Sie sind auf Seite 1von 15

REVIEWS

­Clocking in to immunity
Christoph Scheiermann1,2*, Julie Gibbs3, Louise Ince   1 and Andrew Loudon3*
Abstract | Circadian rhythms are a ubiquitous feature of virtually all living organisms, regulating a
wide diversity of physiological systems. It has long been established that the circadian clockwork
plays a key role in innate immune responses, and recent studies reveal that several aspects of
adaptive immunity are also under circadian control. We discuss the latest insights into the genetic
and biochemical mechanisms linking immunity to the core circadian clock of the cell and
hypothesize as to why the immune system is so tightly controlled by circadian oscillations. Finally ,
we consider implications for human health, including vaccination strategies and the emerging
field of chrono-​immunotherapy.

Circadian
The regular 24 h environmental cycles generated by secretion of hormones (such as glucocorticoids)7 and
A free-​running rhythm with a the planet’s rotation have led to the evolution of daily metabolic cues8, which are driven by rhythmic feeding
period of approximately 24 h circadian rhythms in almost all life forms on Earth. behaviour9,10. Although light is the key entrainment
that persists in the absence of Circadian rhythms are driven by cell-​autonomous bio- factor for the SCN, feeding-​regulated metabolic cues
external entrainment, such as
logical clocks (Box 1), which allow organisms to adapt are now recognized as being pivotal to the regulation of
in constant darkness.
to and anticipate temporal changes in the environment. many peripheral clocks9,10.
Suprachiasmatic nuclei These biological clocks probably evolved a few billion Phase coherence of the organism to the normally
(SCN). A bilateral structure in years ago, perhaps in response to the need for single-​ occurring environmental light–dark cycle is known to
the anterior hypothalamus,
celled eukaryotes to avoid the damaging effects of ioniz- maintain organismal fitness, as animal studies show
home to the central pacemaker,
which processes light input and
ing radiation and oxidative stress during processes such that its disruption by abnormal lighting or feeding
conveys timing information to as cell division in which single-​stranded nucleic acids schedules, or due to genetic mutation of key circa-
the rest of the body. become exposed1,2. In higher organisms such as mam- dian timing genes, induces pathological changes11. In
mals, virtually all aspects of physiology are regulated by support of this, human lifestyles that disrupt inherent
internal circadian clocks, including sleep–wake cycles, timing systems (for example, exposure to abnormal
behaviour and locomotor activity, body temperature lighting schedules in chronic shift work) are associated
cycles, cardiovascular and digestive processes, endocrine with an increased risk of cancer, metabolic disorders
systems and metabolic and immune functions. and cardiovascular and cerebrovascular disease12–15. In
In the mammalian circadian system, peripheral addition, many human inflammatory diseases exhibit
clocks are normally maintained in phase coherence with rhythmicity in their pathology, including myocardial
1
Walter Brendel Centre of the environment by entrainment from daily exposure infarction, asthma and rheumatoid arthritis, as we
Experimental Medicine, to the light–dark cycle. This photic entrainment pathway discuss in detail later9,16,17.
University Hospital, Ludwig-​
operates via retinal innervation of a specialized neural Diurnal host responses to lethal infection and endo-
Maximilians–University
Munich, Biomedical Centre, circadian pacemaker of around 20,000 neurons located toxins were first demonstrated almost 60 years ago18, but
Planegg, Martinsried, in a pair of hypothalamic suprachiasmatic nuclei (SCN)3. it is only recently that studies have uncovered multiple
Germany. The mammalian SCN pacemaker has two unique prop- widespread aspects of immune functions that are under
2
DZHK (German Centre for erties: first, it is the only mammalian circadian struc- the control of the circadian clockwork. These include
Cardiovascular Research), ture that is directly entrainable to light. Second, the the trafficking of immune cells, host–pathogen interac-
partner site Munich Heart
Alliance, Munich, Germany.
SCN neurons remain coupled in ectopic culture con- tions and activation of innate and adaptive immunity,
ditions, persistently generating robust rhythmic circa- which have been discussed in recent reviews19–22. It is
3
School of Medical Sciences,
Faculty of Biology, Medicine dian cycles of electrical activity and gene expression for now apparent that circadian signals operate as a gate to
and Health, University of many months outside the body4,5. By contrast, periph- control the magnitude of immune responses in a pow-
Manchester, Manchester, UK. eral circadian oscillators lose c­ir­ca­dian synchrony in erful time-​of-day manner. In this Review, we describe
*e-​mail: christoph. culture because individual cells are unable to maintain the mechanisms that place the immune system under
scheiermann@ coupled phase coherence. A­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­u­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­t­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­o­­­­­­n­­­­­­o­­m­ous c­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­i­­­­­­­­­­­­­­­­­­­­r­­­­­­­­­­­­­­­­­­­­c­­­a­­­­dian c­­­­l­­o­­­cks clock control. We address what is known of the mole­
med.uni-​muenchen.de;
h­­a­v­e b­­e­e­n d­­e­t­­ec­­ted i­­n a­­l­l major organs and tissues and cular elements that couple the core circadian clockwork
andrew.loudon@
manchester.ac.uk within many cell types of the body. Normal circadian to specific output arms of the immune system and
https://doi.org/10.1038/ synchrony of peripheral cells and tissues is maintained how these new understandings may lead to improved
s41577-018-0008-4 by a complex network involving neuronal signalling6, chronotherapeutic options for treatment of disease.

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Fig. 1 | operation and disruption of the molecular clock ◀


Box 1 | How the clock works
in immune cells. a | The core molecular clockwork (centre)
the mammalian circadian core clockwork consists of a conserved set of transcription consists of interlocked transcription– translation feedback
factors operating as interlocked rhythmic transcription–translation feedback loops. loops within each cell (Box 1). Rhythmic expression of
Central to this are the key regulatory transcriptional activators circadian locomoter these clock genes (and other clock-​controlled genes) has
output cycles protein kaput (CLOCK), brain and muscle arNt-​like 1 (BMaL1) and been described in cells of both the innate and adaptive
neuronal Pas domain-​containing protein 2 (neuronal Pas2), which form the immune system, including macrophages24,25 and
heterodimeric complexes CLOCK–BMaL1 and neuronal Pas2–BMaL1 and act on e-​box microglia129, monocytes23, mast cells26,27, dendritic cells
elements on target genes. these target genes include the circadian transcription (DCs)25, CD4+ T cells54,56, B cells25, natural killer (NK) cells29,
factors period circadian protein homologue 1 (PER1) and PER2, cryptochrome 1 (CRY1) neutrophils28 and eosinophils27 as indicated by the clock
and CRY2, rev-​erBα (NR1D1) and rev-​erBβ (NR1D2) and differentially expressed in symbol. Other cell types such as CD8+ T cells54 and group 2
chondrocytes protein 1 (DEC1) and DEC2 — all of which are repressors of the core innate lymphoid cells (ILC2s)46 exhibit rhythmic function,
clockwork. rhythmic negative feedback by Per and CrY proteins on e-​box sites leads but clock gene expression has not been formally
to the transcriptional repression of CLOCK–BMaL1 complexes on target genes, demonstrated. In addition, circadian oscillations in gene
including PER and CRY genes. this repression is lifted late at night owing to decay of the expression have also been reported in stromal cells such as
Per–CrY complex and structural changes in the BMaL1 protein117. the CLOCK–BMaL1 synoviocytes116, epithelial cells43,80 and blood vessels130,
complex can then reactivate transcription, leading to the generation of an oscillation. which are known to modify immune function. b | Targeted
Clock pace is strongly regulated by post-​translational control of feedback elements. disruption of the clock in specific cell types has been
Genetic mutations altering the phosphorylation status, mobility or degradation of Per achieved largely by using the Cre-​loxP system. Crossing
and CrY proteins can have a profound effect on clock speed118,119. two additional Bmal1flox/flox mice with cell-​specific Cre lines abolishes
circuits cooperate with the core feedback loop to establish robust 24 h rhythms. First, the molecular clockwork in those cells23,31,43,54,55,78,79,131. The
the retinoid-​related orphan receptor (rOr) nuclear receptors drive expression of effects of such a disruption are varied, encompassing both
BMaL1 in a feedforward loop. rOrα shares a DNa-​binding site with rev-​erBs, and pro-​inflammatory and anti-​inflammatory responses, and
rhythmic competitive repression by rev-​erBs generates an oscillation in BMAL1 may depend upon the timing of the immune challenge.
expression. second, BMaL1–CLOCK heterodimers act on a family of Par domain basic BMAL1, brain and muscle ARNT-​like 1; CLOCK , circadian
leucine zipper transcription factors — namely, albumin D box-​binding protein (DBP), locomoter output cycles protein kaput; CNS, central
hepatic leukaemia factor (HLF) and thyrotroph embryonic factor (teF). these act on nervous system; CRY, cryptochrome; CT0, circadian time 0;
D-​box elements in target genes, including those of the core feedback loop; for example, CXCL5, CXC-​chemokine ligand 5; EAE, experimental
DBP positively regulates PER1 expression120. Consequently, the interlocked loops of the autoimmune encephalomyelitis; IEC, intestinal epithelial
core circadian clock act via rhythmic control of e-​box, rOr or D-​box sites to generate cell; LPS, lipopolysaccharide; neuronal PAS2, neuronal PAS
stable oscillations in target gene expression121. thus, the formation, trafficking and domain-​containing protein 2; PER , period circadian protein
degradation of different clock protein complexes throughout this transcriptional cycle homologue; ROR , retinoid-​related orphan receptor ; TNF,
generate the intrinsic nature and stability of the clock. tumour necrosis factor.

Diurnal
Core clockwork in innate immunity (including adhesion, migration and integrin activation)
A pattern that occurs over the Multiple types of innate immune cells have been via direct regulation of CC-​chemokine ligand 2 (Ccl2)
course of a day in which demonstrated to possess intrinsic clocks, including through a REV-​ERB binding motif in the promoter37
external entrainment (such as monocytes23, macrophages24,25, mast cells26,27, neutro- (Fig.  2a) . Although not cell-​s pecific, cryptochromes
light–dark cycles) is used; the
phils28, eosinophils27 and natural killer (NK) cells29 (CRYs) also directly affect inflammatory pathways
onset of the light cycle is
defined as Zeitgeber time (Fig. 1a). These intrinsic timers affect the function of through action on adenylyl cyclase38. In the absence of
0 (ZT0). these cells, driving temporal gating of cell-​type-specific Cry1, basal levels of cAMP rise, leading to increased
processes (Fig. 1b). This temporal gating includes the protein kinase A (PKA) activation. In turn, this induces
Period circadian protein phagocytic activity of macrophages30 and their cytokine phosphorylation of the p65 subunit of nuclear factor-​κB
homologue 1
(PER1). PER1, PER2 and PER3
release24,31, as well as histamine release and allergic reac- (NF-​κ B) at S276, leading to NF-​κ B activation.
are PAS (PER–ARNT–SIM) tions of mast cells32,33. Evidence is emerging to suggest Consequently, basal levels of several pro-​inflammatory
domain-​containing proteins that specific clock genes mediate different elements of cytokines (including IL-6, tumour necrosis factor (TNF)
that associate with CRY immune cell physiology. Logan and colleagues showed and CXC-​chemokine ligand 1 (CXCL1)) are increased
proteins to inhibit BMAL1–
that period circadian protein homologue 1 (PER1) is in fibroblasts and bone-​marrow-derived macrophages
CLOCK-​mediated gene
expression. necessary for efficient function of splenic NK cells34. from Cry1−/−Cry2−/− mice38. Thus, molecular effectors
In the absence of Per1, these cells maintain rhyth- of pro-​inflammatory pathways are directly coupled
REV-​ERB micity but show altered rhythmic expression of IFNγ to clock genes.
REV-​ERBα (encoded by and of the cytotoxic factors perforin and granzyme B.
NR1D1) and REV-​ERBβ
(encoded by NR1D2) are
Furthermore, rhythmicity in these factors and asso- Circadian control of innate immune cell movement.
transcriptional repressors that ciated cytotoxicity can be suppressed by chronic jet The circadian clock is a key regulator of the temporal
bind to ROR response element lag35. In macrophages, the REV-​ERB nuclear receptors abundance of innate immune cells around the body.
(RORE) motifs in the BMAL1 are a critical nodal point between the clock and immu- Studies by several laboratories have highlighted the
promoter to regulate the
nity. For instance, REV-​ERBα (encoded by NR1D1) importance of a functional clock in regulating the daily
rhythmic expression of
BMAL1. and REV-​ERBβ (encoded by NR1D2) suppress gene flux of inflammatory cells between bone marrow, blood
expression (for example, matrix metalloproteinase 9 and immune organs under homeostatic conditions. For
Cryptochromes (Mmp9) and CX3C-​chemokine receptor 1 (Cx3cr1)) instance, leukocyte numbers in blood show time-​of-day-​
(CRYs). CRY1 and CRY2 are in macrophages by repressing transcription from dis- dependent variation in humans39, mice40 and hamsters41.
transcriptional repressors that
associate with PER proteins to
tal enhancers that are selected by macrophage-​lineage This can be abolished by deletion of the core clock gene
inhibit BMAL1–CLOCK-​ determining factors36. Furthermore, REV-​ERBα modu­ brain and muscle ARNT-​like 1 (BMAL1; also known as
mediated gene transcription. lates the inflammatory functions of macrophages ARNTL)40 or by inducing chronic SCN arrhythmia41.

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Resident Mast cell DC CD4+ T cell Circulatory and/or migratory


Macrophage
CD8+ T cell

B cell

Microglia
PERs CRYs

Monocyte
PERs CRYs
PERs
Club cell
(bronchiolar CLOCK–
CRYs
exocrine cell) Neuronal
BMAL1 PAS2
NK cell
RORs
REV-ERBs RORs
Bmal1 REV-
ERBs

IEC
Neutrophil

Blood vessel Eosinophil


Fibroblast-like
synoviocyte
ILC

b
Cell group Cell type Driver Effect Refs
Innate Macrophage Lyz2–Cre 31

and/or (also known


monocyte as LysM–Cre) Increased monocyte recruitment and atherosclerosis lesion size; 79

polarization towards an M1-like phenotype (on ApoE–/– background)


Loss of diurnal variation in Ly6Chi monocyte numbers in spleen, blood 23

and bone marrow; reduced survival and higher inflammatory cytokine


production in Listeria monocytogenes infection; increased weight gain,
adiposity, insulin resistance and metabolic dysfunction on high-fat diet
78

LPS stimulation
Significantly enhanced serum IL-12–p40 under basal conditions, loss of 76
time-of-day susceptibility to EAE and exacerbated EAE pathology,
including increased CNS infiltration of inflammatory monocytes with
increased IL-1β production

Microglia Cd11b–Cre Reduced Il6 upregulation and attenuated neuronal 131

Adaptive T cell Cd4–Cre Differentiation and function unaffected, no impact on EAE severity and 55

reduced percentages of IFNγ+, IL-2+ and TNF+ T cells in L. monocytogenes


infection
Loss of oscillations in T cell numbers in lymph nodes and lymph; loss of 54

oscillations in homing capacity and Ccr7 and S1p1 expression


Lck–Cre Loss of time-of-day variation in susceptibility to EAE and reduced pathology 54

B cell Mb1–Cre Differentiation and function unaffected 55

Cd19–Cre Loss of oscillations in B cell numbers in lymph nodes and lymph 54

Stromal Club cell Ccsp–iCre Increased CXCL5 production and neutrophil influx 43

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Healthy Clock-disrupted
a
Day Night

REV–ERB Il6, ↑ β1 integrin


REV–ERB Il6, ↑ IL-6, IL-12–p40, Ccl2 ↑ Adhesion to
Ccl2 or RORα VCAM1
CCL5, CXCL1
and CCL2 RORα ↑ Chemotaxis
↑ IL-6 and CCL2
Overexpression

TLR9

b LPS LPS
Day Night Day Night
Corticosterone Bmal1-/- Corticosterone
GR Club cells GR

↑ CXCL5 ↓ CXCL5 ↑↑ CXCL5 ↑↑ CXCL5

Fig. 2 | Regulation of innate immunity in the macrophage and the lung. Clock regulation of innate immunity occurs in
an immune cell-​intrinsic and cell-​extrinsic manner. a | In macrophages, for example, the intrinsic clock regulates immune
function at multiple levels. Oscillations in the expression of the pattern-​recognition receptor Toll-​like receptor 9 (TLR9)
occur, along with temporal gating of cytokine release. In this way , the clock regulates not only the ability to sense
pathogens but also the extent of the cell’s response to such stimuli. Clock disruption in the macrophage yields a
pro-​inflammatory phenotype, as a subset of cytokines (for example, IL-6 and CC-​chemokine ligand 2 (CCL2)) loses the
rhythmic repression generated by REV-​ERB signalling. Overexpression of the transcriptional activator retinoid-​related
orphan receptor-​α (RORα), which binds to the same locus as REV-​ERB, generates a similar phenotype. b | Recruitment of
innate immune cells to tissues is also under clock control. In healthy murine lung exposed to lipopolysaccharide (LPS),
enhanced production of the neutrophil chemoattractant CXC-​chemokine ligand 5 (CXCL5) and increased neutrophil
recruitment are observed during the day. At night, endogenous glucocorticoids bind the glucocorticoid receptor (GR),
inhibiting Cxcl5 transcription and reducing neutrophil influx. Targeted ablation of Bmal1 in Club cells (Ccsp–Bmal1−/−)
has a clear pro-​inflammatory effect in this model. Lack of a functional oscillator in these cells greatly increased CXCL5
production and enhanced neutrophil recruitment after LPS at both time points. Although corticosterone still binds the
GR at night, the subsequent repression of Cxcl5 no longer occurs and neutrophilia increases. VCAM1, vascular cell
adhesion protein 1.

Leukocyte numbers also exhibit natural temporal var- human blood but appear to possess a weaker circadian
iation in the lung42. This persists under inflammatory oscillator — perhaps modified as the neutrophil develops
conditions but becomes less regular, with granulocytes from the myeloid progenitor28. Turnover of neutrophils
rather than lymphocytes dominating as the oscillating is known to be under circadian control, with rhythmic
cell type42. Gibbs and colleagues showed that neutro- clearance of aged neutrophils from the blood occurring
Brain and muscle
ARNT-like 1 phil recruitment to the lung after acute inflammatory in the bone marrow (where they are engulfed by bone-​
(BMAL1). A basic helix–loop– challenge is strongly circadian gated, driven by the marrow-resident macrophages)45. This clearance, which
helix PER–ARNT–SIM (bHLH– rhythmic release of the chemokine CXCL5 from non-​ occurs towards the end of the daily resting phase in mice,
PAS) domain transcription ciliated bronchiolar epithelial Club cells43 (Fig. 2b). Also, leads to a subsequent egress of haematopoietic precursor
factor that dimerizes with
CLOCK to bind E-​boxes in gene
neutrophil recruitment to sites infected with Leishmania cells from the bone marrow in a liver X receptor (LXR)-
promoters to induce circadian parasites has been shown to be circadian regulated44. dependent manner. Ella et al. described rhythmic daily
gene expression. Neutrophils are the most abundant leukocytes in changes in superoxide production by human neutrophils

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

and in their ability to engulf bacteria, and this was patho­gen colonization of the colon48. This leads to ele-
attributed to time-​of-day variations in the composition vated pathogen levels following infection in the morning
of the peripheral neutrophil pool, which in turn is a (Zeitgeber time 4 (ZT4) (Box 2)) as compared with infec-
result of circadian changes in circulating chemokine lev- tion in the mid-​dark phase (ZT16)48. This response is
els (specifically in levels of CXCL12 and/or CXCL2)28. dependent on the presence of a functional copy of the
Thus, both trafficking and localization of neutro- circadian-​regulating CLOCK (circadian locomoter
phils and their function are time-​of-day-​dependent. output cycles protein kaput) protein as global disrup-
Similarly, IL-5 and IL-13 cytokine production by a sub- tion of CLOCK alters time-​dependent responses to
set of group 2 innate lymphoid cells (ILC2s) in response S. Typhimurium. In a Leishmania infection model,
to hormonal cues induced by feeding influences the Kiessling and colleagues were able to show that para-
number of blood eosinophils and their recruitment to site burden was circadian in nature. In addition, bone
peripheral tissues46. marrow chimaeras of Rag2–Bmal1−/− mice demonstrated
Under homeostatic conditions, the extravasation that Bmal1 in non-​lymphocyte immune cells (macro­
of innate immune cells from postcapillary venules to phages) was responsible for modulating the magnitude
tissues is under clock control and is regulated by the of Leishmania infection44. Parasites themselves can
rhythmic expression of pro-​migratory factors (such also exhibit circadian behaviour. For example, 10% of
as intercellular adhesion molecule 1 (ICAM1) and the genes of Trypanosoma brucei (parasites responsi-
CCL2) by endothelial cells. This is dependent on local ble for inducing sleeping sickness) are expressed with
delivery of sympathetic tone and signalling through a circadian rhythm49. Furthermore, this circadian tran-
β-​adrenoreceptors40. Rhythmic extravasation persists scriptome affects sensitivity to treatment. The circadian
under inflammatory challenge, and in a model of septic clockwork also regulates cellular innate immunity against
shock in mice, neutrophil recruitment to the liver exhib- viruses. Immortalized lung fibroblasts lacking BMAL1
ited strong circadian oscillations. This influences mouse show altered susceptibility to viruses50. Respiratory syn-
survival following administration of lipopolysaccharide cytial virus (RSV) and parainfluenza virus type 3 (PIV3)
(LPS; a component of the cellular wall of Gram-​negative (which are both non-​segmented negative-​strand RNA
bacteria), with LPS delivery in the evening resulting in viruses) replicate at higher frequencies in the absence of
dramatically increased lethality compared with in the BMAL1 (ref.50). Bmal1−/− mice intranasally infected with
early morning40. RSV had higher viral titres and increased weight loss
The examples discussed above describe how innate than wild-​type controls50. Along the same lines, the time
immune cell trafficking can be regulated by both of day of infection affects the host response to herpes­
intrinsic pathways and circadian signals from other virus or influenza A virus, with enhanced viral titres seen
cells. The trafficking of Ly6Chi inflammatory mono- when levels of Bmal1 are low or in the absence of this
cytes from blood to bone marrow and from blood to gene51. Therefore, the circadian clockwork can regulate
tissue sites of infection is under circadian control23,47. cellular immunity against bacteria, parasites and viruses.
CXC-​chemokine receptor 4 (CXCR4) mediates the
rhythmic fluctuations in overall numbers of circu- Clock control of adaptive immunity
lating Ly6Chi monocytes and their homing to tissue Recently, molecular evidence has emerged to show that
reservoirs 47. During peritoneal infection with the the adaptive immune system is also under circadian
bacterial pathogen Listeria monocytogenes, traffick- control. This evidence is perhaps surprising given that
ing of Ly6Chi monocytes to the peritoneum is regu- adaptive immune responses are mounted over weeks
lated by the rhythmic recruitment of the polycomb and thus were not thought to be heavily dependent
repressor complex 2 (PRC2) to the Ccl2 promoter by on the initial time of day of challenge. However, this is
BMAL1–CLOCK heterodimers23. These studies show that in line with early reports that had initially uncovered
both cell-​autonomous as well as cell-​extrinsic micro­ T cell responses to be rhythmic52,53. In a similar manner
environmental oscillations can govern the distribution to innate immune cells, circadian oscillations have been
of distinct leukocyte populations within the body. described in cells of the adaptive immune system, such
CLOCK as T cells54–56 and B cells25. In addition, dendritic cells
(Circadian locomoter output Innate immune cell clocks in pathogen responses. At (DCs), which are professional antigen-​presenting
cycles kaput). A basic helix– the level of the whole animal, circadian timers clearly cells (APCs) located at the interface between innate
loop–helix PER–ARNT–SIM have an impact on how the host innate immune system and adaptive immunity, show oscillations in core clock
(bHLH–PAS) domain
transcription factor that can
responds to pathogens. In part, this occurs through components25. Generally speaking, clock gene ampli-
dimerize with BMAL1 to the regulation of inflammatory cell recruitment, where tudes appear to be weaker in immune cells than in
regulate circadian gene immune cell clocks are critical for maintaining effec- other cells, such as hepatocytes55. Whether this is due to
expression. tive host responses to disease23,43. Further control is a weaker oscillating system per se, a reduced coupling
exerted through the regulation of tissue-​resident cells capacity or increased heterogeneity in immune cell
Zeitgeber time
Zeitgeber, literally ‘time giver’, at sites of inflammation. Ingestion of the bacterial populations is currently unknown and would need to
is a cue (such as light) that pathogen Salmonella enterica subsp. enterica serovar be addressed using single-​cell analyses.
entrains the circadian clock. Typhimurium (S. Typhimurium) causes intestinal
Zeitgeber time (ZT) is the time inflammation. Microarray analysis of the caecum of Role of clock genes in lymphocyte development.
after light onset; for example,
lights on is ZT0 and lights
mice 72 h post-​infection with this pathogen revealed Circadian clock genes have been associated with the
off is ZT12 in a 12 h light-12 h that genes encoding antimicrobial peptides show development of lymphocytes, but the extent of their
dark cycle. strong circadian oscillations during infection that affect involvement is not yet fully established. Deficiency

