The Circadian Immune System: Review
The Circadian Immune System: Review
The Circadian Immune System: Review
INTRODUCTION in the gene locus of the negative regulators Per1/2/3 and Cry1/2,
The circadian clock uses rhythmic environmental cues, such as light inducing their expression (15). Once expressed, PER and CRY also
and food intake, to establish approximately 24-hour rhythms in the form a heterodimeric transcription factor complex that inhibits the
vast majority of physiological processes. These biological cycles expression of BMAL1 and CLOCK until concentrations of the re-
prepare organisms better to encounter regular, recurring events in pressors are low enough to restart the cycle (15–17). In addition, the
their environment. The immune system is no exception. It is highly REV-ERB nuclear receptor / [nuclear receptor subfamily 1
circadian-regulated and exhibits 24-hour rhythmicity both at steady group D (NR1D); encoded by Nr1d1/2] and the retinoic acid receptor
Table 1. Alteration of circadian clock components in mouse strains and cell lines and their physiological relevance. NP-KLH, 4-hydroxy-3-
nitrophenylacetyl hapten conjugated to KLH (keyhole limpet hemocyanin); NP-CGG, chicken gamma globulin; KO, knockout; iPSCs, induced pluripotent stem
cells; DKO, double KO.
Cell type Mouse strain/cells Experimental model Findings Ref
Time-dependent, endotoxic shock–induced mortality in
Monocyte/
Lyz2cre:Bmal1flox LPS-induced inflammation mice (peak at ZT8) is independent of Bmal1 expression (24)
macrophage
in macrophages, as well as LD cycles.
Bmal1–Hif-1a regulates mitochondrial metabolism in
inflammatory macrophages in the scenario of
Lyz2cre:Bmal1flox M1-stimulated macrophages (32)
LPS-stimulated BMDMs, macrophages from cell-specific
knockout mice, and B16-F10–challenged mice.
Bmal1 deletion enhances macrophage motility and
Lyz2cre:Bmal1flox Cx3cr1cre:Bmal1flox S. pneumoniae (30)
phagocytosis, causing increased antibacterial immunity.
Deletion of Bmal1 abrogates diurnal variations in the
Lyz2cre:Bmal1flox L. monocytogenes numbers of Ly6Chi monocytes in blood, spleen, and (43)
bone marrow during L. monocytogenes infection.
(ZT12), these rhythms are lost in REV-ERB–deficient mice. This the factors that control rhythmic neutrophil infiltration into the
phenomenon was shown to be driven by REV-ERB expression in lungs after LPS challenge.
immune cells using bone marrow chimeric mice. Wild-type mice that
received REV-ERB–deficient bone marrow lost the time-of-day Macrophages and monocytes
difference in neutrophil infiltration, whereas those receiving REV- Among immune cell types, the circadian rhythms in macro-
ERB–sufficient bone marrow retained rhythms after LPS stimula- phages and monocytes have been the most studied to date (Table 1).
tion. Thus, these results indicate that leukocyte REV-ERB is one of In an S. pneumoniae infection model, the phagocytic activity of
macrophages was shown to be regulated by BMAL1 (30). Deletion inactivate NF-B and the NLRP3 inflammasome in macrophages
of Bmal1 in both Lyz2cre:Bmal1flox and Cx3cr1cre:Bmal1flox mice (39). Furthermore, earlier studies reported that REV-ERB can also
showed a strong protective effect, rescuing animals from severe regulate the expression of the cytokine IL-6 and the CC motif
bacteremia and weight loss. Further in vivo and ex vivo experi- chemokine ligand 2 (CCL2) in macrophages (40, 41), as well
ments determined phagocytic function to be enhanced in Bmal1- as CCL2 in microglial cells (42). These data implicate REV-ERB
deficient macrophages because Bmal1 deletion in macrophages as an important mediator of anti-inflammatory roles of the clock
alters the actin cytoskeletal organization and enhances overall anti- machinery.
