You are on page 1of 10

FOCUS | Review Article

FOCUS | Review Article https://doi.org/10.1038/s41590-019-0399-9

https://doi.org/10.1038/s41590-019-0399-9

Adaptation and memory in immune responses


Gioacchino Natoli   1,2* and Renato Ostuni   3,4*

Adaptation is the ability of cells, tissues and organisms to rapidly and reversibly modify their properties to maximize fitness in
a changing environment. The activity of immune-system components unfolds in the remarkably heterogeneous milieus to which
they are exposed in different tissues, during homeostasis or during various acute or chronic pathological states. Therefore,
adaptation is essential for immune cells to tune their responses to a large variety of contexts and conditions. The adaptation of
immune cells reflects the integration of multiple inputs acting simultaneously or in a temporal sequence, which eventually leads
to transcriptional reprogramming and to various functional consequences, some of which extend beyond the duration of the
stimulus. A range of adaptive responses have been observed in both adaptive immune cells and innate immune cells; these are
referred to with terms such as ‘plasticity’, ‘priming’, ‘training’, ‘exhaustion’ and ‘tolerance’, among others, all of which can be
useful for defining a certain immunological process or outcome but whose underlying molecular frameworks are often incom-
pletely understood. Here we review and analyze mechanisms of adaptation and memory in immunity with the aim of providing
basic concepts that rationalize the properties and molecular bases of these essential processes.

I
mmunological memory is defined as the ability of immune cells long-lasting effects of cell stimulation in the immune system as
to specifically ‘remember’ the first encounter with a given anti- forms of adaptation characterized by slow reversibility (Fig.  1).
gen and to mount a secondary response that is faster and greater Notably, lymphocytes also show adaptive responses commonly
in magnitude than the primary response. The specificity of these referred to as ‘plasticity’: the ability of a given lymphocyte to func-
memory responses is very high and is based on irreversible changes tionally adapt its effector responses to a changing environment.
in the DNA sequence during and after rearrangement of the antigen Along the same lines, T cell exhaustion can be considered a form
receptors on B lymphocytes and T lymphocytes. A critical contribu- of adaptation characterized by remarkable stability. Therefore,
tion to the stability of memory responses is provided by changes while bona fide immunological memory is a property of lympho-
in DNA methylation (notably, passive demethylation) that can be cytes, adaptation is a property of essentially all immune-cell types
transmitted to daughter cells during the clonal expansion of anti- (as well as non–immune-cell types), with various degrees of stabil-
gen-stimulated naive lymphocytes and thus enable the fast and effi- ity but without the specificity of true memory. The peculiar case of
cient re-activation of genes encoding critical effector molecules in memory in natural killer (NK) cells has been discussed in excellent
memory cells1,2. reviews6,7. In this specific case, the enhanced responsiveness that
Immunological adaptation is instead the process whereby leuko- follows the initial activation and clonal expansion phase can persist
cytes exposed to environmental stimuli modify their properties in for months8 and resembles memory in the adaptive system (possibly
a way that influences future responses to the same or other stimuli. also from a mechanistic point of view, as discussed below), with the
The adapted state is reversible and dynamic, which means that it obvious difference of the innate-type, limited specificity of NK cell
can be resolved when environmental conditions return to the initial responses. In the next paragraphs, we will focus on the adaptation
state and/or it can be modified in response to subsequent micro- of immune cells, by first defining the properties of the responses to
environmental changes. As discussed below, the reversibility of various acute and chronic stimuli and then by discussing mecha-
adaptive states is linked to specific mechanisms that drive molecu- nisms that underlie those processes.
lar alterations that can be rapidly ‘erased’ even in non-dividing cells,
such as those that occur at the chromatin, transcriptional, post-tran- Properties of eliciting stimuli affect adaptive phenotypes
scriptional or metabolic level. Although they are reversible, adaptive Adaptations in the immune system are extremely diverse in terms of
phenotypes and the associated molecular changes may in some cases biological output, and they affect future responses and mechanisms
persist for variable time windows (up to weeks or even months)3,4. that control the establishment and maintenance of the adapted
Hence, the perpetuation of adaptation in some contexts, particularly state (Fig.  2). This diversity is a direct reflection of the multitude
in innate immunity, has been considered a form of immunological of stimuli that can modify the activity of immune cells as well as
‘memory’ (‘trained immunity’ or ‘short term memory’)3,4. of additional quantitative properties of environmental signals, such
Conceptually, the degree of reversibility of an acquired prop- as their intensity or persistence. It is well known that while distinct
erty of immune cells should define the border between sustained stimuli typically elicit specific adaptations, a given signal may trig-
adaptation and true memory (Fig.  1). The other critical feature ger very different types of adaptations if it is present at a low or high
that distinguishes memory from adaptation is the specificity of the dose or if it is sensed for a short or long period of time. For example,
response. Memory is specific by definition. Conversely, an adaptive repeated or persistent exposure of macrophages to high doses of
process can be pervasive or global in nature. Indeed, while memory inflammatory agents, such as the microbial component lipopoly-
lymphocytes are selectively activated by cognate antigens, adapted saccharide (LPS), often elicits a state of hypo-responsiveness that
myeloid cells can display a general state of hyper- or hypo-respon- prevents further re-induction of genes encoding inflammatory mol-
siveness to a broad set of unrelated stimuli5. ecules (called ‘inflammatory genes’ here)5. On the other hand, very
Because memory implies specificity, we propose to restrict the low doses of LPS or other inflammatory stimuli are generally linked
use of this term to antigen-specific immunity and to consider the to a potentiation of the response to future challenges3.

1
Humanitas University, Pieve Emanuele, Milan, Italy. 2IRCCS Humanitas, Rozzano, Milan, Italy. 3San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget),
IRCCS San Raffaele Scientific Institute, Milan, Italy. 4Vita-Salute San Raffaele University, Milan, Italy. *e-mail: gioacchino.natoli@hunimed.eu;
ostuni.renato@hsr.it

Nature Immunology | www.nature.com/natureimmunology


Review Article | FOCUS
https://doi.org/10.1038/s41590-019-0399-9 NATure Immunology

Adaptation Memory rophages16–19. With some notable exceptions, represented by latent


