Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(10): E2437-E2446, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29463694

RESUMO

Circadian disruption as a result of shift work is associated with adverse metabolic consequences. Internal desynchrony between the phase of the suprachiasmatic nuclei (SCN) and peripheral clocks is widely believed to be a major factor contributing to these adverse consequences, but this hypothesis has never been tested directly. A GABAergic Cre driver combined with conditional casein kinase mutations (Vgat-Cre+CK1δfl/flεfl/+ ) was used to lengthen the endogenous circadian period in GABAergic neurons, including the SCN, but not in peripheral tissues, to create a Discordant mouse model. These mice had a long (27.4 h) behavioral period to which peripheral clocks entrained in vivo, albeit with an advanced phase (∼6 h). Thus, in the absence of environmental timing cues, these mice had internal desynchrony between the SCN and peripheral clocks. Surprisingly, internal desynchrony did not result in obesity in this model. Instead, Discordant mice had reduced body mass compared with Cre-negative controls on regular chow and even when challenged with a high-fat diet. Similarly, internal desynchrony failed to induce glucose intolerance or disrupt body temperature and energy expenditure rhythms. Subsequently, a lighting cycle of 2-h light/23.5-h dark was used to create a similar internal desynchrony state in both genotypes. Under these conditions, Discordant mice maintained their lower body mass relative to controls, suggesting that internal desynchrony did not cause the lowered body mass. Overall, our results indicate that internal desynchrony does not necessarily lead to metabolic derangements and suggest that additional mechanisms contribute to the adverse metabolic consequences observed in circadian disruption protocols.


Assuntos
Caseína Quinase 1 épsilon/genética , Caseína Quinase Idelta/genética , Relógios Circadianos , Neurônios GABAérgicos/enzimologia , Núcleo Supraquiasmático/fisiologia , Animais , Caseína Quinase 1 épsilon/deficiência , Caseína Quinase Idelta/deficiência , Ritmo Circadiano , Feminino , Técnicas de Inativação de Genes , Inativação Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Supraquiasmático/enzimologia
2.
J Pineal Res ; 69(1): e12654, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32243642

RESUMO

Disturbing the circadian regulation of physiology by disruption of the rhythmic environment is associated with adverse health outcomes but the underlying mechanisms are unknown. Here, the response of central and peripheral circadian clocks to an advance or delay of the light-dark cycle was determined in mice. This identified transient damping of peripheral clocks as a consequence of an advanced light-dark cycle. Similar depression of peripheral rhythm amplitude was observed in mice exposed to repeated phase shifts. To assess the metabolic consequences of such peripheral amplitude depression in isolation, temporally chimeric mice lacking a functional central clock (Vgat-Cre+ Bmal1fl/fl ) were housed in the absence of environmental rhythmicity. In vivo PER2::LUC bioluminescence imaging of anesthetized and freely moving mice revealed that this resulted in a state of peripheral amplitude depression, similar in severity to that observed transiently following an advance of the light-dark cycle. Surprisingly, our mice did not show alterations in body mass or glucose tolerance in males or females on regular or high-fat diets. Overall, our results identify transient damping of peripheral rhythm amplitude as a consequence of exposure to an advanced light-dark cycle but chronic damping of peripheral clocks in isolation is insufficient to induce adverse metabolic outcomes in mice.


Assuntos
Comportamento Animal , Relógios Biológicos , Ritmo Circadiano , Intolerância à Glucose , Obesidade , Animais , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/fisiopatologia , Camundongos , Camundongos Transgênicos , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia
3.
J Exp Biol ; 221(Pt 15)2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29903839

