You are on page 1of 16

SCIENCE IMMUNOLOGY | REVIEW

IMMUNE REGULATION Copyright © 2022


The Authors, some
The circadian immune system rights reserved;
exclusive licensee
American Association
Chen Wang1†, Lydia Kay Lutes1†, Coline Barnoud1, Christoph Scheiermann1,2* for the Advancement
of Science. No claim
The immune system is highly time-of-day dependent. Pioneering studies in the 1960s were the first to identify to original U.S.
immune responses to be under a circadian control. Only in the last decade, however, have the molecular factors Government Works
governing circadian immune rhythms been identified. These studies have revealed a highly complex picture
of the interconnectivity of rhythmicity within immune cells with that of their environment. Here, we provide a
global overview of the circadian immune system, focusing on recent advances in the rapidly expanding field of
circadian immunology.

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

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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
state and during activation. (RAR)–related orphan receptors (RORs) make up a secondary feed-
Immune rhythms were first found in the innate immune system back loop that stabilizes the core molecular clock (15, 16). However,
in 1960 (1), and 10 years later, components of the adaptive immune it is important to note that circadian rhythms can also occur in cells
system were shown to be similarly time-of-day dependent (2). More that lack a nucleus, where rhythms are orchestrated by oscillations
than half a century later, we now know that immune cellularity, mi- in the oxidation of peroxiredoxin proteins (18).
gration, and function are all regulated by the circadian clock. The Most circadian studies in the immune system have focused on
number of circulating leukocytes oscillates in the blood of mam- the role of BMAL1, because it is the only clock gene whose sole ab-
mals, peaking during the behavioral rest phase in nocturnal mice sence abrogates rhythmicity (17). Studies that disrupt the molecular
(day) (3) and diurnal humans (night) (Box 1) (4). Circadian clocks clock through removal of BMAL1 must therefore differentiate be-
also modulate leukocyte migration across the body, effectively tween effects mediated specifically by BMAL1 and those mediated
gating the number of leukocytes at specific sites throughout the day by the function of the clock in general, because the latter should
(3, 5–7). In addition, leukocyte effector functions are time-of-day be corroborated with additional clock mutants whenever possible.
dependent as is the capacity of activated immune cells to proliferate
and produce cytokines (8–11). It is highly likely that these immune
rhythms evolved to defend against diurnal peaks of pathogen en-
counter, such as those occurring due to increased social interactions Box 1. Circadian terminologies.
or transmitted via food intake (12) that occur predominantly during Circadian rhythm: From the Latin circa diem (“about a day”), it refers to
the behavioral active period. approximately 24-hour cycles that are perpetuated by recurring
Although the ability to detect and adapt to recurring environ- environmental cues, such as light and food intake, which drive daily
mental rhythms exists in virtually all species, the molecular manner oscillations in the most physiological processes. Circadian rhythms can be
synchronized and are endogenous rhythms.
and complexity of how circadian rhythms are maintained vary be-
Diurnal rhythm: A daily oscillation. It may or may not be a circadian rhythm.
tween organisms (13, 14). In mammals, the core circadian clock
Diurnal: Refers to a species that is active mainly during the daytime, e.g.,
machinery consists of a highly interconnected system of transcrip- Homo sapiens.
tion factors, forming several transcription-translation feedback loops Nocturnal: Refers to a species that is active mainly during the nighttime,
that activate and inhibit one another to drive 24-hour rhythms at e.g., Mus musculus.
the molecular level (15). This network is often referred to as the core Zeitgeber time: Zeitgeber, from the German “time giver,” is an external
clock and consists of the transcription factors BMAL1 [brain and cue (such as light or food intake) that synchronizes the circadian clock of
muscle aryl hydrocarbon receptor nuclear translocator (ARNT)– an organism to its environment. Zeitgeber time (ZT) is the time in hours
like 1; encoded by Arntl], CLOCK (circadian locomotor output after light onset, for example, lights on is ZT0 and lights off is ZT12 in a
cycles kaput), PER1/2/3 (period circadian protein homologs), 12-hour light/12-hour dark cycle.
Circadian time (CT): A measure of subjective time used when organisms
and CRY1/2 (cryptochromes 1/2) (15, 16). At the protein level,
are isolated from Zeitgebers of the environment (for example, in constant
BMAL1 and CLOCK form a heterodimeric transcription factor, which
darkness). CT0 represents the start of the subjective day, and CT12
binds to enhancer (E)–box regions of target genes, including those represents the start of the subjective night.
Synchronization: The process where Zeitgeber signals reset an organism
1
Department of Pathology and Immunology, Faculty of Medicine, University of or cells in culture to the same time or phase.
Geneva, Geneva, Switzerland. 2Biomedical Center (BMC), Institute for Cardiovascular Suprachiasmatic nucleus (SCN): A brain region located within the anterior
Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine hypothalamus that receives environmental light cues and constitutes the
(WBex), Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
*Corresponding author. Email: christoph.scheiermann@unige.ch central clock.
†These authors contributed equally to this work.

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 1 of 15


SCIENCE IMMUNOLOGY | REVIEW

Deficiency in various clock components, such as Bmal1 by itself


or double knockouts of Cry1 and Cry2 or Per1 and Per2, has been Box 2. Key circadian immune concepts.
associated with a variety of altered inflammatory phenotypes, Lethality to strong immune stimuli, such as in the form of high doses of
demonstrating the critical role of the circadian clock in the proper LPS, bacteria, or viruses, exhibits a circadian rhythm, peaking around the
function of the mammalian immune system (Table 1) (16). In this onset of the behavioral activity phase.
Review, we will discuss how circadian regulation of leukocyte cel- Cytokine secretion from stimulated macrophages is circadian.
The numbers of leukocytes in blood are circadian, peaking during the
lularity, trafficking, and effector activity affects the function of
behavioral rest phase in both mice and humans. This is driven by circadian
the immune system.
rhythms in leukocyte trafficking. The mobilization of leukocytes from the
bone marrow into blood, the drainage of leukocytes from tissues, and the
homing of leukocytes to tissues are all rhythmic in a circadian manner.
INNATE IMMUNITY Because these oscillations peak at different times of day, blood leukocyte
Response to bacterial components numbers exhibit an overall circadian oscillation.
Lipopolysaccharides (LPS) are found in the outer membrane of Circadian rhythms in the host and microbiota interact with each other to
Gram-negative bacteria. LPS bind to Toll-like receptor 4 (TLR4) maintain metabolic homeostasis of the host. Host rhythms influence the
and initiate an inflammatory signaling cascade. This has been the most intestinal barrier and microbiota adhesion to the intestinal wall.
widely used model of inflammation to assess circadian rhythms in Microbiota produce metabolites in a time-of-day–dependent manner,
thus regulating the host rhythm.

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


innate immune responses. It was first used in the 1960s, showing
that mice injected with LPS in the evening exhibited a notably
increased mortality compared with those challenged in the morning
(Box 2) (1). A few years later, studies reported similar results using
Gram-positive Streptococcus pneumoniae (19, 20). Many genes in a shift to a new set of genes and immunological processes. In addi-
the LPS-induced signaling cascade are rhythmically expressed in tion, LPS challenge shortened the median period length of the circadian
peritoneal macrophages at steady state, and splenocytes exhibit rhythm in the lung from 24 hours (steady state) to 19.6 hours. Flow
rhythmic tumor necrosis factor– (TNF-) and interleukin-6 (IL-6) cytometry analyses also detected oscillations in B cell and Gr1+
production after LPS challenge (21). Mice treated with LPS at Zeitgeber myeloid cell counts in the lungs of naïve mice, with a peak occurring
time 11 [ZT11; i.e., time (in hours) after light onset in a 12-hour in the morning and a trough in the evening (26). Steady-state
light:12-hour dark (LD) environment, “evening”] exhibited signifi- alveolar macrophages presented an opposite and lower amplitude
cantly higher serum TNF- levels and suffered increased mortality rhythm, peaking in the evening. In contrast, LPS-treated animals
than those treated at ZT19 (“night”) (Box 1) (22). TNF receptor 1 exhibited a dampened amplitude of oscillating B cell numbers,
(TNFR1)–deficient mice exhibited no time-of-day differences in whereas myeloperoxidase-positive (MPO+) granulocytes presented
mortality rate, indicating that the temporal differences observed are circadian-­like variation (26). These data demonstrate the strong
TNF- dependent (22). These data demonstrate that the innate im- interaction between a rhythmic environment and the ensuing im-
mune response to LPS is under circadian control. mune responses.
Disrupting the circadian rhythm of mice affects the immune Circadian oscillation of granulocytes in the lung after LPS stim-
response to LPS challenge. Mice that were desynchronized from ulation is completely abrogated in whole-body Bmal1-deficient mice
their normal circadian environment—by shifting them from the (26). However, it is still unclear which cell types require Bmal1 to
usual 24-hour (12-hour:12-hour LD) cycle to a shorter 20-hour induce this rhythmic granulocyte response. Mice with Bmal1-­deficient
(10-hour:10-hour LD) cycle—exhibited delayed recovery of loco- myeloid cell subsets (Lyz2cre:Bmal1flox) still showed rhythmic re-
motion and weight loss after LPS challenge compared with un- sponses to LPS challenge in the lung (27), indicating a myeloid-­
disrupted mice (23). In addition, mice moved from a 12-hour:12-hour independent mechanism [analogous to the observation discussed
LD schedule to constant darkness (DD; Box 1) exhibited a threefold above (24)]. However, removing BMAL1 expression in bronchiolar
increased susceptibility to LPS compared with mice kept in rhythmic epithelial cells (using Ccspcre:Bmal1flox) ablated their diurnal secre-
LD conditions (24). Although cytokines such as TNF-, monocyte tion of C-X-C motif chemokine ligand 5 (CXCL5) in response to
chemoattractant protein-1 (MCP-1), IL-18, and IL-10 showed a dis- LPS treatment (27). Bronchiolar epithelial cells require BMAL1
tinct time-of-day oscillation in the peripheral blood of mice in LD expression to respond to oscillating glucocorticoid (GC) levels
conditions after LPS challenge, the phase of this oscillation was (discussed further below), and their rhythmic production of CXCL5
changed in mice kept in DD conditions (24). Interestingly, these is dependent on cell-intrinsic expression of the GC receptor (GR)
oscillations and the differences in survival could not be fully abro- (28). Removing GR from the airway epithelium, however, still did
gated in Lyz2cre:Bmal1flox or Lyz2cre:Clockflox mice (24), which target not abolish circadian oscillations in granulocyte recruitment or
the circadian clock in the myeloid cell lineage [mostly granulocytes rhythmic expression of TNF- and IL-6 in the lung after LPS stim-
and some macrophage subsets, as well as a fraction of monocytes (25)]. ulation (28). Thus, although bronchiolar epithelial cell–intrinsic
This indicated that light may exert an anti-inflammatory influence expression of BMAL1 and the GR is sufficient to explain oscillating
on LPS responses in mice and that rhythmicity of this pheno­menon is levels of CXCL5, they do not drive the circadian control of neutro-
at least partially independent of myeloid cells. philic infiltration in response to inhaled LPS, indicating that there
Using a genome-wide screening approach, a recent study inves- are additional circadian mechanisms involved.
tigated the circadian transcriptome in murine lungs, showing that Apart from BMAL1 and CLOCK, the clock component REV-ERB
genes related to immune function in steady state were enriched in has also been implicated in controlling pulmonary LPS-induced
the middle of the day (26). However, upon challenge with LPS inflammation (29). Although wild-type mice exhibit increased neu-
during the evening, gene expression was reprogrammed, exhibiting trophil lung infiltration in the morning (ZT0) versus the evening

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 2 of 15


SCIENCE IMMUNOLOGY | REVIEW

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.

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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.
Relative activity of the NRF2 transcription factor is
Lyz2cre:Bmal1flox LPS-stimulated BMDMs (37)
significantly decreased in Bmal1−/− macrophages.
Circadian-dependent variation in cytokine response to
Lyz2cre:Bmal1flox Rev-erb−/− full KO LPS-induced inflammation (40)
LPS is abrogated in Lyz2cre:Bmal1−/− and Rev-erb−/− mice.
REV-ERB in the murine macrophage suppresses Ccl2
Rev-erb−/− full KO LPS-induced inflammation (41)
expression induced by LPS challenge.
The time-of-day variation in pulmonary neutrophilia is
Rev-erb−/− full KO LPS-induced inflammation (29)
abrogated in Rev-erb−/− mice.
REV-ERB inhibits IL-1 production through the NF-B/
Pharmacological target REV-ERB LPS-induced inflammation (34)
NLRP3 pathway.
REV-ERB regulates diurnal expression and activation of
Rev-erb−/− full KO Induced fulminant hepatitis NLRP3 inflammasome, attenuating NLRP3-driven (35)
inflammation.
REV-ERB represses p65 transcription and indirectly
Rev-erb−/− full KO DSS-induced colitis (38)
represses Nlrp3 via the NF-B pathway.
Loss of Bmal1 abrogates rhythmic recruitment of
Lyz2cre:Bmal1flox TNF-–induced inflammation (84)
myeloid cells to arteries and veins.
Myeloid cell deficiency in Bmal1 abrogates time-of-day
Lyz2cre:Bmal1flox EAE (74)
differences in disease severity of EAE.
REV-ERB/ agonist (SR9009, Macrophages derived from Pharmacological activation of REV-ERB/ inhibits HIV
(108)
GSK2667) administration human iPSCs infected by HIV promoter activity, impeding viral replication.
Diurnal microglial immunoreactivity in the
Microglia Rev-erb−/− full KO LPS-induced inflammation (42)
hippocampus is lost after Rev-erb deletion.
Clec9cre:Bmal1flox Increased DC migration into skin lymphatic vessels occurs
DC DC crawl in (48)
Cdh5creERT2:Bmal1flox at ZT7, which is largely dependent on rhythms in CCL21.
Rev-erb−/− full KO Rev-erb−/− full BMDCs from KO mice showed enhanced expression of
BMDC from DKO mice (50)
KO CD86, MHCII, and proinflammatory cytokines.
DC clock regulates time-of-day–dependent efficiency of
T. muris parasitic helminth
Cd11ccre:Bmal1flox worm expulsion by polarizing the immune system (111)
infection
toward a TH2 response.
Impaired BMDCs migration to spleen after intravenous
Bmal1 full KO DC-OVA vaccination (49)
injection
Similar neutrophils and G-CSF amount in mice with
Neutrophil Lyz2cre:Bmal1flox LPS-induced inflammation (27)
Bmal1-sufficient and Bmal1-deficient myeloid cells.
Neutrophil Bmal1 controls the expression of Cxcl2 in
Cell-intrinsic deletion in
hMRP8cre:Bmal1flox neutrophils, regulating neutrophil aging that peaks (44)
neutrophils
during the day.

continued on next page

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 3 of 15


SCIENCE IMMUNOLOGY | REVIEW

Cell type Mouse strain/cells Experimental model Findings Ref


T cell deficiency in Bmal1 abrogates rhythmic,
cell-intrinsic CCR7 and S1PR1 expression, rhythmic T cell
T cell Cd4cre:Bmal1flox Steady state and EAE (66)
homing to LN, and differences in disease severity in an
EAE model.
Reduced T cell IFN-, IL-2, and TNF- production during
Cd4cre:Bmal1flox L. monocytogenes infection (56)
L. monocytogenes infection.
Abrogation of time-of-day–dependent expansion of
E8Icre:Bmal1flox DC-OVA vaccination OVA-specific T cells in response to vaccination performed (49)
at CT6 versus CT18
REV-ERB/ agonist (SR9009, Jurkat and primary human T Pharmacological activation of REV-ERBs inhibits HIV
(108)
GSK2667) administration cell lines infected by HIV promoter activity, impeding viral replication in vitro.

Rev-erb−/− full KO Rev-erb−/−/−/−


Perturbed light cycles Oscillations in TH17 differentiation are disrupted. (57)
DKO
Loss of rhythmic T cell cellularity in blood, lymph, and
Adrb2−/− full KO Integrin blocking (70)

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


peripheral LN.
Increased activation of the T cell CCR7 and CXCR4
Adrb2−/− full KO 2-AR agonist stimulation (68)
signaling pathways. Increased T cell retention in the LN.
Reduced IL-7Ra expression during the active phase, loss
Cd4cre:Nr3c1flox Steady state of rhythmic T cell CXCR4 expression, and loss of (69)
rhythmic T cell cellularity in blood and LN.
Clock19 In vitro anti-CD3 stimulation Loss of rhythmic T cell proliferative capacity. (10)
T cell deficiency in Bmal1 does not abrogate rhythmic
Cd4cre:Bmal1flox Steady state (87)
CXCR4 expression on Tregs in the spleen or inguinal LN.
Human peripheral blood Reduced peripheral B cell numbers, defective B cell
B cell Bmal1+/−, Bmal1−/− full KO (64)
lymphocytes immunization development, and IgG1 production.
Increased IgG and IgM production. Spontaneous
Cry1−/–Cry2−/− full DKO NP-KLH immunization (65)
development of an autoimmune phenotype.
flox
Cd19cre:Bmal1 Steady state Loss of rhythmic homing to LN. (66)
Loss of rhythmic B cell cellularity in blood, lymph, and
Adrb2−/− full KO Integrin blocking (70)
peripheral LN.
Loss of diurnal variation in IgM and IgG1 antibody
Adrb2−/− full KO NP-CGG immunization (70)
production.
Increased B cell retention in the LN and increased
Adrb2−/− full KO 2-AR agonist stimulation (68)
activation of the CCR7 and CXCR4 signaling pathways.
flox
Gut ILC3s Vav1cre:Bmal1 Mixed bone marrow chimera Reduced ILC3 cellularity in the gut. (95)
Reduced ILC3 cellularity in the gut and the remaining
Rorccre:Bmal1flox Steady state (96)
ILC3s are IL-17+ and IL-22+.
+
Loss of REV-ERB increases the percentage of IL-17 and
Steady-state and C. difficile
Rev-erb−/− full KO IL-22+ gut ILC3s and increases bacterial translocation to (97)
infection
mesenteric LNs during C. difficile infection.
Donor KO-derived ILC3s cells have reduced cellularity in
Il7rcre:Bmal1flox Mixed bone marrow chimera (90)
the gut compared with control Bmal1flox donor cells.
Reduced expression of ILC3 gut-homing molecules
Steady state and C. rodentium altered microbiota. After infection with C. rodentium, KO
Rorccre:Bmal1flox (95)
infection mice exhibit increased clinical score, bacterial burden,
and bacterial translocation.

(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

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 4 of 15


SCIENCE IMMUNOLOGY | REVIEW

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

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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,
and proteome of bone marrow–derived macrophages (BMDMs) as well as in the peritoneum. Furthermore, ChIP analysis showed
showed that only 15% of the oscillatory proteome was found to have that the BMAL1-CLOCK heterodimer binds directly to the E-box of
an oscillating mRNA, indicating that most of the proteome oscillates Ccl2, controlling its rhythmic expression (43). This work provided
because of posttranscriptional regulation (33). Further analy- evidence for a crucial role of Bmal1 in inflammatory monocytes.
sis showed that proteins involved in degradation and translation, In summary, macrophages and monocytes exhibit a strong circadian
such as tRNA-aminoacylation proteins and ubiquitin ligase, are rhythm, particularly with respect to the circadian components BMAL1
rhythmically enriched, which could explain the phenomenon. and REV-ERB, which are implicated in controlling metabolism,
Moreover, tricarboxylic acid–related proteins and oxygen con- function, and cytokine production in these cells.
sumption rate exhibit circadian oscillations, resulting in rhythms of
adenosine triphosphate production (33). Together, these data show Neutrophils
that the clock gene Bmal1 can regulate the metabolic function of The number of neutrophils in blood oscillates with time of day, and
macrophages. their phenotype exhibits a diurnal change that has been associated
Recent studies have focused on the connections between clock with aging. Both phenomena are controlled by Bmal1 in a cell-­
components, particularly REV-ERB, and inflammasome pathways autonomous manner, as demonstrated with the use of an hMRP8Cre:
(Table 1) (34–37). In mouse and human macrophages, NLR family Bmal1flox mouse line that targets neutrophils (44). ChIP assays
pyrin domain containing 3 (NLRP3, a major component of the in- demonstrated that BMAL1 can directly bind to the E-box in the pro-
flammasome) is expressed in a diurnal manner under the control of moter region of Cxcl2, which governs neutrophil aging in a CXCL2-
REV-ERB (35). NLRP3 modulates the expression and secretion of IL-1 C-X-C motif chemokine receptor 2 (CXCR2)–dependent manner.
and IL-18 in the peritoneum, resulting in time-of-day–dependent In contrast, CXCR4 acts as an antagonist to CXCR2-induced aging,
disease severity changes in a mouse model of fulminant hepatitis (38). favoring neutrophil clearance and protection of the cardiovascu-
Mechanistically, chromatin immunoprecipitation (ChIP) assays lar system (44). Surprisingly, although neutrophils only represent
showed that REV-ERB can directly bind to the Nlrp3 promoter re- a minor fraction of leukocytes in nonhematopoietic tissues under
gion in BMDMs (35, 38). Pharmacological activation of REV-ERB steady-state conditions, they were reported to be important regula-
inhibited the NLRP3 inflammasome and reduced the severity of fulmi- tors of the hematopoietic stem cell niche and to alter clock gene ex-
nant hepatitis (35, 38). REV-ERB was further shown to repress the pression in various organs (5, 45). For example, neutrophils were
function of the nuclear factor B (NF-B)/NLRP3 axis, and REV-ERB–­ shown to infiltrate the mouse liver in a time-of-day–dependent man-
deficient mice exhibited a more activated NLRP3 inflammasome (38). ner, which altered the hepatocyte clock because of neutrophil elastase
Enhanced inflammasome activity rendered these mice more sensitive (NE) secretion (46). Neutrophils peaked in the liver during the light
to experimental colitis, demonstrating critical interactions between phase, where they secreted elastase and activated c-Jun N-terminal
circadian rhythms and inflammasome regulation. These studies kinase (JNK) signaling in hepatocytes, ultimately triggering Bmal1
show the strong anti-inflammatory functions of REV-ERB, in expression. Intriguingly, this phenomenon may also relate to the
addition to its role in the circadian clock. human liver, as a strong correlation between activation of JNK, NE
On the basis of this important anti-inflammatory role, REV-ERB levels, and BMAL1 expression was observed in human patient sam-
is currently used as target in therapeutic studies. One study found ples (46). In other tissues, neutrophil infiltration was shown to alter the
that the REV-ERB agonist GSK4112 can inhibit the expression of transcriptional profile of the whole organ (47). Neutrophil infiltration of
NLRP3 and reduce the production of IL-1 in an LPS-induced in- the intestine, for instance, suppressed the expression of IL-23 in
flammation model (34). Another study showed that an NF-B– resident macrophages, thereby reducing the concentration of granu-
driven long noncoding RNA (lncRNA), Lnc-UC, can promote the locyte colony-stimulating factor (G-CSF) in blood plasma (47). These
expression of REV-ERB and, in a negative feedback loop, can studies demonstrate the strong influence of circadian rhythms on

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 5 of 15


SCIENCE IMMUNOLOGY | REVIEW

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 RORt (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

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


expressed CCR7. Synchronization of bone marrow–derived DCs required to assess a potential time-of-day–dependent role for RORt
(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.
autonomous and LEC circadian clocks in these rhythms (Box 1). In contrast to T cells, B cell differentiation appears to be depen-
ChIP analyses indicated that BMAL1 directly controls the expression dent on the molecular clock machinery. Complete BMAL1-deficient
of Ccl21 and Ccr7, as well as the adhesion molecule Lyve1 (48). mice exhibit a partial block in B cell development, as demonstrated
Another study using BMDCs as APCs demonstrated that expan- by a significantly reduced percentage of B220high, mature B cells and
sion of antigen-­specific CD8 T cells is time-of-day dependent a significantly increased percentage of B220low out of the total bone
and partially driven by the DC intrinsic clock, given that dele- marrow cells (64). This effect was demonstrated via adoptive transfer
tion of Bmal1 in BMDCs impaired T cell migration to the spleen experiments to be non–B cell autonomous and to involve the bone
after adoptive transfer (49). An ex vivo study using REV-ERB– and marrow stromal cell compartment (64). This is in line with data from
REV-ERB–deficient BMDCs showed that loss of either REV-ERB mice with a B cell–intrinsic deletion of BMAL1 that exhibit normal
or REV-ERB enhanced the expression of the maturation markers B cell development and function (56). In contrast, mice exhibiting
CD86 and major histocompatibility complex II (MHCII), as well as double deficiency for the BMAL1-repressive clock components Cry1
proinflammatory cytokines such as Il1b, Il6, and Il12b (50). Inversely, and Cry2 show increased serum immunoglobulin G (IgG) levels, in-
the REV-ERB–specific agonist SR9009 inhibited the expression of creased phosphorylation of the proximal B cell receptor signaling
maturation markers and proinflammatory cytokines in BMDCs. pathway, and an overall autoimmune phenotype (65). These results
Consistent with the data in macrophages (34–37), REV-ERB pres- indicate a non–cell-autonomous role for the circadian clock in B cell
ents anti-inflammatory function in DCs, a phenotype that appears development and differentiation.
to be independent of its role within the clock. In summary, these
data demonstrate that DC functions are circadian in nature and Lymphocyte trafficking
can be altered by targeting the clock machinery. Circadian rhythms govern the rhythmic shuttling of lymphocytes
between blood, lymph (both peaking during the behavioral rest
phase), and tissues (peaking around the onset of the active phase)
ADAPTIVE IMMUNITY (3, 66–71). Thus far, three mechanisms have been identified to ex-
Given the longer time frame compared with innate immune re- plain these daily migration patterns, which likely co-govern the
sponses, it is surprising that adaptive immunity exhibits circadian overall phenotype: BMAL1 activity in both lymphocytes and endo-
rhythmicity as well, even weeks after an initial challenge (Table 1) thelial cells (3, 66), the sympathetic nervous system via activation of
(7, 16, 51–55). Here, we provide a summary of the major findings the 2–adrenergic receptor (AR) (70), and GCs (69), the latter two
and highlight the most recent developments. of which are discussed in the next section.
BMAL1 controls lymphocyte trafficking via cell-intrinsic and
Development of lymphocytes cell-extrinsic means. Cell-intrinsic expression of BMAL1 in T cells
Current data indicate that T cell development in the thymus is inde- and B cells governs rhythmic cellularity in lymph nodes (LNs) by
pendent of the circadian clock because T cells with an intrinsic controlling circadian oscillations in the expression of the LN-­
deletion of BMAL1 exhibit no defects in thymic development (56). homing factor CCR7 and the egress factor S1pr1 in lymphocytes
In addition, the frequency of CD4+ T helper 17 (TH17) cells in the (66). Endothelial BMAL1 expression is required for the circadian
intestinal lamina propria (LP) is unchanged in these mice, which protein expression of the adhesion molecules intercellular adhe-
suggested that intestinal TH17 differentiation is also not directed by sion molecule–1 (ICAM-1) and vascular cell adhesion molecule–1
BMAL1 (56). However, several clock genes are involved in or even (VCAM-1) in endothelial cells, which guide rhythmic lymphocyte
master regulators of TH17 differentiation, including Nr1d1 and the homing into tissues (3,  66,  72). Without oscillatory expression of
ROR family of transcription factors (Rorc and Ror in particular). these adhesion receptors, rhythmic tissue recruitment is lost
Using whole-body knockout mice, REV-ERB (encoded by Nr1d1) (3, 66, 72). Thus, BMAL1 is extrinsically (through endothelial cells)

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 6 of 15


SCIENCE IMMUNOLOGY | REVIEW

in the day (ZT2) showed increased IL-2


production compared with infection at
night (ZT14) (56). Together, these data
indicate that, although adaptive immune
responses are clearly rhythmic, different
routes of immunization may be re-
sponsible for some of these observed
time shifts in peak response times.
Oscillations in T cell responses to
antigen stimulations are due, at least in
part, to rhythmic changes in the de-
gree to which naïve T cells are poised
to respond to stimulation throughout
the circadian cycle. The temporal dif-
ferences in T cell vaccination responses
correlated with oscillations in the mRNA

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


expression of T cell receptor (TCR) sig-
naling pathway regulators (49). Positive
regulators of T cell activation (ZAP70,
AKT, and phospholipase C–) peaked
during the day, the time at which
OVA-loaded DC vaccination yielded
the highest T cell responses. In contrast,
negative regulators of TCR signaling
[SHP1 (small heterodimer partner 1),
CSK (C-terminal Src kinase), and PTEN
phosphatase and tensin homolog] peaked
at night (49). These data suggest that
naïve T cells are poised to respond to
antigens in a time-of-day–dependent
Fig. 1. DCs migrate into skin lymphatics in a circadian manner. DC migration peaks during the daytime (left) and
manner and, together with rhythmic DC
troughs during the nighttime (right) in mice. This oscillation is driven by rhythmic secretion of CCL21 by LECs and the trafficking and function, contribute to the
rhythmic expression CCR7 on DCs and adhesion molecules LYVE-1, CD99, and JAM-A on LECs (48). All of these mole- oscillations in vaccination responses.
cules show increased expression during the daytime (left) and decreased expression at nighttime (right). These pro- B cells demonstrate rhythmic effec-
teins are directly regulated by LEC or DC-intrinsic mechanisms, respectively, where BMAL1 binds to promoter regions tor responses through cell-intrinsic, clock-­
in their genes. driven TLR9 protein expression (73).
When mice were stimulated with OVA-
CpG (a TLR9 agonist) at peak TLR9
and intrinsically essential for normal lymphocyte circulation between expression on LN B cells [occurring around ZT19 (night)], they ex-
blood, lymph, and tissues. hibited increased cytokine production and proliferation compared
with stimulation during times of lower TLR9 expression (73). Thus,
Effector functions these correlative data indicate that, similar to T cells, B cells may also
Adaptive immune cells also display rhythmic effector functions. be poised to exhibit strong effector responses in a circadian manner.
T cells exhibit clock-dependent, rhythmic proliferation (10) and Rhythmic effector function can also affect the severity of auto-
show oscillatory IFN- production after in vitro stimulation (11). In immune diseases. Mice lacking BMAL1 in T cells show reduced severity
addition, several studies have demonstrated that the strongest adap- scores after induction of experimental autoimmune encephalomyeli-
tive immune response occurs if antigen is encountered in the skin tis (EAE; an animal model of multiple sclerosis) (66). Furthermore, in
when LNs are near their peak cellularity, which, in mice, occurs be- the skin, it was shown that rhythmic mRNA expression of the IL-23
tween late afternoon and late night (Table 1) (66, 69, 70, 73, 74). In receptor (IL-23R) in T cells correlated with imiquimod (IMQ)–­
contrast, mice immunized intravenously with ovalbumin (OVA)– induced psoriasis severity in murine skin (75). IL-23 is released in
CREDIT: A. MASTIN/SCIENCE IMMUNOLOLGY

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

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 7 of 15


SCIENCE IMMUNOLOGY | REVIEW

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

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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
cancers (80). Across 716 samples and 29 cancer types, negative reg- adhesion molecules within both vascular beds. This has functional
ulators of the circadian clock were down-regulated, whereas the implications for a time-of-day–dependent predisposition to intra-
positive regulator ARNTL2 was up-regulated in most types of can- vascular, thrombotic events, involving rhythmic adhesion of leuko-
cers investigated, suggesting that tumors lose oscillatory mRNA cytes and platelets to endothelial cells (84). These findings show
expression of circadian clock genes (80). Furthermore, high expres- that leukocyte trafficking is heavily controlled by circadian oscil-
sion of clock genes was positively correlated with levels of T cell lations in the sympathetic nervous system.
anergy markers in tumor samples, such as programmed cell death 2-AR signaling is also a crucial regulator of lymphocyte effector
protein 1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 functions after time-of-day–dependent immunizations. Immune re-
(CTLA-4) (80). These findings indicate a link between dysregulated actions were observed to be stronger when immunization was per-
clock gene expression and T cell anergy in tumors, indicating that formed at ZT17 (night; coinciding with enhanced adrenergic tone)
adaptive immune cell senescence is influenced by circadian rhythms than at ZT5 (day), a phenotype that was abrogated in 2-AR–deficient
in aging and in the context of cancer. animals (70). Mice immunized at night displayed higher IgG1 and
IgM-specific antibody titers as well as increased antigen-specific
germinal center B cells and Tfh cells than day-immunized animals.
NEUROENDOCRINE CONTROL OF THE IMMUNE SYSTEM These results thus suggest that adrenergic signaling contributes to a
The nervous system directly conveys external, circadian signals to diurnal variation in the adaptive immune response, particularly with
an organism’s immune system, setting leukocytes in tune with respect to controlling the numbers of lymphocytes in LNs.
rhythms in the environment. The adrenergic branch of the sympa- Together with the sympathetic nervous system, the parasympa-
thetic nervous system is under circadian control and uses the cate- thetic nervous system has been shown to regulate circadian trafficking
cholamines epinephrine and norepinephrine as neurotransmitters of HSPCs and leukocytes (85). At night, cholinergic signals from the
to convey circadian oscillations (81). The central circadian clock, parasympathetic nervous system are antagonizing sympathetic-­
situated in the suprachiasmatic nucleus (SCN) of the hypothalamus derived noradrenergic signals. This has been suggested to repress 3-
(Box 1), synchronizes the rhythmic synthesis and secretion of cate- AR signaling at this time and suggested to result in reduced bone
cholamines, which ultimately results in the peripheral control of marrow egress of both HSPCs and leukocytes, whereas the 2-AR–
immune cell trafficking through AR signaling (52). mediated bone marrow homing capacity of cells is retained. During
One of the first evidence of adrenergic control over hematopoietic daytime, disinhibition of noradrenergic tone triggers egress of the cells
cell migration was uncovered by the mechanistic demonstration that from the bone marrow. Coordination between these two branches of
the circadian clock regulates hematopoietic stem and progenitor cell the nervous system has, thus, been suggested to result in diurnal oscil-
(HSPC) trafficking (82). The circadian clock synchronizes the local lations of HSPCs and leukocyte egress from the bone marrow (85).
secretion of norepinephrine that activates the 3-AR on bone marrow The central nervous system also coordinates circadian GC re-
stromal cells. Adrenergic signaling then results in the degradation of lease from the adrenal cortex in a time-of-day–dependent manner.
the transcription factor S1P and the down-regulation of the bone mar- GC levels peak at the beginning of the active phase in both mice and
row retention factors VCAM-1 and CXCL12 as well as endothelial humans and act by binding to the GR, promoting its translocation
selectins. These changes ultimately lead to the oscillatory mobilization to the nucleus where this complex acts as a transcription factor. GCs
of HSPCs from the bone marrow into the blood (82). In subsequent are best known for their immunosuppressive roles; however, they
years, these data were extended to demonstrate that a similar process are more accurately described as immune modulators that play
occurs in the recruitment of HSPCs to the bone marrow as well as in important immunostimulatory and inhibitory roles in addition to
myeloid cell recruitment to skeletal muscle (72). These rhythms in regulating circadian lymphocyte migration (53, 86). One example
homing are driven by circadian oscillations of VCAM-1, P-selectin, of this is the GC-regulated expression of the interleukin 7 receptor

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 8 of 15


SCIENCE IMMUNOLOGY | REVIEW

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-

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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
lymphocytes throughout the body. with streptomycin are more susceptible to oral Salmonella Typh-
imurium when infected early during the day (ZT4; higher bacterial
count in the colon and mesenteric LNs) compared with the night
GUT IMMUNITY (ZT16) (94). In contrast, nonantibiotic-treated mice infected with
There is a significant amount of evidence connecting circadian Salmonella Typhimurium orally at ZT12 exhibit an increased bacte-
rhythms to the function of the digestive system, gut microbiota, and rial burden in the SI 24 hours later and suffer from significantly in-
intestinal immunity. The timing of food intake exerts a strong creased mortality compared with those infected at ZT0 (morning)
influence on peripheral clocks and causes fluctuations in microbiota, (89). This demonstrates that preserving microbial diversity dramatically
gut cell motility, and nutrient absorption (88, 89). Food intake also changes infection outcome. This is due to food intake–driven oscil-
triggers rhythmic release of vasoactive intestinal peptide (VIP) from lations in the protein expression of antimicrobial peptides (AMPs)
enteric neurons in the intestine (90, 91). VIP binds to G protein– (89). Food intake peaks early in the active phase and promotes SFB
coupled receptor VIP receptor 2 (VIPR2) on immune cells, such as to associate with SIECs. This triggers rhythmic activation of an innate
intestinal innate lymphoid cells group 2 (ILC2s) and ILC3s, and in- immune circuit where a MyD88-dependent response in CD11c+
fluences their function in the gut (88, 90, 91). Although the impact myeloid cells activates ILC3s to secrete IL-22, leading to signal trans-
of food intake and microbiome activity on the circadian rhythm of ducer and activator of transcription 3 (STAT3) activation in SIECs
the organism has been recently reviewed (88, 89), here we will focus and circadian synthesis of AMPs (Reg3g, Lcn2, and S100A8) (89).
on how the circadian rhythm directs immune cells in the gut, par- These oscillations are completely ablated in fasting mice or in animals
ticularly ILC3s (Fig. 2 and Table 1). lacking SFB. They are also dependent on the expression of MyD88 in
CD11c+ cells (Cd11ccre:Myd88flox) and STAT3 in SIECs (Villincre:
Gut tolerance Stat3 flox), as well as the presence of gut ILC3s (89). Because
During periods of food intake (generally the behavioral active phase Salmonella Typhi­murium is resistant to Lcn2, it gains a competitive
of an organism), the gut is poised to absorb a maximum amount of advantage in the early active phase (ZT12), outcompeting the healthy
nutrients, which also facilitates microbiota to traverse the intestinal microbiota that is susceptible to Lcn2 (89). These results demonstrate
epithelial barrier. Recently, a study revealed that MHCII protein ex- how the host circadian rhythm affects microbial rhythms that, in turn,
pression on murine small intestine epithelial cells (SIECs) is rhyth- guide rhythmic host immunity.
mic, is Bmal1 dependent, and peaks in the resting phase (92). This
was paired with rhythmic permeability of the small intestine (SI), Regulation of ILC3s
which, in contrast, peaks during the active phase (92). Rhythmic In 2019, three papers were published exploring the role of the circa-
MHCII expression on SIECs, microbiota composition, and feeding dian clock in ILC3 homeostasis (Table 1) (95–98). All showed that
rhythms all contributed to circadian SI permeability, suggesting that intestinal ILC3s exhibit high clock gene mRNA expression com-
rhythmic SI permeability is due to oscillations in microbiota detec- pared with other immune and ILC cell subsets in the SI-LP, which
tion (92). Indeed, rhythmic MHCII expression on SIECs could be is in line with the critical role that Rorc (a clock gene that encodes
induced in germ-free mice after colonization with mouse segmented the transcription factor RORγt) and Nfil3 (a clock-associated
filamentous bacteria (SFB), but not with rat SFB, which is unable to gene) play as defining transcription factors in ILC3 development
attach to murine SIECs (92). SIECs lacking CDC42 expression, which (90, 95–97, 99–101). ILC3s also exhibit circadian oscillations of the
is essential for SFB to attach to the gut epithelium, also lost diurnal key transcription factors ROR and RUNX1, which are involved
MHCII expression (92). Furthermore, abrogation of MHCII ex- in ILC3 differentiation, as well as the ILC3 signature cytokines IL-17
pression in SIECs (Villin-cre:I-Abflox) reduced the numbers of CD4+ and IL-22 (90, 96, 97). ILC3s receive their circadian cues primarily
intraepithelial T cell lymphocytes in the gut and exacerbated the from light via the SCN, and removal of this brain region by

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 9 of 15


SCIENCE IMMUNOLOGY | REVIEW

A LN to the intestine, because these cells


display reduced surface protein ex-
pression of the gut-homing molecules
47, CCR9, and CXCR4 (95). However,
SI-LP ILC3s with an impaired molec-
ular clock exhibited increased frequency
of IL-17+ and IL-22+ cells and increased
expression of the proapoptotic Bcl-2–
like 11 protein BIM (96, 97). Together,
these data show gut ILC3s to be a highly
rhythmic immune cell population that
is synchronized by light.
Intriguingly, the effect of ablation of
B the circadian clock varies for different
ILC3 subsets. Although NKp46+ ILC3s
exhibited reduced numbers and in-

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


creased IL-22 protein expression, double-­
negative (DN; NKp46−CCR6−) and CCR6+
ILC3s exhibited increased numbers and
IL-17A protein expression when the
circadian clock was disrupted by loss of
REV-ERB. In line with this, REV-ERB–
deficient mice showed an altered chro-
matin landscape in the Il17a locus (97).
This suggests that lack of REV-ERB
C opens up ROR response elements (RORE)
binding sites at that site for additional
transcription factor binding and ulti-
mately increased IL-17 expression in DN
and CCR6+ ILC3s (97).
BMAL1 deficiency also impairs the
relationship between gut ILC3s and in-
testinal bacteria. The circadian oscillations
of microbiota populations are altered in
mice with BMAL1-deficient ILC3s, in-
dicating that the activated phenotype of
ILC3s lacking Bmal1 may be connected
Fig. 2. Circadian rhythms control the number and function of ILC3s in the SI. (A) During the active phase, ILC3s to the intestinal microbiota (95). In ad-
are present in low numbers and exhibit a nonactivated phenotype (low IL-17 and IL-22 expression) in the small intes- dition, when mice with BMAL1-deficient
tinal LP (96, 97). This is partially due to increased feeding-dependent VIP secretion from enteric VIP neurons during ILC3s are given antibiotics for 2 weeks,
the active phase (90, 91), which likely represses IL-22 expression by binding VIPR2 on ILC3s (91). Because IL-22 signals clearing much of the intestinal micro-
epithelial cells to express AMPs, low IL-22 levels allow microbes to thrive during the active/feeding phase, increasing biota, the frequency of IL-17+ and IL-22+
nutrient absorption (91). (B) During the rest phase, there are many ILC3s in the LP (96). IL-17 expression is increased ILC3s decreases and the total number
in ILC3s at this time (96, 97), and reduced food intake causes low VIP levels, promoting IL-22 expression by ILC3s (91).
of ILC3s in the intestine is restored to
These activated ILC3s promote antimicrobial functions and increase barrier maintenance, thus reducing nutrient
the level of ILC3-BMAL1–sufficient
absorption in the SI. (C) In the absence of a circadian rhythm, ILC3 numbers in the gut are dramatically reduced
(90, 95–97), but those that remain have an activated phenotype (96, 97). Mice with clock gene–deficient ILC3s also
animals (96). In agreement with this,
exhibit increased bacterial translocation during infection (95, 97), which may cause the high levels of IL-17 and IL-22 when mice with BMAL1-deficient ILC3s
on the LP ILC3s in these mice. were infected with Clostridium difficile
or Citrobacter rodentium, their ILC3s
electrolytic lesion or genetic ablation of Bmal1 from the forebrain were hyperresponsive and they had more bacterial translocation
CREDIT: A. MASTIN/SCIENCE IMMUNOLOLGY

(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

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 10 of 15


SCIENCE IMMUNOLOGY | REVIEW

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

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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
ant connection between circadian rhythm, VIP production, and CD8+IFN+ T cells compared with those infected at ZT20 (night)
ILC3 homeostasis. However, more research is necessary to elucidate (66). Together, these results highlight the importance of the circadian
the role of this neuroimmune axis in the gut. clock components in antiviral response.
Bacterial infections are also affected by circadian mechanisms.
As mentioned above, mice were shown to be more susceptible to oral
IMMUNE RESPONSES TO PATHOGENS Salmonella Typhimurium infection when gavaged in the morning
Although, initially, most studies focused on sterile inflammation to compared with the evening (94). This increased susceptibility resulted
study the influence of circadian rhythms, the importance of circadian in greater colonization of the colon by Salmonella Typhimurium
rhythms during pathogen infection has emerged as a key factor in and a greater proinflammatory response. Time-of-day–dependent
disease progression. This has become even more obvious in the con- susceptibility to bacterial infection was also shown in the context of
text of the severe acute respiratory syndrome coronavirus 2 (SARS- pulmonary S. pneumoniae, with mice infected in the morning
CoV-2) pandemic. (ZT0) experiencing higher lung and blood bacterial burden 48 hours
Many facets of SARS-CoV-2 infection and subsequent human after infection than mice infected in the evening (ZT12) (27).
immune responses are time-of-day dependent. SARS-CoV-2 viral Another example comes from the genital pathogen Chlamydia
uptake and replication in cultured human monocytes were found to muridarum, responsible for genital tract infection and causing
be regulated in a circadian manner, with higher entry and multipli- infertility (110). Mice infected at ZT3 (morning) had higher
cation of the virus at specific times compared with other times (102). C. muridarum infectivity compared with mice infected at ZT15
Furthermore, the pattern of secreted cytokines was dependent on (evening). This was accompanied by increased pathological lesions
the time of virus encounter. The highest virus load correlated with and higher protein levels of CXCL1, IL-10, IL-1, and IFN- 1 week
higher IL-6, IL-1, and IL-10 protein levels, suggesting that circadian after infection (110). Collectively, these results emphasize time-of-
rhythms affect both viral infection and the host immune response. day–dependent mechanisms in antibacterial defense.
In addition, SARS-CoV-2 entry and replication in human Calu-3 Pathogenic infection with parasites has also been shown to be
lung epithelial cells was shown in vitro to be under circadian regu- clock-regulated. Expulsion of the parasite Trichuris muris is con-
lation of both BMAL1 and REV-ERBs (103). The circadian clock trolled by the DC clock (111). Mice infected with this parasite in the
components repress angiotensin-converting enzyme 2, the primary morning showed a better clearance efficiency because of a stronger
entry receptor of SARS-CoV-2 in human epithelial cells, leading to TH2 immune response in the morning compared with the evening.
the limitation of virus entry and replication. These data make it clear In contrast, mice with BMAL1-­deficient DCs (Cd11ccre:BMAL1flox)
that the circadian oscillations play an important role in viral cell no longer showed this time-of-day difference. The importance of
invasion and the human immune response to the virus. circadian clocks in immune cells has also been demonstrated in
Besides SARS-CoV-2, circadian rhythms play important roles in the context of Leishmania major infection (112). L. major infection
other viral infections (Table 1). Viral replication and dissemination performed in the morning led to lower parasite burden and foot-
of both herpes and influenza A viruses depend on the time of infec- pad swelling compared with infection in the afternoon. This
tion, with mice infected at ZT0 (morning) with a luciferase-expressing was paralleled by a rhythmic infiltration of neutrophils and
herpes virus (M3:luc MuHV4) exhibiting 10-fold higher viral repli- macrophages to the site of infection, due to a rhythmic chemo-
cation than mice infected at ZT10 (evening) (104). However, this kine profile. The implication of the circadian clock in these phe-
time-of-day difference is abrogated in complete Bmal1−/− mice with nomena was confirmed by generating BMDMs from Bmal1−/− mice
levels of viral infection being higher than in wild-type animals. in vitro and in immune cells in vivo (using bone marrow chimera
Similar results were obtained for respiratory syncytial virus (RSV) from Bmal1−/− Rag2–deficient donors), which led to the ablation
and parainfluenza virus type 3, with higher viral replication of of all the described oscillations (112). Together, these findings

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 11 of 15


SCIENCE IMMUNOLOGY | REVIEW

Vaccination responses in humans also


exhibit circadian rhythmicity (Fig. 3). In
a cohort of 298 participants (>65 years
old), morning vaccination with an in-
fluenza vaccine was shown to induce a
higher antibody response compared with
afternoon vaccination (113). Another
influenza vaccination study suggested
that timing of sample collection could
explain the observed differences in
antibody titers between morning- and
afternoon-vaccinated participants (114).
Furthermore, vaccination with the anti-
tuberculosis agent Bacillus Calmette-
Guérin (BCG) in the morning was able
to better induce a nonspecific, trained

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


immunity response compared with later
times in the day (115). In view of the
current SARS-CoV-2 pandemic, adminis-
tration of a protein-based, inactivated
SARS-CoV-2 vaccine (BBIBP-CorV,
Sinopharm) in the morning was demon-
strated to enhance vaccine efficacy (116).
Participants that received morning doses
had a stronger immune response to the
vaccination, with a twofold higher level
of neutralizing antibodies compared
with the afternoon vaccination cohort.
Morning vaccination also yielded stronger
Fig. 3. Benefits of morning vaccination compared with afternoon/evening vaccination in humans. Immune
B cell and Tfh cell responses as well as
response to vaccination is time-of-day dependent with patients vaccinated in the morning experiencing a better
higher frequencies of monocytes and
response to vaccines compared with patients vaccinated in the afternoon/evening. This improved immune response
was observed for three vaccines—BCG (115), influenza (113), and SARS-CoV-2 (116)—and results in (i) greater anti-
DCs. Participants vaccinated in the
body response (113, 116), (ii) enhanced short-term immunity with higher cytokine production (IFN-, TNF-, and IL-1) morning additionally exhibited increased
(115, 116), and (iii) increased long-term immunity with increased frequencies of memory B cells (116). Mono, mono- long-term immunity and had higher
cytes; Mem B, memory B cell. percentages of spike- and RBD-specific
memory B cells (116,  117). A similar
show that oscillations in parasite infectivity and clearance are SARS-CoV-2 vaccination study performed on a U.K. cohort of health
dependent on immune cell clocks. care workers vaccinated with either Pfizer mRNA vaccine or AstraZeneca
adenoviral vaccine found that anti–spike protein responses were higher
in people who received their shot later in the day, suggesting that peak
IMMUNE RESPONSES TO VACCINATION times may vary between different vaccines or vaccination regimes
Given the importance of circadian rhythms in regulating many aspects (118). Overall, these data indicate that circadian rhythms play a role
of immune responses, it follows that circadian rhythms could be used in human vaccination responses and that those circadian rhythms
advantageously in targeted therapies such as vaccination protocols could be harnessed to optimize vaccination strategies.
(Table  1) (55). A pioneering work in mice demonstrated that
vaccination with OVA at ZT19 (night), using the TLR9 ligand
CpG-oligodeoxynucleotides as adjuvant, resulted in increased CONCLUDING REMARKS
antigen-induced lymphocyte proliferation and increased IFN- The recent years have provided substantial new arguments that
production compared with ZT7 (day) vaccination (73). This phe- rhythmicity significantly affects immune responses. In particular,
nomenon was correlated with a rhythmic expression of TLR9 on several recent advances in gut immunity have demonstrated gut-­
CREDIT: A. MASTIN/SCIENCE IMMUNOLOLGY