bacterial function (30). These data suggest that BMAL1 is a strong With respect to monocytes, in a sterile thioglycollate peritonitis
regulator of macrophage morphology and motility. model and after infection with Listeria monocytogenes, the recruit-
An additional key element affecting the inflammatory status of ment of Ly6Chigh inflammatory monocytes to the inflamed site was
macrophages appears to be the circadian regulation of cellular me- demonstrated to exhibit a diurnal variation, regulated by the myeloid
tabolism (31). A recent study showed BMAL1 signaling to regulate cell–intrinsic clock (43). Mice infected at ZT8 (“afternoon”) showed
mitochondrial metabolism in M1 macrophages upon stimulation better control of L. monocytogenes bacterial spread compared with
with interferon- (IFN-) and LPS (32). Bmal1-deficient macro- ZT0 (“morning”) infected mice, with increased levels of the cyto-
phages exhibited dramatically reduced oxygen consumption; on the kines and chemokines IL-1, TNF-, IFN-, and CCL2. Mice with
other hand, succinate dehydrogenase–mediated production of reac- Bmal1-deficient myeloid cell subsets (Lyz2cre:Bmal1flox) were more
tive oxygen species was enhanced compared with control cells. This susceptible to L. monocytogenes, showing reduced survival and an in-
indicates that BMAL1 promotes mitochondrial function. Mechanis- crease in plasma of the same proinflammatory cytokines and chemo-
tically, these effects may be mediated by a posttranscriptional meta- kines (43). Deletion of Bmal1 also impaired the diurnal oscillation
bolic regulation (33). Comprehensive profiling of the transcriptome of Ly6Chigh monocyte numbers in blood, spleen, and bone marrow,
neutrophils, which appear to play much more prominent roles than was shown to promote TH17 differentiation. It does this by inhibit-
previously anticipated in various organs even when their numbers ing the expression of nuclear factor, interleukin 3 regulated (NFIL3),
present in those tissues are low. which itself is a repressor of RORt (encoded by Rorc)—a positive
regulator of TH17 differentiation (57–63). Rhythmicity in these
Dendritic cells transcription factors modulates TH17 differentiation. Thus, TH17
Dendritic cells (DCs) are phagocytic antigen-presenting cells (APCs) that development is favored during the mouse behavioral rest phase
directly link the innate immune system to the adaptive immune because of high NFIL3 activity during the active phase. Disrupting
system by collecting antigen at distal sites and presenting it to circadian rhythms through the use of a jetlag model increased the
cells of the adaptive immune system in immune organs. A recent frequency of TH17 cells and made mice more susceptible to dextran
study found that the migration of mouse DCs into afferent lym- sulfate sodium (DSS)–induced colitis (57). The numbers of TH1
phatic vessels of the skin occurs in a rhythmic manner, with a cells in the intestine were also reduced in Clock mutant mice, sug-
peak around ZT7 (“day”) and a trough at ZT19 (night) (Fig. 1) (48). gesting that the circadian clock also plays a role in intestinal TH1
This rhythmic migration is driven by diurnal expression of several differentiation in addition to TH17 cells (57). Because these tran-
adhesion molecules on lymphatic endothelial cells (LECs) such as scription factors mediating peripheral T cell differentiation are also
CCL21, lymphatic vessel endothelial receptor 1 (LYVE-1), CD99, clock genes, fine-tuning T cell phenotypes in peripheral sites may
and junctional adhesion molecule A (JAM-A), as well as DC- be affected by the circadian rhythm. However, future studies are
expressed CCR7. Synchronization of bone marrow–derived DCs required to assess a potential time-of-day–dependent role for RORt
(BMDCs) and lineage-specific ablation of BMAL1 function in conven- and ROR in stabilizing TH17 commitment and to address whether
tional DCs (Clec9acre:Bmal1flox), LECs (Prox1creERT2:Bmal1flox), or T cell–specific ablation of BMAL1 changes the capacity for TH17
endothelial cells (Cdh5creERT2:Bmal1flox) implicated both DC- differentiation in a circadian manner.