RESUMO

Daily torpor is used by small mammals to reduce daily energy expenditure in response to energetic challenges. Optimizing the timing of daily torpor allows mammals to maximize its energetic benefits and, accordingly, torpor typically occurs in the late night and early morning in most species. However, the regulatory mechanisms underlying such temporal regulation have not been elucidated. Direct control by the circadian clock and indirect control through the timing of food intake have both been suggested as possible mechanisms. Here, feeding cycles outside of the circadian range and brain-specific mutations of circadian clock genes (Vgat-Cre+ CK1δfl/fl εfl/+ ; Vgat-Cre+ Bmal1fl/fl ) were used to separate the roles of the circadian clock and food timing in controlling the timing of daily torpor in mice. These experiments revealed that the timing of daily torpor is transiently inhibited by feeding, while the circadian clock is the major determinant of the timing of torpor. Torpor never occurred during the early part of the circadian active phase, but was preferentially initiated late in the subjective night. Food intake disrupted torpor in the first 4-6 h after feeding by preventing or interrupting torpor bouts. Following interruption, re-initiation of torpor was unlikely until after the next circadian active phase. Overall, these results demonstrate that feeding transiently inhibits torpor while the central circadian clock gates the timing of daily torpor in response to energetic challenges by restricting the initiation of torpor to a specific circadian phase.


Assuntos
Relógios Circadianos/genética , Ingestão de Alimentos/fisiologia , Torpor/fisiologia , Animais , Temperatura Corporal/fisiologia , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Feminino , Locomoção , Masculino , Camundongos , Mutação , Fatores de Tempo
4.
Proc Natl Acad Sci U S A ; 108(41): 17219-24, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21969555

RESUMO

The circadian clock in the mammalian hypothalamic suprachiasmatic nucleus (SCN) is entrained by the ambient light/dark cycle, which differentially acts to cause the clock to advance or delay. Light-induced changes in the rhythmic expression of SCN clock genes are believed to be a critical step in this process, but how the two entrainment modalities--advances vs. delays--engage the molecular clockwork remains incompletely understood. We investigated molecular substrates of photic entrainment of the clock in the SCN by stably entraining hamsters to T cycles (non-24-h light/dark cycles) consisting of a single 1-h light pulse repeated as either a short (23.33-h) or a long (24.67-h) cycle; under these conditions, the light pulse of the short cycle acts as "dawn," whereas that of the long cycle acts as "dusk." Analyses of the expression of the photoinducible and rhythmic clock genes Period 1 and 2 (Per1 and Per2) in the SCN revealed fundamental differences under these two entrainment modes. Light at dawn advanced the clock, advancing the onset of the Per1 mRNA rhythm and acutely increasing mRNA transcription, whereas light at dusk delayed the clock, delaying the offset of the Per2 mRNA rhythm and tonically increasing mRNA stability. The results suggest that the underlying molecular mechanisms of circadian entrainment differ with morning (advancing) or evening (delaying) light exposure, and such differences may reflect how entrainment takes place in nocturnal animals under natural conditions.


Assuntos
Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Proteínas Circadianas Period/genética , Núcleo Supraquiasmático/fisiologia , Animais , Cricetinae , Expressão Gênica , Masculino , Mesocricetus , Estimulação Luminosa , Fotoperíodo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
Proc Natl Acad Sci U S A ; 108(39): 16451-6, 2011 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-21930935

RESUMO

Mounting evidence suggests that PERIOD (PER) proteins play a central role in setting the speed (period) and phase of the circadian clock. Pharmacological and genetic studies have shown that changes in PER phosphorylation kinetics are associated with changes in circadian rhythm period and phase, which can lead to sleep disorders such as Familial Advanced Sleep Phase Syndrome in humans. We and others have shown that casein kinase 1δ and ε (CK1δ/ε) are essential PER kinases, but it is clear that additional, unknown mechanisms are also crucial for regulating the kinetics of PER phosphorylation. Here we report that circadian periodicity is determined primarily through PER phosphorylation kinetics set by the balance between CK1δ/ε and protein phosphatase 1 (PP1). In CK1δ/ε-deficient cells, PER phosphorylation is severely compromised and nonrhythmic, and the PER proteins are constitutively cytoplasmic. However, when PP1 is disrupted, PER phosphorylation is dramatically accelerated; the same effect is not seen when PP2A is disrupted. Our work demonstrates that the speed and rhythmicity of PER phosphorylation are controlled by the balance between CK1δ/ε and PP1, which in turn determines the period of the circadian oscillator. Thus, our findings provide clear insights into the molecular basis of how the period and phase of our daily rhythms are determined.


