Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Metab ; 36(1): 90-102.e7, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38171340

ABSTRACT

Interactions between lineage-determining and activity-dependent transcription factors determine single-cell identity and function within multicellular tissues through incompletely known mechanisms. By assembling a single-cell atlas of chromatin state within human islets, we identified ß cell subtypes governed by either high or low activity of the lineage-determining factor pancreatic duodenal homeobox-1 (PDX1). ß cells with reduced PDX1 activity displayed increased chromatin accessibility at latent nuclear factor κB (NF-κB) enhancers. Pdx1 hypomorphic mice exhibited de-repression of NF-κB and impaired glucose tolerance at night. Three-dimensional analyses in tandem with chromatin immunoprecipitation (ChIP) sequencing revealed that PDX1 silences NF-κB at circadian and inflammatory enhancers through long-range chromatin contacts involving SIN3A. Conversely, Bmal1 ablation in ß cells disrupted genome-wide PDX1 and NF-κB DNA binding. Finally, antagonizing the interleukin (IL)-1ß receptor, an NF-κB target, improved insulin secretion in Pdx1 hypomorphic islets. Our studies reveal functional subtypes of single ß cells defined by a gradient in PDX1 activity and identify NF-κB as a target for insulinotropic therapy.


Subject(s)
Insulin-Secreting Cells , NF-kappa B , Animals , Humans , Mice , Chromatin/metabolism , Genes, Homeobox , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , NF-kappa B/metabolism
2.
Elife ; 112022 02 21.
Article in English | MEDLINE | ID: mdl-35188462

ABSTRACT

The mammalian circadian clock drives daily oscillations in physiology and behavior through an autoregulatory transcription feedback loop present in central and peripheral cells. Ablation of the core clock within the endocrine pancreas of adult animals impairs the transcription and splicing of genes involved in hormone exocytosis and causes hypoinsulinemic diabetes. Here, we developed a genetically sensitized small-molecule screen to identify druggable proteins and mechanistic pathways involved in circadian ß-cell failure. Our approach was to generate ß-cells expressing a nanoluciferase reporter within the proinsulin polypeptide to screen 2640 pharmacologically active compounds and identify insulinotropic molecules that bypass the secretory defect in CRISPR-Cas9-targeted clock mutant ß-cells. We validated hit compounds in primary mouse islets and identified known modulators of ligand-gated ion channels and G-protein-coupled receptors, including the antihelmintic ivermectin. Single-cell electrophysiology in circadian mutant mouse and human cadaveric islets revealed ivermectin as a glucose-dependent secretagogue. Genetic, genomic, and pharmacological analyses established the P2Y1 receptor as a clock-controlled mediator of the insulinotropic activity of ivermectin. These findings identify the P2Y1 purinergic receptor as a diabetes target based upon a genetically sensitized phenotypic screen.


Circadian rhythms ­ 'inbuilt' 24-hour cycles ­ control many aspects of behaviour and physiology. In mammals, they operate in nearly all tissues, including those involved in glucose metabolism. Recent studies have shown that mice with faulty genes involved in circadian rhythms, the core clock genes, can develop diabetes. Diabetes arises when the body struggles to regulate blood sugar levels. In healthy individuals, the hormone insulin produced by beta cells in the pancreas regulates the amount of sugar in the blood. But when beta cells are faulty and do not generate sufficient insulin levels, or when insulin lacks the ability to stimulate cells to take up glucose, diabetes can develop. Marcheva, Weidemann, Taguchi et al. wanted to find out if diabetes caused by impaired clock genes could be treated by targeting pathways regulating the secretion of insulin. To do so, they tested over 2,500 potential drugs on genetically modified beta cells with faulty core clock genes. They further screened the drugs on mice with the same defect in their beta cells. Marcheva et al. identified one potential compound, the anti-parasite drug ivermectin, which was able to restore the secretion of insulin. When ivermectin was applied to both healthy mice and mice with faulty beta cells, the drug improved the control over glucose levels by activating a specific protein receptor that senses molecules important for storing and utilizing energy. The findings reveal new drug targets for treating forms of diabetes associated with deregulation of the pancreatic circadian clock. The drug screening strategy used in the study may also be applied to reveal mechanisms underlying other conditions associated with disrupted circadian clocks, including sleep loss and jetlag.


Subject(s)
Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Islets of Langerhans/metabolism , Receptors, Purinergic P2Y1/metabolism , ARNTL Transcription Factors , Animals , Cell Line , Circadian Clocks , Circadian Rhythm , Cryptochromes/genetics , Cryptochromes/metabolism , Diabetes Mellitus/metabolism , Gene Expression Regulation/drug effects , Glucose/metabolism , High-Throughput Screening Assays , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells , Islets of Langerhans/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Genes Dev ; 34(15-16): 1089-1105, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32616519

ABSTRACT

The circadian clock is encoded by a negative transcriptional feedback loop that coordinates physiology and behavior through molecular programs that remain incompletely understood. Here, we reveal rhythmic genome-wide alternative splicing (AS) of pre-mRNAs encoding regulators of peptidergic secretion within pancreatic ß cells that are perturbed in Clock-/- and Bmal1-/- ß-cell lines. We show that the RNA-binding protein THRAP3 (thyroid hormone receptor-associated protein 3) regulates circadian clock-dependent AS by binding to exons at coding sequences flanking exons that are more frequently skipped in clock mutant ß cells, including transcripts encoding Cask (calcium/calmodulin-dependent serine protein kinase) and Madd (MAP kinase-activating death domain). Depletion of THRAP3 restores expression of the long isoforms of Cask and Madd, and mimicking exon skipping in these transcripts through antisense oligonucleotide delivery in wild-type islets reduces glucose-stimulated insulin secretion. Finally, we identify shared networks of alternatively spliced exocytic genes from islets of rodent models of diet-induced obesity that significantly overlap with clock mutants. Our results establish a role for pre-mRNA alternative splicing in ß-cell function across the sleep/wake cycle.


Subject(s)
Alternative Splicing , Circadian Clocks/genetics , Exocytosis , Glucose/metabolism , Insulin Secretion/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/physiology , Animals , CLOCK Proteins/genetics , CLOCK Proteins/physiology , Cells, Cultured , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Homeostasis , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice, Inbred C57BL , Nuclear Proteins/physiology , Obesity/genetics , Obesity/metabolism , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Transcription Factors/physiology
5.
Cell Metab ; 29(5): 1078-1091.e5, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30827863

ABSTRACT

The alignment of fasting and feeding with the sleep/wake cycle is coordinated by hypothalamic neurons, though the underlying molecular programs remain incompletely understood. Here, we demonstrate that the clock transcription pathway maximizes eating during wakefulness and glucose production during sleep through autonomous circadian regulation of NPY/AgRP neurons. Tandem profiling of whole-cell and ribosome-bound mRNAs in morning and evening under dynamic fasting and fed conditions identified temporal control of activity-dependent gene repertoires in AgRP neurons central to synaptogenesis, bioenergetics, and neurotransmitter and peptidergic signaling. Synaptic and circadian pathways were specific to whole-cell RNA analyses, while bioenergetic pathways were selectively enriched in the ribosome-bound transcriptome. Finally, we demonstrate that the AgRP clock mediates the transcriptional response to leptin. Our results reveal that time-of-day restriction in transcriptional control of energy-sensing neurons underlies the alignment of hunger and food acquisition with the sleep/wake state.


Subject(s)
Agouti-Related Protein/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Hunger/physiology , Neurons/metabolism , Transcription, Genetic/genetics , Agouti-Related Protein/genetics , Animals , Eating/physiology , Fasting/physiology , Gene Regulatory Networks , Glucose/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/genetics , Sleep/physiology , Transcriptome , Wakefulness/physiology
6.
Genes Dev ; 32(21-22): 1367-1379, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30366905

ABSTRACT

The mammalian circadian clock is encoded by an autoregulatory transcription feedback loop that drives rhythmic behavior and gene expression in the brain and peripheral tissues. Transcriptomic analyses indicate cell type-specific effects of circadian cycles on rhythmic physiology, although how clock cycles respond to environmental stimuli remains incompletely understood. Here, we show that activation of the inducible transcription factor NF-κB in response to inflammatory stimuli leads to marked inhibition of clock repressors, including the Period, Cryptochrome, and Rev-erb genes, within the negative limb. Furthermore, activation of NF-κB relocalizes the clock components CLOCK/BMAL1 genome-wide to sites convergent with those bound by NF-κB, marked by acetylated H3K27, and enriched in RNA polymerase II. Abrogation of NF-κB during adulthood alters the expression of clock repressors, disrupts clock-controlled gene cycles, and impairs rhythmic activity behavior, revealing a role for NF-κB in both unstimulated and activated conditions. Together, these data highlight NF-κB-mediated transcriptional repression of the clock feedback limb as a cause of circadian disruption in response to inflammation.


Subject(s)
Circadian Rhythm/genetics , NF-kappa B/physiology , ARNTL Transcription Factors/metabolism , Animals , Behavior, Animal , CLOCK Proteins/metabolism , Cell Line , Chromatin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Repressor Proteins/metabolism , Transcription, Genetic
7.
Genes Dev ; 32(5-6): 321-323, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29593064

ABSTRACT

Each spring, we get out of bed 1 h ahead of our biological wake-up time due to the misalignment of internal clocks with the light-dark cycle. Genetic discoveries revealed that clock genes encode transcription factors that are expressed throughout many tissues, yet a gap has remained in understanding the temporal dynamics of transcription. Two groups now apply circular chromosome conformation capture and high-throughput sequencing to dissect how "time of day"-dependent changes in chromatin drive core clock oscillations. A surprise is the finding that disruption of enhancer-promoter contacts within chromatin leads to an advance in the "wake-up" time of mice. Furthermore, the assembly of transcriptionally active domains of chromatin requires the ordered recruitment of core clock transcription factors each day. These studies show that waking up involves highly dynamic changes in the three-dimensional positioning of genes within the cell.


Subject(s)
Circadian Rhythm/genetics , Enhancer Elements, Genetic/physiology , Promoter Regions, Genetic/physiology , Animals , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Humans , Photoperiod , Promoter Regions, Genetic/genetics
8.
Cell Metab ; 25(1): 86-92, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27773696

ABSTRACT

Circadian clocks are encoded by a transcription-translation feedback loop that aligns energetic processes with the solar cycle. We show that genetic disruption of the clock activator BMAL1 in skeletal myotubes and fibroblasts increased levels of the hypoxia-inducible factor 1α (HIF1α) under hypoxic conditions. Bmal1-/- myotubes displayed reduced anaerobic glycolysis, mitochondrial respiration with glycolytic fuel, and transcription of HIF1α targets Phd3, Vegfa, Mct4, Pk-m, and Ldha, whereas abrogation of the clock repressors CRY1/2 stabilized HIF1α in response to hypoxia. HIF1α bound directly to core clock gene promoters, and, when co-expressed with BMAL1, led to transactivation of PER2-LUC and HRE-LUC reporters. Further, genetic stabilization of HIF1α in Vhl-/- cells altered circadian transcription. Finally, induction of clock and HIF1α target genes in response to strenuous exercise varied according to the time of day in wild-type mice. Collectively, our results reveal bidirectional interactions between circadian and HIF pathways that influence metabolic adaptation to hypoxia.


Subject(s)
Circadian Clocks , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Skeletal/metabolism , Oxygen/metabolism , Anaerobiosis , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Hypoxia/genetics , Hypoxia/metabolism , Mice , Organ Specificity , Oxygen Consumption , Physical Conditioning, Animal , Transcription, Genetic
9.
J Biol Rhythms ; 31(4): 323-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27440914

ABSTRACT

The mammalian circadian clock plays a central role in the temporal coordination of physiology across the 24-h light-dark cycle. A major function of the clock is to maintain energy constancy in anticipation of alternating periods of fasting and feeding that correspond with sleep and wakefulness. While it has long been recognized that humans exhibit robust variation in glucose tolerance and insulin sensitivity across the sleep-wake cycle, experimental genetic analysis has now revealed that the clock transcription cycle plays an essential role in insulin secretion and metabolic function within pancreatic beta cells. This review addresses how studies of the beta cell clock may elucidate the etiology of subtypes of diabetes associated with circadian and sleep cycle disruption, in addition to more general forms of the disease.


Subject(s)
Circadian Rhythm/genetics , Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , Insulin/metabolism , Transcription, Genetic , Animals , Circadian Clocks , Diabetes Mellitus, Type 2/genetics , Fasting , Genomics , Humans , Insulin Resistance/genetics , Insulin Secretion , Mice , Photoperiod , Sleep/physiology , Wakefulness
10.
Science ; 350(6261): aac4250, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26542580

ABSTRACT

The mammalian transcription factors CLOCK and BMAL1 are essential components of the molecular clock that coordinate behavior and metabolism with the solar cycle. Genetic or environmental perturbation of circadian cycles contributes to metabolic disorders including type 2 diabetes. To study the impact of the cell-autonomous clock on pancreatic ß cell function, we examined pancreatic islets from mice with either intact or disrupted BMAL1 expression both throughout life and limited to adulthood. We found pronounced oscillation of insulin secretion that was synchronized with the expression of genes encoding secretory machinery and signaling factors that regulate insulin release. CLOCK/BMAL1 colocalized with the pancreatic transcription factor PDX1 within active enhancers distinct from those controlling rhythmic metabolic gene networks in liver. We also found that ß cell clock ablation in adult mice caused severe glucose intolerance. Thus, cell type-specific enhancers underlie the circadian control of peripheral metabolism throughout life and may help to explain its dysregulation in diabetes.


Subject(s)
Circadian Rhythm/genetics , Enhancer Elements, Genetic/physiology , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Insulin/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Exocytosis/genetics , Glucose Intolerance , Homeodomain Proteins/metabolism , Humans , Insulin Secretion , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Trans-Activators/metabolism , Transcription, Genetic
11.
Science ; 342(6158): 1243417, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24051248

ABSTRACT

Circadian clocks are self-sustained cellular oscillators that synchronize oxidative and reductive cycles in anticipation of the solar cycle. We found that the clock transcription feedback loop produces cycles of nicotinamide adenine dinucleotide (NAD(+)) biosynthesis, adenosine triphosphate production, and mitochondrial respiration through modulation of mitochondrial protein acetylation to synchronize oxidative metabolic pathways with the 24-hour fasting and feeding cycle. Circadian control of the activity of the NAD(+)-dependent deacetylase sirtuin 3 (SIRT3) generated rhythms in the acetylation and activity of oxidative enzymes and respiration in isolated mitochondria, and NAD(+) supplementation restored protein deacetylation and enhanced oxygen consumption in circadian mutant mice. Thus, circadian control of NAD(+) bioavailability modulates mitochondrial oxidative function and organismal metabolism across the daily cycles of fasting and feeding.


Subject(s)
Circadian Clocks/physiology , Energy Metabolism , Mitochondria, Liver/metabolism , NAD/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Acetylation , Animals , Circadian Clocks/genetics , Fasting , Lipid Metabolism , Liver/metabolism , Mice , Mice, Knockout , Oxidation-Reduction , Oxygen Consumption , Sirtuin 3/genetics , Sirtuin 3/metabolism
12.
Methods Mol Biol ; 1077: 285-302, 2013.
Article in English | MEDLINE | ID: mdl-24014414

ABSTRACT

Many of our behavioral and physiological processes display daily oscillations that are under the control of the circadian clock. The core molecular clock network is present in both the brain and peripheral tissues and is composed of a complex series of interlocking transcriptional/translational feedback loops that oscillate with a periodicity of ~24 h. Recent evidence has implicated NAD(+) biosynthesis and the sirtuin family of NAD(+)-dependent protein deacetylases as part of a novel feedback loop within the core clock network, findings which underscore the importance of taking circadian timing into consideration when designing and interpreting metabolic studies, particularly in regard to sirtuin biology. Thus, this chapter introduces both in vivo and in vitro circadian methods to analyze various sirtuin-related endpoints across the light-dark cycle and discusses the transcriptional, biochemical, and physiological outputs of the clock.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , NAD/metabolism , Sirtuins/metabolism , Animals , Feedback, Physiological , Locomotion , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Sirtuins/genetics , Transcription, Genetic
13.
Semin Ophthalmol ; 28(5-6): 406-21, 2013.
Article in English | MEDLINE | ID: mdl-24010846

ABSTRACT

Daily changes in the light-dark cycle are the principal environmental signal that enables organisms to synchronize their internal biology with the 24-hour day-night cycle. In humans, the visual system is integral to photoentrainment and is primarily driven by a specialized class of intrinsically photosensitive retinal ganglion cells (ipRGCs) that express the photopigment melanopsin (OPN4) in the inner retina. These cells project through the retinohypothalamic tract (RHT) to the suprachiasmatic nuclei (SCN) of the hypothalamus, which serves as the body's master biological clock. At the same time, the retina itself possesses intrinsic circadian oscillations, exemplified by diurnal fluctuations in visual sensitivity, neurotransmitter levels, and outer segment turnover rates. Recently, it has been noted that both central and peripheral oscillators share a molecular clock consisting of an endogenous, circadian-driven, transcription-translation feedback loop that cycles with a periodicity of approximately 24 hours. This review will cover the role that melanopsin and ipRGCs play in the circadian organization of the visual system.


Subject(s)
Circadian Rhythm/physiology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Rod Opsins/physiology , Vision, Ocular/physiology , Animals , Humans , Light , Suprachiasmatic Nucleus/physiology
14.
Cell Logist ; 1(1): 32-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21686102

ABSTRACT

The molecular clock controls 24-hour cycles of behavioral and physiological processes across the day-night cycle. Disruption of circadian rhythmicity has been implicated in the pathogenesis of several diseases, including the metabolic syndrome, although the role of clock genes in these disorders is still not well understood. Studies of the etiology of diabetes in circadian mutant mice have revealed a novel role for the clock in pancreatic ß-cell insulin secretion, suggesting that a major cellular function of the circadian network involves control of protein exocytosis.

15.
J Clin Invest ; 121(6): 2133-41, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21633182

ABSTRACT

The discovery of the genetic basis for circadian rhythms has expanded our knowledge of the temporal organization of behavior and physiology. The observations that the circadian gene network is present in most living organisms from eubacteria to humans, that most cells and tissues express autonomous clocks, and that disruption of clock genes results in metabolic dysregulation have revealed interactions between metabolism and circadian rhythms at neural, molecular, and cellular levels. A major challenge remains in understanding the interplay between brain and peripheral clocks and in determining how these interactions promote energy homeostasis across the sleep-wake cycle. In this Review, we evaluate how investigation of molecular timing may create new opportunities to understand and develop therapies for obesity and diabetes.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism/physiology , Sleep/physiology , Animals , Brain/physiology , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/physiology , Diabetes Mellitus, Type 2/physiopathology , Diet , Dietary Fats/adverse effects , Glucose/metabolism , Homeostasis/physiology , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Mutant Strains , Nerve Net/physiology , Neuropeptides/physiology , Obesity/physiopathology , Orexins , Signal Transduction , Sleep Deprivation/physiopathology , Sleep Disorders, Circadian Rhythm/physiopathology
16.
Best Pract Res Clin Endocrinol Metab ; 24(5): 785-800, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21112026

ABSTRACT

Social opportunities and work demands have caused humans to become increasingly active during the late evening hours, leading to a shift from the predominantly diurnal lifestyle of our ancestors to a more nocturnal one. This voluntarily decision to stay awake long into the evening hours leads to circadian disruption at the system, tissue, and cellular levels. These derangements are in turn associated with clinical impairments in metabolic processes and physiology. The use of animal models for circadian disruption provides an important opportunity to determine mechanisms by which disorganization in the circadian system can lead to metabolic dysfunction in response to genetic, environmental, and behavioral perturbations. Here we review recent key animal studies involving circadian disruption and discuss the possible translational implications of these studies for human health and particularly for the development of metabolic disease.


Subject(s)
Circadian Rhythm/physiology , Disease Models, Animal , Metabolic Diseases/etiology , Sleep Disorders, Circadian Rhythm/complications , Animals , Biological Clocks/physiology , Brain/physiopathology , Humans , Metabolic Diseases/pathology , Metabolic Diseases/physiopathology , Models, Biological , Photoperiod , Sleep Disorders, Circadian Rhythm/metabolism , Sleep Disorders, Circadian Rhythm/pathology , Sleep Disorders, Circadian Rhythm/physiopathology
17.
Nature ; 466(7306): 627-31, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20562852

ABSTRACT

The molecular clock maintains energy constancy by producing circadian oscillations of rate-limiting enzymes involved in tissue metabolism across the day and night. During periods of feeding, pancreatic islets secrete insulin to maintain glucose homeostasis, and although rhythmic control of insulin release is recognized to be dysregulated in humans with diabetes, it is not known how the circadian clock may affect this process. Here we show that pancreatic islets possess self-sustained circadian gene and protein oscillations of the transcription factors CLOCK and BMAL1. The phase of oscillation of islet genes involved in growth, glucose metabolism and insulin signalling is delayed in circadian mutant mice, and both Clock and Bmal1 (also called Arntl) mutants show impaired glucose tolerance, reduced insulin secretion and defects in size and proliferation of pancreatic islets that worsen with age. Clock disruption leads to transcriptome-wide alterations in the expression of islet genes involved in growth, survival and synaptic vesicle assembly. Notably, conditional ablation of the pancreatic clock causes diabetes mellitus due to defective beta-cell function at the very latest stage of stimulus-secretion coupling. These results demonstrate a role for the beta-cell clock in coordinating insulin secretion with the sleep-wake cycle, and reveal that ablation of the pancreatic clock can trigger the onset of diabetes mellitus.


Subject(s)
ARNTL Transcription Factors/genetics , CLOCK Proteins/genetics , Circadian Rhythm/physiology , Diabetes Mellitus/metabolism , Insulin/blood , Islets of Langerhans/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/metabolism , Aging/genetics , Aging/pathology , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , CLOCK Proteins/deficiency , CLOCK Proteins/metabolism , Cell Proliferation , Cell Size , Cell Survival , Circadian Rhythm/genetics , Diabetes Mellitus/genetics , Gene Expression Profiling , Glucose Intolerance/genetics , Glucose Tolerance Test , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/pathology , Mice , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Phenotype , Sleep/genetics , Sleep/physiology , Synaptic Vesicles/metabolism , Wakefulness/genetics , Wakefulness/physiology
18.
Circ Res ; 106(3): 447-62, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20167942

ABSTRACT

The incidence of the metabolic syndrome represents a spectrum of disorders that continue to increase across the industrialized world. Both genetic and environmental factors contribute to metabolic syndrome and recent evidence has emerged to suggest that alterations in circadian systems and sleep participate in the pathogenesis of the disease. In this review, we highlight studies at the intersection of clinical medicine and experimental genetics that pinpoint how perturbations of the internal clock system, and sleep, constitute risk factors for disorders including obesity, diabetes mellitus, cardiovascular disease, thrombosis and even inflammation. An exciting aspect of the field has been the integration of behavioral and physiological approaches, and the emerging insight into both neural and peripheral tissues in disease pathogenesis. Consideration of the cell and molecular links between disorders of circadian rhythms and sleep with metabolic syndrome has begun to open new opportunities for mechanism-based therapeutics.


Subject(s)
Circadian Rhythm/physiology , Metabolic Syndrome/physiopathology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/physiology , Adipose Tissue/metabolism , Adult , Animals , CLOCK Proteins/deficiency , CLOCK Proteins/genetics , CLOCK Proteins/physiology , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/physiopathology , Circadian Rhythm/genetics , Circadian Rhythm/immunology , Circadian Rhythm/radiation effects , Cytokines/physiology , Dyssomnias/physiopathology , Eating/physiology , Energy Metabolism/physiology , Feedback, Physiological , Gene Expression Regulation , Hormones/metabolism , Humans , Incidence , Inflammation/physiopathology , Lighting/adverse effects , Liver/metabolism , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Metabolic Syndrome/genetics , Mice , Mice, Knockout , Middle Aged , Nicotinamide Phosphoribosyltransferase/physiology , Sirtuin 1/physiology , Sleep Deprivation/physiopathology
19.
J Appl Physiol (1985) ; 107(5): 1638-46, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19661448

ABSTRACT

The circadian system is a key integrator of behavior and metabolism that synchronizes physiological processes with the rotation of the Earth on its axis. In mammals, the clock is present not only within the central pacemaker neurons of the hypothalamus, but also within extra-suprachiasmatic nucleus (SCN) regions of brain and nearly all peripheral tissues. Recent evidence suggests that the complex feedback networks that encompass both the circadian and metabolic systems are intimately intertwined and that disruption of either system leads to reciprocal disturbances in the other. We anticipate that improved understanding of the interconnections between the circadian and metabolic networks will open new windows on the treatment of sleep and metabolic disorders, including diabetes mellitus and obesity.


Subject(s)
Chronobiology Disorders/complications , Chronobiology Disorders/physiopathology , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Metabolic Diseases/complications , Metabolic Diseases/physiopathology , Models, Biological , Animals , Energy Metabolism , Humans
20.
Science ; 324(5927): 651-4, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19299583

ABSTRACT

The circadian clock is encoded by a transcription-translation feedback loop that synchronizes behavior and metabolism with the light-dark cycle. Here we report that both the rate-limiting enzyme in mammalian nicotinamide adenine dinucleotide (NAD+) biosynthesis, nicotinamide phosphoribosyltransferase (NAMPT), and levels of NAD+ display circadian oscillations that are regulated by the core clock machinery in mice. Inhibition of NAMPT promotes oscillation of the clock gene Per2 by releasing CLOCK:BMAL1 from suppression by SIRT1. In turn, the circadian transcription factor CLOCK binds to and up-regulates Nampt, thus completing a feedback loop involving NAMPT/NAD+ and SIRT1/CLOCK:BMAL1.


Subject(s)
Biological Clocks , Circadian Rhythm , Cytokines/metabolism , Feedback, Physiological , NAD/biosynthesis , Nicotinamide Phosphoribosyltransferase/metabolism , ARNTL Transcription Factors , Acrylamides/pharmacology , Adipose Tissue, White/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Nuclear Proteins/genetics , Period Circadian Proteins , Piperidines/pharmacology , Protein Binding , Sirtuin 1 , Sirtuins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...