Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4078 2022-11-25 09:37:45 |
2 format change Meta information modification 4078 2022-11-28 03:58:03 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Jerigova, V.;  Zeman, M.;  Okuliarova, M. Effects of Circadian Disruption on Innate Immunity. Encyclopedia. Available online: https://encyclopedia.pub/entry/36680 (accessed on 27 July 2024).
Jerigova V,  Zeman M,  Okuliarova M. Effects of Circadian Disruption on Innate Immunity. Encyclopedia. Available at: https://encyclopedia.pub/entry/36680. Accessed July 27, 2024.
Jerigova, Viera, Michal Zeman, Monika Okuliarova. "Effects of Circadian Disruption on Innate Immunity" Encyclopedia, https://encyclopedia.pub/entry/36680 (accessed July 27, 2024).
Jerigova, V.,  Zeman, M., & Okuliarova, M. (2022, November 27). Effects of Circadian Disruption on Innate Immunity. In Encyclopedia. https://encyclopedia.pub/entry/36680
Jerigova, Viera, et al. "Effects of Circadian Disruption on Innate Immunity." Encyclopedia. Web. 27 November, 2022.
Effects of Circadian Disruption on Innate Immunity
Edit

Circadian rhythms control almost all aspects of physiology and behavior, allowing temporal synchrony of these processes between each other, as well as with the external environment. In the immune system, daily rhythms of leukocyte functions can determine the strength of the immune response, thereby regulating the efficiency of defense mechanisms to cope with infections or tissue injury. The natural light/dark cycle is the prominent synchronizing agent perceived by the circadian clock, but this role of light is highly compromised by irregular working schedules and unintentional exposure to artificial light at night (ALAN).

circadian rhythms chronodisruption inflammation innate immunity light at night phase shifts

1. Introduction

Circadian rhythms (circa = about; dies = day) represent endogenous oscillations with a period of approximately 24 h. In most species, circadian rhythms are effectively entrained by external factors, primarily by a light/dark (LD) cycle, allowing the anticipation of daily periodic changes in the environment [1][2]. Mammalian circadian rhythms are governed by a master clock located in the suprachiasmatic nuclei (SCN) of the hypothalamus. The SCN receives photic input from the environment and transmits the information to peripheral oscillators to coordinate the optimal timing of physiological and behavioral processes [3].
Life on Earth has evolved under relatively stable conditions of bright days and dark nights. The sun is the primary light source for the majority of organisms, with daylight illumination varying from 50,000 to 100,000 lx, and low illuminance levels during the night, reaching up to 0.3 lx at the full moon [4][5]. Nowadays, light exposure is no longer limited by the natural LD cycle in the industrialized world. Recent studies show that more than 80% of the world’s population lives in light-polluted areas [6] and increasing exposure to artificial light at night (ALAN) represents a novel challenge for both humans and wildlife [7][8]. The straightforward impact of compromised LD cycles is linked with circadian disruption, which can be manifested at multiple levels, depending on the nature of mistimed light information. Such situations are a common part of modern society and include especially various shift work schedules, time-zone transitions, or unintentional ALAN exposure. Here, circadian disruption refers to transient or chronic misalignment between the external LD cycle and endogenous circadian clocks, which can further lead to internal misalignment (impaired phase relationships) or desynchronization (changes in period) among individual endogenous rhythms, diminished peak-trough differences in these rhythms (changes in amplitude) or complete arrhythmicity [9]. The main result is attenuated or abolished circadian control of important physiological processes, underlying potential links to adverse health effects [10][11]. Many epidemiological studies examining the risk of common lifestyle diseases among shift workers or due to ALAN found a positive correlation with the incidence of sleep disorders [12][13], cancer [14][15], metabolic and cardiovascular diseases [16][17][18]. A common feature of most lifestyle and chronic diseases is low-grade inflammation, which can further potentiate disease progression [19]. Therefore, a better understanding of the mechanisms by which circadian disruption influences the status of the immune system and inflammatory responses can be of importance in the search for strategies to minimize the negative consequences of environmentally induced circadian disruption on health.

2. Mammalian Circadian System

In mammals, circadian timekeeping is organized into a multi-oscillator system operating in a hierarchical manner, with the SCN as a master oscillator [20]. The SCN neurons are located alongside the third ventricle above the optic chiasm and form a unified circadian network [21]. Light information is perceived by the intrinsically photosensitive retinal ganglion cells, containing the photopigment melanopsin, and conveyed via the retinohypothalamic tract into the SCN [22]. Subsequently, the SCN communicates timing information to individual peripheral oscillators via neural and humoral pathways [23].
At the molecular level, circadian rhythms are generated through transcriptional-translational feedback loops of clock genes and their protein products, forming a basis of the self-sustained and cell-autonomous molecular clocks [24]. The core feedback loop consists of positive and negative regulators. The CLOCK and BMAL1 proteins heterodimerize to form the CLOCK/BMAL1 complex, which activates transcription via binding to E-box enhancer elements in the promoters of clock genes, Period (Per1, Per2, and Per3) and Cryptochrome (Cry1 and Cry2). The PER and CRY proteins represent a negative limb of the loop, as they form the repressive PER/CRY complex, which enters the nucleus, combines with CLOCK/BMAL1, and inhibits the transcription of E-box-controlled genes [25]. The availability and stability of PER and CRY proteins are regulated by protein kinases and phosphatases [26].
Additionally, the core loop is stabilized by accessory feedback loops, consisting of transcriptional activators and repressors, which regulate target genes either through ROR response elements (RORE) or D-boxes [27]. In this way, nuclear receptors REV-ERBs (α/β) repress and retinoic acid-related orphan receptors (RORα/β/γ) activate the transcription of Bmal1, which contains RORE in its promoter. On the other hand, the CLOCK/BMAL1 complex can activate the transcription of genes encoding REV-ERBs [28]. The next feedback loop is formed by nuclear factor interleukin-3 (NFIL3, also known as E4BP4) and D-box binding protein (DBP), which competitively repress or activate the transcription of D-box regulated genes, such as those encoding the circadian proteins PER, REV-ERBs, and RORs [24]. Importantly, circadian regulatory elements have also been identified in the promoters of numerous immune genes, underlying direct crosstalk between the components of the molecular clockwork and the immune system [29][30].

3. Circadian Rhythms in Innate Immunity

Innate immune mechanisms represent the first line of defense against invading pathogens. Circulating and tissue-specific innate immune cells recognize pathogens or cell injury via pattern recognition receptors [31]. Subsequently, initiated signaling pathways induce the release of specific immune mediators, such as cytokines, chemokines, and antimicrobial peptides, which are involved in numerous effector functions [32]. Effective host defense against infection is based on tightly regulated immune processes. Inflammation is an essential part of the innate immunity in response to infection or tissue injury. However, deregulated inflammatory responses or disbalance between favoring and limiting factors can lead to chronic inflammation and tissue damage [19].
Immune functions, including innate immune mechanisms, are under circadian control. Leukocyte trafficking, inflammatory responses and susceptibility to pathogens exhibit their peaks and troughs at specific times of the day [33][34]. In steady state, circulating immune cell numbers reach a peak during the day in mice and rats [35][36] and during the night in humans [37]. High and low leukocyte numbers in the blood over 24 h mirror their mobilization from the bone marrow in the passive phase (light phase for rats) and their recruitment to tissues at the onset of the active phase (dark phase for rats) [38]. Leukocyte oscillations persist in an absence of external entraining cues, such as the LD cycle, thereby indicating their endogenous nature [39][40]. Rhythmic leukocyte trafficking is complementary controlled by extrinsic factors, including neural and humoral outputs of the central oscillator, immune cell-autonomous clocks, and tissue-specific microenvironment [35][41][42]. For example, reported data show that β3-adrenergic signaling in the mouse bone marrow down-regulates C-X-C motif chemokine ligand 12 (Cxcl12) expression during the light phase, controlling the rhythmic release of hematopoietic progenitors from the bone marrow into the circulation [43]. Additionally, low corticosterone levels at the onset of the light phase allow proliferation of hematopoietic cells and contribute to their egress into the circulation [44].
The exit of leukocytes from the circulation to the tissues is facilitated by coordinated interactions between adhesion molecules on the endothelium and the surface of leukocytes [45]. In general, rhythmic expression of adhesion molecules, such as intercellular adhesion molecule 1 (ICAM1), vascular cell adhesion molecule 1 (VCAM1), and selectins on endothelial cells promotes time-of-day-dependent leukocyte transmigration into the lymphoid and non-lymphoid tissues [35].
Susceptibility of the immune system to bacterial, viral, and parasitic infections varies across 24 h [46]. One of the first evidence was provided by the experiment, in which mice were administrated a lethal dose of lipopolysaccharide (LPS). An immune challenge given at the end of the rest period led to a mortality rate of 80%, whereas the same LPS dose given in the middle of the active period resulted in a mortality rate of only about 20% [47]. A subsequent study demonstrated that this time-of-day-dependent mortality rate following LPS administration correlates with the increased cytokine response at the end of the light phase (ZT11; ZT—Zeitgeber time) compared to the dark period (ZT19) [48]. Daily variation in susceptibility to inflammatory challenge has also been shown to correlate with nuclear factor kappa B (NF-κB) activation, as mice administrated with a toll-like receptor (TLR) 5 ligand in the middle of their passive phase (ZT6) displayed higher NF-κB activation compared to mice injected in their active phase (ZT18) [49].
Macrophages represent one of the main sources of pro-inflammatory cytokines, and their inflammatory response is controlled by the circadian clock [50]. Mouse peritoneal macrophages show higher LPS-induced expression of inflammatory cytokines, mainly interleukins Il-6, Il-12b, and chemokines Cxcl1 and C-C motif chemokine ligand 2 (Ccl2), when isolated at the end than at the beginning of the subjective passive phase [51]. Moreover, the rhythm of inflammatory monocytes Ly6Chigh in the blood corresponds with the time-of-day-dependent immune response to Listeria monocytogenes infection, reflected by higher levels of CCL2 in the serum and peritoneal fluid upon the induction of infection at ZT8 compared to ZT0 [52].
Neutrophil infiltration into the skeletal muscle was increased upon tumor necrosis factor-alpha (TNFα) challenge at the beginning of the active phase (ZT13) compared to the passive phase (ZT5), and positively correlated with greater Icam1 expression on the muscle endothelial cells [53]. On the other hand, in a mouse model of acute lung inflammation, the recruitment of neutrophils was promoted by the rhythmic release of chemokine CXCL5 from bronchiolar epithelial cells with higher levels upon LPS administration at the beginning of the resting phase compared to the active phase [41].

4. Effects of Circadian Disruption on Innate Immunity

Disruption of the circadian timing system can directly impact daily rhythms in the immune parameters, bearing potential negative consequences on the host’s ability to effectively cope with pathogens or tissue injury. Other complications can include a disturbed balance between anti- and pro-inflammatory mechanisms that can lead to either immunosuppression or promote a pro-inflammatory microenvironment favorable for chronic inflammatory diseases.

4.1. Genetic Circadian Disruption

Many important functional interactions between components of the molecular clock and the immune system have been revealed using animal models with the deletion of clock genes at the systemic or cell-specific levels [29][30]. These studies show that individual clock proteins can differ in their pro-inflammatory and anti-inflammatory properties.
BMAL1 is a central component of the mammalian molecular clock and plays a central role in circadian–immune interactions. Systemic deletion of Bmal1 eliminated circadian rhythmicity in the central pacemaker and periphery, resulting in a complete behavioral arrhythmicity [54][55]. Bmal1−/− mice also lost rhythmicity in the numbers of leukocytes and immature hematopoietic cells in the peripheral blood [43][56]. However, particularly models with targeted Bmal1 deletion in myeloid cell lineages have revealed an essential role of BMAL1 in the control of the time-of-day-dependent effector functions of monocytes and macrophages. Mice with deletion of Bmal1 in myeloid cells lost daily variability in circulating inflammatory Ly6Chigh monocytes, showing higher susceptibility to Listeria monocytogenes infection [52]. In another study, myeloid Bmal1-deficient mice on the Apoe−/− background showed increased recruitment of Ly6Chigh monocytes to atherosclerotic lesions with polarization to pro-inflammatory M1 macrophages [57]. In vitro experiments using bone marrow-derived macrophages (BMDMs) demonstrated that Bmal1 deficiency amplified acute inflammatory response to LPS, as was manifested by enhanced production of pro-inflammatory cytokines, suppressed antioxidant pathways. and increased reactive oxygen species levels [58][59]. Surprisingly, myeloid Bmal1 deficiency was also found to confer protection against pneumococcal infection that was attributed to increased motility and phagocytic activity of Bmal1 deficient macrophages [60]. In neutrophils, specific deletion of Bmal1 eliminated daily variability in granule content and neutrophil extracellular traps formation [61]. In general, the above-mentioned studies demonstrated the anti-inflammatory effects of BMAL1, which are probably mediated by CLOCK/BMAL1-dependent transcriptional regulation of genes containing E-box. For example, circadian monocyte trafficking is driven by time-of-day-dependent expression of chemokines (such as Ccl2), which are under the repressive transcriptional control of BMAL1 through recruitment of the polycomb repressive complex 2 [52].
In contrast to BMAL1, CLOCK protein has been shown to enhance NF-κB-mediated transcription and production of pro-inflammatory cytokines, and these effects were independent of the transactivation capacity of the CLOCK/BMAL1 complex on E-box containing promoters [49]. CLOCK was found in protein complexes with the p65 subunit of NF-κB and CLOCK overexpression was associated with enhanced NF-κB activation [49]. These findings were supported by reduced activation of NF-κB in response to immune challenge in mouse embryonic fibroblasts (MEFs), as well as hepatocytes of Clock-deficient mice compared to wild-type controls [49]. Similarly, reduced induction of pro-inflammatory cytokines upon LPS challenge has been observed in MEFs and BMDMs from Clock-mutant mice [62][63]. Moreover, day/night differences in inflammatory response to Salmonella infection were eliminated in the gut of Clock mutants [62].
Models with genetic disruption of clock genes Per and Cry have revealed distinct roles of these clock components in the regulation of immune functions. A study in Per1 mutant mice showed that they maintained circadian expression of perforin, granzyme B, and interferon-gamma (IFNγ) in splenic NK cells, though these rhythms were either attenuated or phase-shifted [64]. On the other hand, in Per2 mutant mice, serum IFNγ concentrations as well as mRNA and protein levels in the spleen completely lost daily rhythmicity [65]. These eliminated IFNγ rhythms can be translated to impaired IFNγ production by the splenic NK cells upon LPS challenge in Per2 mutant mice, and suppressed response in serum IFNγ and IL-1β levels [66]. Moreover, this study found an increased survival rate of Per2 mutants following a lethal dose of LPS compared to controls [66]. Mutation of Per2, disrupting the ability of PER2 to interact with other clock proteins, can also significantly affect TLR9-mediated immune responses, as peritoneal macrophages from Per2 mutants showed reduced expression of Tlr9 and decreased TLR9 ligand-induced production of TNFα and IL-12 [67]. In contrast to Per2 defects, the absence of Cry genes leads to a pro-inflammatory phenotype. In Cry1 and Cry2 double knockout fibroblasts, enhanced constitutive expression of pro-inflammatory factors was observed, and this was mediated by the constitutive activation of NF-κB and protein kinase A (PKA) signaling [68]. The proposed mechanism shows that CRY1 can inhibit PKA-mediated phosphorylation of p65 through binding to adenylyl cyclase and suppression of cyclic adenosine monophosphate levels [68]. In this study, Cry1−/−Cry2−/− mice exhibited not only enhanced basal expression of Il-6, Cxcl1, and inducible nitric oxide synthase (iNos) in the BMDMs but also elevated cytokine responses to LPS compared to wild-type animals [68].
The nuclear receptors REV-ERBα and RORα represent important regulatory components linking the circadian and immune systems and exert mostly anti-inflammatory effects. Peritoneal macrophages isolated from global REV-ERBα knockout mice (Rev-erbα−/−) displayed augmented pro-inflammatory response to LPS [51][69][70]. Simultaneously, the absence of circadian rhythmicity in LPS-induced IL-6 response was demonstrated in the cultured Rev-erbα−/− macrophages and in vivo upon endotoxin challenge in Rev-erbα−/− mice [51]. Furthermore, these studies showed that REV-ERBα is a direct transcriptional repressor of several pro-inflammatory genes, including Ccl2 and NOD-like receptor family pyrin domain containing 3 (Nlrp3), which contain RORE binding sites in their promoter regions [69][70]. Moreover, Rev-erbα−/− mice have been found to display exaggerated LPS-induced pulmonary inflammation [71], increased severity of dextran sulphate sodium (DSS)-induced colitis [69], as well as a neuroinflammatory phenotype with basal activation of microglia in the hippocampus [72]. Likewise, in mice with deficient Rorα expression (RORαsg/sg, staggerer mutants), several immune defects were described besides typical cerebellar neurodegeneration. For example, splenocytes isolated from these mice were more sensitive to LPS challenge, showing increased expression of pro-inflammatory cytokines compared to wild-type controls [73]. Moreover, similarly to Rev-erbα deficient mice, also RORαsg/sg mice showed increased susceptibility to LPS-induced lung inflammation, higher neutrophil numbers, and increased levels of pro-inflammatory cytokines (IL-1β, IL-6, and macrophage inflammatory protein 2) in the bronchoalveolar lavage compared to wild-type mice [74]. An anti-inflammatory action of RORα can occur through RORE-mediated up-regulation of inhibitor of NF-κB (IκBα) and reduced p65 nuclear translocation [75].
REV-ERBα can transcriptionally regulate and repress another circadian repressor NFIL3 [76], which is also implicated in numerous immune processes. Studies in NFIL3-deficient (Nfil3−/−) mice have shown a critical role of NFIL3 in the development of several types of immune cells, including CD8+ conventional dendritic cells [77], NK cells, as well as all other innate lymphoid cell lineages [78][79]. Later, NFIL3 was identified as an important regulator of macrophage responses via transcriptional repression of Il-12b [80]. Interestingly, the inflammatory response of macrophages has been shown to depend on the phase of circadian oscillations of NFIL3 and DBP, which competitively bind at the Il-12b enhancer [81]. Therefore, desynchronization of the molecular clock in the macrophage population can contribute to the heterogeneity of the inflammatory response [81].
Together, accumulating evidence indicates a complexity of circadian-immune crosstalk, highlighting diverse immunomodulatory effects of individual clock components, which are determined by transcription-dependent mechanisms, direct protein–protein interactions, or the phase of circadian oscillations.

