Soares 2017 PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

REVIEWS

Disease tolerance and immunity


in host protection against infection
Miguel P. Soares, Luis Teixeira and Luis F. Moita
Abstract | The immune system probably evolved to limit the negative effects exerted by
pathogens on host homeostasis. This defence strategy relies on the concerted action of innate
and adaptive components of the immune system, which sense and target pathogens for
containment, destruction or expulsion. Resistance to infection refers to these immune functions,
which reduce the pathogen load of an infected host as the means to preserve homeostasis.
Immune-driven resistance to infection is coupled to an additional, and arguably as important,
defence strategy that limits the extent of dysfunction imposed on host parenchymal tissues
during infection, without exerting a direct negative effect on pathogens. This defence strategy,
known as disease tolerance, relies on tissue damage control mechanisms that prevent the
deleterious effects of pathogens and that uncouples immune-driven resistance mechanisms from
immunopathology and disease. In this Review, we provide a unifying view of resistance and
disease tolerance in the framework of immunity to infection.

Homeostasis The pathological outcome of infection is mainly deter- tolerance is fully operational in animals, including
Maintenance of a stable mined by the degree of metabolic dysfunction and dam- flies9,10, rodents11,12 and humans13, in which it preserves
physiological ‘internal’ state in age inflicted on the host’s parenchymal tissues1,2. Clinical host homeostasis in response to viral14,15, bacterial15–18,
multicellular organisms via signs and symptoms of infectious diseases emerge as host fungal19 and protozoan11,13,20,21 infections.
feedback mechanisms that
homeostasis becomes compromised because of tissue dys- In contrast to resistance to infection, disease tol-
allow physiological functions to
proceed despite variations in function and damage1–3. Current understanding of this erance does not exert a direct negative effect on these
the ‘external’ environment. pathological process is limited, presumably impairing pathogens6 (BOX 1). Disease tolerance is now widely
the ability to treat some infectious diseases that remain studied in experimental models of infection, in which
associated with high human morbidity and mortality 4. the relationship between host health and pathogen load
Immunity provides general protection against dis- can be established at an individual level (BOX 2). In this
ease and particularly against infectious diseases4. This is Review, we discuss the cellular and molecular mech­
achieved by virtue of immune-driven resistance mech­ anisms establishing disease tolerance to infection, and
an­isms that expel, contain or kill pathogens as the means we explore the effects exerted by bona fide immunity
to preserve host homeostasis. Therapeutic approaches on those mechanisms. We also highlight how symbiotic
based on the induction of immune-driven resistance interactions with microorganisms and their regulation
mechanisms, such as vaccination, have proven highly by specific components of innate and adaptive immunity
protective against a broad range of infectious diseases4. affect disease tolerance to infection22. Finally, we sug-
The same is true for the use of antimicrobial agents, gest that the mechanisms underlying disease tolerance
such as antibiotics, which functionally mimic resistance to infection can be therapeutically targeted.
mechanisms by containing or killing pathogens4. The
overwhelming therapeutic success of these approaches Tissue damage control
has probably contributed to the perception that resist- The mechanisms underlying disease tolerance remain
ance mechanisms are the only relevant defence strategy poorly understood but seem to revolve around several
Instituto Gulbenkian de
against infectious diseases. This idea has been challenged evolutionarily conserved stress and damage responses
Ciência, Rua da Quinta
Grande, 6, 2780–156 over the past years by the (re)discovery of disease tol- that confer tissue damage control in the infected host1–3
Oeiras, Portugal. erance5,6. This evolutionarily conserved host defence (FIG. 1). These stress and damage responses sustain the
Correspondence to M.P.S. strategy was originally revealed in plants through the functional outputs of host parenchymal cells under
mpsoares@igc.gulbenkian.pt recognition that variation in disease severity can occur different forms of stress and damage imposed either
doi:10.1038/nri.2016.136 at a population level without a direct correlation to directly by pathogens, that is, virulence, or indirectly
Published online 3 Jan 2017 pathogen load7,8. However, it is now clear that disease by host immune-driven resistance mechanisms, that is

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Immunopathology immunopathology1,3. Stress and damage responses pro- Stress responses in tissue damage control
Refers to a breakdown of vide metabolic adaptation and repair damage to cellular Stress responses are triggered through the engagement
homeostasis in which immunity metabolites, macromolecules and/or organelles, as the of specific sensors that continuously monitor different
functions as the main cause of means to preserve core cellular, tissue and organ func- physiological parameters under homeostatic regula-
disease.
tions required to preserve or restore host homeostasis. tion such as temperature, O2, pH, osmolarity, glucose
Stress However, this often has detrimental effects on accessory and ATP1–3. When these parameters change beyond a
Any variations in the ‘external’ cell functions, and as such stress and damage responses certain threshold, ‘stress sensors’ set off signal transduc-
environment that disrupt the must be tightly regulated1,3. When these responses fail tion pathways that alert cells of a possible disruption to
maintenance of a stable
to sustain the functional outputs of parenchymal cells, homeostasis1–3. The ensuing stress responses provide
physiological environment in
which biological processes are
tissues or organs, the default programme becomes metabolic adaptation in host cells, which confers tissue
allowed to proceed. programmed cell death (FIG. 1). This is coupled to the damage control and contributes to the establishment
induction of cellular and tissue regenerative responses of disease tolerance to infection1–3 (FIGS  1,2). Signal
that have the capacity to restore the functional output of transduction pathways triggered upon recognition of
damaged parenchymal tissues or organs3,23. As discussed pathogen-associated molecular patterns (PAMPs) or
in further detail below, different types of infection impose damage-associated molecular patterns (DAMPs) by pat-
distinct forms of stress and damage, which suggests that tern recognition receptors (PRRs) can also contribute to
tissue damage control mechanisms might function in a tissue damage control and to the establishment of disease
somewhat pathogen class-specific manner that reflects tolerance to infection25, which has been reviewed else-
these differences to effectively establish disease tolerance where24,26 and is not discussed in this Review. Instead,
against diverse types of infection6. Although there are we focus on several bona fide stress responses as tis-
obvious parallels between the protective effect exerted sue damage control mechanisms that contribute to the
by tissue damage control mechanisms in the context of ­establishment of disease tolerance to infection1–3.
infectious and non-infectious diseases1,6,24, we focus our
discussion on infectious diseases. Oxidative stress. The oxidative stress response controlled
by the transcription factor nuclear factor erythroid 2‑
related factor 2 (NRF2)27,28 (FIG. 2) is important for the
establishment of disease tolerance to systemic infec-
Box 1 | Tolerance and disease tolerance tions such as malaria 20,29. Briefly, the blood stage
of Plasmodium spp. infection, the causative agent of
There is a growing interest in understanding disease tolerance, both at a mechanistic malaria, is associated with haemolysis and hence with
level and its potential therapeutic application. However, the concept of disease
generation of extracellular haemoglobin20,21,29,30. Upon
tolerance and its associated terminology can easily be misinterpreted, probably
because tolerance is broadly used to define some core properties of the immune system. oxidation, extracellular haemoglobin releases its pros-
Immunological tolerance is an active process through which specific antigens become thetic haem groups, which can function as catalysts in
non-immunogenic — that is, fail to trigger adaptive immunity in a given individual — on the production of reactive oxygen species (ROS) and
the basis of immunoregulatory mechanisms that eliminate or suppress the activation reactive nitrogen species (RNS). This can lead to oxi-
and proliferation of antigen-specific B and T cells123. Immunological tolerance can dative stress and cellular damage of host parenchymal
explain immune discrimination between self and non-self, which is a concept deeply tissues12,13,16,31, which in turn drive the pathogenesis
rooted in the understanding of immunity. Although some of the mechanisms regulating of severe forms of malaria30. Genetic polymorphisms
immunological tolerance and disease tolerance are functionally related, these two selected through human evolution on the basis of their
phenomena are clearly distinct. protective effect against malaria, such as sickle muta-
Tolerance is also used to describe how innate immune cells, particularly macrophages,
tions in the β-chain of haemoglobin, function via a
modulate responses to bacterial lipopolysaccharide (LPS). The cellular and molecular
bases for LPS tolerance have been extensively studied and relate primarily to the NRF2‑dependent mechanism that counteracts the
induction of epigenetic modifications in enhancers, which modulate gene transcription pathogenic effects of labile haem and establish disease
in response to Toll-like receptor 4 (TLR4) signalling124. This is not specific to TLR4, as it tolerance to the blood stage of Plasmodium spp. infec-
can occur downstream of other pattern recognition receptors (PRRs)125 and other tion20,30,32. Activation of NRF2 is also protective against
sensors18. In some instances this response — which is referred to as ‘trained immunity’ polymicrobial33 and Staphylococcus aureus 34 infections,
(REF. 126) — has been functionally linked to the induction of disease tolerance18. but whether this is due to the establishment of disease
Tolerance also refers to a related phenomenon in which exposure to a sub-lethal dose tolerance is unclear. However, the following two obser-
of a given agonist renders cells, tissues, organs or organisms tolerant to a subsequent vations suggest that disease tolerance could indeed have
higher dosage of the same substance that would otherwise be deleterious. This a role: NRF2 polarizes macrophage responses towards
adaptive response — which is also referred to as hormesis or adaptation — relies on
the promotion of tissue damage control35,36, and NRF2
the activation of several evolutionarily conserved genetic programmes that confer
protection against these agonists. Some of these genetic programmes overlap with induces mitochondrial biogenesis in parenchymal cells34,
those regulating stress and damage responses conferring tissue damage control and which probably also contributes to tissue damage control
disease tolerance to infections1,3. and disease tolerance to infections.
We use the term disease tolerance to refer explicitly to the same concept defined
more than a century ago in the plant literature5,6, in which disease tolerance defines a Hypoxia. Infections can be associated with a local or
defence mechanism that limits ‘damage to functions and structures’ (REF. 8) imposed systemic decrease in O2 supply to host cells, a condition
upon the host during an infection, without interfering with pathogen load5,6. Of note, known as hypoxia. This is sensed and countered by host
although disease tolerance by definition is not associated with modulation of pathogen cells via a stress response controlled by the hypoxia
load, it is possible that this response has an effect on pathogen physiology that is not inducible factor (HIF) family of transcription factors37
revealed by corresponding changes in pathogen load.
(FIG. 2). Whether HIF activation in parenchymal cells

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Experimental identification of mechanisms controlling disease tolerance


By definition, disease tolerance is revealed by variations of host disease severity without a direct correlation with pathogen
load. However, although stress and damage responses regulating tissue damage control and disease tolerance to infection
should not exert direct effects on pathogens, this is often difficult to show because of the inter-relationship between tissue
damage control and immune-driven host resistance mechanisms. For example, tissue damage control should enable
immune-driven resistance mechanisms to operate in a more robust and effective manner without the onset of
immunopathology. As such, tissue damage control can function indirectly to exert a negative effect on pathogens. Hence,
to dissociate disease tolerance from resistance to infection often requires that resistance is stably maintained, for example,
through the use of antimicrobial agents. Under such conditions, experimental modulation of host disease severity is likely
to involve the modulation of tissue damage control mechanisms that contribute to the establishment of disease tolerance.
As most studies investigating mechanisms of host protection against infection are not designed to detect the effect of
disease tolerance, it is often difficult to disentangle disease tolerance from resistance to infection.
When designing experimental approaches to identify mechanisms that regulate disease tolerance, one should consider
quantifying physiological parameters that assess host homeostasis, such as temperature, oxygen, pH, osmolarity or
glucose concentrations. However, these physiological parameters only reveal indirect disruptions of host homeostasis,
a limitation resolved by direct quantification of parameters that estimate the function of tissues and organs such as brain,
cardiovascular system, lungs, kidney and liver. When these parameters are plotted against host pathogen load, they can
reveal variations in disease tolerance. Of note, these parameters should also be quantified at steady state — that is,
before infection — so that host vigour — that is, health status before infection — can be defined. When these parameters
cannot be quantified, incidence of mortality can be used to reveal irreversible homeostasis breakdown13,16,17,20.
The pathogen load of the infected host should ideally be quantified throughout the course of infection, for example,
by sampling tissues in which pathogens accumulate, such as blood in malaria, lungs in pneumonia, liver in hepatitis, and
so on. However, a potential bias of this approach is that it does not account for changes in pathogen behaviour, which can
be reflected by variations in tissue tropism and accumulation at alternative sites. An alternative approach is to quantify
pathogen load in the whole body, as routinely done when infecting flies5,9,10,127. This is more challenging in other species
such as rodents, but it can be achieved using transgenic pathogens expressing reporter genes that can be quantified
throughout the course of an infection by whole-body imaging29.
Disease tolerance is mostly inferred from how a host reacts to infection, which is inferred from the phenotype, that is,
disease severity, produced by a given genotype under a certain pathogen load5,11,128,129. In this analysis of host response
to infection — also known as reaction norm analysis — each individual (genotype) is represented by a single health
parameter and a pathogen load value, which corresponds to the ratio of minimum health parameter to maximum
pathogen load5,11,128,129. However, these reaction norms do not provide information on how the relationship between
phenotype and pathogen load varies over time129. An alternative approach is to plot disease parameters versus pathogen
load over time130. This approach enables mapping of disease trajectories, which are revealed by the concomitant changes
in disease and pathogen load over time130,131. The data from these fitness curves can potentially be used to estimate
individual variations in disease tolerance130,131.

