caused by REV-ERB depletion. In individuals with diabetes, an increased level of blood glucose after waking is a defining feature of the 'extended dawn phenomenon'3,4. Patients with type 2 diabetes with the extended dawn phenomenon exhibit a differential temporal pattern of expression of REV-ERB genes compared to patients with type 2 diabetes who do not have the extended dawn phenomenon. These findings provide mechanistic insights into how the central circadian clock regulates the diurnal rhythm of hepatic insulin sensitivity, with implications for our understanding of the extended dawn phenomenon in type 2 diabetes.


Assuntos
Ritmo Circadiano , Neurônios GABAérgicos/fisiologia , Resistência à Insulina , Fígado/fisiologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/fisiologia , Animais , Glicemia , Relógios Circadianos , Diabetes Mellitus Tipo 2 , Feminino , Glucose/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Fotoperíodo , Núcleo Supraquiasmático/citologia , Transmissão Sináptica
6.
Neuropharmacology ; 185: 108455, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33444638

RESUMO

Suprachiasmatic nucleus (SCN) of the hypothalamus is the master clock that drives circadian rhythms in physiology and behavior and adjusts their timing to external cues. Neurotransmitter glutamate and glutamatergic receptors sensitive to N-methyl-d-aspartate (NMDA) play a dual role in the SCN by coupling astrocytic and neuronal single cell oscillators and by resetting their phase in response to light. Recent reports suggested that signaling by endogenous cannabinoids (ECs) participates in both of these functions. We have previously shown that ECs, such as 2-arachidonoylglycerol (2-AG), act via CB1 receptors to affect the SCN response to light-mimicking NMDA stimulus in a time-dependent manner. We hypothesized that this ability is linked to the circadian regulation of EC signaling. We demonstrate that circadian clock in the rat SCN regulates expression of 2-AG transport, synthesis and degradation enzymes as well as its receptors. Inhibition of the major 2-AG synthesis enzyme, diacylglycerol lipase, enhanced the phase delay and lowered the amplitude of explanted SCN rhythm in response to NMDAR activation. Using microscopic PER2 bioluminescence imaging, we visualized how individual single cell oscillators in different parts of the SCN respond to the DAGL inhibition/NMDAR activation and shape response of the whole pacemaker. Additionally, we present strong evidence that the zero amplitude behavior of the SCN in response to single NMDA stimulus in the middle of subjective night is the result of a loss of rhythm in individual SCN cells. The paper provides new insights into the modulatory role of endocannabinoid signaling during the light entrainment of the SCN.


Assuntos
Ritmo Circadiano/fisiologia , Endocanabinoides/fisiologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Lipase Lipoproteica/antagonistas & inibidores , N-Metilaspartato/farmacologia , Núcleo Supraquiasmático/fisiologia , Animais , Ritmo Circadiano/efeitos dos fármacos , Feminino , Lipase Lipoproteica/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Wistar , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/efeitos dos fármacos
7.
Methods Mol Biol ; 2130: 325-336, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33284455

RESUMO

A fundamental feature of circadian clock neurons across species is that they express circadian rhythms in spontaneous spike frequency. Spike frequency rhythms serve as both output timing signals of clock neurons as well as resonant elements of rhythms generation. Importantly, optogenetics, as applied to clock neurons, can enable investigation of the roles of clock neuron electrical activity in circadian timing. Here we describe protocols for using both in vitro and in vivo optogenetics directed to mammalian clock neurons in the suprachiasmatic nucleus to study circadian physiology and behavior. Optogenetic stimulation via channelrhodopsin, or inhibition via halorhodopsin, allows for the precise manipulation of neuronal firing rates across the SCN, and within specific neuronal subpopulations thereof, and can be combined with actigraphy and gene expression analysis.


Assuntos
Potenciais de Ação , Relógios Circadianos , Neurônios/fisiologia , Optogenética/métodos , Animais , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Camundongos , Neurônios/metabolismo , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia
8.
Neurosci Lett ; 739: 135438, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33132178

RESUMO

Serotonin (5-HT) and its innervation have been implicated in various neural functions including circadian systems. Although classical studies have examined the 5-HT innervation pattern in the adult suprachiasmatic nucleus (SCN), the fine-grained morphological study of the development of pathway and terminal projections to the SCN remains scarce. Here, we utilize transgenic mice expressing GFP under the serotonin transporter (SERT) promoter to subserve our developmental mapping study. We demonstrate that the morphology of 5-HT pathway fibers decussating over the supraoptic commissure that projects to the SCN exhibits two distinct developmental patterns. The punctate fibers at the fetal stage gradually become smooth and filamentous, especially during postnatal one week and remain constant thereafter. The innervation field in the SCN develops properly only during postnatal two weeks. Its ventromedial area remains one of the highest 5-HT innervated areas in the adult brain, whereas the dorsolateral area is less innervated. Thus, we provide novel and specific insights on the developmental map of 5-HT system into the SCN using transgenic mouse.


Assuntos
Neurônios Serotoninérgicos/fisiologia , Neurônios do Núcleo Supraquiasmático/fisiologia , Núcleo Supraquiasmático/crescimento & desenvolvimento , Animais , Feminino , Camundongos Endogâmicos C57BL , Vias Neurais/citologia , Vias Neurais/crescimento & desenvolvimento , Neurônios Serotoninérgicos/citologia , Núcleo Supraquiasmático/citologia , Neurônios do Núcleo Supraquiasmático/citologia
9.
Front Neural Circuits ; 14: 55, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973462

RESUMO

Background: Monochromatic blue light (MBL), with a wavelength between 400-490 nm, can regulate non-image-forming (NIF) functions of light in the central nervous system. The suprachiasmatic nucleus (SCN) in the brain is involved in the arousal-promoting response to blue light in mice. Animal and human studies showed that the responsiveness of the brain to visual stimuli is partly preserved under general anesthesia. Therefore, this study aimed to investigate whether MBL promotes arousal from sevoflurane anesthesia via activation of the SCN in mice. Methods: The induction and emergence time of sevoflurane anesthesia under MBL (460 nm and 800 lux) exposure was measured. Cortical electroencephalograms (EEGs) were recorded and the burst-suppression ratio (BSR) was calculated under MBL during sevoflurane anesthesia. The EEGs and local field potential (LFP) recordings with or without locally electrolytic ablated bilateral SCN were used to further explore the role of SCN in the arousal-promoting effect of MBL under sevoflurane anesthesia. Immunofluorescent staining of c-Fos was conducted to reveal the possible downstream mechanism of SCN activation. Results: Unlike the lack of effect on the induction time, MBL shortened the emergence time and the EEG recordings showed cortical arousal during the recovery period. MBL resulted in a significant decrease in BSR and a marked increase in EEG power at all frequency bands except for the spindle band during 2.5% sevoflurane anesthesia. MBL exposure under sevoflurane anesthesia enhances the neuronal activity of the SCN. These responses to MBL were abolished in SCN lesioned (SCNx) mice. MBL evoked a high level of c-Fos expression in the prefrontal cortex (PFC) and lateral hypothalamus (LH) compared to polychromatic white light (PWL) under sevoflurane anesthesia, while it exerted no effect on c-Fos expression in the ventrolateral preoptic area (VLPO) and locus coeruleus (LC) c-Fos expression. Conclusions: MBL promotes behavioral and electroencephalographic arousal from sevoflurane anesthesia via the activation of the SCN and its associated downstream wake-related nuclei. The clinical implications of this study warrant further study.


Assuntos
Anestésicos Inalatórios/farmacologia , Nível de Alerta/efeitos da radiação , Hipotálamo/efeitos da radiação , Luz , Neurônios/efeitos da radiação , Córtex Pré-Frontal/efeitos da radiação , Sevoflurano/farmacologia , Núcleo Supraquiasmático/efeitos da radiação , Anestesia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Eletroencefalografia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos da radiação , Reflexo de Endireitamento/efeitos dos fármacos , Reflexo de Endireitamento/efeitos da radiação , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/metabolismo
10.
Nat Commun ; 11(1): 4410, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32879310

RESUMO

The hypothalamic suprachiasmatic (SCN) clock contains several neurochemically defined cell groups that contribute to the genesis of circadian rhythms. Using cell-specific and genetically targeted approaches we have confirmed an indispensable role for vasoactive intestinal polypeptide-expressing SCN (SCNVIP) neurons, including their molecular clock, in generating the mammalian locomotor activity (LMA) circadian rhythm. Optogenetic-assisted circuit mapping revealed functional, di-synaptic connectivity between SCNVIP neurons and dorsomedial hypothalamic neurons, providing a circuit substrate by which SCNVIP neurons may regulate LMA rhythms. In vivo photometry revealed that while SCNVIP neurons are acutely responsive to light, their activity is otherwise behavioral state invariant. Single-nuclei RNA-sequencing revealed that SCNVIP neurons comprise two transcriptionally distinct subtypes, including putative pacemaker and non-pacemaker populations. Altogether, our work establishes necessity of SCNVIP neurons for the LMA circadian rhythm, elucidates organization of circadian outflow from and modulatory input to SCNVIP cells, and demonstrates a subpopulation-level molecular heterogeneity that suggests distinct functions for specific SCNVIP subtypes.