4.2. Light-Phase Shifts

Shift work and jet lag represent frequent circadian challenges associated with a modern lifestyle that lead to desynchronization of the SCN and downstream oscillators with the external environment [82]. Shift work refers to work outside the regular daytime hours and involves non-standard work schedules, such as night shifts, early morning shifts, or rotating shifts, which are also associated with alterations in the sleep/wake cycle [83]. Reduced amplitude or disturbance of the key circadian rhythms, such as melatonin, cortisol, and body temperature, has been observed among shift workers [84]. Misalignment between endogenous circadian rhythms and the LD cycle in shift workers can also predispose to an increased risk of negative health outcomes, such as cancer, and metabolic and cardiovascular diseases [85][86]. Moreover, shift workers are at a higher risk of common respiratory infections, including cold, flu, or COVID-19 [87][88][89].
Circadian clocks have been studied as an important player in many aspects of cancer-immune cell interactions [90]. Experimental research has shown that circadian disruption induced by different jet lag models can accelerate tumor growth and the incidence of metastasis as compared to a normal lighting regime [91][92][93][94]. Innate lymphoid NK cells are an integral part of anti-tumor immunity and provide effective immune surveillance by destroying tumor cells [95]. This ability is ensured by a stable count of NK cells and their production of various cytolytic factors and cytokines, mainly perforin, granzyme B, and IFNγ [96][97]. In mice, chronic shifts in the LD cycle reduced the numbers of NK cells in the spleen and lungs [92] and attenuated their cytolytic activity through suppressed expression of CD107a, a sensitive indicator of NK cell cytotoxicity and degranulation [98]. Another study in rats showed that repeated phase advances of the LD cycle suppressed rhythmic cytotoxicity of splenic NK cells, and modified circadian expression of granzyme B, perforin, and IFNγ in NK cells [93]. In addition to NK cells, tumor progression is controlled by the tumor microenvironment, which contains a variety of immune cells with a tumor-promoting or tumor-suppressing phenotype [99]. In a melanoma mouse model, circadian disruption induced by CJL abolished daily variability and decreased the M1 (pro-inflammatory)/M2 (anti-inflammatory) macrophage ratio in the tumor, promoting immunosuppression of the tumor microenvironment [91]. These effects accelerated tumor growth, and were also associated with increased mortality [91]. Other studies found reduced survival in aged mice exposed to chronic phase-advances for 8 weeks [100] or even as a result of long-term exposure (for 85 weeks) to phase-advances in 4-day intervals [101]. Additionally, epigenetic changes are known to participate in carcinogenesis, and they can also have the potential to mediate deregulation of immune mechanisms induced by circadian disruption. For example, rats, experienced chronic circadian disruption, exhibited aberrant changes in the expression of several cancer-related microRNAs in mammary tissues and, these changes were associated with increased protein levels of pro-inflammatory transcription factors, phosphorylated NF-κB and STAT3 [102].

4.3. Dim ALAN

The advancement of lighting technologies, including the implementation of light-emitting diode (LED) technology, goes in parallel with increasing levels of light pollution [103]. Moreover, evening use of devices with light-emitting screens as well as the use of night lamps, especially for small children while sleeping, considerably contribute to unintentional exposure to ALAN [104].

Several experimental studies have demonstrated that ALAN can affect innate immune mechanisms, including inflammatory response. However, in most of these studies, the immune status was evaluated only at one time point, neglecting consequences on circadian rhythms in the immune system. Indeed, a recent study showed that rats exposed to dim ALAN (2 lx) for 5 weeks exhibited impaired daily variation of the main leukocyte subsets in the blood, especially monocytes and T cells [138]. Moreover, ALAN reduced blood monocyte counts and altered gene expression of macrophage marker Cd68 and chemokine Ccl2 in the kidney, indicating that weakened circadian control of circulating leukocyte numbers was associated with disturbed renal immune homeostasis [138].

Immune disbalance caused by ALAN is considered one of the key mechanisms that can promote a pro-inflammatory state or accelerate various pathologies. For example, mice exposed to either ALAN (5 lx) or an HFD for 4 weeks showed up-regulated expression of inflammatory markers Tnfα and macrophage-1 antigen (Mac-1) in white adipose tissue, while ALAN further potentiated HFD-induced inflammation [105]. In cancer research, dim ALAN has been shown to favor tumor growth, especially in models of mammary cancer [106][107]. C3H mice exposed to ALAN (5 lx) for 3 weeks and then injected with FM3A mammary carcinoma cells displayed earlier tumor onset and increased terminal tumor volume compared to tumor-bearing mice housed in the LD regime [106]. In another study in nude rats, chronic ALAN even with a very low light intensity of 0.2 lx accelerated mammary tumor growth [107].
Another process that can drive the impact of ALAN on the progression of diseases is the ability of ALAN to promote neuroinflammation. Exposure to ALAN for 4 weeks increased hippocampal Tnfα and Il-6 expression simultaneously with depression-like behavior in female Siberian hamsters (Phodopus sungorus) [108], and up-regulated Il-6 mRNA levels in the medulla of mice that concomitantly exhibited cold hyperalgesia and mechanical allodynia [109]. Moreover, mice that underwent global cerebral ischemia and were subsequently exposed to ALAN showed decreased survival associated with increased neuronal damage that was preceded by amplified neuroinflammation, compared to animals in the control regime [110].
Till now, the effects of ALAN on inflammatory response were examined only in a limited number of studies. In mice challenged with LPS following 4 weeks of dim ALAN (5 lx), exaggerated changes in body temperature, prolonged sickness responses, and elevated pro-inflammatory cytokine expression (Tnfα and Il-6) in microglia were found compared to controls [111]. Additionally, diminished bactericidal capacity of blood upon LPS challenge and reduced delayed-type hypersensitivity response was observed in Siberian hamsters exposed to dim ALAN compared to animals in the standard LD regime [112]. Interestingly, the opposite effects of ALAN were obtained in a diurnal rodent model, Nile grass rats (Arvicanthis niloticus), which exhibited enhanced delayed-type hypersensitivity response and elevated basal bactericidal capacity when exposed to ALAN for 3 weeks [113]. Thus, the currently available data demonstrate that ALAN affects the responsiveness of the immune system to challenges, but clearly more studies are needed to reveal potentially differential responses between diurnal and nocturnal mammals, and to evaluate whether immune responses are impacted by ALAN in a time-of-day-dependent manner. Moreover, surprisingly limited data are available on the effects of ALAN on innate immunity and inflammation in humans.

4.4. Constant Light

Exposure to LL and low-intensity ALAN are often considered interchangeable conditions. However, circadian disruption caused by LL differs from that induced by low-intensity ALAN in several ways [114]. In general, LL leads to the complete loss of locomotor activity rhythms [115], and this behavioral arrhythmicity develops as soon as one month after changed lighting conditions in rats [116][117]. In the master clock, LL causes desynchronization of SCN neurons [115] and reduces the amplitude of SCN neuronal activity rhythm [118], which is further attenuated by long-term LL exposure [119]. Suppressed nocturnal melatonin levels have been found under both LL and dim ALAN regimes [120][121] but corticosterone is arrhythmic in LL [122][123], and preserves its rhythmicity with decreased amplitude in the dim ALAN regime [124].
Circadian disruption induced by LL was shown to facilitate a pro-inflammatory state even under unchallenged conditions. Specifically, in rats, 4-week LL exposure up-regulated the expression of the pro-inflammatory markers Stat3, Il-17ra, and Il-1α in the colonic mucosa [116]. In another study, rats exposed to LL for 5 weeks displayed amplified plasma TNFα response and sickness symptoms, such as febrile reaction and food intake reduction, following LPS administration [117]. Interestingly, in rats, 24 h leukocyte rhythms in the circulation persisted 8 weeks after LL exposure, despite suppressed circadian rhythms in body temperature and locomotor activity [125]. Nevertheless, in the same study, prolonged LL exposure (for 11 and 16 weeks) did eliminate the circadian rhythm in blood leukocytes that was not restored even 16 weeks after re-synchronization in the LD regime [125]. In mice exposed to LL for 8 weeks, increased numbers of blood neutrophils and reduced numbers of lymphocytes were found together with an enhanced response of pro-inflammatory cytokines to LPS challenge [119]. Interestingly, these effects were transient, as no further changes were observed in mice after 24 weeks of the LL regime [119]. However, the study did not monitor the whole 24 h profile in white blood cells.

References

  1. Albrecht, U. Timing to perfection: The biology of central and peripheral circadian clocks. Neuron 2012, 74, 246–260.
  2. Golombek, D.A.; Rosenstein, R.E. Physiology of circadian entrainment. Physiol. Rev. 2010, 90, 1063–1102.
  3. Pilorz, V.; Astiz, M.; Heinen, K.O.; Rawashdeh, O.; Oster, H. The concept of coupling in the mammalian circadian clock network. J. Mol. Biol. 2020, 432, 3618–3638.
  4. Blume, C.; Garbazza, C.; Spitschan, M. Effects of light on human circadian rhythms, sleep and mood. Somnologie 2019, 23, 147–156.
  5. Gaston, K.J.; Bennie, J.; Davies, T.W.; Hopkins, J. The ecological impacts of nighttime light pollution: A mechanistic appraisal. Biol. Rev. 2013, 88, 912–927.
  6. Falchi, F.; Cinzano, P.; Duriscoe, D.; Kyba, C.C.; Elvidge, C.D.; Baugh, K.; Portnov, B.A.; Rybnikova, N.A.; Furgoni, R. The new world atlas of artificial night sky brightness. Sci. Adv. 2016, 2, e1600377.
  7. Kernbach, M.E.; Miller, C.; Alaasam, V.; Ferguson, S.; Francis, C.D. Introduction to the symposium: Effects of light pollution across diverse natural systems. Integr. Comp. Biol. 2021, 61, 1089–1097.
  8. Davies, T.W.; Smyth, T. Why artificial light at night should be a focus for global change research in the 21st century. Glob. Chang. Biol. 2018, 24, 872–882.
  9. Vetter, C. Circadian disruption: What do we actually mean? Eur. J. Neurosci. 2020, 51, 531–550.
  10. Lunn, R.M.; Blask, D.E.; Coogan, A.N.; Figueiro, M.G.; Gorman, M.R.; Hall, J.E.; Hansen, J.; Nelson, R.J.; Panda, S.; Smolensky, M.H.; et al. Health consequences of electric lighting practices in the modern world: A report on the National Toxicology Program’s workshop on shift work at night, artificial light at night, and circadian disruption. Sci. Total Environ. 2017, 607, 1073–1084.
  11. Stevens, R.G.; Brainard, G.C.; Blask, D.E.; Lockley, S.W.; Motta, M.E. Adverse health effects of nighttime lighting: Comments on American Medical Association policy statement. Am. J. Prev. Med. 2013, 45, 343–346.
  12. Yazdi, Z.; Sadeghniiat-Haghighi, K.; Loukzadeh, Z.; Elmizadeh, K.; Abbasi, M. Prevalence of sleep disorders and their impacts on occupational performance: A comparison between shift workers and nonshift workers. Sleep Disord. 2014, 2014, 870320.
  13. Ohayon, M.M.; Milesi, C. Artificial outdoor nighttime lights associate with altered sleep behavior in the American general population. Sleep 2016, 39, 1311–1320.
  14. Garcia-Saenz, A.; Sánchez de Miguel, A.; Espinosa, A.; Valentin, A.; Aragonés, N.; Llorca, J.; Amiano, P.; Martín Sánchez, V.; Guevara, M.; Capelo, R.; et al. Evaluating the association between artificial light-at-night exposure and breast and prostate cancer risk in Spain (MCC-Spain study). Environ. Health Perspect. 2018, 126, 047011.
  15. Wang, X.; Ji, A.; Zhu, Y.; Liang, Z.; Wu, J.; Li, S.Q.; Meng, S.; Zheng, X.Y.; Xie, L.P. A meta-analysis including dose-response relationship between night shift work and the risk of colorectal cancer. Oncotarget 2015, 6, 25046–25060.
  16. Sun, S.; Cao, W.; Ge, Y.; Ran, J.; Sun, F.; Zeng, Q.; Guo, M.; Huang, J.; Lee, R.S.-Y.; Tian, L.; et al. Outdoor light at night and risk of coronary heart disease among older adults: A prospective cohort study. Eur. Heart J. 2021, 42, 822–830.
  17. Vetter, C.; Devore, E.E.; Wegrzyn, L.R.; Massa, J.; Speizer, F.E.; Kawachi, I.; Rosner, B.; Stampfer, M.J.; Schernhammer, E.S. Association between rotating night shift work and risk of coronary heart disease among women. JAMA-J. Am. Med. Assoc. 2016, 315, 1726–1734.
  18. Koo, Y.S.; Song, J.-Y.; Joo, E.-Y.; Lee, H.-J.; Lee, E.; Lee, S.-K.; Jung, K.-Y. Outdoor artificial light at night, obesity, and sleep health: Cross-sectional analysis in the KoGES study. Chronobiol. Int. 2016, 33, 301–314.
  19. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832.
  20. Honma, S. The mammalian circadian system: A hierarchical multi-oscillator structure for generating circadian rhythm. J. Physiol. Sci. 2018, 68, 207–219.
  21. Welsh, D.K.; Takahashi, J.S.; Kay, S.A. Suprachiasmatic nucleus: Cell autonomy and network properties. Annu. Rev. Physiol. 2010, 72, 551–577.
  22. Lucas, R.J.; Peirson, S.N.; Berson, D.M.; Brown, T.M.; Cooper, H.M.; Czeisler, C.A.; Figueiro, M.G.; Gamlin, P.D.; Lockley, S.W.; O’Hagan, J.B.; et al. Measuring and using light in the melanopsin age. Trends Neurosci. 2014, 37, 1–9.
  23. Buijs, F.N.; León-Mercado, L.; Guzmán-Ruiz, M.; Guerrero-Vargas, N.N.; Romo-Nava, F.; Buijs, R.M. The circadian system: A regulatory feedback network of periphery and brain. Physiology 2016, 31, 170–181.
  24. Takahashi, J.S. Transcriptional architecture of the mammalian circadian clock. Nat. Rev. Genet. 2017, 18, 164–179.
  25. Reppert, S.M.; Weaver, D.R. Molecular analysis of mammalian circadian rhythms. Annu. Rev. Physiol. 2001, 63, 647–676.
  26. Alessandro, M.S.; Golombek, D.A.; Chiesa, J.J. Protein kinases in the photic signaling of the mammalian circadian clock. Yale J. Biol. Med. 2019, 92, 241–250.
  27. Ueda, H.R.; Hayashi, S.; Chen, W.; Sano, M.; Machida, M.; Shigeyoshi, Y.; Iino, M.; Hashimoto, S. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat. Genet. 2005, 37, 187–192.
  28. Preitner, N.; Damiola, F.; Luis Lopez, M.; Zakany, J.; Duboule, D.; Albrecht, U.; Schibler, U. The orphan nuclear receptor REV-ERBα controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 2002, 110, 251–260.
  29. Curtis, A.M.; Bellet, M.M.; Sassone-Corsi, P.; O’Neill, L.A. Circadian clock proteins and immunity. Immunity 2014, 40, 178–186.
  30. Hergenhan, S.; Holtkamp, S.; Scheiermann, C. Molecular interactions between components of the circadian clock and the immune system. J. Mol. Biol. 2020, 432, 3700–3713.
  31. Li, D.; Wu, M. Pattern recognition receptors in health and diseases. Signal Transduct. Target. Ther. 2021, 6, 291.
  32. Newton, K.; Dixit, V.M. Signaling in innate immunity and inflammation. Cold Spring Harb Perspect. Biol. 2012, 4, a006049.
  33. Abele, S.H.; Meadows, K.E.; Medeiros, D.; Silver, A.C. Time is on the immune system’s side, Yes it is. Yale J. Biol. Med. 2019, 92, 225–231.
  34. Waggoner, S.N. Circadian rhythms in immunity. Curr. Allergy Asthma Rep. 2020, 20, 2.
  35. He, W.; Holtkamp, S.; Hergenhan, S.M.; Kraus, K.; de Juan, A.; Weber, J.; Bradfield, P.; Grenier, J.M.P.; Pelletier, J.; Druzd, D.; et al. Circadian expression of migratory factors establishes lineage-specific signatures that guide the homing of leukocyte subsets to tissues. Immunity 2018, 49, 1175–1190.e7.
  36. Pelegrí, C.; Vilaplana, J.; Castellote, C.; Rabanal, M.; Franch, À.; Castell, M. Circadian rhythms in surface molecules of rat blood lymphocytes. Am. J. Physiol. Cell Physiol. 2003, 284, C67–C76.
  37. Haus, E.; Smolensky, M.H. Biologic rhythms in the immune system. Chronobiol. Int. 1999, 16, 581–622.
  38. Pick, R.; He, W.; Chen, C.-S.; Scheiermann, C. Time-of-day-dependent trafficking and function of leukocyte subsets. Trends Immunol. 2019, 40, 524–537.
  39. Casanova-Acebes, M.; Pitaval, C.; Weiss, L.A.; Nombela-Arrieta, C.; Chèvre, R.; A-González, N.; Kunisaki, Y.; Zhang, D.; van Rooijen, N.; Silberstein, L.E.; et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 2013, 153, 1025–1035.
  40. Druzd, D.; Matveeva, O.; Ince, L.; Harrison, U.; He, W.; Schmal, C.; Herzel, H.; Tsang, A.H.; Kawakami, N.; Leliavski, A.; et al. Lymphocyte circadian clocks control lymph node trafficking and adaptive immune responses. Immunity 2017, 46, 120–132.
  41. Gibbs, J.; Ince, L.; Matthews, L.; Mei, J.; Bell, T.; Yang, N.; Saer, B.; Begley, N.; Poolman, T.; Pariollaud, M.; et al. An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat. Med. 2014, 20, 919–926.
  42. Palomino-Segura, M.; Hidalgo, A. Circadian immune circuits. J. Exp. Med. 2020, 218, e20200798.
  43. Méndez-Ferrer, S.; Lucas, D.; Battista, M.; Frenette, P.S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature 2008, 452, 442–447.
  44. Kollet, O.; Vagima, Y.; D’Uva, G.; Golan, K.; Canaani, J.; Itkin, T.; Gur-Cohen, S.; Kalinkovich, A.; Caglio, G.; Medaglia, C.; et al. Physiologic corticosterone oscillations regulate murine hematopoietic stem/progenitor cell proliferation and CXCL12 expression by bone marrow stromal progenitors. Leukemia 2013, 27, 2006–2015.
  45. Yadav, R.; Larbi, K.Y.; Young, R.E.; Nourshargh, S. Migration of leukocytes through the vessel wall and beyond. Thromb. Haemost. 2003, 90, 598–606.
  46. Nobis, C.C.; Labrecque, N.; Cermakian, N. From immune homeostasis to inflammation, a question of rhythms. Curr. Opin. Physiol. 2018, 5, 90–98.
  47. Halberg, F.; Johnson, E.A.; Brown, B.W.; Bittner, J.J. Susceptibility rhythm to E. coli endotoxin and bioassay. Proc. Soc. Exp. Biol. Med. 1960, 103, 142–144.
  48. Marpegan, L.; Leone, M.J.; Katz, M.E.; Sobrero, P.M.; Bekinstein, T.A.; Golombek, D.A. Diurnal variation in endotoxin-induced mortality in mice: Correlation with proinflammatory factors. Chronobiol. Int. 2009, 26, 1430–1442.
  49. Spengler, M.L.; Kuropatwinski, K.K.; Comas, M.; Gasparian, A.V.; Fedtsova, N.; Gleiberman, A.S.; Gitlin, I.I.; Artemicheva, N.M.; Deluca, K.A.; Gudkov, A.V.; et al. Core circadian protein CLOCK is a positive regulator of NF-κB–mediated transcription. Proc. Natl. Acad. Sci. USA 2012, 109, E2457–E2465.
  50. Timmons, G.A.; O’Siorain, J.R.; Kennedy, O.D.; Curtis, A.M.; Early, J.O. Innate rhythms: Clocks at the center of monocyte and macrophage function. Front. Immunol. 2020, 11, 1743.
  51. Gibbs, J.E.; Blaikley, J.; Beesley, S.; Matthews, L.; Simpson, K.D.; Boyce, S.H.; Farrow, S.N.; Else, K.J.; Singh, D.; Ray, D.W.; et al. The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc. Natl. Acad. Sci. USA 2012, 109, 582.
  52. Nguyen, K.D.; Fentress, S.J.; Qiu, Y.; Yun, K.; Cox, J.S.; Chawla, A. Circadian gene Bmal1 regulates diurnal oscillations of Ly6Chi inflammatory monocytes. Science 2013, 341, 1483–1488.
  53. Scheiermann, C.; Kunisaki, Y.; Lucas, D.; Chow, A.; Jang, J.E.; Zhang, D.; Hashimoto, D.; Merad, M.; Frenette, P.S. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 2012, 37, 290–301.
  54. Bunger, M.K.; Wilsbacher, L.D.; Moran, S.M.; Clendenin, C.; Radcliffe, L.A.; Hogenesch, J.B.; Simon, M.C.; Takahashi, J.S.; Bradfield, C.A. Mop3 is an essential component of the master circadian pacemaker in mammals. Cell 2000, 103, 1009–1017.
  55. Baggs, J.E.; Price, T.S.; DiTacchio, L.; Panda, S.; FitzGerald, G.A.; Hogenesch, J.B. Network features of the mammalian circadian clock. PLoS Biol. 2009, 7, e1000052.
  56. Stenzinger, M.; Karpova, D.; Unterrainer, C.; Harenkamp, S.; Wiercinska, E.; Hoerster, K.; Pfeffer, M.; Maronde, E.; Bonig, H. Hematopoietic-extrinsic cues dictate circadian redistribution of mature and immature hematopoietic cells in blood and spleen. Cells 2019, 8, 1033.
  57. Huo, M.; Huang, Y.; Qu, D.; Zhang, H.; Wong, W.T.; Chawla, A.; Huang, Y.; Tian, X.Y. Myeloid Bmal1 deletion increases monocyte recruitment and worsens atherosclerosis. FASEB J. 2017, 31, 1097–1106.
  58. Curtis, A.M.; Fagundes, C.T.; Yang, G.; Palsson-McDermott, E.M.; Wochal, P.; McGettrick, A.F.; Foley, N.H.; Early, J.O.; Chen, L.; Zhang, H.; et al. Circadian control of innate immunity in macrophages by miR-155 targeting Bmal1. Proc. Natl. Acad. Sci. USA 2015, 112, 7231–7236.
  59. Early, J.O.; Menon, D.; Wyse, C.A.; Cervantes-Silva, M.P.; Zaslona, Z.; Carroll, R.G.; Palsson-McDermott, E.M.; Angiari, S.; Ryan, D.G.; Corcoran, S.E.; et al. Circadian clock protein BMAL1 regulates IL-1β in macrophages via NRF2. Proc. Natl. Acad. Sci. USA 2018, 115, E8460–E8468.
  60. Kitchen, G.B.; Cunningham, P.S.; Poolman, T.M.; Iqbal, M.; Maidstone, R.; Baxter, M.; Bagnall, J.; Begley, N.; Saer, B.; Hussell, T.; et al. The clock gene Bmal1 inhibits macrophage motility, phagocytosis, and impairs defense against pneumonia. Proc. Natl. Acad. Sci. USA 2020, 117, 1543–1551.
  61. Adrover, J.M.; Aroca-Crevillén, A.; Crainiciuc, G.; Ostos, F.; Rojas-Vega, Y.; Rubio-Ponce, A.; Cilloniz, C.; Bonzón-Kulichenko, E.; Calvo, E.; Rico, D.; et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat. Immunol. 2020, 21, 135–144.
  62. Bellet, M.M.; Deriu, E.; Liu, J.Z.; Grimaldi, B.; Blaschitz, C.; Zeller, M.; Edwards, R.A.; Sahar, S.; Dandekar, S.; Baldi, P.; et al. Circadian clock regulates the host response to Salmonella. Proc. Natl. Acad. Sci. USA 2013, 110, 9897–9902.
  63. Bellet, M.M.; Zocchi, L.; Sassone-Corsi, P. The RelB subunit of NFκB acts as a negative regulator of circadian gene expression. Cell Cycle 2012, 11, 3304–3311.
  64. Logan, R.W.; Wynne, O.; Levitt, D.; Price, D.; Sarkar, D.K. Altered circadian expression of cytokines and cytolytic factors in splenic natural killer cells of Per1−/− mutant mice. J. Interferon Cytokine Res. 2013, 33, 108–114.
  65. Arjona, A.; Sarkar, D.K. The circadian gene mPer2 regulates the daily rhythm of IFN-gamma. J. Interferon Cytokine Res. 2006, 26, 645–649.
  66. Liu, J.G.; Mankani, G.; Shi, X.Y.; Meyer, M.; Cunningham-Runddles, S.; Ma, X.J.; Sun, Z.S. The circadian clock Period 2 gene regulates gamma interferon production of NK cells in host response to lipopolysaccharide-induced endotoxic shock. Infect. Immun. 2006, 74, 4750–4756.
