Skip to main content
  • Review
  • Published:

The role of the circadian clock in rheumatoid arthritis

Abstract

Rheumatoid arthritis exhibits diurnal variation in symptoms, with patients suffering with increased painful joint stiffness in the early morning. This correlates with an early morning rise in circulating levels of pro-inflammatory cytokines, such as interleukin-6. This temporal variation in disease pathology is directed by the circadian clock, both at a systemic level, through signalling pathways derived in the central clock, and at a local level by autonomous clocks found within inflammatory organs and cells. Indeed, many cellular components of the immune system, which are involved in the pathogenesis of rheumatoid arthritis, possess independent clocks that facilitate temporal gating of their functions. Furthermore, the circadian clock regulates the expression and activity of several genes and proteins that have demonstrated roles in progression of this autoimmune disease. These include a number of nuclear receptors and also fat-derived adipokines. Employing the knowledge we have about how the inflammatory response is regulated by the clock will facilitate the development of chronotherapy regimens to improve the efficacy of current treatment strategies. Furthermore, a full understanding of the mechanisms by which the clock couples to the immune system may provide novel therapeutic targets for the treatment of this debilitating disease.

Introduction

Symptoms of rheumatoid arthritis (RA) frequently show diurnal variation, with exacerbations in the morning (Figure 1) [1]. This variation in disease expression is accompanied by daily oscillations in circulating concentrations of disease-mediating cytokines [2]. In particular, IL-6 shows robust oscillations, and fluctuations in serum IL-6 levels correlate with changes in disease symptoms [3]. This review summarises the evidence for a primary role for the circadian clock in the observed diurnal variations in disease activity, and its role in further aspects of RA disease manifestation. We consider how this information can be utilised, not only to modify existing treatment regimens, but to develop new therapeutic strategies to treat RA.

Figure 1
figure 1

Rheumatoid arthritis shows diurnal variation in disease symptoms and markers. Joint stiffness and pain are more pronounced in the early morning [1], and this correlates with the early morning rise in plasma IL-6 levels [3]. The circadian hormone melatonin (which is considered to exacerbate the inflammatory response) is released only during the night, and circulating levels peak in the mid-night. The anti-inflammatory glucocorticoid - cortisol - is also under circadian control, peaking in the early morning.

The circadian clock

The circadian clock drives daily rhythms in physiology necessary to synchronise the function of an organism with the 24-hour environment. Physiological functions under circadian control include the sleep-wake cycle, body temperature, heart rate, blood pressure, hormone regulation and immunity. These daily oscillations are orchestrated by a central pacemaker, which is found within the brain, in a hypothalamic region located above the optic chiasm called the suprachiasmatic nucleus (SCN). The SCN receives light input from the eyes via the retinohypothalamic tract. The central pacemaker synchronises additional peripheral oscillators found locally within organs, tissues and cells (reviewed in [4]). These secondary clocks are synchronised by the central clock, but are self-sustaining and can be entrained by external cues such as temperature and feeding schedules.

The molecular machinery required to enable a cell to oscillate comprises a transcription/translation feedback loop (Figure 2). Central to this loop are the genes clock and bmal, whose encoded proteins dimerise (CLOCK/BMAL) and bind to E-box elements on the promoters of the clock genes period (per), cryptochrome (cry), rev-erb and ror to activate their transcription. Translated PER and CRY form a dimeric complex (PER/CRY), enter the nucleus and inhibit CLOCK/BMAL transactivation. Subsequent degradation of PER/CRY allows CLOCK/BMAL to start a new cycle of transcriptional activation. A second feedback loop is formed by the action of ROR and REV-ERB proteins on bmal transcription; these nuclear receptors activate (ROR) and repress (REV-ERB) transcription through their competitive action on response elements (ROREs) on the bmal promoter. Initially thought of as an auxillary stabilising loop, it is now considered that this circuit is required for circadian oscillation [5]. In addition to these core clock genes, numerous other 'clock-controlled genes' show circadian patterns of expression as a result of action on E boxes, D box enhancers, and RORE sites.

Figure 2
figure 2

The molecular clock. CLOCK and BMAL heterodimers activate the transcription of per, cry, rev-erb and ror via E box elements on the promoters. The transcribed PER and CRY proteins form a multimeric complex, enter the nucleus and inhibit CLOCK/BMAL-mediated transcription. The PER/CRY complex is degraded during the night, thereby releasing this inhibitory effect and allowing a new cycle of transcription to begin. REV-ERB and ROR proteins compete for a response element (RORE) on the promoter of bmal, thereby repressing and activating transcription accordingly.

Circadian regulation of the immune system

The biological clock regulates many aspects of the immune system. As a consequence, immune responses often demonstrate measurable circadian variation. Several lines of evidence highlight the contribution of the circadian clock to the function of the immune system. Firstly, multiple immune cells possess the clockwork machinery and show daily variation in their function [6–8]. Secondly, disruption of the circadian clock has a detrimental effect on the function of the immune system. Finally, deletion of core clock genes can impact on immune responses. These established interactions between the clock and the immune system all have implications for RA.

Oscillating immune cells; a source for rhythms in IL-6 levels

Studies have revealed that the individual cellular components of the immune system that initiate and perpetuate inflammatory pathways are tightly regulated by the circadian clock. These include mast cells, natural killer cells, eosinophils, basophils, T lymphocytes and macrophages. As yet, the dominant cell type driving circadian disease expression in RA remains undetermined. One candidate that may underlie the rhythmic IL-6 secretion is the CD4+ T lymphocyte, which is considered a key mediator of RA with an established role in disease initiation and perpetuation. It has been shown that CD4+ T lymphocytes possess a circadian oscillator that drives rhythmic responses to activating stimuli, as manifest by altered cell proliferation and cytokine secretion [7, 9]. In addition, macrophages possess an intrinsic clock, and over 8% of the macrophage transcriptome is under circadian control [6]. Although macrophages are not thought to be involved in the initiation of RA, their pro-inflammatory and tissue destructive actions contribute to disease expression and these phagocytes mediate joint inflammation and tissue destruction in erosive disease. A recent study demonstrated rhythmic IL-6 responses to stimulation by both human and mouse macrophages, and established the importance of the clock gene (and nuclear receptor) rev-erb α in generating these rhythms [10]. Finally, fibroblast like synoviocytes, located within the intimal lining of the synovium, also have a circadian oscillator [11]. These cells are a major source of pro-inflammatory cytokines in RA, although in active disease they may no longer oscillate. To summarise, a number of candidate cells may orchestrate the circadian oscillation in IL-6 secretion. However, the mechanism by which the clock exerts this control is not yet fully established. The production of IL-6 by immune cells may fall under central clock control as a result of circulating mediators (such as glucocorticoids), or alternatively, local clocks may directly drive IL-6 oscillations. Of course, both possibilities may co-exist.

Disruption of the circadian clock and rheumatoid arthritis

Recent studies have identified an intriguing bi-directional interaction between inflammation and the circadian clock. Disruption of the clock has a significant effect on the performance of the immune system, and there is a suggestion that this might impact negatively on the pathogenesis of RA. In converse, inflammation can directly alter cellular expression of core clock genes [12]. Disruption of the clock is exemplified by jet-lag, a consequence of de-synchrony between the internal clock and the environment, resulting in the need to re-set the phase of the clock. Depending on the number of time zones crossed and direction of travel, it can take a number of days to re-synchronise the body clock. Shift-workers experience the same phenomenon. These constant shifts in the daily schedule are detrimental to health and have been linked with an increased incidence of a number of chronic diseases such as cancer, cardiovascular disease, metabolic syndromes, diabetes and irritable bowel syndrome [13]. Circadian disruption can be modelled by inducing 'chronic jet-lag' in animals through exposure to regular shifts in the lighting schedule. This type of environmental disturbance has negative effects on the function of immune cells, including macrophages and natural killer cells, and has detrimental effects on the survival of mice after immune challenge [14, 15]. Interestingly, a study in 2010 provided a significant link between shift work and an increased risk of RA (in women) [16]. This is the first observation of its kind, supporting the concept that the body clock not only impacts on the symptoms of this disease, but is also involved in the pathogenesis.

Core clock genes - links with rheumatoid arthritis

A direct link has been made between core components of the molecular clock and inflammatory pathways known to be relevant to RA. Mice lacking two core clock genes, cry1 and cry2, show disruptions in their circadian clock. Where wild-type mice are able to maintain strong circadian behaviour in the absence of light cues, cry1-/-cry2-/- mice are behaviourally arrhythmic under constant darkness. These double knockout mice have a more aggressive inflammatory arthritis in response to collagen induction (collagen-induced arthritis) [17]. This is attributed to the function of CRY as a transcriptional inhibitor of the tnfα gene. It is now established that CRY acts on a broad range of pro-inflammatory target genes, which it modulates through the NFκB pathway [18]. Whilst this is the only established direct regulatory pathway linking the core clock and the pathogenesis of RA, several clock outputs (including hormones and clock-controlled genes) have been attributed roles, and these are expanded on below.

Circadian control of the endocrine system and links with RA

The endocrine system mediates the dissemination of timing signals from the SCN throughout the body. Two hormones in particular act as circadian agents - melatonin and glucocorticoids. Both are important in inflammation and regulation of the immune response, and may contribute to the pathogenesis of RA.

Melatonin

Melatonin is primarily secreted from the pineal gland (other sources include the retina, Harderian glands, intestine, skin, leukocytes and bone marrow). The SCN regulates melatonin synthesis through tonic inhibition of noradrenergic stimulation of the pineal gland. This hormone is only synthesized during the night (as synthesis is inhibited by daylight; Figure 1). Melatonin affects several aspects of the immune response, and is known to modulate pro-inflammatory cytokines, inhibit proliferation of fibroblast-like synoviocytes and regulate leukocyte function [19, 20]. Interestingly, RA patients have altered levels of circulating melatonin [21]. Sulli and colleagues [21] report higher baseline levels of serum melatonin in RA patients (at the start and end of the dark phase) and an altered temporal profile with a more rapid increase at the start of the night and an earlier peak. These observations suggest that the effects of melatonin may be more pro-inflammatory than anti-inflammatory. In support, in vivo studies clearly indicate that melatonin has adverse effects on the development and severity of inflammatory arthritis. Administration of melatonin in a collagen-induced arthritis model resulted in enhanced disease incidence and severity [22]. This is concordant with studies showing that removal of the pineal gland, the main source of melatonin, has a beneficial effect on disease progression in the collagen-induced arthritis model [23]. It is important to consider that melatonin is widely available and often utilised by shift-workers and long-haul travellers, where it is thought to result in faster adaptation to a new time zone. Given that a clinical study of the use of melatonin in RA resulted in poor outcomes, with an increase in some markers of inflammation [24, 25], this current widespread use of melatonin may need to be addressed.

Glucocorticoids

Glucocorticoids are endogenous anti-inflammatory agents (cortisol in humans and corticosterone in rodents). Circulating levels fluctuate throughout the day in a circadian manner, peaking just before waking in mammals (Figure 1). The diurnal variation in circulating glucocorticoid concentrations is driven by the circadian clock through direct neural connections between the central clock in the SCN, and the paraventricular nucleus in the hypothalamus, the site of central control for the hypothalamic pituitary adrenal axis. Glucocorticoids act via the ubiquitously expressed glucocorticoid receptor (GR), a ligand-activated transcription factor, belonging to the nuclear receptor superfamily (see below). The activated, hormone bound GR translocates from the cytoplasm to the nucleus, and binds to target gene regulatory regions. Binding to positive glucocorticoid response elements as a homodimer results in transcriptional activation; and binding to negative glucocorticoid response elements results in transcriptional inhibition [26]. Additionally, ligand-bound GR can function as a transrepressor by binding (either directly or indirectly) to other transcription factors (such as activator protein-1 and NFκB) in a so-called tethering mechanism, to inhibit their trans-activating function and prevent their association with DNA [27–29]. Glucocorticoids can also exert rapid effects by coupling the GR to intracellular signalling cascades, including mitogen-activated protein and phosphoinositide-3 kinases [30]. This mechanism of action is less well-characterised, but offers potential cross-talk between growth factors and cytokine signal ling, relevant to inflammatory arthritis, and glucocorticoid action.

In RA patients, the natural morning peak in cortisol levels occurs approximately 40 minutes after the morning rise in IL-6 levels [2]. It is a striking paradox that the stress-responsive hypothalamic pituitary adrenal axis is not constitutively activated in RA patients, and in fact, the dynamic responses to activation have been shown to be blunted, for reasons that remain obscure, but with obvious clinical implications [2]. Recent insights suggest that merely tracking circulating cortisol levels is inadequate as a means to assess glucocorticoid action. Indeed, in certain cell-types, expression of the GR shows daily rhythms; furthermore, the function of the receptor is under clock control, thereby suggesting alternative mechanisms by which the clock regulates glucocorticoid sensitivity.

Nuclear receptors

Nuclear receptors are ligand-dependent transcription factors, which modulate gene expression through direct binding to DNA response elements. Over half of the nuclear receptor family exhibit rhythmic expression in a tissue-specific manner [31], and many can feedback directly onto the clock itself. Nuclear receptors are recognised as key intermediaries between the molecular clock and a wide array of physiological processes, including immunity [32]. In the context of RA, three nuclear receptors are of particular interest: GR, retinoid-related orphan receptors (RORs) and peroxisome proliferator activated receptors (PPARs).

Glucocorticoid receptor

There are three protein isoforms of the GR (α, β and γ), of which GRα is the dominant form, mediating most of the actions of glucocorticoids. Evidence suggests that receptor levels show circadian variation in some tissues, but not others [31]. In addition, the cellular clock can drive post-translational modification of the GR protein, so affecting its function. This includes GR interaction with NFκB, which consequently affects the anti-inflammatory capacity of glucocorticoids. CLOCK and BMAL proteins can acetylate a cluster of lysine residues in the hinge region of the GR, which selectively attenuates the ability of the GR to bind to glucocorticoid response elements. The naturally occurring oscillations in CLOCK/BMAL levels consequently translate into circadian fluctuations in the acetylation of the GR; for example, in human peripheral blood mononuclear cells, acetylated GR is almost three-fold higher in the day [33]. An additional insight is provided by the recent discovery that GR and the core clock protein CRY form a dimer, which is critical to normal GR function. As CRY is only present for part of the circadian cycle this imposes indirect clock control upon the GR, with the suggestion that only specific aspects of GR function (including gluconeogenesis and hypothalamo-pituitary axis tone) show a circadian component [34]. This opens up a potential therapeutic opportunity whereby through altering the timing of delivery (chronotherapy) it may be possible to maximise the anti-inflammatory effects of glucocorticoids whilst alleviating the adverse metabolic effects.

Retinoid-related orphan receptors

RORs are a family of nuclear receptors with three members, RORα, RORβ and RORγ (of which there are two isoforms γ and γt). Transcription of ror genes is rhythmic, although to varying degrees, and the ROR proteins have a well established role in the molecular clock via transcriptional control of bmal (mice deficient in either RORα or RORβ show aberrant circadian behaviour). The circadian rhythmic RORα has a role in promoting the differentiation of T cells into TH17 cells. These are a subset of T helper cells (distinct from TH1, TH2 and regulatory T cells) characterised by their expression of the pro-inflammatory cytokines IL-17 and IL-17F, which represent a substantial population of infiltrating CD4+ T cells in inflamed synovial tissue [35]. In addition, RORα has a role in regulating inflammation. Staggerer (sg) mice, which carry a microdeletion in the rorα gene preventing translation of the ligand binding homology domain, have immune system deficiencies. Macrophages isolated from sg mice are hyper-responsive to lipopolysaccharide, producing elevated levels of cytokines (IL-1β, TNFα and IL-1α) [36]. Further studies have identified direct interactions between RORα and inflammatory genes. RORα1 can activate IκBα (the main inhibitory protein in the NFκB pathway) via a response element on the promoter, thereby suppressing inflammatory responses [37]. Conversely, RORα can up-regulate the inflammatory response through binding to a response element on the promoter of the il6 gene to enhance IL-6 production [38]. Of note, RORγt also plays a key role in the differentiation of T cells into TH17 cells [39]; however, RORγt expression is clock independent.