Reversibility Yes No
or de novo enhancers20,21, the set of enhancers made accessible by
lineage-determining transcription factors serves as a platform onto
Short-term or
Persistence
long-term
Long-term which stimulus-responsive transcription factors land after activa-
tion, which thus links cell identity to activation14,15. Tissue-resident
Specificity No Yes
macrophages are exposed to cues that are linked to the specific prop-
• Receptor signaling/recycling • DNA sequence alterations
erties of the organ, such as heme in the spleen, retinoic acid in the
• Metabolic reprogramming • Epigenetic modifications peritoneum or the microbiota in the intestine13. Such signals in turn
• Chromatin/histone (DNA methylation) stimulate the activity of tissue-restricted transcription factors, such
Mechanisms modification • Self-sustaining feedback
• TF occupancy/distribution loops
as GATA-6 in the peritoneum22–24, SPI-C in the spleen25, LXRα and
• Induction of long-lived ID3 in the liver26,27 and many others, which control the selection of
mediators tissue-specific enhancer landscapes and transcriptional programs in
the tissue-resident macrophages28,29. Despite being conditioned by
Innate, adaptive immune cells
Target cells
Non-immune cells
Adaptive immune cells local cues, the regulatory repertoire of resident macrophages, and
eventually the tissue-specific transcriptional output of these cells, is
to a large extent plastic and reversible, as indicated by experiments
Fig. 1 | Discriminative properties of adaptation and memory in the
in which macrophages are transferred from one tissue to another28.
immune system. Adaptation to and memory of environmental stimuli
The distinct genomic organizations of resident macrophages
should be defined mainly by the degree of reversibility or persistence of
represent paradigmatic adaptations that allow context-dependent
the elicited state and by the specificity of the response after secondary
responses to stimulation. For example, microglia have high expres-
stimulation. In this framework, adaptation is controlled by molecular
sion of transcription factors such as MEF2C30, MAFB31 and NR4A132,
mechanisms that involve reversible biochemical, metabolic or chromatin
which act to limit the induction of inflammatory genes in response
alterations that are diluted across cell division, while memory relies on
to stimulation and thus prevent neuronal damage. Expression of
stable genetic or epigenetic changes, as well as on self-sustaining loops.
the transcription factor SPI-C in some tissue macrophages, such as
Because almost all types of cells are equipped with molecular machinery
spleen macrophages, Küpffer cells and intestinal macrophages, sup-
able to trigger reversible changes, adaptation can be considered a
presses induction of the inflammatory cytokines interleukin 6 (IL-6)
universal cellular property. Enzymes (such as the RAG recombinases or
and IL-1α (but not that of TNF); here, SPI-C acts by interfering with
the activation-induced cytidine deaminase AID) that can elicit irreversible
interaction of the transcription factor NF-κB subunit p65 with the
changes are instead active only in cells of the adaptive immune system,
transcription factor IRF5 in response to LPS33. Conversely, GATA-6
which underlies memory in this compartment.
inhibits the expression of genes encoding molecules linked to type 2
immunity, such as Mrc1, Cd163, Arg1 and Chi3l323, which indicates
The divergent outputs of possible adaptations to a common that this transcription factor might boost inflammatory responses
stimulus probably reflect the associated functional requirements: in peritoneal macrophages, whose activation reflects critical emer-
preventing immune system–mediated tissue damage and excessive gencies that require rapid and effective responses. Emerging evi-
inflammatory responses on the one hand and enhancing immune dence indicates that the ability to adapt to local signals is a general
responses to concomitant or future threats on the other. In addi- feature of immune cells, although it is unclear whether the underly-
tion to modulating the outputs of the adapted states, the intensity ing molecular mechanisms follow the same rules as those described
of stimulation can underlie the extent of the persistence and revers- above for macrophages. For example, neutrophils infiltrate tissues at
ibility of the states. Transient and/or low-dose stimulation typically steady state34 and display phenotypic and functional heterogeneity35.
elicits reversible adaptations that may be resolved in short time Phagocytosis of neutrophils in the bone marrow regulates circadian
frames (hours to days) or long time frames (weeks to months); this oscillations of the number of mobilized hematopoietic progenitor
is exemplified by the short-term priming effect of inflammatory cells36, while their phagocytosis in the intestine suppresses sys-
signals on macrophages (described below) or by the long-term, but temic levels of the cytokine G-CSF and controls overall bone mar-
flexible, adaptations of CD4+ helper T cells after transient exposure row activity34. Additional tissue-specific functions of neutrophils
to polarizing cytokines9. On the other hand, persistent and/or high- include the support of B cell maturation in the spleen37 or the regu-
dose stimulation often elicits persistent adaptations that, to some lation of diurnal transcriptome changes in the lungs34. Along the
extent, may be not easily, rapidly or completely reverted. For exam- same lines, conventional T lymphocytes, innate lymphoid cells and
ple, chronic antigen stimulation of CD8+ T cells leads to a state of NK cells show tissue-specific profiles and functions38.
exhaustion that is associated with stable transcriptional reprogram- Even if located within the same tissue, immune cells may still
ming and acquisition of specific functional features, which can be function differently according to their ontogeny, as they often rep-
reverted only partially after blockade of inhibitory immunological resent the progeny of distinct hematopoietic precursor cells and
checkpoints10,11. include cell populations that are maintained independently from
adult hematopoietic stem cells (HSCs)39. This is now well estab-
Adaptation to tissue contexts lished for tissue-resident macrophages40, as well as for mast cells41,42,
A common output of adaptation is the acquisition of context- and for lymphocyte populations such as innate-like B-1 cells43, γδ
dependent activities, as exemplified by the tissue-specific functions T cells44 or CD8+ T cells45. Emerging evidence indicates that ontog-
of immune cells and of macrophages in particular12,13 (Fig. 2). The eny affects how immune cells interact with the environment, which
diverse activities of tissue macrophages are enabled by specific tran- in turn implies that different regulatory networks operate in cells
scriptional programs that reflect differential selection and usage of of different origin, even if the cells are exposed to the same tissue
the available cis-regulatory repertoire (specifically, the complement microenvironment. For example, adult monocyte-derived macro-
of accessible genomic regulatory elements) in response to tissue- phages that engraft the brain post-natally acquire transcriptional
specific cues14,15. Mechanistically, the first layer of genomic regu- and chromatin features that only partially resemble those of yolk
lation is provided by myeloid lineage–determining transcription sac–derived microglia and display different capacities to respond
factors such as PU.1, IRF8, C/EBPα, C/EBPβ and others, which act to inflammatory insults46,47. Distinct molecular organization and
in a combinatorial fashion during differentiation to establish and functional properties have been reported for monocyte-derived
maintain the basal and inducible cis-regulatory landscape of mac- macrophages and embryonic resident macrophages in pancreatic

Nature Immunology | www.nature.com/natureimmunology


FOCUS | Review
| Article
NATure Immunology FOCUShttps://doi.org/10.1038/s41590-019-0399-9
Review Article
NF-κB NF-κB STAT1
Ac Ac
Hyper-responsive
a Termination of stimulus
adaptation

Induction of signaling components


Induction of transcription factors (e.g., STAT1)
Metabolic reprogramming
1st stimulus Higher chromatin accessibility 2nd stimulus
(LPS) (e.g., LPS)
Receptor desensitization NF-κB
Induction of repressive factors
(e.g., miR-221/miR-222)
NF-κB Metabolic reprogramming Me3
Ac
Lower chromatin accessibility
Hypo-responsive
adaptation
Persistence of stimulus

TF A NF-κB TF A
b Ac

Poised Active
Ac
Active Active

Inactive Inactive

Inactive (e.g., tissue signal A)


Context-dependent
Inactive 2nd stimulus adaptation
1st stimulus
(e.g., LPS)
Inactive
(e.g., tissue signal B) NF-κB
Me3

Repressed
Repressed
Inactive
Inactive

Active
Active
TF B
TF B
Selection of tissue-specific enhancers
Tissue-specific response to stimulation
Tissue-specific basal gene expression

Fig. 2 | Different outputs of immunological adaptations dictated by the properties of the eliciting stimulus. a, A hypothetical locus of inflammatory
genes (blue) in macrophages showing hyper-responsive adaptations (top) or hypo-responsive adaptations (bottom) elicited by a transient (low-dose)
exposure or persistent (high-dose) exposure, respectively, to an inflammatory agent. Transient inflammatory stimulation of macrophages, such as with
LPS, triggers complex changes at the signaling, metabolic or transcriptional level that often result in increased histone acetylation (Ac) and greater
chromatin accessibility at regulatory elements of adapted genes, which in turn favors the binding of transcription factors and gene expression after
secondary exposure to another inflammatory stimulus (‘priming’ or ‘training’). On the other hand, persistent inflammatory stimulation of macrophages
often elicits negative feedback loops that lead to a reduction in signaling strength, induction of anti-inflammatory factors, such as miR-221 and miR-222,
and metabolic rewiring. Such alterations are associated with reduced chromatin accessibility and, in some cases, deposition of repressive histone marks
such as H3K27me3 (Me3) at regulatory elements of inflammatory genes, which prevents binding of transcription factors and efficient locus re-induction
(‘LPS tolerance’). b, Hypothetical context-dependent adaptation at a locus of inflammatory genes (blue) in tissue-resident macrophages. After exposure
to local cues, tissue-specific transcription factors control the selection and activation of the cis-regulatory landscape (orange or purple loci) in resident
macrophages. Transcription factor A (TF A; orange), activated by tissue-associated signal A (tissue signal A), also binds to regulatory elements of an
inflammatory gene and primes chromatin at this locus, while transcription factor B (TF B; purple), activated by tissue-associated signal B (tissue signal
B), promotes deposition of repressive histone marks at this locus. The differential chromatin organization at this same locus, imparted by the distinct
transcription factors in tissue A versus tissue B, underlies the different transcriptional outputs in resident macrophages that lead to context-dependent
adaptations.

cancer48 and glioblastoma49. Similarly, CD8+ T  cells that originate methylation profiles at regulatory elements of key genes — which
during fetal development or adult development display distinct results in distinct adaptations to identical tissue environments.
transcriptional and chromatin landscapes, which underlie differ-
ential effector capacities and dynamics45. Such observations clearly Hyper-responsive adaptation
indicate that the ontogeny of immune cells is associated with persis- Macrophages acutely exposed to LPS upregulate hundreds of
tent molecular properties that are only partially reversible through- transcripts in a highly dynamic and coordinated fashion50,51.
out the cellular life cycle. While the underlying mechanisms remain Among those, a limited set of transcripts originally referred to as
largely unknown, a sound hypothesis is that the source of progeni- ‘non-tolerized’ transcripts are induced at higher levels and with
tor cells (such as primitive HSCs versus definitive HSCs39) may faster kinetics after re-challenge with LPS 24 hours after the pri-
impart stable epigenetic features — specifically, changes in DNA- mary stimulation52. That set of genes shows enrichment for genes

Nature Immunology | www.nature.com/natureimmunology


Review Article | FOCUS
https://doi.org/10.1038/s41590-019-0399-9 NATure Immunology

Signal response Signal integration


Interestingly, immunological stimuli can elicit hyper-responsive
adapted states in non-immune cells as well, as reported for skin
State A epithelial stem cells that, after exposure to the innate immunore-
ceptor TLR7 or activators of the NLRP3 inflammasome, gained
Stimulus regenerative capacities after subsequent tissue damage59, as well
A
B as for lung basal cells, in which adaptations to IL-4 or IL-13 drive
Stimulus A
barrier-tissue dysfunction during allergy60.
B
B Hypo-responsive adaptation
C
C ? Persistent or high-dose stimulation of immune cells often elicits
C
states of adaptation characterized by reduced responsiveness to fur-
Naive D ther challenges (Fig. 2). This reflects the existence of auto-inhibitory
macrophage loops aimed at preventing excessive inflammatory responses and
D D
E immune system–mediated pathology. For example, macrophages
exposed to sustained stimulation with LPS or stimulation with a
E high dose of LPS acquire a hypo-responsive state, known as ‘LPS
tolerance’, in which a large number of inflammatory genes are not
E
efficiently re-induced after secondary stimulation with LPS or other
Principles of inducible gene expression Principles of context-dependent
inflammatory cytokines5. The blunted immune reactions by cells
gene expression that acquired a hypo-responsive state are associated with pathologi-
cal conditions such as chronic infection, in which prolonged anti-
Fig. 3 | Signal-response and signal-integration models of macrophage gen stimulation of T lymphocytes causes a gradual decrease in T cell
activation. In the current model of macrophage activation in response to effector functions and unresponsiveness to stimulation, a state often
environmental signals (left), exposure to diverse stimuli elicits specific referred to as ‘exhaustion’61. Analogously, the hypo-responsiveness
molecular changes and functional states. Analyses of the transcriptional of innate immune cells in patients with sepsis underlies immune-
and epigenomic effects of individual stimuli have provided fundamental system paralysis, a state of immunosuppression that persists after
information on the principles governing inducible gene expression50,51, clearance of the pathogen and is associated with a higher risk of
as well as on the diversity of activation states of macrophages95,96. The secondary infection, organ failure and mortality in the first weeks
model of signal integration at right shows the ability of macrophages to after resolution of sepsis62.
integrate multiple and even contrasting environmental signals into specific A variety of mechanisms have been shown to contribute to the
genomic and functional programs. Mechanistic understanding of signal LPS-tolerant state of macrophages, including receptor desensiti-
integration are expected to yield new paradigms of context-dependent gene zation, secondary activation of negative signaling modules and
expression. induction of anti-inflammatory microRNAs5. In this context, the
organization and dynamics of chromatin at regulatory elements of
inflammatory genes seem to be relevant to LPS tolerance. The lack
encoding anti-microbial effector molecules, which suggests that this of re-induction of tolerized genes correlates with defective depo-
type of adaptation may boost the protective actions of macrophages sition of histone marks associated with active transcription, chro-
directed against infection while preventing the sustained expression matin accessibility and nucleosome remodeling at promoters and
of typical inflammatory genes, whose products might cause severe enhancers of tolerized genes52,57. On the one hand, such defective
damage5. Potentiated re-induction of specific sets of genes has been deposition of histone marks at tolerized genes might reflect the
reported in macrophages or fibroblasts after repeated exposure to lower signaling strength observed after re-stimulation of toler-
diverse cytokines such as interferon-γ (IFN-γ)20, IFN-β53 or IL-420,54. ized macrophages. On the other hand, chromatin organization at
The state of heightened responsiveness acquired by macrophages in the promoters of tolerized genes differs in several ways from that
inflammatory conditions often extends to secondary exposure to of non-tolerized genes even in unstimulated macrophages, which
related, but not identical, signals (Fig. 2). For example, pre-exposure suggests that the propensity for or refractoriness to tolerization may
of macrophages to IFN-γ boosts the expression of many inflamma- be an intrinsic property of LPS-responsive genomic regulatory ele-
tory genes after subsequent stimulation with LPS55; innate immune ments52. In this context, the capacity of some stimuli such as inter-
cells stimulated with β-glucans, key components of the fungal cell ferons or β-glucans to revert tolerance63,64 might be related to their
wall, mount more-efficient inflammatory responses to a variety of ability to induce chromatin changes that make some inflammatory
bacterial pathogens in vitro and in vivo3. genes capable of responding efficiently to the diminished signals
Mechanistically, increased expression of inflammatory genes observed in tolerized cells and thus in fact compensate for the lower
after re-stimulation of macrophages relies on various processes, signaling strength65. Along the same lines, non-tolerized genes
including increased expression of signaling-pathway com- — specifically, genes that are efficiently re-induced by secondary
ponents, metabolic reprogramming, such as the mTOR- and stimulation of tolerized macrophages — may be characterized by
HIF1-α-dependent glycolytic switch induced by β-glucans56, a chromatin state that enables efficient activation in response to
and the deposition at cis-regulatory elements of histone modi- weaker upstream signals.
fications, whose persistence after removal of the initial stimu-
lus might facilitate or potentiate secondary activation20,52,57. In Mechanisms for the persistence of adaptive states
principle, therapeutic strategies that promote adapted states of Most adaptations in the immune system are plastic and are thus rap-
hyper-responsiveness in innate immune cells could be exploited idly reversible. However, in specific cases, these states can persist
to boost vaccination efficacy as well as to promote anti-tumor and display memory-like features. In principle, persistent adapta-
immunity. On the other hand, this type of adaptation may also tion to a transient stimulation may be acquired and maintained via
exacerbate immune reactions in chronic inflammatory diseases at least three non–mutually exclusive mechanisms.
or in autoimmunity. Repeated systemic administration of low The first mechanism is the activation of a self-sustaining positive
doses of LPS boosts the expression of inflammatory genes in feedback mechanism able to independently propagate the initial
brain macrophages, which leads to exacerbated neuropathology58. response, even after removal of the eliciting stimulus. Mathematical

Nature Immunology | www.nature.com/natureimmunology


FOCUS | Review
| Article
NATure Immunology FOCUShttps://doi.org/10.1038/s41590-019-0399-9
Review Article
models have shown that memory based on transcriptional circuits The role of histone modifications
can typically develop when the system is ‘bi-stable’; that is, when, in The current evidence of memory-like, long-lasting adaptive phe-
response to stimulation, the system can move from one stable state notypes in the innate immune system has been linked to the per-
to an alternative, self-maintained, stable state66. This memory is sistence of stimulus-induced histone modifications associated with
thus restricted to systems that can exist in only two main states (for gene activation3. Given the current knowledge on the turnover of
example, on and off, or active and repressed), such as the lac operon, nucleosomes and their modifications, the amino termini (‘tails’)
which can be present in an active state or in a repressed state, and of nucleosomal histones are subjected to a large number of modi-
the phage lambda, which exists in a lysogenic phase (in which the fications, usually characterized by a very fast turnover (minutes
prophage genome is repressed) and a lytic phase (in which genes to hours), determined by the antagonistic activity of ‘writer’ and
encoding molecules required for replication are active)67. The exis- ‘eraser’ enzyme pairs, such as histone acetyltransferases and histone
tence of such bi-stable switches in the context of immune-cell adap- deacetylases73. Some of these modifications, notably acetylation, act
tation has never been reported, to our knowledge. to physically relax the chromatin fiber and thus make the underly-
A second mechanism, the induction of long-lived mediators, ing DNA more accessible and prone to transcription, while others,
includes the expression of microRNAs, whose half-life can be such as histone methylation, act as platforms that might favor or
extremely long and result in long-term cellular effects. An interest- stabilize the recruitment of various enzymatic complexes that con-
ing example is provided by the microRNAs miR-221 and mi-R222, trol chromatin organization, transcription and DNA replication. In
which are involved in LPS tolerance68. These microRNAs negatively addition to the rapid turnover of histone modifications determined
control the expression of Brg1, a catalytic subunit of the BAF (SWI/ by erasers (such as deacetylases and demethylases), nucleosomes
SNF) chromatin-remodeling complex. Reduced expression of Brg1 themselves are rapidly turned over by dedicated ‘machines’ at active
prevents efficient re-induction of those inflammatory genes that are genomic regions74–76.
highly dependent on SWI/SNF-mediated nucleosome remodeling, The highly dynamic nature of nucleosomes, which represents the
such as the gene encoding IFN-β69. In keeping with this observation, basis for restoration of the basal state in a continuously changing
genes tolerized by the miR221–miR222–Brg1 axis include many micro-environment, is an obvious barrier to the long-term main-
genes encoding interferon-stimulated transcripts, as a consequence tenance of a stimulus-induced chromatin state, such as a gain in
of defective autocrine and paracrine IFN-β signaling and secondary acetylation at an activated promoter or enhancer. The gene-specific
activation of the STAT and IRF families of transcription factors68. mechanisms that overcome such high turnover rates have not yet
An extension of this mechanism is the evolution of the response been identified, although some genomic regions might display an
over time due to the reduced concentration of some mediators and intrinsically different ability to undergo erasure events; a slower era-
the increased concentration of others, as exemplified by coordinated sure rate might thus result in the short-term persistence of newly
and dynamic changes in transcription factors of the REL–NF-kB, deposited histone modifications20. Among known histone modifi-
AP-1, STAT and IRF families over many hours after initial stimula- cations, the only one shown to be associated with a maintenance
tion. For example, interferons induce the expression of STAT1 and mechanism similar to that of DNA methylation is the trimethyl-
STAT2, which can potentiate responses at re-stimulation. An inter- ation of histone H3 at Lys27 (H3K27me3), a repressive modifica-
esting category of potential mediators of persistent adaptation is tion77. Conversely, there is currently no mechanistic support for the
rare proteins with exceptionally long lifespans, although their pos- notion that in mammalian cells, modifications such as acetylation
sible effect on immune responses is largely unknown70. and activation-associated methylations can be propagated across
A third mechanism is a stable and self-propagating molecular DNA synthesis or can be self-sustained for a long period of time.
change, typically a change in DNA methylation that can be trans- In the absence of such a mechanism, or until it is discovered, the
mitted across subsequent DNA syntheses because of coupling of long-term persistence of a stimulus-induced activation-associated
the DNA-methylation machinery with the replication fork. In the histone modification, such as trimethylation of histone H3 at Lys4
context of typical memory responses in lymphocytes, changes in (H3K4me3) in response to stimulation with β-glucans78, might be
DNA methylation provide a critical contribution. Specifically, the accounted for by two mechanisms: the maintenance of a transcrip-
proliferation of antigen-stimulated naive lymphocytes provides an tion factor–dependent regulatory circuit that self-sustains after
opportunity for the passive loss of repressive methyl groups at CpG stimulus termination and continues to promote the deposition of
dinucleotides, if the modification on the parental DNA strand is not active histone modifications at target genes, or incomplete removal
copied onto the daughter strand. This is the mechanism by which of the stimulus.
genes encoding critical effector cytokines undergo enhanced acti-
vation in memory cells. Loci such as those encoding IL-4, IL-13 or Sustained adaptation of the innate immune system
IFN-γ are methylated in naive cells and undergo passive demeth- Probably the best-known case of sustained adaptation in the
ylation after activation, which thus enables stronger and faster immune system is trained immunity: the short- to medium-term
responses at re-stimulation1,2. Changes in DNA methylation may (weeks to months) persistence of a state of enhanced responsiveness
also contribute to the persistence of adaptation, as shown in T cell to microbial challenges of innate immune cells previously exposed
exhaustion, in which de  novo deposition of DNA methylation is to an initial stimulation4. The existence of such a hyper-responsive
required for the maintenance of T  cell hypo-responsiveness71. We state of the innate immune system is supported by a plethora of epi-
are tempted to speculate that dilution of DNA methylation at criti- demiological and experimental evidence that has been discussed in
cal regulatory elements after the proliferation of progenitor cells detail elsewhere3. One mechanistic issue that has not received due
might contribute to persistent changes in differentiated cells during attention in this context is the role of the persistence of the trigger
emergency myelopoiesis72 and thus might account for the mainte- of a trained-immunity phenotype.
nance of the observed effects. Along the same lines, passive dilu- The two most extensively studied triggers of trained immu-
tion of DNA methylation might contribute to memory in the NK nity are β-glucans and mycobacteria, notably the bacillus
cell system, as NK cell memory responses are preceded by a phase Calmette-Guérin (BCG) used for vaccination against tuberculosis.
of clonal expansion8. Adaptive responses in proliferating cells (NK Chemically, β-glucans are polymers that share with cellulose their
cells and hematopoietic progenitor cells) and non-proliferating cells basic component, β-glucose, but differ from cellulose in the nature
might thus be critically different, because DNA demethylation dur- of the chemical bonds that link the β-glucose units79. β-glucans are
ing replication provides the opportunity for the efficient propaga- structural components of fungi (such as Candida albicans), yeast,
tion of a bona fide, long-lasting epigenetic change. edible mushrooms and some plants (mainly seaweed) and show

Nature Immunology | www.nature.com/natureimmunology


Review Article | FOCUS
https://doi.org/10.1038/s41590-019-0399-9 NATure Immunology

a Synergy b Antagonism

IFN-γ IFN-γ

Ac Ac Me3
STAT1 STAT1

LPS IL-4

STAT6
NF-κB

IFN-γ
IFN-γ LPS IL-4

Synergized locus Antagonized locus A Antagonized locus B

Fig. 4 | Chromatin control of signal integration in macrophages exposed to synergistic or antagonistic stimuli. a, Stimulation of macrophages with
IFN-γ or LPS alone (top and middle) triggers partial chromatin accessibility and limited transcriptional induction of a hypothetical locus of inflammatory
genes (blue). After combined stimulation (bottom), transcription factors induced by the two stimuli act together at these regulatory elements to increase
accessibility and promote the deposition of histone acetylation and of other features of permissive chromatin, which leads to potent gene expression of the
synergized locus. b, In addition to eliciting chromatin accessibility and transcriptional induction at target genes, IFN-γ and IL-4 (signals with antagonistic
biological outputs) actively promote de-activation of enhancers and promoters that control reciprocal gene targets. While the underlying molecular
mechanisms are not entirely clear (as discussed in the text), the net effect of this antagonism is that combined macrophage stimulation with IFN-γ plus
IL-4 leads to broad and selective impairment of both IFN-γ target genes and IL-4 target genes. Additional gene-specific activities can be observed in these
experimental settings100–102, including genes whose expression is not antagonized or is even synergized by the antagonistic stimulus, but are not depicted
here.