Assuntos
Caseína Quinase I/metabolismo , Ritmo Circadiano , Proteínas Circadianas Period/fisiologia , Proteína Fosfatase 1/metabolismo , Animais , Eletroforese em Gel de Poliacrilamida , Camundongos , Proteínas Circadianas Period/metabolismo , Fosforilação
6.
J Biol Rhythms ; 39(2): 135-165, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38366616

RESUMO

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.


Assuntos
Ritmo Circadiano , Neuropeptídeos , Ritmo Circadiano/fisiologia , Neuropeptídeos/metabolismo , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Liberador de Gastrina/metabolismo
7.
Am J Physiol Endocrinol Metab ; 303(6): E762-76, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22785238

RESUMO

Insulinoma-associated protein (IA)-2 and IA-2ß are transmembrane proteins involved in neurotransmitter secretion. Mice with targeted disruption of both IA-2 and IA-2ß (double-knockout, or DKO mice) have numerous endocrine and physiological disruptions, including disruption of circadian and diurnal rhythms. In the present study, we have assessed the impact of disruption of IA-2 and IA-2ß on molecular rhythms in the brain and peripheral oscillators. We used in situ hybridization to assess molecular rhythms in the hypothalamic suprachiasmatic nuclei (SCN) of wild-type (WT) and DKO mice. The results indicate significant disruption of molecular rhythmicity in the SCN, which serves as the central pacemaker regulating circadian behavior. We also used quantitative PCR to assess gene expression rhythms in peripheral tissues of DKO, single-knockout, and WT mice. The results indicate significant attenuation of gene expression rhythms in several peripheral tissues of DKO mice but not in either single knockout. To distinguish whether this reduction in rhythmicity reflects defective oscillatory function in peripheral tissues or lack of entrainment of peripheral tissues, animals were injected with dexamethasone daily for 15 days, and then molecular rhythms were assessed throughout the day after discontinuation of injections. Dexamethasone injections improved gene expression rhythms in liver and heart of DKO mice. These results are consistent with the hypothesis that peripheral tissues of DKO mice have a functioning circadian clockwork, but rhythmicity is greatly reduced in the absence of robust, rhythmic physiological signals originating from the SCN. Thus, IA-2 and IA-2ß play an important role in the regulation of circadian rhythms, likely through their participation in neurochemical communication among SCN neurons.


Assuntos
Ritmo Circadiano , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/metabolismo , Vesículas Secretórias/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano/efeitos dos fármacos , Cruzamentos Genéticos , Dexametasona/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Coração/efeitos dos fármacos , Coração/inervação , Fígado/efeitos dos fármacos , Fígado/inervação , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Especificidade de Órgãos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/genética , Vesículas Secretórias/efeitos dos fármacos
8.
J Biol Rhythms ; 37(1): 53-77, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35023384

RESUMO

Circadian rhythms are endogenously generated physiological and molecular rhythms with a cycle length of about 24 h. Bioluminescent reporters have been exceptionally useful for studying circadian rhythms in numerous species. Here, we report development of a reporter mouse generated by modification of a widely expressed and highly rhythmic gene encoding D-site albumin promoter binding protein (Dbp). In this line of mice, firefly luciferase is expressed from the Dbp locus in a Cre recombinase-dependent manner, allowing assessment of bioluminescence rhythms in specific cellular populations. A mouse line in which luciferase expression was Cre-independent was also generated. The Dbp reporter alleles do not alter Dbp gene expression rhythms in liver or circadian locomotor activity rhythms. In vivo and ex vivo studies show the utility of the reporter alleles for monitoring rhythmicity. Our studies reveal cell-type-specific characteristics of rhythms among neuronal populations within the suprachiasmatic nuclei ex vivo. In vivo studies show Dbp-driven bioluminescence rhythms in the liver of Albumin-Cre;DbpKI/+ "liver reporter" mice. After a shift of the lighting schedule, locomotor activity achieved the proper phase relationship with the new lighting cycle more rapidly than hepatic bioluminescence did. As previously shown, restricting food access to the daytime altered the phase of hepatic rhythmicity. Our model allowed assessment of the rate of recovery from misalignment once animals were provided with food ad libitum. These studies confirm the previously demonstrated circadian misalignment following environmental perturbations and reveal the utility of this model for minimally invasive, longitudinal monitoring of rhythmicity from specific mouse tissues.


