Sunteți pe pagina 1din 14

REVIEWS

Role of innate and adaptive immune


mechanisms in cardiac injury and
repair
Slava Epelman1, Peter P. Liu2 and Douglas L. Mann3
Abstract | Despite the advances that have been made in developing new therapeutics,
cardiovascular disease remains the leading cause of worldwide mortality. Therefore,
understanding the mechanisms underlying cardiovascular tissue injury and repair is of
prime importance. Following cardiac tissue injury, the immune system has an important
and complex role in driving both the acute inflammatory response and the regenerative
response. This Review summarizes the role of the immune system in cardiovascular
disease — focusing on the idea that the immune system evolved to promote tissue
homeostasis following injury and/or infection, and that the inherent cost of this
evolutionary development is unwanted inflammatory damage.

The cardiovascular system evolved ~600 million years Immune anatomy of the resting heart
ago as a means to transport nutrients and cells within As in most tissues, the primary immune cells that
multicellular organisms1. Primitive organisms such as reside in the heart are macrophages, which are typically
Drosophila possess a single chamber that functions as observed near endothelial cells or within the interstitial
both a pumping tube and a simple vascular system2. More space, while very few, if any, monocytes are found within
complex organisms have compartmentalized cardio­ cardiac tissue4–7 (FIG. 1). Sparse populations of dendritic
vascular systems, comprising venous and arterial vascular cells (DCs) have been found within cardiac tissue, some
systems that are connected to a multi-chamber muscular of which are localized in cardiac valves, where they
myocardium that continually receives and ejects blood presumably sample antigens5,8. Mast cells are found in
components. Despite the more complex nature of the resting cardiac tissue, and are thought to be important
mammalian cardiovascular system, its primary functions early triggers of immune responses9. A small number of
remain the same, and its importance to health and dis­ B cells and regulatory T cell subsets are also present in
ease is highlighted by the fact that cardiovascular disease cardiac tissue in resting conditions, whereas neutrophils
1
Toronto Medical Discovery is the leading cause of death worldwide, with an increasing are typically not found within non-inflamed cardiac tis­
Tower, 101 College Street, burden over the past decade3. Therefore, understanding sue5,10,11. Whether from sterile or infectious triggers,
TMDT 3903 Toronto, Ontario, both how cardiac tissue is injured and how cardiac tissue cardiac tissue injury initiates a dynamic cellular cascade
M5G 1L7, Canada. regenerates is of prime importance to global health. that initially activates resident immune cells, and over
2
University of Ottawa Heart
Institute, 40 Ruskin Street,
The immune system evolved to enable host defence time this evolves in a coordinated manner that leads to
Ottawa, Ontario, K1Y 4W7, against invading pathogens but also to promote tissue the recruitment of diverse leukocyte populations into
Canada. growth and repair during development or following sterile inflamed tissue.
3
Center for Cardiovascular tissue injury, such as that which can occur within the
Research, Division of
myocardium (BOX 1). In this Review, we detail the roles of Mechanisms of cardiac injury
Cardiology, Department of
Medicine, Washington individual immune cell subsets and immune signalling The myocardium can be injured by various pathophysio­
University School of Medicine, pathways in both sterile and infectious cardiac injury. logical processes, which can be grouped broadly into
St. Louis, Missouri 63110, We also use examples from the cardiac system to suggest ischaemic and non-ischaemic aetiologies. In terms of
USA. the idea that the immune system evolved to promote global disease burden, ischaemic injury is the primary
Correspondence to S.E.
e-mail: slava.epelman@
tissue homeostasis, but that this beneficial activity comes pathophysiological mechanism of injury 3,12. Occlusion
uhnresearch.ca at a cost of increased ‘bystander damage’ when the of a coronary vessel after acute plaque rupture can lead
doi:10.1038/nri3800 immune system overreacts to internal injury. to two potential outcomes: a completed myocardial

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 117

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | The immune system during tissue growth and regeneration


the zymosans of yeast, the glycolipids of mycobacterium,
or the double-stranded RNAs of viruses (FIG. 2). More
A careful examination of examples from the natural world reveals temporal and recently it has become clear that cardiac PRRs also rec­
phylogenetic characteristics that predict the ability of tissues to regenerate in diverse ognize the molecular patterns of endogenous material
organisms. More primitive organisms such as invertebrates, reptiles and amphibians released by dying or injured myocardial cells. Cells that
have a striking regenerative potential when compared with mammals. For example,
die by accidental necrosis, regulated necrosis (that is,
both the zebrafish and newt heart can fully re‑grow after experiencing substantial
injury, and the salamander can fully re‑grow its limbs after amputation — functions that
necroptosis) and/or secondary apoptosis release their
are not possessed by adult mammals141–143. Very young mammals also retain this cytosolic contents into the extracellular space, thereby
regenerative capacity. The neonatal heart can fully regenerate after apical resection initiating a brisk inflammatory response through the
of the left ventricle, myocardial infarction or necrotic injury, but this capacity is lost engagement of an ensemble of extracellular or intra­
after the first weeks of life83–85. One important similarity between more primitive cellular PRRs14. The time course of the inflamma­
organisms and very young mammals is a more limited (primitive) immune system144. tory response that ensues following tissue injury is
The macrophage is a specialized mononuclear phagocyte that resides in all tissues remarkably consistent, irrespective of the specific cause
from the earliest stages of development57,145. Loss of macrophages leads to of the injury, and it is associated with the rapid influx of
abnormalities in growth of complex vascular and neuronal networks, increased neutrophils, and subsequently monocytes, into the area
mortality and stunted growth146–152. Beyond supporting growth, macrophages also have
of tissue injury. This inflammatory response has been
an important and more generalized role in the clearance of senescent cells during
embryonic development87,88. Importantly, non-selective depletion of all macrophages
referred to as ‘sterile inflammation’, insofar as the inflam­
impairs the ability of primitive organisms and young mammals to regenerate, mation following tissue injury occurs in the absence of
highlighting the crucial role these cells have in tissue growth and repair83–85. Although a known pathogenic infection15,16.
macrophages possess important regenerative functions, they can also mediate Many PRRs encountering PAMPs and DAMPs trig­
pathology. Excessive expansion of macrophage populations during ischaemic injury ger signalling cascades that activate nuclear factor‑κB
impairs tissue healing, indicating that certain macrophage subsets can interfere with (NF‑κB), activator protein 1 (AP-1), and interferon
the regenerative process78. Understanding when macrophages do or do not promote regulatory factor (IRF) transcription factors, that in
tissue repair is a crucial first step if we are to understand why the adult human heart has turn regulate target genes that encode pro-inflammatory
only a limited regenerative capacity. cytokines and interferons (IFNs) in the heart 17. Another
subset of PRRs in the heart trigger a distinct pro-
inflammatory mechanism that requires assembly of
infarction occurs if blood flow is not restored, result­ cytosolic protein complexes called inflammasomes18.
ing in permanent anoxic and low-nutrient tissue injury. Canonical inflammasomes convert pro-caspase 1 into
If blood flow is re-established to ischaemic (yet viable) the catalytically active caspase 1 protease that is respon­
tissue, inflammatory ‘reperfusion injury’ can also occur sible the production of interleukin‑1β (IL‑1β) and
(discussed below). Non-ischaemic cardiomyopathy is IL‑18 — cytokines that are sufficient to trigger
a composite diagnosis that includes myocarditis that inflammatory responses in the heart 18.
occurs after viral or bacterial infections or toxin admin­ PRRs can be subdivided into two major classes
istration. In addition, there are cardiomyopathies that based on their subcellular localization. Toll-like recep­
develop secondary to chronic hypertension. All these tors (TLRs) and C‑type lectin receptors are found on
forms of injury are influenced by genetic predisposition, plasma membranes or endosomes, where they can
which can itself lead to early onset cardiac dysfunction13. detect the presence of PAMPs or DAMPs. A second
Whether through acute ischaemic injury or through class of PRRs resides in intracellular compartments,
the gradual impairment of cardiac function following a and includes retinoic-acid inducible gene I (RIG-I)‑like
variety of clinical pathologies, irreversible heart failure receptors (RLRs), NOD-like receptors (NLRs) and
often develops. As we are coming to understand, the absent-in‑melanoma 2 (AIM2) receptors19,20.
immune system can contribute both during the initial Little is known of how TLR expression is regulated in
insult and during the chronic phase of cardiac injury the heart. However, TLR4 seems to be upregulated in the
— and despite the investment of substantial research failing human heart and on circulating monocytes at
into understanding the contribution of immune cells the time of myocardial infarction21–23. In animal studies,
to cardiac injury and repair, much remains unknown. loss of TLR4 in haematopoietic cells is protective
in the setting of sepsis-induced cardiac dysfunction and
Innate sensing of cardiac injury the loss of TLR2 in haematopoietic cells is protective
Mammalian hearts use both innate and adaptive during ischaemic injury 24–26. The loss of TLR4 is also
immunity to respond to tissue injury resulting from protective following ischaemic injury, although it is not
pathogens or environmental injury (for example, known whether this is attributable to the absence of
ischaemia or haemodynamic overloading). Resident TLR4 activity in haematopoietic or other cell types25–28.
cardiac immune cells are triggered by the detection During haemodynamic stress, mitochondria are typi­
of pathogen-associated molecular patterns (PAMPs) cally damaged; if the degradation of mitochondrial
or damage-associated molecular patterns (DAMPs) by DNA is inhibited in this setting, a TLR9‑dependent
a fixed number of germline encoded pattern recogni­ inflammation-induced cardiomyopathy develops29.
tion receptors (PRRs). Classic examples of pathogen- NLRs function as cytosolic sensors of intra­cellular
associated molecular patterns include the lipopolysac­ DAMPs and PAMPs. In humans, the NLR family is
charides (LPSs) of Gram-negative microorganisms, composed of 22 intracellular PRRs that share a central
the teichoic acids of Gram-positive microorganisms, NACHT domain and a carboxy-terminal leucine-rich