Assuntos
Ritmo Circadiano/fisiologia , Neurônios/metabolismo , Núcleo Supraquiasmático , Animais , Mapeamento Encefálico , Relógios Circadianos/fisiologia , Locomoção/fisiologia , Camundongos , Optogenética/métodos , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo
11.
FASEB J ; 34(10): 13685-13695, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32869393

RESUMO

The central circadian pacemaker in mammals, the suprachiasmatic nucleus (SCN), is important for daily as well as seasonal rhythms. The SCN encodes seasonal changes in day length by adjusting phase distribution among oscillating neurons thereby shaping the output signal used for adaptation of physiology and behavior. It is well-established that brief light exposure at the beginning and end of the day, also referred to as "skeleton" light pulses, are sufficient to evoke the seasonal behavioral phenotype. However, the effect of skeleton light exposure on SCN network reorganization remains unknown. Therefore, we exposed mice to brief morning and evening light pulses that mark the time of dawn and dusk in a short winter- or a long summer day. Single-cell PER2::LUC recordings, electrophysiological recordings of SCN activity, and measurements of GABA response polarity revealed that skeleton light-regimes affected the SCN network to the same degree as full photoperiod. These results indicate the powerful, yet potentially harmful effects of even relatively short light exposures during the evening or night for nocturnal animals.


Assuntos
Ritmo Circadiano , Núcleo Supraquiasmático/fisiologia , Animais , Potenciais Evocados , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fotoperíodo , Núcleo Supraquiasmático/citologia , Ácido gama-Aminobutírico/metabolismo
12.
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
13.
Nat Commun ; 11(1): 3394, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32636383

RESUMO

The hypothalamic suprachiasmatic nuclei (SCN) are the principal mammalian circadian timekeeper, co-ordinating organism-wide daily and seasonal rhythms. To achieve this, cell-autonomous circadian timing by the ~20,000 SCN cells is welded into a tight circuit-wide ensemble oscillation. This creates essential, network-level emergent properties of precise, high-amplitude oscillation with tightly defined ensemble period and phase. Although synchronised, regional cell groups exhibit differentially phased activity, creating stereotypical spatiotemporal circadian waves of cellular activation across the circuit. The cellular circuit pacemaking components that generate these critical emergent properties are unknown. Using intersectional genetics and real-time imaging, we show that SCN cells expressing vasoactive intestinal polypeptide (VIP) or its cognate receptor, VPAC2, are neurochemically and electrophysiologically distinct, but together they control de novo rhythmicity, setting ensemble period and phase with circuit-level spatiotemporal complexity. The VIP/VPAC2 cellular axis is therefore a neurochemically and topologically specific pacemaker hub that determines the emergent properties of the SCN timekeeper.


Assuntos
Ritmo Circadiano , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Relógios Circadianos , Criptocromos/genética , Feminino , Genes Reporter , Teste de Complementação Genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Optogenética , Oscilometria , Transdução de Sinais , Núcleo Supraquiasmático/citologia
14.
Nature ; 583(7816): 421-424, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32641825