Assuntos
Ritmo Circadiano , Núcleo Supraquiasmático , Albuminas/genética , Albuminas/metabolismo , Animais , Ritmo Circadiano/genética , Genes Reporter , Luciferases/genética , Luciferases/metabolismo , Camundongos , Fotoperíodo , Núcleo Supraquiasmático/metabolismo
9.
J Biol Rhythms ; 37(1): 78-93, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34873943

RESUMO

Circadian rhythms are driven by daily oscillations of gene expression. An important tool for studying cellular and tissue circadian rhythms is the use of a gene reporter, such as bioluminescence from the reporter gene luciferase controlled by a rhythmically expressed gene of interest. Here we describe methods that allow measurement of circadian bioluminescence from a freely moving mouse housed in a standard cage. Using a LumiCycle In Vivo (Actimetrics), we determined conditions that allow detection of circadian rhythms of bioluminescence from the PER2 reporter, PER2::LUC, in freely behaving mice. The LumiCycle In Vivo applies a background subtraction that corrects for effects of room temperature on photomultiplier tube (PMT) output. We tested delivery of d-luciferin via a subcutaneous minipump and in the drinking water. We demonstrate spikes in bioluminescence associated with drinking bouts. Further, we demonstrate that a synthetic luciferase substrate, CycLuc1, can support circadian rhythms of bioluminescence, even when delivered at a lower concentration than d-luciferin, and can support longer-term studies. A small difference in phase of the PER2::LUC bioluminescence rhythms, with females phase leading males, can be detected with this technique. We share our analysis scripts and suggestions for further improvements in this method. This approach will be straightforward to apply to mice with tissue-specific reporters, allowing insights into responses of specific peripheral clocks to perturbations such as environmental or pharmacological manipulations.


Assuntos
Ritmo Circadiano , Proteínas Circadianas Period , Animais , Ritmo Circadiano/fisiologia , Feminino , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/fisiologia
10.
Nat Neurosci ; 10(5): 543-5, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17417633

RESUMO

Heterodimers of CLOCK and BMAL1, bHLH-PAS transcription factors, are believed to be the major transcriptional regulators of the circadian clock mechanism in mammals. However, a recent study shows that CLOCK-deficient mice continue to exhibit robust behavioral and molecular rhythms. Here we report that the transcription factor NPAS2 (MOP4) is able to functionally substitute for CLOCK in the master brain clock in mice to regulate circadian rhythmicity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Ritmo Circadiano/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Núcleo Supraquiasmático/fisiologia , Transativadores/metabolismo , Análise de Variância , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Comportamento Animal , Proteínas CLOCK , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , Atividade Motora/genética , Proteínas do Tecido Nervoso/deficiência , Transativadores/deficiência
11.
Neuron ; 50(3): 465-77, 2006 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-16675400

RESUMO

The circadian clock mechanism in the mouse is composed of interlocking transcriptional feedback loops. Two transcription factors, CLOCK and BMAL1, are believed to be essential components of the circadian clock. We have used the Cre-LoxP system to generate whole-animal knockouts of CLOCK and evaluated the resultant circadian phenotypes. Surprisingly, CLOCK-deficient mice continue to express robust circadian rhythms in locomotor activity, although they do have altered responses to light. At the molecular and biochemical levels, clock gene mRNA and protein levels in both the master clock in the suprachiasmatic nuclei and a peripheral clock in the liver show alterations in the CLOCK-deficient animals, although the molecular feedback loops continue to function. Our data challenge a central feature of the current mammalian circadian clock model regarding the necessity of CLOCK:BMAL1 heterodimers for clock function.