contributes to the establishment of disease tolerance to (NFAT5)43 (FIG. 2). This osmotic stress response acts in a
infection has not been established, but HIF activation cytoprotective manner in parenchymal cells44, confer-
in macrophages provides metabolic adaptation towards ring tissue damage control in the kidney during systemic
ATP production via anaerobic glycolytic metabolism polymicrobial infections45 and probably in the heart
and modulates macrophage polarization38,39 towards an ­during infection with coxsackievirus46 in mice (FIG. 2).
effector response that promotes tissue damage control
and disease tolerance to infection39,40, for example, in the Damage responses and tissue damage control
context of Helicobacter pylori infection in mice40. When cellular stress associated with a given infection
persists in strength and/or time, metabolic adaptation
Metabolic stress. Some stress sensors directly or indi- is no longer sufficient to preserve core cellular func-
rectly monitor variations in the relative concentration tions in the parenchymal tissues and organs that sustain
of essential metabolites such as ATP or glucose, respec- homeostasis. The ensuing damage inflicted to cellular
tively 1,3. These activate stress responses that adjust metabolites or macromolecules such as DNA, proteins
host cellular metabolism to the relative availability of and lipids, and eventually to organelles, activates spe-
the metabolites sensed (FIG. 2). In strong support of the cific damage responses that contribute to tissue damage
idea that metabolic stress responses can substantially control and to the establishment of disease tolerance to
contribute to the establishment of disease tolerance to infection1–3 (FIGS 1,2).
infection is the finding that regulation of host glucose
metabolism is essential to establish disease tolerance to Metabolite damage and extracellular release. Metabolite
viral and bacterial infections in mice41. damage refers to modifications of metabo­lites that com-
promise their original function47. Such modified metabo­
Osmotic stress. Systemic infections can be associated lites can have pathological effects via the impairment of
with osmotic stress42, which is sensed and counteracted host cellular functions and the generation of toxic prod-
by osmotic stress responses, such as the one regulated ucts that promote inflammation, cellular stress and dam-
via the activation of nuclear factor of activated T cells 5 age47. For example, sustained modifications of soluble

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Immunity Pathogen Immunity activity. Haem catabo­lism by the NRF2-regulated haem


oxygenase 1 (HO1) also contributes to prevent lipid
Stress
Stress ­peroxidation31 (FIG. 2).
responses
Proteotoxic damage. Proteotoxic damage responses aim
Damage to repair or eventually to degrade unfolded nascent or
Damage
responses
mature proteins as a means to preserve essential cellu-
lar functions53,54. These can function as tissue damage
Cellular control mechanisms and promote the establishment of
Cytotoxicity
regeneration
disease tolerance to bacterial infection53,55–58 (FIG. 2). Of
note, proteotoxic damage responses are also involved in
Tissue Tissue the regulation of immune-driven resistance mechanisms
damage control dysfunction against intracellular bacteria and viruses, some of which
in turn evolved to modulate these responses as a strategy
Disease Infectious to repress host resistance53,59.
tolerance disease

Figure 1 | Tissue damage control and disease tolerance. Immunity exerts a negative DNA damage. Infections are associated with host
effect on pathogens and triggers stress and damage in hostNature Reviewstissues,
parenchymal | Immunology
which DNA damage and consequently with the activation
can lead to cytotoxicity, tissue dysfunction and disease. Tissue damage control of DNA damage responses60 (FIG. 2). This probably occurs
mechanisms involve several stress and damage responses that function in a concerted via a mechanism involving the MRE11–RAD50–NBS1
manner to protect parenchymal cells and tissues from dysfunction or damage caused by complex, which activates ataxia-telangiectasia mutated
pathogens or by immune-driven host resistance mechanisms. Tissue damage control kinase (ATM) — a master regulator of the double-­
initially relies on stress responses that rewire metabolic pathways as the means to stranded DNA damage response61. Activation of ATM
preserve the core functional outputs of parenchymal cells1,3,151. If stress persists over time, confers tissue damage control and disease tolerance
damage to intracellular metabolites, macromolecules and cellular organelles develops1. to polymicrobial infections in mice17, which suggests
This activates damage responses that repair these different types of damage to preserve
that DNA damage responses contribute to maintain
core functional outputs of parenchymal cells. Programmed cell death is triggered if this
second layer of tissue damage control fails to preserve the functional outputs of both the genetic integrity and the functional outputs of
parenchymal cells. When this occurs, the last layer of tissue damage control becomes ­parenchymal cells during infection.
cellular regeneration and tissue repair. If this still fails to preserve or to restore the
functional outputs of parenchymal cells, the outcome is tissue dysfunction and damage, Damage to cellular organelles. Autophagy is an evolu-
which is revealed by the appearance of the clinical signs and symptoms of infectious tionarily conserved damage response that supports cell
diseases. Immune responses regulate stress, damage and regenerative responses that function under stress conditions that elicit damage to
underlie tissue damage control mechanisms and that contribute to the establishment of cellular organelles62 (FIG. 2). Autophagy regulates inflam-
disease tolerance to infection. matory responses and modulates disease tolerance to
infections caused by pathogens such as Sindbis virus63
metabolites that promote their phase transition into crys- and S. aureus 64 as well as to polymicrobial17 infections in
tals are sensed by PRRs, which can trigger inflammatory mice. Of note, autophagy can also function as a resist-
Ataxia-telangiectasia responses that are deleterious to the host 48. Thus, lim- ance mechanism against intracellular pathogens, so
mutated kinase iting the pathological effects of metabolite d ­ amage can called xenophagy 62.
(ATM kinase). A serine and ­promote disease tolerance to infection (FIG. 2).
threonine protein kinase that is
recruited and activated by
Cell death in tissue damage control
DNA double-strand breaks and Lipid damage. Lipid peroxidation can impair cellu- When stress and damage responses fail to preserve cel-
that has an important role in lar membrane function, which eventually leads to the lular functional outputs, the default response is, in most
the activation of DNA damage release of intracellular content 49,50 (FIG. 2). Moreover, cases, programmed cell death (FIG. 1). Different forms of
responses.
sensing of lipid peroxidation products by PRRs50, such stress and damage are associated with distinctive forms of
Necroptosis as Toll-like receptor 4 (TLR4), can exacerbate tissue programmed cell death and ensuing pathophysio­logi­cal
A specific form of programmed damage, as shown during pathogenesis of influenza consequences65. For example, failure to resolve oxidative
cell death mediated via a virus infection51. Expression of phospholipid hydro­ stress can lead to necroptosis66 and failure to repair lipid
genetically encoded mechanism peroxide glutathione peroxidase 4 (GPX4) has a central peroxidation induces ferroptosis67. Irreparable DNA dam-
involving receptor-interacting
serine/threonine-protein
role in inhibiting lipid peroxidation28,52. This salutary age results in the induction of different programmed cell
kinase 1 (RIPK1) and RIPK3 effect occurs via a mechanism that relies on glutathione death pathways, including apoptosis68, which is ­regulated
and the mixed lineage kinase supply from an NRF2‑regulated pathway involving by the caspase family of cysteine proteases.
domain-like (MLKL) solute carrier family 7 member 11 (SLC7A11; which Although programmed cell death can damage host
pseudokinase.
together with SLC3A2 encodes the cystine/glutamate parenchymal tissues and lead to organ dysfunction, this
Ferroptosis antiporter), glutamate–cysteine ligase catalytic sub­ relationship is not always straightforward because pro-
Genetically encoded form of unit  (GCLC),  ­g lutamate-­c ysteine ligase modifier grammed cell death is also part of a resistance mechanism
programmed cell death driven ­subunit (GCLM, which encodes the γ‑glutamylcysteine against intracellular pathogens69. However, the trade-off
by loss of activity of the lipid synthetase γGCS) and GSS (encoding the glutathione of this resistance mechanism can be particularly high,
repair enzyme glutathione
peroxidase 4 (GPX4) and by
synthetase)28,52. This suggests that activation of the depending on the relative capacity of different tissues to
the accumulation of lipid transcription factor NRF2 contributes to counter lipid withstand cell loss without compromising tissue function
hydroperoxides. peroxidation via a mechanism that supports GPX4 and homeostasis6. The pathophysiological relevance of

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Figure 2 | Relative contribution of stress and damage responses to the Toxins