loaded DCs produce more OVA-specific T cells in the spleen when psoriatic lesions by DCs and macrophages, which stimulates IL-23R–
immunized in the day (ZT6) versus at night (ZT18) (10). Mice vac- expressing cells to release IL-22 and IL-17, further exacerbating the
cinated with OVA-loaded DCs during the day have a significantly disease (76). ChIP assays demonstrated that CLOCK binds to the
increased percentage of OVA-specific, IFN-producing effector promoter region of IL-23R in T cells (75). On the other hand, mice
T cells in the spleen 7 days after vaccination compared with vaccina- with a loss-of-function mutation in Clock (Clock 19) lost rhythmic
tion during the night (CT18) (49). These mice further exhibit a sig- IL-23R expression on T cells, displayed reduced IL-22 expression,
nificantly lower bacterial load when challenged with a lethal dose of and exhibited significantly reduced psoriasis severity compared
L. monocytogenes 7 days after DC-OVA vaccination (49), corrobo- with wild-type mice (75). REV-ERB has also been shown to con-
rating previous observations where L. monocytogenes–infected mice trol inflammatory cytokine production in T cells in the context
of IMQ-induced psoriasis (77). REV-ERB–deficient mice exhibited and E-selectin expression in bone marrow sinusoids, as well as
significantly increased numbers of IL-17+ T cells. In contrast, stim- ICAM-1 and CCL2 expression in skeletal muscle endothelial cells
ulating REV-ERB with the synthetic agonist SR9009 suppressed both at the mRNA and at the protein level (72). Removal of nerve-
IL-17 production by T cells and reduces the severity of IMQ- derived adrenergic signals via surgical, chemical, or genetic denerva-
induced psoriasis (77). These data point to a crucial role for clock tran- tion abrogates the circadian differences in leukocyte migration to the
scription factors in autoimmune diseases. bone marrow, the muscle, and the LN (68, 70, 72). An additional study
There is evidence that long-term disruption of an organism’s provided mechanistic insight into how adrenergic signaling regulates
rhythmic environment affects adaptive immune cell function. Aged two distinct peaks of HSPC activity, in the blood and bone marrow,
mice who undergo chronic jetlag (CJL) display a reduced life span across the 24-hour period (83). The first peak, driven by the onset of
(78), and extended CJL (~85 weeks) starting from a young age exac- light, increases norepinephrine levels. This triggers HSPC prolifera-
erbates immune senescence and mortality (79). CJL resulted in tion and differentiation that, together with increased vascular perme-
significantly increased numbers of PD-1 +CD44 +CD4 + T cells, ability, lead to blood replenishment. The onset of darkness induces a
PD-1+CD44+CD153+CD4+ T cells, T follicular helper cells (Tfh), and second peak in HSPC activity via increased TNF- secretion, which
T regulatory cells (Tregs) in the spleen—all phenotypes associated mediates HSPC maintenance (83). These data provide insights into
with T cell senescence (79). In addition, this phenomenon was ac- how adrenergic signaling can account for two opposite events, name-
companied by significantly higher percentages and numbers of ly, rhythmic egress and homing of hematopoietic cells to and from the
germinal center B cells (B220+CD19+CD95+GL7+) (79). A second bone marrow. Local adrenergic signaling also plays a critical role in
study used high-throughput RNA sequencing data from the Cancer oscillatory inflammatory leukocyte recruitment to arteries and veins
Genome Atlas to show that disrupted circadian rhythms in tumors (72, 84). In a TNF-–induced inflammation model, it was demon-
correlate with increased tumor-associated T cell anergy in human strated that 2-AR signaling regulates the rhythmic expression of
alpha (IL-7R) on T cells, which is required for normal T cell severity of a Crohn’s disease model in mice (92). These data
proliferation, differentiation, and survival. GC binding to the GR in demonstrate that there is a delicate circadian balance between pro-
T cells promotes the expression of IL-7R protein by binding to GR moting gut permeability for nutrient absorbance during feeding
elements (GREs) in the Il7r enhancer (69). Removing GR or the hours and increasing intestinal immune surveillance of microbiota
GREs in the Il7r locus in T cells ablates the normally observed during the resting hours, which allows organisms to anticipate
rhythmic IL-7R expression, and mice lacking T cell–intrinsic GR nutrient uptake while maintaining homeostasis.