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

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 12 of 15


SCIENCE IMMUNOLOGY | REVIEW

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

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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.
Nat. Med. 20, 919–926 (2014).
are sure to provide answers to these exciting questions.
28. L. M. Ince, Z. Zhang, S. Beesley, R. M. Vonslow, B. R. Saer, L. C. Matthews, N. Begley,
J. E. Gibbs, D. W. Ray, A. S. I. Loudon, Circadian variation in pulmonary inflammatory
responses is independent of rhythmic glucocorticoid signaling in airway epithelial cells.
REFERENCES AND NOTES FASEB J. 33, 126–139 (2019).
1. F. Halberg, E. A. Johnson, B. W. Brown, J. J. Bittner, Susceptibility rhythm to E. coli 29. M. Pariollaud, J. E. Gibbs, T. W. Hopwood, S. Brown, N. Begley, R. Vonslow, T. Poolman,
endotoxin and bioassay. Proc. Soc. Exp. Biol. Med. 103, 142–144 (1960).
B. Guo, B. Saer, D. H. Jones, J. P. Tellam, S. Bresciani, N. C. Tomkinson, J. Wojno-Picon,
2. G. Fernandes, F. Halberg, E. J. Yunis, R. A. Good, Circadian rhythmic plaque-forming cell
A. W. Cooper, D. A. Daniels, R. P. Trump, D. Grant, W. Zuercher, T. M. Willson,
response of spleens from mice immunized with SRBC. J. Immunol. 117, 962–966 (1976).
A. S. MacDonald, B. Bolognese, P. L. Podolin, Y. Sanchez, A. S. Loudon, D. W. Ray, Circadian
3. W. He, S. Holtkamp, S. M. Hergenhan, K. Kraus, A. de Juan, J. Weber, P. Bradfield,
clock component REV-ERB controls homeostatic regulation of pulmonary inflammation.
J. M. P. Grenier, J. Pelletier, D. Druzd, C. S. Chen, L. M. Ince, S. Bierschenk, R. Pick,
J. Clin. Invest. 128, 2281–2296 (2018).
M. Sperandio, M. Aurrand-Lions, C. Scheiermann, Circadian expression of migratory
30. G. B. Kitchen, P. S. Cunningham, T. M. Poolman, M. Iqbal, R. Maidstone, M. Baxter,
factors establishes lineage-specific signatures that guide the homing of leukocyte
J. Bagnall, N. Begley, B. Saer, T. Hussell, L. C. Matthews, D. H. Dockrell, H. J. Durrington,
subsets to tissues. Immunity 49, 1175–1190.e7 (2018).
J. E. Gibbs, J. F. Blaikley, A. S. Loudon, D. W. Ray, The clock gene Bmal1 inhibits
4. C. Wyse, G. O'Malley, A. N. Coogan, S. McConkey, D. J. Smith, Seasonal and daytime
macrophage motility, phagocytosis, and impairs defense against pneumonia. Proc. Natl.
variation in multiple immune parameters in humans: Evidence from 329,261 participants
Acad. Sci. U.S.A. 117, 1543–1551 (2020).
of the UK Biobank cohort. iScience 24, 102255 (2021).
31. D. Guan, M. A. Lazar, Interconnections between circadian clocks and metabolism. J. Clin.
5. I. Cossio, D. Lucas, A. Hidalgo, Neutrophils as regulators of the hematopoietic niche. Blood
Invest. 131, e148278 (2021).
133, 2140–2148 (2019).
32. R. K. Alexander, Y. H. Liou, N. H. Knudsen, K. A. Starost, C. Xu, A. L. Hyde, S. Liu, D. Jacobi,
6. M. Baxter, D. W. Ray, Circadian rhythms in innate immunity and stress responses.
N. S. Liao, C. H. Lee, Bmal1 integrates mitochondrial metabolism and macrophage
Immunology 161, 261–267 (2020).
activation. eLife 9, e54090 (2020).
7. R. Pick, W. He, C. S. Chen, C. Scheiermann, Time-of-day-dependent trafficking
and function of leukocyte subsets. Trends Immunol. 40, 524–537 (2019). 33. E. J. Collins, M. P. Cervantes-Silva, G. A. Timmons, J. R. O'Siorain, A. M. Curtis, J. M. Hurley,
8. S. Wang, F. Li, Y. Lin, B. Wu, Targeting REV-ERB for therapeutic purposes: Promises Post-transcriptional circadian regulation in macrophages organizes temporally distinct
and challenges. Theranostics 10, 4168–4182 (2020). immunometabolic states. Genome Res. 31, 171–185 (2021).
9. G. A. Timmons, J. R. O'Siorain, O. D. Kennedy, A. M. Curtis, J. O. Early, Innate rhythms: Clocks 34. D. Yu, X. Fang, Y. Xu, H. Xiao, T. Huang, Y. Zhang, Y. Ge, Y. Li, L. Zong, J. Gao, Rev-erb can
at the center of monocyte and macrophage function. Front. Immunol. 11, 1743 (2020). regulate the NF-B/NALP3 pathway to modulate lipopolysaccharide-induced acute lung
10. E. E. Fortier, J. Rooney, H. Dardente, M. P. Hardy, N. Labrecque, N. Cermakian, Circadian injury and inflammation. Int. Immunopharmacol. 73, 312–320 (2019).
variation of the response of T cells to antigen. J. Immunol. 187, 6291–6300 (2011). 35. B. Pourcet, M. Zecchin, L. Ferri, J. Beauchamp, S. Sitaula, C. Billon, S. Delhaye, J. Vanhoutte,
11. T. Bollinger, A. Leutz, A. Leliavski, L. Skrum, J. Kovac, L. Bonacina, C. Benedict, T. Lange, A. Mayeuf-Louchart, Q. Thorel, J. T. Haas, J. Eeckhoute, D. Dombrowicz, C. Duhem,
J. Westermann, H. Oster, W. Solbach, Circadian clocks in mouse and human CD4+ T cells. A. Boulinguiez, S. Lancel, Y. Sebti, T. P. Burris, B. Staels, H. M. Duez, Nuclear receptor
PLOS ONE 6, e29801 (2011). subfamily 1 group d member 1 regulates circadian activity of NLRP3 inflammasome to reduce
12. E. Challet, The circadian regulation of food intake. Nat. Rev. Endocrinol. 15, 393–405 (2019). the severity of fulminant hepatitis in mice. Gastroenterology 154, 1449–1464.e20 (2018).
13. K. Wager-Smith, S. A. Kay, Circadian rhythm genetics: From flies to mice to humans. 36. B. Pourcet, H. Duez, Circadian control of inflammasome pathways: Implications
Nat. Genet. 26, 23–27 (2000). for circadian medicine. Front. Immunol. 11, 1630 (2020).
14. D. Stoleru, Y. Peng, P. Nawathean, M. Rosbash, A resetting signal between Drosophila 37. J. O. Early, D. Menon, C. A. Wyse, M. P. Cervantes-Silva, Z. Zaslona, R. G. Carroll,
pacemakers synchronizes morning and evening activity. Nature 438, 238–242 (2005). E. M. Palsson-McDermott, S. Angiari, D. G. Ryan, S. E. Corcoran, G. Timmons, S. S. Geiger,
15. J. S. Takahashi, Transcriptional architecture of the mammalian circadian clock. Nat. Rev. D. J. Fitzpatrick, D. O'Connell, R. J. Xavier, K. Hokamp, L. A. J. O'Neill, A. M. Curtis, Circadian
Genet. 18, 164–179 (2017). clock protein BMAL1 regulates IL-1 in macrophages via NRF2. Proc. Natl. Acad. Sci. U.S.A.
16. C. Scheiermann, J. Gibbs, L. Ince, A. Loudon, Clocking in to immunity. Nat. Rev. Immunol. 115, E8460–E8468 (2018).
18, 423–437 (2018). 38. S. Wang, Y. Lin, X. Yuan, F. Li, L. Guo, B. Wu, REV-ERB integrates colon clock with
17. M. K. Bunger, L. D. Wilsbacher, S. M. Moran, C. Clendenin, L. A. Radcliffe, J. B. Hogenesch, experimental colitis through regulation of NF-B/NLRP3 axis. Nat. Commun. 9, 4246 (2018).
M. C. Simon, J. S. Takahashi, C. A. Bradfield, Mop3 is an essential component of the master 39. S. Wang, Y. Lin, F. Li, Z. Qin, Z. Zhou, L. Gao, Z. Yang, Z. Wang, B. Wu, An NF-B-driven
circadian pacemaker in mammals. Cell 103, 1009–1017 (2000). lncRNA orchestrates colitis and circadian clock. Sci. Adv. 6, eabb5202 (2020).
18. J. S. O'Neill, G. van Ooijen, L. E. Dixon, C. Troein, F. Corellou, F. Y. Bouget, A. B. Reddy, A. J. Millar, 40. J. E. Gibbs, J. Blaikley, S. Beesley, L. Matthews, K. D. Simpson, S. H. Boyce, S. N. Farrow,
Circadian rhythms persist without transcription in a eukaryote. Nature 469, 554–558 (2011). K. J. Else, D. Singh, D. W. Ray, A. S. Loudon, The nuclear receptor REV-ERB mediates
19. R. D. Feigin, V. H. San Joaquin, M. W. Haymond, R. G. Wyatt, Daily periodicity circadian regulation of innate immunity through selective regulation of inflammatory
of susceptibility of mice to pneumococcal infection. Nature 224, 379–380 (1969). cytokines. Proc. Natl. Acad. Sci. U.S.A. 109, 582–587 (2012).

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 13 of 15


SCIENCE IMMUNOLOGY | REVIEW

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 RORt 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,

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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).
M. Leiva, G. Sabio, Neutrophil infiltration regulates clock-gene expression to organize 67. E. Haus, M. H. Smolensky, Biologic rhythms in the immune system. Chronobiol. Int. 16,
daily hepatic metabolism. Elife 9, e59258 (2020). 581–622 (1999).
47. M. Casanova-Acebes, J. A. Nicolas-Avila, J. L. Li, S. Garcia-Silva, A. Balachander, 68. A. Nakai, Y. Hayano, F. Furuta, M. Noda, K. Suzuki, Control of lymphocyte egress from
A. Rubio-Ponce, L. A. Weiss, J. M. Adrover, K. Burrows, N. A-González, I. Ballesteros, S. Devi, lymph nodes through 2-adrenergic receptors. J. Exp. Med. 211, 2583–2598 (2014).
J. A. Quintana, G. Crainiciuc, M. Leiva, M. Gunzer, C. Weber, T. Nagasawa, O. Soehnlein, 69. A. Shimba, G. Cui, S. Tani-Ichi, M. Ogawa, S. Abe, F. Okazaki, S. Kitano, H. Miyachi,
M. Merad, A. Mortha, L. G. Ng, H. Peinado, A. Hidalgo, Neutrophils instruct homeostatic H. Yamada, T. Hara, Y. Yoshikai, T. Nagasawa, G. Schutz, K. Ikuta, Glucocorticoids drive
and pathological states in naive tissues. J. Exp. Med. 215, 2778–2795 (2018). diurnal oscillations in T cell distribution and responses by inducing interleukin-7 receptor
48. S. J. Holtkamp, L. M. Ince, C. Barnoud, M. T. Schmitt, F. Sinturel, V. Pilorz, R. Pick, S. Jemelin, and CXCR4. Immunity 48, 286–298.e6 (2018).
M. Muhlstadt, W. H. Boehncke, J. Weber, D. Laubender, J. Philippou-Massier, C. S. Chen, 70. K. Suzuki, Y. Hayano, A. Nakai, F. Furuta, M. Noda, Adrenergic control of the adaptive
L. Holtermann, D. Vestweber, M. Sperandio, B. U. Schraml, C. Halin, C. Dibner, H. Oster, immune response by diurnal lymphocyte recirculation through lymph nodes. J. Exp. Med.
J. Renkawitz, C. Scheiermann, Circadian clocks guide dendritic cells into skin lymphatics. 213, 2567–2574 (2016).
Nat. Immunol. 22, 1375–1381 (2021). 71. S. D. House, S. Ruch, W. F. Koscienski III, C. W. Rocholl, R. L. Moldow, Effects
49. C. C. Nobis, G. Dubeau Laramee, L. Kervezee, D. Maurice De Sousa, N. Labrecque, of the circadian rhythm of corticosteroids on leukocyte-endothelium interactions
N. Cermakian, The circadian clock of CD8 T cells modulates their early response in the AM and PM. Life Sci. 60, 2023–2034 (1997).
to vaccination and the rhythmicity of related signaling pathways. Proc. Natl. Acad. Sci. 72. C. Scheiermann, Y. Kunisaki, D. Lucas, A. Chow, J. E. Jang, D. Zhang, D. Hashimoto,
U.S.A. 116, 20077–20086 (2019). M. Merad, P. S. Frenette, Adrenergic nerves govern circadian leukocyte recruitment
50. M. Amir, S. Campbell, T. M. Kamenecka, L. A. Solt, Pharmacological modulation to tissues. Immunity 37, 290–301 (2012).
and genetic deletion of REV-ERB and REV-ERB regulates dendritic cell development. 73. A. C. Silver, A. Arjona, W. E. Walker, E. Fikrig, The circadian clock controls toll-like receptor
Biochem. Biophys. Res. Commun. 527, 1000–1007 (2020). 9-mediated innate and adaptive immunity. Immunity 36, 251–261 (2012).
51. A. Nakai, K. Suzuki, Adrenergic control of lymphocyte trafficking and adaptive immune 74. C. E. Sutton, C. M. Finlay, M. Raverdeau, J. O. Early, J. DeCourcey, Z. Zaslona, L. A. J. O'Neill,
responses. Neurochem. Int. 130, 104320 (2019). K. H. G. Mills, A. M. Curtis, Loss of the molecular clock in myeloid cells exacerbates
52. C. Scheiermann, Y. Kunisaki, P. S. Frenette, Circadian control of the immune system. T cell-mediated CNS autoimmune disease. Nat. Commun. 8, 1923 (2017).
Nat. Rev. Immunol. 13, 190–198 (2013). 75. N. Ando, Y. Nakamura, R. Aoki, K. Ishimaru, H. Ogawa, K. Okumura, S. Shibata, S. Shimada,
53. A. Shimba, K. Ikuta, Glucocorticoids regulate circadian rhythm of innate and adaptive A. Nakao, Circadian gene clock regulates psoriasis-like skin inflammation in mice.
immunity. Front. Immunol. 11, 2143 (2020). J. Invest. Dermatol. 135, 3001–3008 (2015).
54. N. Labrecque, N. Cermakian, Circadian clocks in the immune system. J. Biol. Rhythms 30, 76. A. Chiricozzi, R. Saraceno, M. S. Chimenti, E. Guttman-Yassky, J. G. Krueger, Role of IL-23
277–290 (2015). in the pathogenesis of psoriasis: A novel potential therapeutic target? Expert Opin. Ther.
55. N. Cermakian, S. K. Stegeman, K. Tekade, N. Labrecque, Circadian rhythms in adaptive Targets 18, 513–525 (2014).
immunity and vaccination. Semin. Immunopathol. 44, 193–207 (2021). 77. S. Wang, M. Kozai, H. Mita, Z. Cai, M. A. Masum, O. Ichii, K. Takada, M. Inaba, REV-ERB
56. S. Hemmers, A. Y. Rudensky, The cell-intrinsic circadian clock is dispensable agonist suppresses IL-17 production in T cells and improves psoriatic dermatitis
for lymphocyte differentiation and function. Cell Rep. 11, 1339–1349 (2015). in a mouse model. Biomed. Pharmacother. 144, 112283 (2021).
57. X. Yu, D. Rollins, K. A. Ruhn, J. J. Stubblefield, C. B. Green, M. Kashiwada, P. B. Rothman, 78. A. J. Davidson, M. T. Sellix, J. Daniel, S. Yamazaki, M. Menaker, G. D. Block, Chronic jet-lag
J. S. Takahashi, L. V. Hooper, TH17 cell differentiation is regulated by the circadian clock. increases mortality in aged mice. Curr. Biol. 16, R914–R916 (2006).
Science 342, 727–730 (2013). 79. H. Inokawa, Y. Umemura, A. Shimba, E. Kawakami, N. Koike, Y. Tsuchiya, M. Ohashi,
58. C. Chang, C. S. Loo, X. Zhao, L. A. Solt, Y. Liang, S. P. Bapat, H. Cho, T. M. Kamenecka, Y. Minami, G. Cui, T. Asahi, R. Ono, Y. Sasawaki, E. Konishi, S. H. Yoo, Z. Chen, S. Teramukai,
M. Leblanc, A. R. Atkins, R. T. Yu, M. Downes, T. P. Burris, R. M. Evans, Y. Zheng, The nuclear K. Ikuta, K. Yagita, Chronic circadian misalignment accelerates immune senescence
receptor REV-ERB modulates Th17 cell-mediated autoimmune disease. Proc. Natl. Acad. and abbreviates lifespan in mice. Sci. Rep. 10, 2569 (2020).
Sci. U.S.A. 116, 18528–18536 (2019). 80. Y. Wu, B. Tao, T. Zhang, Y. Fan, R. Mao, Pan-cancer analysis reveals disrupted circadian
59. M. Amir, S. Chaudhari, R. Wang, S. Campbell, S. A. Mosure, L. B. Chopp, Q. Lu, J. Shang, clock associates with T cell exhaustion. Front. Immunol. 10, 2451 (2019).
O. B. Pelletier, Y. He, C. Doebelin, M. D. Cameron, D. J. Kojetin, T. M. Kamenecka, L. A. Solt, 81. S. Leach, K. Suzuki, Adrenergic signaling in circadian control of immunity. Front. Immunol.
REV-ERBalpha regulates TH17 cell development and autoimmunity. Cell Rep. 25, 11, 1235 (2020).
3733–3749.e8 (2018). 82. S. Mendez-Ferrer, D. Lucas, M. Battista, P. S. Frenette, Haematopoietic stem cell release is
60. M. Ciofani, A. Madar, C. Galan, M. Sellars, K. Mace, F. Pauli, A. Agarwal, W. Huang, regulated by circadian oscillations. Nature 452, 442–447 (2008).
C. N. Parkhurst, M. Muratet, K. M. Newberry, S. Meadows, A. Greenfield, Y. Yang, P. Jain, 83. K. Golan, A. Kumari, O. Kollet, E. Khatib-Massalha, M. D. Subramaniam, Z. S. Ferreira,
F. K. Kirigin, C. Birchmeier, E. F. Wagner, K. M. Murphy, R. M. Myers, R. Bonneau, F. Avemaria, S. Rzeszotek, A. Garcia-Garcia, S. Xie, E. Flores-Figueroa, S. Gur-Cohen,
D. R. Littman, A validated regulatory network for Th17 cell specification. Cell 151, T. Itkin, A. Ludin-Tal, H. Massalha, B. Bernshtein, A. K. Ciechanowicz, A. Brandis,
289–303 (2012). T. Mehlman, S. Bhattacharya, M. Bertagna, H. Cheng, E. Petrovich-Kopitman, T. Janus,

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 14 of 15


SCIENCE IMMUNOLOGY | REVIEW

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).

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


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
109. S. Sengupta, S. Y. Tang, J. C. Devine, S. T. Anderson, S. Nayak, S. L. Zhang, A. Valenzuela,
neuropeptide VIP confers anticipatory mucosal immunity by regulating ILC3 activity. Nat.
D. G. Fisher, G. R. Grant, C. B. Lopez, G. A. FitzGerald, Circadian control of lung
Immunol. 21, 168–177 (2020).
inflammation in influenza infection. Nat. Commun. 10, 4107 (2019).
91. J. Talbot, P. Hahn, L. Kroehling, H. Nguyen, D. Li, D. R. Littman, Feeding-dependent VIP 110. S. R. Lundy, T. Ahmad, T. Simoneaux, I. Benyeogor, Y. Robinson, Z. George, D. Ellerson,
neuron-ILC3 circuit regulates the intestinal barrier. Nature 579, 575–580 (2020). W. Kirlin, T. Omosun, F. O. Eko, C. M. Black, U. Blas-Machado, J. P. DeBruyne,
92. T. Tuganbaev, U. Mor, S. Bashiardes, T. Liwinski, S. P. Nobs, A. Leshem, M. Dori-Bachash, J. U. Igietseme, Q. He, Y. O. Omosun, Effect of time of day of infection on chlamydia
C. A. Thaiss, E. Y. Pinker, K. Ratiner, L. Adlung, S. Federici, C. Kleimeyer, C. Moresi, infectivity and pathogenesis. Sci. Rep. 9, 11405 (2019).
T. Yamada, Y. Cohen, X. Zhang, H. Massalha, E. Massasa, Y. Kuperman, P. A. Koni, 111. T. W. Hopwood, S. Hall, N. Begley, R. Forman, S. Brown, R. Vonslow, B. Saer, M. C. Little,
A. Harmelin, N. Gao, S. Itzkovitz, K. Honda, H. Shapiro, E. Elinav, Diet diurnally regulates E. A. Murphy, R. J. Hurst, D. W. Ray, A. S. MacDonald, A. Brass, D. A. Bechtold, J. E. Gibbs,
small intestinal microbiome-epithelial-immune homeostasis and enteritis. Cell 182, A. S. Loudon, K. J. Else, The circadian regulator BMAL1 programmes responses to parasitic
1441–1459.e21 (2020). worm infection via a dendritic cell clock. Sci. Rep. 8, 3782 (2018).
93. C. A. Thaiss, M. Levy, T. Korem, L. Dohnalova, H. Shapiro, D. A. Jaitin, E. David, D. R. Winter, 112. S. Kiessling, G. Dubeau-Laramee, H. Ohm, N. Labrecque, M. Olivier, N. Cermakian, The
M. Gury-BenAri, E. Tatirovsky, T. Tuganbaev, S. Federici, N. Zmora, D. Zeevi, M. Dori-Bachash, circadian clock in immune cells controls the magnitude of Leishmania parasite infection.
M. Pevsner-Fischer, E. Kartvelishvily, A. Brandis, A. Harmelin, O. Shibolet, Z. Halpern, Sci. Rep. 7, 10892 (2017).
K. Honda, I. Amit, E. Segal, E. Elinav, Microbiota diurnal rhythmicity programs host 113. J. E. Long, M. T. Drayson, A. E. Taylor, K. M. Toellner, J. M. Lord, A. C. Phillips, Morning
transcriptome oscillations. Cell 167, 1495–1510.e12 (2016). vaccination enhances antibody response over afternoon vaccination: A cluster-
94. M. M. Bellet, E. Deriu, J. Z. Liu, B. Grimaldi, C. Blaschitz, M. Zeller, R. A. Edwards, S. Sahar, randomised trial. Vaccine 34, 2679–2685 (2016).
S. Dandekar, P. Baldi, M. D. George, M. Raffatellu, P. Sassone-Corsi, Circadian clock regulates 114. R. K. Kurupati, A. Kossenkoff, S. Kannan, L. H. Haut, S. Doyle, X. Yin, K. E. Schmader, Q. Liu,
the host response to Salmonella. Proc. Natl. Acad. Sci. U.S.A. 110, 9897–9902 (2013). L. Showe, H. C. J. Ertl, The effect of timing of influenza vaccination and sample collection
95. C. Godinho-Silva, R. G. Domingues, M. Rendas, B. Raposo, H. Ribeiro, J. A. da Silva, on antibody titers and responses in the aged. Vaccine 35, 3700–3708 (2017).
A. Vieira, R. M. Costa, N. L. Barbosa-Morais, T. Carvalho, H. Veiga-Fernandes, Light- 115. L. C. J. de Bree, V. P. Mourits, V. A. Koeken, S. J. Moorlag, R. Janssen, L. Folkman,
entrained and brain-tuned circadian circuits regulate ILC3s and gut homeostasis. Nature D. Barreca, T. Krausgruber, V. Fife-Gernedl, B. Novakovic, R. J. Arts, H. Dijkstra,
574, 254–258 (2019). H. Lemmers, C. Bock, L. A. Joosten, R. van Crevel, C. S. Benn, M. G. Netea, Circadian
96. F. Teng, J. Goc, L. Zhou, C. Chu, M. A. Shah, G. Eberl, G. F. Sonnenberg, A circadian clock is rhythm influences induction of trained immunity by BCG vaccination. J. Clin. Invest. 130,
essential for homeostasis of group 3 innate lymphoid cells in the gut. Sci. Immunol. 4, 5603–5617 (2020).
eaax1215 (2019). 116. H. Zhang, Y. Liu, D. Liu, Q. Zeng, L. Li, Q. Zhou, M. Li, J. Mei, N. Yang, S. Mo, Q. Liu, M. Liu,
97. Q. Wang, M. L. Robinette, C. Billon, P. L. Collins, J. K. Bando, J. L. Fachi, C. Secca, S. I. Porter, S. Peng, H. Xiao, Time of day influences immune response to an inactivated vaccine
A. Saini, S. Gilfillan, L. A. Solt, E. S. Musiek, E. M. Oltz, T. P. Burris, M. Colonna, Circadian against SARS-CoV-2. Cell Res. 31, 1215–1217 (2021).
rhythm–dependent and circadian rhythm–independent impacts of the molecular clock 117. C. Barnoud, C. Wang, C. Scheiermann, Timing vaccination against SARS-CoV-2. Cell Res.
on type 3 innate lymphoid cells. Sci. Immunol. 4, eaay7501 (2019). 31, 1146–1147 (2021).
98. Q. Wang, M. Colonna, Keeping time in group 3 innate lymphoid cells. Nat. Rev. Immunol. 118. W. Wang, P. Balfe, D. W. Eyre, S. F. Lumley, D. O'Donnell, F. Warren, D. W. Crook, K. Jeffery,
20, 720–726 (2020). P. C. Matthews, E. B. Klerman, J. A. McKeating, Time of day of vaccination affects
99. C. Seillet, L. C. Rankin, J. R. Groom, L. A. Mielke, J. Tellier, M. Chopin, N. D. Huntington, SARS-CoV-2 antibody responses in an observational study of health care workers.
G. T. Belz, S. Carotta, Nfil3 is required for the development of all innate lymphoid cell J. Biol. Rhythms 37, 124–129 (2022).
subsets. J. Exp. Med. 211, 1733–1740 (2014).
100. T. L. Geiger, M. C. Abt, G. Gasteiger, M. A. Firth, M. H. O'Connor, C. D. Geary, T. E. O'Sullivan,
M. R. van den Brink, E. G. Pamer, A. M. Hanash, J. C. Sun, Nfil3 is crucial for development Funding: Research in the laboratory is funded by the European Research Council (ERC CoG
of innate lymphoid cells and host protection against intestinal pathogens. J. Exp. Med. 101001233, CIRCADYN), the Swiss National Science Foundation (SNF; 310030_182417/1), the
211, 1723–1731 (2014). Swiss Cancer League (KLS-4836-08-2019), the Geneva Cancer league (2106), the Fondation
101. W. Xu, R. G. Domingues, D. Fonseca-Pereira, M. Ferreira, H. Ribeiro, S. Lopez-Lastra, privée des HUG (RC05-03), EU ITN (813284, INTEGRATA), and the Novartis Foundation for
Y. Motomura, L. Moreira-Santos, F. Bihl, V. Braud, B. Kee, H. Brady, M. C. Coles, Medical-Biological Research (20A019); C.W. is the recipient of a fellowship of the Chinese
C. Vosshenrich, M. Kubo, J. P. Di Santo, H. Veiga-Fernandes, NFIL3 orchestrates Scholarship Council (CSC). Competing interests: The authors declare that they have no
the emergence of common helper innate lymphoid cell precursors. Cell Rep. 10, competing interests.
2043–2054 (2015).
102. A. B. Diallo, L. Gay, B. Coiffard, M. Leone, S. Mezouar, J. L. Mege, Daytime variation Submitted 15 December 2021
in SARS-CoV-2 infection and cytokine production. Microb. Pathog. 158, 105067 (2021). Accepted 9 May 2022
103. X. Zhuang, S. Tsukuda, F. Wrensch, P. A. C. Wing, M. Schilling, J. M. Harris, H. Borrmann, Published 3 June 2022
S. B. Morgan, J. L. Cane, L. Mailly, N. Thakur, C. Conceicao, H. Sanghani, L. Heydmann, 10.1126/sciimmunol.abm2465

Wang et al., Sci. Immunol. 7, eabm2465 (2022) 3 June 2022 15 of 15


The circadian immune system
Chen Wang, Lydia Kay Lutes, Coline Barnoud, and Christoph Scheiermann

Sci. Immunol., 7 (72), eabm2465.


DOI: 10.1126/sciimmunol.abm2465

Downloaded from https://www.science.org at Universidade de Sao Paulo on May 03, 2023


View the article online
https://www.science.org/doi/10.1126/sciimmunol.abm2465
Permissions
https://www.science.org/help/reprints-and-permissions

Use of this article is subject to the Terms of service

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

You might also like