establishment of disease tolerance to infection. Pathogen-derived toxins, such as Oxidative KEAP1 and NRF2
pore-forming toxins, can function as a major driving force in the pathogenesis of Hypoxic PHD2 and HIF
Stress
infectious disease by inducing stress and damage to host cells, which eventually leads Stress Metabolic AMPK, PI3K, responses
to programmed cell death152. Production of reactive oxygen species (ROS) and reactive AKT and FOXO
nitrogen species (RNS) by macrophages or polymorphonuclear cells is a common host Osmotic NLRP3, NLRC4,
immune resistance mechanism against various pathogens153,154, which can lead to AKAP13 and NFAT5
oxidative stress153,154. This is sensed by Kelch-like ECH-associated protein 1 (KEAP1)27,28 Metabolites NLRP3, P2X,
— which under homeostatic conditions constitutively promotes the ubiquitylation and CD39 and CD73
proteolytic degradation of the transcription factor nuclear factor erythroid 2‑related Lipids HO1 and GPX4 Damage
Damage
factor 2 (NRF2)27,28. Both ROS and RNS repress KEAP1 activity and enable NRF2 Proteins AMPK, HSF1 responses
activation, which triggers the expression of effector genes regulating oxidative stress and UPR
responses27,28. Hypoxia is counteracted by a stress response triggered by the O2 sensor DNA ATM and p53
prolyl hydroxylase domain protein 2 (PHD2)37, which constitutively represses the Organelles Autophagy
activity of the hypoxia inducible factor (HIF) family of transcription factors, under Tissue Tissue damage
normoxic conditions37. HIF activation can also occur under normoxic conditions37, dysfunction control
in response to pattern recognition receptor signalling, reduced intracellular iron
availability or ROS. Hence, HIF activation might confer metabolic adaptation to
different types of stress associated with infection37. Metabolic stress resulting from Infectious Disease
reduced intracellular ATP content is sensed and countered by a metabolic stress disease tolerance
response controlled by AMP-activated protein kinase (AMPK)155. Metabolic
stress resulting from a systemic reduction in glucose concentration is sensed indirectly Nature Reviews | Immunology
by insulin receptor156. When insulin levels drop, phosphoinositide 3‑kinase (PI3K) and this defence strategy is supported by the realisation that
AKT activation are reduced, promoting the activation of the forkhead box O (FOXO) intracellular pathogens evolved a range of mechanisms
family of transcription factors156. Activation of FOXO family members functions in a that can modulate host genetic programmes controlling
tissue-specific manner to promote or to repress disease tolerance to infection. programmed cell death, presumably as a strategy to
For example, FOXO activation in epithelial cells regulates the expression of genes
escape resistance to infection70.
encoding antimicrobial peptides157 and antioxidant enzymes158, which contribute to
maintain epithelial barrier integrity and as such are protective against infection in
Programmed cell death of infected cells is coupled
flies157,158 and mice159. Conversely, activation of FOXO1 and FOXO3α in muscle cells to phagocytosis by bystander macrophages or dendritic
promotes myofibre atrophy and muscle wasting associated with sepsis in mice160 and cells (DCs), which present pathogen-associated antigens
Mycobacterium marinum infection in flies127. Osmotic stress can be sensed by to CD4+ T helper (TH) cells or CD8+ cytotoxic T lym-
inflammasomes161,162, containing NLRP3 (NOD-, LRR- and pyrin domain-containing 3) phocytes (CTLs) and thereby elicit an antigen-­specific
or NLRC4 (NOD-, LRR- and CARD-containing 4)162 as well as by the A‑kinase anchor adaptive immune response that targets and kills the
protein 13 (AKAP13), which activates nuclear factor of activated T cells 5 (NFAT5)43, remaining reservoir of infected cells. Clearance of dam-
a transcription factor that provides metabolic adaptation to osmotic stress and confers aged and dying cells is associated with macrophage
tissue damage control during infection45. Metabolite damage, such as imposed by polarization towards the production of cytokines and
phase transition into crystals, functions in a pro-inflammatory manner as shown, for
lipid mediators that limit the extent of inflammatory
example, for uric acid-driven monosodium urate crystals163 or cholesterol crystals164.
This is sensed by NLRP3 inflammasomes, triggering a downstream signal transduction
reactions71. These include interleukin‑10 (IL‑10) and
pathway, which involves the apoptosis-associated speck-like protein containing a 15‑deoxy‑Δ‑­prostaglandin J2 (15d‑PGJ2), which induce
CARD (ASC), caspase 1 and interleukin‑18 (IL‑18)48. Extracellular release of the expression of HO1 (REFS 72,73). Haem catabolism by
metabolites, such as ATP, is sensed by the P2X purinergic receptor165, which signals via HO1 modulates macrophage responses towards inhibi-
inflammasomes161 to promote inflammation and tissue damage165. This is countered by tion of pro-­inflammatory gene expression74 and probably
the catabolism of extracellular ATP and ADP into AMP and adenosine by CD39 and exerts cytoprotective effects in bystander cells75. This pro-­
CD73, respectively165, which exert immunoregulatory effects, promoting the resolving response is also associated with the production
establishment of disease tolerance to Toxoplasma gondii166 or Helicobacter spp.167 of growth factors, such as platelet-derived growth factor
infections in mice. Presumably these effects are mediated via adenosine-driven (PDGF), and cytokines, including transforming growth
signalling through transmembrane adenosine G protein-coupled cell surface
factor‑β1 (TGFβ1), which can directly affect paren­
receptors110,168. Lipid peroxidation is counteracted by several damage responses49
including the induction of haem oxygenese 1 (HO1)31, which promotes the
chymal cells to promote tissue regeneration, regulating
establishment of disease tolerance to bloodstream infections16,20,21,29–31. Whether yet another layer of tissue damage control23.
inhibition of lipid peroxidation by the glutathione–glutathione peroxidase 4 (GPX4) Although some tissue damage control mechanisms
pathway also promotes disease tolerance to infection has not been established. function in a cell-autonomous manner, other mech-
Damage to proteins or proteotoxic damage is sensed by heat shock proteins, which anisms seem to rely on bystander innate and adap-
activate heat shock factor 1 (HSF1)54, and is sensed in the endoplasmic reticulum by the tive immune cells. As discussed in the section below,
78 kDa glucose-regulated protein (GRP78) chaperone and inositol-requiring protein 1 tissue-resident leukocytes probably have a predom-
(IRE1)53, which activate the unfolded protein response (UPR). HSF1 activation confers inant role in this non-cell autonomous mechanism of
a survival advantage against Enterococcus faecalis and Pseudomonas aeruginosa ­establishment of disease tolerance to infection.
infections in Caenorhabditis elegans55,56 and against Listeria monocytogenes infection in
mice57. The UPR prevents immunopathology associated with P. aeruginosa infection in
C. elegans58 and preserves homeostatic control of microbiota interactions with the host
Immune regulation of tissue damage control
gut epithelium53. The DNA damage responses controlled by Ataxia-telangiectasia In this section we discuss how tissue-resident macro­
mutated kinase (ATM)17 and by p53 (REF. 169) confer tissue damage control and phages, innate lymphoid cells (ILCs) and regulatory T
disease tolerance to pulmonary bacterial infections in mice. Damage to cellular (Treg) cells affect tissue damage control mechanisms and
organelles is counteracted by autophagy, which modulates disease tolerance to a thereby contribute in a non cell-autonomous manner to
variety of infections17,62–64. the establishment of disease tolerance to infection (FIG. 3).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Alarmins Macrophages. Tissue-resident macrophages express high IL‑18 and IL‑33, which polarize macrophages towards
Endogenous molecules levels of PRRs and various other receptors that sense tissue-healing regenerative responses and, as discussed
released from damaged cells PAMPs, DAMPs or alarmins released from damaged cells, in the previous section, can contribute to the establish-
and sensed by receptors of the including cytokines that alert cells to the disruption of ment of disease tolerance to infection (FIG. 3a). Signalling
immune system that alert for
tissue dysfunction or damage,
homeostasis76,77 (FIG. 3a). These cytokines include IL‑1α, via other sensors, such as the aryl hydrocarbon receptor
associated with disruption of (AHR) — a ligand-dependent transcription factor that
homeostasis. senses exo­genous environmental toxins and endogenous
a Parenchymal cells ligands — can also polarize macrophages towards the
establishment of disease tolerance to bacterial infections18.
Macrophage antimicrobial responses are associated
NO with the expression of genes that can promote tissue
CO
H2S damage control in parenchymal cells. These include
IL-1, inducible nitric oxide synthase (iNOS), which generates
PDGFR ILR IL-18
and nitric oxide (NO), HO1, which generates carbon mon-
IL-33 oxide (CO), and cystathionine β‑synthase (CBS), which
PDGF, TGFβ, generates hydrogen sulphide (H2S) (FIG. 3a). These gaso­
TNF, IL-6 and ILR
IL-10 transmitters78 diffuse across cellular membranes to sense
DAMP
the metabolic status of microorganisms79 while driving
Macrophage PRR metabolic adaptation in host parenchymal cells78,80, sup-
Stress L-Kynurenine porting tissue damage control and contributing to the
AHR
Sensor establishment of disease tolerance to infection13,29 (FIG. 3a).
PRR Sensor Cytokines produced during these responses have a dual
Dying infected cells role, for example, tumour necrosis factor (TNF) can trig-
Pathogen undergoing PCD ger programmed cell death and also activate pro-survival
(PAMP)
responses via activation of the nuclear factor‑κB (NF‑κB)
family of transcription factors81 (FIG. 3a).
b Parenchymal cells Phagocytosis is also associated with the produc-
tion of ROS by macrophages, which supports stem cell
division and differentiation towards tissue healing and

IL-22R EGFRs IL-33 Figure 3 | Immune regulation of tissue damage control.


a | Tissue-resident macrophages use pattern recognition
IL-1
receptors (PRRs), interleukin receptors (ILRs) and/or aryl
EGFs ST2 hydrocarbon receptors (AHRs) to sense environmental cues,
ILC2
(e.g. AREG)
including pathogens, dying infected cells and alarmins —
Stress
such as damage-associated molecular patterns (DAMPs),
interleukin‑1 (IL‑1), IL‑18, IL‑33 or l‑kynurenine generated
IL-22 by host parenchymal cells via tryptophan 2,3‑dioxygenase18
IL-1R
ILC3 — that alert cells to disruption of homeostasis. Signalling by
Stress these sensors polarizes macrophage responses to resolve
NCR inflammation and to assist parenchymal cells in restoring
c Parenchymal cells homeostasis18,170,171. This non cell-autonomous mechanism
supporting tissue damage control is mediated by the
production of gasotransmitters, including nitric oxide (NO),
carbon monoxide (CO) and probably hydrogen sulphide
(H2S), and by the secretion of cytokines, such as tumour
necrosis factor (TNF), IL‑6 and IL‑10, and growth factors,
EGFRs Stress including transforming growth factor‑β (TGFβ) and members
IL-18 of the platelet-derived growth factor (PDGF) family.
and b | Tissue-resident innate lymphoid cells (ILCs) can sense
IL-33 alarmins, such as IL‑33 and IL‑1, via the corresponding ILRs
EGFs Treg cell ILR
(e.g. AREG) or can sense cellular stress and damage via natural
cytotoxicity receptors (NCRs)85. These trigger the production
MHC TCR of cytokines such as IL‑22 and epidermal growth factors
class II (EGFs), including amphiregulin (AREG), which promote
ILR tissue damage control in parenchymal cells, such as
epithelial cells85,86,88. c | Tissue-resident regulatory T (Treg) cells
Macrophage sense alarmins, such as IL‑18 and IL‑33, which elicit the
Stress production of AREG91–93, affecting parenchymal cells to
promote tissue damage control91–93 and to establish
Sensor
PRR
disease tolerance to infection92. PCD, programmed cell
Pathogen death; PDGFR, PDGR receptor; EGFRs, EGF receptors;
(DAMP) TCR, T cell receptor.

Nature Reviews | Immunology


6 | ADVANCE ONLINE PUBLICATION www.nature.com/nri
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

regeneration82. This process involves repression of the the balance between TH17 and Treg cells96, and thereby
transcriptional programme activated by NRF2 (REF. 83), modu­lates immune-driven resistance and possibly dis-
revealing a tight integration of stress and damage ease tolerance to infection. Activation of other stress and
responses with subsequent tissue repair and regenera- damage responses in tissue-resident leukocytes probably
tion programmes, which restore host homeostasis23,82,84 exerts similar effects, but this is yet to be determined
(FIGS 1,2). How these apparently conflicting activities experimentally.
exerted by cytokines such as TNF or ROS are resolved
remains to be understood23. This suggests, nevertheless, Pathogen-specific tissue damage control
that antimicrobial macrophage responses have built‑in Multicellular organisms establish stable symbiotic inter-
feedback loops promoting tissue damage control. actions with a wide range of microorganisms, which
affects different aspects of their physiology 97 and also
Innate lymphoid cells. All three classes of ILCs, that is modulates resistance and disease tolerance to patho-
ILC1, ILC2 and ILC3, have a crucial role in sensing tissue gens22 (BOX 3). Infection by viruses, bacteria, fungi, pro-
dysfunction and damage, regulating tissue damage con- tozoan parasites or helminths imposes distinct forms
trol responses85, which contribute to the establishment of stress and damage on host parenchymal cells that
of disease tolerance to viral86 and helminth87 infections. are driven by intrinsic differences in virulence mech-
These cells are also operational during intestinal inflam- anisms and countervailing host immune-driven resist-
mation88. For example, ILC2s can sense alarmins, such ance mechanisms elicited. Thus, tissue damage control
as IL‑33, and promote tissue damage control in epithelia mechanisms probably function in a pathogen-specific
via a mechanism that involves the production of the epi- manner to confer disease tolerance to different types of
dermal growth factor (EGF)-like molecule amphiregu- infection, as originally shown in flies98 and thereafter
lin85,86,88 (FIG. 3b). Natural cytotoxicity receptors (NCRs) in mice6,99,100. In this section we discuss how immune-
expressed by ILC3s can sense ligands expressed by path- driven resistance to a specific pathogen class regulates
ogens as well as self ligands upregulated in response to tissue damage control mechanisms that contribute to the
cellular stress85, which leads to the production of IL‑22, establishment of disease tolerance to pathogens within
an IL‑10 family member that promotes epithelia repair the same or other classes.
and regeneration89 (FIG. 3b).
Intracellular pathogens. Host resistance to intra­cellular
Regulatory T cells. Natural loss of function mutations pathogens, such as viruses and certain bacteria, relies to
in the gene encoding the Treg cell lineage commitment a large extent on targeting infected cells for programmed
transcription factor forkhead box protein P3 (FOXP3) cell death by type 1 immunity-driven cytotoxic mechan­
are associated with development of severe immuno­ isms101 (FIG. 4a). Hence, tissue damage control mech­anisms
pathology in both mice and humans90. This suggests opposing type 1 immunopathology should not rely on
that Treg cells contribute to tissue damage control and cytoprotection of infected cells, as this would compro-
presumably therefore regulate the establishment of dis- mise resistance to intracellular pathogens. Instead, these
ease tolerance to infection. The physiological functions tissue damage control mechanisms are more likely to
of Treg cells have mostly been assigned to their capacity promote cell regeneration and tissue repair as a means
to restrain adaptive and to a lesser extent innate immune to compensate for loss of infected and bystander paren-
responses90. More recently, tissue-resident Treg cells were chymal cells, restoring tissue homeo­stasis15,92 (FIG. 4a). The
shown to also promote tissue damage control mechan­ mechanisms by which this occurs are not completely
isms91–93, which can contribute to the establishment of established but are probably tissue specific6 and involve
disease tolerance to influenza virus infection92. This pro- the production of EGF and PDGF family members as
tective effect involves sensing of alarmins, such as IL‑18 well as TGFβ, which promote stem cell proliferation
or IL‑33, which elicits the production and secretion of and/or differentiation into parenchymal cells and thereby
amphiregulin15,91,92 by tissue-resident Treg cells91–93 (FIG. 3c). restore tissue integrity and f­ unction23 (FIG. 4a).
Whether this protective effect functions via direct inter-
action of tissue-resident Treg cells with parenchymal cells Extracellular parasites. Host resistance to extracellular
or indirectly through immunoregulatory mechanisms metazoan parasites mainly depends on type 2 immu-
involving tissue-resident macrophages or ILCs has not nity 101,102. Some of these parasites are damaging to host
been established (FIG. 3c). parenchymal tissues, which presumably explains why
type 2 immunity encompasses tissue damage control
Stress and damage responses in immune cells. When mechanisms that contribute to the establishment of dis-
exposed to different forms of infection-associated stress, ease tolerance103 (FIG. 4b). Type 2 immunity targets patho­
tissue-resident macrophages, ILCs and Treg cells should gens primarily for containment or expulsion, probably
activate the same stress and damage responses that because of the inherent failure of most immune-driven
operate in parenchymal cells to promote tissue damage cytotoxic molecules to kill large metazoan parasites
Nutritional immunity control and disease tolerance to infection. For example, such as helminths (FIG. 4b). In addition, type 2 cytokines
An evolutionarily conserved NRF2 activation in macrophages35 or in T cells94 exerts promote macrophage polarization towards granuloma
resistance mechanism against
infection based on the host’s
immunoregulatory effects that promote tissue damage formation and thereby contain pathogens and deprive
ability to withhold nutrients, control. Furthermore, activation of HIF1α in macro­ them of essential nutrients. This resistance mechanism,
such as iron, from pathogens. phages controls a metabolic response39,95 that shifts known as nutritional immunity, is perhaps best illustrated

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

by the modulation of host iron metabolism as a strat- immunity can impose metabolic stress on host parenchy-
egy to deprive pathogens of this essential nutrient 104. mal cells, thereby activating stress responses that confer
Macrophage polarization by type  2 cytokines also meta­bolic adaption and possibly tissue damage control
deprives pathogens of access to other essential nutrients and disease tolerance to extracellular parasites (FIG. 4b).
such as arginine, which is depleted via the expression of
high levels of arginase in macrophages105. Arginine deple- Extracellular bacteria and fungi. Host resistance to
tion also prevents sustained T cell activation and thereby pathogenic extracellular bacteria and fungi relies to a
limits immunopathology 105. As a trade-off, nutritional large extent on TH17 immunity 101. Most of the immune

Box 3 | Microbiota and disease tolerance


There are numerous examples of symbiotic microorganisms that enhance resistance to infection in animals22. The
bacterial endosymbiont Wolbachia, for example, enhances resistance to Drosophila C virus in flies10,132 and to dengue,
chikungunya as well as Zika virus infections in mosquitoes133,134 (see the figure, part a). This protective effect has been
linked to priming of the insect innate immune system and possibly competition for resources supporting pathogen
replication135,136. Symbiotic interactions between bacteria and mice can also promote immune-driven resistance
mechanisms against viral137, bacterial138 or protozoan139,140 infections (see the figure, part a). This suggests that the
establishment of stable symbiotic interactions between microcellular and multicellular organisms is a widespread
recurrent trait that modulates host resistance against a range of pathogens.
Symbiotic bacteria can also modulate tissue damage control and disease tolerance to infection in multicellular
organisms10,22,141,142 (see the figure, part b). For example, Wolbachia enhance disease tolerance to flock house virus
infection in flies10 and against Plasmodium relictum infection in mosquitoes142. In mice, bacteria that colonize the gut
— such as the human symbiont Bacteroides fragilis — confer disease tolerance to Helicobacter hepaticus infection143,
whereas the gut symbiont Escherichia coli O21:H+ confers disease tolerance to Burkholderia thailadensis or Salmonella
enterica subsp. enterica serovar Typhimurium infections141 (see the figure, part b).
Although the mechanisms by which endosymbiotic bacteria promote disease tolerance to viral infections in insects
are not completely understood, some mechanistic insight is emerging in mice22. For example, the gut commensal E. coli
O21:H+ confers disease tolerance to systemic bacterial infections via a mechanism that involves the activation of the
NLRC4 (NOD-, LRR- and CARD-containing 4) inflammasome, which induces the production of interleukin‑18 (IL‑18)141.
This alarmin sustains the production of insulin-like growth factor 1 (IGF1), which activates the phosphoinositide
3‑kinase (PI3K)–AKT signal transduction pathway and represses the expression of the E3 ubiquitin ligases atrogin 1 (also
known as FBXO32) and MURF1 (also known as TRIM63) in muscle cells141. This prevents the development of muscle
atrophy and the loss of body weight, which are common pathological outcomes of infection. Components of the
microbiota can also modulate disease tolerance by altering the glycosylation profile of the gut epithelia lumen — as
shown for regulation of fucose expression under inflammatory conditions — which modulates microbiota composition,
metabolism and gene expression in a manner that promotes disease tolerance to enteric infection by Citrobacter
rodentium144. Modulation of host metabolism by bacterial components of the microbiota145 probably also modulates
tissue damage control mechanisms and disease tolerance to infection22. Whereas bacterial symbionts such as
Clostridium strains146 can promote the development of regulatory T (Treg) cells in mice, it is not clear whether this in turn
modulates disease tolerance to infections.
In the same way that pathogens can modulate host resistance mechanisms, some pathogens can also promote disease
tolerance as the means to support their own survival, proliferation and/or transmission. For example, S. Typhimurium
induces the activation of host RHO GTPases and
mitogen-activated protein kinases (MAPKs), which leads
to the activation of nuclear factor‑κB (NF‑κB) and to the a
expression of downstream pro-inflammatory genes in the Endosymbiotic bacteria Symbiotic bacteria
gut epithelium147. This activation enables S. Typhimurium e.g. Wolbachia i.e. microbiota
to overcome immune-driven host resistance imposed by
the resident gut microbiota147 and promotes invasion of Enhanced resistance to
gut epithelial cells and systemic infection148. During
bacterial invasion of the epithelium, S. Typhimurium Drosophila C virus, Influenza A virus, E. coli
dengue virus and and Plasmodium spp.
secrete proteases that target the NF‑κB family members chikungunya virus infections in mice
RELA and RELB and inhibit the transcription of infections in insects
downstream pro-inflammatory genes149. This
immunoregulatory effect limits tissue damage and
disease severity without interfering with host pathogen b
load149; thus, S. Typhimurium promotes disease tolerance Symbiotic bacteria
to infection. Endosymbiotic bacteria
e.g. B. fragilis and
e.g. Wolbachia
Another example of how bacteria can promote disease E. coli O21:H+
tolerance is the induction of IL‑10 by Staphylococcus
Enhanced disease tolerance to
aureus150. Recognition of staphylococcal peptidoglycan
by Toll-like receptor 2 (TLR2) induces IL‑10, which Flock house virus and H. hepaticus, B. thailadensis,
prevents unfettered T cell activation; hence, the P. relictum infections Salmonella spp. and
response to S. aureus infection promotes tissue damage in insects C. rodentium infections
control and disease tolerance to infection150. in mice

8 | ADVANCE ONLINE PUBLICATION Nature Reviews | Immunology


www.nature.com/nri
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

resistance mechanisms associated with TH17 immunity, Therapeutic modulation of disease tolerance
including pathogen killing, are mediated by neutro- In some cases, effective pathogen elimination by
phils101. This can be associated with the development immune-driven resistance mechanisms fails to over-
of immunopathology, as shown at epithelial barriers come the morbidity or mortality associated with infec-
where ROS and elastase produced by neutrophils can tion. Moreover, current antimicrobial approaches can
cause epithelial cell damage and compromise disease also fail to treat infectious diseases. In these cases, a
tolerance to pathogenic Gram-negative bacteria, for rational pharmacological targeting of stress and dam-
example, in the case of infection with Burkholderia age responses regulating tissue damage control may act
pseudomallei 106 (FIG.  4c). TH17 immunopathology is therapeutically through the establishment of disease
counteracted by tissue damage control mechanisms tolerance to infection. Examples of proof‑of‑concept
that protect epithelial barriers against oxidative stress for this approach are provided by pharmacological
and damage as well as by additional mechanisms that targeting of adenosine receptors110 or labile haem16,
promote tissue repair and regeneration (FIG. 4c). Some which promote the establishment of disease tolerance
of these mechanisms are regulated directly or indirectly to ­sepsis16,110 or malaria20,21,30 in mice.
by IL‑22 produced by TH17 cells or by ILC3s, which When targeting labile haem, several pharmaco­logical
signals via the IL-22 receptor expressed by epithelial strategies can be envisioned. For example, administra-
cells86,88,107 (FIG. 4c). tion of haemopexin — which is a plasma protein that
binds avidly to labile haem and neutralizes its dele-
Disease tolerance to co‑infections terious effects — confers tissue damage control and
Pathogen class-specific tissue damage control mech- disease tolerance to sepsis in mice16. Gasotransmitters
anisms are particularly relevant in the context of co‑­ such as CO21 can also be used pharmacologically to
infections, as shown for bacterial pneumonia following block haem–iron oxidation (Fe2+ to Fe3+) and inhibit
influenza virus infection108. Several mechanisms may haem release from haemoproteins, thereby preventing
contribute to worsen the clinical outcome of these co‑­ its accumulation in plasma during bloodstream infec-
infections. Viral infections can compromise immune- tions20,21. Remarkably, this is sufficient to confer disease
driven resistance99 or tissue damage control15 mechanisms tolerance to malaria in mice20,21,30. When used pharma-
against bacterial co‑infections, as is the case for influenza cologically, the gasotransmitter NO also promotes the
virus and Legionella pneumophila co-­infections15,99. This establishment of disease tolerance to malaria in mice29.
is probably also the case for co‑­infections by rhinovirus, This protective effect of NO occurs via an indirect
adenovirus or parainfluenza and Streptococcus pneumo- mechanism involving the activation of the transcrip-
niae, as well as for Haemophilus influenza and Moraxella tion factor NRF2, the induction of HO1 expression and
catarrhalis co-infections108. The mechanisms by which the downstream generation of CO, which consequently
viruses compromise disease tolerance to secondary bac­ establishes disease tolerance to malaria29–31. In addition
terial infections are probably multi-factorial and have to preventing haem release from extracellular haemo-
been associated with the dysregulated production of globin, the cytoprotective effects of CO75 should also
EGF-like factors, such as a­ mphiregulin15, by ­lung-­resident contribute to promote tissue damage control and dis-
ILCs86 and Treg cells92. ease tolerance to malaria. As labile haem is a potent
Immune-driven resistance mechanisms targeting pro-oxidant, pharmacological use of antioxidants such
viruses can also promote, rather than impair, disease as N‑acetyl cysteine can prevent this pathogenic effect
tolerance to bacterial infections. For example, regula- and thereby confer tissue damage control and dis-
tion of IL‑1β‑induced inflammation by type I interferon ease tolerance to malaria12,13. Moreover, downstream
(IFN) prevents the lethality of systemic Streptococcus events in the signalling transduction pathways driving
pyogenes infection, without affecting bacterial load109. haem cytotoxicity, such as sustained activation of JUN
In this case, signalling via type I IFN receptor in macro­ amino‑­terminal kinases, can also be targeted pharmaco­
phages, DCs and neutrophils represses IL‑1β tran- logically to promote recovery and survival through
scription through signal transducer and activator of ­tissue ­damage control during malaria infection13.
transcription 1 (STAT1), and to a lesser extent through Pharmacological targeting of metabolic stress
STAT2, which ultimately prevents the development of responses, such as the one regulated by 5′ AMP-
lethal IL‑1β‑driven inflammation109. activated protein kinase (AMPK), confers protection
Disease tolerance against one class of pathogens has against the development of organ dysfunction associ­
also been shown to antagonize immune-driven resist- ated with the pathogenesis of sepsis in mice111. This
ance against other pathogen classes. For example, the treatment is not associated with overt modulation of
induction of HO1 expression promotes disease tol- the host pathogen load, which suggests that AMPK
erance to the blood stage of Plasmodium spp. infec- activation contributes to the establishment of disease
tion12,20,21,30 but impairs resistance to Salmonella enterica tolerance to sepsis111. In further support of this idea,
subsp. enterica serovar Typhimurium co-infection100. pharmacological AMPK activation in the brain is
This is thought to involve dysregulated mobilization of sufficient to confer protection against polymicrobial
granulocytes from the bone marrow with concomitant ­infections in mice112.
induction of HO1 during Plasmodium spp. infection, Another possible approach to modulate disease tol-
which reduces their subsequent capacity to generate erance involves targeting the inflammatory response
ROS and to clear S. Typhimurium infection100. to infection without interfering with immune-driven

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

resistance mechanisms. This can be achieved, for exam- tissue regeneration, such as EGF-like factors, includ-
ple, by pharmacological inhibition of topoisomerase 1, ing amphiregulin15,92, have also been shown to promote
which suppresses the recruitment of RNA polymer- ­disease tolerance to infections in mice.
ase II to the promoter of PRR-responsive genes113. This Furthermore, pharmacological targeting of specific
approach confers a net survival advantage, presum- host resistance mechanisms associated with the devel-
ably through the establishment of disease tolerance, opment of immunopathology is another therapeutic
against viral and bacterial infections as well sterile tis- approach to establish disease tolerance to infection116.
sue injury in mice113. Pharmacological use of cytokines However, as a trade-off, this strategy can be associ-
that are cytoprotective in parenchymal cells, such as ated with reduced host resistance and hence increased
IL‑10 (REF. 114) and IL‑22 (REF. 115), or that promote pathogen load, as shown for influenza A virus117 and

Intracellular Proliferation and


a pathogen Infected cell PCD differentiation

NK Tissue
Tissue
cell damage
damage
control
IFNγR MHC
TCR class I ROS EGFs,
RNS Macrophage ? PDGFs
or DC and
CTL TGFβ
IL-12 IFNγR
IFNγR MHC
TCR class II
Stem cell
B cell
TH1
cell IFNγ
IFNγR
IFNγ

b TSLP, ILR EGFs? Tissue


Tissue IL-25
and ILC2 damage
damage
IL-33 EGFR control
Eosinophil Parasite ST2
Mast cell VEGF, TGFβ,
Basophil PRR RELMα and
Macrophage ?
RELMβ
IgG1 or DC
and
IgE IL-4Rα
MHC
TCR class II
Granuloma B cell IL-4
TH2
cell
ILR
IL-5 and IL-13

Extracellular
c pathogen
PRR

PRR Tissue
Tissue ROS Macrophage damage
damage Elastase DAMP or DC control

IL-1β IL-22R FGF2R


MHC
TCR class II IL-23 FGF2
IL-1R
TH17
cell
IL-23R
IL-17 Treg cell
Neutrophil
IL-1R IL-22
IL-1β ILC3
IL-17
NCR
Stress

Nature Reviews | Immunology


10 | ADVANCE ONLINE PUBLICATION www.nature.com/nri
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Anthracycline Trypanosoma cruzi 118, as well as for human rhinovirus119 tolerance to infection. One possible way forward is the
A class of red aromatic infections in mice. In some cases, however, pharmaco- use of functional ‘drug screens’, which have already
polyketide drugs derived from logical targeting of host resistance mechanisms can con- been successful in identifying anthracycline as a family of
Streptomyces spp. bacteria fer tissue damage control without impairing resistance chemotherapeutic agents inducing disease tolerance to
that intercalate into DNA,
arresting transcription and cell
to infection. This is shown for Mycobacterium tubercu- sepsis in mice17. The therapeutic effect of anthra­cyclines
division, a property widely losis infection in mice116, in which cyclooxygenase inhi- was associated with the induction of tissue damage con-
used therapeutically against bition using ibuprofen decreases both lung lesions and trol via a mechanism involving the ATM kinase17. This
cancers. pathogen load. These, among other studies, provide the and a subsequent study 113 suggest that targeting specific
proof‑of‑principle that immunoregulatory mech­anisms components of the host DNA damage response machin-
can be targeted pharmacologically to confer tissue ery has therapeutic potential in the establishment of
damage control and disease tolerance to different types ­disease tolerance to sepsis.
of infection when antimicrobial therapy alone fails to Finally, a few classes of antibiotics — macrolides,
­overcome host morbidity or mortality. fluoroquinolones, tetracyclines and polymyxines — are
An important question to address is how to ration- labelled as immunomodulatory on the basis of their
ally design therapeutic approaches that would target ability to improve the outcome of chronic disorders via
tissue damage control mechanisms and confer disease mechanisms not readily explained by their antimicro-
bial activity 120. Macrolides have the best-documented
immuno­modulatory activity; for example, the therapeu-
◀ Figure 4 | Pathogen class-specific tissue damage control mechanisms. a | Type 1 tic effects of the macrolide azithromycin in cystic fibrosis
immunity drives resistance to viruses and intracellular bacteria, such as Listeria can be dissociated from its antibacterial activity 120. The
monocytogenes, Salmonella spp. and Mycobacteria spp., as well as to intracellular immunomodulatory effect of these antibiotics probably
protozoan parasites such as Leishmania spp.101. The T helper 1 (TH1) signature cytokine goes beyond inhibition of pro-inflammatory medi­
interferon‑γ (IFNγ) has a central role in triggering cytotoxic mechanisms that, although ators and other immune processes and could involve
directed against intracellular pathogens, can lead to tissue damage through various the modu­lation of molecular pathways associated with
means, including macrophage polarization towards an antimicrobial response the regulation of host lifespan. In support of this idea,
associated with the production of high levels of reactive oxygen species (ROS) and
tetracyclines increase longevity in Caenorhabditis ele-
reactive nitrogen species (RNS), activation of CD8+ cytotoxic T lymphocytes (CTLs)
and natural killer (NK) cells to kill infected cells via the perforin and/or granzyme gans via a mechanism involving the unfolded protein
B‑dependent lytic pathway or via the ligation of surface death receptors; and B cell response (UPR)121, which is a damage response that
activation towards the production of cytolytic antibodies that target infected cells for promotes disease tolerance to infections (as discussed
complement and Fc receptor-mediated cellular cytotoxicity. Tissue damage control above). Colistin induces the activation of the forkhead
mechanisms counteracting type 1 immunopathology rely on cellular regeneration and box O (FOXO) pathway, which is another lifespan exten-
tissue repair to restore homeostasis15,92. The mechanisms by which type 1 immunity sion pathway in C. elegans that confers disease tolerance
contributes to this tissue damage control response are not clear but probably involve the to Gram-negative infections122. Of note, these anti­
production of epidermal growth factors (EGFs), transforming growth factor‑β (TGFβ) and biotics have not been shown to exert a direct effect on
platelet-derived growth factor (PDGF), which drive the proliferation and differentiation tissue damage control mechanisms and more definitive
of stem cells into functional parenchymal cells, restoring tissue integrity and function23.
conclusions need to be experimentally confirmed, for
b | Resistance to extracellular metazoan parasites and other large parasites is mediated
and/or involves type 2 immunity101,102. Pathogen neutralization is achieved via different ­example, by using germ-free organisms.
mechanisms controlled by TH2 signature cytokines, including interleukin‑4 (IL‑4), IL‑5 and
IL‑13, and by additional type 2 cytokines such as thymic stromal lymphopoietin (TSLP), Concluding remarks and perspectives
IL‑25 or IL‑33, secreted by damaged cells101,102. TH2 signature cytokines drive B cell It is now clear that multicellular organisms use two
activation towards the production of high-affinity pathogen-specific IgG1 and IgE genetically distinct defence strategies to limit the patho-
antibodies that function via Fc‑dependent mechanisms to trigger the activation of genicity of microorganisms. The prevailing view has long
eosinophils, mast cells and basophils, expelling pathogens across epithelia102. Some of been that immune-driven host resistance mechanisms
these parasites, for example, helminths, are damaging to parenchymal cells and type 2 that contain, kill or expel invading microorganisms are
immunity encompasses tissue damage control mechanisms that confer disease tolerance the prevailing defence strategy against infectious dis-
to infection by these parasites103. These mechanisms involve the production of EGFs,
eases. However, disease tolerance is an equally impor-
vascular endothelial growth factor (VEGF), TGFβ, resistin-like molecule-α (RELMα) and
RELMβ. c | TH17 immunity confers resistance to extracellular bacteria such as Klebsiella tant host defence strategy against infection, which
pneumoniae, Escherichia coli, Citrobacter rodentium, Bordetella pertussis, Porphyromonas does not have a direct negative effect on pathogens and
gingivalis and Streptococcus pneumoniae, and also to fungi such as Candida albicans, functionally interacts with immune-driven resistance
Coccidioides posadasii, Histoplasma capsulatum and Blastomyces dermatitidis101. mechanisms to limit the severity of infectious diseases.
Activation of TH17 cells by cognate T cell receptor (TCR–MHC class II interactions and The cellular and molecular bases of these interactions
activation of group 3 innate lymphoid cells (ILC3s) via engagement of IL‑1 receptor are only now starting to be appreciated. A series of
(IL‑1R) by IL‑1β secreted from damaged cells lead to the recruitment and activation experimental approaches have been used to identify
of neutrophils106. TH17 immunopathology is driven to a large extent by products of and characterize the molecular and cellular basis of
neutrophil activation, such as ROS and elastase. This is countered by tissue damage tissue damage control mechanisms conferring disease
control mechanisms regulated directly or indirectly by IL‑22, originating from TH17 cells,
tolerance to various pathogens. These mechanisms
TH22 cells (not shown) or ILC3s, and promoting tissue damage control. Other cytokines
produced by TH17 cells, including IL‑17, can amplify this protective response, working have so far been restricted to numerous evolutionarily
together with fibroblast growth factor 2 (FGF2) produced by regulatory T (Treg) cells to conserved stress and damage responses, which have in
promote tissue damage control at epithelial barriers172. DC, dendritic cell; EGFR, EGF some cases been associated with immunoregulatory
receptor; FGF2R, FGF2 receptor; IFNγR, IFNγ receptor; ILR, interleukin receptor; responses controlling host resistance mechanisms.
PCD, programmed cell death; PRR, pattern recognition receptor. How immuno­regulatory mechanisms modulate these

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

stress and damage responses to confer disease tolerance nature of these interactions is crucial for understanding,
to infection is fairly unexplored. A growing number and perhaps subsequently shaping, therapeutic strat­egies
of experimental evidence also suggests that symbiotic to manipulate protection against major infectious dis-
microorganisms can regulate disease tolerance to pre- eases in which host resistance mechanisms fail to limit
vent their pathogenicity as well as that of other patho- disease severity. Such therapeutic approaches will prob-
gens22. However, whether this regulation occurs via the ably also be important to overcome the growing global
induction of immunoregulatory mechanisms and/or health threat imposed by the emergence of multidrug-­
the activation of stress and damage responses remains resistant pathogens as well as to treat co‑infections
to be established. Deciphering the cellular and molecular ­associated with high levels of morbidity and mortality.

1. Soares, M. P., Gozzelino, R. & Weis, S. Tissue damage 16. Larsen, R. et al. A central role for free heme in the cytokine transcription. Nat. Commun. 7, 11624
control in disease tolerance. Trends Immunol. 35, pathogenesis of severe sepsis. Sci. Transl Med. 2, (2016).
483–494 (2014). 51ra71 (2010). 36. Eichenfield, D. Z. et al. Tissue damage drives
2. Kotas, M. E. & Medzhitov, R. Homeostasis, 17. Figueiredo, N. et al. Anthracyclines induce DNA co‑localization of NF‑κB, SMAD3, and NRF2 to direct
inflammation, and disease susceptibility. Cell 160, damage response-mediated protection against severe Rev-erb sensitive wound repair in mouse
816–827 (2015). sepsis. Immunity 39, 874–884 (2013). macrophages. eLife 5, e13024 (2016).
3. Chovatiya, R. & Medzhitov, R. Stress, inflammation, 18. Bessede, A. et al. Aryl hydrocarbon receptor control 37. Greer, S. N., Metcalf, J. L., Wang, Y. & Ohh, M.
and defense of homeostasis. Mol. Cell 54, 281–288 of a disease tolerance defence pathway. Nature 511, The updated biology of hypoxia-inducible factor.
(2014). 184–190 (2014). EMBO J. 31, 2448–2460 (2012).
References 1–3 provide an overview of how 19. Romani, L. Immunity to fungal infections. Nat. Rev. 38. Cheng, S. C. et al. mTOR- and HIF‑1α‑mediated
different stress and damage reponses contribute Immunol. 11, 275–288 (2011). aerobic glycolysis as metabolic basis for trained
to re‑establishment of homeostasis after infection 20. Ferreira, A. et al. Sickle hemoglobin confers tolerance immunity. Science 345, 1250684 (2014).
and other pathological conditions. to plasmodium infection. Cell 145, 398–409 (2011). 39. Kelly, B. & O’Neill, L. A. Metabolic reprogramming in
4. Fauci, A. S. & Morens, D. M. The perpetual challenge 21. Pamplona, A. et al. Heme oxygenase‑1 and carbon macrophages and dendritic cells in innate immunity.
of infectious diseases. N. Engl. J. Med. 366, 454–461 monoxide suppress the pathogenesis of experimental Cell Res. 25, 771–784 (2015).
(2012). cerebral malaria. Nat. Med. 13, 703–710 (2007). 40. Matak, P. et al. Myeloid HIF‑1 is protective in
5. Schneider, D. S. & Ayres, J. S. Two ways to survive 22. Ayres, J. S. Cooperative microbial tolerance behaviors Helicobacter pylori-mediated gastritis. J. Immunol.
infection: what resistance and tolerance can teach us in host-microbiota mutualism. Cell 165, 1323–1331 194, 3259–3266 (2015).
about treating infectious diseases. Nat. Rev. Immunol. (2016). 41. Wang, A. et al. Opposing effects of fasting
8, 889–895 (2008). 23. Karin, M. & Clevers, H. Reparative inflammation takes metabolism on tissue tolerance in bacterial and
6. Medzhitov, R., Schneider, D. S. & Soares, M. P. charge of tissue regeneration. Nature 529, 307–315 viral inflammation. Cell 166, 1512–1525.e12
Disease tolerance as a defense strategy. Science 335, (2016). (2016).
936–941 (2012). 24. Ayres, J. S. & Schneider, D. S. Tolerance of infections. 42. Jantsch, J. et al. Cutaneous Na+ storage strengthens
References 5 and 6 provide a conceptual overview Annu. Rev. Immunol. 30, 271–294 (2012). the antimicrobial barrier function of the skin and
of disease tolerance to infection, bridging its initial 25. Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., boosts macrophage-driven host defense. Cell Metab.
description in plants and insects to mice and Edberg, S. & Medzhitov, R. Recognition of commensal 21, 493–501 (2015).
possibly humans. microflora by toll-like receptors is required for 43. Kino, T. et al. Brx mediates the response of
7. Schaefer, J. F. Tolerance to plant disease. Annu. Rev. intestinal homeostasis. Cell 118, 229–241 (2004). lymphocytes to osmotic stress through the activation
Phytopathol. 9, 235–252 (1971). This seminal article demonstrates that PRRs sense of NFAT5. Sci. Signal. 2, ra5 (2009).
8. Caldwell, R. M., Schafer, J. F., Compton, L. E. & components of the gut microbiota and help to 44. Brocker, C., Thompson, D. C. & Vasiliou, V. The role
Patterson, F. L. Tolerance to cereal leaf rusts. Science maintain a homeostatic balance in host–microbiota of hyperosmotic stress in inflammation and disease.
128, 714–715 (1958). interactions. The article also suggests that PRRs Biomol. Concepts 3, 345–364 (2012).
9. Ayres, J. S., Freitag, N. & Schneider, D. S. contribute to tissue damage control in the host 45. Kuper, C., Fraek, M. L., Muller, H. H., Beck, F. X. &
Identification of Drosophila mutants altering defense gut epithelium. Neuhofer, W. Sepsis-induced urinary concentration
of and endurance to Listeria monocytogenes infection. 26. Ayres, J. S. Inflammasome-microbiota interplay in host defect is related to nitric oxide-dependent
Genetics 178, 1807–1815 (2008). physiologies. Cell Host Microbe 14, 491–497 (2013). inactivation of TonEBP/NFAT5, which downregulates
10. Teixeira, L., Ferreira, A. & Ashburner, M. The bacterial 27. Sykiotis, G. P. & Bohmann, D. Stress-activated renal medullary solute transport proteins and
symbiont Wolbachia induces resistance to RNA viral cap’n’collar transcription factors in aging and human aquaporin‑2. Crit. Care Med. 40, 1887–1895
infections in Drosophila melanogaster. PLoS Biol. 6, disease. Sci. Signal. 3, re3 (2010). (2012).
e2 (2008). 28. Hayes, J. D. & Dinkova-Kostova, A. T. The NRF2 46. Qiu, Y., Hansen, P. J., Zhang, M. & Yang, D. in
References 9 and 10 are the first demonstrations regulatory network provides an interface between Experimental Biology 2016 Meeting Faseb Journal.30,
of disease tolerance in insects, suggesting that this redox and intermediary metabolism. Trends Biochem. (Suppl.) 920.10 (2016).
host defence strategy against infection, identified Sci. 39, 199–218 (2014). 47. Linster, C. L., Van Schaftingen, E. & Hanson, A. D.
originally in plants, can be extrapolated to animals. 29. Jeney, V. et al. Control of disease tolerance to malaria Metabolite damage and its repair or pre-emption.
Reference 10 also demonstrates that host by nitric oxide and carbon monoxide. Cell Rep. 8, Nat. Chem. Biol. 9, 72–80 (2013).
symbiotic interactions modulate disease tolerance 126–136 (2014). This article proposes a new concept — metabolite
to pathogens. 30. Ferreira, A., Balla, J., Jeney, V., Balla, G. & damage.
11. Raberg, L., Sim, D. & Read, A. F. Disentangling Soares, M. P. A central role for free heme in the 48. Franklin, B. S., Mangan, M. S. & Latz, E. Crystal
genetic variation for resistance and tolerance to pathogenesis of severe malaria: the missing link? formation in inflammation. Annu. Rev. Immunol. 34,
infectious diseases in animals. Science 318, 812–814 J. Mol. Med. 86, 1097–1111 (2008). 173–202 (2016).
(2007). This article proposes that labile haem generated This article provides a detailed conceptual and
This is the original demonstration of disease through haemolysis functions as a central mechanistic framework for phase transition as a
tolerance in mammals. component in the pathogenesis of severe forms universal ‘danger signal’ sensed by PRRs.
12. Seixas, E. et al. Heme oxygenase‑1 affords protection of malaria. The article also proposes how 49. Gutteridge, J. M. Lipid peroxidation and antioxidants
against noncerebral forms of severe malaria. physiological mechanisms that control the as biomarkers of tissue damage. Clin. Chem. 41,
Proc. Natl Acad. Sci. USA 106, 15837–15842 pathogenic effects of labile haem counteract 1819–1828 (1995).
(2009). the pathogenesis of severe forms of malaria. 50. Binder, C. J., Papac-Milicevic, N. & Witztum, J. L.
This is the first mechanistic study on disease 31. Gozzelino, R., Jeney, V. & Soares, M. P. Mechanisms Innate sensing of oxidation-specific epitopes in health
tolerance in mammals using the same experimental of cell protection by heme oxygenase‑1. Annu. Rev. and disease. Nat. Rev. Immunol. 16, 485–497
model as in reference 11. Pharmacol. Toxicol. 50, 323–354 (2010). (2016).
13. Gozzelino, R. et al. Metabolic adaptation to tissue iron 32. Soares, M. P. & Ribeiro, A. M. NRF2 as a master 51. Imai, Y. et al. Identification of oxidative stress and
overload confers tolerance to malaria. Cell Host regulator of tissue damage control and disease Toll-like receptor 4 signaling as a key pathway of acute
Microbe 12, 693–704 (2012). tolerance to infection. Biochem. Soc. Trans. 43, lung injury. Cell 133, 235–249 (2008).
14. Rodrigue-Gervais, I. G. et al. Cellular inhibitor of 663–668 (2015). This article shows the extremely potent
apoptosis protein cIAP2 protects against pulmonary 33. Thimmulappa, R. K. et al. NRF2 is a critical regulator pathological effect exerted by lipid peroxidation
tissue necrosis during influenza virus infection to of the innate immune response and survival during via TLR4 signalling.
promote host survival. Cell Host Microbe 15, 23–35 experimental sepsis. J. Clin. Invest. 116, 984–995 52. Suzuki, T., Motohashi, H. & Yamamoto, M. Toward
(2014). (2006). clinical application of the KEAP1‑NRF2 pathway.
15. Jamieson, A. M. et al. Role of tissue protection in 34. Athale, J. et al. NRF2 promotes alveolar mitochondrial Trends Pharmacol. Sci. 34, 340–346 (2013).
lethal respiratory viral-bacterial coinfection. Science biogenesis and resolution of lung injury in 53. Bettigole, S. E. & Glimcher, L. H. Endoplasmic
340, 1230–1234 (2013). Staphylococcus aureus pneumonia in mice. reticulum stress in immunity. Annu. Rev. Immunol. 33,
This is the first mechanistic demonstration of how Free Radic. Biol. Med. 53, 1584–1594 (2012). 107–138 (2015).
deregulation of disease tolerance affects the 35. Kobayashi, E. H. et al. NRF2 suppresses macrophage 54. Lindquist, S. The heat-shock response. Annu. Rev.
pathological outcome of co‑infections. inflammatory response by blocking proinflammatory Biochem. 55, 1151–1191 (1986).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

55. Mohri-Shiomi, A. & Garsin, D. A. Insulin signaling 80. Soares, M. P. & Hamza, I. Macrophages and iron 105. Murray, P. J. Amino acid auxotrophy as a system of
and the heat shock response modulate protein metabolism. Immnunity 44, 492–504 (2016). immunological control nodes. Nat. Immunol. 17,
homeostasis in the Caenorhabditis elegans intestine 81. Beg, A. A. & Baltimore, D. An essential role for NF‑kB 132–139 (2016).
during infection. J. Biol. Chem. 283, 194–201 in preventing TNFα ‑induced cell death. Science 274, 106. Sahoo, M., Del Barrio, L., Miller, M. A. & Re, F.
(2008). 782–784 (1996). Neutrophil elastase causes tissue damage that
56. Singh, V. & Aballay, A. Heat-shock transcription 82. Buchon, N., Broderick, N. A., Poidevin, M., decreases host tolerance to lung infection with
factor (HSF)-1 pathway required for Caenorhabditis Pradervand, S. & Lemaitre, B. Drosophila intestinal Burkholderia species. PLoS Pathog. 10, e1004327
elegans immunity. Proc. Natl Acad. Sci. USA 103, response to bacterial infection: activation of host (2014).
13092–13097 (2006). defense and stem cell proliferation. Cell Host Microbe 107. Gimblet, C. et al. IL‑22 protects against tissue damage
57. Murapa, P., Ward, M. R., Gandhapudi, S. K., 5, 200–211 (2009). during cutaneous leishmaniasis. PLoS ONE 10,
Woodward, J. G. & D’Orazio, S. E. Heat shock factor 1 This is the original finding of the crucial part played e0134698 (2015).
protects mice from rapid death during Listeria by regenerative responses of the gut epithelium as 108. Robinson, K. M., Kolls, J. K. & Alcorn, J. F.
monocytogenes infection by regulating expression of a defence strategy against enteric pathogens. The immunology of influenza virus-associated
tumor necrosis factor-α during fever. Infect. Immun. 83. Hochmuth, C. E., Biteau, B., Bohmann, D. & Jasper, H. bacterial pneumonia. Curr. Opin. Immunol. 34,
79, 177–184 (2011). Redox regulation by KEAP1 and NRF2 controls 59–67 (2015).
58. Richardson, C. E., Kooistra, T. & Kim, D. H. intestinal stem cell proliferation in Drosophila. 109. Castiglia, V. et al. Type I interferon signaling prevents
An essential role for XBP‑1 in host protection against Cell Stem Cell 8, 188–199 (2011). IL‑1β‑driven lethal systemic hyperinflammation during
immune activation in C. elegans. Nature 463, 84. Bonfini, A., Liu, X. & Buchon, N. From pathogens to invasive bacterial infection of soft tissue. Cell Host
1092–1095 (2010). microbiota: how Drosophila intestinal stem cells react Microbe 19, 375–387 (2016).
59. Filone, C. M. et al. The master regulator of the cellular to gut microbes. Dev. Comp. Immunol. 64, 22–38 110. Sullivan, G. W., Fang, G., Linden, J. & Scheld, W. M.
stress response (HSF1) is critical for orthopoxvirus (2016). A2A adenosine receptor activation improves survival
infection. PLoS Pathog. 10, e1003904 (2014). 85. Artis, D. & Spits, H. The biology of innate lymphoid in mouse models of endotoxemia and sepsis.
60. Weitzman, M. D. & Weitzman, J. B. What’s the cells. Nature 517, 293–301 (2015). J. Infect. Dis. 189, 1897–1904 (2004).
damage? The impact of pathogens on pathways that 86. Monticelli, L. A. et al. Innate lymphoid cells promote 111. Escobar, D. A. et al. Adenosine monophosphate-
maintain host genome integrity. Cell Host Microbe 15, lung-tissue homeostasis after infection with influenza activated protein kinase activation protects against
283–294 (2014). virus. Nat. Immunol. 12, 1045–1054 (2011). sepsis-induced organ injury and inflammation.
61. Shiloh, Y. & Ziv, Y. The ATM protein kinase: regulating 87. Turner, J. E. et al. IL‑9‑mediated survival of type 2 J. Surg. Res. 194, 262–272 (2015).
the cellular response to genotoxic stress, and more. innate lymphoid cells promotes damage control in 112. Mulchandani, N. et al. Stimulation of brain AMP-
Nat. Rev. Mol. Cell Biol. 14, 197–210 (2013). helminth-induced lung inflammation. J. Exp. Med. activated protein kinase attenuates inflammation and
62. Deretic, V., Saitoh, T. & Akira, S. Autophagy in 210, 2951–2965 (2013). acute lung injury in sepsis. Mol. Med. 21, 637–644
infection, inflammation and immunity. Nat. Rev. 88. Monticelli, L. A. et al. IL‑33 promotes an innate (2015).
Immunol. 13, 722–737 (2013). immune pathway of intestinal tissue protection 113. Rialdi, A. et al. Topoisomerase 1 inhibition
63. Orvedahl, A. et al. Autophagy protects against dependent on amphiregulin-EGFR interactions. suppresses inflammatory genes and protects from
sindbis virus infection of the central nervous system. Proc. Natl Acad. Sci. USA 112, 10762–10767 (2015). death by inflammation. Science 352, aad7993
Cell Host Microbe 7, 115–127 (2010). 89. Lindemans, C. A. et al. Interleukin‑22 promotes (2016).
64. Maurer, K. et al. Autophagy mediates tolerance to intestinal-stem-cell-mediated epithelial regeneration. 114. Couper, K. N., Blount, D. G. & Riley, E. M. IL‑10: the
Staphylococcus aureus alpha-toxin. Cell Host Microbe Nature 528, 560–564 (2015). master regulator of immunity to infection. J. Immunol.
17, 429–440 (2015). 90. Josefowicz, S. Z., Lu, L. F. & Rudensky, A. Y. Regulatory 180, 5771–5777 (2008).
65. Medzhitov, R. Origin and physiological roles of T cells: mechanisms of differentiation and function. 115. Muhl, H. et al. IL‑22 in tissue-protective therapy.
inflammation. Nature 454, 428–435 (2008). Annu. Rev. Immunol. 30, 531–564 (2012). Br. J. Pharmacol. 169, 761–771 (2013).
This article is a ‘must read’ and probably the best 91. Burzyn, D. et al. A special population of regulatory 116. Vilaplana, C. et al. Ibuprofen therapy resulted in
succinct review so far on inflammation. T cells potentiates muscle repair. Cell 155, 1282–1295 significantly decreased tissue bacillary loads and
66. Pasparakis, M. & Vandenabeele, P. Necroptosis and its (2013). increased survival in a new murine experimental
role in inflammation. Nature 517, 311–320 (2015). 92. Arpaia, N. et al. A distinct function of regulatory T cells model of active tuberculosis. J. Infect. Dis. 208,
67. Dixon, S. J. et al. Ferroptosis: an iron-dependent form in tissue protection. Cell 162, 1078–1089 (2015). 199–202 (2013).
of nonapoptotic cell death. Cell 149, 1060–1072 References 91 and 92 are the original descriptions 117. Carey, M. A. et al. Contrasting effects of
(2012). of an unsuspected physiological role for cyclooxygenase‑1 (COX‑1) and COX‑2 deficiency on
68. Fridman, J. S. & Lowe, S. W. Control of apoptosis by tissue-resident Treg cells in conferring tissue the host response to influenza A viral infection.
p53. Oncogene 22, 9030–9040 (2003). damage control and disease tolerance to infections J. Immunol. 175, 6878–6884 (2005).
69. Ashida, H. et al. Cell death and infection: a double- (reference 92). 118. Hideko Tatakihara, V. L. et al. Effects of
edged sword for host and pathogen survival. J. Cell 93. Schiering, C. et al. The alarmin IL‑33 promotes cyclooxygenase inhibitors on parasite burden, anemia
Biol. 195, 931–942 (2011). regulatory T‑cell function in the intestine. Nature 513, and oxidative stress in murine Trypanosoma cruzi
70. Gillet, G. & Brun, G. Viral inhibition of apoptosis. 564–568 (2014). infection. FEMS Immunol. Med. Microbiol. 52, 47–58
Trends Microbiol. 4, 312–317 (1996). 94. Noel, S. et al. T lymphocyte-specific activation of (2008).
71. Serhan, C. N. Pro-resolving lipid mediators are leads NRF2 protects from AKI. J. Am. Soc. Nephrol. 26, 119. Stanley, E. D., Jackson, G. G., Panusarn, C.,
for resolution physiology. Nature 510, 92–101 2989–3000 (2015). Rubenis, M. & Dirda, V. Increased virus shedding with
(2014). 95. Cramer, T. et al. HIF1α is essential for myeloid cell- aspirin treatment of rhinovirus infection. JAMA 231,
72. Lee, T. S. & Chau, L. Y. Heme oxygenase‑1 mediates mediated inflammation. Cell 112, 645–657 (2003). 1248–1251 (1975).
the anti-inflammatory effect of interleukin‑10 in mice. 96. Dang, E. V. et al. Control of TH17/Treg balance by 120. Tauber, S. C. & Nau, R. Immunomodulatory properties
Nat. Med. 8, 240–246 (2002). hypoxia-inducible factor 1. Cell 146, 772–784 (2011). of antibiotics. Curr. Mol. Pharmacol. 1, 68–79
73. Chen, Y. C. et al. Nitric oxide and prostaglandin E2 97. McFall-Ngai, M. et al. Animals in a bacterial world, a (2008).
participate in lipopolysaccharide/interferon-gamma- new imperative for the life sciences. Proc. Natl Acad. 121. Houtkooper, R. H. et al. Mitonuclear protein
induced heme oxygenase 1 and prevent RAW264.7 Sci. USA 110, 3229–3236 (2013). imbalance as a conserved longevity mechanism.
macrophages from UV‑irradiation-induced cell death. 98. Ayres, J. S. & Schneider, D. S. The role of anorexia in Nature 497, 451–457 (2013).
J. Cell. Biochem. 86, 331–339 (2002). resistance and tolerance to infections in Drosophila. 122. Cai, Y., Cao, X. & Aballay, A. Whole-animal chemical
74. Otterbein, L. E. et al. Carbon monoxide mediates anti- PLoS Biol. 7, e1000150 (2009). screen identifies colistin as a new immunomodulator
inflammatory effects via the mitogen activated protein 99. Jamieson, A. M., Yu, S., Annicelli, C. H. & that targets conserved pathways. mBio 5, e01235‑14
kinase pathway. Nat. Med. 6, 422–428 (2000). Medzhitov, R. Influenza virus-induced glucocorticoids (2014).
75. Brouard, S. et al. Carbon monoxide generated by compromise innate host defense against a secondary 123. Billingham, R. E., Brent, L. & Medawar, P. B. Actively
heme oxygenase 1 suppresses endothelial cell bacterial infection. Cell Host Microbe 7, 103–114 acquired tolerance of foreign cells. Nature 172,
apoptosis. J. Exp. Med. 192, 1015–1026 (2000). (2010). 603–606 (1953).
This is the original description of the cytoprotective 100. Cunnington, A. J., de Souza, J. B., Walther, M. & 124. Foster, S. L., Hargreaves, D. C. & Medzhitov, R.
effects exerted by the gasotransmitter carbon Riley, E. M. Malaria impairs resistance to Salmonella Gene-specific control of inflammation by TLR-induced
monoxide. through heme- and heme oxygenase-dependent chromatin modifications. Nature 447, 972–978
76. Chen, G. Y. & Nunez, G. Sterile inflammation: sensing dysfunctional granulocyte mobilization. Nat. Med. 18, (2007).
and reacting to damage. Nat. Rev. Immunol. 10, 120–127 (2012). 125. Saeed, S. et al. Epigenetic programming of monocyte-
826–837 (2010). References 99 and 100 are the original findings to‑macrophage differentiation and trained innate
77. Bianchi, M. E. DAMPs, PAMPs and alarmins: all we that disease tolerance to one class of pathogens immunity. Science 345, 1251086 (2014).
need to know about danger. J. Leukoc. Biol. 81, 1–5 can compromise immune-driven resistance to 126. Netea, M. G., Latz, E., Mills, K. H. & O’Neill, L. A.
(2007). co‑infections by another class of pathogens. Innate immune memory: a paradigm shift in
78. Mustafa, A. K., Gadalla, M. M. & Snyder, S. H. 101. Kara, E. E. et al. Tailored immune responses: novel understanding host defense. Nat. Immunol. 16,
Signaling by gasotransmitters. Sci. Signal. 2, re2 effector helper T cell subsets in protective immunity. 675–679 (2015).
(2009). PLoS Pathog. 10, e1003905 (2014). Reference 124 provides a mechanistic basis for
79. Wegiel, B. et al. Macrophages sense and kill bacteria 102. Wynn, T. A. Type 2 cytokines: mechanisms and lipopolysaccharide tolerance in macrophages,
through carbon monoxide-dependent inflammasome therapeutic strategies. Nat. Rev. Immunol. 15, which was later expanded to other PRR agonists
activation. J. Clin. Invest. 124, 4926–4940 (2014). 271–282 (2015). and coined as ‘trained immunity’ (references 125
This article proposes that the gasotransmitter 103. Allen, J. E. & Wynn, T. A. Evolution of TH2 immunity: and 126).
carbon monoxide, produced by macrophages via a rapid repair response to tissue destructive 127. Dionne, M. S., Pham, L. N., Shirasu-Hiza, M. &
haem catabolism by HO1, provides metabolic pathogens. PLoS Pathog. 7, e1002003 (2011). Schneider, D. S. Akt and FOXO dysregulation
sensing of microbes and adjusts macrophage 104. Soares, M. P. & Weiss, G. The Iron age of host-microbe contribute to infection-induced wasting in Drosophila.
antimicrobial responses accordingly. interactions. EMBO Rep. 16, 1482–1500 (2015). Curr. Biol. 16, 1977–1985 (2006).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