extreme chemical heterogeneity, from linear molecules to highly frequently detected in asymptomatic people and in patients with
branched molecules with a different number of subunits and a vari- chronic diseases88–91.
able percentage of branching80. The most relevant β-glucans for the Similar considerations could apply to mycobacteria. BCG vac-
induction of trained immunity are those with (1→3)-β-d-glycosidic cination induces T  cell–independent protection against unrelated
bonds in the linear polymer and side chains bound by (1→6)-β-d- pathogens, including fungi and parasites3. Because BCG is a live
glycosidic linkages, which are all recognized by the pattern-recogni- attenuated vaccine, its persistence in the host may represent an
tion receptor dectin-181,82. The immune system–stimulating effects important factor in the generation of sustained training of the innate
of β-glucans vary greatly on the basis of the number of units and the immune system92,93. Moreover, many of the lipids of mycobacteria,
branching patterns, but 11 units are in general sufficient to trigger complex structures critical for the various phases of their life cycle
macrophage activation83. and in many cases endowed with biological activity94, are extremely
Although they are chemically heterogeneous, such bioactive, difficult to digest; therefore, their persistence after the clearance of
immune system–stimulating polysaccharides share the prop- live bacteria might generate long-lasting priming effects.
erty of not being degraded in mammals, which lack β-glucanase
enzymes. In mammals, the only pathway available for their degra- Signal integration as driver of adaptive responses
dation is the uptake by macrophages, in which they are subjected The current knowledge of how cell activity is influenced by envi-
to a slow oxidative process84. Such slow oxidation explains the ronmental signals is built substantially on the effects of individual
retention of β-glucans within macrophages for extended periods stimuli on cell populations, as exemplified by refined models of
of time after internalization85. The lack of highly active, dedicated macrophage activation95. However, it is clear that the cellular and
degradation pathways explains the persistence of intravenously molecular effects of individual stimuli can be heavily influenced by
injected (1→3)-β-glucans in the liver and spleen of mice for more co-existing environmental cues, such as those that are acutely co-
than 1 month (refs. 86,87), although it is unclear whether (or for delivered to cells or that are part of the local milieu in which cells
how long) these persistent β-glucans retain their biological activ- are embedded96. We refer to this feature as ‘signal integration’, mean-
ity. The persistence of β-glucans in cells and tissues raises the pos- ing the ability of cells to integrate multiple and complex inputs into
sibility that one critical component of trained immunity, at least unique and specific outputs that underlie the multifaceted functions
in some contexts and/or in response to some inducers, is the per- of immune cells in homeostasis and disease (Fig. 3). As discussed
sistence of the trigger, which would create a long-lasting primed below, the possible outputs of signal integration range from neu-
state due to low-level, continuous triggering of immune cells. This trality, when two or more stimuli do not influence each other, to
condition of sustained exposure to an innate-immune-response reciprocal modulation, synergism or antagonism. Signal integration
trigger would be evocative of the persistent hyper-activation state in immune cells occurs at multiple levels, including signal-trans-
induced by super-low concentrations of LPS, such as those that are duction pathways and especially genomic cis-regulatory elements,

Nature Immunology | www.nature.com/natureimmunology


FOCUS | Review
| Article
NATure Immunology FOCUShttps://doi.org/10.1038/s41590-019-0399-9
Review Article
where multiple transcription factors regulated by lineage- and stim- Box 1 | Known unknowns of adaptation and memory in the
ulus-specific signals land in close proximity (Fig. 4). immune system
Combined exposure to suboptimal doses of functionally coher-
ent stimuli (for example, inflammatory stimuli such as TNF and
To what extent are persistent adaptive phenotypes driven by sus-
IFN-γ) often leads to the potentiation of cellular responses. Signal
tained stimulation rather than by bona fide transcriptional and/
integration in this context is key to tailoring the potency of immune
or epigenetic memory?
reactions to the context in which cells are activated. For example,
What are the discriminating properties of stimuli that induce
combined sensing of microbial and damage-associated signals, such
rapidly reversible adaptive responses versus those that induce
as ATP and many others, by innate immune cells leads to increased
sustained adaptive responses?
production of the cytokine precursor pro-IL-1β, inflammasome
What is the specific contribution of transcription factor–
activation and, eventually, increased secretion of mature IL-1β97.
mediated memory to the adapted state, relative to that of
Along the same lines, dendritic cells exposed to LPS together with
chromatin mark–mediated memory?
oxidized phospholipids from the host, which are released by dying
What are the mechanisms that link signaling, metabolic
cells, enter a state of hyper-activation with more secretion of IL-1β
and chromatin reprogramming in the establishment and
and powerful stimulation of adaptive immunity98. A paradigmatic
maintenance of the adapted state?
example of synergism in the innate immune system is the ‘priming’
effect of IFN-γ: its ability to increase the production of inflammatory
cytokines by macrophages exposed to other stimuli, such as Toll-like
receptor agonists55. Mechanistically, stimulation of human macro- sites in co-stimulated mouse macrophages102. Such data indicate
phages with IFN-γ increases the accessibility of regulatory elements that MAF transcription factors might contribute to the ability of
of inflammatory genes, via STAT1 and IRF1, and thus allows greater inflammatory stimuli to antagonize gene expression induced by
transcription of these genes in response to LPS55,99. Other inflam- immunomodulatory stimuli. The co-existence of inflammatory
matory cytokines, such as TNF and IFN-α, have also been shown signals and anti-inflammatory signals that antagonistically con-
to increase chromatin accessibility, transcription-factor occupancy trol macrophage activities is common in vivo, in contexts such as
and deposition of H3K4me3 at promoters of a specific set of inflam- co-infection with helminths (which activate type 2 immunity) and
matory genes, which leads to amplified transcriptional responses bacteria or viruses (which activate type 1 immune responses)107–109,
after stimulation with a suboptimal concentration of LPS65. cancer110 and tissue repair111. Furthermore, while the examples
Signal integration at the chromatin level also seems to govern reported here have focused mostly on macrophages, many other
antagonistic responses in cells exposed to stimuli with oppos- immune-cell and non–immune-cell types can be subjected to simi-
ing biological functions, such as pro-inflammatory cytokines and lar regulation. For example, commensal-specific skin T lymphocytes
anti-inflammatory cytokines. Genomic analyses of macrophages co-express RORγt and GATA-3, two transcription factors that gov-
co-exposed to antagonistic signals such as IFN-γ and IL-4 or LPS ern the antagonistic type 17 helper T cell–differentiation program
and IL-4 have revealed a selective transcriptional antagonism that and type 2 helper T cell–differentiation program, respectively112. A
leads to the defective induction of dozens of genes from the pro- hybrid chromatin landscape in these cells supports the concomi-
grams elicited by each stimulus alone100–102. Reciprocal repression tant transcription of genes such as Il17a, Il23r and Ccr6 (type 17)
occurs in the absence of detectable interference with proximal sig- as well as Il5, Il3 and Ccr8 (type 2), which allows plastic adaptations
naling but is linked to altered chromatin remodeling at regulatory of commensal-specific T cells between homeostatic conditions and
elements of antagonized genes100–103. Treatment with IL-4 elicits loss tissue-injury conditions112.
of histone acetylation and of chromatin accessibility at promoters
and enhancers of inflammatory genes, which are induced at lower Conclusions
levels after concomitant or subsequent stimulation with IFN-γ or Data obtained in recent years have contributed to appreciation for
LPS100,102. While this effect is almost entirely dependent on STAT6, the relevance of adaptive processes to immunological homeostasis
only a minority of the antagonized enhancers are bound by this and to the tuning of responses to stimuli. Long-term alterations
transcription factor100,102, which suggests that repression of inflam- in gene expression and associated functional outcomes in innate
matory genes may occur via indirect binding of STAT6 to DNA and/ immune cells exposed to particular stimuli have often been inter-
or through the recruitment of transcriptional repressors such as preted as evidence of memory of the adapted state. Here we propose
HDAC3, PPAR-γ and components of nuclear receptor–co-repressor a revision of the current ideas in the field, based on the concept that
complexes100,104,105. Notably, binding of STAT1, C/EBPα, C/EBPβ or bona fide memory in biological systems should possess two essential
the NF-κB subunit p65 in response to IFN-γ or LPS is antagonized properties: specificity of the response, and the presence of dedicated
by IL-4 at only small sets of enhancers100,102, which indicates that maintenance mechanisms that make memory a stable and potentially
direct interference with occupancy by inflammatory transcrip- irreversible state. In B cell and T cell memory, specificity is provided
tion factors is not the main mechanism of repression. In a recip- by antigen receptors, and maintenance of the memory state is linked
rocal fashion, IFN-γ has been reported to suppress IL-4-inducible to DNA hypomethylation of the promoters and enhancers of genes
genes via interference with chromatin remodeling101,102,106. In human encoding critical effector molecules, which maximizes their induc-
macrophages, 24 hours of exposure to IFN-γ elicits deposition of tion at re-stimulation. Passive DNA demethylation is generated by
the repressive chromatin mark H3K27me3 at regulatory elements the clonal expansion that occurs after naive lymphocytes encounter
of genes encoding molecules with immunomodulatory functions, antigen and is stable over decades. Conversely, the enhanced second-
such as MERTK, PPARG and RANK106. This effect is mediated by ary responses in primed (or trained) innate immune cells tend to
EZH2, the catalytic subunit of the polycomb repressive complex occur after exposure to unrelated stimuli, and the enhanced activity
PRC2, and prevents subsequent induction of those genes by IL-4 state reverts to the basal state in a (relatively) short time. We suggest
and glucocorticoids106. Prolonged (24-hour) stimulation of human that such responses are better described as adaptive phenotypes that
macrophages with IFN-γ disassembles the enhancers of genes persist because the underlying molecular processes are not efficiently
encoding molecules with reparative and immunosuppressive func- and rapidly reverted except if cells are exposed to a novel stimulus,
tions that were bound by transcription factors of the MAF family in as shown by the swift tissue-instructed transcriptional changes
unstimulated macrophages101, consistent with the observation that observed in macrophages in transfer experiments in  vivo28,29. The
enhancers antagonized by IL-4 show enrichment for MAFB binding persistence of the adapted state is in many cases an advantageous

Nature Immunology | www.nature.com/natureimmunology


Review Article | FOCUS
https://doi.org/10.1038/s41590-019-0399-9 NATure Immunology

and desirable outcome of the primary stimulation, as exemplified 18. Mancino, A. et al. A dual cis-regulatory code links IRF8 to constitutive and
by the reduced production of inflammatory and potentially noxious inducible gene expression in macrophages. Genes Dev. 29, 394–408 (2015).
19. Garber, M. et al. A high-throughput chromatin immunoprecipitation
cytokines by tolerized macrophages generated by sustained exposure approach reveals principles of dynamic gene regulation in mammals. Mol.
to large amounts of activating stimuli. In other cases, persistence of Cell 47, 810–822 (2012).
the adaptive phenotype might anticipate future likely events, such 20. Ostuni, R. et al. Latent enhancers activated by stimulation in differentiated
as re-infection with similar microbes, to which the ‘trained’ innate cells. Cell 152, 157–171 (2013).
21. Kaikkonen, M. U. et al. Remodeling of the enhancer landscape during
immune cells would respond faster and more efficiently.
macrophage activation is coupled to enhancer transcription. Mol. Cell 51,
Within the conceptual framework outlined above, many mecha- 310–325 (2013).
nistic details remain to be addressed. The understanding of how 22. Okabe, Y. & Medzhitov, R. Tissue-specific signals control reversible
immune cells integrate complex environmental signals into spe- program of localization and functional polarization of macrophages. Cell
cific functional states is incomplete, and understanding of how 157, 832–844 (2014).
23. Gautier, E. L. et al. Gata6 regulates aspartoacylase expression in resident
some inputs generate sustained adaptations is even less complete peritoneal macrophages and controls their survival. J. Exp. Med. 211,
(Box  1). A critical issue that is still to be solved at the molecular 1525–1531 (2014).
level is the role of sustained stimulation relative to that of bona fide 24. Rosas, M. et al. The transcription factor Gata6 links tissue macrophage
transcriptional or epigenetic memory mechanisms in the genera- phenotype and proliferative renewal. Science 344, 645–648 (2014).
tion of persistent adaptive phenotypes. It remains possible that the 25. Haldar, M. et al. Heme-mediated SPI-C induction promotes monocyte
differentiation into iron-recycling macrophages. Cell 156, 1223–1234 (2014).
persistence of some stimuli, possibly because of the absence of dedi- 26. Scott, C. L. et al. The transcription factor ZEB2 is required to maintain the
cated degradation systems, may contribute to sustained phenotypes tissue-specific identities of macrophages. Immunity 49, 312–325.e315 (2018).
in the innate immune system, at least in some settings. From an 27. Mass, E. et al. Specification of tissue-resident macrophages during
evolutionary perspective, we are tempted to speculate that adapta- organogenesis. Science 353, aaf4238 (2016).
tions to constant exposure to signals such as β-glucans and other 28. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped
by the local microenvironment. Cell 159, 1312–1326 (2014).
bioactive polysaccharides that are widely present in food, parasites 29. Gosselin, D. et al. Environment drives selection and function of enhancers
or commensals113 may have contributed to the optimization of controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).
immune responses through low-level priming of innate immune 30. Deczkowska, A. et al. Mef2C restrains microglial inflammatory response
cells. Delineating the rules of signal integration will surely lead to and is lost in brain ageing in an IFN-I-dependent manner. Nat. Commun. 8,
better understanding of immunological adaptations in vivo, when 717 (2017).
31. Matcovitch-Natan, O. et al. Microglia development follows a stepwise
cells are concomitantly or sequentially exposed to a changing envi- program to regulate brain homeostasis. Science 353, aad8670 (2016).
ronment milieu. Such knowledge may in turn translate into novel 32. Saijo, K. et al. A Nurr1/CoREST pathway in microglia and astrocytes
strategies for modulating immune responses in conditions such as protects dopaminergic neurons from inflammation-induced death. Cell 137,
chronic infection, autoimmunity or cancer. 47–59 (2009).
33. Kayama, H. et al. Heme ameliorates dextran sodium sulfate-induced colitis
through providing intestinal macrophages with noninflammatory profiles.
Received: 8 March 2019; Accepted: 12 April 2019;
Proc. Natl Acad. Sci. USA 115, 8418–8423 (2018).
Published: xx xx xxxx 34. Casanova-Acebes, M. et al. Neutrophils instruct homeostatic and
pathological states in naive tissues. J. Exp. Med. 215, 2778–2795 (2018).
References 35. Ng, L. G., Ostuni, R. & Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev.
1. Wilson, C. B., Rowell, E. & Sekimata, M. Epigenetic control of T-helper-cell Immunol. 19, 255–265 (2019).
differentiation. Nat. Rev. Immunol. 9, 91–105 (2009). 36. Casanova-Acebes, M. et al. Rhythmic modulation of the hematopoietic
2. Monticelli, S. DNA (hydroxy)methylation in T helper lymphocytes. Trends niche through neutrophil clearance. Cell 153, 1025–1035 (2013).
Biochem. Sci. https://doi.org/10.1016/j.tibs.2019.01.009 (2019). 37. Puga, I. et al. B cell-helper neutrophils stimulate the diversification and
3. Netea, M. G. et al. Trained immunity: A program of innate immune production of immunoglobulin in the marginal zone of the spleen. Nat.
memory in health and disease. Science 352, aaf1098 (2016). Immunol. 13, 170–180 (2011).
4. Monticelli, S. & Natoli, G. Short-term memory of danger signals and 38. Mackay, L. K. & Kallies, A. Transcriptional regulation of tissue-resident
environmental stimuli in immune cells. Nat. Immunol. 14, 777–784 (2013). lymphocytes. Trends Immunol. 38, 94–103 (2017).
5. Seeley, J. J. & Ghosh, S. Molecular mechanisms of innate memory and 39. Ditadi, A., Sturgeon, C. M. & Keller, G. A view of human haematopoietic
tolerance to LPS. J. Leukoc. Biol. 101, 107–119 (2017). development from the Petri dish. Nat. Rev. Mol. Cell Biol. 18, 56–67 (2017).
6. Sun, J. C., Ugolini, S. & Vivier, E. Immunological memory within the innate 40. Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and
immune system. EMBO J. 33, 1295–1303 (2014). homeostasis. Immunity 44, 439–449 (2016).
7. O’Sullivan, T. E., Sun, J. C. & Lanier, L. L. Natural killer cell memory. 41. Gentek, R. et al. Hemogenic endothelial fate mapping reveals dual
Immunity 43, 634–645 (2015). developmental origin of mast cells. Immunity 48, 1160–1171.e1165 (2018).
8. Sun, J. C., Beilke, J. N. & Lanier, L. L. Adaptive immune features of natural 42. Li, Z. et al. Adult connective tissue-resident mast cells originate from late
killer cells. Nature 457, 557–561 (2009). erythro-myeloid progenitors. Immunity 49, 640–653.e645 (2018).
9. DuPage, M. & Bluestone, J. A. Harnessing the plasticity of CD4+ T cells to 43. Smith, F. L. & Baumgarth, N. B-1 cell responses to infections. Curr. Opin.
treat immune-mediated disease. Nat. Rev. Immunol. 16, 149–163 (2016). Immunol. 57, 23–31 (2019).
10. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 44. Gentek, R. et al. Epidermal γ δ T cells originate from yolk sac
1165–1169 (2016). hematopoiesis and clonally self-renew in the adult. J. Exp. Med. 215,
11. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability 2994–3005 (2018).
of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016). 45. Smith, N. L. et al. Developmental origin governs CD8 + T cell fate decisions
12. Matzinger, P. Friendly and dangerous signals: is the tissue in control? Nat. during infection. Cell 174, 117–130.e114 (2018).
Immunol. 8, 11–13 (2007). 46. Shemer, A. et al. Engrafted parenchymal brain macrophages differ from
13. Varol, C., Mildner, A. & Jung, S. Macrophages: development and tissue microglia in transcriptome, chromatin landscape and response to challenge.
specialization. Annu. Rev. Immunol. 33, 643–675 (2015). Nat. Commun. 9, 5206 (2018).
14. Monticelli, S. & Natoli, G. Transcriptional determination and functional 47. Cronk, J. C. et al. Peripherally derived macrophages can engraft the brain
specificity of myeloid cells: making sense of diversity. Nat. Rev. Immunol. independent of irradiation and maintain an identity distinct from
17, 595–607 (2017). microglia. J. Exp. Med. 215, 1627–1647 (2018).
15. Glass, C. K. & Natoli, G. Molecular control of activation and priming in 48. Zhu, Y. et al. Tissue-resident macrophages in pancreatic ductal
macrophages. Nat. Immunol. 17, 26–33 (2016). adenocarcinoma originate from embryonic hematopoiesis and promote
16. Ghisletti, S. et al. Identification and characterization of enhancers tumor progression. Immunity 47, 597 (2017).
controlling the inflammatory gene expression program in macrophages. 49. Bowman, R. L. et al. Macrophage ontogeny underlies differences in
Immunity 32, 317–328 (2010). tumor-specific education in brain malignancies. Cell Rep. 17,
17. Heinz, S. et al. Simple combinations of lineage-determining transcription 2445–2459 (2016).
factors prime cis-regulatory elements required for macrophage and B cell 50. Pope, S. D. & Medzhitov, R. Emerging Principles of Gene Expression
identities. Mol. Cell 38, 576–589 (2010). Programs and Their Regulation. Mol. Cell 71, 389–397 (2018).

Nature Immunology | www.nature.com/natureimmunology


FOCUS | Review
| Article
NATure Immunology FOCUShttps://doi.org/10.1038/s41590-019-0399-9
Review Article
51. Smale, S. T., Tarakhovsky, A. & Natoli, G. Chromatin contributions 83. Palma, A. S. et al. Ligands for the β-glucan receptor, Dectin-1, assigned
to the regulation of innate immunity. Annu. Rev. Immunol. 32, using “designer” microarrays of oligosaccharide probes (neoglycolipids)
489–511 (2014). generated from glucan polysaccharides. J. Biol. Chem. 281,
52. Foster, S. L., Hargreaves, D. C. & Medzhitov, R. Gene-specific control of 5771–5779 (2006).
inflammation by TLR-induced chromatin modifications. Nature 447, 84. Nono, I., Ohno, N., Masuda, A., Oikawa, S. & Yadomae, T. Oxidative
972–978 (2007). degradation of an antitumor (1→3)-β-D-glucan, grifolan. J. Pharmacobiodyn.
53. Kamada, R. et al. Interferon stimulation creates chromatin marks and 14, 9–19 (1991).
establishes transcriptional memory. Proc. Natl Acad. Sci. USA 115, 85. Ozment, T. R., Goldman, M. P., Kalbfleisch, J. H. & Williams, D. L. Soluble
E9162–E9171 (2018). glucan is internalized and trafficked to the Golgi apparatus in macrophages
54. Daniel, B. et al. The nuclear receptor PPARγ controls progressive via a clathrin-mediated, lipid raft-regulated mechanism. J. Pharmacol. Exp.
macrophage polarization as a ligand-insensitive epigenomic ratchet of Ther. 342, 808–815 (2012).
transcriptional memory. Immunity 49, 615–626.e616 (2018). 86. Miura, T., Ohno, N., Miura, N. N., Shimada, S. & Yadomae, T. Inactivation
55. Qiao, Y. et al. Synergistic activation of inflammatory cytokine genes by of a particle β-glucan by proteins in plasma and serum. Biol. Pharm. Bull.
interferon-γ-induced chromatin remodeling and toll-like receptor signaling. 20, 1103–1107 (1997).
Immunity 39, 454–469 (2013). 87. Miura, T. et al. Inactivation of (1→3)-β-D-glucan in mice. Biol. Pharm.
56. Cheng, S. C. et al. mTOR- and HIF-1α-mediated aerobic glycolysis as Bull. 18, 1797–1801 (1995).
metabolic basis for trained immunity. Science 345, 1250684 (2014). 88. Chen, K. et al. Super-low dose endotoxin pre-conditioning exacerbates
57. Saeed, S. et al. Epigenetic programming of monocyte-to-macrophage sepsis mortality. EBioMedicine 2, 324–333 (2015).
differentiation and trained innate immunity. Science 345, 1251086 (2014). 89. Lira, F. S. et al. Endotoxin levels correlate positively with a sedentary
58. Wendeln, A. C. et al. Innate immune memory in the brain shapes lifestyle and negatively with highly trained subjects. Lipids Health Dis. 9,
neurological disease hallmarks. Nature 556, 332–338 (2018). 82 (2010).
59. Naik, S. et al. Inflammatory memory sensitizes skin epithelial stem cells to 90. Szeto, C. C. et al. Endotoxemia is related to systemic inflammation and
tissue damage. Nature 550, 475–480 (2017). atherosclerosis in peritoneal dialysis patients. Clin. J. Am. Soc. Nephrol. 3,
60. Ordovas-Montanes, J. et al. Allergic inflammatory memory in human 431–436 (2008).
respiratory epithelial progenitor cells. Nature 560, 649–654 (2018). 91. Wiedermann, C. J. et al. Association of endotoxemia with carotid
61. McLane, L.M., Abdel-Hakeem, M.S. & Wherry, E.J. CD8 T cell exhaustion atherosclerosis and cardiovascular disease: prospective results from the
during chronic viral infection and cancer. Annu. Rev. Immunol. 37, Bruneck Study. J. Am. Coll. Cardiol. 34, 1975–1981 (1999).
457–495 (2019). 92. Olsen, A. W., Brandt, L., Agger, E. M., van Pinxteren, L. A. & Andersen, P.
62. Hotchkiss, R. S., Monneret, G. & Payen, D. Sepsis-induced The influence of remaining live BCG organisms in vaccinated mice on
immunosuppression: from cellular dysfunctions to immunotherapy. the maintenance of immunity to tuberculosis. Scand. J. Immunol. 60,
Nat. Rev. Immunol. 13, 862–874 (2013). 273–277 (2004).
63. Chen, J. & Ivashkiv, L. B. IFN-γ abrogates endotoxin tolerance by 93. Kaveh, D. A., Garcia-Pelayo, M. C. & Hogarth, P. J. Persistent BCG bacilli
facilitating Toll-like receptor-induced chromatin remodeling. Proc. Natl perpetuate CD4 T effector memory and optimal protection against
Acad. Sci. USA 107, 19438–19443 (2010). tuberculosis. Vaccine 32, 6911–6918 (2014).
64. Novakovic, B. et al. β-Glucan reverses the epigenetic state of LPS-induced 94. Queiroz, A. & Riley, L. W. Bacterial immunostat: Mycobacterium
immunological tolerance. Cell 167, 1354–1368.e1314 (2016). tuberculosis lipids and their role in the host immune response. Rev. Soc.
65. Park, S. H. et al. Type I interferons and the cytokine TNF cooperatively Bras. Med. Trop. 50, 9–18 (2017).
reprogram the macrophage epigenome to promote inflammatory activation. 95. Xue, J. et al. Transcriptome-based network analysis reveals a spectrum
Nat. Immunol. 18, 1104–1116 (2017). model of human macrophage activation. Immunity 40, 274–288 (2014).
66. Burrill, D. R. & Silver, P. A. Making cellular memories. Cell 140, 96. Murray, P. J. et al. Macrophage activation and polarization: nomenclature
13–18 (2010). and experimental guidelines. Immunity 41, 14–20 (2014).
67. Ptashne, M. Regulation of transcription: from lambda to eukaryotes. Trends 97. Schroder, K. & Tschopp, J. The inflammasomes. Cell 140, 821–832 (2010).
Biochem. Sci. 30, 275–279 (2005). 98. Zanoni, I. et al. An endogenous caspase-11 ligand elicits interleukin-1
68. Seeley, J. J. et al. Induction of innate immune memory via microRNA release from living dendritic cells. Science 352, 1232–1236 (2016).
targeting of chromatin remodelling factors. Nature 559, 114–119 (2018). 99. Saccani, S., Pantano, S. & Natoli, G. Two waves of nuclear factor kB
69. Ramirez-Carrozzi, V. R. et al. A unifying model for the selective regulation recruitment to target promoters. J. Exp. Med. 193, 1351–1359 (2001).
of inducible transcription by CpG islands and nucleosome remodeling. Cell 100. Czimmerer, Z. et al. The transcription factor STAT6 mediates direct
138, 114–128 (2009). repression of inflammatory enhancers and limits activation of alternatively
70. Toyama, B. H. et al. Identification of long-lived proteins reveals exceptional polarized macrophages. Immunity 48, 75–90.e76 (2018).
stability of essential cellular structures. Cell 154, 971–982 (2013). 101. Kang, K. et al. Interferon-γ represses m2 gene expression in human
71. Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 macrophages by disassembling enhancers bound by the transcription factor
blockade-mediated T cell rejuvenation. Cell 170, 142–157.e119 (2017). MAF. Immunity 47, 235–250.e234 (2017).
72. Mitroulis, I. et al. Modulation of myelopoiesis progenitors is an integral 102. Piccolo, V. et al. Opposing macrophage polarization programs show
component of trained immunity. Cell 172, 147–161.e112 (2018). extensive epigenomic and transcriptional cross-talk. Nat. Immunol. 18,
73. Kouzarides, T. Chromatin modifications and their function. Cell 128, 530–540 (2017).
693–705 (2007). 103. Schleicher, U. et al. TNF-mediated restriction of arginase 1 expression in
74. Deal, R. B., Henikoff, J. G. & Henikoff, S. Genome-wide kinetics of myeloid cells triggers type 2 NO synthase activity at the site of infection.
nucleosome turnover determined by metabolic labeling of histones. Science Cell Rep. 15, 1062–1075 (2016).
328, 1161–1164 (2010). 104. Nelson, V. L. et al. PPARγ is a nexus controlling alternative activation of
75. Dion, M. F. et al. Dynamics of replication-independent histone turnover in macrophages via glutamine metabolism. Genes Dev. 32, 1035–1044 (2018).
budding yeast. Science 315, 1405–1408 (2007). 105. Glass, C. K. & Saijo, K. Nuclear receptor transrepression pathways that
76. Kraushaar, D. C. et al. Genome-wide incorporation dynamics reveal regulate inflammation in macrophages and T cells. Nat. Rev. Immunol. 10,
distinct categories of turnover for the histone variant H3.3. Genome Biol. 365–376 (2010).
14, R121 (2013). 106. Qiao, Y., Kang, K., Giannopoulou, E., Fang, C. & Ivashkiv, L. B. IFN-γ
77. Margueron, R. et al. Role of the polycomb protein EED in the propagation induces histone 3 lysine 27 trimethylation in a small subset of promoters to
of repressive histone marks. Nature 461, 762–767 (2009). stably silence gene expression in human macrophages. Cell Rep. 16,
78. Quintin, J. et al. Candida albicans infection affords protection against 3121–3129 (2016).
reinfection via functional reprogramming of monocytes. Cell Host Microbe 107. Ezenwa, V. O. & Jolles, A. E. Epidemiology. Opposite effects of anthelmintic
12, 223–232 (2012). treatment on microbial infection at individual versus population scales.
79. Stone, B.A. Chemistry of β-glucans. in Chemistry, Biochemistry, and Biology Science 347, 175–177 (2015).
of 1→3-β-Glucans and Related Polysaccharides (eds. Bacic, A. et al.) Ch. 2.1 108. Osborne, L. C. et al. Coinfection. Virus-helminth coinfection reveals a
(Academic Press, 2009). microbiota-independent mechanism of immunomodulation. Science 345,
80. Williams, D. L. Overview of (1→3)-β-D-glucan immunobiology. Mediators 578–582 (2014).
Inflamm. 6, 247–250 (1997). 109. Rückerl, D. et al. Macrophage origin limits functional plasticity in
81. Ariizumi, K. et al. Identification of a novel, dendritic cell-associated helminth-bacterial co-infection. PLoS Pathog. 13, e1006233 (2017).
molecule, dectin-1, by subtractive cDNA cloning. J. Biol. Chem. 275, 110. Kratochvill, F. et al. TNF counterbalances the emergence of M2 tumor
20157–20167 (2000). macrophages. Cell Rep. 12, 1902–1914 (2015).
82. Brown, G. D. & Gordon, S. Immune recognition. A new receptor for 111. Bosurgi, L. et al. Macrophage function in tissue repair and remodeling
β-glucans. Nature 413, 36–37 (2001). requires IL-4 or IL-13 with apoptotic cells. Science 356, 1072–1076 (2017).

Nature Immunology | www.nature.com/natureimmunology


Review Article | FOCUS
https://doi.org/10.1038/s41590-019-0399-9 NATure Immunology
112. Harrison, O.J. et al. Commensal-specific T cell plasticity promotes rapid Ministry of Health (GR-2016-02362156), the Associazione Italiana per la Ricerca sul
tissue adaptation to injury. Science 363, eaat6280 (2019). Cancro (AIRC MFAG, 20247), the Cariplo Foundation (2015–0990) and the European
113. Blander, J. M., Longman, R. S., Iliev, I. D., Sonnenberg, G. F. & Artis, D. Union (Infect-ERA 126).
Regulation of inflammation by microbiota interactions with the host.
Nat. Immunol. 18, 851–860 (2017). Competing interests
The authors declare no competing interests.
Acknowledgements
We are grateful to members of the G.N. and R.O. laboratories for scientific discussions
and to S. Monticelli for critical reading of the manuscript. Research on related topics in Additional information
the G.N. laboratory is supported by the European Research Council (ERC Advanced Reprints and permissions information is available at www.nature.com/reprints.
Grant 692789, MEDICI), the European Commission (Consortium grant SYSCYD), the Correspondence should be addressed to G.N. or R.O.
Cariplo Foundation and the Italian Ministry of University and Research (MIUR, grant
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
METRIC (METabolic Regulation of Inflammatory Cells). Research in the R.O. laboratory
published maps and institutional affiliations.
is supported by grants from the European Research Council (ERC Starting Grant
759532, X-TAM), the Italian Telethon Foundation (SR-Tiget grant award F04), the Italian © Springer Nature America, Inc. 2019

Nature Immunology | www.nature.com/natureimmunology

You might also like