or Il7r GREs lose circadian rhythmicity in T cell cellularity in the
blood and lymphoid organs (69). This is due to loss of rhythmic, Gut microbiota
IL-7–driven CXCR4 expression, and mice lacking T cell–intrinsic In recent years, the gut microbiota has been shown to be highly in-
GR or Il7r GREs exhibit a similar phenotype to CXCR4-deficient fluenced by host circadian rhythm and rhythmic food intake. Gut
T cells (69). Because CXCR4 is the receptor for CXCL12 and crucial bacteria rhythmically adhere to the intestinal wall and exhibit time-
for driving leukocyte trafficking, interrupting this axis disrupts of-day–dependent changes in species composition. The microbiota
circadian oscillations in leukocyte migration (3). There is additional also has oscillatory production of metabolites that rhythmically
evidence that GC-induced CXCR4 expression preferentially recruits affect host rhythms, both locally in the gut and in relatively distant
Tregs specifically during the active phase to inflamed joints in a mouse sites such as the liver (93). Changes in diet affect the gut microbiome
model for arthritis (87). Although Tregs show no overt oscillations in and can weaken immune responses against bacteria or even alter im-
circadian clock genes (87), during both homeostasis and chronic mune cell migration patterns in the blood and other tissues (88).
inflammation, these cells exhibit circadian CXCR4 expression that The circadian immune response against Salmonella Typhimurium
matches the rhythmic oscillations of serum GCs (87). These data has recently been demonstrated to be driven by oscillations in feed-
reveal a critical role for GCs in guiding the circadian distribution of ing behavior and the activity of the gut microbiota. Mice pretreated
(using Camk2acre:Bmal1flox mice) completely abrogated rhyth- from the gut to the mesenteric LN or spleen, respectively (95, 97).
micity in ILC3s (95). In contrast, ILC3s appear to be only mildly In addition, mice with BMAL1-deficient ILC3s showed overall
influenced by microbial or rhythmic feeding cues (95), which is sur- reduced survival during C. rodentium infection compared with mice
prising, given that food intake strongly influences gut bacterial with BMAL1-sufficient ILC3s (95, 97). Mice with BMAL1-deficient
composition. Ablating the circadian clock in Rorc-expressing cells ILC3s also exhibited more severe gut inflammation during infection.
(using Rorccre:Bmal1flox mice), which includes gut ILC3s, causes a re- Furthermore, human patients with inflammatory bowel disease
duction in the frequency and number of ILC3s in the intestine, but exhibit ILC3 populations with dysregulated clock genes, compared
not the spleen, blood, or lung (90, 95–97). This is likely due to an with that of control patients (96). Together, these data make a
inability of clock-deficient ILC3s to migrate from the mesenteric strong case that circadian regulation of ILC3s is crucial for
maintaining gut homeostasis with the microbiota and managing both viruses in Bmal1−/− cells and a more severe RSV infection in
gut infection. Bmal1−/− mice (105). The importance of BMAL1 in antiviral re-
Although the number of ILC3s in the gut appears to be con- sponse was also highlighted in the context of Sendai virus (SeV)
trolled by the central clock and light, the function of ILC3s may be infection (106). Bmal1−/− mice were more susceptible to SeV infec-
influenced by food-induced expression of VIP. In early 2020, two tion because of a defective control of viral replication, resulting in
papers showed that ILC3s express the VIPR2, which binds VIP (90, 91). higher cytokine expression and an overall increased lung inflam-
ILC3s and VIP-expressing neurons colocalize in the intestine, and mation. In the context of hepatitis B infection, REV-ERB plays a
food consumption, which induces VIP production, changes the ex- direct role in controlling the expression of NTCP (sodium taurocho-
pression of IL-22 by ILC3s (90, 91). Although one paper showed late cotransporting polypeptide), the primary receptor used by
that in vitro and in vivo VIPR2 stimulation caused reduced IL-22 hepatitis B virus (HBV) to enter cells (107). Pharmacological activa-
protein expression in ILC3s (90), the other demonstrated the tion of REV-ERBs with the agonist SR9009 resulted in the inhibi-
opposite phenomenon, namely, increased IL-22 (91). Both groups tion of HBV entry into cells, whereas BMAL1, in contrast, was
argued that the VIP-induced changes they found were important shown to bind to HBV DNA and enhance viral promoter activity.
for maintaining gut homeostasis during feeding, either for reducing REV-ERBs were additionally shown to regulate HIV-1 replication
inflammation via high IL-22 levels (90) or for supporting efficient through the modulation of HIV-1 long-terminal repeat promoter
nutrient absorption and balance with the microbiota via low IL-22 activity (108). Furthermore, susceptibility to influenza infection
levels (91). Despite the differences in the role for VIPR2 stimula- is time-of-day dependent in mice, with animals infected at ZT11
tion, IL-22 production was shown to be rhythmic in ILC3s, and this (evening) experiencing a more severe disease and a higher mortality
rhythmicity could not be entirely abolished by removing cell-intrinsic compared with animals infected at ZT23 (morning) (109). This is
BMAL1 (90). It is clear from these findings that there is an import- despite the fact that mice infected at ZT8 (afternoon) feature more
macrophages and B cells. Circadian vaccination responses were fur- associated immunity, principally ILC3s, to exhibit time-of-day
ther observed in the CD8 T cell response to an intravenously injected differences. Because Nr1d1 and Rorc play important roles in both
DC vaccine (49). This was largely mediated by the CD8 T cell– circadian rhythms and as master regulators of TH17 and ILC3
intrinsic clock and circadian modulation of the TCR signaling path- development, this indicates a strong circadian control of the develop-
way, which enhanced the activation and proliferation potential of ment of these subsets. It will be important to distinguish between
T cells during the day. These studies demonstrate that circadian the circadian and developmental roles of these transcription factors
changes in the steady-state expression of immune molecules and in the future.
activation potential of immune cells can have a profound impact on Clinical data indicate time-of-day differences in long-term immune
the strength of vaccination responses. responses in humans, indicating that chronobiological considerations
are finding their way into the clinic. Although most of these datasets 20. P. G. Shackelford, R. D. Feigin, Periodicity of susceptibility to pneumococcal infection:
are still assessed retrospectively for potential time-of-day effects, a Influence of light and adrenocortical secretions. Science 182, 285–287 (1973).
21. M. Keller, J. Mazuch, U. Abraham, G. D. Eom, E. D. Herzog, H. D. Volk, A. Kramer, B. Maier,
few studies are now already incorporating circadian aspect into the
A circadian clock in macrophages controls inflammatory immune responses. Proc. Natl.
study design for prospective trials. Although a lot of ground has Acad. Sci. U.S.A. 106, 21407–21412 (2009).
been made, these advances have also yielded additional unanswered 22. M. L. Mul Fedele, I. Aiello, C. S. Caldart, D. A. Golombek, L. Marpegan, N. Paladino,
questions. One question is the relevance for circadian rhythmicity Differential thermoregulatory and inflammatory patterns in the circadian response
in vaccination responses. Do the observed differences in antibody to LPS-induced septic shock. Front. Cell. Infect. Microbiol. 10, 100 (2020).
23. G. L. Pearson, M. Savenkova, J. J. Barnwell, I. N. Karatsoreos, Circadian desynchronization
titers provide a better outcome after challenge? Assessing this in an
alters metabolic and immune responses following lipopolysaccharide inoculation in male
experimental and clinical setting will be essential. Furthermore, where mice. Brain Behav. Immun. 88, 220–229 (2020).
is rhythmicity in adaptive immunity defined and how is it maintained? 24. V. Lang, S. Ferencik, B. Ananthasubramaniam, A. Kramer, B. Maier, Susceptibility rhythm
It may be that the very initial response at the site of challenge to bacterial endotoxin in myeloid clock-knockout mice. eLife 10, e62469 (2021).
defines how the ensuing response unfolds in a circadian manner. It 25. C. L. Abram, G. L. Roberge, Y. Hu, C. A. Lowell, Comparative analysis of the efficiency
is currently completely unclear how circadian rhythmicity in im- and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice.
J. Immunol. Methods 408, 89–100 (2014).
mune responses seen months after the initial stimulus is maintained
26. J. A. Haspel, S. Chettimada, R. S. Shaik, J. H. Chu, B. A. Raby, M. Cernadas, V. Carey,
by the body, and why these differences do not wane or even become V. Process, G. M. Hunninghake, E. Ifedigbo, J. A. Lederer, J. Englert, A. Pelton, A. Coronata,
bigger. An answer may lie in the importance of circadian rhythms L. E. Fredenburgh, A. M. Choi, Circadian rhythm reprogramming during lung
in balancing physiology, maintaining the correct level of the immune inflammation. Nat. Commun. 5, 4753 (2014).
responses. Studies showing that lack of circadian rhythmicity can 27. J. Gibbs, L. Ince, L. Matthews, J. Mei, T. Bell, N. Yang, B. Saer, N. Begley, T. Poolman,
lead to exaggerated responses support this notion. Given the recent M. Pariollaud, S. Farrow, F. DeMayo, T. Hussell, G. S. Worthen, D. Ray, A. Loudon, An
major developments in this relatively new field, the coming years epithelial circadian clock controls pulmonary inflammation and glucocorticoid action.
41. S. Sato, T. Sakurai, J. Ogasawara, M. Takahashi, T. Izawa, K. Imaizumi, N. Taniguchi, H. Ohno, 61. J. R. Huh, M. W. Leung, P. Huang, D. A. Ryan, M. R. Krout, R. R. Malapaka, J. Chow, N. Manel,
T. Kizaki, A circadian clock gene, Rev-erb, modulates the inflammatory function of macrophages M. Ciofani, S. V. Kim, A. Cuesta, F. R. Santori, J. J. Lafaille, H. E. Xu, D. Y. Gin, F. Rastinejad,
through the negative regulation of Ccl2 expression. J. Immunol. 192, 407–417 (2014). D. R. Littman, Digoxin and its derivatives suppress TH17 cell differentiation by
42. P. Griffin, J. M. Dimitry, P. W. Sheehan, B. V. Lananna, C. Guo, M. L. Robinette, M. E. Hayes, antagonizing RORt activity. Nature 472, 486–490 (2011).
M. R. Cedeno, C. J. Nadarajah, L. A. Ezerskiy, M. Colonna, J. Zhang, A. Q. Bauer, T. P. Burris, 62. J.-Y. Lee, J. A. Hall, M. Pokrovskii, L. Kroehling, L. Wu, D. R. Littman, ROR enforces stability
E. S. Musiek, Circadian clock protein Rev-erb regulates neuroinflammation. Proc. Natl. of the T-helper-17 cell effector program. bioRxiv 2020.12.15.422921 [Preprint].
Acad. Sci. U.S.A. 116, 5102–5107 (2019). 22 December 2020. https://doi.org/10.1101/2020.12.15.422921.
43. K. D. Nguyen, S. J. Fentress, Y. Qiu, K. Yun, J. S. Cox, A. Chawla, Circadian gene Bmal1 regulates 63. X. O. Yang, B. P. Pappu, R. Nurieva, A. Akimzhanov, H. S. Kang, Y. Chung, L. Ma, B. Shah,
diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science 341, 1483–1488 (2013). A. D. Panopoulos, K. S. Schluns, S. S. Watowich, Q. Tian, A. M. Jetten, C. Dong, T helper 17
44. J. M. Adrover, C. Del Fresno, G. Crainiciuc, M. I. Cuartero, M. Casanova-Acebes, L. A. Weiss, lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR
H. Huerga-Encabo, C. Silvestre-Roig, J. Rossaint, I. Cossio, A. V. Lechuga-Vieco, gamma. Immunity 28, 29–39 (2008).
J. Garcia-Prieto, M. Gomez-Parrizas, J. A. Quintana, I. Ballesteros, S. Martin-Salamanca, 64. Y. Sun, Z. Yang, Z. Niu, J. Peng, Q. Li, W. Xiong, A. N. Langnas, M. Y. Ma, Y. Zhao, MOP3,
A. Aroca-Crevillen, S. Z. Chong, M. Evrard, K. Balabanian, J. Lopez, K. Bidzhekov, F. Bachelerie, a component of the molecular clock, regulates the development of B cells. Immunology
F. Abad-Santos, C. Munoz-Calleja, A. Zarbock, O. Soehnlein, C. Weber, L. G. Ng, 119, 451–460 (2006).
C. Lopez-Rodriguez, D. Sancho, M. A. Moro, B. Ibanez, A. Hidalgo, A neutrophil timer 65. Q. Cao, X. Zhao, J. Bai, S. Gery, H. Sun, D. C. Lin, Q. Chen, Z. Chen, L. Mack, H. Yang,
coordinates immune defense and vascular protection. Immunity 50, 390–402.e10 (2019). R. Deng, X. Shi, L. W. Chong, H. Cho, J. Xie, Q. Z. Li, M. Muschen, A. R. Atkins, C. Liddle,
45. M. Casanova-Acebes, C. Pitaval, L. A. Weiss, C. Nombela-Arrieta, R. Chevre, N. A-González, R. T. Yu, S. Alkan, J. W. Said, Y. Zheng, M. Downes, R. M. Evans, H. P. Koeffler, Circadian
Y. Kunisaki, D. Zhang, N. van Rooijen, L. E. Silberstein, C. Weber, T. Nagasawa, clock cryptochrome proteins regulate autoimmunity. Proc. Natl. Acad. Sci. U.S.A. 114,
P. S. Frenette, A. Castrillo, A. Hidalgo, Rhythmic modulation of the hematopoietic niche 12548–12553 (2017).
through neutrophil clearance. Cell 153, 1025–1035 (2013). 66. D. Druzd, O. Matveeva, L. Ince, U. Harrison, W. He, C. Schmal, H. Herzel, A. H. Tsang,
46. M. Crespo, B. Gonzalez-Teran, I. Nikolic, A. Mora, C. Folgueira, E. Rodriguez, L. Leiva-Vega, N. Kawakami, A. Leliavski, O. Uhl, L. Yao, L. E. Sander, C. S. Chen, K. Kraus, A. de Juan,
A. Pintor-Chocano, M. Fernandez-Chacon, I. Ruiz-Garrido, B. Cicuendez, A. Tomas-Loba, S. M. Hergenhan, M. Ehlers, B. Koletzko, R. Haas, W. Solbach, H. Oster, C. Scheiermann,
N. A-González, A. Caballero-Molano, D. Beiroa, L. Hernandez-Cosido, J. L. Torres, Lymphocyte circadian clocks control lymph node trafficking and adaptive immune
N. J. Kennedy, R. J. Davis, R. Benedito, M. Marcos, R. Nogueiras, A. Hidalgo, N. Matesanz, responses. Immunity 46, 120–132 (2017).
N. Kaushansky, T. Cheng, I. Sagi, M. Z. Ratajczak, S. Mendez-Ferrer, J. E. Dick, R. P. Markus, C. Bach, A. Ashton, S. Walsh, T. K. Tan, L. Schimanski, K. A. Huang, C. Schuster, K. Watashi,
T. Lapidot, Daily onset of light and darkness differentially controls hematopoietic stem T. S. C. Hinks, A. Jagannath, S. R. Vausdevan, D. Bailey, T. F. Baumert, J. A. McKeating, The
cell differentiation and maintenance. Cell Stem Cell 23, 572–585.e7 (2018). circadian clock component BMAL1 regulates SARS-CoV-2 entry and replication in lung
84. A. de Juan, L. M. Ince, R. Pick, C. S. Chen, F. Molica, G. Zuchtriegel, C. Wang, D. Zhang, epithelial cells. iScience 24, 103144 (2021).