128. Raberg, L., Graham, A. L. & Read, A. F. Decomposing 144. Pickard, J. M. et al. Rapid fucosylation of intestinal and induces hypertrophy. FASEB J. 26, 987–1000
health: tolerance and resistance to parasites in epithelium sustains host-commensal symbiosis in (2012).
animals. Phil. Trans. R. Soc. B 364, 37–49 (2009). sickness. Nature 514, 638–641 (2014). 161. Latz, E., Xiao, T. S. & Stutz, A. Activation and
129. Simms, E. L. Defining tolerance as a norm of reaction. 145. Brestoff, J. R. & Artis, D. Commensal bacteria at the regulation of the inflammasomes. Nat. Rev. Immunol.
Evol. Ecol. 14, 563–570 (2000). interface of host metabolism and the immune system. 13, 397–411 (2013).
130. Schneider, D. S. Tracing personalized health curves Nat. Immunol. 14, 676–684 (2013). 162. Ip, W. K. & Medzhitov, R. Macrophages monitor tissue
during infections. PLoS Biol. 9, e1001158 (2011). 146. Atarashi, K. et al. Induction of colonic regulatory osmolarity and induce inflammatory response through
This is the original description of disease curves T cells by indigenous Clostridium species. Science NLRP3 and NLRC4 inflammasome activation.
and their interpretation as a novel way to 331, 337–341 (2011). Nat. Commun. 6, 6931 (2015).
understand host–pathogen interactions. 147. Buffie, C. G. & Pamer, E. G. Microbiota-mediated 163. Martinon, F., Petrilli, V., Mayor, A., Tardivel, A. &
131. Torres, B. Y. et al. Tracking resilience to infections by colonization resistance against intestinal pathogens. Tschopp, J. Gout-associated uric acid crystals activate
mapping disease space. PLoS Biol. 14, e1002436 Nat. Rev. Immunol. 13, 790–801 (2013). the NALP3 inflammasome. Nature 440, 237–241
(2016). 148. Stecher, B. et al. Salmonella enterica serovar (2006).
132. Hedges, L. M., Brownlie, J. C., O’Neill, S. L. & Typhimurium exploits inflammation to compete with 164. Duewell, P. et al. NLRP3 inflammasomes are required
Johnson, K. N. Wolbachia and virus protection in the intestinal microbiota. PLoS Biol. 5, 2177–2189 for atherogenesis and activated by cholesterol
insects. Science 322, 702 (2008). (2007). crystals. Nature 464, 1357–1361 (2010).
133. Moreira, L. A. et al. A Wolbachia symbiont in Aedes 149. Sun, H., Kamanova, J., Lara-Tejero, M. & 165. Idzko, M., Ferrari, D. & Eltzschig, H. K. Nucleotide
aegypti limits infection with dengue, chikungunya, Galan, J. E. A. Family of Salmonella type III secretion signalling during inflammation. Nature 509, 310–317
and Plasmodium. Cell 139, 1268–1278 (2009). effector proteins selectively targets the NF‑κB (2014).
134. Dutra, H. L. et al. Wolbachia blocks currently signaling pathway to preserve host homeostasis. 166. Mahamed, D. A., Toussaint, L. E. & Bynoe, M. S.
circulating zika virus isolates in Brazilian Aedes PLoS Pathog. 12, e1005484 (2016). CD73‑generated adenosine is critical for immune
aegypti mosquitoes. Cell Host Microbe 19, 771–774 150. Chau, T. A. et al. Toll-like receptor 2 ligands on the regulation during Toxoplasma gondii infection.
(2016). staphylococcal cell wall downregulate superantigen- Infect. Immun. 83, 721–729 (2015).
135. Pan, X. et al. Wolbachia induces reactive oxygen induced T cell activation and prevent toxic shock 167. Alam, M. S. et al. CD73 is expressed by human
species (ROS)-dependent activation of the Toll syndrome. Nat. Med. 15, 641–648 (2009). regulatory T helper cells and suppresses
pathway to control dengue virus in the mosquito 151. Naquet, P., Giessner, C. & Galland, F. Metabolic proinflammatory cytokine production and
Aedes aegypti. Proc. Natl Acad. Sci. USA 109, adaptation of tissues to stress releases metabolites Helicobacter felis-induced gastritis in mice.
E23–E31 (2012). influencing innate immunity. Curr. Opin. Immunol. 38, J. Infect. Dis. 199, 494–504 (2009).
136. Caragata, E. P. et al. Dietary cholesterol modulates 30–38 (2016). 168. Ohta, A. & Sitkovsky, M. Role of G‑protein-coupled
pathogen blocking by Wolbachia. PLoS Pathog. 9, 152. Dal Peraro, M. & van der Goot, F. G. Pore-forming adenosine receptors in downregulation of
e1003459 (2013). toxins: ancient, but never really out of fashion. inflammation and protection from tissue damage.
137. Ichinohe, T. et al. Microbiota regulates immune Nat. Rev. Microbiol. 14, 77–92 (2016). Nature 414, 916–920 (2001).
defense against respiratory tract influenza A virus 153. Nathan, C. & Shiloh, M. U. Reactive oxygen and 169. Madenspacher, J. H. et al. p53 Integrates host
infection. Proc. Natl Acad. Sci. USA 108, 5354–5359 nitrogen intermediates in the relationship between defense and cell fate during bacterial pneumonia.
(2011). mammalian hosts and microbial pathogens. J. Exp. Med. 210, 891–904 (2013).
138. Zeng, Melody, Y. et al. Gut microbiota-induced Proc. Natl Acad. Sci. USA 97, 8841–8848 (2000). 170. Okabe, Y. & Medzhitov, R. Tissue biology perspective
immunoglobulin G controls systemic infection by 154. Beckman, J. S. & Koppenol, W. H. Nitric oxide, on macrophages. Nat. Immunol. 17, 9–17 (2015).
symbiotic bacteria and pathogens. Immunity 44, superoxide, and peroxynitrite: the good, the bad, and 171. Davies, L. C., Jenkins, S. J., Allen, J. E. & Taylor, P. R.
647–658 (2016). ugly. Am. J. Physiol. 271, C1424–C1437 (1996). Tissue-resident macrophages. Nat. Immunol. 14,
139. Yilmaz, B. et al. Gut microbiota elicits a protective 155. Hardie, D. G., Ross, F. A. & Hawley, S. A. AMPK: 986–995 (2013).
immune response against malaria transmission. Cell a nutrient and energy sensor that maintains energy 172. Song, X. et al. Growth factor FGF2 cooperates with
159, 1277–1289 (2014). homeostasis. Nat. Rev. Mol. Cell Biol. 13, 251–262 interleukin‑17 to repair intestinal epithelial damage.
140. Villarino, N. F. et al. Composition of the gut microbiota (2012). Immunity 43, 488–501 (2015).
modulates the severity of malaria. Proc. Natl Acad. 156. Eijkelenboom, A. & Burgering, B. M. FOXOs: signalling
Sci. USA 113, 2235–2240 (2016). integrators for homeostasis maintenance. Nat. Rev. Acknowledgements
Articles 137–140 provide mechanistic Mol. Cell Biol. 14, 83–97 (2013). The authors thank J. Thompson and F. Brazza for insightful com-
demonstrations of how symbiotic interactions with 157. Becker, T. et al. FOXO-dependent regulation of ments and S. Ramos for careful review and editing of the manu-
different bacteria in the microbiota modulate innate immune homeostasis. Nature 463, 369–373 script (Instituto Gulbenkian de Ciência). M.P.S. is supported by
resistance to infections by different blood-borne (2010). Fundação Calouste Gulbenkian, Fundação para a Ciência e
pathogens. 158. Ahn, H. M., Lee, K. S., Lee, D. S. & Yu, K. JNK/FOXO Tecnologia (FCT; PTDC/SAU-TOX/116627/2010 and HMSP-
141. Schieber, A. M. et al. Disease tolerance mediated by mediated peroxiredoxin V expression regulates redox ICT/0022/2010) and the European Community 7th Framework
microbiome E. coli involves inflammasome and IGF‑1 homeostasis during Drosophila melanogaster gut program (ERC‑2011‑AdG 294709‑DAMAGECONTROL). L.F.M.
signaling. Science 350, 558–563 (2015). infection. Dev. Comp. Immunol. 38, 466–473 is an Fundação para a Ciência e Tecnologia Investigator and is
142. Zele, F., Nicot, A., Duron, O. & Rivero, A. Infection with (2012). supported by the European Community Horizon 2020
Wolbachia protects mosquitoes against Plasmodium- 159. Seiler, F. et al. FOXO transcription factors regulate (ERC‑2014‑CoG 647888‑­iPROTECTION). L.T. is supported by
induced mortality in a natural system. J. Evol. Biol. 25, innate immune mechanisms in respiratory epithelial Fundação Calouste Gulbenkian and Fundação para a Ciência e
1243–1252 (2012). cells. J. Immunol. 190, 1603–1613 (2013). Tecnologia PTDC/BEX-GMG/3128/2014.
143. Mazmanian, S. K., Round, J. L. & Kasper, D. L. 160. Reed, S. A., Sandesara, P. B., Senf, S. M. &
A microbial symbiosis factor prevents intestinal Judge, A. R. Inhibition of FOXO transcriptional Competing interests statement
inflammatory disease. Nature 453, 620–625 (2008). activity prevents muscle fiber atrophy during cachexia The authors declare no competing interests.

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like