  67. Silver, A.C.; Arjona, A.; Walker, W.E.; Fikrig, E. The circadian clock controls toll-like receptor 9-mediated innate and adaptive immunity. Immunity 2012, 36, 251–261.
  68. Narasimamurthy, R.; Hatori, M.; Nayak, S.K.; Liu, F.; Panda, S.; Verma, I.M. Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc. Natl. Acad. Sci. USA 2012, 109, 12662–12667.
  69. Wang, S.; Lin, Y.; Yuan, X.; Li, F.; Guo, L.; Wu, B. REV-ERBα integrates colon clock with experimental colitis through regulation of NF-κB/NLRP3 axis. Nat. Commun. 2018, 9, 4246.
  70. Sato, S.; Sakurai, T.; Ogasawara, J.; Takahashi, M.; Izawa, T.; Imaizumi, K.; Taniguchi, N.; Ohno, H.; Kizaki, T. A circadian clock gene, Rev-erbα, modulates the inflammatory function of macrophages through the negative regulation of Ccl2 expression. J. Immunol. 2014, 192, 407–417.
  71. Pariollaud, M.; Gibbs, J.E.; Hopwood, T.W.; Brown, S.; Begley, N.; Vonslow, R.; Poolman, T.; Guo, B.; Saer, B.; Jones, D.H.; et al. Circadian clock component REV-ERBα controls homeostatic regulation of pulmonary inflammation. J. Clin. Investig. 2018, 128, 2281–2296.
  72. Griffin, P.; Dimitry, J.M.; Sheehan, P.W.; Lananna, B.V.; Guo, C.; Robinette, M.L.; Hayes, M.E.; Cedeño, M.R.; Nadarajah, C.J.; Ezerskiy, L.A.; et al. Circadian clock protein Rev-erbα regulates neuroinflammation. Proc. Natl. Acad. Sci. USA 2019, 116, 5102–5107.
  73. Kopmels, B.; Mariani, J.; Delhaye-Bouchaud, N.; Audibert, F.; Fradelizi, D.; Wollman, E.E. Evidence for a hyperexcitability state of staggerer mutant mice macrophages. J. Neurochem. 1992, 58, 192–199.
  74. Stapleton, C.M.; Jaradat, M.; Dixon, D.; Kang, H.S.; Kim, S.-C.; Liao, G.; Carey, M.A.; Cristiano, J.; Moorman, M.P.; Jetten, A.M. Enhanced susceptibility of staggerer (RORαsg/sg) mice to lipopolysaccharide-induced lung inflammation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005, 289, L144–L152.
  75. Delerive, P.; Monté, D.; Dubois, G.; Trottein, F.; Fruchart-Najib, J.; Mariani, J.; Fruchart, J.-C.; Staels, B. The orphan nuclear receptor RORα is a negative regulator of the inflammatory response. EMBO Rep. 2001, 2, 42–48.
  76. Duez, H.; van der Veen, J.N.; Duhem, C.; Pourcet, B.; Touvier, T.; Fontaine, C.; Derudas, B.; Baugé, E.; Havinga, R.; Bloks, V.W.; et al. Regulation of bile acid synthesis by the nuclear receptor Rev-erbα. Gastroenterology 2008, 135, 689–698.
  77. Kashiwada, M.; Pham, N.-L.L.; Pewe, L.L.; Harty, J.T.; Rothman, P.B. NFIL3/E4BP4 is a key transcription factor for CD8α+ dendritic cell development. Blood 2011, 117, 6193–6197.
  78. Gascoyne, D.M.; Long, E.; Veiga-Fernandes, H.; de Boer, J.; Williams, O.; Seddon, B.; Coles, M.; Kioussis, D.; Brady, H.J.M. The basic leucine zipper transcription factor E4BP4 is essential for natural killer cell development. Nat. Immunol. 2009, 10, 1118–1124.
  79. Geiger, T.L.; Abt, M.C.; Gasteiger, G.; Firth, M.A.; O’Connor, M.H.; Geary, C.D.; O’Sullivan, T.E.; van den Brink, M.R.; Pamer, E.G.; Hanash, A.M.; et al. Nfil3 is crucial for development of innate lymphoid cells and host protection against intestinal pathogens. J. Exp. Med. 2014, 211, 1723–1731.
  80. Kobayashi, T.; Matsuoka, K.; Sheikh, S.Z.; Elloumi, H.Z.; Kamada, N.; Hisamatsu, T.; Hansen, J.J.; Doty, K.R.; Pope, S.D.; Smale, S.T.; et al. NFIL3 is a regulator of IL-12 p40 in macrophages and mucosal immunity. J. Immunol. 2011, 186, 4649.
  81. Allen, N.C.; Philip, N.H.; Hui, L.; Zhou, X.; Franklin, R.A.; Kong, Y.; Medzhitov, R. Desynchronization of the molecular clock contributes to the heterogeneity of the inflammatory response. Sci. Signal. 2019, 12, eaau1851.
  82. Vosko, A.M.; Colwell, C.S.; Avidan, A.Y. Jet lag syndrome: Circadian organization, pathophysiology, and management strategies. Nat. Sci. Sleep 2010, 2, 187–198.
  83. Wickwire, E.M.; Geiger-Brown, J.; Scharf, S.M.; Drake, C.L. Shift work and shift work sleep disorder: Clinical and organizational perspectives. Chest 2017, 151, 1156–1172.
  84. Boivin, D.B.; Boudreau, P.; Kosmadopoulos, A. Disturbance of the circadian system in shift work and its health impact. J. Biol. Rhythm 2021, 37, 3–28.
  85. Hemmer, A.; Mareschal, J.; Dibner, C.; Pralong, J.A.; Dorribo, V.; Perrig, S.; Genton, L.; Pichard, C.; Collet, T.-H. The effects of shift work on cardio-metabolic diseases and eating patterns. Nutrients 2021, 13, 4178.
  86. Wang, X.S.; Armstrong, M.E.G.; Cairns, B.J.; Key, T.J.; Travis, R.C. Shift work and chronic disease: The epidemiological evidence. Occup. Med. 2011, 61, 78–89.
  87. Loef, B.; van Baarle, D.; van der Beek, A.J.; Sanders, E.A.M.; Bruijning-Verhagen, P.; Proper, K.I. Shift work and respiratory infections in health-care workers. Am. J. Epidemiol. 2019, 188, 509–517.
  88. Mohren, D.C.L.; Jansen, N.W.H.; Kant, I.; Galama, J.M.D.; van den Brandt, P.A.; Swaen, G.M.H. Prevalence of common infections among employees in different work schedules. J. Occup. Environ. Med. 2002, 44, 1003–1011.
  89. Fatima, Y.; Bucks, R.S.; Mamun, A.A.; Skinner, I.; Rosenzweig, I.; Leschziner, G.; Skinner, T.C. Shift work is associated with increased risk of COVID-19: Findings from the UK Biobank cohort. J. Sleep Res. 2021, 30, e13326.
  90. Zhang, Z.; Zeng, P.; Gao, W.; Zhou, Q.; Feng, T.; Tian, X. Circadian clock: A regulator of the immunity in cancer. Cell Commun. Signal. 2021, 19, 37.
  91. Aiello, I.; Fedele, M.L.M.; Román, F.; Marpegan, L.; Caldart, C.; Chiesa, J.J.; Golombek, D.A.; Finkielstein, C.V.; Paladino, N. Circadian disruption promotes tumor-immune microenvironment remodeling favoring tumor cell proliferation. Sci. Adv. 2020, 6, eaaz4530.
  92. Zeng, X.; Liang, C.; Yao, J. Chronic shift-lag promotes NK cell ageing and impairs immunosurveillance in mice by decreasing the expression of CD122. J. Cell. Mol. Med. 2020, 24, 14583–14595.
  93. Logan, R.W.; Zhang, C.; Murugan, S.; O’Connell, S.; Levitt, D.; Rosenwasser, A.M.; Sarkar, D.K. Chronic shift-lag alters the circadian clock of NK cells and promotes lung cancer growth in rats. J. Immunol. 2012, 188, 2583–2591.
  94. Filipski, E.; Delaunay, F.; King, V.M.; Wu, M.-W.; Claustrat, B.; Gréchez-Cassiau, A.; Guettier, C.; Hastings, M.H.; Francis, L.V. Effects of chronic jet lag on tumor progression in mice. Cancer Res. 2004, 64, 7879–7885.
  95. Pierce, S.; Geanes, E.S.; Bradley, T. Targeting natural killer cells for improved immunity and control of the adaptive immune response. Front. Cell. Infect. Microbiol. 2020, 10, 231.
  96. Kim, S.; Iizuka, K.; Aguila, H.L.; Weissman, I.L.; Yokoyama, W.M. In vivo natural killer cell activities revealed by natural killer cell-deficient mice. Proc. Natl. Acad. Sci. USA 2000, 97, 2731–2736.
  97. Paul, S.; Lal, G. The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front. Immunol. 2017, 8, 1124.
  98. Aktas, E.; Kucuksezer, U.C.; Bilgic, S.; Erten, G.; Deniz, G. Relationship between CD107a expression and cytotoxic activity. Cell. Immunol. 2009, 254, 149–154.
  99. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437.
  100. Davidson, A.J.; Sellix, M.T.; Daniel, J.; Yamazaki, S.; Menaker, M.; Block, G.D. Chronic jet-lag increases mortality in aged mice. Curr. Biol. 2006, 16, R914–R916.
  101. Inokawa, H.; Umemura, Y.; Shimba, A.; Kawakami, E.; Koike, N.; Tsuchiya, Y.; Ohashi, M.; Minami, Y.; Cui, G.; Asahi, T.; et al. Chronic circadian misalignment accelerates immune senescence and abbreviates lifespan in mice. Sci. Rep. 2020, 10, 2569.
  102. Kochan, D.Z.; Ilnytskyy, Y.; Golubov, A.; Deibel, S.H.; McDonald, R.J.; Kovalchuk, O. Circadian disruption-induced microRNAome deregulation in rat mammary gland tissues. Oncoscience 2015, 2, 428–442.
  103. Sánchez de Miguel, A.; Bennie, J.; Rosenfeld, E.; Dzurjak, S.; Gaston, K.J. First estimation of global trends in nocturnal power emissions reveals acceleration of light pollution. Remote Sens. 2021, 13, 3311.
  104. Cajochen, C.; Frey, S.; Anders, D.; Späti, J.; Bues, M.; Pross, A.; Mager, R.; Wirz-Justice, A.; Stefani, O. Evening exposure to a light-emitting diodes (LED)-backlit computer screen affects circadian physiology and cognitive performance. J. Appl. Physiol. 2011, 110, 1432–1438.
  105. Fonken, L.K.; Lieberman, R.A.; Weil, Z.M.; Nelson, R.J. Dim light at night exaggerates weight gain and inflammation associated with a high-fat diet in male mice. Endocrinology 2013, 154, 3817–3825.
  106. Walker, W.H.; Kvadas, R.M.; May, L.E.; Liu, J.A.; Bumgarner, J.R.; Walton, J.C.; DeVries, A.C.; Dauchy, R.T.; Blask, D.E.; Nelson, R.J. Artificial light at night reduces anxiety-like behavior in female mice with exacerbated mammary tumor growth. Cancers 2021, 13, 4860.
  107. Blask, D.E.; Dauchy, R.T.; Dauchy, E.M.; Mao, L.; Hill, S.M.; Greene, M.W.; Belancio, V.P.; Sauer, L.A.; Davidson, L. Light exposure at night disrupts host/cancer circadian regulatory dynamics: Impact on the Warburg effect, lipid signaling and tumor growth prevention. PLoS ONE 2014, 9, e102776.
  108. Bedrosian, T.A.; Weil, Z.M.; Nelson, R.J. Chronic dim light at night provokes reversible depression-like phenotype: Possible role for TNF. Mol. Psychiatry 2013, 18, 930–936.
  109. Bumgarner, J.R.; Walker, W.H.; Liu, J.A.; Walton, J.C.; Nelson, R.J. Dim light at night exposure induces cold hyperalgesia and mechanical allodynia in male mice. Neuroscience 2020, 434, 111–119.
  110. Fonken, L.K.; Bedrosian, T.A.; Zhang, N.; Weil, Z.M.; DeVries, A.C.; Nelson, R.J. Dim light at night impairs recovery from global cerebral ischemia. Exp. Neurol. 2019, 317, 100–109.
  111. Fonken, L.K.; Weil, Z.M.; Nelson, R.J. Mice exposed to dim light at night exaggerate inflammatory responses to lipopolysaccharide. Brain Behav. Immun. 2013, 34, 159–163.
  112. Bedrosian, T.A.; Fonken, L.K.; Walton, J.C.; Nelson, R.J. Chronic exposure to dim light at night suppresses immune responses in Siberian hamsters. Biol. Lett. 2011, 7, 468–471.
  113. Fonken, L.K.; Kitsmiller, E.; Smale, L.; Nelson, R.J. Dim nighttime light impairs cognition and provokes depressive-like responses in a diurnal rodent. J. Biol. Rhythm 2012, 27, 319–327.
  114. Rumanova, V.S.; Okuliarova, M.; Zeman, M. Differential effects of constant light and dim light at night on the circadian control of metabolism and behavior. Int. J. Mol. Sci. 2020, 21, 5478.
  115. Ohta, H.; Yamazaki, S.; McMahon, D.G. Constant light desynchronizes mammalian clock neurons. Nat. Neurosci. 2005, 8, 267–269.
  116. Polidarová, L.; Houdek, P.; Sumová, A. Chronic disruptions of circadian sleep regulation induce specific proinflammatory responses in the rat colon. Chronobiol. Int. 2017, 34, 1273–1287.
  117. Guerrero-Vargas, N.N.; Navarro-Espíndola, R.; Guzmán-Ruíz, M.A.; Basualdo, M.D.C.; Espitia-Bautista, E.; López-Bago, A.; Lascurain, R.; Córdoba-Manilla, C.; Buijs, R.M.; Escobar, C. Circadian disruption promotes tumor growth by anabolic host metabolism; experimental evidence in a rat model. BMC Cancer 2017, 17, 625.
  118. Coomans, C.P.; van den Berg, S.A.A.; Houben, T.; van Klinken, J.-B.; van den Berg, R.; Pronk, A.C.M.; Havekes, L.M.; Romijn, J.A.; van Dijk, K.W.; Biermasz, N.R.; et al. Detrimental effects of constant light exposure and high-fat diet on circadian energy metabolism and insulin sensitivity. FASEB J. 2013, 27, 1721–1732.
  119. Lucassen, E.A.; Coomans, C.P.; van Putten, M.; de Kreij, S.R.; van Genugten, J.H.L.T.; Sutorius, R.P.M.; de Rooij, K.E.; van der Velde, M.; Verhoeve, S.L.; Smit, J.W.A.; et al. Environmental 24-hr cycles are essential for health. Curr. Biol. 2016, 26, 1843–1853.
  120. Báez-Ruiz, A.; Guerrero-Vargas, N.N.; Cázarez-Márquez, F.; Sabath, E.; Basualdo, M.d.C.; Salgado-Delgado, R.; Escobar, C.; Buijs, R.M. Food in synchrony with melatonin and corticosterone relieves constant light disturbed metabolism. J. Endocrinol. 2017, 235, 167–178.
  121. Gale, J.E.; Cox, H.I.; Qian, J.Y.; Block, G.D.; Colwell, C.S.; Matveyenko, A.V. Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction. J. Biol. Rhythm 2011, 26, 423–433.
  122. Dauchy, R.T.; Dauchy, E.M.; Tirrell, R.P.; Hill, C.R.; Davidson, L.K.; Greene, M.W.; Tirrell, P.C.; Wu, J.; Sauer, L.A.; Blask, D.E. Dark-phase light contamination disrupts circadian rhythms in plasma measures of endocrine physiology and metabolism in rats. Comp. Med. 2010, 60, 348–356.
  123. Claustrat, B.; Valatx, J.L.; Harthé, C.; Brun, J. Effect of constant light on prolactin and corticosterone rhythms evaluated using a noninvasive urine sampling protocol in the rat. Horm. Metab. Res. 2008, 40, 398–403.
  124. Okuliarova, M.; Dzirbikova, Z.; Rumanova, V.S.; Foppen, E.; Kalsbeek, A.; Zeman, M. Disrupted circadian control of hormonal rhythms and anticipatory thirst by dim light at night. Neuroendocrinology 2022, 112, 1116–1128.
  125. Deprés-Brummer, P.; Bourin, P.; Pages, N.; Metzger, G.; Lévi, F. Persistent T lymphocyte rhythms despite suppressed circadian clock outputs in rats. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1997, 273, R1891–R1899.
More
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 344
Revisions: 2 times (View History)
Update Date: 28 Nov 2022
1000/1000
Video Production Service