D. Druzd, M. E. T. Hessenauer, G. Pelli, I. Kolbe, H. Oster, C. Prophete, S. M. Hergenhan, 104. R. S. Edgar, A. Stangherlin, A. D. Nagy, M. P. Nicoll, S. Efstathiou, J. S. O'Neill, A. B. Reddy,
U. Albrecht, J. Ripperger, E. Montanez, C. A. Reichel, O. Soehnlein, B. R. Kwak, Cell autonomous regulation of herpes and influenza virus infection by the circadian
P. S. Frenette, C. Scheiermann, Artery-associated sympathetic innervation drives clock. Proc. Natl. Acad. Sci. U.S.A. 113, 10085–10090 (2016).
rhythmic vascular inflammation of arteries and veins. Circulation 140, 1100–1114 (2019). 105. T. Majumdar, J. Dhar, S. Patel, R. Kondratov, S. Barik, Circadian transcription factor
85. A. Garcia-Garcia, C. Korn, M. Garcia-Fernandez, O. Domingues, J. Villadiego, D. Martin-Perez, BMAL1 regulates innate immunity against select RNA viruses. Innate Immun. 23,
J. Isern, J. A. Bejarano-Garcia, J. Zimmer, J. A. Perez-Simon, J. J. Toledo-Aral, T. Michel, 147–154 (2017).
M. S. Airaksinen, S. Mendez-Ferrer, Dual cholinergic signals regulate daily migration 106. A. Ehlers, W. Xie, E. Agapov, S. Brown, D. Steinberg, R. Tidwell, G. Sajol, R. Schutz,
of hematopoietic stem cells and leukocytes. Blood 133, 224–236 (2019).
R. Weaver, H. Yu, M. Castro, L. B. Bacharier, X. Wang, M. J. Holtzman, J. A. Haspel, BMAL1
86. R. Dumbell, O. Matveeva, H. Oster, Circadian clocks, stress, and immunity. Front.
links the circadian clock to viral airway pathology and asthma phenotypes. Mucosal
Endocrinol. (Lausanne) 7, 37 (2016).
Immunol. 11, 97–111 (2018).
87. L. E. Hand, K. J. Gray, S. H. Dickson, D. A. Simpkins, D. W. Ray, J. E. Konkel, M. R. Hepworth,
107. X. Zhuang, D. Forde, S. Tsukuda, V. D'Arienzo, L. Mailly, J. M. Harris, P. A. C. Wing,
J. E. Gibbs, Regulatory T cells confer a circadian signature on inflammatory arthritis.
H. Borrmann, M. Schilling, A. Magri, C. O. Rubio, R. J. Maidstone, M. Iqbal, M. Garzon,
Nat. Commun. 11, 1658 (2020).
R. Minisini, M. Pirisi, S. Butterworth, P. Balfe, D. W. Ray, K. Watashi, T. F. Baumert,
88. D. Zheng, K. Ratiner, E. Elinav, Circadian influences of diet on the microbiome J. A. McKeating, Circadian control of hepatitis B virus replication. Nat. Commun. 12, 1658
and immunity. Trends Immunol. 41, 512–530 (2020). (2021).
89. J. F. Brooks II, C. L. Behrendt, K. A. Ruhn, S. Lee, P. Raj, J. S. Takahashi, L. V. Hooper, The
108. H. Borrmann, R. Davies, M. Dickinson, I. Pedroza-Pacheco, M. Schilling, A. Vaughan-Jackson,
microbiota coordinates diurnal rhythms in innate immunity with the circadian clock. Cell
A. Magri, W. James, P. Balfe, P. Borrow, J. A. McKeating, X. Zhuang, Pharmacological
184, 4154–4167.e12 (2021).
activation of the circadian component REV-ERB inhibits HIV-1 replication. Sci. Rep. 10,
90. C. Seillet, K. Luong, J. Tellier, N. Jacquelot, R. D. Shen, P. Hickey, V. C. Wimmer,
13271 (2020).
L. Whitehead, K. Rogers, G. K. Smyth, A. L. Garnham, M. E. Ritchie, G. T. Belz, The
Science Immunology (ISSN ) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW,
Washington, DC 20005. The title Science Immunology is a registered trademark of AAAS.
Copyright © 2022 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works