Assuntos
Relógios Biológicos/genética , Ritmo Circadiano/genética , Núcleo Supraquiasmático/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição ARNTL , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Relógios Biológicos/efeitos da radiação , Proteínas CLOCK , Ritmo Circadiano/efeitos da radiação , Dimerização , Retroalimentação Fisiológica/genética , Luz , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Fenótipo , Estimulação Luminosa , RNA Mensageiro/metabolismo , Núcleo Supraquiasmático/citologia
12.
FASEB J ; 23(9): 3226-32, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19433624

RESUMO

Targeted deletion of IA-2 and IA-2beta, major autoantigens in type 1 diabetes and transmembrane secretory vesicle proteins, results in impaired secretion of hormones and neurotransmitters. In the present study, we evaluated the effect of these deletions on daily rhythms in blood pressure, heart rate, core body temperature, and spontaneous physical and neuronal activity. We found that deletion of both IA-2 and IA-2beta profoundly disrupts the usual diurnal variation of each of these parameters, whereas the deletion of either IA-2 or IA-2beta alone did not produce a major change. In situ hybridization revealed that IA-2 and IA-2beta transcripts are highly but nonrhythmically expressed in the suprachiasmatic nuclei, the site of the brain's master circadian oscillator. Electrophysiological studies on tissue slices from the suprachiasmatic nuclei showed that disruption of both IA-2 and IA-2beta results in significant alterations in neuronal firing. From these studies, we concluded that deletion of IA-2 and IA-2beta, structural proteins of secretory vesicles and modulators of neuroendocrine secretion, has a profound effect on the circadian system.


Assuntos
Ritmo Circadiano , Eletrofisiologia , Hemodinâmica/fisiologia , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/fisiologia , Vesículas Secretórias/química , Animais , Camundongos , RNA Mensageiro/análise , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/deficiência , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/genética , Núcleo Supraquiasmático/fisiologia
13.
Cell Host Microbe ; 27(2): 176-187, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32053788

RESUMO

Biological rhythms appear to be an elegant solution to the challenge of coordinating activities with the consequences of the Earth's daily and seasonal rotation. The genes and molecular mechanisms underpinning circadian clocks in multicellular organisms are well understood. In contrast, the regulatory mechanisms and fitness consequences of biological rhythms exhibited by parasites remain mysterious. Here, we explore how periodicity in parasite traits is generated and why daily rhythms matter for parasite fitness. We focus on malaria (Plasmodium) parasites which exhibit developmental rhythms during replication in the mammalian host's blood and in transmission to vectors. Rhythmic in-host parasite replication is responsible for eliciting inflammatory responses, the severity of disease symptoms, and fueling transmission, as well as conferring tolerance to anti-parasite drugs. Thus, understanding both how and why the timing and synchrony of parasites are connected to the daily rhythms of hosts and vectors may make treatment more effective and less toxic to hosts.


Assuntos
Ritmo Circadiano/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Plasmodium/fisiologia , Animais , Evolução Biológica , Relógios Circadianos/fisiologia , Eritrócitos/parasitologia , Humanos , Imunidade/fisiologia , Inflamação/parasitologia , Malária , Camundongos , Mosquitos Vetores/parasitologia , Mosquitos Vetores/fisiologia , Parasitos/fisiologia
14.
Eur J Neurosci ; 28(12): 2443-50, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19087172