RESUMO

The suprachiasmatic nucleus (SCN) serves as the body's master circadian clock that adaptively coordinates changes in physiology and behaviour in anticipation of changing requirements throughout the 24-h day-night cycle1-4. For example, the SCN opposes overnight adipsia by driving water intake before sleep5,6, and by driving the secretion of anti-diuretic hormone7,8 and lowering body temperature9,10 to reduce water loss during sleep11. These responses can also be driven by central osmo-sodium sensors to oppose an unscheduled rise in osmolality during the active phase12-16. However, it is unknown whether osmo-sodium sensors require clock-output networks to drive homeostatic responses. Here we show that a systemic salt injection (hypertonic saline) given at Zeitgeber time 19-a time at which SCNVP (vasopressin) neurons are inactive-excited SCNVP neurons and decreased non-shivering thermogenesis (NST) and body temperature. The effects of hypertonic saline on NST and body temperature were prevented by chemogenetic inhibition of SCNVP neurons and mimicked by optogenetic stimulation of SCNVP neurons in vivo. Combined anatomical and electrophysiological experiments revealed that osmo-sodium-sensing organum vasculosum lamina terminalis (OVLT) neurons expressing glutamic acid decarboxylase (OVLTGAD) relay this information to SCNVP neurons via an excitatory effect of γ-aminobutyric acid (GABA). Optogenetic activation of OVLTGAD neuron axon terminals excited SCNVP neurons in vitro and mimicked the effects of hypertonic saline on NST and body temperature in vivo. Furthermore, chemogenetic inhibition of OVLTGAD neurons blunted the effects of systemic hypertonic saline on NST and body temperature. Finally, we show that hypertonic saline significantly phase-advanced the circadian locomotor activity onset of mice. This effect was mimicked by optogenetic activation of the OVLTGAD→ SCNVP pathway and was prevented by chemogenetic inhibition of OVLTGAD neurons. Collectively, our findings provide demonstration that clock time can be regulated by non-photic physiologically relevant cues, and that such cues can drive unscheduled homeostatic responses via clock-output networks.


Assuntos
Relógios Circadianos/fisiologia , Vias Neurais , Neurônios/metabolismo , Sódio/metabolismo , Núcleo Supraquiasmático/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Relógios Circadianos/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Ingestão de Líquidos/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Vias Neurais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Optogenética , Organum Vasculosum/citologia , Organum Vasculosum/efeitos dos fármacos , Organum Vasculosum/enzimologia , Organum Vasculosum/fisiologia , Concentração Osmolar , Solução Salina Hipertônica/administração & dosagem , Solução Salina Hipertônica/metabolismo , Solução Salina Hipertônica/farmacologia , Sódio/administração & dosagem , Sódio/farmacologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/efeitos dos fármacos , Vasopressinas/metabolismo
15.
Int J Mol Sci ; 21(10)2020 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-32408622

RESUMO

The circadian system is an internal time-keeping system that synchronizes the behavior and physiology of an organism to the 24 h solar day. The master circadian clock, the suprachiasmatic nucleus (SCN), resides in the hypothalamus. It receives information about the environmental light/dark conditions through the eyes and orchestrates peripheral oscillators. Purinergic signaling is mediated by extracellular purines and pyrimidines that bind to purinergic receptors and regulate multiple body functions. In this review, we highlight the interaction between the circadian system and purinergic signaling to provide a better understanding of rhythmic body functions under physiological and pathological conditions.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Neurônios/fisiologia , Receptores Purinérgicos/fisiologia , Transdução de Sinais/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Humanos , Hipotálamo/citologia , Hipotálamo/fisiologia , Modelos Neurológicos , Neurônios/citologia , Núcleo Supraquiasmático/citologia
16.
Nature ; 581(7807): 194-198, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32404998

RESUMO

Daily changes in light and food availability are major time cues that influence circadian timing1. However, little is known about the circuits that integrate these time cues to drive a coherent circadian output1-3. Here we investigate whether retinal inputs modulate entrainment to nonphotic cues such as time-restricted feeding. Photic information is relayed to the suprachiasmatic nucleus (SCN)-the central circadian pacemaker-and the intergeniculate leaflet (IGL) through intrinsically photosensitive retinal ganglion cells (ipRGCs)4. We show that adult mice that lack ipRGCs from the early postnatal stages have impaired entrainment to time-restricted feeding, whereas ablation of ipRGCs at later stages had no effect. Innervation of ipRGCs at early postnatal stages influences IGL neurons that express neuropeptide Y (NPY) (hereafter, IGLNPY neurons), guiding the assembly of a functional IGLNPY-SCN circuit. Moreover, silencing IGLNPY neurons in adult mice mimicked the deficits that were induced by ablation of ipRGCs in the early postnatal stages, and acute inhibition of IGLNPY terminals in the SCN decreased food-anticipatory activity. Thus, innervation of ipRGCs in the early postnatal period tunes the IGLNPY-SCN circuit to allow entrainment to time-restricted feeding.


Assuntos
Ritmo Circadiano/fisiologia , Comportamento Alimentar/fisiologia , Luz , Vias Neurais , Retina/fisiologia , Animais , Axônios/fisiologia , Axônios/efeitos da radiação , Ritmo Circadiano/efeitos da radiação , Sinais (Psicologia) , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/efeitos da radiação , Comportamento Alimentar/efeitos da radiação , Feminino , Corpos Geniculados/citologia , Corpos Geniculados/fisiologia , Corpos Geniculados/efeitos da radiação , Masculino , Camundongos , Vias Neurais/efeitos da radiação , Neuropeptídeo Y/metabolismo , Retina/citologia , Retina/efeitos da radiação , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia , Núcleo Supraquiasmático/efeitos da radiação , Fatores de Tempo
17.
PLoS One ; 15(5): e0232748, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32379800

RESUMO

The brain's biological clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus and generates circadian rhythms in physiology and behavior. The circadian clock needs daily adjustment by light to stay synchronized (entrained) with the astronomical 24 h light/dark cycle. Light entrainment occurs via melanopsin expressing retinal ganglion cells (mRGCs) and two neurotransmitters of the retinohypothalamic tract (RHT), PACAP and glutamate, which transmit light information to the SCN neurons. In SCN neurons, light signaling involves the immediate-early genes Fos, Egr1 and the clock genes Per1 and Per2. In this study, we used PACAP deficient mice to evaluate PACAP's role in light induced gene expression of EGR1 in SCN neurons during early (ZT17) and late (ZT23) subjective night at high (300 lux) and low (10 lux) white light exposure. We found significantly lower levels of both EGR1 mRNA and protein in the SCN in PACAP deficient mice compared to wild type mice at early subjective night (ZT17) exposed to low but not high light intensity. No difference was found between the two genotypes at late night (ZT23) at neither light intensities. In conclusion, light mediated EGR1 induction in SCN neurons at early night at low light intensities is dependent of PACAP signaling. A role of PACAP in shaping synaptic plasticity during light stimulation at night is discussed.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano , Feminino , Luz , Masculino , Camundongos , Fotoperíodo , Núcleo Supraquiasmático/citologia
18.
Nat Commun ; 11(1): 1453, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32193397

RESUMO

The suprachiasmatic nucleus (SCN) circadian clock is critical for optimising daily cycles in mammalian physiology and behaviour. The roles of the various SCN cell types in communicating timing information to downstream physiological systems remain incompletely understood, however. In particular, while vasoactive intestinal polypeptide (VIP) signalling is essential for SCN function and whole animal circadian rhythmicity, the specific contributions of VIP cell output to physiological control remains uncertain. Here we reveal a key role for SCN VIP cells in central clock output. Using multielectrode recording and optogenetic manipulations, we show that VIP neurons provide coordinated daily waves of GABAergic input to target cells across the paraventricular hypothalamus and ventral thalamus, supressing their activity during the mid to late day. Using chemogenetic manipulation, we further demonstrate specific roles for this circuitry in the daily control of heart rate and corticosterone secretion, collectively establishing SCN VIP cells as influential regulators of physiological timing.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Neurônios GABAérgicos/metabolismo , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Bicuculina/farmacologia , Channelrhodopsins/química , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Relógios Circadianos/efeitos dos fármacos , Corticosterona/sangue , Corticosterona/metabolismo , Eletrodos Implantados , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Núcleo Supraquiasmático/citologia , Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/genética
19.
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
20.
J Neuroendocrinol ; 31(12): e12806, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31677199

RESUMO

Physiological circadian rhythms are orchestrated by the hypothalamic suprachiasmatic nucleus (SCN). The activity of SCN cells is synchronised by environmental signals, including light information from retinal ganglion cells (RGCs). We recently described a population of vasopressin-expressing RGCs (VP-RGC) that send axonal projections to the SCN. To determine how these VP-RGCs influence the activity of cells in the SCN, we used optogenetic tools to specifically activate their axon terminals within the SCN. Rats were intravitreally injected with a recombinant adeno-associated virus to express the channelrhodopsin-2 and the red fluorescent protein mCherry under the vasopressin promoter (VP-ChR2mCherry). In vitro recordings in acute brain slices showed that approximately 30% of ventrolateral SCN cells responded to optogenetic stimulation with an increase in firing rate that progressively increased during the first 200 seconds of stimulation and which persisted after the end of stimulation. Finally, application of a vasopressin V1A receptor antagonist dampened the response to optogenetic stimulation. Our data suggest that optogenetic stimulation of VP-RGC axons within the SCN influences the activity of SCN cells in a vasopressin-dependent manner.


Assuntos
Neurônios/fisiologia , Optogenética , Células Ganglionares da Retina/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia , Vasopressinas/fisiologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Channelrhodopsins/genética , Feminino , Masculino , Ratos , Ratos Transgênicos , Receptores de Vasopressinas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...