Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nat Rev Mol Cell Biol ; 20(4): 227-241, 2019 04.
Article in English | MEDLINE | ID: mdl-30635659

ABSTRACT

Humans, like all mammals, partition their daily behaviour into activity (wakefulness) and rest (sleep) phases that differ largely in their metabolic requirements. The circadian clock evolved as an autonomous timekeeping system that aligns behavioural patterns with the solar day and supports the body functions by anticipating and coordinating the required metabolic programmes. The key component of this synchronization is a master clock in the brain, which responds to light-darkness cues from the environment. However, to achieve circadian control of the entire organism, each cell of the body is equipped with its own circadian oscillator that is controlled by the master clock and confers rhythmicity to individual cells and organs through the control of rate-limiting steps of metabolic programmes. Importantly, metabolic regulation is not a mere output function of the circadian system, but nutrient, energy and redox levels signal back to cellular clocks in order to reinforce circadian rhythmicity and to adapt physiology to temporal tissue-specific needs. Thus, multiple systemic and molecular mechanisms exist that connect the circadian clock with metabolism at all levels, from cellular organelles to the whole organism, and deregulation of this circadian-metabolic crosstalk can lead to various pathologies.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Animals , Brain/physiology , Humans , Mammals/physiology
2.
Cell ; 161(1): 84-92, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25815987

ABSTRACT

The circadian clock, a highly specialized, hierarchical network of biological pacemakers, directs and maintains proper rhythms in endocrine and metabolic pathways required for organism homeostasis. The clock adapts to environmental changes, specifically daily light-dark cycles, as well as rhythmic food intake. Nutritional challenges reprogram the clock, while time-specific food intake has been shown to have profound consequences on physiology. Importantly, a critical role in the clock-nutrition interplay appears to be played by the microbiota. The circadian clock appears to operate as a critical interface between nutrition and homeostasis, calling for more attention on the beneficial effects of chrono-nutrition.


Subject(s)
Circadian Clocks , Eating , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology , Animals , Humans , Metabolomics
3.
PLoS Biol ; 21(11): e3002367, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37967106

ABSTRACT

In mammals, O2 and CO2 levels are tightly regulated and are altered under various pathological conditions. While the molecular mechanisms that participate in O2 sensing are well characterized, little is known regarding the signaling pathways that participate in CO2 signaling and adaptation. Here, we show that CO2 levels control a distinct cellular transcriptional response that differs from mere pH changes. Unexpectedly, we discovered that CO2 regulates the expression of cholesterogenic genes in a SREBP2-dependent manner and modulates cellular cholesterol accumulation. Molecular dissection of the underlying mechanism suggests that CO2 triggers SREBP2 activation through changes in endoplasmic reticulum (ER) membrane cholesterol levels. Collectively, we propose that SREBP2 participates in CO2 signaling and that cellular cholesterol levels can be modulated by CO2 through SREBP2.


Subject(s)
Carbon Dioxide , Cholesterol , Animals , Cholesterol/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Mammals/metabolism
4.
Cell ; 142(6): 943-53, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20832105

ABSTRACT

Circadian clocks in peripheral organs are tightly coupled to cellular metabolism and are readily entrained by feeding-fasting cycles. However, the molecular mechanisms involved are largely unknown. Here we show that in liver the activity of PARP-1, an NAD(+)-dependent ADP-ribosyltransferase, oscillates in a daily manner and is regulated by feeding. We provide biochemical evidence that PARP-1 binds and poly(ADP-ribosyl)ates CLOCK at the beginning of the light phase. The loss of PARP-1 enhances the binding of CLOCK-BMAL1 to DNA and leads to a phase-shift of the interaction of CLOCK-BMAL1 with PER and CRY repressor proteins. As a consequence, CLOCK-BMAL1-dependent gene expression is altered in PARP-1-deficient mice, in particular in response to changes in feeding times. Our results show that Parp-1 knockout mice exhibit impaired food entrainment of peripheral circadian clocks and support a role for PARP-1 in connecting feeding with the mammalian timing system.


Subject(s)
Biological Clocks , Circadian Rhythm , Feeding Behavior , Poly(ADP-ribose) Polymerases/metabolism , Animals , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Liver/metabolism , Mice , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics
5.
Proc Natl Acad Sci U S A ; 119(44): e2209933119, 2022 11.
Article in English | MEDLINE | ID: mdl-36279450

ABSTRACT

Circadian clocks are synchronized by external timing cues to align with one another and the environment. Various signaling pathways have been shown to independently reset the phase of the clock. However, in the body, circadian clocks are exposed to a multitude of potential timing cues with complex temporal dynamics, raising the question of how clocks integrate information in response to multiple signals. To investigate different modes of signal integration by the circadian clock, we used Circa-SCOPE, a method we recently developed for high-throughput phase resetting analysis. We found that simultaneous exposure to different combinations of known pharmacological resetting agents elicits a diverse range of responses. Often, the response was nonadditive and could not be readily predicted by the response to the individual signals. For instance, we observed that dexamethasone is dominant over other tested inputs. In the case of signals administered sequentially, the background levels of a signal attenuated subsequent resetting by the same signal, but not by signals acting through a different pathway. This led us to examine whether the circadian clock is sensitive to relative rather than absolute levels of the signal. Importantly, our analysis revealed the involvement of a signal-specific fold-change detection mechanism in the clock response. This mechanism likely stems from properties of the signaling pathway that are upstream to the clock. Overall, our findings elucidate modes of input integration by the circadian clock, with potential relevance to clock resetting under both physiological and pathological conditions.


Subject(s)
Circadian Clocks , Circadian Clocks/physiology , Circadian Rhythm/physiology , Signal Transduction , Cues , Dexamethasone/pharmacology
6.
Hepatology ; 77(3): 1022-1035, 2023 03 01.
Article in English | MEDLINE | ID: mdl-35591797

ABSTRACT

The mammalian liver must cope with various metabolic and physiological changes that normally recur every day and primarily stem from daily cycles of rest-activity and fasting-feeding. Although a large body of evidence supports the reciprocal regulation of circadian rhythms and liver function, the research on the hepatic ultradian rhythms have largely been lagging behind. However, with the advent of more cost-effective high-throughput omics technologies, high-resolution time-lapse imaging, and more robust and powerful mathematical tools, several recent studies have shed new light on the presence and functions of hepatic ultradian rhythms. In this review, we will first very briefly discuss the basic principles of circadian rhythms, and then cover in greater details the recent literature related to ultradian rhythms. Specifically, we will highlight the prevalence and mechanisms of hepatic 12-h rhythms, and 8-h rhythms, which cycle at the second and third harmonics of circadian frequency. Finally, we also refer to ultradian rhythms with other frequencies and examine the limitations of the current approaches as well as the challenges related to identifying ultradian rhythm and addressing their molecular underpinnings.


Subject(s)
Ultradian Rhythm , Animals , Activity Cycles/physiology , Circadian Rhythm/physiology , Fasting , Liver , Mammals
7.
PLoS Biol ; 19(12): e3001492, 2021 12.
Article in English | MEDLINE | ID: mdl-34968386

ABSTRACT

Rhythmicity of biological processes can be elicited either in response to environmental cycles or driven by endogenous oscillators. In mammals, the circadian clock drives about 24-hour rhythms of multitude metabolic and physiological processes in anticipation to environmental daily oscillations. Also at the intersection of environment and metabolism is the protein kinase-AKT. It conveys extracellular signals, primarily feeding-related signals, to regulate various key cellular functions. Previous studies in mice identified rhythmicity in AKT activation (pAKT) with elevated levels in the fed state. However, it is still unknown whether rhythmic AKT activation can be driven through intrinsic mechanisms. Here, we inspected temporal changes in pAKT levels both in cultured cells and animal models. In cultured cells, pAKT levels showed circadian oscillations similar to those observed in livers of wild-type mice under free-running conditions. Unexpectedly, in livers of Per1,2-/- but not of Bmal1-/- mice we detected ultradian (about 16 hours) oscillations of pAKT levels. Importantly, the liver transcriptome of Per1,2-/- mice also showed ultradian rhythms, corresponding to pAKT rhythmicity and consisting of AKT-related genes and regulators. Overall, our findings reveal ultradian rhythms in liver gene expression and AKT phosphorylation that emerge in the absence of environmental rhythms and Per1,2-/- genes.


Subject(s)
Gene Expression Regulation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Ultradian Rhythm/genetics , Animals , Cells, Cultured , Circadian Clocks/genetics , Gene Expression/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Transcription Factors/metabolism , Transcriptome/genetics
8.
Mol Cell ; 62(4): 636-48, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27161994

ABSTRACT

Cells have evolved mechanisms to handle incompatible processes through temporal organization by circadian clocks and by spatial compartmentalization within organelles defined by lipid bilayers. Recent advances in lipidomics have led to identification of plentiful lipid species, yet our knowledge regarding their spatiotemporal organization is lagging behind. In this study, we quantitatively characterized the nuclear and mitochondrial lipidome in mouse liver throughout the day, upon different feeding regimens, and in clock-disrupted mice. Our analyses revealed potential connections between lipid species within and between lipid classes. Remarkably, we uncovered diurnal oscillations in lipid accumulation in the nucleus and mitochondria. These oscillations exhibited opposite phases and readily responded to feeding time. Furthermore, we found that the circadian clock coordinates the phase relation between the organelles. In summary, our study provides temporal and spatial depiction of lipid organization and reveals the presence and coordination of diurnal rhythmicity in intracellular organelles.


Subject(s)
Cell Nucleus/metabolism , Circadian Rhythm , Feeding Behavior , Lipid Metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Periodicity , Animals , Circadian Rhythm/genetics , Genotype , Male , Mice, Knockout , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Phenotype , Time Factors
9.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34426495

ABSTRACT

Exercise and circadian biology are closely intertwined with physiology and metabolism, yet the functional interaction between circadian clocks and exercise capacity is only partially characterized. Here, we tested different clock mutant mouse models to examine the effect of the circadian clock and clock proteins, namely PERIODs and BMAL1, on exercise capacity. We found that daytime variance in endurance exercise capacity is circadian clock controlled. Unlike wild-type mice, which outperform in the late compared with the early part of their active phase, PERIODs- and BMAL1-null mice do not show daytime variance in exercise capacity. It appears that BMAL1 impairs and PERIODs enhance exercise capacity in a daytime-dependent manner. An analysis of liver and muscle glycogen stores as well as muscle lipid utilization suggested that these daytime effects mostly relate to liver glycogen levels and correspond to the animals' feeding behavior. Furthermore, given that exercise capacity responds to training, we tested the effect of training at different times of the day and found that training in the late compared with the early part of the active phase improves exercise performance. Overall, our findings suggest that clock proteins shape exercise capacity in a daytime-dependent manner through changes in liver glycogen levels, likely due to their effect on animals' feeding behavior.


Subject(s)
CLOCK Proteins/physiology , Exercise Tolerance/physiology , Physical Conditioning, Animal/physiology , ARNTL Transcription Factors/physiology , Animals , CLOCK Proteins/genetics , Feeding Behavior , Female , Light , Liver Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Muscles/metabolism , Mutation , Period Circadian Proteins/physiology , Photoperiod , Sex Characteristics , Time Factors
10.
Cell ; 134(2): 317-28, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18662546

ABSTRACT

The mammalian circadian timing system is composed of a central pacemaker in the suprachiasmatic nucleus of the brain that synchronizes countless subsidiary oscillators in peripheral tissues. The rhythm-generating mechanism is thought to rely on a feedback loop involving positively and negatively acting transcription factors. BMAL1 and CLOCK activate the expression of Period (Per) and Cryptochrome (Cry) genes, and once PER and CRY proteins accumulate to a critical level they form complexes with BMAL1-CLOCK heterodimers and thereby repress the transcription of their own genes. Here, we show that SIRT1, an NAD(+)-dependent protein deacetylase, is required for high-magnitude circadian transcription of several core clock genes, including Bmal1, Rorgamma, Per2, and Cry1. SIRT1 binds CLOCK-BMAL1 in a circadian manner and promotes the deacetylation and degradation of PER2. Given the NAD(+) dependence of SIRT1 deacetylase activity, it is likely that SIRT1 connects cellular metabolism to the circadian core clockwork circuitry.


Subject(s)
Cell Cycle Proteins/metabolism , Circadian Rhythm , Nuclear Proteins/metabolism , Sirtuins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , ARNTL Transcription Factors , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Liver/metabolism , Mice , NIH 3T3 Cells , Period Circadian Proteins , Sirtuin 1
11.
Proc Natl Acad Sci U S A ; 117(1): 779-786, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31848250

ABSTRACT

The occurrence and sequelae of disorders that lead to hypoxic spells such as asthma, chronic obstructive pulmonary disease, and obstructive sleep apnea (OSA) exhibit daily variance. This prompted us to examine the interaction between the hypoxic response and the circadian clock in vivo. We found that the global transcriptional response to acute hypoxia is tissue-specific and time-of-day-dependent. In particular, clock components differentially responded at the transcriptional and posttranscriptional level, and these responses depended on an intact circadian clock. Importantly, exposure to hypoxia phase-shifted clocks in a tissue-dependent manner led to intertissue circadian clock misalignment. This differential response relied on the intrinsic properties of each tissue and could be recapitulated ex vivo. Notably, circadian misalignment was also elicited by intermittent hypoxia, a widely used model for OSA. Given that phase coherence between circadian clocks is considered favorable, we propose that hypoxia leads to circadian misalignment, contributing to the pathophysiology of OSA and potentially other diseases that involve hypoxia.


Subject(s)
Circadian Clocks/physiology , Hypoxia/physiopathology , Photoperiod , Sleep Apnea, Obstructive/physiopathology , Animals , Disease Models, Animal , Gene Expression Regulation/physiology , Humans , Hypoxia/etiology , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Mice , Oxygen/metabolism , RNA-Seq , Sleep Apnea, Obstructive/etiology
12.
Proc Natl Acad Sci U S A ; 113(12): E1673-82, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26862173

ABSTRACT

Mitochondria are major suppliers of cellular energy through nutrients oxidation. Little is known about the mechanisms that enable mitochondria to cope with changes in nutrient supply and energy demand that naturally occur throughout the day. To address this question, we applied MS-based quantitative proteomics on isolated mitochondria from mice killed throughout the day and identified extensive oscillations in the mitochondrial proteome. Remarkably, the majority of cycling mitochondrial proteins peaked during the early light phase. We found that rate-limiting mitochondrial enzymes that process lipids and carbohydrates accumulate in a diurnal manner and are dependent on the clock proteins PER1/2. In this conjuncture, we uncovered daily oscillations in mitochondrial respiration that peak during different times of the day in response to different nutrients. Notably, the diurnal regulation of mitochondrial respiration was blunted in mice lacking PER1/2 or on a high-fat diet. We propose that PERIOD proteins optimize mitochondrial metabolism to daily changes in energy supply/demand and thereby, serve as a rheostat for mitochondrial nutrient utilization.


Subject(s)
Circadian Rhythm/physiology , Mitochondria, Liver/physiology , Mitochondrial Proteins/metabolism , Period Circadian Proteins/physiology , Animals , Circadian Rhythm/genetics , Citric Acid Cycle , Diet, High-Fat , Dietary Fats/metabolism , Electron Transport , Fatty Acids/metabolism , Feeding Behavior/physiology , Gene Expression Profiling , Male , Mice , Mice, Knockout , Mitochondria, Liver/enzymology , Motor Activity , Period Circadian Proteins/deficiency , Period Circadian Proteins/genetics , Proteome , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
13.
Gastroenterology ; 150(3): 574-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26657326

ABSTRACT

The circadian clock is an endogenous biological timekeeping system that synchronizes physiology and behavior to day/night cycles. A wide variety of processes throughout the entire gastrointestinal tract and notably the liver appear to be under circadian control. These include various metabolic functions such as nutrient uptake, processing, and detoxification, which align organ function to cycle with nutrient supply and demand. Remarkably, genetic or environmental disruption of the circadian clock can cause metabolic diseases or exacerbate pathological states. In addition, modern lifestyles force more and more people worldwide into asynchrony between the external time and their circadian clock, resulting in a constant state of social jetlag. Recent evidence indicates that interactions between altered energy metabolism and disruptions in the circadian clock create a downward spiral that can lead to diabetes and other metabolic diseases. In this review, we provide an overview of rhythmic processes in the liver and highlight the functions of circadian clock genes under physiological and pathological conditions; we focus on their roles in regulation of hepatic glucose as well as lipid and bile acid metabolism and detoxification and their potential effects on the development of fatty liver and nonalcoholic steatohepatitis.


Subject(s)
Circadian Rhythm , Energy Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Homeostasis , Humans , Inactivation, Metabolic , Liver/pathology , Liver/physiopathology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Signal Transduction , Time Factors
14.
Biochim Biophys Acta ; 1851(8): 1017-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25483623

ABSTRACT

Lipids play vital roles in a wide variety of cellular functions. They act as structural components in cell membranes, serve as a major form of energy storage, and function as key signaling molecules. Mounting evidence points towards a tight interplay between lipids and circadian clocks. In mammals, circadian clocks regulate the daily physiology and metabolism, and disruption of circadian rhythmicity is associated with altered lipid homeostasis and pathologies such as fatty liver and obesity. Concomitantly, emerging evidence suggest that lipids are embedded within the core clock circuitry and participate in circadian control. Recent advances in lipidomics methodologies and their application in chronobiology studies have shed new light on the cross talk between circadian clocks and lipid homeostasis. We review herein the latest literature related to the involvement of lipids in circadian clock's function and highlight the contribution of circadian lipidomics studies to our understanding of circadian rhythmicity and lipid homeostasis. This article is part of a Special Issue entitled Brain Lipids.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/genetics , Fatty Liver/metabolism , Lipid Metabolism/genetics , Obesity/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression Regulation , Homeostasis , Humans , Lipids/chemistry , Obesity/genetics , Obesity/pathology , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction
16.
Nucleic Acids Res ; 42(19): 11879-90, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25260595

ABSTRACT

The circadian core clock circuitry relies on interlocked transcription-translation feedback loops that largely count on multiple protein interactions. The molecular mechanisms implicated in the assembly of these protein complexes are relatively unknown. Our bioinformatics analysis of short linear motifs, implicated in protein interactions, reveals an enrichment of the Pro-X-Asp-Leu-Ser (PXDLS) motif within circadian transcripts. We show that the PXDLS motif can bind to BMAL1/CLOCK and disrupt circadian oscillations in a cell-autonomous manner. Remarkably, the motif is evolutionary conserved in the core clock protein REV-ERBα, and additional proteins implicated in the clock's function (NRIP1, CBP). In this conjuncture, we uncover a novel cross talk between the two principal core clock feedback loops and show that BMAL/CLOCK and REV-ERBα interact and that the PXDLS motif of REV-ERBα participates in their binding. Furthermore, we demonstrate that the PXDLS motifs of NRIP1 and CBP are involved in circadian rhythmicity. Our findings suggest that the PXDLS motif plays an important role in circadian rhythmicity through regulation of protein interactions within the clock circuitry and that short linear motifs can be employed to modulate circadian oscillations.


Subject(s)
ARNTL Transcription Factors/metabolism , CLOCK Proteins/metabolism , Circadian Rhythm , Nuclear Receptor Subfamily 1, Group D, Member 1/chemistry , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding Sites , CREB-Binding Protein/chemistry , CREB-Binding Protein/metabolism , Circadian Rhythm/genetics , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Protein Interaction Domains and Motifs , Transcription, Genetic
18.
Hepatology ; 67(1): 441-443, 2018 01.
Article in English | MEDLINE | ID: mdl-28873232

Subject(s)
Liver , Humans
19.
Nat Rev Gastroenterol Hepatol ; 20(11): 695-707, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37291279

ABSTRACT

A wide variety of liver functions are regulated daily by the liver circadian clock and via systemic circadian control by other organs and cells within the gastrointestinal tract as well as the microbiome and immune cells. Disruption of the circadian system, as occurs during jetlag, shift work or an unhealthy lifestyle, is implicated in several liver-related pathologies, ranging from metabolic diseases such as obesity, type 2 diabetes mellitus and nonalcoholic fatty liver disease to liver malignancies such as hepatocellular carcinoma. In this Review, we cover the molecular, cellular and organismal aspects of various liver pathologies from a circadian viewpoint, and in particular how circadian dysregulation has a role in the development and progression of these diseases. Finally, we discuss therapeutic and lifestyle interventions that carry health benefits through support of a functional circadian clock that acts in synchrony with the environment.


Subject(s)
Circadian Clocks , Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Humans , Circadian Rhythm/physiology , Diabetes Mellitus, Type 2/metabolism , Liver/metabolism , Circadian Clocks/physiology , Non-alcoholic Fatty Liver Disease/metabolism
20.
Acta Physiol (Oxf) ; 234(2): e13770, 2022 02.
Article in English | MEDLINE | ID: mdl-34984824

ABSTRACT

In mammals, physiology and metabolism are shaped both by immediate and anticipatory responses to environmental changes through the myriad of molecular mechanisms. Whilst the former is mostly mediated through different acute signalling pathways the latter is primarily orchestrated by the circadian clock. Oxygen is vital for life and as such mammals have evolved different mechanisms to cope with changes in oxygen levels. It is widely accepted that oxygen sensing through the HIF-1 signalling pathway is paramount for the acute response to changes in oxygen levels. Circadian clocks are molecular oscillators that control 24 hours rhythms in various aspects of physiology and behaviour. Evidence emerging in recent years points towards pervasive molecular and functional interactions between these two pathways on multiple levels. Daily oscillations in oxygen levels are circadian clock-controlled and can reset the clock through HIF-1. Furthermore, the circadian clock appears to modulate the hypoxic response. We review herein the literature related to the crosstalk between the circadian clockwork and the oxygen-signalling pathway in mammals at the molecular and physiological level both under normal and pathologic conditions.


Subject(s)
Circadian Clocks , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Mammals/physiology , Oxygen , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL