Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Cell ; 177(4): 896-909.e20, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31030999

ABSTRACT

In mammals, endogenous circadian clocks sense and respond to daily feeding and lighting cues, adjusting internal ∼24 h rhythms to resonate with, and anticipate, external cycles of day and night. The mechanism underlying circadian entrainment to feeding time is critical for understanding why mistimed feeding, as occurs during shift work, disrupts circadian physiology, a state that is associated with increased incidence of chronic diseases such as type 2 (T2) diabetes. We show that feeding-regulated hormones insulin and insulin-like growth factor 1 (IGF-1) reset circadian clocks in vivo and in vitro by induction of PERIOD proteins, and mistimed insulin signaling disrupts circadian organization of mouse behavior and clock gene expression. Insulin and IGF-1 receptor signaling is sufficient to determine essential circadian parameters, principally via increased PERIOD protein synthesis. This requires coincident mechanistic target of rapamycin (mTOR) activation, increased phosphoinositide signaling, and microRNA downregulation. Besides its well-known homeostatic functions, we propose insulin and IGF-1 are primary signals of feeding time to cellular clocks throughout the body.


Subject(s)
Circadian Clocks/physiology , Feeding Behavior/physiology , Period Circadian Proteins/metabolism , Animals , Circadian Rhythm/physiology , Female , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Mammals/metabolism , Mice , Mice, Inbred C57BL , Receptor, IGF Type 1/metabolism , Signal Transduction
2.
Cell ; 163(5): 1214-1224, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26590423

ABSTRACT

Circadian clocks integrate light and temperature input to remain synchronized with the day/night cycle. Although light input to the clock is well studied, the molecular mechanisms by which circadian clocks respond to temperature remain poorly understood. We found that temperature phase shifts Drosophila circadian clocks through degradation of the pacemaker protein TIM. This degradation is mechanistically distinct from photic CRY-dependent TIM degradation. Thermal TIM degradation is triggered by cytosolic calcium increase and CALMODULIN binding to TIM and is mediated by the atypical calpain protease SOL. This thermal input pathway and CRY-dependent light input thus converge on TIM, providing a molecular mechanism for the integration of circadian light and temperature inputs. Mammals use body temperature cycles to keep peripheral clocks synchronized with their brain pacemaker. Interestingly, downregulating the mammalian SOL homolog SOLH blocks thermal mPER2 degradation and phase shifts. Thus, we propose that circadian thermosensation in insects and mammals share common principles.


Subject(s)
Circadian Clocks , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Nerve Tissue Proteins/metabolism , Animals , Biological Clocks , Calcium Signaling , Calmodulin/metabolism , Calpain , Circadian Rhythm , Male , Mammals/physiology , Proteolysis
3.
Nature ; 623(7988): 842-852, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37853127

ABSTRACT

Optimum protein function and biochemical activity critically depends on water availability because solvent thermodynamics drive protein folding and macromolecular interactions1. Reciprocally, macromolecules restrict the movement of 'structured' water molecules within their hydration layers, reducing the available 'free' bulk solvent and therefore the total thermodynamic potential energy of water, or water potential. Here, within concentrated macromolecular solutions such as the cytosol, we found that modest changes in temperature greatly affect the water potential, and are counteracted by opposing changes in osmotic strength. This duality of temperature and osmotic strength enables simple manipulations of solvent thermodynamics to prevent cell death after extreme cold or heat shock. Physiologically, cells must sustain their activity against fluctuating temperature, pressure and osmotic strength, which impact water availability within seconds. Yet, established mechanisms of water homeostasis act over much slower timescales2,3; we therefore postulated the existence of a rapid compensatory response. We find that this function is performed by water potential-driven changes in macromolecular assembly, particularly biomolecular condensation of intrinsically disordered proteins. The formation and dissolution of biomolecular condensates liberates and captures free water, respectively, quickly counteracting thermal or osmotic perturbations of water potential, which is consequently robustly buffered in the cytoplasm. Our results indicate that biomolecular condensation constitutes an intrinsic biophysical feedback response that rapidly compensates for intracellular osmotic and thermal fluctuations. We suggest that preserving water availability within the concentrated cytosol is an overlooked evolutionary driver of protein (dis)order and function.


Subject(s)
Macromolecular Substances , Proteins , Solvents , Thermodynamics , Water , Cell Death , Cytosol/chemistry , Cytosol/metabolism , Homeostasis , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Osmolar Concentration , Pressure , Proteins/chemistry , Proteins/metabolism , Solvents/chemistry , Solvents/metabolism , Temperature , Time Factors , Water/chemistry , Water/metabolism
4.
EMBO J ; 43(13): 2813-2833, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38778155

ABSTRACT

Although costly to maintain, protein homeostasis is indispensable for normal cellular function and long-term health. In mammalian cells and tissues, daily variation in global protein synthesis has been observed, but its utility and consequences for proteome integrity are not fully understood. Using several different pulse-labelling strategies, here we gain direct insight into the relationship between protein synthesis and abundance proteome-wide. We show that protein degradation varies in-phase with protein synthesis, facilitating rhythms in turnover rather than abundance. This results in daily consolidation of proteome renewal whilst minimising changes in composition. Coupled rhythms in synthesis and turnover are especially salient to the assembly of macromolecular protein complexes, particularly the ribosome, the most abundant species of complex in the cell. Daily turnover and proteasomal degradation rhythms render cells and mice more sensitive to proteotoxic stress at specific times of day, potentially contributing to daily rhythms in the efficacy of proteasomal inhibitors against cancer. Our findings suggest that circadian rhythms function to minimise the bioenergetic cost of protein homeostasis through temporal consolidation of protein turnover.


Subject(s)
Circadian Rhythm , Proteome , Animals , Circadian Rhythm/physiology , Proteome/metabolism , Mice , Protein Biosynthesis , Humans , Proteasome Endopeptidase Complex/metabolism , Ribosomes/metabolism , Proteolysis , Proteostasis , Mice, Inbred C57BL
5.
EMBO J ; 42(19): e114164, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37554073

ABSTRACT

Cellular circadian rhythms confer temporal organisation upon physiology that is fundamental to human health. Rhythms are present in red blood cells (RBCs), the most abundant cell type in the body, but their physiological function is poorly understood. Here, we present a novel biochemical assay for haemoglobin (Hb) oxidation status which relies on a redox-sensitive covalent haem-Hb linkage that forms during SDS-mediated cell lysis. Formation of this linkage is lowest when ferrous Hb is oxidised, in the form of ferric metHb. Daily haemoglobin oxidation rhythms are observed in mouse and human RBCs cultured in vitro, or taken from humans in vivo, and are unaffected by mutations that affect circadian rhythms in nucleated cells. These rhythms correlate with daily rhythms in core body temperature, with temperature lowest when metHb levels are highest. Raising metHb levels with dietary sodium nitrite can further decrease daytime core body temperature in mice via nitric oxide (NO) signalling. These results extend our molecular understanding of RBC circadian rhythms and suggest they contribute to the regulation of body temperature.


Subject(s)
Erythrocytes , Hemoglobins , Humans , Mice , Animals , Erythrocytes/metabolism , Hemoglobins/metabolism , Oxidation-Reduction , Heme/metabolism , Circadian Rhythm
7.
EMBO J ; 41(1): e108883, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34842284

ABSTRACT

The daily organisation of most mammalian cellular functions is attributed to circadian regulation of clock-controlled protein expression, driven by daily cycles of CRYPTOCHROME-dependent transcriptional feedback repression. To test this, we used quantitative mass spectrometry to compare wild-type and CRY-deficient fibroblasts under constant conditions. In CRY-deficient cells, we found that temporal variation in protein, phosphopeptide, and K+ abundance was at least as great as wild-type controls. Most strikingly, the extent of temporal variation within either genotype was much smaller than overall differences in proteome composition between WT and CRY-deficient cells. This proteome imbalance in CRY-deficient cells and tissues was associated with increased susceptibility to proteotoxic stress, which impairs circadian robustness, and may contribute to the wide-ranging phenotypes of CRY-deficient mice. Rather than generating large-scale daily variation in proteome composition, we suggest it is plausible that the various transcriptional and post-translational functions of CRY proteins ultimately act to maintain protein and osmotic homeostasis against daily perturbation.


Subject(s)
Circadian Rhythm/physiology , Cryptochromes/metabolism , Proteostasis , Animals , Cryptochromes/deficiency , Ion Transport , Mice , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteome/metabolism , Proteomics , Reproducibility of Results , Stress, Physiological , Time Factors
8.
Mol Cell ; 69(4): 581-593.e7, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452638

ABSTRACT

The bioenergetics and molecular determinants of the metabolic response to mitochondrial dysfunction are incompletely understood, in part due to a lack of appropriate isogenic cellular models of primary mitochondrial defects. Here, we capitalize on a recently developed cell model with defined levels of m.8993T>G mutation heteroplasmy, mTUNE, to investigate the metabolic underpinnings of mitochondrial dysfunction. We found that impaired utilization of reduced nicotinamide adenine dinucleotide (NADH) by the mitochondrial respiratory chain leads to cytosolic reductive carboxylation of glutamine as a new mechanism for cytosol-confined NADH recycling supported by malate dehydrogenase 1 (MDH1). We also observed that increased glycolysis in cells with mitochondrial dysfunction is associated with increased cell migration in an MDH1-dependent fashion. Our results describe a novel link between glycolysis and mitochondrial dysfunction mediated by reductive carboxylation of glutamine.


Subject(s)
Cytosol/metabolism , Glutamine/metabolism , Malate Dehydrogenase/metabolism , Mitochondria/pathology , NAD/metabolism , Osteosarcoma/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Citric Acid Cycle , DNA, Mitochondrial/genetics , Energy Metabolism , Female , Glucose/metabolism , Glycolysis , Humans , Mitochondria/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , Oxidation-Reduction , Tumor Cells, Cultured
9.
EMBO J ; 40(7): e106745, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33491228

ABSTRACT

Circadian rhythms are a pervasive property of mammalian cells, tissues and behaviour, ensuring physiological adaptation to solar time. Models of cellular timekeeping revolve around transcriptional feedback repression, whereby CLOCK and BMAL1 activate the expression of PERIOD (PER) and CRYPTOCHROME (CRY), which in turn repress CLOCK/BMAL1 activity. CRY proteins are therefore considered essential components of the cellular clock mechanism, supported by behavioural arrhythmicity of CRY-deficient (CKO) mice under constant conditions. Challenging this interpretation, we find locomotor rhythms in adult CKO mice under specific environmental conditions and circadian rhythms in cellular PER2 levels when CRY is absent. CRY-less oscillations are variable in their expression and have shorter periods than wild-type controls. Importantly, we find classic circadian hallmarks such as temperature compensation and period determination by CK1δ/ε activity to be maintained. In the absence of CRY-mediated feedback repression and rhythmic Per2 transcription, PER2 protein rhythms are sustained for several cycles, accompanied by circadian variation in protein stability. We suggest that, whereas circadian transcriptional feedback imparts robustness and functionality onto biological clocks, the core timekeeping mechanism is post-translational.


Subject(s)
Circadian Rhythm , Cryptochromes/metabolism , Animals , Cells, Cultured , Cryptochromes/deficiency , Cryptochromes/genetics , Drosophila melanogaster , Female , Locomotion , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
11.
Brain ; 145(6): 2031-2048, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35691613

ABSTRACT

Patients undergo interventions to achieve a 'normal' brain temperature; a parameter that remains undefined for humans. The profound sensitivity of neuronal function to temperature implies the brain should be isothermal, but observations from patients and non-human primates suggest significant spatiotemporal variation. We aimed to determine the clinical relevance of brain temperature in patients by establishing how much it varies in healthy adults. We retrospectively screened data for all patients recruited to the Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) High Resolution Intensive Care Unit Sub-Study. Only patients with direct brain temperature measurements and without targeted temperature management were included. To interpret patient analyses, we prospectively recruited 40 healthy adults (20 males, 20 females, 20-40 years) for brain thermometry using magnetic resonance spectroscopy. Participants were scanned in the morning, afternoon, and late evening of a single day. In patients (n = 114), brain temperature ranged from 32.6 to 42.3°C and mean brain temperature (38.5 ± 0.8°C) exceeded body temperature (37.5 ± 0.5°C, P < 0.0001). Of 100 patients eligible for brain temperature rhythm analysis, 25 displayed a daily rhythm, and the brain temperature range decreased in older patients (P = 0.018). In healthy participants, brain temperature ranged from 36.1 to 40.9°C; mean brain temperature (38.5 ± 0.4°C) exceeded oral temperature (36.0 ± 0.5°C) and was 0.36°C higher in luteal females relative to follicular females and males (P = 0.0006 and P < 0.0001, respectively). Temperature increased with age, most notably in deep brain regions (0.6°C over 20 years, P = 0.0002), and varied spatially by 2.41 ± 0.46°C with highest temperatures in the thalamus. Brain temperature varied by time of day, especially in deep regions (0.86°C, P = 0.0001), and was lowest at night. From the healthy data we built HEATWAVE-a 4D map of human brain temperature. Testing the clinical relevance of HEATWAVE in patients, we found that lack of a daily brain temperature rhythm increased the odds of death in intensive care 21-fold (P = 0.016), whilst absolute temperature maxima or minima did not predict outcome. A warmer mean brain temperature was associated with survival (P = 0.035), however, and ageing by 10 years increased the odds of death 11-fold (P = 0.0002). Human brain temperature is higher and varies more than previously assumed-by age, sex, menstrual cycle, brain region, and time of day. This has major implications for temperature monitoring and management, with daily brain temperature rhythmicity emerging as one of the strongest single predictors of survival after brain injury. We conclude that daily rhythmic brain temperature variation-not absolute brain temperature-is one way in which human brain physiology may be distinguished from pathophysiology.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Hypothermia, Induced , Adult , Aged , Body Temperature/physiology , Brain/physiology , Brain Injuries/complications , Brain Injuries, Traumatic/complications , Female , Humans , Male , Retrospective Studies , Temperature
12.
PLoS Comput Biol ; 17(5): e1008987, 2021 05.
Article in English | MEDLINE | ID: mdl-34048425

ABSTRACT

Modification of the Per2 clock gene in mPer2Luc reporter mice significantly alters circadian function. Behavioral period in constant dark is lengthened, and dissociates into two distinct components in constant light. Rhythms exhibit increased bimodality, enhanced phase resetting to light pulses, and altered entrainment to scheduled feeding. Mechanistic mathematical modelling predicts that enhanced protein interactions with the modified mPER2 C-terminus, combined with differential clock regulation among SCN subregions, can account for effects on circadian behavior via increased Per2 transcript and protein stability. PER2::LUC produces greater suppression of CLOCK:BMAL1 E-box activity than PER2. mPer2Luc carries a 72 bp deletion in exon 23 of Per2, and retains a neomycin resistance cassette that affects rhythm amplitude but not period. The results show that mPer2Luc acts as a circadian clock mutation illustrating a need for detailed assessment of potential impacts of c-terminal tags in genetically modified animal models.


Subject(s)
Circadian Rhythm , Luciferases/genetics , Period Circadian Proteins/genetics , Animals , Behavior, Animal , Feeding Behavior , Locomotion , Mice , Mice, Inbred C57BL , Mutation
13.
Nature ; 532(7599): 375-9, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27074515

ABSTRACT

Circadian clocks are fundamental to the biology of most eukaryotes, coordinating behaviour and physiology to resonate with the environmental cycle of day and night through complex networks of clock-controlled genes. A fundamental knowledge gap exists, however, between circadian gene expression cycles and the biochemical mechanisms that ultimately facilitate circadian regulation of cell biology. Here we report circadian rhythms in the intracellular concentration of magnesium ions, [Mg(2+)]i, which act as a cell-autonomous timekeeping component to determine key clock properties both in a human cell line and in a unicellular alga that diverged from each other more than 1 billion years ago. Given the essential role of Mg(2+) as a cofactor for ATP, a functional consequence of [Mg(2+)]i oscillations is dynamic regulation of cellular energy expenditure over the daily cycle. Mechanistically, we find that these rhythms provide bilateral feedback linking rhythmic metabolism to clock-controlled gene expression. The global regulation of nucleotide triphosphate turnover by intracellular Mg(2+) availability has potential to impact upon many of the cell's more than 600 MgATP-dependent enzymes and every cellular system where MgNTP hydrolysis becomes rate limiting. Indeed, we find that circadian control of translation by mTOR is regulated through [Mg(2+)]i oscillations. It will now be important to identify which additional biological processes are subject to this form of regulation in tissues of multicellular organisms such as plants and humans, in the context of health and disease.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Energy Metabolism , Magnesium/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Chlorophyta/cytology , Chlorophyta/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Feedback, Physiological , Gene Expression Regulation , Humans , Intracellular Space/metabolism , Male , Mice , TOR Serine-Threonine Kinases/metabolism , Time Factors
14.
Proc Natl Acad Sci U S A ; 113(36): 10085-90, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27528682

ABSTRACT

Viruses are intracellular pathogens that hijack host cell machinery and resources to replicate. Rather than being constant, host physiology is rhythmic, undergoing circadian (∼24 h) oscillations in many virus-relevant pathways, but whether daily rhythms impact on viral replication is unknown. We find that the time of day of host infection regulates virus progression in live mice and individual cells. Furthermore, we demonstrate that herpes and influenza A virus infections are enhanced when host circadian rhythms are abolished by disrupting the key clock gene transcription factor Bmal1. Intracellular trafficking, biosynthetic processes, protein synthesis, and chromatin assembly all contribute to circadian regulation of virus infection. Moreover, herpesviruses differentially target components of the molecular circadian clockwork. Our work demonstrates that viruses exploit the clockwork for their own gain and that the clock represents a novel target for modulating viral replication that extends beyond any single family of these ubiquitous pathogens.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks/genetics , Herpes Simplex/virology , Herpesviridae Infections/virology , Host-Pathogen Interactions , Orthomyxoviridae Infections/virology , Tumor Virus Infections/virology , ARNTL Transcription Factors/deficiency , Animals , Biological Transport , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Line , Chromatin Assembly and Disassembly , Circadian Rhythm/genetics , Cricetinae , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Gene Expression Regulation , Genes, Reporter , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpesviridae Infections/genetics , Herpesviridae Infections/metabolism , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Rhadinovirus/pathogenicity , Rhadinovirus/physiology , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism , Virus Replication
15.
Nat Chem Biol ; 17(4): 373-374, 2021 04.
Article in English | MEDLINE | ID: mdl-33686295
16.
Nature ; 485(7399): 459-64, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22622569

ABSTRACT

Cellular life emerged ∼3.7 billion years ago. With scant exception, terrestrial organisms have evolved under predictable daily cycles owing to the Earth's rotation. The advantage conferred on organisms that anticipate such environmental cycles has driven the evolution of endogenous circadian rhythms that tune internal physiology to external conditions. The molecular phylogeny of mechanisms driving these rhythms has been difficult to dissect because identified clock genes and proteins are not conserved across the domains of life: Bacteria, Archaea and Eukaryota. Here we show that oxidation-reduction cycles of peroxiredoxin proteins constitute a universal marker for circadian rhythms in all domains of life, by characterizing their oscillations in a variety of model organisms. Furthermore, we explore the interconnectivity between these metabolic cycles and transcription-translation feedback loops of the clockwork in each system. Our results suggest an intimate co-evolution of cellular timekeeping with redox homeostatic mechanisms after the Great Oxidation Event ∼2.5 billion years ago.


Subject(s)
Circadian Rhythm/physiology , Conserved Sequence , Evolution, Molecular , Peroxiredoxins/metabolism , Amino Acid Sequence , Animals , Archaea/metabolism , Bacteria/metabolism , Biomarkers/metabolism , Catalytic Domain , Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/genetics , Eukaryotic Cells/metabolism , Feedback, Physiological , Homeostasis , Humans , Models, Biological , Molecular Sequence Data , Oxidation-Reduction , Peroxiredoxins/chemistry , Phylogeny , Prokaryotic Cells/metabolism , Protein Biosynthesis , Transcription, Genetic
17.
Nature ; 469(7331): 498-503, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21270888

ABSTRACT

Circadian (∼24 hour) clocks are fundamentally important for coordinated physiology in organisms as diverse as cyanobacteria and humans. All current models of the molecular circadian clockwork in eukaryotic cells are based on transcription-translation feedback loops. Non-transcriptional mechanisms in the clockwork have been difficult to study in mammalian systems. We circumvented these problems by developing novel assays using human red blood cells, which have no nucleus (or DNA) and therefore cannot perform transcription. Our results show that transcription is not required for circadian oscillations in humans, and that non-transcriptional events seem to be sufficient to sustain cellular circadian rhythms. Using red blood cells, we found that peroxiredoxins, highly conserved antioxidant proteins, undergo ∼24-hour redox cycles, which persist for many days under constant conditions (that is, in the absence of external cues). Moreover, these rhythms are entrainable (that is, tunable by environmental stimuli) and temperature-compensated, both key features of circadian rhythms. We anticipate that our findings will facilitate more sophisticated cellular clock models, highlighting the interdependency of transcriptional and non-transcriptional oscillations in potentially all eukaryotic cells.


Subject(s)
Circadian Clocks/physiology , Erythrocytes/physiology , Cell Nucleus/metabolism , Erythrocytes/metabolism , Hemoglobins/metabolism , Humans , Oxidation-Reduction , Peroxiredoxins/metabolism , Phenotype , Temperature
18.
Nature ; 469(7331): 554-8, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21270895

ABSTRACT

Circadian rhythms are ubiquitous in eukaryotes, and coordinate numerous aspects of behaviour, physiology and metabolism, from sleep/wake cycles in mammals to growth and photosynthesis in plants. This daily timekeeping is thought to be driven by transcriptional-translational feedback loops, whereby rhythmic expression of 'clock' gene products regulates the expression of associated genes in approximately 24-hour cycles. The specific transcriptional components differ between phylogenetic kingdoms. The unicellular pico-eukaryotic alga Ostreococcus tauri possesses a naturally minimized clock, which includes many features that are shared with plants, such as a central negative feedback loop that involves the morning-expressed CCA1 and evening-expressed TOC1 genes. Given that recent observations in animals and plants have revealed prominent post-translational contributions to timekeeping, a reappraisal of the transcriptional contribution to oscillator function is overdue. Here we show that non-transcriptional mechanisms are sufficient to sustain circadian timekeeping in the eukaryotic lineage, although they normally function in conjunction with transcriptional components. We identify oxidation of peroxiredoxin proteins as a transcription-independent rhythmic biomarker, which is also rhythmic in mammals. Moreover we show that pharmacological modulators of the mammalian clock mechanism have the same effects on rhythms in Ostreococcus. Post-translational mechanisms, and at least one rhythmic marker, seem to be better conserved than transcriptional clock regulators. It is plausible that the oldest oscillator components are non-transcriptional in nature, as in cyanobacteria, and are conserved across kingdoms.


Subject(s)
Chlorophyta/physiology , Circadian Rhythm/physiology , Transcription, Genetic , Biomarkers/analysis , Chlorophyta/drug effects , Chlorophyta/metabolism , Cycloheximide/pharmacology , Deoxyadenosines/pharmacology , Gene Expression Regulation/drug effects , Oxidation-Reduction , Peroxiredoxins/metabolism , Protein Synthesis Inhibitors/pharmacology
19.
Proc Natl Acad Sci U S A ; 110(4): 1554-9, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23297224

ABSTRACT

Daily cyclical expression of thousands of genes in tissues such as the liver is orchestrated by the molecular circadian clock, the disruption of which is implicated in metabolic disorders and cancer. Although we understand much about the circadian transcription factors that can switch gene expression on and off, it is still unclear how global changes in rhythmic transcription are controlled at the genomic level. Here, we demonstrate circadian modification of an activating histone mark at a significant proportion of gene loci that undergo daily transcription, implicating widespread epigenetic modification as a key node regulated by the clockwork. Furthermore, we identify the histone-remodelling enzyme mixed lineage leukemia (MLL)3 as a clock-controlled factor that is able to directly and indirectly modulate over a hundred epigenetically targeted circadian "output" genes in the liver. Importantly, catalytic inactivation of the histone methyltransferase activity of MLL3 also severely compromises the oscillation of "core" clock gene promoters, including Bmal1, mCry1, mPer2, and Rev-erbα, suggesting that rhythmic histone methylation is vital for robust transcriptional oscillator function. This highlights a pathway by which the clockwork exerts genome-wide control over transcription, which is critical for sustaining temporal programming of tissue physiology.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , ARNTL Transcription Factors/genetics , Animals , Cell Line , Cryptochromes/deficiency , Cryptochromes/genetics , Epigenomics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Period Circadian Proteins/genetics , Promoter Regions, Genetic , Systems Biology , Transcription, Genetic
20.
Proc Natl Acad Sci U S A ; 110(23): 9547-52, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690615

ABSTRACT

The suprachiasmatic nucleus (SCN) coordinates circadian rhythms that adapt the individual to solar time. SCN pacemaking revolves around feedback loops in which expression of Period (Per) and Cryptochrome (Cry) genes is periodically suppressed by their protein products. Specifically, PER/CRY complexes act at E-box sequences in Per and Cry to inhibit their transactivation by CLOCK/BMAL1 heterodimers. To function effectively, these closed intracellular loops need to be synchronized between SCN cells and to the light/dark cycle. For Per expression, this is mediated by neuropeptidergic and glutamatergic extracellular cues acting via cAMP/calcium-responsive elements (CREs) in Per genes. Cry genes, however, carry no CREs, and how CRY-dependent SCN pacemaking is synchronized remains unclear. Furthermore, whereas reporter lines are available to explore Per circadian expression in real time, no Cry equivalent exists. We therefore created a mouse, B6.Cg-Tg(Cry1-luc)01Ld, carrying a transgene (mCry1-luc) consisting of mCry1 elements containing an E-box and E'-box driving firefly luciferase. mCry1-luc organotypic SCN slices exhibited stable circadian bioluminescence rhythms with appropriate phase, period, profile, and spatial organization. In SCN lacking vasoactive intestinal peptide or its receptor, mCry1 expression was damped and desynchronized between cells. Despite the absence of CREs, mCry1-luc expression was nevertheless (indirectly) sensitive to manipulation of cAMP-dependent signaling. In mPer1/2-null SCN, mCry1-luc bioluminescence was arrhythmic and no longer suppressed by elevation of cAMP. Finally, an SCN graft procedure showed that PER-independent as well as PER-dependent mechanisms could sustain circadian expression of mCry1. The mCry1-luc mouse therefore reports circadian mCry1 expression and its interactions with vasoactive intestinal peptide, cAMP, and PER at the heart of the SCN pacemaker.


Subject(s)
Circadian Rhythm/physiology , Cryptochromes/metabolism , Feedback, Physiological/physiology , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Cyclic AMP/metabolism , DNA Primers/genetics , Luciferases , Mice , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL