Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.614
Filtrar
1.
J Pineal Res ; 76(4): e12956, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38695262

RESUMEN

The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.


Asunto(s)
Ritmo Circadiano , Glucosa , Humanos , Animales , Ritmo Circadiano/fisiología , Glucosa/metabolismo , Relojes Circadianos/fisiología , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiología
2.
Chronobiol Int ; 41(6): 802-816, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38757583

RESUMEN

Stable and entrainable physiological circadian rhythms are crucial for overall health and well-being. The suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals, consists of diverse neuron types that collectively generate a circadian profile of electrical activity. However, the mechanisms underlying the regulation of endogenous neuronal excitability in the SCN remain unclear. Two-pore domain potassium channels (K2P), including TASK-3, are known to play a significant role in maintaining SCN diurnal homeostasis by inhibiting neuronal activity at night. In this study, we investigated the role of TASK-3 in SCN circadian neuronal regulation and behavioural photoentrainment using a TASK-3 global knockout mouse model. Our findings demonstrate the importance of TASK-3 in maintaining SCN hyperpolarization during the night and establishing SCN sensitivity to glutamate. Specifically, we observed that TASK-3 knockout mice lacked diurnal variation in resting membrane potential and exhibited altered glutamate sensitivity both in vivo and in vitro. Interestingly, despite these changes, the mice lacking TASK-3 were still able to maintain relatively normal circadian behaviour.


Asunto(s)
Ritmo Circadiano , Ratones Noqueados , Canales de Potasio de Dominio Poro en Tándem , Núcleo Supraquiasmático , Animales , Ritmo Circadiano/fisiología , Núcleo Supraquiasmático/fisiología , Núcleo Supraquiasmático/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Ratones , Masculino , Ratones Endogámicos C57BL , Conducta Animal/fisiología , Ácido Glutámico/metabolismo , Neuronas/fisiología , Neuronas/metabolismo , Potenciales de la Membrana/fisiología , Canales de Potasio
3.
Int J Mol Sci ; 25(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38732079

RESUMEN

Long-term spaceflight is known to induce disruptions in circadian rhythms, which are driven by a central pacemaker located in the suprachiasmatic nucleus (SCN) of the hypothalamus, but the underlying molecular mechanisms remain unclear. Here, we developed a rat model that simulated microgravity and isolation environments through tail suspension and isolation (TSI). We found that the TSI environment imposed circadian disruptions to the core body temperature, heart rate, and locomotor-activity rhythms of rats, especially in the amplitude of these rhythms. In TSI model rats' SCNs, the core circadian gene NR1D1 showed higher protein but not mRNA levels along with decreased BMAL1 levels, which indicated that NR1D1 could be regulated through post-translational regulation. The autophagosome marker LC3 could directly bind to NR1D1 via the LC3-interacting region (LIR) motifs and induce the degradation of NR1D1 in a mitophagy-dependent manner. Defects in mitophagy led to the reversal of NR1D1 degradation, thereby suppressing the expression of BMAL1. Mitophagy deficiency and subsequent mitochondrial dysfunction were observed in the SCN of TSI models. Urolithin A (UA), a mitophagy activator, demonstrated an ability to enhance the amplitude of core body temperature, heart rate, and locomotor-activity rhythms by prompting mitophagy induction to degrade NR1D1. Cumulatively, our results demonstrate that mitophagy exerts circadian control by regulating NR1D1 degradation, revealing mitophagy as a potential target for long-term spaceflight as well as diseases with SCN circadian disruption.


Asunto(s)
Factores de Transcripción ARNTL , Ritmo Circadiano , Mitofagia , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Animales , Ratas , Ritmo Circadiano/fisiología , Masculino , Factores de Transcripción ARNTL/metabolismo , Factores de Transcripción ARNTL/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Simulación de Ingravidez , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Temperatura Corporal , Frecuencia Cardíaca , Ratas Sprague-Dawley , Proteolisis
4.
Fluids Barriers CNS ; 21(1): 46, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802875