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 2 | Circadian terminologies


be suppressed by the circadian-​regulated basic leucine
zipper transcription factor nuclear factor IL-3-regulated
a circadian rhythm is an oscillation with a period (time from peak to peak) of protein (NFIL3) in a cell-​intrinsic manner, and Nfil3−/−
approximately 24 h that is able to be entrained by external factors and persists in mice exhibit increased TH17 cell frequencies in spleen,
constant conditions (see the Figure). amplitudes (difference between peak and mean small intestine and colon58. NFIL3 is crucial for the
values) of these oscillations vary, as does acrophase (the timing of the peak value), such
development of ILCs59 and is a major regulator of TH17
that some parameters may peak during the day (for example, leukocyte numbers in
blood in mice) and some during the night (for example, leukocyte numbers in lymph
cell frequency via rhythmic suppression of Rorc, which
node in mice). experiments assessing rhythmicity are often performed under one (or encodes RORγt, a key transcription factor necessary for
both) of two conditions: light–dark or constant darkness. in the light–dark condition, TH17 cell differentiation58. Loss of ILC3s in the Nfil3−/−
abbreviated as LD, animals or participants are exposed to controlled cycles of light and mice would already skew towards a higher propensity
dark (usually 12 h light:12 h dark) to mimic a day–night cycle. in this situation, the timing for TH17 cell differentiation as ILC3s are a significant
nomenclature used is the ‘Zeitgeber time’ (Zt). a Zeitgeber (German for ‘time giver’) is a source of IL-22, which inhibits TH17 cell differentiation.
stimulus that provides a timing signal, such as light onset, and Zt0 is simply the timing Depletion of ILCs thus enhances TH17 cell responses60.
of this stimulus. all other times are measured in a 24 h cycle relative to this stimulus. For Within CD4+ T cells themselves, Nfil3 and Rorγt are
example, in a cycle with 12 h light:12 h dark, Zt6 is the middle of the light phase, Zt12 is rhythmically expressed in an anti-​phase manner, with
then the onset of the dark phase, Zt18 is the middle of the dark phase and Zt24/0 is the
Nfil3 expression peaking during the dark phase (night)
onset of the light phase once more. an oscillation under these conditions would be
termed diurnal. in constant darkness, however, no such light stimulus exists. in this case,
and Rorγt expression peaking during the light phase
times are noted as ‘circadian time’ (Ct) and better reflect the animal and/or (day). T cells isolated during the day are therefore more
participant’s subjective time in the absence of external cues. thus, in dark–dark (DD) prone to differentiate into TH17 cells upon stimulation
experiments, Ct0 is the start of the subjective day (that is, when the lights would have than those isolated at night58. This propensity may addi-
come on), Ct6 the middle of the subjective day, Ct12 the start of the subjective night tionally be due to the fact that during the day levels of the
(that is, when the lights would have gone off), Ct18 the middle of subjective night and night-​signalling hormone melatonin are low, because
Ct24/0 the start of the next subjective cycle. an oscillation under these conditions melatonin treatment of human T cells has been shown
would be termed circadian. to inhibit TH17 cell differentiation via the NFIL3–RORγt
Period pathway61. Instead, melatonin promotes the develop-
ment of IL-10-producing type 1 regulatory (TR1) cells61.
Amplitude
The circadian clock protein REV-​ERBα directly inhibits
transcription of Nfil3, and Nfil3 expression is ele-
Mean value vated in both REV-​E RBα −/− T  cells and in T  cells
Acrophase
from ClockΔ19/Δ19 mice, which exhibit a lengthened
circadian period and arrhythmicity in complete dark-
ness 58. ClockΔ19/Δ19 mice also displayed reduced
frequencies of TH1 cells in the intestines, implying
ZT LD additional pathways by which the circadian clock can
CT DD affect T cell polarization. The circadian regulation of
T cell development may be particularly important for
0 12 24/0 12 24/0
maintenance of an appropriate balance of cell types
by time of day, as chronic circadian disruption via
of BMAL1 negatively affects mouse B cell develop- simulated jet lag resulted in increased proportions
ment, with Bmal1−/− animals showing significantly of TH17 cells, rendering animals more susceptible to
reduced B cell numbers in blood and spleen57. Bmal1- experimental colitis58.
deficient B cells also showed a specific deficit in pro- In addition, the circadian-​regulated proteins differ-
duction of IgG1 (but not in IgG2a, IgG2b, IgG3 or IgM) entially expressed in chondrocytes protein 1 (DEC1;
antibodies after immunization, but they exhibited no also known as BHLHE40) and DEC2 (also known as
functional difference in response to LPS in vitro, suggest- BHLHE41) are driven by E-​box mediated BMAL1–
ing that those cells that do develop are still largely func- CLOCK binding. DEC1 is involved in essential CD4+
tional. Adoptive transfer experiments revealed that it is T cell effector functions, including in the regulation
not the B cell intrinsic clock but rather loss of Bmal1 in the of a large group of CD28-dependent genes62. Antigen-​
bone marrow microenvironment that hampers B cell dif- specific DEC1-deficient CD4+ T cells have cell-​intrinsic
ferentiation57. This finding is also consistent with the data defects in survival and proliferation62. Meanwhile, DEC2
of Hemmers and Rudensky, who showed that mice with is critical for the development of B-1a cells, and DEC2-
a B cell-​specific Bmal1 deficiency do not show altered deficient B-1a cells show an abnormal B cell receptor
B cell differentiation or function55. These experiments (BCR) repertoire63. Taken together, the proteins of
suggest that the influence of clock genes in the genera- the circadian clock clearly play an important role in the
tion of B cells is not cell-​autonomous but dependent on function of lymphocytes.
Circadian time
(CT). A measure of subjective the microenvironment.
time used when organisms are With respect to the influence of the circadian clock Lymphocyte trafficking rhythms. Similarly to innate
isolated from Zeitgebers (for on the development of T cells, the overall T cell number immune cells, T cells and B cells exhibit strong cir-
example, constant darkness). appears to be unaffected by global or cell-​specific Bmal1 cadian oscillations in the blood, with peak numbers
CT0 represents the start of
subjective day and CT12
ablation55,57. However, Yu and colleagues reported a role during the respective behavioural rest phase of the
represents the start of for the clock in regulating the differentiation of T helper 17 organism (that is, during the day in mice and at night
subjective night. (TH17) cells58. TH17 cell development has been shown to in humans)39,54,64–66. These rhythms have been linked to

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Day Night

Blood Blood
↓ CCR7 ↑ CCR7
(leukocytes) (leukocytes)

↑ NA

↓ Sympathetic tone ↑ Sympathetic tone


↓ Leukocyte content ↑ Leukocyte content

↑ S1p1 ↓ S1p1
(leukocytes) HEV (leukocytes)

Efferent Efferent
lymph lymph

B cell CD4+ T cell CD8+ T cell

Fig. 3 | Regulation of adaptive immunity in the lymph node. A coordinated oscillation in pro-​migratory factors drives
leukocyte oscillations in blood, lymph node and lymph.  During the active phase, sympathetic tone (noradrenaline (NA))
and concentrations of CC-​chemokine ligand 21 (CCL21) are high in the lymph node. In addition, expression of the
receptor for CCL21, CC-​chemokine receptor 7 (CCR7), is at its peak on T and B cells. The combination of high receptor
expression on the cells and high chemokine content in the lymph node provides a strong attraction signal, leading to
increased lymph node cellularity during this phase. During the rest phase, CCL21 and CCR7 expression decrease and
retention signals are reduced. At the same time, leukocytes begin to upregulate expression of the sphingosine
1-phosphate receptor 1 (S1P1), a critical mediator of cell egress. Thus, the reduction in sympathetic tone and homing
drive coupled with an increased egress potential lead to an emptying of the lymph node during the day. HEV, high
endothelial venule.

oscillations in CXCR4 and CX3CR1 expression, regulated the time-​dependent difference in lymph node homing
by glucocorticoids and catecholamines64,65, as well as and cellularity54.
hypoxia-​inducible factor 1α (HIF1α) signalling67. Recent Interestingly, the egress of cells into efferent lym-
papers found that under steady-​state conditions, lympho­ phatics was also found to be rhythmic, with lymphocyte
cytes within the lymph node also exhibit circadian counts highest in lymph around ZT9 (refs54,66). This
oscillations in mice, with the highest numbers present finding was dependent on rhythmic expression of
at the beginning of the active phase (that is, at night in sphingosine 1-phosphate receptor 1 (S1P1)54, which
mice)54,66,68 (Fig. 3). These higher numbers were not due promotes lymphocyte egress from lymph nodes
to local intranodal proliferation but depended on both in response to the chemotactic lipid sphingosine
the rhythmic homing of cells from blood into lymph 1-phosphate 71. Cells that migrated into the lymph
nodes and the rhythmic egress of cells from lymph nodes node at night also stayed longer within this tissue than
into efferent lymph. A key regulatory element is the the (fewer) cells that had immigrated during the day.
CC-​chemokine receptor 7 (CCR7), which exhibited diur- Suzuki and colleagues demonstrated that this is depend-
nal oscillations in T cells and B cells, with its expression ent on lymphocyte expression of β2-adrenergic recep-
peaking in phase with CCL21 levels on high endothelial tors72 and rhythmic environmental sympathetic tone66.
venules in lymph nodes54. This receptor–ligand pair is β2-Adrenergic-​receptor-deficient lymphocytes transited
critical for control of T cell migration into lymph nodes, through lymph nodes more quickly and failed to show a
whereas B cells also rely on additional receptors such as diurnal pattern66. This finding indicates that enhanced
CXCR4 and CXCR5 (refs69,70). Peak expression levels sympathetic tone and engagement of the β2-adren-
of both CCR7 and CCL21 were observed around night ergic receptor at night helps maintain cells in lymph
onset, when the highest recruitment of CD4+ T cells, node compartments, likely by boosting lymphocyte
CD8+ T cells and B cells occurred54. Both the microen- expression of the retention factors CCR7 and CXCR4
vironment and lymphocytes themselves contribute to (ref. 72) . This longer lymph node dwell time plays an
this process, but T cell-​specific genetic deletion of the important role in the generation of adaptive immune
circadian clock regulator BMAL1 was sufficient to ablate responses54,66. Thus, time of day both dictates expression

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

of critical pro-​migratory molecules on lymphocytes and Recent vaccination studies also showed diur-
endothelial cells and governs the trafficking behaviour nal rhythmicity in adaptive immune responses.
of lymphocytes from blood through lymph nodes and Responses to nitrophenyl (NP), a soluble antigen that
into efferent lymphatics. is directly transported to lymph nodes, or to myelin-​
oligodendrocyte glycoprotein (MOG), which is an
Rhythmic adaptive immune responses. Initial obser- antigenic target in certain models of experimental
vations by Esquifino and colleagues showed that the autoimmune encephalomyelitis (EAE), were higher
adaptive immune system exhibits functional rhythmi­ at the times when more cells were present in lymph
city. They demonstrated that proliferative responses of nodes 54,66,76. Stimulation at high cellularity times
rat lymph node cells following treatment with LPS or resulted in a stronger humoral response, as demon-
concanavalin A were strongly rhythmic, with signifi- strated by higher NP-​specific antibody titres, as well as
cantly stronger proliferation occurring at midday than at more NP-​specific IgM+ germinal centre B cells 7 days
midnight73. Subsequent studies by Fortier and colleagues after immunization66. In addition, in the EAE model a
confirmed these observations and further demonstrated stronger cellular immune response was observed with
that this was dependent on the circadian clock. Mice more IL-17-producing TH17 cells being present, more
bearing the circadian mutation ClockΔ19/Δ19 exhib- IL-2 being made, higher leukocyte infiltration into the
ited strongly blunted rhythms of proliferative responses central nervous system and ultimately a higher dis-
of T cells after direct stimulation of the T cell receptor ease score 14 days after inoculation54. Importantly, the
(TCR)74. Furthermore, they found that intravenous humoral immune response to NP became arrhythmic in
injections of ovalbumin (OVA)-loaded DCs into recip- mice lacking the β2-adrenergic receptor66, and animals
ient mice at ZT6 (daytime), as opposed to at ZT18 with a myeloid-​cell-specific or T cell-​specific deficiency
(night-​time), generated higher numbers of OVA-​specific in BMAL1 became arrhythmic in the EAE model of
T cells within the spleen. These data provide evidence autoimmunity54,76. Rhythmicity was not restricted solely
for the importance of circadian timing in the interaction to sterile immunization responses but was also observed
between T cells and APCs. following infection with pathogens, such as influenza A
For an adaptive immune response to be generated, virus and the bacterium Helicobacter pylori54.
APCs must functionally interact with lymphocytes. Current research efforts now aim to take these find-
Silver and colleagues were the first to investigate a ings into the clinic and establish novel ways to enhance
rhythmic link between the adaptive and innate immune responses to vaccines. A randomized trial of different
system. They demonstrated that the expression of Toll-​ times of day of vaccination of three strains of influenza
like receptor 9 (TLR9) — which recognizes bacterial virus yielded promising results. When 276 patients (over
CpG DNA — but not of other TLRs by macrophages, 65 years of age) were vaccinated in the morning they had
DCs and B cells, exhibited oscillations that were con- greater antibody titres 1 month later than patients vacci-
trolled by the circadian clock gene Per2 (ref.75). In line nated in the afternoon77. These results are encouraging,
with this, mice treated with OVA coupled to CpG at but they will need to be validated in other preclinical
the time of enhanced TLR9 expression (ZT19 versus laboratory models and clinical trials using larger patient
ZT7) showed elevated OVA-​specific adaptive immune cohorts. Taken together, this points to a strong role of
responses several weeks later75. Lymph node cultures the circadian clock in the polarization of the adaptive
from these mice showed higher levels of OVA-​induced immune response, which is strongly dependent on the
lymphocyte proliferation and production of IFNγ at initial time of day of antigen challenge.
ZT19. In addition, lymphocytes from mice lacking a
functional PER2 protein showed a stronger immune Innate versus adaptive immune clocks. Whereas
response than wild-​type cells75. myeloid-​specific ablation of the circadian gene Bmal1
These data demonstrate that circadian differences leads to a general pro-​inflammatory phenotype in mice
in the expression of pattern-​recognition receptors that is characterized by higher levels of TNF, IL-6, IL-1β
(PRRs) by APCs can drive rhythmic adaptive immune and CCL2 (refs23,31,78,79), the phenotype of T cell-​specific
responses and are thus under the control of the cir- Bmal1 deficiency appears to be more time-​of-day-​
cadian clock. However, it is currently unclear how dependent54,55. Whether this is because the currently
the observed differences in the time of day of the employed genetic targeting strategies for lymphocytes
acrophase of the response arise. Whereas Silver and (CD4–Cre for T cells54,55) are more specific than for mye-
Suzuki described stronger immune responses at night loid cells (that is, where Lyz2–Cre is currently used, thus
(ZT17 and ZT19, respectively)66,75, Fortier and Druzd targeting most myeloid cell populations23) is unclear. To
observed peak responses during the day (ZT6 and ZT8, better compare this, future studies will need to use pro-
respectively)54,74. An explanation for these discrepan- moters that are specific for neutrophils, monocytes or
cies might lie in the type of response elicited: injec- macrophages for Bmal1 ablation. Infection of mice with
tion sites (subcutaneous, intraperitoneal, intravenous L. monocytogenes resulted in a lower percentage of IL-2+,
or intradermal), readout tissue (lymph node versus IFNγ+ and TNF+ T cells in mice with a T cell-​specific
spleen), lighting conditions, responding cell type and deficiency in Bmal1 than in control animals55. In addi-
antigen choice all contribute to such variations, par- tion, in models of EAE, mice with a T cell-​specific Bmal1
ticularly whether the antigen needs to be taken up by deficiency had lower disease scores than controls54
APCs to be transported to lymph nodes or can move whereas mice with myeloid cell-​specific loss of Bmal1
there directly in a soluble form. showed exacerbated pathology76. This finding indicates

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

that the outcome of Bmal1 deletion on inflammation is Many bacterial genera within the gut microflora of
lineage dependent. mice and humans exhibit circadian oscillations them-
selves. For example, Lactobacillaceae show strong oscil-
Microbial regulation of circadian immunity lations in general abundance over the course of the day,
Recent evidence demonstrates that symbiosis between peaking at the onset of the mouse active phase85,86. This
microbiota and intestinal epithelial cells is critical in rhythmicity is driven by feeding rhythms and can be
maintaining immune homeostasis in the gut. Intriguingly, ablated by jet-​lag protocols (repeated shifts in timing of
this interaction is orchestrated by the circadian clock light–dark cycles) or antibiotic treatment85,86. Chronic
and cues from the microbiota that activate intestinal jet lag in mice leads to obesity, whereas in humans jet-​
TLRs. Mukherji and colleagues showed that the anti-​ lag protocols result in a type 2 diabetic phenotype86.
phasic binding of RORα and REV-​ERBα to TLR genes This finding indicated that aberrant circadian rhythms
acts as an activator and repressor, respectively, to medi- in humans can lead to metabolic disturbances. Gut
ate the circadian expression of TLR1–TLR5 and TLR9 bacteria exhibit circadian rhythmicity in terms of their
(but not TLR6 and TLR7)80. This oscillatory promoter numbers and activity, and their rhythmic function is
occupancy drives rhythmic activation of the key pro-​ also important for the systemic regulation of host tis-
inflammatory transcription factors activator protein 1 sues8. This is achieved by a specific rhythmic signature
(AP-1) and NF-​κB to exert their functions at diurnal of metabolites present in blood and is important in
times ZT20–ZT4 (which is the active phase in mice). driving epigenetic changes in host tissues distant from
Antibiotic-​mediated ablation of the microbiota disrupts the gut, such as the liver8. Thus, rhythmic release of
this rhythmicity and induces a pre-​diabetic syndrome in metabolites and LPS from the microbiota may be key
mice, characterized by hyperglycaemia and hypoinsuli- in directly driving important rhythmic immune-​related
naemia80. Intriguingly, rhythmicity could be restored by phenotypes, such as neutrophil ageing and associated
administration of LPS. Additional recent findings pro- activation45,82. In addition, rhythms in commensal bac-
vide a link between rhythmic Nfil3 expression in intesti- teria appear to help synchronize the body to the external
nal epithelial cells, microbiota and host metabolism that environment and thus indirectly influence a rhythmic
is mediated by immune cells81. The induction of TLR– immune system.
MYD88 signalling by bacteria stimulates DCs to secrete
IL-23 and activate ILC3s. This activates signal transducer Synchronization of the immune system. Microbial
and activator of transcription 3 (STAT3), leading to an products and feeding rhythms can synchronize periph-
inhibition of REV-​ERBα, a negative regulator of NFIL3. eral clocks in tissues87 but it is yet unclear how immune
Epithelial NFIL3 regulates lipid uptake and export in cells — most of which are migratory — are synchro-
intestinal epithelial cells so that mice with an intestinal-​ nized in  vivo to the external environment. Initial
specific deletion of Nfil3 are resistant to obesity induced studies focusing on circulating hormones were able
by a high-​fat diet81. These data indicate that the micro­ to demonstrate a correlative link between glucocorti-
biota is a previously unrecognized and important general coids and adrenaline and numbers of circulating T cells
regulator of immune rhythmicity in the host (Fig. 4). in blood by modulating surface CXCR4 and CX3CR1
levels65. It has now been shown using cell-​specific dis-
Microbial products shape circadian immunity. LPS ruption of glucocorticoid signalling that rhythmic T cell
also appears to be a critical factor in shaping the acti- trafficking is regulated via glucocorticoid receptor
vation status of neutrophils that are present in blood. (GR)-dependent induction of the IL-7 receptor (IL-7R)
Although blood neutrophils have long been thought to and CXCR4 (ref.68). Dexamethasone88 and high tran-
be a relatively homogeneous population, heterogeneity sient serum concentrations (a so-​called serum shock)87
arises owing to differences in their age and activation are used to synchronize cells in culture, indicating that
level. Neutrophils are short-​lived and — after having both glucocorticoids and feeding rhythms are impor-
been mobilized into the blood in the morning45 — age tant for the synchronization of migratory cells in vivo.
in blood over the course of the day45,82. Aged neutro- With respect to the sympathetic nervous system, lack
phils exhibit higher levels of CXCR4 on their surface of β2-adrenergic or β3-adrenergic receptors resulted
and lower levels of L-​selectin45,83,84 and are present at in an ablation of the rhythmic trafficking behaviour
higher concentrations during the day in mice45. This is of leuko­c ytes to tissues40,66. However, it is currently
associated with an enhanced activation status83, includ- unclear whether the clockwork in immune cells and/or
ing an increased capacity to form neutrophil extra- stromal cells was also affected in this case. Furthermore,
cellular traps82. Microbiota-​derived LPS drives this a recent surprising study has found that the cell-​intrinsic
ageing process82. Mechanistically, this is modulated by clock is sufficient for the rhythmic migration behaviour
TLR2- and TLR4-expressing haematopoietic cells, as of leuko­cytes67. Zhao and colleagues showed that in
well as by MYD88-expressing myeloid cells. Ablating humanized mice, human and mouse leukocyte num-
the microbiota dramatically reduced the number of bers within the same organism exhibited inverted oscil-
RORα aged neutrophils and was also effective in alleviating lations in blood, reproducing the trafficking pattern
The nuclear receptors RORα, the pathogenesis of sickle cell disease and endotoxin-​ previously observed in both species. This cyclic pattern
RORβ and RORγ are induced septic shock82. Thus, rhythmically recurring of trafficking correlated with rhythmic CXCR4 expres-
transcriptional activators that
bind to ROR response element
cycles of neutrophil activation in the circulation can have sion, which was regulated by a HIF1α–BMAL1 hetero­
(RORE) sites in target gene dramatic consequences on the time-​of-day-​dependent dimer. p38 mitogen-​activated protein kinases (MAPKs)
promoters. worsening of inflammatory disorders. and MAPK kinase activated protein kinase 2 (MK2)

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Outer layers Epithelium

Day Muscularis Night


mucosae
DC
Lamina propria
IL-23
MYD88
ILC3

IL-22 IEC
REV–ERBα
Lumen CD4+
T cell
CD4 T cell
+
Neutrophil Microbiome
REV–ERBα NFIL3
Blood MYD88 Bacteria
vessel
Metabolites
↑ CXCR4 ↓ CXCR4

NFIL3
RORγ t

RORγ t

↓ TH17 cell
↑ TH17 cell
in vitro in vitro

Fig. 4 | Interactions of the clock , microbiome and immune function. Circadian oscillations occur not only in host cells
but also in the composition of microbiota.  The relative abundance of microorganisms fluctuates across a day , as does the
quantity and variety of metabolites released from the gut into the bloodstream, which can, in turn, signal to peripheral
tissues or circulating cells (for example, via Toll-​like receptors (TLRs)). Expression of a subset of TLRs is rhythmic in intestinal
epithelial cells (IECs) and drives increased pro-​inflammatory responses via activator protein 1 (AP-1) and nuclear factor-​κB
(NF-​κB) at the light onset (Zeitgeber times ZT20–ZT4). Activation of the MYD88 pathway , a downstream cascade common
to many TLRs, was recently shown to regulate neutrophil ageing. This process occurs more prominently during the rest
phase, with aged neutrophils increasing CXC-​chemokine receptor 4 (CXCR4) expression and preferentially homing to
bone marrow. Homing of such senescent neutrophils triggers mobilization of haematopoietic progenitor cells, thereby
driving rhythmic modulation of this niche. Cell differentiation may also be modulated rhythmically , as in the case of T
helper 17 (TH17) cells and their key differentiation factor retinoid-​related orphan receptor γt (RORγt). In cells isolated
during the night, RORγt is suppressed by nuclear factor IL-3-regulated protein (NFIL3) and polarization towards TH17 cell
differentiation is reduced. However, in cells isolated during the day , the clock protein REV-​ERBα inhibits this suppression,
allowing RORγt levels to increase along with increased TH17 cell differentiation. In this way , T cell populations cross this
developmental checkpoint in a coordinated manner. Furthermore, MYD88-dependent activation of dendritic cells (DCs)
can trigger IL-22 release by type 3 innate lymphoid cells (ILC3s), which in turn promotes signal transducer and activator of
transcription 3 (STAT3)-dependent inhibition of REV-​ERBα expression in the IECs. Therefore, microbiota components may
further fine-​tune diurnal rhythms in the gut.

exhibited opposite effects between mice and humans in when administered at a more physiological dose that
generating intracellular reactive oxygen species, which is more likely to be encountered in nature23. Thus, this
subsequently regulated HIF1α expression67. This indi- heightened immune sensitivity would be an adapta-
cates that immune cells use reactive oxygen species as tion to potentially recurring environmental challenges,
their synchronization cue. Taken together, a complex with the additional effect of reducing detrimental
network of factors appears to be important for immune immune responses (such as a high activation status of
cells to be synchronized to external time. neutrophils) at times when they are not needed. Thus,
a timed immune response — with a refractory phase
Potential benefits of rhythmic immune responses in between, possibly for opposing immune processes
A key question in our field is why is the immune sys- to occur — may ultimately increase the survival of an
tem so tightly controlled by circadian oscillations? organism. Evidence for this lies in the fact that both
The most striking examples of timing in the immune immune cell-​i ntrinsic and cell-​e xtrinsic (such as
response are mice that succumb to high doses of pro-​ endothelial and stromal cell) rhythms are coordinated
inflammatory mediators or pathogen more promi- in a highly complex fashion to balance the recruitment
nently in the afternoon18,89. However, at the same time, and activation of immune cells. An alternative expla-
mice are able to fight off bacteria more effectively nation could be that rhythmic immune responses may

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

have evolved as a secondary consequence of rhythmic with an increased risk in developing inflammatory dis-
metabolic processing within the immune cell itself. eases such as psoriasis95 and irritable bowel syndrome96.
For the latter, the key benefit of circadian rhythmicity In keeping with this, Cuesta et al. report desynchrony in
would be the ability of cells to partition metabolic flux rhythmic immune parameters after simulated night-​shift
and detrimental redox processes in time and space9. work97. Of course, rotating shift work affects not just
Clearly, metabolic and immune processes are intricately biological rhythms but also other lifestyle habits (sleep
linked, and both affect each other also with respect to quality and quantity, diet, mealtimes and exercise) that
circadian timing. For example, metabolic cues are crit- may potentially affect the development of such disor-
ical for the entrainment of peripheral clocks, such as ders. Nevertheless, this is a major concern for society.
those present in immune cells. In addition, disruption Together, animal studies and clinical data corroborate
of clock gene function results in a host of metabolic the notion that a healthy intact clock is necessary to
disorders, which are often associated with immune regulate immune homeostasis and to help prevent the
phenotypes90, such as mice with a myeloid cell-​specific development of immune disorders. In addition to this
deletion of Bmal1 that present with a pro-​inflammatory role under homeostatic conditions, the clock continues
phenotype and over time become obese23. Thus, par- to be critical in regulating inflammation once disease
titioning of immune cell function to specific times of is established. It is well recognized that human chronic
the day likely demonstrates an adaptation of the organ- inflammatory diseases can show time-​of-day variation in
ism to pathogen exposure in addition to detrimental not only symptoms but also disease markers. Most often
endogenous processes. cited is the nocturnal component to asthma, whereby
symptoms show exacerbations in the early morning98.
Therapeutic implications Similarly, patients suffering from rheumatoid arthritis
Numerous animal models show that genetic or envi- often report increased joint stiffness and pain in the early
ronmental disruption of the clock can expedite the morning99. These periods of increased symptoms corre-
development of inflammatory disease or exacerbate late with peaks in disease markers, indicating that these
disease pathology91–94. This translates to humans, as phenomena are not simply a consequence of sleep itself
employment in rotating shift work has been associated (for example, caused by limited movement and altered
breathing patterns). There is an early morning peak in
the number of pulmonary eosinophils in patients with
Box 3 | Circadian clock links to cancer asthma100 and in circulating cytokine levels in patients
with rheumatoid arthritis 101. The onset of cardio­
the circadian clock regulates daily rhythms in cell division, metabolism, inflammation vascular events often shows time-​of-day variation, with
and DNa damage response, and disruption of the circadian clock is known to increase
acute myocardial infarction and ventricular arrhythmias
the incidence of cancer122,123. specifically, the core clock machinery is tightly coupled to
metabolic cycles and is able to adapt to shifting metabolic rhythms, such as those
more common in the morning102. This timing is likely a
imposed by enforced feeding schedules, leading to reprogramming of circadian consequence of circadian variation in blood pressure, the
transcriptional circuits and altered cyclic metabolism124–126. Both central and peripheral coagulation cascade and vascular function. Furthermore,
circadian homeostasis are known to be disrupted in a number of pathological states, there is evidence that in the days following the transition
including inflammation and cancer. Further insight into this process has been shown in to or from daylight savings time, there is an increase in
a recent study of lung adenocarcinoma in mice, which explored the impact of tissue the incidence of acute coronary syndrome related to the
malignancy on the circadian organization of body organs127. Lung tumours driven by desynchrony between the internal clock and the external
conditional activation of KrasG12D mutation and loss of p53 tumour suppressor were environment103. Given the wide range of effects of the
associated with widespread disruption of hepatic glucose production and altered circadian clock on immune function, it is not surprising
insulin signalling and lipid metabolism, mediated by alterations in aKt, aMPK and
that in a chronic setting, there are rhythmic fluctuations
sreBP pathways. surprisingly, these distal effects were not mediated by hepatic
malignancy, disruption of the core circadian clockwork or indeed behavioural
in inflammatory pathways, sufficient to cause physio­
rhythmicity. rather, the authors proposed that circadian remodelling of the hepatic logical effects. Our current understanding of these
transcriptome was mediated by the release of immune signals, such as tumour-​derived interac­tions is very limited, but it is clearly important
cytokines (for example, iL-6) from malignant lung tissue, which then acted as distal that we gain deeper knowledge as this is likely to lead to
reorganizers of circadian hepatic metabolism. novel treatment options.
the link between the circadian clock, inflammation and metabolism raises the
intriguing possibility as to whether pharmacological targeting of specific elements of Seasonal diseases and underlying mechanisms.
the core circadian clockwork may regulate metabolic and inflammatory pathways and There is emerging evidence that the immune system
indirectly provides effective therapeutic options for the treatment of malignancy and also shows seasonal fluctuations in man and animals;
cancer. a recent study has shown that pharmacological activation of rev-​erB using
indeed, this may explain why some inflammatory dis-
small-​molecule agonists is lethal in cancer and oncogene-​induced senescence128. these
circadian-​regulated rev-​erB proteins are constitutive repressors and nuclear hormone
eases have an additional seasonal component. Within
receptors and act as important outputs coupling the core clock to physiology, playing a circulating immune cells, approximately 23% of the
central role in the regulation of rhythmic hepatic metabolism125 and inflammation31. genome shows seasonal variation in expression 104;
Sulli et al. showed that REV-​ERB agonists evoked an apoptotic response, confined to furthermore, the cellular makeup of the peripheral
malignant cells, and critically also revealed selective targeting of slowly proliferating immune system varies according to season104. It is not
tumorigenic populations. importantly, this study now links for the first time key yet established how annual changes in the external envi-
hallmarks of cancer — de novo lipogenesis and apoptosis, important in meeting ronment modulate seasonal changes or to what extent
metabolic demands of cancer cells — to a specific circadian clock circuit. this suggests seasonal changes affect immune function over the year.
a hitherto unsuspected link between elements of the core clock, cellular metabolism However, this new information may explain in part why
and malignancy.
some immune disorders exhibit seasonal variation in

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

the occurrence of exacerbations. Dopico and colleagues activity in patients with multiple sclerosis peaks in
noted a seasonal fluctuation in Bmal1 expression, with early spring and troughs in autumn106. This trend may
lower levels of expression during the winter months104. be attributed in part to seasonal variation in the noc-
Herpesviruses and influenza A viruses replicate more turnal pattern of secretion of pineal melatonin, with
efficiently in the absence of BMAL1, perhaps in part longer nocturnal durations in the autumn and winter61.
explaining why viral dissemination is more common These examples demonstrate that there may be impor-
in winter51. Individuals diagnosed with the debilitating tant seasonal changes in the phasing and exposure of
lung condition chronic obstructive pulmonary disease circadian-​regulated signals, tuned to the environment,
(COPD) are also more prone to exacerbation during the that can affect our physiology and health.
winter months105. This is thought to be the result of a
combination of increased prevalence of respiratory viral Chrono-​immunotherapy. Our knowledge regard-
infections, lifestyle changes during the colder months ing circadian variation in the expression and activ-
and reduced vitamin D levels. Conversely, disease ity of drug targets is steadily growing. A recent study
has shown that a high proportion of the best-​selling
drugs used in the treatment of inflammatory diseases
(as well as drugs for metabolic diseases, mental health
Targeting Timed disorders and cancer (Box 3)) directly target the prod-
the clock delivery ucts of genes that are themselves under clock control107.
Neurodegeneration Many of these drugs typically have short half-​lives
(multiple sclerosis) (less than 6 h); consequently, timed application in line
Melatonin decreases Vaccination
severity and/or incidence of Increased influenza antibody with target expression may markedly improve treatment
experimental autoimmune titres in morning versus efficacy (Fig. 5). Surprisingly, as yet there is very little use
encephalomyelitis132 evening in vaccinated elderly
population (≥ 65 years)77 of chronotherapy for the treatment of inflammatory
disorders in a clinical setting. One success involves the
Asthma use of glucocorticoids in the treatment of rheumatoid
Timed dosing increases arthritis. A slow-​release formula of prednisone has been
effectiveness of multiple manufactured, designed to be taken at bedtime, result-
treatments134
ing in release into the bloodstream 4 h later, and phased
Atherosclerosis to the peak in expression of inflammatory markers,
Anti-inflammatory effects
Chemotherapy of REV–ERB agonists112 which reduced the duration of morning stiffness of the
Timed treatment increases
therapeutic index133 joints108. We now know that the circadian clock (through
the actions of the CRY and REV-​ERB proteins) regu-
lates the response of the GR to its ligands109,110. Thus,
timed application of anti-​inflammatory glucocorticoids
may enable maximal therapeutic benefit while at the
same time reducing undesirable hepatic metabolic side
effects of these heavily used drugs109.
Bone marrow transplant
Arthritis
Slow-release prednisolone The clock as a therapeutic target for inflammation.
allows peak drug availability mouse model40 Our growing understanding of how components of the
at time of greatest need108
molecular clock interact with critical elements of inflam-
Cryptochrome-stabilizing matory pathways offers the exciting potential for the use
drugs are anti-inflammatory of pharmacological agents that target these clock pro-
(in vitro)116 teins as anti-​inflammatory agents. The challenge now
is to produce high-​affinity, high-​efficacy molecules that
enhance the activity of clock proteins. Recently, a num-
ber of tool compounds have been developed that show,
in principle, the benefits of pharmacological modula-
tion of clock proteins on the outcome of inflammatory
challenge. SR9009 is a synthetic REV-​ERB agonist111 that
Fig. 5 | Chronotherapy and the clock as a therapeutic target. The knowledge of has been shown to decrease atherosclerotic plaque bur-
circadian regulation of various aspects of immunity can inform treatment in two ways: den after chronic administration in mice112. Conversely,
modifying treatment regimens to the time of greatest efficacy (green boxes) and application of a synthetic REV-​E RB antagonist
targeting the clock itself (blue boxes).  Timed delivery of compounds has already been (SR82778)113 has detrimental effects in a mouse model of
shown to increase effectiveness of multiple drugs already in use, with the benefit of viral-​induced encephalitis114. In addition, the compound
increasing the therapeutic index77,108,132–134. Timing of surgical procedures such as bone KL001 acts to stabilize CRY proteins by preventing
marrow transplantation is also a key factor modulating success and survival and should
ubiquitin-​driven degradation115. Application of KL001
be carefully considered in clinical applications40. On the other hand, targeting
components of the molecular clock may yield novel treatment options in some diseases. in vitro has anti-​inflammatory effects on fibroblast-​like
Increasing the stability of the clock protein cryptochrome has shown anti-​inflammatory synoviocytes116, and this presents as another potential
effects in vitro116, and REV-​ERB agonists have also proved anti-​inflammatory in vivo112. anti-​inflammatory target. Taken together, the future will
However, given the broad influence of the clock upon physiology and pathology , care see the development of novel tools targeting the clock in
must be taken when developing compounds that manipulate this network. inflammatory disorders.

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Conclusions in chronic responses manifested several days or even


The immune system is heavily influenced by time-​of-day weeks later. Despite the rapid recent progress made in
cues, both under steady-​state conditions and in response understanding the mechanisms involved, the role of the
to inflammatory challenges. The regulation of immune circadian-​controlled expression signatures of specific
responses according to the time of the day is likely opti- organs and crosstalk to other body systems remains
mized to provide protection at specific times. In addi- very poorly understood. An improved understand-
tion, time-​partitioning of metabolic and redox processes ing of these pathways is necessary in order to advance
in immune cells may help minimize detrimental effects chronotherapeutic options. From this, novel drugs and
on the organism. Shifts of a few hours in the timing of treatments that could maximize therapeutic benefits at
an inflammatory challenge targeting the innate or the specific phases while minimizing off-​target side effects
adaptive immune system have dramatic consequences may then be developed.
on subsequent immune responses and pathological
Published online xx xx xxxx
outcomes, not only in acute immune responses but also

1. Edgar, R. S. et al. Peroxiredoxins are conserved 23. Nguyen, K. D. et al. Circadian gene Bmal1 regulates 40. Scheiermann, C. et al. Adrenergic nerves govern
markers of circadian rhythms. Nature 485, 459–464 diurnal oscillations of Ly6C(hi) inflammatory circadian leukocyte recruitment to tissues. Immunity
(2012). monocytes. Science 341, 1483–1488 (2013). 37, 290–301 (2012).
2. Pittendrigh, C. S. Temporal organization: reflections This paper shows that oscillations in the number of 41. Prendergast, B. J. et al. Impaired leukocyte trafficking
of a Darwinian clock-​watcher. Annu. Rev. Physiol. 55, inflammatory monocytes in tissues are driven by and skin inflammatory responses in hamsters lacking
16–54 (1993). the myeloid cell clock. a functional circadian system. Brain Behav. Immun.
3. Brown, T. M. & Piggins, H. D. Electrophysiology of the 24. Keller, M. et al. A circadian clock in macrophages 32, 94–104 (2013).
suprachiasmatic circadian clock. Prog. Neurobiol. 82, controls inflammatory immune responses. Proc. Natl 42. Haspel, J. A. et al. Circadian rhythm reprogramming
229–255 (2007). Acad. Sci. USA 106, 21407–21412 (2009). during lung inflammation. Nat. Commun. 5, 4753
4. Brancaccio, M. et al. Network-​mediated encoding of 25. Silver, A. C., Arjona, A., Hughes, M. E., Nitabach, M. N. (2014).
circadian time: the suprachiasmatic nucleus (SCN) from & Fikrig, E. Circadian expression of clock genes in 43. Gibbs, J. et al. An epithelial circadian clock controls
genes to neurons to circuits, and back. J. Neurosci. 34, mouse macrophages, dendritic cells, and B cells. pulmonary inflammation and glucocorticoid action.
15192–15199 (2014). Brain Behav. Immun. 26, 407–413 (2012). Nat. Med. 20, 919–926 (2014).
5. Herzog, E. D., Hermanstyne, T., Smyllie, N. J. & 26. Wang, X., Reece, S. P., Van Scott, M. R. & Brown, J. M. This paper shows circadian regulation of neutrophil
Hastings, M. H. Regulating the suprachiasmatic nucleus A circadian clock in murine bone marrow-​derived mast influx via rhythmic inhibition of chemoattractant
(SCN) circadian clockwork: interplay between cells modulates IgE-​dependent activation in vitro. production by bronchial epithelial cells in the lung.
cell-​autonomous and circuit-​level mechanisms. Brain Behav. Immun. 25, 127–134 (2011). 44. Kiessling, S. et al. The circadian clock in immune cells
Cold Spring Harb. Perspect. Biol. 9, a027706 (2017). 27. Baumann, A. et al. The circadian clock is functional controls the magnitude of Leishmania parasite
6. Buijs, F. N. et al. The circadian system: a regulatory in eosinophils and mast cells. Immunology 140, infection. Sci. Rep. 7, 10892 (2017).
feedback network of periphery and brain. Physiology 465–474 (2013). 45. Casanova-​Acebes, M. et al. Rhythmic modulation of
31, 170–181 (2016). 28. Ella, K., Csepanyi-​Komi, R. & Kaldi, K. Circadian the hematopoietic niche through neutrophil clearance.
7. Dumbell, R., Matveeva, O. & Oster, H. Circadian clocks, regulation of human peripheral neutrophils. Brain Cell 153, 1025–1035 (2013).
stress, and immunity. Front. Endocrinol. 7, 37 (2016). Behav. Immun. 57, 209–221 (2016). This study describes how rhythmic homing of aged
8. Thaiss, C. A. et al. Microbiota diurnal rhythmicity 29. Arjona, A. & Sarkar, D. K. Circadian oscillations neutrophils generates homeostatic cues that
programs host transcriptome oscillations. Cell 167, of clock genes, cytolytic factors, and cytokines trigger release of haematopoietic stem cells from
1495–1510 (2016). in rat NK cells. J. Immunol. 174, 7618–7624 the bone marrow.
9. Bass, J. & Lazar, M. A. Circadian time signatures of (2005). 46. Nussbaum, J. C. et al. Type 2 innate lymphoid cells
fitness and disease. Science 354, 994–999 (2016). 30. Oliva-​Ramirez, J., Moreno-​Altamirano, M. M., Pineda-​ control eosinophil homeostasis. Nature 502,
10. Panda, S. Circadian physiology of metabolism. Science Olvera, B., Cauich-​Sanchez, P. & Sanchez-​Garcia, F. J. 245–248 (2013).
354, 1008–1015 (2016). Crosstalk between circadian rhythmicity, mitochondrial 47. Chong, S. Z. et al. CXCR4 identifies transitional bone
11. Spoelstra, K., Wikelski, M., Daan, S., Loudon, A. S. & dynamics and macrophage bactericidal activity. marrow premonocytes that replenish the mature
Hau, M. Natural selection against a circadian clock Immunology 143, 490–497 (2014). monocyte pool for peripheral responses. J. Exp. Med.
gene mutation in mice. Proc. Natl Acad. Sci. USA 113, 31. Gibbs, J. E. et al. The nuclear receptor REV-​ERBalpha 213, 2293–2314 (2016).
686–691 (2016). mediates circadian regulation of innate immunity 48. Bellet, M. M. et al. Circadian clock regulates the host
12. McHill, A. W. & Wright, K. P. Jr. Role of sleep and through selective regulation of inflammatory response to Salmonella. Proc. Natl Acad. Sci. USA
circadian disruption on energy expenditure and in cytokines. Proc. Natl Acad. Sci. USA 109, 582–587 110, 9897–9902 (2013).
metabolic predisposition to human obesity and (2012). 49. Rijo-​Ferreira, F., Pinto-​Neves, D., Barbosa-​Morais, N. L.,
metabolic disease. Obes. Rev. 18 (Suppl. 1), 15–24 This article presents the first demonstration that Takahashi, J. S. & Figueiredo, L. M. Trypanosoma
(2017). synthetic ligands targeting a circadian clock protein brucei metabolism is under circadian control.
13. Qian, J. & Scheer, F. A. Circadian system and glucose modify inflammatory responses. Nat. Microbiol. 2, 17032 (2017).
metabolism: implications for physiology and disease. 32. Nakamura, Y., Ishimaru, K., Shibata, S. & Nakao, A. 50. Majumdar, T., Dhar, J., Patel, S., Kondratov, R. &
Trends Endocrinol. Metab. 27, 282–293 (2016). Regulation of plasma histamine levels by the mast cell Barik, S. Circadian transcription factor BMAL1
14. Reitz, C. J. & Martino, T. A. Disruption of circadian clock and its modulation by stress. Sci. Rep. 7, 39934 regulates innate immunity against select RNA viruses.
rhythms and sleep on critical illness and the impact (2017). Innate Immun. 23, 147–154 (2017).
on cardiovascular events. Curr. Pharm. Des. 21, 33. Nakamura, Y. et al. Circadian regulation of allergic 51. Edgar, R. S. et al. Cell autonomous regulation of
3505–3511 (2015). reactions by the mast cell clock in mice. J. Allergy Clin. herpes and influenza virus infection by the circadian
15. Yuan, X. et al. Night shift work increases the risks of Immunol. 133, 568–575 (2014). clock. Proc. Natl Acad. Sci. USA 113, 10085–10090
multiple primary cancers in women: a systematic 34. Logan, R. W., Wynne, O., Levitt, D., Price, D. & (2016).
review and meta-​analysis of 61 articles. Cancer Sarkar, D. K. Altered circadian expression of cytokines 52. Fernandes, G., Halberg, F., Yunis, E. J. & Good, R. A.
Epidemiol. Biomarkers Prev. 27, 25–40 (2018). and cytolytic factors in splenic natural killer cells of Circadian rhythmic plaque-​forming cell response of
16. Berenbaum, F. & Meng, Q. J. The brain-​joint axis in Per1(−/−) mutant mice. J. Interferon Cytokine Res. 33, spleens from mice immunized with SRBC. J. Immunol.
osteoarthritis: nerves, circadian clocks and beyond. 108–114 (2013). 117, 962–966 (1976).
Nat. Rev. Rheumatol. 12, 508–516 (2016). 35. Logan, R. W. et al. Chronic shift-​lag alters the circadian 53. Kaplan, M. S. et al. Circadian rhythm of stimulated
17. Cutolo, M. Rheumatoid arthritis: circadian and clock of NK cells and promotes lung cancer growth in lymphocyte blastogenesis. A 24 h cycle in the
circannual rhythms in RA. Nat. Rev. Rheumatol. 7, rats. J. Immunol. 188, 2583–2591 (2012). mixed leukocyte culture reaction and with SKSD
500–502 (2011). 36. Lam, M. T. et al. Rev-​Erbs repress macrophage gene stimulation. J. Allergy Clin. Immunol. 58, 180–189
18. Halberg, F., Johnson, E. A., Brown, B. W. & Bittner, J. J. expression by inhibiting enhancer-​directed (1976).
Susceptibility rhythm to E. coli endotoxin and transcription. Nature 498, 511–515 (2013). 54. Druzd, D. et al. Lymphocyte circadian clocks control
bioassay. Proc. Soc. Exp. Biol. Med. 103, 142–144 37. Sato, S. et al. A circadian clock gene, Rev-​erbα, lymph node trafficking and adaptive immune
(1960). modulates the inflammatory function of macrophages responses. Immunity 46, 120–132 (2017).
19. Curtis, A. M., Bellet, M. M., Sassone-​Corsi, P. & through the negative regulation of Ccl2 expression. This study shows dynamic regulation of lymphocyte
O’Neill, L. A. Circadian clock proteins and immunity. J. Immunol. 192, 407–417 (2014). numbers in blood, lymph node and lymph, driven
Immunity 40, 178–186 (2014). 38. Narasimamurthy, R. et al. Circadian clock protein by rhythmic expression of migration and egress
20. Labrecque, N. & Cermakian, N. Circadian clocks in the cryptochrome regulates the expression of factors.
immune system. J. Biol. Rhythms 30, 277–290 (2015). proinflammatory cytokines. Proc. Natl Acad. Sci. USA 55. Hemmers, S. & Rudensky, A. Y. The cell-​intrinsic
21. Man, K., Loudon, A. & Chawla, A. Immunity around 109, 12662–12667 (2012). circadian clock is dispensable for lymphocyte
the clock. Science 354, 999–1003 (2016). 39. Born, J., Lange, T., Hansen, K., Molle, M. & Fehm, H. L. differentiation and function. Cell Rep. 11,
22. Scheiermann, C., Kunisaki, Y. & Frenette, P. S. Effects of sleep and circadian rhythm on human 1339–1349 (2015).
Circadian control of the immune system. Nat. Rev. circulating immune cells. J. Immunol. 158, 56. Bollinger, T. et al. Circadian clocks in mouse and
Immunol. 13, 190–198 (2013). 4454–4464 (1997). human CD4 + T cells. PLoS ONE 6, e29801 (2011).

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

57. Sun, Y. et al. MOP3, a component of the molecular 78. Curtis, A. M. et al. Circadian control of innate 102. Takeda, N. & Maemura, K. Circadian clock and the
clock, regulates the development of B cells. immunity in macrophages by miR-155 targeting onset of cardiovascular events. Hypertens. Res. 39,
Immunology 119, 451–460 (2006). Bmal1. Proc. Natl Acad. Sci. USA 112, 7231–7236 383–390 (2016).
58. Yu, X. et al. TH17 cell differentiation is regulated by (2015). 103. Culic, V. Daylight saving time transitions and acute
the circadian clock. Science 342, 727–730 (2013). 79. Huo, M. et al. Myeloid Bmal1 deletion increases myocardial infarction. Chronobiol. Int. 30, 662–668
59. Yu, X. et al. The basic leucine zipper transcription monocyte recruitment and worsens atherosclerosis. (2013).
factor NFIL3 directs the development of a common FASEB J. 31, 1097–1106 (2016). 104. Dopico, X. C. et al. Widespread seasonal gene
innate lymphoid cell precursor. eLife 3, e04406 80. Mukherji, A., Kobiita, A., Ye, T. & Chambon, P. expression reveals annual differences in human
(2014). Homeostasis in intestinal epithelium is orchestrated immunity and physiology. Nat. Commun. 6, 7000
60. Qiu, J. et al. Group 3 innate lymphoid cells inhibit by the circadian clock and microbiota cues transduced (2015).
T-​cell-mediated intestinal inflammation through aryl by TLRs. Cell 153, 812–827 (2013). 105. Donaldson, G. C. & Wedzicha, J. A. The causes and
hydrocarbon receptor signaling and regulation of 81. Wang, Y. et al. The intestinal microbiota regulates consequences of seasonal variation in COPD
microflora. Immunity 39, 386–399 (2013). body composition through NFIL3 and the circadian exacerbations. Int. J. Chron. Obstruct Pulmon. Dis. 9,
61. Farez, M. F. et al. Melatonin contributes to the clock. Science 357, 912–916 (2017). 1101–1110 (2014).
seasonality of multiple sclerosis relapses. Cell 162, Along with previous work from the same authors 106. Spelman, T. et al. Seasonal variation of relapse rate in
1338–1352 (2015). (reference 58), this study links the clock protein multiple sclerosis is latitude dependent. Ann. Neurol.
62. Martinez-​Llordella, M. et al. CD28-inducible REV-​ERBα with rhythmic expression of NFIL3, a 76, 880–890 (2014).
transcription factor DEC1 is required for efficient critical regulator of both TH17 cell differentiation 107. Zhang, R., Lahens, N. F., Ballance, H. I., Hughes, M. E.
autoreactive CD4 + T cell response. J. Exp. Med. 210, and enterocyte lipid metabolism. & Hogenesch, J. B. A circadian gene expression atlas
1603–1619 (2013). 82. Zhang, D. et al. Neutrophil ageing is regulated by the in mammals: implications for biology and medicine.
63. Kreslavsky, T. et al. Essential role for the transcription microbiome. Nature 525, 528–532 (2015). Proc. Natl Acad. Sci. USA 111, 16219–16224
factor Bhlhe41 in regulating the development, 83. Martin, C. et al. Chemokines acting via CXCR2 and (2014).
self-​renewal and BCR repertoire of B-1a cells. CXCR4 control the release of neutrophils from the This article presents a comprehensive analysis of
Nat. Immunol. 18, 442–455 (2017). bone marrow and their return following senescence. oscillating genes and non-​coding RNAs in murine
64. Besedovsky, L., Born, J. & Lange, T. Endogenous Immunity 19, 583–593 (2003). organs, highlighting the relationships between
glucocorticoid receptor signaling drives rhythmic 84. Tanji-​Matsuba, K. et al. Functional changes in aging rhythmic genes, disease-​associated genes and
changes in human T-​cell subset numbers and the polymorphonuclear leukocytes. Circulation 97, 91–98 current drug targets.
expression of the chemokine receptor CXCR4. (1998). 108. Buttgereit, F. et al. Efficacy of modified-​release versus
FASEB J. 28, 67–75 (2014). 85. Liang, X., Bushman, F. D. & FitzGerald, G. A. standard prednisone to reduce duration of morning
65. Dimitrov, S. et al. Cortisol and epinephrine control Rhythmicity of the intestinal microbiota is regulated by stiffness of the joints in rheumatoid arthritis
opposing circadian rhythms in T cell subsets. gender and the host circadian clock. Proc. Natl Acad. (CAPRA-1): a double-​blind, randomised controlled
Blood 113, 5134–5143 (2009). Sci. USA 112, 10479–10484 (2015). trial. Lancet 371, 205–214 (2008).
66. Suzuki, K., Hayano, Y., Nakai, A., Furuta, F. & Noda, M. 86. Thaiss, C. A. et al. Transkingdom control of microbiota 109. Lamia, K. A. et al. Cryptochromes mediate rhythmic
Adrenergic control of the adaptive immune response diurnal oscillations promotes metabolic homeostasis. repression of the glucocorticoid receptor. Nature 480,
by diurnal lymphocyte recirculation through lymph Cell 159, 514–529 (2014). 552–556 (2011).
nodes. J. Exp. Med. 213, 2567–2574 (2016). 87. Balsalobre, A., Damiola, F. & Schibler, U. A serum 110. Okabe, T. et al. REV-​ERBalpha influences the stability
This study shows how adrenergic tone modulates shock induces circadian gene expression in and nuclear localization of the glucocorticoid receptor.
lymphocyte trafficking rhythms and humoral mammalian tissue culture cells. Cell 93, 929–937 J. Cell Sci. 129, 4143–4154 (2016).
immune responses through β2-adrenergic (1998). 111. Solt, L. A. et al. Regulation of circadian behaviour
receptors expressed by lymphocytes. 88. Balsalobre, A. et al. Resetting of circadian time in and metabolism by synthetic REV-​ERB agonists.
67. Zhao, Y. et al. Uncovering the mystery of opposite peripheral tissues by glucocorticoid signaling. Science Nature 485, 62–68 (2012).
circadian rhythms between mouse and human 289, 2344–2347 (2000). 112. Sitaula, S., Billon, C., Kamenecka, T. M., Solt, L. A. &
leukocytes in humanized mice. Blood 130, 89. Shackelford, P. G. & Feigin, R. D. Periodicity of Burris, T. P. Suppression of atherosclerosis by
1995–2005 (2017). susceptibility to pneumococcal infection: influence of synthetic REV-​ERB agonist. Biochem. Biophys. Res.
This is a very interesting report of cell-​intrinsic light and adrenocortical secretions. Science 182, Commun. 460, 566–571 (2015).
phase encoding of leukocytes, driven by opposite 285–287 (1973). 113. Kojetin, D., Wang, Y., Kamenecka, T. M. & Burris, T. P.
effects of p38MAPK–MK2 signalling upon HIF1α 90. Early, J. O. & Curtis, A. M. Immunometabolism: Is it Identification of SR8278, a synthetic antagonist of the
induction and CXCR4 expression. under the eye of the clock? Semin. Immunol. 28, nuclear heme receptor REV-​ERB. ACS Chem. Biol. 6,
68. Shimba, A. et al. Glucocorticoids drive diurnal 478–490 (2016). 131–134 (2011).
oscillations in T cell distribution and responses 91. Ando, N. et al. Circadian gene clock regulates 114. Gagnidze, K. et al. Nuclear receptor REV-​ERBα
by inducing interleukin-7 receptor and CXCR4. psoriasis-​like skin inflammation in mice. J. Invest. mediates circadian sensitivity to mortality in murine
Immunity 48, 286–298.e6 (2018). Dermatol. 135, 3001–3008 (2015). vesicular stomatitis virus-​induced encephalitis.
This work illustrates an immune-​enhancing role of 92. Castanon-​Cervantes, O. et al. Dysregulation of Proc. Natl Acad. Sci. USA 113, 5730–5735 (2016).
glucocorticoids via upregulation of homing inflammatory responses by chronic circadian 115. Hirota, T. et al. Identification of small molecule
receptors, promoting rhythmic T cell accumulation disruption. J. Immunol. 185, 5796–5805 (2010). activators of cryptochrome. Science 337, 1094–1097
and heightened responses to systemic infection. 93. Pagel, R. et al. Circadian rhythm disruption impairs (2012).
69. Forster, R., Davalos-​Misslitz, A. C. & Rot, A. CCR7 tissue homeostasis and exacerbates chronic 116. Hand, L. E. et al. The circadian clock regulates
and its ligands: balancing immunity and tolerance. inflammation in the intestine. FASEB J. 31, inflammatory arthritis. FASEB J. 30, 3759–3770
Nat. Rev. Immunol. 8, 362–371 (2008). 4707–4719 (2017). (2016).
70. Stein, J. V. & Nombela-​Arrieta, C. Chemokine control 94. Summa, K. C. et al. Disruption of the circadian clock in 117. Partch, C. L., Green, C. B. & Takahashi, J. S. Molecular
of lymphocyte trafficking: a general overview. mice increases intestinal permeability and promotes architecture of the mammalian circadian clock. Trends
Immunology 116, 1–12 (2005). alcohol-​induced hepatic pathology and inflammation. Cell Biol. 24, 90–99 (2014).
71. Cyster, J. G. & Schwab, S. R. Sphingosine-1-phosphate PLoS ONE 8, e67102 (2013). 118. Guilding, C. et al. Suppressed cellular oscillations
and lymphocyte egress from lymphoid organs. 95. Li, W. Q., Qureshi, A. A., Schernhammer, E. S. & in after-​hours mutant mice are associated with
Annu. Rev. Immunol. 30, 69–94 (2012). Han, J. Rotating night-​shift work and risk of psoriasis enhanced circadian phase-​resetting. J. Physiol. 591,
72. Nakai, A., Hayano, Y., Furuta, F., Noda, M. & Suzuki, K. in US women. J. Invest. Dermatol. 133, 565–567 1063–1080 (2013).
Control of lymphocyte egress from lymph nodes (2013). 119. Meng, Q. J. et al. Setting clock speed in mammals: the
through β2-adrenergic receptors. J. Exp. Med. 211, 96. Nojkov, B., Rubenstein, J. H., Chey, W. D. & CK1 epsilon tau mutation in mice accelerates circadian
2583–2598 (2014). Hoogerwerf, W. A. The impact of rotating shift pacemakers by selectively destabilizing PERIOD
73. Esquifino, A. I., Selgas, L., Arce, A., Maggiore, V. D. & work on the prevalence of irritable bowel syndrome proteins. Neuron 58, 78–88 (2008).
Cardinali, D. P. Twenty-​four-hour rhythms in immune in nurses. Am. J. Gastroenterol. 105, 842–847 120. Yamaguchi, S. et al. Role of DBP in the circadian
responses in rat submaxillary lymph nodes and (2010). oscillatory mechanism. Mol. Cell. Biol. 20,
spleen: effect of cyclosporine. Brain Behav. Immun. 10, 97. Cuesta, M., Boudreau, P., Dubeau-​Laramee, G., 4773–4781 (2000).
92–102 (1996). Cermakian, N. & Boivin, D. B. Simulated night shift 121. Ueda, H. R. et al. System-​level identification of
74. Fortier, E. E. et al. Circadian variation of the response disrupts circadian rhythms of immune functions in transcriptional circuits underlying mammalian
of T cells to antigen. J. Immunol. 187, 6291–6300 humans. J. Immunol. 196, 2466–2475 (2016). circadian clocks. Nat. Genet. 37, 187–192
(2011). 98. Durrington, H. J., Farrow, S. N., Loudon, A. S. & (2005).
75. Silver, A. C., Arjona, A., Walker, W. E. & Fikrig, E. The Ray, D. W. The circadian clock and asthma. Thorax 69, 122. Fu, L. & Lee, C. C. The circadian clock: pacemaker
circadian clock controls toll-​like receptor 9-mediated 90–92 (2014). and tumour suppressor. Nat. Rev. Cancer 3, 350–361
innate and adaptive immunity. Immunity 36, 99. Olsen, N. J., Brooks, R. H. & Furst, D. Variability of (2003).
251–261 (2012). immunologic and clinical features in patients with 123. Yu, E. A. & Weaver, D. R. Disrupting the circadian
76. Sutton, C. E. et al. Loss of the molecular clock in rheumatoid arthritis studied over 24 h. J. Rheumatol. clock: gene-​specific effects on aging, cancer, and other
myeloid cells exacerbates T cell-​mediated CNS 20, 940–943 (1993). phenotypes. Aging 3, 479–493 (2011).
autoimmune disease. Nat. Commun. 8, 1923 (2017). 100. Panzer, S. E., Dodge, A. M., Kelly, E. A. & Jarjour, N. N. 124. Asher, G. & Sassone-​Corsi, P. Time for food: the
This report shows the importance of appropriate Circadian variation of sputum inflammatory cells in intimate interplay between nutrition, metabolism,
immune cell crosstalk in an EAE model, as mild asthma. J. Allergy Clin. Immunol. 111, 308–312 and the circadian clock. Cell 161, 84–92 (2015).
disruption of the myeloid clock increases TH1 and (2003). 125. Cho, H. et al. Regulation of circadian behaviour and
TH17 cell responses in the central nervous system. 101. Perry, M. G., Kirwan, J. R., Jessop, D. S. & Hunt, L. P. metabolism by REV-​ERB-alpha and REV-​ERB-beta.
77. Long, J. E. et al. Morning vaccination enhances Overnight variations in cortisol, interleukin 6, tumour Nature 485, 123–127 (2012).
antibody response over afternoon vaccination: a necrosis factor alpha and other cytokines in people 126. Zhang, Y. et al. Discrete functions of nuclear receptor
cluster-​randomised trial. Vaccine 34, 2679–2685 with rheumatoid arthritis. Ann. Rheum. Dis. 68, Rev-​erbα couple metabolism to the clock. Science
(2016). 63–68 (2009). 348, 1488–1492 (2015).

www.nature.com/nri
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews
127. Masri, S. et al. Lung adenocarcinoma distally rewires 133. Borniger, J. C. et al. Time-​of-day dictates transcriptional Competing interests
hepatic circadian homeostasis. Cell 165, 896–909 inflammatory responses to cytotoxic chemotherapy. The authors declare no competing interests.
(2016). Sci. Rep 7, 41220 (2017).
128. Sulli, G. et al. Pharmacological activation of REV-​ERBs 134. Durrington, H. J., Farrow, S. N. & Ray, D. Recent Publisher’s note
is lethal in cancer and oncogene-​induced senescence. advances in chronotherapy for the management of Springer Nature remains neutral with regard to jurisdictional
Nature 553, 351–355 (2018). asthma. ChronoPhysiology Ther. 4, 125–135 (2014). claims in published maps and institutional affiliations.
This study shows that agonism of REV-​ERBs is
specifically lethal to cancer cells via inhibition of Acknowledgements Supplementary information
autophagy and lipogenesis The authors thank V. Lavilla for creating the video and Supplementary information is available for this paper at
129. Fonken, L. K. et al. Microglia inflammatory responses M. F. Loudon for providing the voice-​over to it. C.S. is funded https://doi.org/10.1038/s41577-018-0008-4.
are controlled by an intrinsic circadian clock. Brain by the German Research Foundation (DFG) (Emmy-​Noether
Behav. Immun. 45, 171–179 (2015). grant (SCHE 1645/2-1) and SFB914 projects B09 and Z03), Related links
130. Rudic, R. D. et al. Bioinformatic analysis of circadian the European Research Council (ERC; starting grant 635872, Manchester Biological Timing:
gene oscillation in mouse aorta. Circulation 112, CIRCODE), the DZHK (German Centre for Cardiovascular https://www.bmh.manchester.ac.uk/research/biological-​timing/
2716–2724 (2005). Research) and the BMBF (German Ministry of Education and Loudon Laboratory:
131. Nakazato, R. et al. The intrinsic microglial clock system Research). J.G. is an Arthritis Research UK Career http://www.manchester.ac.uk/research/Andrew.loudon/
regulates interleukin-6 expression. Glia 65, 198–208 Development Fellow (Ref. 20629). A.L. acknowledges the personaldetails
(2017). support of the Wellcome Trust (grant 107851/Z/15/Z). scheiermann Laboratory:
132. Alvarez-​Sanchez, N. et al. Melatonin controls http://scheiermannlab.de/
experimental autoimmune encephalomyelitis by Author contributions Gibbs Laboratory:
altering the T effector/regulatory balance. Brain Behav. All authors contributed to the research, discussion of content, https://www.research.manchester.ac.uk/portal/Julie.Gibbs.html
Immun. 50, 101–114 (2015). writing and review of this manuscript.

Nature Reviews | Immunology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

Das könnte Ihnen auch gefallen