RESUMO

Reproductive physiology is regulated by the photoperiod in many mammals. Decoding of the photoperiod involves circadian clock mechanisms, although the molecular basis remains unclear. Recent studies have shown that the ependymal cell layer lining the infundibular recess of the third ventricle (EC) is a key structure for the photoperiodic gonadal response. The EC exhibits daylength-dependent changes in the expression of photoperiodic output genes, including the type 2 deiodinase gene (Dio2 ). Here we investigated whether clock genes (Per1 and Bmal1) and the albumin D-binding protein gene (Dbp) are expressed in the EC of Syrian hamsters, and whether their expression differs under long-day and short-day conditions. Expression of all three genes followed a diurnal rhythm; expression of Per1 and Dbp in the EC peaked around lights-off, and expression of Bmal1 peaked in the early light phase. The amplitude of Per1 and Dbp expression was higher in hamsters kept under long-day conditions than in those kept under short-day conditions. Notably, the expression of these genes was not modified by exogenous melatonin within 25 h after injection, whereas Dio2 expression was inhibited 19 h after injection. Targeted melatonin receptor (MT1, MT2, and both MT1 and MT2) disruption in melatonin-proficient C3H mice did not affect the rhythmic expression of Per1 in the EC. These data show the existence of a molecular clock in the rodent EC. In the hamster, this clock responds to long-term changes in the photoperiod, but is independent of acute melatonin signals. In mice, the EC clock is not affected by deletion of melatonin receptors.


Assuntos
Relógios Biológicos/genética , Ritmo Circadiano/fisiologia , Epêndima , Regulação da Expressão Gênica , Melatonina/metabolismo , Transdução de Sinais/fisiologia , Terceiro Ventrículo/citologia , Fatores de Transcrição ARNTL , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epêndima/citologia , Epêndima/fisiologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C3H , Proteínas Circadianas Period , Fotoperíodo , Receptores de Melatonina/genética , Receptores de Melatonina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Nat Neurosci ; 5(3): 234-8, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11836530

RESUMO

In mammals, many daily cycles are driven by a central circadian clock, which is based on the cell-autonomous rhythmic expression of clock genes. It is not clear, however, how peripheral cells are able to interpret the rhythmic signals disseminated from this central oscillator. Here we show that cycling expression of the clock gene Period1 in rodent pituitary cells depends on the heterologous sensitization of the adenosine A2b receptor, which occurs through the nocturnal activation of melatonin mt1 receptors. Eliminating the impact of the neurohormone melatonin simultaneously suppresses the expression of Period1 and evokes an increase in the release of pituitary prolactin. Our findings expose a mechanism by which two convergent signals interact within a temporal dimension to establish high-amplitude, precise and robust cycles of gene expression.


Assuntos
Relógios Biológicos/fisiologia , Regulação da Expressão Gênica/fisiologia , Melatonina/metabolismo , Proteínas Nucleares/metabolismo , Neuro-Hipófise/fisiologia , Receptores Purinérgicos P1/metabolismo , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Animais , Proteínas de Ciclo Celular , Ritmo Circadiano/fisiologia , Cricetinae , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hibridização In Situ , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C3H , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Circadianas Period , Phodopus , Glândula Pineal/cirurgia , Neuro-Hipófise/citologia , Receptor A2B de Adenosina , Receptores de Superfície Celular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Melatonina , Transdução de Sinais/fisiologia
16.
J Biol Rhythms ; 33(2): 179-191, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29671710

RESUMO

Mice with targeted gene disruption have provided important information about the molecular mechanisms of circadian clock function. A full understanding of the roles of circadian-relevant genes requires manipulation of their expression in a tissue-specific manner, ideally including manipulation with high efficiency within the suprachiasmatic nuclei (SCN). To date, conditional manipulation of genes within the SCN has been difficult. In a previously developed mouse line, Cre recombinase was inserted into the vesicular GABA transporter (Vgat) locus. Since virtually all SCN neurons are GABAergic, this Vgat-Cre line seemed likely to have high efficiency at disrupting conditional alleles in SCN. To test this premise, the efficacy of Vgat-Cre in excising conditional (fl, for flanked by LoxP) alleles in the SCN was examined. Vgat-Cre-mediated excision of conditional alleles of Clock or Bmal1 led to loss of immunostaining for products of the targeted genes in the SCN. Vgat-Cre+; Clockfl/fl; Npas2m/m mice and Vgat-Cre+; Bmal1fl/fl mice became arrhythmic immediately upon exposure to constant darkness, as expected based on the phenotype of mice in which these genes are disrupted throughout the body. The phenotype of mice with other combinations of Vgat-Cre+, conditional Clock, and mutant Npas2 alleles also resembled the corresponding whole-body knockout mice. These data indicate that the Vgat-Cre line is useful for Cre-mediated recombination within the SCN, making it useful for Cre-enabled technologies including gene disruption, gene replacement, and opto- and chemogenetic manipulation of the SCN circadian clock.


Assuntos
Alelos , Proteínas CLOCK/genética , Núcleo Supraquiasmático , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Animais , Relógios Circadianos/genética , Ritmo Circadiano/genética , Feminino , Integrases , Masculino , Camundongos , Camundongos Knockout
17.
Mol Cell Biol ; 24(2): 584-94, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14701732

RESUMO

The mPER1 and mPER2 proteins have important roles in the circadian clock mechanism, whereas mPER3 is expendable. Here we examine the posttranslational regulation of mPER3 in vivo in mouse liver and compare it to the other mPER proteins to define the salient features required for clock function. Like mPER1 and mPER2, mPER3 is phosphorylated, changes cellular location, and interacts with other clock proteins in a time-dependent manner. Consistent with behavioral data from mPer2/3 and mPer1/3 double-mutant mice, either mPER1 or mPER2 alone can sustain rhythmic posttranslational events. However, mPER3 is unable to sustain molecular rhythmicity in mPer1/2 double-mutant mice. Indeed, mPER3 is always cytoplasmic and is not phosphorylated in the livers of mPer1-deficient mice, suggesting that mPER3 is regulated by mPER1 at a posttranslational level. In vitro studies with chimeric proteins suggest that the inability of mPER3 to support circadian clock function results in part from lack of direct and stable interaction with casein kinase Iepsilon (CKIepsilon). We thus propose that the CKIepsilon-binding domain is critical not only for mPER phosphorylation but also for a functioning circadian clock.


Assuntos
Ritmo Circadiano/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Quinases/fisiologia , Animais , Sítios de Ligação , Caseína Quinases , Proteínas de Ciclo Celular , Ritmo Circadiano/genética , Técnicas In Vitro , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Nucleares/química , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Circadianas Period , Fosforilação , Ligação Proteica , Proteínas Quinases/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Fatores de Transcrição
18.
Mol Cell Biol ; 23(3): 1054-60, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12529409

RESUMO

Two high-affinity, G protein-coupled melatonin receptor subtypes have been identified in mammals. Targeted disruption of the Mel(1a) melatonin receptor prevents some, but not all, responses to the hormone, suggesting functional redundancy among receptor subtypes (Liu et al., Neuron 19:91-102, 1997). In the present work, the mouse Mel(1b) melatonin receptor cDNA was isolated and characterized, and the gene has been disrupted. The cDNA encodes a receptor with high affinity for melatonin and a pharmacological profile consistent with its assignment as encoding a melatonin receptor. Mice with targeted disruption of the Mel(1b) receptor have no obvious circadian phenotype. Melatonin suppressed multiunit electrical activity in the suprachiasmatic nucleus (SCN) in Mel(1b) receptor-deficient mice as effectively as in wild-type controls. The neuropeptide, pituitary adenylyl cyclase activating peptide, increases the level of phosphorylated cyclic AMP response element binding protein (CREB) in SCN slices, and melatonin reduces this effect. The Mel(1a) receptor subtype mediates this inhibitory response at moderate ligand concentrations (1 nM). A residual response apparent in Mel(1a) receptor-deficient C3H mice at higher melatonin concentrations (100 nM) is absent in Mel(1a)-Mel(1b) double-mutant mice, indicating that the Mel(1b) receptor mediates this effect of melatonin. These data indicate that there is a limited functional redundancy between the receptor subtypes in the SCN. Mice with targeted disruption of melatonin receptor subtypes will allow molecular dissection of other melatonin receptor-mediated responses.


Assuntos
Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Marcação de Genes , Melatonina/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Fenótipo , Fosforilação , Receptores de Melatonina , Homologia de Sequência de Aminoácidos , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/metabolismo
19.
Cell Mol Gastroenterol Hepatol ; 4(1): 95-114, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28593182

RESUMO

BACKGROUND & AIMS: The gastrointestinal syndrome is an illness of the intestine caused by high levels of radiation. It is characterized by extensive loss of epithelial tissue integrity, which initiates a regenerative response by intestinal stem and precursor cells. The intestine has 24-hour rhythms in many physiological functions that are believed to be outputs of the circadian clock: a molecular system that produces 24-hour rhythms in transcription/translation. Certain gastrointestinal illnesses are worsened when the circadian rhythms are disrupted, but the role of the circadian clock in gastrointestinal regeneration has not been studied. METHODS: We tested the timing of regeneration in the mouse intestine during the gastrointestinal syndrome. The role of the circadian clock was tested genetically using the BMAL1 loss of function mouse mutant in vivo, and in vitro using intestinal organoid culture. RESULTS: The proliferation of the intestinal epithelium follows a 24-hour rhythm during the gastrointestinal syndrome. The circadian clock runs in the intestinal epithelium during this pathologic state, and the loss of the core clock gene, BMAL1, disrupts both the circadian clock and rhythmic proliferation. Circadian activity in the intestine involves a rhythmic production of inflammatory cytokines and subsequent rhythmic activation of the JNK stress response pathway. CONCLUSIONS: Our results show that a circadian rhythm in inflammation and regeneration occurs during the gastrointestinal syndrome. The study and treatment of radiation-induced illnesses, and other gastrointestinal illnesses, should consider 24-hour timing in physiology and pathology.

20.
J Biol Rhythms ; 20(3): 206-18, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15851527

RESUMO

Prokineticin 2 (PK2) is a putative output molecule from the SCN. PK2 RNA levels are rhythmic in the mouse SCN, with high levels during the day, and PK2 administration suppresses nocturnal locomotor activity in rats. The authors examined the PK2 system in a diurnal rodent, Arvicanthis niloticus, to determine whether PK2 or PK2 receptors differ between diurnal and nocturnal species. The major transcript variant of A. niloticus PK2 (AnPK2) encodes a 26-residue signal peptide followed by the presumed mature peptide of 81 residues. Within the grass rat signal sequence, polymorphic sequences and amino acid substitutions were observed relative to mouse and laboratory rats, but the hydrophobic core and cleavage site of the signal sequence were preserved. The mature PK2 peptide is identical among A. niloticus, rat, and mouse. AnPK2 mRNA is rhythmically expressed in the SCN, with peak RNA levels occurring in the morning, preceding peaks of Per1 and Per2 as in mouse SCN. Analysis of prokineticin receptor 2 (PKR2) sequences revealed polymorphisms among the grass rats studied. PKR2 mRNA was expressed in the SCN and paraventricular nuclei of the thalamus and hypothalamus. While further analysis is necessary, there is no clear evidence indicating that a difference in the PK2 ligand/receptor system accounts for diurnality in this rodent species. These data contribute to a growing body of evidence suggesting that the key to diurnality lies downstream of the SCN in A. niloticus.


Assuntos
Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Regulação da Expressão Gênica/fisiologia , Sequência de Aminoácidos , Animais , Química Encefálica/genética , DNA Complementar/biossíntese , DNA Complementar/genética , Hibridização In Situ , Luz , Masculino , Dados de Sequência Molecular , Polimorfismo Genético/genética , Ratos , Roedores , Transcrição Gênica , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA