Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
J Biol Rhythms ; 39(2): 135-165, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38366616

ABSTRACT

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.


Subject(s)
Circadian Rhythm , Neuropeptides , Circadian Rhythm/physiology , Neuropeptides/metabolism , Suprachiasmatic Nucleus/physiology , Vasoactive Intestinal Peptide/metabolism , Gastrin-Releasing Peptide/metabolism
2.
J Biol Rhythms ; 37(1): 53-77, 2022 02.
Article in English | MEDLINE | ID: mdl-35023384

ABSTRACT

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.


Subject(s)
Circadian Rhythm , Suprachiasmatic Nucleus , Albumins/genetics , Albumins/metabolism , Animals , Circadian Rhythm/genetics , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Mice , Photoperiod , Suprachiasmatic Nucleus/metabolism
3.
J Biol Rhythms ; 37(1): 78-93, 2022 02.
Article in English | MEDLINE | ID: mdl-34873943

ABSTRACT

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.


Subject(s)
Circadian Rhythm , Period Circadian Proteins , Animals , Circadian Rhythm/physiology , Female , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology
4.
J Pineal Res ; 69(1): e12654, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32243642

ABSTRACT

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.


Subject(s)
Behavior, Animal , Biological Clocks , Circadian Rhythm , Glucose Intolerance , Obesity , Animals , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Mice , Mice, Transgenic , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology
5.
Cell Host Microbe ; 27(2): 176-187, 2020 02 12.
Article in English | MEDLINE | ID: mdl-32053788

ABSTRACT

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.


Subject(s)
Circadian Rhythm/physiology , Host-Parasite Interactions/physiology , Plasmodium/physiology , Animals , Biological Evolution , Circadian Clocks/physiology , Erythrocytes/parasitology , Humans , Immunity/physiology , Inflammation/parasitology , Malaria , Mice , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Parasites/physiology
6.
J Exp Biol ; 221(Pt 15)2018 08 06.
Article in English | MEDLINE | ID: mdl-29903839

ABSTRACT

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.


Subject(s)
Circadian Clocks/genetics , Eating/physiology , Torpor/physiology , Animals , Body Temperature/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Female , Locomotion , Male , Mice , Mutation , Time Factors
7.
J Biol Rhythms ; 33(2): 179-191, 2018 04.
Article in English | MEDLINE | ID: mdl-29671710

ABSTRACT

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.


Subject(s)
Alleles , CLOCK Proteins/genetics , Suprachiasmatic Nucleus , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Female , Integrases , Male , Mice , Mice, Knockout
8.
Proc Natl Acad Sci U S A ; 115(10): E2437-E2446, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463694

ABSTRACT

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.


Subject(s)
Casein Kinase 1 epsilon/genetics , Casein Kinase Idelta/genetics , Circadian Clocks , GABAergic Neurons/enzymology , Suprachiasmatic Nucleus/physiology , Animals , Casein Kinase 1 epsilon/deficiency , Casein Kinase Idelta/deficiency , Circadian Rhythm , Female , Gene Knockout Techniques , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Suprachiasmatic Nucleus/enzymology
9.
Cell Mol Gastroenterol Hepatol ; 4(1): 95-114, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28593182

ABSTRACT

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.

10.
Front Neurosci ; 10: 481, 2016.
Article in English | MEDLINE | ID: mdl-27826225

ABSTRACT

Single-cell heterogeneity confounds efforts to understand how a population of cells organizes into cellular networks that underlie tissue-level function. This complexity is prominent in the mammalian suprachiasmatic nucleus (SCN). Here, individual neurons exhibit a remarkable amount of asynchronous behavior and transcriptional heterogeneity. However, SCN neurons are able to generate precisely coordinated synaptic and molecular outputs that synchronize the body to a common circadian cycle by organizing into cellular networks. To understand this emergent cellular network property, it is important to reconcile single-neuron heterogeneity with network organization. In light of recent studies suggesting that transcriptionally heterogeneous cells organize into distinct cellular phenotypes, we characterized the transcriptional, spatial, and functional organization of 352 SCN neurons from mice experiencing phase-shifts in their circadian cycle. Using the community structure detection method and multivariate analytical techniques, we identified previously undescribed neuronal phenotypes that are likely to participate in regulatory networks with known SCN cell types. Based on the newly discovered neuronal phenotypes, we developed a data-driven neuronal network structure in which multiple cell types interact through known synaptic and paracrine signaling mechanisms. These results provide a basis from which to interpret the functional variability of SCN neurons and describe methodologies toward understanding how a population of heterogeneous single cells organizes into cellular networks that underlie tissue-level function.

11.
J Biol Rhythms ; 29(4): 277-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25238856

ABSTRACT

The circadian clock generates daily cycles of gene expression that regulate physiological processes. The liver plays an important role in xenobiotic metabolism and also has been shown to possess its own cell-based clock. The liver clock is synchronized by the master clock in the brain, and a portion of rhythmic gene expression can be driven by behavior of the organism as a whole even when the hepatic clock is suppressed. So far, however, there is relatively little evidence indicating whether the liver clock is functionally important in modulating xenobiotic metabolism. Thus, mice lacking circadian clock function in the whole body or specifically in liver were challenged with pentobarbital and acetaminophen, and pentobarbital sleep time (PBST) and acetaminophen toxicity, respectively, was assessed at different times of day in mutant and control mice. The results suggest that the liver clock is essential for rhythmic changes in xenobiotic detoxification. Surprisingly, it seems that the way in which the clock is disrupted determines the rate of xenobiotic metabolism in the liver. CLOCK-deficient mice are remarkably resistant to acetaminophen and exhibit a longer PBST, while PERIOD-deficient mice have a short PBST. These results indicate an essential role of the tissue-intrinsic peripheral circadian oscillator in the liver in regulating xenobiotic metabolism.


Subject(s)
Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Liver/metabolism , Liver/physiology , Xenobiotics/metabolism , Acetaminophen/pharmacology , Animals , Gene Expression/genetics , Male , Mice , Mice, Inbred C57BL , Pentobarbital/pharmacology , Periodicity
12.
J Clin Invest ; 123(12): 5389-400, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24270424

ABSTRACT

Brain aging is associated with diminished circadian clock output and decreased expression of the core clock proteins, which regulate many aspects of cellular biochemistry and metabolism. The genes encoding clock proteins are expressed throughout the brain, though it is unknown whether these proteins modulate brain homeostasis. We observed that deletion of circadian clock transcriptional activators aryl hydrocarbon receptor nuclear translocator-like (Bmal1) alone, or circadian locomotor output cycles kaput (Clock) in combination with neuronal PAS domain protein 2 (Npas2), induced severe age-dependent astrogliosis in the cortex and hippocampus. Mice lacking the clock gene repressors period circadian clock 1 (Per1) and period circadian clock 2 (Per2) had no observed astrogliosis. Bmal1 deletion caused the degeneration of synaptic terminals and impaired cortical functional connectivity, as well as neuronal oxidative damage and impaired expression of several redox defense genes. Targeted deletion of Bmal1 in neurons and glia caused similar neuropathology, despite the retention of intact circadian behavioral and sleep-wake rhythms. Reduction of Bmal1 expression promoted neuronal death in primary cultures and in mice treated with a chemical inducer of oxidative injury and striatal neurodegeneration. Our findings indicate that BMAL1 in a complex with CLOCK or NPAS2 regulates cerebral redox homeostasis and connects impaired clock gene function to neurodegeneration.


Subject(s)
ARNTL Transcription Factors/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Brain/pathology , CLOCK Proteins/physiology , Circadian Rhythm/physiology , Gliosis/genetics , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/physiology , Neurons/metabolism , ARNTL Transcription Factors/deficiency , Aging/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Brain/physiopathology , CLOCK Proteins/deficiency , Cerebral Cortex/pathology , Circadian Rhythm/genetics , Corpus Striatum/pathology , Gene Expression Regulation/physiology , Gliosis/pathology , Hippocampus/pathology , Homeostasis/genetics , Homeostasis/physiology , Locomotion/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Nerve Degeneration/genetics , Nerve Tissue Proteins/deficiency , Neuroglia/metabolism , Neuroglia/pathology , Neurons/pathology , Oxidation-Reduction , Oxidative Stress , Period Circadian Proteins/deficiency , Period Circadian Proteins/physiology , RNA Interference , Sleep Disorders, Circadian Rhythm/physiopathology
13.
Am J Physiol Endocrinol Metab ; 303(6): E762-76, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22785238

ABSTRACT

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.


Subject(s)
Circadian Rhythm , Gene Expression Regulation , Membrane Proteins/metabolism , Neurons/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Secretory Vesicles/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Circadian Rhythm/drug effects , Crosses, Genetic , Dexamethasone/pharmacology , Female , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Heart/drug effects , Heart/innervation , Liver/drug effects , Liver/innervation , Liver/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Secretory Vesicles/drug effects
14.
PLoS One ; 7(5): e37833, 2012.
Article in English | MEDLINE | ID: mdl-22662235

ABSTRACT

We use the multigenic pattern of gene expression across suprachiasmatic nuclei (SCN) regions and time to understand the dynamics within the SCN in response to a circadian phase-resetting light pulse. Global gene expression studies of the SCN indicate that circadian functions like phase resetting are complex multigenic processes. While the molecular dynamics of phase resetting are not well understood, it is clear they involve a "functional gene expression program", e.g., the coordinated behavior of functionally related genes in space and time. In the present study we selected a set of 89 of these functionally related genes in order to further understand this multigenic program. By use of high-throughput qPCR we studied 52 small samples taken by anatomically precise laser capture from within the core and shell SCN regions, and taken at time points with and without phase resetting light exposure. The results show striking regional differences in light response to be present in the mouse SCN. By using network-based analyses, we are able to establish a highly specific multigenic correlation between genes expressed in response to light at night and genes normally activated during the day. The light pulse triggers a complex and highly coordinated network of gene regulation. The largest differences marking neuroanatomical location are in transmitter receptors, and the largest time-dependent differences occur in clock-related genes. Nighttime phase resetting appears to recruit transcriptional regulatory processes normally active in the day. This program, or mechanism, causes the pattern of core region gene expression to transiently shift to become more like that of the shell region.


Subject(s)
Gene Expression Regulation/radiation effects , Gene Regulatory Networks , Light , Suprachiasmatic Nucleus/metabolism , Animals , Biological Clocks/genetics , Circadian Rhythm/genetics , Cluster Analysis , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL
15.
PLoS One ; 7(4): e35938, 2012.
Article in English | MEDLINE | ID: mdl-22558277

ABSTRACT

Research on the mechanisms underlying circadian rhythmicity and the response of brain and body clocks to environmental and physiological challenges requires assessing levels of circadian clock proteins. Too often, however, it is difficult to acquire antibodies that specifically and reliably label these proteins. Many of these antibodies also lack appropriate validation. The goal of this project was to generate and characterize antibodies against several circadian clock proteins. We examined mice and hamsters at peak and trough times of clock protein expression in the suprachiasmatic nucleus (SCN). In addition, we confirmed specificity by testing the antibodies on mice with targeted disruption of the relevant genes. Our results identify antibodies against PER1, PER2, BMAL1 and CLOCK that are useful for assessing circadian clock proteins in the SCN by immunocytochemistry.


Subject(s)
Antibodies/immunology , CLOCK Proteins/immunology , Circadian Clocks/immunology , Suprachiasmatic Nucleus/metabolism , Animals , Cricetinae , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Staining and Labeling , Suprachiasmatic Nucleus/cytology
16.
Proc Natl Acad Sci U S A ; 108(41): 17219-24, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969555

ABSTRACT

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.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Period Circadian Proteins/genetics , Suprachiasmatic Nucleus/physiology , Animals , Cricetinae , Gene Expression , Male , Mesocricetus , Photic Stimulation , Photoperiod , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Biol Rhythms ; 26(5): 390-401, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21921293

ABSTRACT

Mice lacking the CLOCK protein have a relatively subtle circadian phenotype, including a slightly shorter period in constant darkness, differences in phase resetting after 4-hour light pulses in the early and late night, and a variably advanced phase angle of entrainment in a light-dark (LD) cycle. The present series of experiments was conducted to more fully characterize the circadian phenotype of Clock(-/-) mice under various lighting conditions. A phase-response curve (PRC) to 4-hour light pulses in free-running mice was conducted; the results confirm that Clock(-/-) mice exhibit very large phase advances after 4-hour light pulses in the late subjective night but have relatively normal responses to light at other phases. The abnormal shape of the PRC to light may explain the tendency of CLOCK-deficient mice to begin activity before lights-out when housed in a 12-hour light:12-hour dark lighting schedule. To assess this relationship further, Clock(-/-) and wild-type control mice were entrained to skeleton lighting cycles (1L:23D and 1L:10D:1L:12D). Comparing entrainment under the 2 types of skeleton photoperiods revealed that exposure to 1-hour light in the morning leads to a phase advance of activity onset (expressed the following afternoon) in Clock(-/-) mice but not in the controls. Constant light typically causes an intensity-dependent increase in circadian period in mice, but this did not occur in CLOCK-deficient mice. The failure of Clock(-/-) mice to respond to the period-lengthening effect of constant light likely results from the increased functional impact of light falling in the phase advance zone of the PRC. Collectively, these experiments reveal that alterations in the response of CLOCK-deficient mice to light in several paradigms are likely due to an imbalance in the shape of the PRC to light.


Subject(s)
CLOCK Proteins/deficiency , Circadian Rhythm/physiology , Motor Activity/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , CLOCK Proteins/genetics , Female , Male , Mice , Motor Activity/radiation effects , Nerve Tissue Proteins/physiology , Photic Stimulation , Photoperiod
18.
Proc Natl Acad Sci U S A ; 108(39): 16451-6, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21930935

ABSTRACT

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.


Subject(s)
Casein Kinase I/metabolism , Circadian Rhythm , Period Circadian Proteins/physiology , Protein Phosphatase 1/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Mice , Period Circadian Proteins/metabolism , Phosphorylation
19.
Aging (Albany NY) ; 3(5): 479-93, 2011 May.
Article in English | MEDLINE | ID: mdl-21566258

ABSTRACT

The circadian clock imparts 24-hour rhythmicity on gene expression and cellular physiology in virtually all cells. Disruption of the genes necessary for the circadian clock to function has diverse effects, including aging-related phenotypes. Some circadian clock genes have been described as tumor suppressors, while other genes have less clear functions in aging and cancer. In this Review, we highlight a recent study [Dubrovsky et al., Aging 2: 936-944, 2010] and discuss the much larger field examining the relationship between circadian clock genes, circadian rhythmicity, aging-related phenotypes, and cancer.


Subject(s)
Aging/genetics , Circadian Clocks/genetics , Circadian Rhythm/physiology , Neoplasms/genetics , Aging/physiology , Gene Expression Regulation , Humans , Light , Periodicity , Phenotype , Trans-Activators/physiology
20.
Chronobiol Int ; 27(6): 1317-28, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20653457

ABSTRACT

The circadian clock orchestrates most physiological processes in mammals. Disruption of circadian rhythms appears to contribute to the development of obesity and metabolic syndrome. The Period genes mPer1 and mPer2, but not mPer3, are essential for core clock function in mice. To assess the impact of mPer genes on body mass regulation, mPer mutant and control mice were fed a high-fat diet. Here the authors report that male mPer1/2/3 triple-deficient mice gain significantly more body mass than wild-type controls on high-fat diet. Surprisingly, mPer3 single-deficient animals mimicked this phenotype, suggesting a previously unrecognized role for mPer3 in body mass regulation.


Subject(s)
Body Weight/genetics , Body Weight/physiology , Period Circadian Proteins/genetics , Period Circadian Proteins/physiology , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Dietary Fats/administration & dosage , Eating/genetics , Eating/physiology , Female , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Mutation , Period Circadian Proteins/deficiency , Sex Characteristics , Weight Gain/genetics , Weight Gain/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...