Peroxisome proliferator activated receptors

PPARs are a further class of nuclear receptors closely interlinked with the circadian clock. They are ligand-activated transcription regulators, which act by forming heterodimers with retinoic acid receptors (RXRs) and binding to response elements on target genes. All three PPARs (α, β/δ and γ) show tissue-specific patterns of circadian expression. Clock regulation of both the expression and function of PPARs is multifaceted. PPARα transcription is directly regulated by CLOCK/BMAL via an Ebox element in the promoter. In comparison, the activity of PPARγ is regulated through the activity of circadian proteins, which include the transcriptional enhancer nocturin, the co-activator protein PGC-1 α, and the core clock protein PER2 [40–42]. Endogenous ligands for PPARs include free fatty acids and eicosanoids and so in response to a high fat diet or fasting (when lipolysis in adipose tissue is active) these receptors become activated. The PPARs play a key role in lipid metabolism, energy partition, and also in regulating macrophage activity. Importantly, PPARγ and δ facilitate 'alternative' activation of macrophages - a cellular state that plays a key role in the resolution of inflammation [43]. In addition to modulating macrophage function, PPARs are regulators of T-cell function. Both PPAR α and γ are expressed in T cells, α is down-regulated following T-cell activation, whilst γ is up-regulated [44]. PPARγ is known to mediate the proliferative response of T cells, inhibits their differentiation into TH17 cells, and induces T-cell apoptosis [45–47].

Adipokines

The circadian clock is fundamental in the regulation of metabolic processes; controlling expression of genes involved in metabolic pathways but also responding to metabolic cues [32]. Circadian disruption has detrimental effects on metabolism, and is an exacerbating factor in the incidence of metabolic syndrome, which itself is associated with the risk of developing RA [48]. It is proposed that the link between metabolic syndrome and RA is due to the inflammatory milieu associated with metabolic syndrome, much of which is provided by the increased quantities of adipose tissue, which secretes elevated levels of cytokines (TNFα and IL-6) and adipokines. Adipokines are signalling molecules produced primarily by adipose tissue and include adiponectin, leptin, resistin and visfatin. White adipose tissue and its constituent adipocytes are both circadian rhythmic, and many adipokines exhibit 24-hour variation in plasma concentrations [49]. Recent studies have identified clear associations between adipokines and the pathophysiology of rheumatic diseases. For example, visfatin (also known as nicotinamide phosphoribosyltransferase (NAMPT) or pre-B cell colony-enhancing factor (PBEF)) is released by visceral fat, macrophages, liver, skeletal muscle and leukocytes in a rhythmic manner to produce diurnal rhythms in circulating levels [50]. Elevated levels of visfatin are associated with RA; furthermore, levels detected in serum and synovial fluid correlate with clinical disease severity [51]. Visfatin is pro-inflammatory, and although the underlying mechanisms are not fully understood, it stimulates synovial fibroblasts and monocytes to release pro-inflammatory cytokines and matrix metalloproteinases. The well-established and complex relationship between the circadian clock and metabolic processes provides another link between the clock and the pathogenesis of RA.

Targeting clock output genes as therapeutics

Synthetic glucocorticoids, such as dexamethasone and prednisone, have long been a cornerstone in the clinical treatment of RA. Yet, prolonged usage is associated with a number of adverse effects, including immune suppression, metabolic imbalance and diabetes. Although the recent development of biologics (which specifically target components of the immune system that play pivotal roles in driving inflammation) has made great advances in the treatment of RA, these unfortunately are known to increase the risk of infection (that is, tuberculosis and hepatitis B). Consequently, there remains a need to develop new therapies for the treatment of this inflammatory disorder.

Given that RORs and PPARs have significant input into key events that contribute to the pathogenesis of RA, both present as potential therapeutic targets in this, and other, inflammatory diseases. Many synthetic PPAR ligands are already in clinical use. The thiazolidinediones are a group of synthetic PPARγ ligands that have anti-hyperglycemic actions and are used in the management of type 2 diabetes. They include rosiglitazone, troglitazone and pioglitazone. Synthetic PPARγ ligands, and the proposed endogenous ligand 15-deoxy-delta (12,14)-prostoglandin J2 are anti-inflammatory, and improve outcome in animal models of inflammatory arthritis [52–54]. Recently, a clinical trial has suggested that the concomitant use of pioglitazone and methotrexate may have synergistic effects in RA [55]. However, although these compounds have potent anti-inflammatory properties, off-target effects, possibly through activation of the glucocorticoid receptor [56], can result in osteoporosis or cardiac failure, which is likely to limit their application. Similarly, there is growing evidence that PPARα agonists may be useful in the treatment of RA. Synthetic PPARα ligands include fibrates (fenofibrate, gemfibrozil and clofibrate), hypolipidemic compounds already utilised in the treatment of metabolic disorders. Promisingly, fenofibrate has been shown to be effective at reducing inflammation in experimental arthritis models [57].

Novel therapeutic targets

A full understanding of the network through which the circadian clock regulates inflammatory pathways is likely to provide novel therapeutic targets for treating RA and other inflammatory disorders and autoimmune conditions. Given its role in T-cell function, one such target is ROR. A novel synthetic ligand for ROR (α and γ) has recently been synthesised (SR1001) that inhibits TH17 cell differentiation and reduces the severity of murine experimental autoimmune encephalomyelitis, a TH17 cell-mediated autoimmune disease [58]. In addition, another key area for therapeutic targeting is the molecular clock .Given the role of CRY in regulating inflammatory pathways, the recently identified CRY activator KL001 may prove to have anti-inflammatory properties beneficial in the treatment of RA [59]. Finally, our relatively recent understanding of the role of adipokines in rheumatic disease suggests that these circadian-regulated molecules could be explored as a new avenue of treatment.

Chronotherapy for the treatment of rheumatic arthritis

Chronotherapy refers to the application of information about circadian rhythms in therapeutics. An understanding of how the clock influences expression of the drug target or elements of the drug's metabolic pathway can be utilised to optimise dosing times to make treatment more effective; this might maximise effectiveness and minimise side effects. Given the strong circadian component to RA, there is obvious potential for the use of chronotherapy in the treatment of this disease. As proof of principle, a study in 1997 demonstrated the advantages of administering prednisone overnight, just prior to the rise in IL-6 levels and joint stiffness [60]. This small study identified a significant improvement in joint stiffness when this glucocorticoid was administered at 2:00 am versus 7:30 am. In response to the impractical nature of dosing during the night-time, modified release prednisone (Lodotra) has been developed. The CAPRA-1 study compared the effectiveness of Lodotra with immediate release prednisone in patients with active RA receiving disease-modifying anti-rheumatic drugs [61, 62]. The group receiving Lodotra demonstrated significant improvements in the duration of their morning joint stiffness after just 2 weeks. Lodotra significantly reduced morning IL-6 levels, which was not seen in the immediate-release prednisone group. These beneficial effects over immediate-release prednisone were sustained over a 12 month period [63]. Furthermore, in a placebo-controlled study (CAPRA-2) the addition of low-dose (5 mg) Lodotra to existing treatment regimes proved to be beneficial [62]. These trials highlight the positive effects of chronotherapy on relieving morning joint pain and stiffness, and are the first of their kind to show that modification of pharmacokinetics and the application of timed-dosing can boost the effectiveness of established RA therapies. It is expected that application of chronotherapy to alternative types of RA therapies may prove similarly beneficial.

Conclusion

RA has long been known to have a circadian component, not just in its symptoms, but in disease biomarkers. It is only in recent years, however, that the importance of the circadian clock in regulating the immune response has been recognised. This control is exerted both at a systemic level, through signalling pathways originating in the central clock, and at the local level by autonomous clocks in inflammatory cells and tissues (Figure 3). The biological clock directly regulates processes that contribute to the pathogenesis of RA, such as the production of inflammatory cytokines by immune cells. In addition, clock-regulated adipokines and nuclear receptors contribute to processes integral to disease progression, such as inflammation and T-cell activation. A greater understanding of how the clock couples to the immune system, and the involvement of clock outputs in disease pathogenesis, is likely to produce novel therapeutic targets for the treatment of not only RA but also other inflammatory disorders. Harnessing the knowledge we have about how the inflammatory response is regulated by the clock to develop chronotherapy regimens has already proved valuable, and there is much scope to develop this area to improve the efficacy and reduce adverse effects of both current and new treatment strategies.

Figure 3
figure 3

Interactions between the circadian clock and the immune system that contribute to disease pathogenesis in rheumatoid arthritis. GR, glucocorticoid receptor; PPAR, peroxisome proliferator activated receptor; ROR, retinoic acid related orphan receptor.

Abbreviations

GR:

glucocorticoid receptor

IL:

interleukin

NF:

nuclear factor

PPAR:

peroxisome proliferator activated receptor

RA:

rheumatoid arthritis

ROR:

retinoic acid related orphan receptor

RORE:

response element

SCN:

suprachiasmatic nucleus

TNF:

tumour necrosis factor.

References

  1. Ingpen ML: The quantitative measurement of joint changes in rheumatoid arthritis. Ann Phys Med. 1968, 9: 322-327.

    CAS  PubMed  Google Scholar 

  2. Perry MG, Kirwan JR, Jessop DS, Hunt LP: Overnight variations in cortisol, interleukin 6, tumour necrosis factor alpha and other cytokines in people with rheumatoid arthritis. Ann Rheum Dis. 2009, 68: 63-68.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Arvidson NG, Gudbjornsson B, Elfman L, Ryden AC, Totterman TH, Hallgren R: Circadian rhythm of serum interleukin-6 in rheumatoid arthritis. Ann Rheum Dis. 1994, 53: 521-524. 10.1136/ard.53.8.521.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Albrecht U: Timing to perfection: the biology of central and peripheral circadian clocks. Neuron. 2012, 74: 246-260. 10.1016/j.neuron.2012.04.006.

    Article  CAS  PubMed  Google Scholar 

  5. Relogio A, Westermark PO, Wallach T, Schellenberg K, Kramer A, Herzel H: Tuning the mammalian circadian clock: robust synergy of two loops. PLoS Comput Biol. 2011, 7: e1002309-10.1371/journal.pcbi.1002309.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Keller M, Mazuch J, Abraham U, Eom GD, Herzog ED, Volk HD, Kramer A, Maier B: A circadian clock in macrophages controls inflammatory immune responses. Proc Natl Acad Sci USA. 2009, 106: 21407-21412. 10.1073/pnas.0906361106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Bollinger T, Leutz A, Leliavski A, Skrum L, Kovac J, Bonacina L, Benedict C, Lange T, Westermann J, Oster H, Solbach W: Circadian clocks in mouse and human CD4+ T cells. PLoS One. 2011, 6: e29801-10.1371/journal.pone.0029801.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Wang X, Reece SP, Van Scott MR, Brown JM: A circadian clock in murine bone marrow-derived mast cells modulates IgE-dependent activation in vitro. Brain Behav Immun. 2011, 25: 127-134. 10.1016/j.bbi.2010.09.007.

    Article  PubMed  Google Scholar 

  9. Fortier EE, Rooney J, Dardente H, Hardy MP, Labrecque N, Cermakian N: Circadian variation of the response of T cells to antigen. J Immunol. 2011, 187: 6291-6300. 10.4049/jimmunol.1004030.