RESUMEN

Choroid plexus (ChP), the brain structure primarily responsible for cerebrospinal fluid production, contains a robust circadian clock, whose role remains to be elucidated. The aim of our study was to [1] identify rhythmically controlled cellular processes in the mouse ChP and [2] assess the role and nature of signals derived from the master clock in the suprachiasmatic nuclei (SCN) that control ChP rhythms. To accomplish this goal, we used various mouse models (WT, mPer2Luc, ChP-specific Bmal1 knockout) and combined multiple experimental approaches, including surgical lesion of the SCN (SCNx), time-resolved transcriptomics, and single cell luminescence microscopy. In ChP of control (Ctrl) mice collected every 4 h over 2 circadian cycles in darkness, we found that the ChP clock regulates many processes, including the cerebrospinal fluid circadian secretome, precisely times endoplasmic reticulum stress response, and controls genes involved in neurodegenerative diseases (Alzheimer's disease, Huntington's disease, and frontotemporal dementia). In ChP of SCNx mice, the rhythmicity detected in vivo and ex vivo was severely dampened to a comparable extent as in mice with ChP-specific Bmal1 knockout, and the dampened cellular rhythms were restored by daily injections of dexamethasone in mice. Our data demonstrate that the ChP clock controls tissue-specific gene expression and is strongly dependent on the presence of a functional connection with the SCN. The results may contribute to the search for a novel link between ChP clock disruption and impaired brain health.


Asunto(s)
Plexo Coroideo , Relojes Circadianos , Núcleo Supraquiasmático , Animales , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiología , Plexo Coroideo/metabolismo , Plexo Coroideo/fisiología , Relojes Circadianos/fisiología , Ratones , Ratones Endogámicos C57BL , Ritmo Circadiano/fisiología , Masculino , Ratones Noqueados , Factores de Transcripción ARNTL/metabolismo , Factores de Transcripción ARNTL/genética
5.
J Neurosci Res ; 102(4): e25331, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38651314

RESUMEN

Circadian rhythms synchronize to light through the retinohypothalamic tract (RHT), which is a bundle of axons coming from melanopsin retinal ganglion cells, whose synaptic terminals release glutamate to the ventral suprachiasmatic nucleus (SCN). Activation of AMPA-kainate and NMDA postsynaptic receptors elicits the increase in intracellular calcium required for triggering the signaling cascade that ends in phase shifts. During aging, there is a decline in the synchronization of circadian rhythms to light. With electrophysiological (whole-cell patch-clamp) and immunohistochemical assays, in this work, we studied pre- and postsynaptic properties between the RHT and ventral SCN neurons in young adult (P90-120) and old (P540-650) C57BL/6J mice. Incremental stimulation intensities (applied on the optic chiasm) induced much lesser AMPA-kainate postsynaptic responses in old animals, implying a lower recruitment of RHT fibers. Conversely, a higher proportion of old SCN neurons exhibited synaptic facilitation, and variance-mean analysis indicated an increase in the probability of release in RHT terminals. Moreover, both spontaneous and miniature postsynaptic events displayed larger amplitudes in neurons from aged mice, whereas analysis of the NMDA and AMPA-kainate components (evoked by RHT electrical stimulation) disclosed no difference between the two ages studied. Immunohistochemistry revealed a bigger size in the puncta of vGluT2, GluN2B, and GluN2A of elderly animals, and the number of immunopositive particles was increased, but that of PSD-95 was reduced. All these synaptic adaptations could be part of compensatory mechanisms in the glutamatergic signaling to ameliorate the loss of RHT terminals in old animals.


Asunto(s)
Envejecimiento , Ácido Glutámico , Ratones Endogámicos C57BL , Núcleo Supraquiasmático , Transmisión Sináptica , Animales , Ratones , Núcleo Supraquiasmático/fisiología , Núcleo Supraquiasmático/metabolismo , Transmisión Sináptica/fisiología , Envejecimiento/fisiología , Ácido Glutámico/metabolismo , Masculino , Potenciales Postsinápticos Excitadores/fisiología , Vías Visuales/fisiología , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Técnicas de Placa-Clamp , Receptores de N-Metil-D-Aspartato/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo
6.
Peptides ; 177: 171229, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38663583

RESUMEN

Circadian rhythms optimally regulate numerous physiological processes in an organism and synchronize them with the external environment. The suprachiasmatic nucleus (SCN), the center of the circadian clock in mammals, is composed of multiple cell types that form a network that provides the basis for the remarkable stability of the circadian clock. Among the neuropeptides expressed in the SCN, arginine vasopressin (AVP) has attracted much attention because of its deep involvement in the function of circadian rhythms, as elucidated in particular by studies using genetically engineered mice. This review briefly summarizes the current knowledge on the peptidergic distribution and topographic neuronal organization in the SCN, the molecular mechanisms of the clock genes, and the relationship between the SCN and peripheral clocks. With respect to the physiological roles of AVP and AVP-expressing neurons, in addition to a sex-dependent action of AVP in the SCN, studies using AVP receptor knockout mice and mice genetically manipulated to alter the clock properties of AVP neurons are summarized here, highlighting its importance in maintaining circadian homeostasis and its potential as a target for therapeutic interventions.


Asunto(s)
Arginina Vasopresina , Ritmo Circadiano , Homeostasis , Núcleo Supraquiasmático , Animales , Arginina Vasopresina/metabolismo , Arginina Vasopresina/genética , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiología , Homeostasis/genética , Ritmo Circadiano/fisiología , Ritmo Circadiano/genética , Humanos , Ratones , Relojes Circadianos/genética , Relojes Circadianos/fisiología , Neuronas/metabolismo , Ratones Noqueados , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo
7.
J Neurosci ; 44(18)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38485259

RESUMEN

Sleep is regulated by homeostatic sleep drive and the circadian clock. While tremendous progress has been made in elucidating the molecular components of the core circadian oscillator, the output mechanisms by which this robust oscillator generates rhythmic sleep behavior remain poorly understood. At the cellular level, growing evidence suggests that subcircuits in the master circadian pacemaker suprachiasmatic nucleus (SCN) in mammals and in the clock network in Drosophila regulate distinct aspects of sleep. Thus, to identify novel molecules regulating the circadian timing of sleep, we conducted a large-scale screen of mouse SCN-enriched genes in Drosophila Here, we show that Tob (Transducer of ERB-B2) regulates the timing of sleep onset at night in female fruit flies. Knockdown of Tob pan-neuronally, either constitutively or conditionally, advances sleep onset at night. We show that Tob is specifically required in "evening neurons" (the LNds and the fifth s-LNv) of the clock network for proper timing of sleep onset. Tob levels cycle in a clock-dependent manner in these neurons. Silencing of these "evening" clock neurons results in an advanced sleep onset at night, similar to that seen with Tob knockdown. Finally, sharp intracellular recordings demonstrate that the amplitude and kinetics of LNd postsynaptic potentials (PSPs) cycle between day and night, and this cycling is attenuated with Tob knockdown in these cells. Our data suggest that Tob acts as a clock output molecule in a subset of clock neurons to potentiate their activity in the evening and enable the proper timing of sleep onset at night.


Asunto(s)
Ritmo Circadiano , Proteínas de Drosophila , Drosophila , Sueño , Animales , Femenino , Animales Modificados Genéticamente , Ritmo Circadiano/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neuronas/fisiología , Sueño/fisiología , Núcleo Supraquiasmático/fisiología
8.
J Biol Rhythms ; 39(2): 135-165, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38366616

RESUMEN

It has been 50 years since the suprachiasmatic nucleus (SCN) was first identified as the central circadian clock and 25 years since the last overview of developments in the field was published in the Journal of Biological Rhythms. Here, we explore new mechanisms and concepts that have emerged in the subsequent 25 years. Since 1997, methodological developments, such as luminescent and fluorescent reporter techniques, have revealed intricate relationships between cellular and network-level mechanisms. In particular, specific neuropeptides such as arginine vasopressin, vasoactive intestinal peptide, and gastrin-releasing peptide have been identified as key players in the synchronization of cellular circadian rhythms within the SCN. The discovery of multiple oscillators governing behavioral and physiological rhythms has significantly advanced our understanding of the circadian clock. The interaction between neurons and glial cells has been found to play a crucial role in regulating these circadian rhythms within the SCN. Furthermore, the properties of the SCN network vary across ontogenetic stages. The application of cell type-specific genetic manipulations has revealed components of the functional input-output system of the SCN and their correlation with physiological functions. This review concludes with the high-risk effort of identifying open questions and challenges that lie ahead.


Asunto(s)
Ritmo Circadiano , Neuropéptidos , Ritmo Circadiano/fisiología , Neuropéptidos/metabolismo , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/metabolismo , Péptido Liberador de Gastrina/metabolismo
9.
Eur J Neurosci ; 59(7): 1723-1742, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38326974

RESUMEN

The circadian clock orchestrates many physiological and behavioural rhythms in mammals with 24-h periodicity, through a hierarchical organisation, with the central clock located in the suprachiasmatic nucleus (SCN) in the hypothalamus. The circuits of the SCN generate circadian rhythms with precision, relying on intrinsic coupling mechanisms, for example, neurotransmitters like arginine vasopressin (AVP), vasoactive intestinal peptide (VIP), neuronal gamma-aminobutyric acid (GABA) signalling and astrocytes connected by gap junctions composed of connexins (Cx). In female rodents, the presence of estrogen receptors (ERs) in the dorsal SCN suggests an influence of estrogen (E2) on the circuit timekeeping that could regulate circadian rhythm and coupling. To investigate this, we used SCN explants together with hypothalamic neurons and astrocytes. First, we showed that E2 stabilised the circadian amplitude in the SCN when rAVPs (receptor-associated vasopressin peptides) were inhibited. However, the phase delay induced by VIPAC2 (VIP receptors) inhibition remained unaffected by E2. We then showed that E2 exerted its effects in the SCN via ERß (estrogen receptor beta), resulting in increased expression of Cx36 and Cx43. Notably, specific inhibition of both connexins resulted in a significant reduction in circadian amplitude within the SCN. Remarkably, E2 restored the period with inhibited Cx36 but not with Cx43 inhibition. This implies that the network between astrocytes and neurons, responsible for coupling in the SCN, can be reinforced through E2. In conclusion, these findings provide new insights into how E2 regulates circadian rhythms ex vivo in an ERß-dependent manner, underscoring its crucial role in fortifying the SCN's rhythm.


Asunto(s)
Conexina 43 , Receptor beta de Estrógeno , Animales , Femenino , Conexina 43/metabolismo , Receptor beta de Estrógeno/metabolismo , Núcleo Supraquiasmático/fisiología , Ritmo Circadiano/fisiología , Uniones Comunicantes/metabolismo , Conexinas/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Péptido Intestinal Vasoactivo/metabolismo , Estrógenos/farmacología , Mamíferos/metabolismo
10.
J Neurosci ; 44(8)2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38238074

RESUMEN

The suprachiasmatic nucleus (SCN) is the central clock for circadian rhythms. Animal studies have revealed daily rhythms in the neuronal activity in the SCN. However, the circadian activity of the human SCN has remained elusive. In this study, to reveal the diurnal variation of the SCN activity in humans, we localized the SCN by employing an areal boundary mapping technique to resting-state functional images and investigated the SCN activity using perfusion imaging. In the first experiment (n = 27, including both sexes), we scanned each participant four times a day, every 6 h. Higher activity was observed at noon, while lower activity was recorded in the early morning. In the second experiment (n = 20, including both sexes), the SCN activity was measured every 30 min for 6 h from midnight to dawn. The results showed that the SCN activity gradually decreased and was not associated with the electroencephalography. Furthermore, the SCN activity was compatible with the rodent SCN activity after switching off the lights. These results suggest that the diurnal variation of the human SCN follows the zeitgeber cycles of nocturnal and diurnal mammals and is modulated by physical lights rather than the local time.


Asunto(s)
Ritmo Circadiano , Núcleo Supraquiasmático , Masculino , Animales , Femenino , Humanos , Ritmo Circadiano/fisiología , Núcleo Supraquiasmático/fisiología , Roedores , Mamíferos , Neuronas
11.
FASEB J ; 38(1): e23348, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38084798

RESUMEN

A robust endogenous clock is required for proper function of many physiological processes. The suprachiasmatic nucleus (SCN) constitutes our central circadian clock and allows us to adapt to daily changes in the environment. Aging can cause a decline in the amplitude of circadian rhythms in SCN and peripheral clocks, which contributes to increased risk of several chronic diseases. Strengthening clock function would therefore be an effective strategy to improve health. A high-throughput chemical screening has identified clock-enhancing molecule 3 (CEM3) as small molecule that increases circadian rhythm amplitude in cell lines and SCN explants. It is, however, currently not known whether CEM3 acts by enhancing the amplitude of individual single-cell oscillators or by enhancing synchrony among neurons. In view of CEM3's potential, it is of evident importance to clarify the mode of action of CEM3. Here, we investigated the effects of CEM3 on single-cell PERIOD2::LUCIFERASE rhythms in mouse SCN explants. CEM3 increased the amplitude in approximately 80%-90% of the individual cells in the SCN without disrupting the phase and/or period of their rhythms. Noticeably, CEM3's effect on amplitude is independent of the cell's initial amplitude. These findings make CEM3 a potential therapeutic candidate to restore compromised amplitude in circadian rhythms and will boost the development of other molecular approaches to improve health.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Ratones , Animales , Ritmo Circadiano/fisiología , Núcleo Supraquiasmático/fisiología , Relojes Circadianos/fisiología , Luciferasas/metabolismo , Neuronas/metabolismo
12.
Endocrinology ; 165(1)2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38128120

RESUMEN

Repeated or chronic stress can change the phase of peripheral circadian rhythms. Melatonin (Mel) is thought to be a circadian clock-controlled signal that might play a role in synchronizing peripheral rhythms, in addition to its direct suppressing effects on the stress axis. In this study we test whether Mel can reduce the social-defeat stress-induced phase shifts in peripheral rhythms, either by modulating circadian phase or by modulating the stress axis. Two experiments were performed with male Mel-deficient C57BL/6J mice carrying the circadian reporter gene construct (PER2::LUC). In the first experiment, mice received night-restricted (ZT11-21) Mel in their drinking water, resulting in physiological levels of plasma Mel peaking in the early dark phase. This treatment facilitated re-entrainment of the activity rhythm to a shifted light-dark cycle, but did not prevent the stress-induced (ZT21-22) reduction of activity during stress days. Also, this treatment did not attenuate the phase-delaying effects of stress in peripheral clocks in the pituitary, lung, and kidney. In a second experiment, pituitary, lung, and kidney collected from naive mice (ZT22-23), were treated with Mel, dexamethasone (Dex), or a combination of the two. Dex application affected PER2 rhythms in the pituitary, kidney, and lung by changing period, phase, or both. Administering Mel did not influence PER2 rhythms nor did it alleviate Dex-induced delays in PER2 rhythms in those tissues. We conclude that exogenous Mel is insufficient to affect peripheral PER2 rhythms and reduce stress effects on locomotor activity and phase changes in peripheral tissues.


Asunto(s)
Relojes Circadianos , Melatonina , Ratones , Masculino , Animales , Melatonina/farmacología , Luz , Núcleo Supraquiasmático/fisiología , Ratones Endogámicos C57BL , Ritmo Circadiano/fisiología , Relojes Circadianos/fisiología
13.
J Physiol ; 601(21): 4737-4749, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37777993

RESUMEN

Many neurons of the mammalian master circadian oscillator in the suprachiasmatic nuclei (SCN) respond to light pulses with irradiance-dependent changes in firing. Here, we set out to better understand this irradiance coding ability by considering how the SCN tracks more continuous changes in irradiance at both population and single unit level. To this end, we recorded extracellular activity in the SCN of anaesthetised mice presented with up + down irradiance staircase stimuli covering moonlight to daylight conditions and incorporating epochs with steady light or superimposed higher frequency modulations (temporal white noise (WN) and frequency/contrast chirps). Single unit activity was extracted by spike sorting. The population response of SCN units to this stimulus was a progressive increase in firing rate at higher irradiances. This relationship was symmetrical for up vs. down phases of the ramp in the presence of white noise or chirps but exhibited hysteresis for steady light, with firing systematically higher during increasing irradiance. Single units also showed a monotonic relationship between firing and irradiance but exhibited diversity not only in response polarity (increases vs. decreases in firing), but also in the sensitivity (EC50 ) and slope of fitted functions. These data show that individual SCN neurons exhibit monotonic relationships between irradiance and firing rate but differ in the irradiance range over which they respond. This property may help the SCN to encode the large differences in irradiance found in nature using neurons with a constrained range of firing rates. KEY POINTS: Daily changes in environmental light (irradiance) entrain the suprachiasmatic nucleus (SCN) circadian clock. The mouse SCN shows graded increases in neurophysiological activity with light pulses of increasing irradiance. We show that this monotonic relationship between firing rate and irradiance is retained at population and single unit level when probed with more naturalistic staircase increases and decreases in irradiance. The irradiance response is more reliable in the presence of ongoing higher temporal frequency modulations in light intensity than under steady light. Single units varied in sensitivity allowing the population to cover a wide range of irradiances. Irradiance coding in the SCN has characteristics of a sparse code with individual neurons tracking different portions of the natural irradiance range. This property may address the challenge of encoding a 109 -fold day:night difference in irradiance within the constrained range of firing rates available to individual neurons.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Ratones , Animales , Ritmo Circadiano/fisiología , Núcleo Supraquiasmático/fisiología , Neuronas/fisiología , Luz , Mamíferos
14.
Sci Rep ; 13(1): 16974, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37813987

RESUMEN

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.


Asunto(s)
Calcio , Neuronas del Núcleo Supraquiasmático , Ratones , Animales , Calcio/metabolismo , Núcleo Supraquiasmático/fisiología , Ritmo Circadiano/fisiología , Neuronas del Núcleo Supraquiasmático/metabolismo , Neuronas/metabolismo , Transactivadores/metabolismo , Calcio de la Dieta/metabolismo , Mamíferos/metabolismo
15.
Rejuvenation Res ; 26(6): 229-241, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37847148

RESUMEN

Circadian rhythms (CRs) are 24-hour periodic oscillations governed by an endogenous circadian pacemaker located in the suprachiasmatic nucleus (SCN), which organizes the physiology and behavior of organisms. Circadian rhythm disruption (CRD) is also indicative of the aging process. In mammals, melatonin is primarily synthesized in the pineal gland and participates in a variety of multifaceted intracellular signaling networks and has been shown to synchronize CRs. Endogenous melatonin synthesis and its release tend to decrease progressively with advancing age. Older individuals experience frequent CR disruption, which hastens the process of aging. A profound understanding of the relationship between CRs and aging has the potential to improve existing treatments and facilitate development of novel chronotherapies that target age-related disorders. This review article aims to examine the circadian regulatory mechanisms in which melatonin plays a key role in signaling. We describe the basic architecture of the molecular circadian clock and its functional decline with age in detail. Furthermore, we discuss the role of melatonin in regulation of the circadian pacemaker and redox homeostasis during aging. Moreover, we also discuss the protective effect of exogenous melatonin supplementation in age-dependent CR disruption, which sheds light on this pleiotropic molecule and how it can be used as an effective chronotherapeutic medicine.


Asunto(s)
Relojes Circadianos , Melatonina , Humanos , Animales , Melatonina/farmacología , Melatonina/fisiología , Ritmo Circadiano/fisiología , Relojes Circadianos/fisiología , Núcleo Supraquiasmático/fisiología , Envejecimiento/fisiología , Mamíferos
16.
Curr Biol ; 33(20): 4343-4352.e4, 2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37725978

RESUMEN

Short sleep is linked to disturbances in glucose metabolism and may induce a prediabetic condition. The biological clock in the suprachiasmatic nucleus (SCN) regulates the glucose rhythm in the circulation and the sleep-wake cycle. SCN vasopressin neurons (SCNVP) control daily glycemia by regulating the entrance of glucose into the arcuate nucleus (ARC). Thus, we hypothesized that sleep delay may influence SCN neuronal activity. We, therefore, investigated the role of SCNVP when sleep is disrupted by forced locomotor activity. After 2 h of sleep delay, rats exhibited decreased SCNVP neuronal activity, a decrease in the glucose transporter GLUT1 expression in tanycytes lining the third ventricle, lowered glucose entrance into the ARC, and developed hyperglycemia. The association between reduced SCNVP neuronal activity and hyperglycemia in sleep-delayed rats was evidenced by injecting intracerebroventricular vasopressin; this increased GLUT1 immunoreactivity in tanycytes, thus promoting normoglycemia. Following sleep recovery, glucose levels decreased, whereas SCNVP neuronal activity increased. These results imply that sleep-delay-induced changes in SCNVP activity lead to glycemic impairment, inferring that disruption of biological clock function might represent a critical step in developing type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperglucemia , Ratas , Animales , Transportador de Glucosa de Tipo 1/metabolismo , Ritmo Circadiano/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Núcleo Supraquiasmático/fisiología , Sueño , Glucosa/metabolismo , Hiperglucemia/metabolismo , Vasopresinas/metabolismo
17.
STAR Protoc ; 4(4): 102618, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37756154

RESUMEN

The mammalian suprachiasmatic nucleus (SCN) is the principal circadian clock that synchronizes daily behavioral and physiological responses in response to environmental cues. Here, we present a protocol for harvesting mouse SCN by vibrating microtome for diurnal transcriptome analysis. We describe steps for mouse entrainment, isolation of the SCN, tissue preparation, slicing with a vibratome, and handling of the harvested SCN for RNA extraction. This protocol can also be used for harvesting other mammalian brain regions for genomic studies.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Ratones , Animales , Ritmo Circadiano/genética , Núcleo Supraquiasmático/fisiología , Perfilación de la Expresión Génica , Técnicas Histológicas , Mamíferos
18.
J Biol Rhythms ; 38(6): 601-616, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37529986

RESUMEN

Synchronizing the circadian timing system (CTS) to external light/dark cycles is crucial for homeostasis maintenance and environmental adaptation. The CTS is organized hierarchically, with the central pacemaker located in the suprachiasmatic nuclei (SCN) generating coherent oscillations that are entrained to light/dark cycles. These oscillations regulate the release of glucocorticoids by the hypothalamus-pituitary-adrenal (HPA) axis, which acts as a systemic entrainer of peripheral clocks throughout the body. The SCN adjusts its network plasticity in response to variations in photoperiod, leading to changes in the rhythmic release of glucocorticoids and ultimately impacting peripheral clocks. However, the effects of photoperiod-induced variations of glucocorticoids on the synchronization of peripheral clocks are not fully understood, and the interaction between jetlag adaption and photoperiod changes is unclear. This study presents a semi-mechanistic mathematical model to investigate how the CTS responds to changes in photoperiod. Specifically, the study focuses on the entrainment properties of a system composed of the SCN, HPA axis, and peripheral clocks. The results show that high-amplitude glucocorticoid rhythms lead to a more coherent phase distribution in the periphery. In addition, our study investigates the effect of photoperiod exposure on jetlag recovery time and phase shift, proposing different interventional strategies for eastward and westward jetlag. The findings suggest that decreasing photic exposure before jetlag during eastward traveling and after jetlag during westward traveling can accelerate jetlag readaptation. The study provides insights into the mechanisms of CTS organization and potential recovery strategies for transitions between time zones and lighting zones.


Asunto(s)
Relojes Circadianos , Fotoperiodo , Ritmo Circadiano/fisiología , Relojes Circadianos/fisiología , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Núcleo Supraquiasmático/fisiología , Glucocorticoides/farmacología , Modelos Teóricos
19.
J Gen Physiol ; 155(9)2023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37584659

RESUMEN

JGP study (Hermanstyne et al. 2023. J. Gen. Physiol.https://doi.org/10.1085/jgp.202213310) shows that Kv12-encoded K+ currents reduce the repetitive firing rates of SCN neurons at night, thereby regulating daily oscillations in the master circadian pacemaker.


Asunto(s)
Ritmo Circadiano , Núcleo Supraquiasmático , Ritmo Circadiano/fisiología , Núcleo Supraquiasmático/fisiología , Neuronas/fisiología
20.
J Neurosci Res ; 101(11): 1737-1756, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37551165

RESUMEN

The circadian clock is one of the most important homeostatic systems regulating the majority of physiological functions. Its proper development contributes significantly to the maintenance of health in adulthood. Methadone is recommended for the treatment of opioid use disorders during pregnancy, increasing the number of children prenatally exposed to long-acting opioids. Although early-life opioid exposure has been studied for a number of behavioral and physiological changes observed later in life, information on the relationship between the effects of methadone exposure and circadian system development is lacking. Using a rat model, we investigated the effects of prenatal and early postnatal methadone administration on the maturation of the circadian clockwork in the suprachiasmatic nucleus (SCN) and liver, the rhythm of aralkylamine N-acetyltransferase (AA-NAT) activity in the pineal gland, and gene expression in the livers of 20-day-old rats. Our data show that repeated administration of methadone to pregnant and lactating mothers has significant effect on rhythmic gene expression in the SCN and livers and on the rhythm of AA-NAT in the offspring. Similar to previous studies with morphine, the rhythm amplitudes of the clock genes in the SCN and liver were unchanged or enhanced. However, six of seven specific genes in the liver showed significant downregulation of their expression, compared to the controls in at least one experimental group. Importantly, the amplitude of the AA-NAT rhythm was significantly reduced in all methadone-treated groups. As there is a strong correlation with melatonin levels, this result could be of importance for clinical practice.


Asunto(s)
Melatonina , Glándula Pineal , Embarazo , Femenino , Ratas , Animales , Metadona/metabolismo , Metadona/farmacología , Lactancia , Ritmo Circadiano/fisiología , Glándula Pineal/metabolismo , Melatonina/farmacología , Núcleo Supraquiasmático/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA