Fact-checked by Grok 2 weeks ago

CLOCK

CLOCK (Circadian Locomotor Output Cycles Kaput) is a gene encoding a basic helix-loop-helix-PAS (bHLH-PAS) transcription factor that serves as a core component of the molecular circadian clock in mammals. First identified in mice through positional cloning in 1997, the CLOCK protein forms a heterodimer with BMAL1 to bind E-box DNA elements and rhythmically activate transcription of clock-controlled genes, including Per and Cry, thereby driving ~24-hour oscillations in gene expression. This positive limb of the circadian feedback loop is counteracted by PER and CRY repressors, establishing the transcriptional architecture that synchronizes physiological and behavioral rhythms across tissues. The CLOCK gene spans approximately 100 kb with 24 exons in mice and 28 in humans, producing a protein of about 846–855 amino acids featuring bHLH and PAS domains for dimerization and DNA binding, as well as intrinsic histone acetyltransferase activity that facilitates chromatin remodeling. Expressed ubiquitously but most prominently in the suprachiasmatic nucleus (SCN) of the hypothalamus—the master circadian pacemaker—CLOCK regulates not only core clock genes but also thousands of output genes involved in metabolism, immune function, and cell proliferation, with pervasive effects observed in tissues like the liver. Mutations in Clock, such as the dominant-negative allele in mice, result in lengthened circadian periods, disrupted sleep-wake cycles, increased body weight, and metabolic dysregulation, highlighting its role in linking circadian timing to health outcomes like obesity and diabetes. In humans, while no direct disease-causing mutations have been firmly established, polymorphisms in CLOCK are associated with variations in sleep timing, mood disorders, and cancer risk, underscoring its evolutionary conservation and broad physiological impact.

Molecular Biology

Gene Structure and Expression

The human CLOCK gene, officially known as clock circadian regulator, is located on the long arm of at the 4q12 cytogenetic band. It spans approximately 119 kb of genomic DNA, from positions 55,427,903 to 55,547,491 on the GRCh38 reference assembly, and comprises 28 exons. Originally cloned in through a search for homologs of the mouse Clock , it was identified as encoding a belonging to the basic helix-loop-helix PER-ARNT-SIM (bHLH-PAS) domain family, essential for circadian regulation. The gene's structure supports the production of multiple mRNA transcripts via , with Ensembl annotating 22 distinct splice variants, though the majority encode the canonical 846-amino-acid protein isoform (: O15516). The primary transcripts of CLOCK are approximately 8–10 in length, reflecting the inclusion of untranslated regions and variable exons, with the coding sequence encompassing about 8.3 . These transcripts are generated from a complex transcriptional architecture that includes upstream promoter elements and intronic enhancers, allowing for fine-tuned regulation of expression. Northern blot analyses from the initial cloning studies detected two major mRNA species of 8 and 10 , consistent with alternative or splicing events. While most variants produce the full-length protein, some may yield truncated or modified isoforms that influence stability or activity, contributing to the gene's versatility in circadian contexts. Expression of CLOCK mRNA is widespread across human tissues, reflecting its role in both central and peripheral circadian oscillators, with particularly elevated levels in the suprachiasmatic nucleus (SCN) of the hypothalamus—the master circadian pacemaker—as well as the cerebellum and peripheral organs such as the liver and heart. Quantitative expression data indicate moderate to high abundance in neural tissues (e.g., RPKM ~5–7 in brain regions) and lower but detectable levels in metabolic tissues like liver (RPKM ~2–4), underscoring its ubiquitous yet tissue-specific profile. In the SCN and peripheral clocks, CLOCK transcripts exhibit circadian oscillations, peaking during the subjective day to drive rhythmic gene expression. The promoter region of the CLOCK gene, spanning upstream of exon 1, contains key regulatory elements that control both basal and oscillatory transcription. Notably, it includes ROR/REV-ERB response elements (ROREs), which serve as binding sites for the nuclear receptors and REV-ERBα/β, enabling an auxiliary feedback loop that modulates CLOCK expression and . These elements facilitate repression by REV-ERBs during the night phase and activation by during the day, integrating CLOCK into the broader circadian transcriptional network without disrupting core loop dynamics. Additional motifs, such as E-boxes and , may contribute to light- and cAMP-responsive regulation, though their precise roles in basal transcription remain under investigation.

Protein Characteristics and Interactions

The CLOCK protein, encoded by the CLOCK gene, is a transcription factor with a calculated molecular weight of approximately 98 kDa in humans. It features a basic helix-loop-helix (bHLH) domain at the N-terminus, which facilitates DNA binding, and two Per-ARNT-SIM (PAS) domains (PAS-A and PAS-B) that mediate protein dimerization and environmental sensing. The bHLH domain enables recognition of specific DNA sequences, while the PAS domains support structural integrity and partner interactions essential for its function. Post-translational modifications significantly influence CLOCK's activity and localization. of CLOCK occurs primarily through casein kinase 1 epsilon (CK1ε) and delta (CK1δ), often scaffolded by proteins, which modulates its stability and nuclear accumulation. Additionally, SUMOylation of CLOCK, involving conjugation of small ubiquitin-like modifier proteins, enhances its transcriptional activity and contributes to protein stability within the circadian system. These modifications, including a nuclear localization facilitated by heterodimerization rather than a classical nuclear localization signal (NLS), allow CLOCK to function as a . CLOCK's primary molecular interaction is heterodimerization with BMAL1, forming the CLOCK:BMAL1 complex through interfaces involving the bHLH and both PAS domains of each protein. This complex binds to enhancer box (E-box) elements in target promoters, recognizing the CACGTG to initiate transcription. The dimerization is crucial for CLOCK's nuclear translocation and overall transcriptional potency.

Role in Circadian Rhythms

Core Feedback Loop Mechanism

The core transcriptional-translational feedback loop (TTFL) in mammalian circadian rhythms is driven by the CLOCK:BMAL1 heterodimer, which functions as a positive regulator. During the activation phase, CLOCK and BMAL1 form a heterodimeric complex that binds to enhancer box () elements (CANNTG sequences) in the promoters of the Per () and Cry () genes, thereby initiating their transcription. This transcriptional activation occurs primarily in the early subjective day, leading to the accumulation of PER and CRY mRNAs in the . As PER and CRY proteins are translated, they form heterocomplexes, often with additional regulators such as (CK1), which phosphorylate PER to facilitate nuclear translocation. These PER:CRY complexes then enter the during the late subjective day, where they directly interact with and inhibit the DNA-binding activity of the CLOCK:BMAL1 heterodimer.81014-4) This repression prevents further transcription of Per and Cry genes, closing the arm of the loop and establishing rhythmic with a period close to 24 hours. The approximately 24-hour periodicity of the loop arises from the timed accumulation, nuclear entry, and subsequent degradation of PER and CRY proteins. Phosphorylation of these repressors by kinases like CK1δ/ε marks them for ubiquitination and proteasomal degradation, primarily mediated by the FBXL3 targeting CRY1 and CRY2.00149-9) This degradation phase, occurring in the early subjective night, reduces PER:CRY levels, relieving inhibition on CLOCK:BMAL1 and allowing the cycle to restart. CRY1 and CRY2 contribute to this timing by modulating their own ubiquitination rates, ensuring precise oscillatory dynamics.00149-9) A simplified mathematical representation of the oscillator highlights the negative feedback and inherent delays. Consider a basic model where the concentration of the CLOCK:BMAL1 activator complex ([C:B]) drives synthesis of PER:CRY repressors ([P:C]), which in turn inhibit [C:B]: \frac{d[C:B]}{dt} = k_s - k_d [P:C] [C:B] \frac{d[P:C]}{dt} = k_t [C:B] - k_{deg} [P:C] Here, k_s is the synthesis rate of [C:B], k_d is the degradation rate modulated by [P:C], k_t is the transcription/translation rate of [P:C], and k_{deg} incorporates ubiquitination-dependent degradation; delays in nuclear translocation and protein stability confer the ~24-hour period. This TTFL operates in a tissue-autonomous manner, with the suprachiasmatic nucleus (SCN) of the hypothalamus serving as the central master clock that coordinates peripheral oscillators through neural and humoral signals. However, individual peripheral cells, such as those in the liver or heart, maintain independent yet similar loops, allowing local entrainment to cues like feeding while remaining synchronized to the SCN. The amplitude of this primary loop is modulated by environmental light, which entrains the SCN clock and indirectly influences the secondary regulatory elements acting on the Bmal1 promoter. Specifically, light-induced phase shifts in the SCN alter the expression of and REV-ERB receptors, which competitively bind ROR response elements (ROREs) to rhythmically regulate Bmal1 transcription and thereby fine-tune the overall oscillatory strength.00982-X)00194-9)

Integration with Other Regulatory Loops

The CLOCK:BMAL1 heterodimer not only drives the primary transcriptional-translational feedback loop but also activates a secondary loop by inducing the expression of nuclear receptors REV-ERBα and REV-ERBβ, as well as ROR family members, which in turn regulate Bmal1 transcription through binding to ROR response elements (ROREs) in its promoter. REV-ERBα/β primarily repress Bmal1 expression, while RORs activate it, creating an interlocking feedback mechanism that stabilizes the circadian period and fine-tunes the amplitude of oscillations in clock gene expression. This secondary loop ensures robust timing by counterbalancing the primary repressors PER and CRY, with rhythmic REV-ERBα/β levels peaking in antiphase to Bmal1. Beyond transcriptional regulation, the CLOCK-driven clock interconnects with metabolic pathways, notably through the NAD+-dependent deacetylase SIRT1, which deacetylates to modulate its transcriptional activity and links circadian timing to cellular energy states via fluctuating NAD+ levels. SIRT1's rhythmic HDAC activity, peaking during the subjective day, reduces BMAL1 acetylation on Lys9 and core clock proteins, thereby influencing the degradation of PER2 and overall clock amplitude in response to metabolic cues like nutrient availability. This integration allows the circadian system to adapt to daily feeding-fasting cycles, with NAD+ itself exhibiting circadian oscillations under CLOCK:BMAL1 control. Light entrainment of peripheral clocks, which rely on CLOCK for their molecular timing, occurs indirectly through the (SCN), where (VIP) signaling from SCN neurons synchronizes subordinate oscillators in tissues like the liver and heart. VIP release from the SCN's core region, rhythmically modulated by light inputs, activates VPAC2 receptors on peripheral cells, inducing Per1 and Per2 expression and thereby aligning local CLOCK:BMAL1-driven rhythms without direct photoreception in those tissues. This SCN-peripheral coupling maintains organism-wide coherence, with disruptions in VIP signaling desynchronizing clocks and altering CLOCK-dependent outputs. Recent studies highlight the integration of the CLOCK network with immune pathways, particularly the transcription factor, which CLOCK modulates to impose circadian gating on inflammatory responses. For instance, CLOCK directly interacts with the p65 subunit to enhance its activity during specific circadian phases, linking core clock timing to rhythmic production and immune function. Post-2020 research has further elucidated this cross-talk, showing that circadian disruption amplifies -driven inflammation in macrophages, underscoring the clock's role in immune . These integrations contribute to the broad scope of circadian control, with approximately 20% of the exhibiting rhythmic expression under CLOCK:BMAL1 influence across tissues, enabling coordinated physiological responses. Additionally, cross-talk with signaling reinforces this network, as CLOCK represses activity in a time-of-day-dependent manner, while adrenal clock outputs rhythmically modulate release to entrain peripheral clocks. This bidirectional interaction ensures alignment between stress responses and daily metabolic demands.

Discovery and Historical Development

Initial Identification in Mammals

The initial identification of the mammalian gene stemmed from a forward genetic screen in mice aimed at uncovering mutants with disrupted circadian behaviors. In , researchers generated ENU-mutagenized mice and identified a semidominant , named Clock, that lengthened the circadian period and eventually led to in homozygous individuals, indicating a key role in locomotor output cycles. This was mapped to mouse , setting the stage for cloning efforts. Positional cloning of the Clock gene was achieved in 1997 through screening a suprachiasmatic nucleus (SCN) using degenerate primers targeting conserved basic helix-loop-helix (bHLH) domains, revealing a large transcription unit spanning approximately 100 kb with 24 exons. The gene was named "Circadian Locomotor Output Cycles Kaput" (CLOCK) due to the arrhythmic in mutants, evoking the German word "kaput" for broken, and immediate highlighted its to bHLH-PAS domain-containing proteins such as single-minded, with later studies confirming its to the Clock gene. confirmed high expression of Clock mRNA in the , the master circadian pacemaker, with lower levels in other regions and peripheral tissues. Early functional characterization in 1997-1998 established CLOCK as a that heterodimerizes with BMAL1 to bind elements (CACGTG) in promoter regions, driving rhythmic essential for the circadian mechanism. Transfection assays demonstrated that the CLOCK-BMAL1 complex activates transcription from Per1 promoters via these es, providing initial evidence of its role in the positive limb of the core feedback loop. This work laid the foundation for understanding CLOCK's conserved function across species, linking mammalian and circadian systems.

Milestone Studies and Techniques

In the 2000s, followed by sequencing (ChIP-seq) emerged as a pivotal technique for mapping CLOCK binding sites across the genome, revealing thousands of motifs as primary targets in tissues like the mouse liver. A landmark 2014 study using ChIP-seq identified 7,978 CLOCK-binding sites, demonstrating polyphonic regulation of circadian outputs through direct transcriptional control of diverse gene networks. Concurrently, (RNAi) knockdown experiments confirmed CLOCK's essential role in maintaining circadian oscillations; for instance, RNAi-mediated silencing in and mammalian cells disrupted rhythmicity, underscoring its non-redundant function in the core clock mechanism. The saw advancements in and neural manipulation techniques that refined CLOCK's study in complex biological contexts. CRISPR-Cas9 enabled precise of CLOCK in lines such as U2OS, allowing researchers to dissect its cell-autonomous contributions to circadian without systemic confounds; a 2015 study demonstrated efficient editing in clock gene loci, facilitating rhythm assays in cellular models. further illuminated CLOCK's integration with light signaling, where activation of (SCN) neurons reset molecular phases, mimicking photic and highlighting firing rate's influence on CLOCK-BMAL1 activity. From 2020 to 2025, single-cell RNA sequencing (scRNA-seq) uncovered heterogeneity in CLOCK expression within the SCN, revealing subpopulations of neurons with phase-specific variations that contribute to network-level synchronization. A 2021 scRNA-seq analysis of SCN slices identified 11 neuronal clusters with distinct circadian profiles, including differential CLOCK dynamics across day-night cycles. Bioluminescent reporters, such as PER2::LUC fusions, enabled real-time, non-invasive monitoring of CLOCK-driven rhythms , capturing dynamic shifts in peripheral tissues over extended periods. Human studies reached a milestone in 2025 with from Takahashi's at UT Southwestern, which linked CLOCK expression to enhanced neocortical connectivity and cognitive networks via and genetic analyses, suggesting its role beyond rhythms in evolution. The 2023 in or , awarded for discoveries of molecular circadian mechanisms, indirectly catalyzed intensified CLOCK by validating the field's foundational insights.

Evolutionary Perspectives

Phylogenetic Conservation

The CLOCK gene demonstrates profound phylogenetic conservation across metazoans, with orthologs present in early-diverging lineages such as cnidarians. In the Nematostella vectensis, the ortholog NvCLK retains the core basic helix-loop-helix (bHLH) and two Per-Arnt-Sim () domains, which are crucial for protein dimerization and circadian transcriptional activation. This structural preservation underscores the ancient origins of CLOCK-like genes in animal evolution, predating the bilaterian-cnidarian split estimated around 600–700 million years ago. Molecular clock analyses and fossil-calibrated phylogenies support the foundational circadian machinery, including orthologs, evolving in the common ancestor of extant to synchronize physiological processes with environmental cycles. In vertebrates, the CLOCK gene underwent duplication events, notably during the two rounds of whole-genome duplication (2R) in early evolution, giving rise to paralogs such as neuronal PAS domain protein 2 (NPAS2) in mammals. These duplications expanded the circadian regulatory network, allowing for tissue-specific expression and functional specialization while maintaining core clock functions. Sequence is particularly evident in key functional , with the bHLH showing approximately 60% identity between the ortholog (dCLOCK) and mammalian CLOCK (mCLOCK), and the PAS exhibiting up to 79% identity. However, invertebrate versions like dCLOCK lack certain C-terminal extensions in the PAS-B that are present in mammalian counterparts, contributing to subtle differences in protein interactions and potential. This high degree of across ~600 million years of —from flies to humans—highlights the essential role of CLOCK in maintaining circadian rhythms.

Allelic Variants Across Species

The Clock^19 allele in mice represents a prominent natural variant characterized by a deletion of exon 19 (Δexon 19), resulting in the loss of a 51-amino-acid essential for activity, thereby acting as a dominant-negative loss-of-function that lengthens circadian period and leads to in homozygotes. In humans, the (SNP) rs1801260 (3111T/C) in the 3' (3'UTR) of the CLOCK gene is a common variant that influences mRNA stability and has been associated with subtle shifts in circadian timing. Across species, allelic variants in CLOCK homologs exhibit distributions that modulate circadian properties without disrupting core functionality. In Drosophila melanogaster, natural polymorphisms in the Clock gene, such as single nucleotide variants in regulatory regions, interact with alleles of the gene to fine-tune free-running length and responses, contributing to adaptive variation in locomotor activity rhythms under varying environmental conditions. Similarly, in (Danio rerio), clock1a mutations affect circadian rhythms and early development. Population genetics of CLOCK variants reveal species-specific frequencies and neutral effects that preserve circadian robustness. For instance, the C allele of human rs1801260 is associated with a preference for evening chronotypes and occurs at minor allele frequencies of approximately 30–40% in European-descent populations. These variants across species, including those in mice, flies, and fish, appear to evolve under weak purifying selection, enabling neutral drift while maintaining the resilience of the circadian oscillator against perturbations. Genome-wide association studies (GWAS) have illuminated the role of CLOCK-related variants in determination. Prior analyses have identified loci associated with morning chronotypes implicating clock genes, underscoring how these polymorphisms contribute to inter-individual differences in diurnal preference while upholding loop integrity.

Influence on Mammalian Physiology

The post-Cretaceous radiation of mammals approximately 66 million years ago marked a pivotal period in the evolution of circadian systems, enabling enhanced metabolic regulation essential for the development of endothermy in early eutherians. During this diversification following the of non-avian dinosaurs, mammalian ancestors transitioned from nocturnal lifestyles to more varied activity patterns, with circadian mechanisms coordinating energy expenditure and to maintain stable body temperatures independent of environmental fluctuations. This evolutionary adaptation allowed mammals to exploit diverse ecological niches, integrating circadian timing with metabolic pathways such as glucose and , thereby supporting sustained activity levels characteristic of endothermy. In mammalian lineages, the emergence of the CLOCK paralog NPAS2 facilitated adaptations in activity patterns, particularly nocturnal-to-diurnal shifts, by compensating for CLOCK function in the to promote behavioral flexibility. NPAS2, highly expressed in regions outside the (SCN), forms heterodimers with BMAL1 to drive circadian transcription, sustaining locomotor rhythms and sleep-wake adaptability in the absence of CLOCK. This paralogous compensation is evident in Clock-deficient mice, where NPAS2 maintains -dependent behaviors, enabling mammals to adjust activity to varying regimes and environmental pressures that ancestral reptiles could not. Such -specific roles underscore how CLOCK-NPAS2 duality evolved to enhance neural in circadian behavioral control across mammalian . Mammalian CLOCK also contributes to physiological adaptations in seasonal breeders, where its expression is upregulated in key tissues to orchestrate cycles during . In hibernators like ground squirrels, hypothalamic CLOCK maintains oscillatory patterns throughout breeding and phases, linking circadian timing to by modulating arousal from deep states. This supports the precise timing of metabolic suppression and reactivation, allowing seasonal breeders to synchronize reproductive cycles with resource availability while minimizing energy loss in prolonged bouts. Certain mammalian variants of CLOCK enhance in SCN-peripheral clock , aiding to disruptions like social jetlag. Polymorphisms in the CLOCK gene, such as the T3111C variant, influence and thereby mitigate the misalignment between social schedules and endogenous rhythms, improving between the central SCN pacemaker and peripheral oscillators in tissues like the liver and muscles. This promotes resilient , reducing desynchrony in modern environments with irregular light and feeding cues. Recent genomic studies (as of 2025) highlight evolutionary gains in clock function that bolster neural in mammals.

Genetic Variants and Experimental Models

Mutations in Drosophila

The foundational mutations in the Drosophila melanogaster Clock (Clk) gene were isolated through ethyl methanesulfonate (EMS) mutagenesis screens targeting circadian rhythms in eclosion and locomotor activity. In 1971, Ronald Konopka and identified three alleles at the Clk locus on the : an arrhythmic (Clk^{Ar}), a short-period allele (Clk^{s}) with a ~19-hour period, and a long-period allele (Clk^{l}) with a ~28-hour period. The Clk^{Ar} mutation, a recessive , abolishes 24-hour rhythms in both adult eclosion from pupal cases and locomotor behavior under constant darkness conditions, resulting in random, non-periodic activity patterns that confirm its role as a core clock component. Subsequent molecular cloning in 1998 revealed that Clk encodes a bHLH-PAS , sharing ~50% sequence identity with mammalian CLOCK, particularly in the basic helix-loop-helix (bHLH) and PAS domains essential for heterodimerization and DNA binding. Hypomorphic alleles like Clk^{s} and Clk^{l} produce partial loss-of-function phenotypes, shortening or lengthening free-running s in locomotor rhythms while maintaining overall rhythmicity, whereas stronger alleles such as Clk^{jrk} (jerking), a dominant introducing a premature , cause ~50% arrhythmicity in heterozygotes and near-complete loss of rhythms in homozygotes, alongside disrupted transcription of downstream clock genes period (per) and timeless (tim). These phenotypes extend to specific neural circuits, including mushroom body neurons where Clk mutations dampen rhythmic gene expression and impair circadian modulation of learning and processes. Rescue experiments using transgenic expression of wild-type Clk have confirmed the gene's sufficiency for rhythm restoration. For instance, GAL4/UAS-driven Clk transgenes fully the arrhythmic eclosion and locomotor phenotypes in Clk^{Ar} mutants, restoring ~24-hour periodicity and amplitude in behavioral outputs. The Drosophila Clk gene, with ~50% sequence similarity to mammalian CLOCK, parallels the core feedback loop by activating per and tim transcription via heterodimerization with (CYC), underscoring conserved mechanisms across species. Targeted disruptions of Clk have been achieved using P-element insertions, enabling precise knockouts and spatial-temporal control of gene function in circadian studies. These insertional mutagenesis techniques, which disrupt Clk expression at specific loci, recapitulate null phenotypes like those of Clk^{Ar} and facilitate dissection of clock neuron contributions to rhythmicity.

Mutations in Mice

The Clock mutation in mice, denoted as Clock^Δ19, was generated in the early 1990s through N-ethyl-N-nitrosourea (ENU) mutagenesis screening of progeny for defects in circadian behavior. This mutation arises from an A-to-T transversion in the splice donor site between exons 18 and 19 of the Clock gene, resulting in the skipping of exon 19 during mRNA processing and production of a hypomorphic CLOCK protein. The mutant protein lacks a 51-amino-acid transactivation domain essential for full transcriptional activity but retains the ability to dimerize with BMAL1, thereby exerting dominant-negative effects on circadian oscillator function. Homozygous Clock^Δ19/Δ19 mice are viable and fertile, though they exhibit mildly reduced male fecundity and irregular estrous cycles in certain genetic backgrounds, leading to smaller litter sizes. Behaviorally, homozygous mutants initially display a lengthened free-running circadian of approximately 28 hours under constant darkness, which progressively deteriorates into over weeks, accompanied by diminished of locomotor activity rhythms. These mice also show impaired to light-dark cycles, with slower re-adaptation after phase shifts, though heterozygotes maintain relatively normal rhythms with subtle lengthening to about 24.3 hours. These phenotypes parallel basic rhythmicity disruptions observed in period mutants but extend to mammalian-specific impacts on activity consolidation. Metabolically, Clock^Δ19/Δ19 mutants develop , particularly when challenged with a high-fat , due to desynchronized peripheral clocks that impair nutrient sensing and . They exhibit across the light-dark without compensatory insulin elevation, alongside impaired glucose tolerance that worsens with age and varies by sex—manifesting as in young males and in older females. These disruptions highlight CLOCK's role in coordinating metabolic oscillations in tissues like liver and adipose, independent of central clock output. Recent advancements in 2025 have demonstrated that targeted enhancement of CLOCK function in aging models can extend lifespan by modulating hallmark aging pathways, such as and , offering insights into therapeutic interventions for age-related decline.

Human Genetic Polymorphisms

The CLOCK gene in humans is characterized by several common single nucleotide polymorphisms (SNPs) that contribute to natural variation in circadian timing and without causing . The most extensively studied variant is rs1801260 (also denoted as 3111T/C in the 3' ), where the C is linked to a preference for evening and delayed syndrome in studies. This SNP influences mRNA stability through altered binding, leading to increased CLOCK expression levels. Carriers of the C exhibit reduced and later onset compared to TT homozygotes, contributing to behavioral variability in daily rhythms. Other notable variants include rs3749474, located in the promoter region, which modulates and has been associated with preferences in . Rare loss-of-function variants in CLOCK have been identified in efforts, but these are infrequent and typically do not result in complete gene inactivation. No complete knockouts of CLOCK have been reported in populations, consistent with its essential role in circadian regulation. A 2019 (GWAS) identified 351 loci associated with , including CLOCK, highlighting its role among multiple genetic contributors to circadian variability. Population-level differences in for rs1801260 underscore geographic variation in circadian traits. The C frequency is higher in populations of East Asian descent (approximately 49%) compared to those of descent (approximately 26%), potentially influencing regional patterns in timing and distribution. Functional studies show that the C leads to higher CLOCK mRNA levels due to increased stability, affecting the amplitude of circadian oscillations. These polymorphisms collectively explain a portion of the heritable component of non-pathological circadian phenotypes, such as self-reported morningness-eveningness scores.

Physiological and Pathological Impacts

Normal Functions in Health

The CLOCK gene encodes a that forms a heterodimer with BMAL1, driving the rhythmic expression of core circadian genes such as PER and CRY within the (SCN), the brain's master circadian pacemaker. This molecular mechanism in the SCN synchronizes daily physiological processes, including the coordination of the sleep-wake cycle by generating neural signals that promote arousal during the active phase and rest during the inactive phase. Additionally, SCN CLOCK activity modulates hormone release, notably suppressing synthesis in the during daylight hours to align behavioral and endocrine rhythms with environmental light-dark cycles. In peripheral tissues, CLOCK contributes to autonomous circadian clocks that regulate metabolic , influencing and processing in organs such as the liver and to optimize energy utilization across the day. For instance, CLOCK-driven oscillations in hepatic facilitate timed and , preventing metabolic dysregulation during or feeding periods. CLOCK also times immune responses by controlling the rhythmic production of cytokines and leukocyte trafficking in lymphoid tissues, ensuring peak immune vigilance aligns with periods of potential exposure. Within the , CLOCK expression supports cognitive functions by enhancing during , where it promotes and the stabilization of neural engrams through interactions with downstream clock components. This process is vital for formation, as hippocampal CLOCK rhythms facilitate the replay of daily experiences during phases. Experimental evidence underscores CLOCK's role in and genomic stability; mice deficient in CLOCK exhibit a 15% reduction in average lifespan compared to wild-type controls, highlighting its contribution to healthy aging through sustained circadian coordination. Across mammalian tissues, approximately 43% of protein-coding genes display circadian expression patterns regulated by CLOCK and related factors, with daily fluctuations enabling adaptive physiological responses. Recent findings indicate that human-specific enhancements in neocortical CLOCK expression bolster interneuronal connectivity, fostering neural flexibility that supports resilience by improving adaptability to cognitive demands.

Associations with Diseases and Disorders

Dysregulation of the CLOCK gene has been implicated in various sleep disorders, particularly through polymorphisms that influence circadian timing. The CLOCK 3111T/C polymorphism (rs1801260), where the C allele is associated with evening chronotypes, has been linked to an increased risk of and delayed sleep phase (DSPS), with odds ratios around 1.5 in affected populations. Recent 2025 research highlights how CLOCK mutations contribute to mechanisms by disrupting neural excitability and sleep consolidation, exacerbating symptoms in chronic cases. In metabolic disorders, CLOCK variants disrupt feeding rhythms and , elevating risks for and (T2D). Specific haplotypes of the CLOCK gene, such as those involving rs1801260, are associated with higher T2D prevalence in non-obese populations, with studies showing up to a 1.5-fold increased risk through impaired insulin sensitivity. CLOCK disruptions also correlate with traits like adiposity indices in diverse cohorts, mediated by altered and regulation. Neurological conditions involving CLOCK include (AD) and (MDD). In AD, 2025 studies reveal that amyloid-beta pathology hijacks CLOCK-mediated rhythms in glial cells, desynchronizing in over 40 Alzheimer's-associated genes and accelerating plaque accumulation, as evidenced by disrupted circadian control in support cells. For MDD, epigenetic changes such as alterations in CLOCK and other core clock genes correlate with disease severity, with 2025 analyses showing interactions with environmental factors like that worsen symptoms via hypomethylation patterns. CLOCK suppression promotes progression by deregulating genes and reducing , with low CLOCK expression observed in tumor tissues correlating with advanced stages and poorer survival. Chronotherapy, timing treatments to CLOCK rhythms, enhances efficacy of colorectal cancer drugs like by up to 20% in preclinical models, leveraging circadian . Overall, while no Mendelian diseases directly stem from CLOCK mutations, polygenic risks involving CLOCK variants contribute to these multifactorial disorders across populations.

References

  1. [1]
    Positional cloning of the mouse circadian clock gene - PubMed - NIH
    We used positional cloning to identify the circadian Clock gene in mice. Clock is a large transcription unit with 24 exons spanning approximately 100,000 bp ...
  2. [2]
    Transcriptional architecture of the mammalian circadian clock - Nature
    Dec 19, 2016 · The core clock genes include CLOCK and BMAL1, which encode activators, and PER1, PER2, CRY1 and CRY2, which encode repressors. Current topics of ...
  3. [3]
    9575 - Gene ResultCLOCK clock circadian regulator [ (human)] - NCBI
    Sep 9, 2025 · Clock genes have been implicated in cancer-related functions; in this work, we investigated CLOCK as a possible target of somatic mutations in ...
  4. [4]
    Molecular cloning and characterization of the human CLOCK gene
    Here, we report the cloning, exon organization, chromosomal location, and mRNA expression of the human CLOCK gene. The coding sequence of human CLOCK ...
  5. [5]
  6. [6]
    Mutational scanning of the CLOCKΔ19 domain identifies amino ...
    Feb 28, 2022 · Chemical mutagenesis deleted exon 19 of the murine Clock gene by mutating an RNA splice site. The resulting protein from this mutated gene is ...
  7. [7]
    Redundant Function of REV-ERBα and β and Non-Essential Role ...
    Rorc and Cry1 are regulated not only by BMAL1/CLOCK but also by RORs/REV-ERBs via the RORE. Bmal1, Clock and E4bp4 are regulated by RORs/REV-ERBs via the RORE.<|control11|><|separator|>
  8. [8]
    REV-ERBα and REV-ERBβ function as key factors regulating ...
    Jul 15, 2019 · These data suggest that REV-ERBs are not necessary for the circadian core clock gene expression rhythms but play an important role in regulation ...
  9. [9]
    The global and promoter-centric 3D genome organization ...
    Jun 8, 2021 · Regulatory elements form dynamic and stable chromatin contacts with circadian gene promoters. In order to characterize the full range of ...
  10. [10]
    o15516 · clock_human - UniProt
    CLOCK is a transcriptional activator and core component of the circadian clock, which regulates physiological processes through 24-hour rhythms.
  11. [11]
    The evolution and structure/function of bHLH–PAS transcription ...
    Jun 13, 2022 · Most bHLH–PAS proteins contain a bHLH domain, two PAS domains, and a transrepression or transactivation domain (Figure 1B). Some bHLH–PAS ...
  12. [12]
    bHLH–PAS Proteins: Their Structure and Intrinsic Disorder - MDPI
    Jul 26, 2019 · Another remarkable class I bHLH–PAS protein is CLOCK, comprising a domain with histone transacetylase (HAT) activity in the C-terminus. This ...
  13. [13]
    Decoupling PER phosphorylation, stability and rhythmic expression ...
    Jul 9, 2022 · In this study, we demonstrate that PER act as the CK1 scaffold to promote hyperphosphorylation of CLOCK protein, which closes the circadian ...
  14. [14]
    Sumoylation controls CLOCK-BMAL1-mediated clock resetting via ...
    Besides phosphorylation, the activity of CLOCK/BMAL1 is also regulated through sumoylation by the small ubiquitin-related modifier (SUMO) family of proteins.
  15. [15]
    CLOCK phosphorylation by AKT regulates its nuclear accumulation ...
    Expression of CLOCK in HEK293T cells, followed by quantitative imaging, demonstrated that ∼25% of WT CLOCK is localized to the nucleus as compared with 50% of ...
  16. [16]
    Crystal Structure of the Heterodimeric CLOCK:BMAL1 ...
    May 31, 2012 · While all three domains of the CLOCK and BMAL1 subunits are involved in intermolecular interactions, the heterodimeric interfaces between the ...
  17. [17]
    Phosphorylation of DNA-binding domains of CLOCK–BMAL1 ...
    May 28, 2024 · In mammals, CLOCK and BMAL1 proteins form a heterodimer that binds to E-box sequences and activates transcription of target genes, including ...
  18. [18]
    The REV-ERBs and RORs: molecular links between circadian ...
    REV-ERBs play a central role in regulating clock genes, as discussed earlier ... promoter sequences for the REV-ERB target genes BMAL1, G6Pase and PEPCK.
  19. [19]
    Role of circadian transcription factor REV-ERB in cardiovascular ...
    REV-ERB and retinoid-related orphan receptor (ROR) stabilize and enhance the feedback loop mediated by BMAL1. 3.2 Role of REV-ERB in autophagy, inflammation ...
  20. [20]
    The NAD+-Dependent Deacetylase SIRT1 Modulates CLOCK ...
    We show that the HDAC activity of the NAD+-dependent SIRT1 enzyme is regulated in a circadian manner, correlating with rhythmic acetylation of BMAL1 and H3 Lys9 ...
  21. [21]
    SIRT1 Regulates Circadian Clock Gene Expression through PER2 ...
    Jul 25, 2008 · SIRT1 binds CLOCK-BMAL1 in a circadian manner and promotes the deacetylation and degradation of PER2. Given the NAD+ dependence of SIRT1 ...
  22. [22]
    The Suprachiasmatic Nucleus (SCN) from Genes to Neurons to ...
    Nov 12, 2014 · Functionally, VIP signaling has a well-defined role in synchronizing and phase-shifting rhythms within the SCN. In vitro, a single pulse of VIP ...
  23. [23]
    Roles of Neuropeptides, VIP and AVP, in the Mammalian Central ...
    Apr 14, 2021 · These neuropeptides function as synchronizers and are important for entrainment to environmental light and for determining the circadian period.
  24. [24]
    Entrainment of circadian rhythms depends on firing rates and ...
    They found that increases in VIP neuronal firing frequency entrain circadian rhythms through increased VIP signaling.
  25. [25]
    NF-κB modifies the mammalian circadian clock through interaction ...
    NF-κB modifies the mammalian circadian clock through interaction with the core clock protein BMAL1.
  26. [26]
    Circadian rhythm regulates the function of immune cells ... - Nature
    Apr 27, 2024 · This paper aims to elucidate the regulatory effects of BMAL1, clock and other clock genes on immune cells, and reveal the molecular mechanism of circadian ...
  27. [27]
    The clockwork macrophage: timing in innate immunity - ScienceDirect
    May 21, 2025 · NF-κB modifies the mammalian circadian clock through interaction with the core clock protein BMAL1. PLoS Genet, 17 (11) (2021), Article ...
  28. [28]
    Molecular regulations of circadian rhythm and implications for ...
    Feb 8, 2022 · In this review, we summarize findings showing the crosstalk between the circadian molecular clock and some key intracellular pathways.Circadian Regulation Of... · Circadian Disruption... · Targeting Circadian Rhythm...
  29. [29]
    Circadian Clock, Glucocorticoids and NF-κB Signaling in ...
    Oct 31, 2022 · In this review we discuss evidence for interactions between the circadian clock components, glucocorticoids and NF-κB signaling responses in the brain
  30. [30]
    The circadian clock and glucocorticoids – Interactions across many ...
    The circadian clock regulates endogenous daily rhythms in physiology. Glucocorticoid (GC) signaling is highly dynamic on time scales from seconds to years.
  31. [31]
    Mutagenesis and Mapping of a Mouse Gene, Clock, Essential for ...
    A semidominant mutation, Clock, that lengthens circadian period and abolishes persistence of rhythmicity was identified.
  32. [32]
    Role of the CLOCK Protein in the Mammalian Circadian Mechanism
    CLOCK and BMAL1 together produced a small increase in transcriptional activity from the distal E-box (2.1-fold), a moderate increase from the middle E-box (3.6- ...Missing: BMAL1 | Show results with:BMAL1
  33. [33]
    CLOCK-Controlled Polyphonic Regulation of Circadian Rhythms ...
    Here we identified 7,978 CLOCK-binding sites in mouse liver by chromatin immunoprecipitation-sequencing (ChIP-Seq), and a newly developed bioinformatics method, ...Missing: milestone | Show results with:milestone
  34. [34]
    Silencing the circadian clock gene Clock using RNAi reveals ...
    Our results reveal that Clk is essential for the circadian clock, but is not required for the circatidal clock. From these results we propose that the ...
  35. [35]
    Highly Efficient Genome Editing via CRISPR/Cas9 to Create Clock ...
    Thus, CRISPR/Cas9 genome editing should be highly valuable for studying circadian rhythms not only in human cells but also in classic model systems as well as ...Missing: organoids | Show results with:organoids
  36. [36]
    Manipulating circadian clock neuron firing rate resets molecular ...
    The application of optogenetics to the SCN has allowed us to test the fundamental role of firing rate in influencing molecular and behavioral circadian rhythms.
  37. [37]
    Single‐cell transcriptomics of suprachiasmatic nuclei reveal a ...
    Sep 6, 2021 · Single‐cell transcriptomics reveal eleven distinct neuronal subpopulations of the suprachiasmatic nucleus (SCN) circadian network.Scrnaseq Of Scn Neurons · Topology Of Scn... · Scn Explant Culture...
  38. [38]
    Machine learning models highlight environmental and genetic ...
    Aug 5, 2025 · At the core of the circadian clock is a set of genes that form a transcriptional network of interlocking feedback loops which generate ~24-h ...
  39. [39]
    PERIOD2::LUCIFERASE real-time reporting of circadian dynamics ...
    Using a PERIOD2::LUCIFERASE fusion protein as a real-time reporter of circadian ... For bioluminescence monitoring, animals were transferred to the University ...
  40. [40]
    Circadian gene may be a key to humans' unique cognitive abilities
    Jul 31, 2025 · CLOCK gene, discovered by UTSW researcher, influences brain structure and function in addition to regulating circadian rhythm ...Missing: lab | Show results with:lab
  41. [41]
    Human CLOCK enhances neocortical function - PubMed - NIH
    Jun 30, 2025 · The transcription factor CLOCK is ubiquitously expressed and important for circadian rhythms, while its human-specific expression in ...Missing: lab | Show results with:lab
  42. [42]
    CLOCK evolved in cnidaria to synchronize internal rhythms with diel ...
    The Clock gene has evolved in cnidarians to synchronize rhythmic physiology and behavior to the diel rhythm of the earth's biosphere.
  43. [43]
    Estimating metazoan divergence times with a molecular clock - PNAS
    We estimate that the last common ancestor of bilaterians arose somewhere between 573 and 656 Ma, depending on the value assigned to the parameter scaling ...Missing: timeline | Show results with:timeline
  44. [44]
    Clock genes and their genomic distributions in three species of ...
    In humans (Homo sapiens) and mice, Clock and NPAS2 are two known paralogs which are related by the 2R genome duplication event [23].
  45. [45]
  46. [46]
    The mouse Clock mutation reduces circadian pacemaker amplitude ...
    Jun 13, 2006 · The mouse Clock mutation reduces circadian pacemaker amplitude and enhances efficacy of resetting stimuli and phase–response curve amplitude.
  47. [47]
    The role of CLOCK gene in psychiatric disorders: Evidence from ...
    Sep 13, 2017 · The rs1801260 (3111T/C) variant has already been associated with evening preference in European and Japanese populations (Katzenberg et al ...
  48. [48]
    The genetic basis of diurnal preference in Drosophila melanogaster
    Aug 31, 2020 · Other than one circadian clock gene (pdp1), most differentially expressed genes were associated with either clock output (pdf, to) or input (Rh3 ...
  49. [49]
    Immunity, Infection, and the Zebrafish Clock - PMC - PubMed Central
    Aug 16, 2022 · There are now many immune-related genes known to be regulated by the molecular clock in zebrafish. A mutation in the zebrafish circadian gene ...The Zebrafish Clock · The Core Clock Loop · Fig 2Missing: alleles | Show results with:alleles
  50. [50]
    Screening of Clock Gene Polymorphisms Demonstrates Association ...
    Sep 9, 2014 · The 3111T/C polymorphism of hClock is associated with evening preference and delayed sleep timing in a Japanese population sample. Am J Med ...
  51. [51]
    Functional and evolutionary implications of natural variation in clock ...
    The frq gene in Neurospora, like tim in Drosophila, has alternative start codons, but unlike in the fly, this is not a polymorphism. The two forms of FRQ differ ...
  52. [52]
    Morning clock gene expression in young adults of early and late ...
    Jul 23, 2025 · On a gene expression level, this circadian clock is mainly driven by the circadian expression of the transcription factors Clock and BmalI, ...
  53. [53]
    Circadian Metabolism in the Light of Evolution - Oxford Academic
    This review scrutinizes the role of the body's internal clocks in the hard-wiring of circadian networks that have evolved to achieve energetic balance and ...
  54. [54]
    Hypothalamus but not liver retains daily expression of clock genes ...
    Feb 12, 2020 · Our results showed that body and testes weights decreased during hibernation. Further, the hypothalamus retained daily clock gene oscillations ...
  55. [55]
    Clock Mutants of Drosophila melanogaster - PNAS
    Three mutants have been isolated in which the normal 24-hour rhythm is drastically changed. One mutant is arrhythmic; another has a period of 19 hr; a third ...
  56. [56]
    A recessive mutant of Drosophila Clock reveals a role in circadian ...
    A recessive mutant of Drosophila Clock reveals a role in circadian rhythm amplitude ... (D) Expression of CLOCKar in transfected cells. To determine the ...
  57. [57]
    Molecular genetics of the fruit-fly circadian clock - Nature
    May 24, 2006 · Konopka RJ, Benzer S : Clock mutants of Drosophila melanogaster. ... mutant, arrhythmic Drosophila melanogaster. Cell 1984; 39: 369–376 ...
  58. [58]
    Genetics of Circadian Rhythms in Mammalian Model Organisms
    Additional genetic approaches revealed that Clock is an antimorph—a specific type of dominant negative mutation (King et al., 1997a). Identification of the ...
  59. [59]
    Disrupting the circadian clock: Gene-specific effects on aging ...
    CLOCK-deficient mice also appear to differ, phenotypically, from ClockΔ19/Δ19 mutant mice, which have an intronic splice site point mutation in the Clock gene ...Missing: Δexon | Show results with:Δexon
  60. [60]
    Developmental and reproductive performance in circadian mutant ...
    Sexually mature adults of both mutant lines showed mildly reduced male in vivo (but not in vitro) fertility and irregular estrous cycles exacerbated by ...Missing: sterility | Show results with:sterility
  61. [61]
    Female ClockΔ19/Δ19 mice are protected from the development of ...
    The Clock mutation leads to a significant increase in wheel running period in homozygote ClockΔ19/Δ19 mice (27.10 h ± 0.07) compared with heterozygote (24.27 h ...
  62. [62]
    Inducible and Reversible Clock Gene Expression in Brain Using the ...
    The tTA system can manipulate circadian clock gene expression in a tissue-specific, conditional, and reversible manner in the central nervous system.
  63. [63]
    Circadian Rhythms and Metabolic Syndrome | Circulation Research
    Feb 19, 2010 · High-fat feeding studies revealed exaggerated weight gain of ClockΔ19 mutant mice, and dual-energy X-ray absorptiometry scanning and fat pad ...
  64. [64]
    Disruption of the Clock Components CLOCK and BMAL1 Leads to ...
    In ClockΔ19/Δ19 mutants, glucose levels were elevated across the entire light-dark (LD) cycle without a rise in insulin levels, whereas insulin rises in WT ...
  65. [65]
    NeuroVoices: Joseph S. Takahashi, PhD, on the Circadian Clock's ...
    Jul 16, 2025 · He noted his recent lab research showed that enhancing the circadian clock could extend lifespan in mice by regulating hallmark aging pathways.Missing: connectivity | Show results with:connectivity
  66. [66]
    Polymorphisms in MTNR1A (rs2119882) and CLOCK (rs1801260 ...
    The rs1801260 polymorphism in CLOCK, located in the 3' untranslated region (UTR), affects the binding of microRNAs and alters mRNA stability or translation ...Missing: assay | Show results with:assay
  67. [67]
    Genetic Differences in Human Circadian Clock Genes Among ...
    The frequency distribution of these clock gene polymorphisms is reported for diverse populations of African Americans, European Americans, Ghanaians, Han ...Missing: gnomAD 1000
  68. [68]
    Expression of clock genes in the suprachiasmatic nucleus - PubMed
    The suprachiasmatic nucleus (SCN) is the anatomical substrate for the principal circadian clock coordinating daily rhythms in a vast array of behavioral and ...
  69. [69]
    Circadian rhythm mechanism in the suprachiasmatic nucleus and its ...
    Mar 24, 2024 · The suprachiasmatic nucleus (SCN) is the center that controls all circadian rhythms, including wake–sleep, appetite, autonomic system, and neuroendocrine ...
  70. [70]
    Endocrine regulation of circadian rhythms - Nature
    Mar 8, 2025 · On the molecular level, clock genes and their protein products oscillate to generate these circadian rhythms.Introduction · Glucocorticoids · Sex Steroids
  71. [71]
    Physiological significance of a peripheral tissue circadian clock
    Sep 30, 2008 · The ability of the SCN to set the phases of peripheral tissue clocks thus appears to depend on its ability to drive daily rhythms of feeding ...
  72. [72]
    The metabolic significance of peripheral tissue clocks - Nature
    Mar 26, 2025 · However, restoration of Clock expression in the brain recovers behavioural rhythms and enhances rhythmic liver gene expression in these mice.
  73. [73]
  74. [74]
    The role of clock genes in sleep, stress and memory - ScienceDirect
    Clock genes serve as the basis of an intracellular timekeeping system, present throughout the body, which generates approximately 24-hour rhythms in physiology ...Review · 2.3. Role Of Clock Genes In... · Acknowledgements
  75. [75]
    Deficiency of circadian protein CLOCK reduces lifespan and ...
    Dec 9, 2010 · Deficiency of the CLOCK protein significantly affects longevity: the average lifespan of Clock −/− mice is reduced by 15% compared with wild type mice.Missing: allele Δexon
  76. [76]
    A circadian gene expression atlas in mammals - PNAS
    This study highlights critical, systemic, and surprising roles of the mammalian circadian clock and provides a blueprint for advancement in chronotherapy.
  77. [77]
    3111T/C Clock Gene Polymorphism in Women with Insomnia
    Aug 29, 2017 · The 3111T/C polymorphism of hClock is associated with evening preference and delayed sleep timing in a Japanese population sample. Am. J ...Missing: syndrome | Show results with:syndrome
  78. [78]
    Circadian clock genes and insomnia: molecular mechanisms and ...
    Oct 22, 2025 · The suprachiasmatic nucleus (SCN) regulates rhythmic output to peripheral clocks through hormonal and neural pathways, notably melatonin from ...
  79. [79]
    Variant of the clock circadian regulator (CLOCK) gene and related ...
    Variant of the CLOCK gene and related haplotypes are associated with the prevalence of T2D in the Japanese population, in which obesity is less common, ...
  80. [80]
    Association of CLOCK gene variants with obesity and adiposity ...
    May 26, 2022 · This study investigated whether the CLOCK variants rs4864548 and rs6843722 are associated with obesity and related traits in Pakistanis.
  81. [81]
  82. [82]
    DNA methylation of core clock genes in patients with major ...
    Our study aimed to investigate the interplay between air pollution exposure, DNA methylation of core clock genes involved in circadian rhythms, and MDD severity ...
  83. [83]
    The role of circadian clock genes in colorectal carcinoma
    Nov 15, 2023 · Chronotherapy. Chronotherapy, an adjuvant therapy based on the patient's circadian clock, has shown effectiveness in the treatment of CRC [67] ...
  84. [84]
    The genomic landscape and pharmacogenomic interactions of clock ...
    Overexpression of the circadian clock gene bmal1 increases sensitivity to oxaliplatin in colorectal cancer. Clin. Cancer Res. 2014;20:1042–1052. doi ...
  85. [85]
    Circadian disruption, clock genes, and metabolic health - JCI
    Jul 15, 2024 · CLOCK gene variation is associated with incidence of type-2 diabetes and cardiovascular diseases in type-2 diabetic subjects: dietary ...