    Article  CAS  PubMed  Google Scholar 

  10. Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, Farrow SN, Else KJ, Singh D, Ray DW, Loudon AS: The nuclear receptor REV-ERBalpha mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci USA. 2012, 109: 582-587. 10.1073/pnas.1106750109.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  11. Haas S, Straub RH: Disruption of rhythms of molecular clocks in primary synovial fibroblasts of patients with osteoarthritis and rheumatoid arthritis, role of IL-1beta/TNF. Arthritis Res Ther. 2012, 14: R122-10.1186/ar3852.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Coogan AN, Wyse CA: Neuroimmunology of the circadian clock. Brain Res. 2008, 1232: 104-112.

    Article  CAS  PubMed  Google Scholar 

  13. Wang XS, Armstrong ME, Cairns BJ, Key TJ, Travis RC: Shift work and chronic disease: the epidemiological evidence. Occup Med (Lond). 2011, 61: 78-89. 10.1093/occmed/kqr001.

    Article  Google Scholar 

  14. Castanon-Cervantes O, Wu M, Ehlen JC, Paul K, Gamble KL, Johnson RL, Besing RC, Menaker M, Gewirtz AT, Davidson AJ: Dysregulation of inflammatory responses by chronic circadian disruption. J Immunol. 2010, 185: 5796-5805. 10.4049/jimmunol.1001026.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Logan RW, Zhang C, Murugan S, O'Connell S, Levitt D, Rosenwasser AM, Sarkar DK: Chronic shift-lag alters the circadian clock of NK cells and promotes lung cancer growth in rats. J Immunol. 2012, 188: 2583-2591. 10.4049/jimmunol.1102715.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Puttonen S, Oksanen T, Vahtera J, Pentti J, Virtanen M, Salo P, Kivimaki M: Is shift work a risk factor for rheumatoid arthritis? The Finnish Public Sector study. Ann Rheum Dis. 2010, 69: 779-780.

    Article  PubMed  Google Scholar 

  17. Hashiramoto A, Yamane T, Tsumiyama K, Yoshida K, Komai K, Yamada H, Yamazaki F, Doi M, Okamura H, Shiozawa S: Mammalian clock gene Cryptochrome regulates arthritis via proinflammatory cytokine TNF-alpha. J Immunol. 2010, 184: 1560-1565. 10.4049/jimmunol.0903284.

    Article  CAS  PubMed  Google Scholar 

  18. Narasimamurthy R, Hatori M, Nayak SK, Liu F, Panda S, Verma IM: Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc Natl Acad Sci USA. 2012, 109: 12662-12667. 10.1073/pnas.1209965109.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Radogna F, Diederich M, Ghibelli L: Melatonin: a pleiotropic molecule regulating inflammation. Biochem Pharmacol. 2010, 80: 1844-1852. 10.1016/j.bcp.2010.07.041.

    Article  CAS  PubMed  Google Scholar 

  20. Nah SS, Won HJ, Park HJ, Ha E, Chung JH, Cho HY, Baik HH: Melatonin inhibits human fibroblast-like synoviocyte proliferation via extracellular signal-regulated protein kinase/P21(CIP1)/P27(KIP1) pathways. J Pineal Res. 2009, 47: 70-74. 10.1111/j.1600-079X.2009.00689.x.

    Article  CAS  PubMed  Google Scholar 

  21. Sulli A, Maestroni GJ, Villaggio B, Hertens E, Craviotto C, Pizzorni C, Briata M, Seriolo B, Cutolo M: Melatonin serum levels in rheumatoid arthritis. Ann NY Acad Sci. 2002, 966: 276-283. 10.1111/j.1749-6632.2002.tb04227.x.

    Article  CAS  PubMed  Google Scholar 

  22. Hansson I, Holmdahl R, Mattsson R: The pineal hormone melatonin exaggerates development of collagen-induced arthritis in mice. J Neuroimmunol. 1992, 39: 23-30. 10.1016/0165-5728(92)90171-G.

    Article  CAS  PubMed  Google Scholar 

  23. Hansson I, Holmdahl R, Mattsson R: Pinealectomy ameliorates collagen II-induced arthritis in mice. Clin Exp Immunol. 1993, 92: 432-436.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Forrest CM, Mackay GM, Stoy N, Stone TW, Darlington LG: Inflammatory status and kynurenine metabolism in rheumatoid arthritis treated with melatonin. Br J Clin Pharmacol. 2007, 64: 517-526. 10.1111/j.1365-2125.2007.02911.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Cutolo M, Straub RH, Buttgereit F: Circadian rhythms of nocturnal hormones in rheumatoid arthritis: translation from bench to bedside. Ann Rheum Dis. 2008, 67: 905-908.

    Article  CAS  PubMed  Google Scholar 

  26. Surjit M, Ganti KP, Mukherji A, Ye T, Hua G, Metzger D, Li M, Chambon P: Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor. Cell. 2011, 145: 224-241. 10.1016/j.cell.2011.03.027.

    Article  CAS  PubMed  Google Scholar 

  27. McMaster A, Ray DW: Modelling the glucocorticoid receptor and producing therapeutic agents with anti-inflammatory effects but reduced side-effects. Exp Physiol. 2007, 92: 299-309. 10.1113/expphysiol.2006.036194.

    Article  CAS  PubMed  Google Scholar 

  28. Rhen T, Cidlowski JA: Antiinflammatory action of glucocorticoids--new mechanisms for old drugs. N Engl J Med. 2005, 353: 1711-1723. 10.1056/NEJMra050541.

    Article  CAS  PubMed  Google Scholar 

  29. Uhlenhaut NH, Barish GD, Yu RT, Downes M, Karunasiri M, Liddle C, Schwalie P, Hubner N, Evans RM: Insights into negative regulation by the glucocorticoid receptor from genome-wide profiling of inflammatory cistromes. Mol Cell. 2013, 49: 1-14.

    Article  Google Scholar 

  30. Stahn C, Buttgereit F: Genomic and nongenomic effects of glucocorticoids. Nat Clin Pract Rheumatol. 2008, 4: 525-533.

    Article  CAS  PubMed  Google Scholar 

  31. Yang X, Downes M, Yu RT, Bookout AL, He W, Straume M, Mangelsdorf DJ, Evans RM: Nuclear receptor expression links the circadian clock to metabolism. Cell. 2006, 126: 801-810. 10.1016/j.cell.2006.06.050.

    Article  CAS  PubMed  Google Scholar 

  32. Bechtold DA, Gibbs JE, Loudon AS: Circadian dysfunction in disease. Trends Pharmacol Sci. 2010, 31: 191-198. 10.1016/j.tips.2010.01.002.

    Article  CAS  PubMed  Google Scholar 

  33. Charmandari E, Chrousos GP, Lambrou GI, Pavlaki A, Koide H, Ng SS, Kino T: Peripheral CLOCK regulates target-tissue glucocorticoid receptor transcriptional activity in a circadian fashion in man. PLoS One. 2011, 6: e25612-10.1371/journal.pone.0025612.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Lamia KA, Papp SJ, Yu RT, Barish GD, Uhlenhaut NH, Jonker JW, Downes M, Evans RM: Cryptochromes mediate rhythmic repression of the glucocorticoid receptor. Nature. 2011, 480: 552-556.

    PubMed Central  CAS  PubMed  Google Scholar 

  35. McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR: The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006, 126: 1121-1133. 10.1016/j.cell.2006.07.035.

    Article  PubMed  Google Scholar 

  36. Kopmels B, Mariani J, Delhaye-Bouchaud N, Audibert F, Fradelizi D, Wollman EE: Evidence for a hyperexcitability state of staggerer mutant mice macrophages. J Neurochem. 1992, 58: 192-199. 10.1111/j.1471-4159.1992.tb09295.x.

    Article  CAS  PubMed  Google Scholar 

  37. Delerive P, Monte D, Dubois G, Trottein F, Fruchart-Najib J, Mariani J, Fruchart JC, Staels B: The orphan nuclear receptor ROR alpha is a negative regulator of the inflammatory response. EMBO Rep. 2001, 2: 42-48. 10.1093/embo-reports/kve007.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Journiac N, Jolly S, Jarvis C, Gautheron V, Rogard M, Trembleau A, Blondeau JP, Mariani J, Vernet-der GB: The nuclear receptor ROR(alpha) exerts a bi-directional regulation of IL-6 in resting and reactive astrocytes. Proc Natl Acad Sci USA. 2009, 106: 21365-21370. 10.1073/pnas.0911782106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Eberl G, Littman DR: The role of the nuclear hormone receptor RORgammat in the development of lymph nodes and Peyer's patches. Immunol Rev. 2003, 195: 81-90. 10.1034/j.1600-065X.2003.00074.x.

    Article  CAS  PubMed  Google Scholar 

  40. Kawai M, Green CB, Lecka-Czernik B, Douris N, Gilbert MR, Kojima S, Ackert-Bicknell C, Garg N, Horowitz MC, Adamo ML, Clemmons DR, Rosen CJ: A circadian-regulated gene, Nocturnin, promotes adipogenesis by stimulating PPAR-gamma nuclear translocation. Proc Natl Acad Sci USA. 2010, 107: 10508-10513. 10.1073/pnas.1000788107.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Liu C, Li S, Liu T, Borjigin J, Lin JD: Transcriptional coactivator PGC-1alpha integrates the mammalian clock and energy metabolism. Nature. 2007, 447: 477-481. 10.1038/nature05767.

    Article  CAS  PubMed  Google Scholar 

  42. Grimaldi B, Bellet MM, Katada S, Astarita G, Hirayama J, Amin RH, Granneman JG, Piomelli D, Leff T, Sassone-Corsi P: PER2 controls lipid metabolism by direct regulation of PPARgamma. Cell Metab. 2010, 12: 509-520. 10.1016/j.cmet.2010.10.005.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  43. Chawla A: Control of macrophage activation and function by PPARs. Circ Res. 2010, 106: 1559-1569. 10.1161/CIRCRESAHA.110.216523.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Jones DC, Ding X, Daynes RA: Nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is expressed in resting murine lymphocytes. The PPARalpha in T and B lymphocytes is both transactivation and transrepression competent. J Biol Chem. 2002, 277: 6838-6845. 10.1074/jbc.M106908200.

    Article  CAS  PubMed  Google Scholar 

  45. Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, Hucke S, Alferink J, Nowak N, Beyer M, Mayer G, Langhans B, Klockgether T, Waisman A, Eberl G, Schultze J, Famulok M, Kolanus W, Glass C, Kurts C, Knolle PA: The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity. J Exp Med. 2009, 206: 2079-2089. 10.1084/jem.20082771.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  46. Clark RB, Bishop-Bailey D, Estrada-Hernandez T, Hla T, Puddington L, Padula SJ: The nuclear receptor PPAR gamma and immunoregulation: PPAR gamma mediates inhibition of helper T cell responses. J Immunol. 2000, 164: 1364-1371.

    Article  CAS  PubMed  Google Scholar 

  47. Harris SG, Phipps RP: Induction of apoptosis in mouse T cells upon peroxisome proliferator-activated receptor gamma (PPAR-gamma) binding. Adv Exp Med Biol. 2002, 507: 421-425.

    Article  CAS  PubMed  Google Scholar 

  48. Gremese E, Ferraccioli G: The metabolic syndrome: the crossroads between rheumatoid arthritis and cardiovascular risk. Autoimmun Rev. 2011, 10: 582-589. 10.1016/j.autrev.2011.04.018.

    Article  CAS  PubMed  Google Scholar 

  49. Johnston JD: Adipose circadian rhythms: translating cellular and animal studies to human physiology. Mol Cell Endocrinol. 2012, 349: 45-50. 10.1016/j.mce.2011.05.008.

    Article  CAS  PubMed  Google Scholar 

  50. Benedict C, Shostak A, Lange T, Brooks SJ, Schioth HB, Schultes B, Born J, Oster H, Hallschmid M: Diurnal rhythm of circulating nicotinamide phosphoribosyltransferase (Nampt/visfatin/PBEF): impact of sleep loss and relation to glucose metabolism. J Clin Endocrinol Metab. 2012, 97: E218-222. 10.1210/jc.2011-2241.

    Article  CAS  PubMed  Google Scholar 

  51. Gomez R, Conde J, Scotece M, Gomez-Reino JJ, Lago F, Gualillo O: What's new in our understanding of the role of adipokines in rheumatic diseases?. Nat Rev Rheumatol. 2011, 7: 528-536. 10.1038/nrrheum.2011.107.

    Article  CAS  PubMed  Google Scholar 

  52. Tomita T, Kakiuchi Y, Tsao PS: THR0921, a novel peroxisome proliferator-activated receptor gamma agonist, reduces the severity of collagen-induced arthritis. Arthritis Res Ther. 2006, 8: R7-10.1186/ar1856.

    Article  PubMed Central  PubMed  Google Scholar 

  53. Song JS, Kim CH, Heo JY, Cho YS: Rosiglitazone reduces a wide range of proinflammatory profiles in synovial fibroblast SW982 under spheroid culture. Immunol Lett. 2010, 131: 81-88. 10.1016/j.imlet.2010.02.010.

    Article  CAS  PubMed  Google Scholar 

  54. Lin TH, Tang CH, Wu K, Fong YC, Yang RS, Fu WM: 15-deoxy-Delta(12,14) -prostaglandin-J2 and ciglitazone inhibit TNF-alpha-induced matrix metalloproteinase 13 production via the antagonism of NF-kappaB activation in human synovial fibroblasts. J Cell Physiol. 2011, 226: 3242-3250. 10.1002/jcp.22685.

    Article  CAS  PubMed  Google Scholar 

  55. Shahin D, Toraby EE, Abdel-Malek H, Boshra V, Elsamanoudy AZ, Shaheen D: Effect of peroxisome proliferator-activated receptor gamma agonist (pioglitazone) and methotrexate on disease activity in rheumatoid arthritis (experimental and clinical study). Clin Med Insights Arthritis Musculoskelet Disord. 2011, 4: 1-10.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  56. Matthews L, Berry A, Tersigni M, D'Acquisto F, Ianaro A, Ray D: Thiazolidinediones are partial agonists for the glucocorticoid receptor. Endocrinology. 2009, 150: 75-86.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  57. Okamoto H, Iwamoto T, Kotake S, Momohara S, Yamanaka H, Kamatani N: Inhibition of NF-kappaB signaling by fenofibrate, a peroxisome proliferator-activated receptor-alpha ligand, presents a therapeutic strategy for rheumatoid arthritis. Clin Exp Rheumatol. 2005, 23: 323-330.

    CAS  PubMed  Google Scholar 

  58. Solt LA, Kumar N, Nuhant P, Wang Y, Lauer JL, Liu J, Istrate MA, Kamenecka TM, Roush WR, Vidović D, Schürer SC, Xu J, Wagoner G, Drew PD, Griffin PR, Burris TP: Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature. 2011, 472: 491-494. 10.1038/nature10075.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  59. Hirota T, Lee JW, St John PC, Sawa M, Iwaisako K, Noguchi T, Pongsawakul PY, Sonntag T, Welsh DK, Brenner DA, Doyle FJ 3rd, Schultz PG, Kay SA: Identification of small molecule activators of cryptochrome. Science. 2012, 337: 1094-1097. 10.1126/science.1223710.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  60. Arvidson NG, Gudbjornsson B, Larsson A, Hallgren R: The timing of glucocorticoid administration in rheumatoid arthritis. Ann Rheum Dis. 1997, 56: 27-31. 10.1136/ard.56.1.27.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, Jeka S, Krueger K, Szechinski J, Alten R: Efficacy of modified-release versus standard prednisone to reduce duration of morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet. 2008, 371: 205-214. 10.1016/S0140-6736(08)60132-4.

    Article  CAS  PubMed  Google Scholar 

  62. Buttgereit F, Mehta D, Kirwan J, Szechinski J, Boers M, Alten RE, Supronik J, Szombati I, Romer U, Witte S, Saag KG: Low-dose prednisone chronotherapy for rheumatoid arthritis: a randomised clinical trial (CAPRA-2). Ann Rheum Dis. 2012, 72: 204-210.

    Article  PubMed Central  PubMed  Google Scholar 

  63. Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, Jeka S, Krueger K, Szechinski J, Alten R: Targeting pathophysiological rhythms: prednisone chronotherapy shows sustained efficacy in rheumatoid arthritis. Ann Rheum Dis. 2010, 69: 1275-1280. 10.1136/ard.2009.126888.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We wish to thank Professor Andrew Loudon, University of Manchester, for his appraisal of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Julie E Gibbs.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ original submitted files for images

Rights and permissions

Reprints and permissions

About this article

Cite this article

Gibbs, J.E., Ray, D.W. The role of the circadian clock in rheumatoid arthritis. Arthritis Res Ther 15, 205 (2013). https://doi.org/10.1186/ar4146

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/ar4146

Keywords