118 | FEBRUARY 2015 | VOLUME 15 www.nature.com/reviews/immunol

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Monocyte Immune cell activation in cardiac injury


Acute ischaemic injury is the best characterized model
of cardiac injury and repair. Following injury, necrotic
Cardiomyocyte
cell death leads to the activation of tissue-resident
immune and non-immune cells. These cells produce
Macrophage pro-inflammatory cytokines and chemokines that are
responsible for recruiting inflammatory leukocytes
Blood DC from the blood into the area of tissue injury 35. The initial
vessel inflammatory phase is followed by a proliferative phase
Endothelial TReg cell
that is characterized by the expansion of neutrophil
cell and macrophage populations that are responsible for
Mast cell removing dead cells and matrix debris, as well as releas­
Neutrophil ing cytokines and growth factors that lead to the forma­
B cell tion of a highly vascularized granulation tissue, which is
composed of connective tissue and new blood vessels.
The final maturation phase is characterized by fibroblast
activation and endothelial cell proliferation, culminating
in reparative myocardial fibrosis and angiogenesis36.
During the very early stages of ischaemic injury, mast
cells and soluble complement proteins become impor­
Figure 1 | Immune cells in the resting heart.  The majority of immune cells in the resting
tant initiators of inflammation — a process that is ampli­
heart are macrophages, which are found primarily surrounding endothelial
Nature Reviewscells but are
| Immunology fied following coronary reperfusion. Immediately after
also seen in the interstitium amongst cardiomyocytes. Mast cells, dendritic cells (DCs), blood flow is restored, resident cardiac mast cells release
B cells and regulatory T (TReg) cells are found sparsely in cardiac tissue, while neutrophils pre-formed pro-inflammatory mediators (for example,
and monocytes are not observed within myocardial tissue, but can be observed as tumour necrosis factor (TNF), histamine and mast cell
contaminants found in the vasculature during steady state5. proteases) that initiate an amplification loop involving
adjacent cells, such as endothelial cells, resident macro­
phages and, subsequently, infiltrating neutrophils9.
repeat region . Analysis of human tissue has revealed that Timely restoration of blood flow to viable tissue is cru­
30

NOD1, NOD2 and the NLR family members NOD-, LRR- cial to prevent cardiomyocyte death; however, reperfu­
and pyrin domain-containing 2 (NLRP2) and NLRP3 sion comes at the cost of exposing complement proteins
are expressed in the heart. Both NOD1 and NLRP3 have to injured endothelial cells and myocardium (see REF. 37
been shown to activate canonical inflammasomes in for a review). Activated complement proteins trigger
the heart, and these molecules have an important role further mast cell degranulation, release of histamine
in the adverse cardiac remodelling events that follow and vasogenic oedema37. Cleaved complement proteins
ischaemia–reperfusion injury and myocardial infarction; such as C5a both attract neutrophils and induce their
however, the cell types involved are not known18,31. transendothelial migration into the injured tissue via the
The RLR family is composed of RIG‑I, melanoma CD11b–CD18 complex 38 (FIG. 3a). If reperfusion is not
differentiation-associated gene 5 (MDA5) and LGP2. established, cardiomyocyte cell death ensues through
RLRs are localized in the cytoplasm and recognize the a variety of pathways that lead to additional DAMP
genomic RNA of double-stranded RNA (dsRNA) viruses release. The exact role of resident mast cells is in part
and the dsRNA that is generated as the replication inferred, as models of mast cell deficiency generally use
intermediate of single-stranded RNA (ssRNA) viruses. Kit−/− animals, which have other immune-cell deficits39–41.
MDA5 is the best characterized receptor in this family, However, mast cells, the complement cascade, oxidative
and the loss of MDA5 expression in cardiomyocytes stress and pro-inflammatory cytokine and chemokine
leads to uncontrolled viral replication and rapid death in production immediately after injury initiate a complex
mice infected with encephalomyocarditis virus (EMCV), interplay between innate and adaptive immune cells.
whereas over-expression of MDA5 in the heart protects
against lethal myocarditis32,33. A final set of PRRs that Neutrophil recruitment during cardiac injury
are expressed in human and mouse heart tissue are the Following either cardiac ischaemic injury or pres­
C‑type lectin receptors, which are calcium-dependent sure overload, neutrophils are the first innate immune
carbohydrate-binding receptors34. However, very little is cells recruited to the myocardium in large numbers42.
known about their roles in cardiac tissue injury. Patients deficient in neutrophils or neutrophil function
One important issue that has been only partially suffer from devastating disseminated bacterial infec­
Granulation tissue addressed is the delineation of cell-type-specific roles tions, indicating a clear requirement for this cell type to
This is tissue that arises for PRRs. Beyond the few examples given here, it is not prevent expansion of otherwise harmless pathogens43.
after cardiac tissue injury, clear what differential roles various PRRs have within However, their role in the response to cardiac injury is
when the replacement of individual immune and non-immune cell subsets almost entirely pathological and, as such, neutrophils
cardiomyocytes with collagen
and extracellular matrix occurs
during the process of cardiac tissue injury and repair, are the best example of a cell type that promotes overall
in order to maintain the which represents an important avenue for further longevity, but in the setting of sterile injury, they have no
integrity of the myocardial wall. investigations. known protective role.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 119

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Heart sites, where viable tissue is present. Initially, migration is


dependent on chemokines, and subsequently neutrophils
follow necrotic signals, such as liberated intra­cellular
ATP (which is a DAMP), in order to precisely home to
the site of tissue injury 45. The reason neutrophils (and
perhaps other recruited immune cells) take this more
DAMPs convoluted path is that necrotic tissue is usually non- (or
Receptors
• dsRNA
• TLR3 under) perfused, and transendothelial migration at the
Thrombus • CpG site of injury is not possible. Unfortunately, such detailed
• TLR9
• Uric acid
Cell necrosis crystal
• NLRP3 imaging of migrating neutrophils has yet to be done in
•TLR2–TLR4–RAGE
• HMGB1
• P2Y11 and P2X7
the injured beating heart, but techniques to achieve this
• ATP are being developed46. Such studies will be highly inform­
• TLR4
• HSP
• IL-1α
• IL-1R ative for understanding the spatial dynamics of resident
Ischaemic area macrophages, recruited neutrophils and monocytes
during the progression of ischaemic cardiac injury.
DAMPs An important early mediator of tissue injury seems
• Hyaluronan Receptors to be IL‑6, which is produced in an autocrine fashion by
ECM
degradation
fragment • CD44–TLR4 cardiomyocytes and recruited myeloid cells (both neutro­
• Heparan • TLR4
sulphate
phils and macrophages)47,48. IL‑6 upregulates intracellular
adhesion molecule 1 (ICAM1) expression on cardio­
Immune cell recruitment
and activation
myocytes, inducing neutrophil binding and stimulating
cytotoxic activity 49–51. The role of neutrophils following
Figure 2 | Cardiac injury and sensing damaged tissue.  The figure shows a coronary cardiac injury may be more pronounced when major
artery occlusion (black) that leads to ischaemic tissue injuryNature
(grey zone).
ReviewsFrom within the
| Immunology areas of the myocardium are at risk of cell death (but have
ischaemic area, cell necrosis, extracellular matrix (ECM) degradation and recruitment
not yet died), such as following ischaemia–reperfusion
of immune cells all lead to the production of specific damage-associated molecular
patterns (DAMPs), which are recognized by pattern recognition receptors. This leads
injury. Neutrophils produce numerous proteases that
to the generation of inflammatory responses to internal injury signals. CpG, CpG contribute to injury, and blockade of neutrophil recruit­
dinucleotides; dsRNA, double-stranded RNA; HMGB1, high-mobility group box 1; ment seems to be most effective during more limited
HSP, heat shock protein; IL, interleukin; IL-1R, IL-1 receptor; NLRP3, NOD-, LRR- episodes of ischaemia, rather than during longer episodes
and pyrin domain-containing 3; P2Y, P2Y purinoceptor; P2X, P2X purinoceptor; in which cardiomyocyte death is largely secondary to
RAGE, receptor for advanced glycation end-products; TLR, Toll-like receptor. anoxia and nutrient deprivation52–54.

Cardiac macrophages
Neutrophil recruitment is mediated in two phases The dominant view for the past half century has been
(FIG. 3a); the first phase is peripheral activation prior to that bone marrow-derived haematopoietic stem cells
infiltration. Mitochondria in all cell types, including (HSCs) give rise to circulating blood monocytes, which
cardiomyocytes, contain formylated peptides and mito­ enter into tissues and become tissue macrophages55.
chondrial DNA, both of which are structurally similar However, in the past few years a series of more definitive
to bacterial components. In an analogous system (skel­ publications have drastically revised our understand­
etal muscle necrosis), these mitochondrial DAMPs are ing of monocyte and macrophage origin by showing
released. Formylated peptides and mitochondrial DNA that many tissue-resident macrophage populations are
are sensed by formyl peptide receptor 1 (FPR1) and established during embryonic development, and they
TLR9, respectively, and the triggering of these receptors are subsequently maintained by self-renewal, rather than
promotes neutrophil activation and their recruitment to through blood monocyte input 5,56–61. Given the impor­
inflamed tissues29. It is important to confirm whether tance of monocytes and macrophages to cardio­vascular
similar mechanisms are involved in neutrophil activation disease, we review recent insights into the origin of
following cardiomyocyte necrosis. The ability to sense functions of these subsets and how they contribute to
mitochondrial motifs is not surprising as mitochondria cardiac tissue injury and repair.
are endosymbionts and related to microorganisms in There are two main subsets of circulating mono­
many ways. However, the exact role of mitochondrial cytes in mice, LY6Chi monocytes and LY6Clow monocytes.
motifs in cardiac injury remains to be defined. LY6C+ monocyte progenitors give rise to LY6Chi mono­
The second phase of neutrophil activation depends on cytes and, through a nuclear receptor subfamily  4
cardiac endothelial cells. Pro-inflammatory mediators, group A member 1 (NR4A1)-dependent transcrip­
such as TNF, IL‑1β and histamine, activate endothelium tional programme, LY6C hi monocytes differentiate
and induce the upregulation of adhesion molecules that into LY6Clow monocytes58,62,63. Global transcriptional
enable neutrophil transmigration between and through profiling has shown these monocyte subsets are con­
endothelial cells to reach the site of tissue injury 9,44. served in humans64 and they have very different roles
Detailed imaging studies in other organ systems have in vivo. LY6Clow monocytes adhere to and move along
aided our understanding of how neutrophils enter the endothelium, both clearing damaged cells and
damaged tissue. For example, following necrotic tissue triggering inflammatory responses without entering
injury in the liver, neutrophils adhere to more remote tissue65,66. During cardiac stress, LY6Chi monocytes are

120 | FEBRUARY 2015 | VOLUME 15 www.nature.com/reviews/immunol

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a 24 hours post ischaemic injury DAMPs

Necrotic Endothelial
cardiomyocyte cell activation

Histamine,
DAMPs TNF and IL-1β
Infarct
zone

Mast cell Peripheral


degranulation C5a
C5 neutrophil
activation
Proteinases and
Phagocytosis of collagenases
necrotic cells

Neutrophil
recruitment

Ischaemia and anoxia

b 3–4 days post ischaemic injury IL-17A promotes


neutrophil production
in bone marrow
IL-17A induces
apoptosis IL-17A TReg cell

Apoptotic γδ T cell IL-10 IL-10 suppresses


cardiomyocyte IL-17A monocyte activation
Apoptotic
neutrophil IL-23
Activated Endothelial cell
monocyte

Monocytes produce CCL7


pro-inflammatory
cytokines and
IL-10 chemokines
and TGFβ CCR2-dependent
TGFβ monocyte recruitment
Innate
B cell
Monocytes ingest
apoptotic cells and Innate B cells activated
release anti-inflammatory in response to DAMPs
cytokines Fibroblast
TGFβ induces
fibrosis

Nature Reviews | Immunology


Figure 3 | Immune response to ischaemic injury in adult animals.  a | Early after ischaemic injury (within 24 hours),
endogenous danger-associated molecular patterns (DAMPs) are released from necrotic cardiomyocytes and activate
resident mast cells, causing mast cell degranulation and release of preformed pro-inflammatory cytokines and vasogenic
compounds (such as histamine, tumour necrosis factor (TNF) and interleukin‑1β (IL‑1β)), which activate endothelial cells.
Necrotic cardiomyocytes also release mitochondrial DAMPs (such as formylated peptides and mitochondrial DNA) into
the circulation, which causes systemic neutrophil activation. Activated neutrophils adhere to activated endothelium
and transmigrate into the cardiac tissue following a chemokine gradient. Neutrophils secrete proteases that digest
tissue (and also activate chemoattractants, such as complement component C5a), which further potentiates leukocyte
recruitment. Neutrophils are directed to ischaemic areas by following gradients of DAMPs (such as ATP). Neutrophils
may then phagocytose dying cells, but they can also induce apoptosis in healthy cardiomyocytes themselves through
the release of reactive oxygen species. b | In the later phases of ischaemic injury (3–4 days), there is recruitment of LY6Chi
monocytes from the blood into ischaemic cardiac tissue. Some of these monocytes originate from ‘reservoirs’ in the
spleen. Innate B cells that express immunoglobulins IgM and IgD are also recruited into the myocardium and produce
CC‑chemokine ligand 7 (CCL7), which promotes further monocyte recruitment10. Innate B cell activation is MYD88
(myeloid differentiation primary response protein 88) dependent (suggesting Toll-like receptor and DAMP involvement).
Recruited monocytes secrete pro-inflammatory cytokines and chemokines, and they drive inflammatory processes.
A proportion of recruited monocytes ingest apoptotic cells, including neutrophils, which increases the secretion of
anti-inflammatory cytokines, such as transforming growth factor-β (TGFβ) and IL‑10, and thereby decrease leukocyte
recruitment. Monocytes produce IL‑23, which drives the production of IL‑17A by γδ T cells. IL‑17A has two main roles
in perpetuating the inflammatory response: it drives neutrophil production in the bone marrow and causes cardiomyocyte
death. As inflammatory responses diminish, less IL‑23 is produced in the tissue. CCR2, CC-chemokine receptor 2;
TReg, regulatory T cell.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 121

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

the primary subset recruited to the heart, either follow­ against invading pathogens, but their in­appropriate
ing ischaemic injury or hypertensive stress; whereas activation in the setting of sterile inflammation may
LY6Clow monocytes do not seem to be directly recruited lead to unwanted pathology 75,76. Prior studies have used
into the myocardium5,59,61,67,68. Monocytes recruited into dichotomous expression of two classical myeloid cell
ischaemic myocardium are found in the blood, but the markers (F4/80 and CD11b) in order discern embry­
spleen is also a monocyte reservoir that can be used onic versus adult monocyte origin57. However, differ­
when blood and bone marrow stores are insufficient69,70. ent cardiac macrophage lineages (embryonic and adult)
Splenic monocytes also seem to have a protective role, as cannot be distinguished by these cell surface markers
splenectomy leads to impaired infarct healing 71. Monocyte alone and, as such, genetic fate mapping remains the
recruitment is in part dependent on innate B cells, which most accurate method for lineage discrimination of
are also recruited to the ischaemic myocardium, and cardiac macrophages in adult mice5,72.
these B cells drive monocyte population expansion
through a CC‑chemokine ligand 7 (CCL7)-dependent Functions of cardiac macrophages. One of the interest­
process10 (FIG. 3b). Once monocytes enter the tissue they ing challenges in the setting of cardiac tissue injury is to
begin to differentiate into macrophages. For many years, define the roles of recruited monocytes and monocyte-
it has been very challenging to separate recruited mono­ derived macrophages. Non-selective depletion strategies
cytes from resident macrophages and, as a result, they have shown that in the absence of both monocytes and
have been analysed as a single population. However, macrophages, scar formation is impaired after cardiac
as we describe below, these populations have different ischaemic injury, with decreased collagen production,
ontological lineages, and this has important functional decreased angiogenesis and increased mortality due
implications. to myocardial rupture6,77. However, experiments using
ApoE‑deficient mice (apolipoprotein-E-deficient mice) sug­
Cardiac macrophage subsets. Recently, studies using gest that excessive expansion of macrophage populations
genetic fate mapping, parabiosis and adoptive transfer can have a detrimental effect on infarct healing, leading to
techniques have defined resident cardiac macrophage excessive inflammation and impaired cardiac function78.
populations in much more detail. Rather than being a Furthermore, studies of cardiac injury in Ccr2−/− mice,
homogenous population, resident cardiac macrophages which lack circulating monocytes, indicate that monocyte
comprise three discrete subsets that have different ori­ recruitment and the associated inflammatory response
gins and functions5 (FIG. 4a). These three macrophage leads to increased cardiac pathology 79–81. The loss of
subsets are defined by their differential expression LY6Chi monocytes also prevents hypertension-induced
of MHC class II and CC chemokine receptor 2 (CCR2). cardiac fibrosis and improves left ventricle function after
MHC class IIhi and MHC class IIlow cardiac macrophages myocardial infarction. Taken together, these data suggest
are both CCR2− and are, numerically, the dominant that recruited monocytes have a pathological role in the
subsets in the heart 5. These macrophage subsets are setting of sterile cardiac injury, but that resident embry­
primarily derived from embryonic progenitors, with onically derived macrophages are likely to have important
a substantial number arising from embryonic yolk sac roles in the tissue-repair response6,67,82.
precursors, and they renew through in situ proliferation, Interestingly, as already mentioned, the neonatal heart
rather than through monocyte input. After birth, there has a remarkable capacity to regenerate in response to
is some dilution of embryonically derived macrophages multiple forms of tissue injury, but the regenerative pro­
by recruited monocyte-derived macrophages in the cess is lost if resident embryonically derived macrophages
heart, but in adult mice the majority of resident cardiac are eliminated83–85. In fact, the neonatal heart supports
macrophages remain of embryonic origin5,72. the expansion of tissue-resident embryonically derived
The third cardiac macrophage subset is made up macro­phage populations following injury, as opposed
of CCR2+ macrophages, which are derived from, and to the recruitment of monocytes, and this seems to be a
slowly replenished by, circulating blood monocytes. fundamental difference between the neonatal and adult
Innate B cells Detailed studies during hypertensive stress suggest that heart. Neonatal cardiac macrophages promote endothelial
These cells are thought to embryonically derived macrophage populations expand cell activation and cardiomyocyte growth, and they gener­
become rapidly activated in solely through in situ proliferation, whereas monocyte- ate minimal inflammation after stimulation through TLR
the absence of classical derived macrophages require monocyte input prior to and inflammasome pathways85 (FIG. 4b). These data suggest
T cell-dependent antigen
presentation mechanisms.
proliferative expansion in the tissue5. CCR2+ macro­ that that the neonatal heart avoids excessive inflamma­
They are mobilized by other phages express high levels of pro-inflammatory genes, tory responses, which may be a crucial factor that aids the
cell types and/or inflammatory including those associated with the NLPR3 inflamma­ regenerative process, although this benefit may come at
triggers. some, which is required to process and deliver IL‑1β to the cost of being more susceptible to infections86.
the heart during cardiac stress5. Inflammasome acti­ Macrophage subsets that are primarily derived from
ApoE-deficient mice
These mice are used to model vation promotes adverse cardiac remodelling follow­ embryonic precursors are more efficient at internaliz­
atherosclerosis. They have ing ischaemic injury, genetic cardiac hypertrophy and ing debris and engulfing apoptotic cardiomyocytes, sug­
increased total plasma hyper­tensive cardiac disease18,73,74; it is likely that CCR2+ gesting they have important homeostatic roles. Indeed,
cholesterol levels and cardiac macrophages are involved in inflamma­some acti­ these functions are reminiscent of embryonic macrophage
increased bone marrow
production of monocytes and
vation in each of these settings. The robust pro-inflam­ functions during development and could suggest that
heightened inflammatory matory gene signature of CCR2+ cardiac macro­phages some of these functions are ‘hard-wired’ into the adult
responses. suggests that they are important for immune protection macrophages5,87,88. The uptake of dead or dying cells is

122 | FEBRUARY 2015 | VOLUME 15 www.nature.com/reviews/immunol

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Early in embryonic development Later in embryonic development Adult heart


MHC class IIhi
MHC class IIlow yolk sac- MHC class IIlow fetal macrophage MHC class IIlow
monocyte-derived macrophage macrophage
Cardiomyocyte derived macrophage

Monocyte

Blood
vessel

Embryonically
Developing heart seeded by derived macrophages
yolk sac-derived macrophages proliferate in situ

Heart seeded by macrophages MHC class IIhi macrophages develop


derived from fetal monocytes from MHC class IIlow populations
Adult monocytes give rise to MHC class IIhi
and MHC class IIlow macrophages and
short-lived CCR2+ macrophages

b
Neonatal heart Adult heart

Receptive
neonatal
cardiomyocyte
Quiescent
adult
cardiomyocyte

Tissue injury Expansion


of resident Tissue injury
macrophage
populations

↑ Angiogenesis ↓ Angiogenesis
↑ Cardiomyocyte proliferation ↓ Cardiomyocyte proliferation
↓ Inflammation ↑ Inflammation

Regeneration of • Impeded regeneration


cardiac tissue of cardiac tissue
• Scar formation

Figure 4 | Role of embryonically derived macrophages during tissue injury and adult animals (where there is minimal tissue regeneration). In the neonate,
Nature Reviews | Immunology
and repair.  a | During embryonic development, extra-embryonic yolk-sac resident embryonically derived macrophage populations expand without
derived macrophages (shown in blue) initially seed the developing heart. The notable monocyte input. Embryonically derived macrophages promote
cells express low levels of MHC class II molecules. Later during development, angiogenesis, cardiomyocyte proliferation and drive minimal inflammation
fetal monocyte-derived macrophages (shown in orange) also infiltrate the when stimulated by damage-associated molecular patterns (DAMPs). These
heart, and both of these embryonic macrophage populations expand further properties of neonatal macrophage populations facilitate cardiac
during development, and after birth, by in situ proliferation. MHC class IIhi regeneration85. Following cardiac injury in the adult, there is marked expansion
macrophages develop from these populations of MHC class IIlow macrophages of CCR2 + monocyte and macrophage populations. Notably, these
after weaning5,72. During this time, haematopoietic stem cell-derived macrophages possess a limited ability to promote angiogenesis and
monocytes infiltrate the heart and also differentiate into MHC class IIhi cardiomyocyte population expansion, but have notable capacity to drive
macrophages (dark green) and MHC class IIlow macrophages (light green). inflammatory responses, which impedes tissue regeneration5,85. Importantly,
MHC class IIhi and MHC class IIlow macrophages persist in the tissue as an neonatal cardiomyocytes are primed to divide and therefore are receptive to
ontologically mixed population, primarily composed of embryonically derived regenerative signals84,153,154. Adult cardiomyocytes are much less receptive
macrophages5. Monocytes that infiltrate the heart can also become short-lived to regenerative signals, and growth signals from embryonically derived cardiac
CC-chemokine receptor 2 (CCR2)+ macrophages, which are entirely derived macrophages are one of many important components of cardiac regeneration
from blood monocytes5. b | The figure shows a comparison of the macrophage promotion in adult tissues. A key therapeutic goal would be to recapture the
response to tissue injury in neonatal animals (which regenerate cardiac tissue) neonatal response to injury in the injured adult heart.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 123

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

an important function in the setting of ischaemic injury. In addition to these roles of T cells, other lympho­
Expression of the MER tyrosine kinase (MERTK) — a cytes have been shown to shape inflammatory responses
phagocytic receptor that is a highly specific marker in cardiac tissue. Natural killer (NK) cells were reported
of tissue macrophages — is upregulated on cardiac to infiltrate the myocardium and have protective effects
macrophages after myocardial infarction, and loss of following ischaemic injury 103. The adoptive transfer of
MERTK expression leads to the accumulation of apop­ activated NK cells into mice was shown to protect against
totic cardiomyocytes, increased neutrophil persistence left ventricle dysfunction following ischaemia by pre­
and decreased levels of the anti-inflammatory cytokine venting fibrosis and enhancing angiogenesis through a
IL‑10 in the myocardium89,90. This ongoing inflamma­ contact-dependent interaction with endothelial cells.
tion ultimately results in decreased cardiac function89,90.
Phagocytosis of apoptotic cells (efferocytosis) in skeletal Transition from acute inflammation to fibrosis
muscle results in the transition of macrophages to a more One of the first important steps that limits tissue injury
anti-inflammatory state through the upregulation of the and promotes the transition to tissue healing is the
intracellular signalling kinase AMPK, which is associated resolution of neutrophil recruitment. Macrophages
with downregulation of pro-inflammatory genes (such as are believed to initiate the process through two key
Tnf) and the upregulation of anti-inflammatory genes, mechanisms that have been linked to their phagocytic
such as Il10 (REFS 91,92). Together, these data suggest that capacity. First, it has been well described in vitro and
strategies that target newly recruited monocytes while in vivo that macrophages that have ingested apoptotic
sparing resident macrophages may yield therapeutic cells increase their production of anti-inflammatory
benefit during cardiac injury. and pro-fibrotic cytokines, such as IL‑10 and TGFβ,
while they decrease production of pro-inflammatory
Adaptive immune cells and heart injury cytokines, such as IL‑1β and TNF 104,105. Following
Lymphocytes have been reported to have diverse roles ischaemic injury, if all cardiac macrophages are absent,
during cardiac tissue injury and repair. CD4‑deficient or they if they lack the phagocytic receptor MERTK,
mice, MHC class II‑deficient mice and mice that express there is neutrophil persistence within infarcted myo­
a single transgenic T cell receptor show impaired wound cardial tissue and ongoing inflammation. This suggests
healing and increased monocyte population expansion that the activation of cardiac macrophage phagocytic
after ischaemic injury. This suggests that CD4+ T cells pathways limits inflammation, and that the loss of the
have protective functions during cardiac injury, and phagocytic macrophages results in an accumulation of
that auto-antigens may be presented to CD4+ T cells by apoptotic material in the infarct zone5,6,89.The transi­
MHC class II‑expressing cells (such as macrophages and tion from inflammatory to pro-fibrotic macrophages
DCs), which then drives immunosuppressive responses has been classically thought to involve polarization
in the myocardium93. Interestingly, it may be that regu­ changes (from an ‘M1’ to an ‘M2’ macrophage pheno­
latory T cells are the key CD4+ T cell subset involved type); however, it is becoming clear that macrophages
here, and regulatory T cells have been shown to promote are very heterogeneous and fit along a broad spectrum
myo­cardial recovery through an IL‑10‑dependent path­ of possibilities, rather than into clearly defined subsets.
way 94,95. Another study found that recombination acti­ Further work is required to define where individual
vating gene (RAG)-deficient mice (which lack T cells cardiac macrophage subsets fit into the M1 and M2
and B cells) have decreased infarct sizes after ischaemic framework106.
injury, and that the transfer of CD4+ T cells into the RAG- The second mechanism involves a complex cytokine
deficient mice had a negative effect on infarct healing. cascade that depends on γδ T cells and their production
Therefore, in certain situations CD4+ T cells may have of IL‑23 and IL‑17A. The production of IL‑17A leads to
pathogenic roles following ischaemic injury96. In addi­ neutrophil production and release from bone marrow
tion, RAG-deficient mice have decreased levels of circu­ stores107. Interestingly, IL‑17A is regulated by secretion
lating IgG antibodies, and these antibodies are known to of IL‑23 by macrophages, and the ingestion of apoptotic
have immunosuppressive effects (as reviewed in REF. 97). neutrophils decreases IL‑23 production, which leads to
CD8+ T cells have also been shown to have regula­ decreased IL‑17A levels and decreased neutrophil pro­
tory roles following ischaemic injury. CD8+ T cells duction in the bone marrow 107–109. Following ischaemic
express type 2 angiotensin II receptor (AGTR2), which cardiac injury, γδ T cells and neutrophils themselves seem
is known to drive anti-inflammatory responses98,99. to be important sources of IL‑17A production within the
Angiotensin II is part of the renin–angiotensin system, myocardium, and mice that lack IL‑23 have reduced
which has long been known to be a crucial pathological IL‑17A levels in cardiac tissue, reduced neutrophil accu­
pathway in heart disease100,101. Angiotensin II is thought mulation, increased γδ T cell population expansion and
to primarily induce cardiomyocyte hypertrophy and improved infarct healing 110. Indeed, IL‑17A seems to also
increase vascular tone by signalling through AGTR1, be directly toxic to cardiomyocytes, and neutralization
in a process that is amplified by blockade of AGTR2 of IL‑17A improves infarct healing 111 (FIG. 3b).
(as reviewed in REF. 102). After ischaemia–reperfusion Additional work has shown an important link between
injury AGTR2‑expressing CD8+ T cells, inhibit inflam­ IL‑17A production and activation of fibrotic pathways
mation and decrease infarct size by producing IL‑10 in an within the heart that lead to collagen deposition, both fol­
angiotensin II‑dependent fashion, and this subsequently lowing ischaemia and haemodynamic strain112,113. In this
dampens the immune response98. context, and in contrast to the work described above, the

124 | FEBRUARY 2015 | VOLUME 15 www.nature.com/reviews/immunol

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Coxsackievirus B myocardial ischaemic injury that we discussed earlier, in


which either too few or too many infiltrating monocytes
impaired infarct healing 6,78. These data indicate that there
TLR2 CAR DAF are yet-to-be determined, context-dependent factors
or TLR4 that modulate whether a particular cell type or pathway
Cardiomyocyte mediates cardiac tissue repair or damage.
FYN
The activation of cardiac fibroblasts and subse­
MYD88 LCK
or ABL quent extracellular matrix deposition is an important
Viral entry Endosome component of cardiac ischaemic injury and repair.
Interestingly, cardiac fibroblasts can also become acti­
Viral nucleic vated in more subtle forms of cardiac stress that also
acids
lead to impaired cardiac function owing to increased
MDA5 RIG-I
myocardial stiffness (such as hypertension caused
TLR3 TLR7 by infusion of angiotensin II). In this setting there is
Activation of rapid recruitment of neutrophils and monocytes into
cytosolic PRRs TRIF
Activation of the myocardium and also expansion of resident car­
endosomal PRRs
IRAK4 diac macrophage populations5,114–118. Cardiac fibroblasts
become activated, leading to interstitial cardiac fibrosis,
a process dependent on input and possible differen­
tiation from monocytes117. The activation of cardiac
fibroblasts is also amplified by CD4+ and CD8+ T cells
NF-κB IRF3 through an IFNγ-dependent pathway 119,120 (reviewed
p50 p65 in detail in REF. 121).

NF-κB activation
Myocarditis as a model of cardiac inflammation
IRF activation
Myocarditis is an excellent model for dissecting the
inflammatory processes that occur in the heart. It
shows how the balance of innate and adaptive immune
Type I IFN mechanisms that occur following tissue injury deter­
production mine whether there is progression to heart failure or
to repair of cardiac tissue. Myocarditis is most often
induced by infection with viruses, although other infec­
Activation of immuno- Protective tious pathogens can also be involved, such as bacteria or
pathological response, immune response the protozoan parasite Trypanosoma cruzii, which is the
tissue damage
cause of Chagas disease, the commonest cause of heart
Figure 5 | Interaction of coxsackievirus B with the host innate and acquired immune failure in South America122,123. Myocarditis can also
system.  Coxsackievirus B (CVB) engages the internalizingNaturereceptor coxsackievirus
Reviews and
| Immunology result from non-infectious triggers of DAMP release,
adenovirus receptor (CAR), and the receptor-associated tyrosine kinases FYN and ABL which activate the innate immune response leading to
can facilitate viral remodelling of the cell cytoskeleton to promote viral cell entry. This sterile inflammation, such as allergic reactions to drugs
process is further aided by the co‑receptor decay accelerating factor (DAF) and its
or chemicals124.
associated tyrosine kinase LCK. Viral components can be detected by the intracellular
pattern recognition receptors (PRRs) melanoma differentiation-associated gene 5 Myocarditis accounts for about one in nine cases
(MDA5) and retinoic acid-inducible gene I (RIG‑I) leading to the activation of nuclear of heart failure and remains one of the most common
factor‑κB (NF‑κB)‑dependent inflammatory pathways. NF-κB‑dependent responses can reasons for heart transplantation worldwide, as spe­
also be activated following the engagement of CVB with cell surface-expressed Toll-like cific treatments are lacking 125. The most common viral
receptors (TLRs), which signal through the myeloid differentiation primary response triggers of myocarditis include enteroviruses, such as
protein 88 (MYD88) adaptor and downstream signal intermediates such as IL‑1-receptor- coxsackievirus B3 (CVB3) and CVB4, and adenovi­
associated kinase 4 (IRAK4) and TNF receptor-associated factor 6 (TRAF6). These ruses. Virus-induced myocarditis shows a periodicity
pathways facilitate viral proliferation and host immune-mediated tissue damage. By in its prevalence in human populations, which is pos­
contrast, activation of interferon regulatory factor 3 (IRF3) following the triggering of sibly related to herd immunity. European studies also
the TIR-domain-containing adapter-inducing IFNβ (TRIF) pathway by CVB leads to the
show a substantial contribution of parvovirus B19,
production of protective type I interferons (IFNs). There is mutual counter-regulation of
the MYD88 –IRAK4 and TRIF–IRF3 pathways. The subsequent maturation of CD4+ and which shows tropism for endothelial cells and the bone
CD8+ T cell subsets is also detrimental for the host, whereas regulatory T cells are marrow 126. Coxsackieviruses and adenoviruses target
host-protective. Cross-talk between acquired and innate immune signalling pathways host tissues, including the immune, cardiovascular and
also modulates the host response to CVB infection. neurological systems, through their binding and inter­
nalization by coxsackievirus and adenovirus receptor
(CAR) — which is an immune-regulated tight junc­
loss of IL‑23 was demonstrated to impair infarct healing, tion protein127,128. Internalization of the virus is assisted
which was mechanistically linked to a failure to activate by co‑binding to decay accelerating factor (DAF; also
fibroblasts, leading to impaired scar formation and myo­ known as CD55), a ubiquitously expressed host pro­
cardial rupture113. These data are reminiscent of the rela­ tein that inhibits complement activation129 (FIG. 5). Loss
tionship between monocyte influx and tissue healing after of CAR expression in cardiomyocytes prevent cardiac

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 125

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

dysfunction and inflammation following viral infection, The activation of innate immune signalling path­
suggesting that entry into cardiomyocytes is required ways also sets the stage for subsequent T cell maturation
for tissue damage130. However, although viral entry and and participation in the host inflammatory response.
subsequent viral proliferation trigger disease, they are Genetic deletion of LCK, which impairs T cell matura­
not necessarily the primary determinants of disease out­ tion, results in almost complete protection of the host
comes. Both the innate and adaptive immune responses against CVB3 infection131. A similar outcome is observed
have crucial roles in the progression of viral myocar­ following CD4+ or CD8+ T cell subset deletion, or fol­
ditis, as shown by the fact that genetic deletion of the lowing deletion of the leukocyte tyrosine phosphatase
TLR-signalling proteins myeloid differentiation pri­ CD45, which is associated with upregulation of type I
mary response protein 88 (MYD88) or IL‑1R-associated IFNs132,137. Adoptive transfer of regulatory T cells into
kinase 4 (IRAK4), or deletion of the T cell receptor- CVB3‑infected hosts also had a marked protective
associated tyrosine kinase LCK, ameliorates myocardial effect, again associated with a decrease in viral prolif­
inflammation and improves survival in mice, despite eration and upregulation of type I IFNs138. Surprisingly,
increased levels of viral proliferation131–134. there was a general downregulation of molecules asso­
Following viral entry into the target cell, such as an ciated with TLR signalling pathways and even TLR4
immune cell or cardiomyocyte, the virus can engage itself. Moreover, beyond their role during infections,
intracellular NLRs, including RIG‑I and MDA5. The regulatory T cells also promote recovery through an
endosomal degradation of the virus can also lead to the IL‑10‑dependent pathway following ischaemic injury 95.
activation of TLR3 and TLR732,133. The signalling cas­ These data suggest that there is close cross-talk between
cades induced by such PRR activation can have detri­ the regulatory T cell subsets and the innate immune sig­
mental consequences for the host. As stated above, the nalling pathways that operate during both infectious and
MYD88–IRAK4 pathway, showed a very marked protec­ sterile injury.
tive benefit in mice during CVB3 infection. At least four The above progress in understanding myocarditic
different mechanisms are thought to mediate protection processes has moved the field forward from the ear­
in this setting. First, the loss of MYD88–IRAK4 signal­ lier failed attempts to use immunosuppression to treat
ling decreases downstream activation of TNF receptor- patients with myocarditis139. Subsequently, a Phase II
associated factor 6 (TRAF6) and nuclear translocation of trial has demonstrated that in patients with persistent
the NF‑κB complexes, resulting in reduced pro-inflam­ viral proliferation, intravenous delivery of IFNβ clears
matory cytokine production and T cell activation135. the virus and improves symptoms of myocarditis140.
Second, increased IRF3 or IRF5 homodimerization and Targeted immunosuppressive therapy may be indi­
phosphorylation of signal transducers and activators of cated for patients with persistent immune activation, in
transcription 1 (STAT1) and STAT5 occurs in the absence which the viral proliferation phase has already passed.
of MYD88 or IRAK4, leading to increased production of Nevertheless, there are still large gaps in our knowledge,
protective antiviral type I IFNs133–135. Third, IRAK4 dele­ including why there are periodic enteroviral outbreaks
tion facilitates the mobilization of protective CCR5+ in the population. Furthermore, why do some patients
monocytes from the bone marrow into the myocar­ develop severe viral myocarditis that requires transplan­
dium133. Fourth, the absence of MYD88 or IRAK4 leads tation, while others are able to rebalance the immune
to downregulation of CAR expression and decreased response and promptly recover from viral mycocarditis?
viral proliferation135. However, the genetic deletion of Are there biomarkers to permit one to predict an indi­
IRF3, which is activated by the TIR-domain-containing vidual patient’s susceptibility and, for those at high risk,
adaptor-inducing IFNβ (TRIF)-dependent TLR path­ could vaccination be a viable option?
way, leads to much worse outcome during virus-induced
myocarditis, with mice showing increased mortality and Future directions
greater levels of viral proliferation136. Mechanistically, this The ultimate goal in terms of understanding how the
is associated with decreased type I interferon production immune system directs inflammatory and reparative
and increased activation and translocation of NF-κB136. programmes following cardiac injury is the develop­
These observations have several important implica­ ment of therapeutic strategies that promote tissue
tions. The first is that increased expression of viral entry regeneration. Evolutionary pressures drove both the
receptors and enhanced viral proliferation are, paradoxi­ development of primitive phagocytic cells that promote
cally, facilitated by the host innate immune response — tissue growth and wound healing, but also drove the
probably an evolutionarily selected advantage for the development of innate and adaptive immune mecha­
pathogenic virus at the expense of the host. The second nisms that promote survival of the host in the face
is that intracellular immune signalling pathways seem to of infectious threats. Adaptations that both enhance
induce functional responses that regulate trafficking of patho­gen clearance and promote tissue repair may
inflammatory cells, such as CCR5+ monocyte-derived result in a zero-sum game, whereby the very adapta­
macrophages, from the bone marrow. Indeed this is also tions that enhance host defence result in a loss of repar­
the case in sterile inflammation post myocardial infarction, ative potential owing to excessive inflammation. The
in which IRAK4 signalling seems to regulate the traffick­ ability to selectively suppress excessive inflammation
ing and maturation of dendritic cells into the myocardium, while preserving reparative functions is a novel thera­
with subsequent orchestration of host inflammatory peutic avenue that can enhance recovery in patients
responses that dictate tissue remodelling and host survival. with a wide range of cardiovascular diseases.

126 | FEBRUARY 2015 | VOLUME 15 www.nature.com/reviews/immunol

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

1. Ma, X., Cong, P., Hou, X., Edgecombe, G. D. & 24. Arslan, F. et al. Myocardial ischemia/reperfusion injury 48. Youker, K. et al. Neutrophil adherence to isolated
Strausfeld, N. J. An exceptionally preserved arthropod is mediated by leukocytic toll-like receptor‑2 and adult cardiac myocytes. Induction by cardiac lymph
cardiovascular system from the early Cambrian. reduced by systemic administration of a novel anti-toll- collected during ischemia and reperfusion.
Nature Commun. 5, 3560 (2014). like receptor‑2 antibody. Circulation 121, 80–90 J. Clin. Invest. 89, 602–609 (1992).
2. Bier, E. & Bodmer, R. Drosophila, an emerging model (2010). This important early study demonstrated the role
for cardiac disease. Gene 342, 1–11 (2004). 25. Fallach, R. et al. Cardiomyocyte Toll-like receptor 4 is of neutrophils in cardiomyocyte injury.
3. Lozano, R. et al. Global and regional mortality from involved in heart dysfunction following septic shock 49. Entman, M. L. et al. Neutrophil induced oxidative
235 causes of death for 20 age groups in 1990 and or myocardial ischemia. J. Mol. Cell Cardiol. 48, injury of cardiac myocytes. A compartmented system
2010: a systematic analysis for the Global Burden of 1236–1244 (2010). requiring CD11b/CD18–ICAM‑1 adherence.
Disease Study 2010. Lancet 380, 2095–2128 26. Binck, B. W. et al. Bone marrow-derived cells J. Clin. Invest. 90, 1335–1345 (1992).
(2012). contribute to contractile dysfunction in endotoxic 50. Entman, M. L. et al. Neutrophil adherence to isolated
4. Pinto, A. R. et al. An abundant tissue macrophage shock. Am. J. Physiol. Heart Circ. Physiol. 288, adult canine myocytes. Evidence for a
population in the adult murine heart with a distinct H577–H583 (2005). CD18‑dependent mechanism. J. Clin. Invest. 85,
alternatively-activated macrophage profile. 27. Oyama, J. et al. Reduced myocardial ischemia- 1497–1506 (1990).
PLoS ONE. 7, e36814 (2012). reperfusion injury in toll-like receptor 4‑deficient mice. 51. Tyagi, S., Klickstein, L. B. & Nicholson-Weller, A.
5. Epelman, S. et al. Embryonic and adult-derived Circulation 109, 784–789 (2004). C5a‑stimulated human neutrophils use a subset of β2
resident cardiac macrophages are maintained through 28. Tavener, S. A. et al. Immune cell Toll-like receptor 4 integrins to support the adhesion-dependent phase of
distinct mechanisms at steady state and during is required for cardiac myocyte impairment during superoxide production. J. Leukoc. Biol. 68, 679–686
inflammation. Immunity 40, 91–104 (2014). endotoxemia. Circ. Res. 95, 700–707 (2004). (2000).
This is the first study to show that cardiac 29. Oka, T. et al. Mitochondrial DNA that escapes from 52. Kawakami, R. et al. Overexpression of brain
macrophages are not a single population but are autophagy causes inflammation and heart failure. natriuretic peptide facilitates neutrophil infiltration
composed of distinct subsets, with different origins Nature 485, 251–255 (2012). and cardiac matrix metalloproteinase‑9 expression
and functions. This is the first study to show that endogenous after acute myocardial infarction. Circulation 110,
6. Nahrendorf, M. et al. The healing myocardium DAMPs are released from the myocardium during 3306–3312 (2004).
sequentially mobilizes two monocyte subsets with haemodynamic strain, a process that impairs 53. Romson, J. L. et al. Reduction of the extent of
divergent and complementary functions. J. Exp. Med. cardiac function. ischemic myocardial injury by neutrophil depletion in
204, 3037–3047 (2007). 30. Geddes, K., Magalhaes, J. G. & Girardin, S. E. the dog. Circulation 67, 1016–1023 (1983).
This report shows the initial characterization of Unleashing the therapeutic potential of NOD-like 54. Jolly, S. R. et al. Reduction of myocardial infarct size
cardiac macrophages in the resting heart and after receptors. Nature Rev. Drug Discov. 8, 465–479 by neutrophil depletion: effect of duration of occlusion.
ischaemic injury. (2009). Am. Heart J. 112, 682–690 (1986).
7. Swirski, F. K. & Nahrendorf, M. Leukocyte behavior in 31. Kawaguchi, M. et al. Inflammasome activation of 55. Van Furth, R. & Cohn, Z. A. The origin and kinetics
atherosclerosis, myocardial infarction, and heart cardiac fibroblasts is essential for myocardial of mononuclear phagocytes. J. Exp. Med. 128,
failure. Science 339, 161–166 (2013). ischemia/reperfusion injury. Circulation 123, 415–435 (1968).
8. Choi, J. H. et al. Identification of antigen-presenting 594–604 (2011). 56. Ginhoux, F. et al. Fate mapping analysis reveals that
dendritic cells in mouse aorta and cardiac valves. 32. McCartney, S. A. et al. RNA sensor-induced type I IFN adult microglia derive from primitive macrophages.
J. Exp. Med. 206, 497–505 (2009). prevents diabetes caused by a β cell-tropic virus in Science 330, 841–845 (2010).
9. Frangogiannis, N. G. et al. Resident cardiac mast cells mice. J. Clin. Invest. 121, 1497–1507 (2011). 57. Schulz, C. et al. A lineage of myeloid cells independent
degranulate and release preformed TNF-α, initiating 33. Philip, J., Xu, Z., Bowles, N. E. & Vallejo, J. G. of Myb and hematopoietic stem cells. Science 336,
the cytokine cascade in experimental canine Cardiac-specific overexpression of melanoma 86–90 (2012).
myocardial ischemia/reperfusion. Circulation 98, differentiation-associated gene‑5 protects mice from 58. Yona, S. et al. Fate mapping reveals origins and
699–710 (1998). lethal viral myocarditis. Circ. Heart Fail. 6, 326–334 dynamics of monocytes and tissue macrophages
10. Zouggari, Y. et al. B lymphocytes trigger monocyte (2013). under homeostasis. Immunity. 38, 79–91 (2013).
mobilization and impair heart function after acute 34. Lech, M. et al. Quantitative expression of C‑type lectin 59. Hashimoto, D. et al. Tissue-resident macrophages
myocardial infarction. Nature Med. 19, 1273–1280 receptors in humans and mice. Int. J. Mol. Sci. 13, self-maintain locally throughout adult life with
(2013). 10113–10131 (2012). minimal contribution from circulating monocytes.
11. Saxena, A. et al. Regulatory T cells are recruited in 35. Frangogiannis, N. G. & Entman, M. L. Chemokines in Immunity. 38, 792–804 (2013).
the infarcted mouse myocardium and may modulate myocardial ischemia. Trends Cardiovasc. Med. 15, 60. Guilliams, M. et al. Alveolar macrophages develop
fibroblast phenotype and function. Am. J. Physiol. 163–169 (2005). from fetal monocytes that differentiate into long-lived
Heart Circ. Physiol. 307, H1233–H1242 (2014). 36. Frangogiannis, N. G. The mechanistic basis of infarct cells in the first week of life via GM‑CSF. J. Exp. Med.
12. Roger, V. L. et al. Heart disease and stroke healing. Antioxid. Redox. Signal. 8, 1907–1939 (2006). 210, 1977–1992 (2013).
statistics—2012 update: a report from the American 37. Chakraborti, T., Mandal, A., Mandal, M., Das, S. & 61. Jakubzick, C. et al. Minimal differentiation of classical
Heart Association. Circulation 125, e2–e220 Chakraborti, S. Complement activation in heart monocytes as they survey steady-state tissues and
(2012). diseases. Role of oxidants. Cell Signal. 12, 607–617 transport antigen to lymph nodes. Immunity. 39,
13. Jacoby, D. & McKenna, W. J. Genetics of inherited (2000). 599–610 (2013).
cardiomyopathy. Eur. Heart J. 33, 296–304 (2012). 38. Foreman, K. E., Glovsky, M. M., Warner, R. L., 62. Hanna, R. N. et al. The transcription factor NR4A1
14. Sangiuliano, B., Perez, N. M., Moreira, D. F. & Horvath, S. J. & Ward, P. A. Comparative effect of C3a (Nur77) controls bone marrow differentiation and the
Belizario, J. E. Cell death-associated molecular- and C5a on adhesion molecule expression on survival of Ly6C- monocytes. Nature Immunol. 12,
pattern molecules: inflammatory signaling and neutrophils and endothelial cells. Inflammation 20, 778–785 (2011).
control. Mediators Inflamm. 2014, 821043 1–9 (1996). 63. Hettinger, J. et al. Origin of monocytes and
(2014). 39. Bhattacharya, K. et al. Mast cell deficient W/Wv mice macrophages in a committed progenitor.
15. Matzinger, P. Tolerance, danger, and the extended have lower serum IL‑6 and less cardiac tissue necrosis Nature Immunol. 14, 821–830 (2013).
family. Annu. Rev. Immunol. 12, 991–1045 (1994). than their normal littermates following myocardial 64. Ingersoll, M. A. et al. Comparison of gene expression
16. Janeway, C. A. Jr. Approaching the asymptote? ischemia-reperfusion. Int. J. Immunopathol. profiles between human and mouse monocyte
Evolution and revolution in immunology. Pharmacol. 20, 69–74 (2007). subsets. Blood 115, e10–e19 (2010).
Cold Spring Harb. Symp. Quant. Biol. 54 Pt. 1, 1–13 40. Ayach, B. B. et al. Stem cell factor receptor induces 65. Auffray, C. et al. Monitoring of blood vessels and
(1989). progenitor and natural killer cell-mediated cardiac tissues by a population of monocytes with patrolling
17. Mann, D. L., Topkara, V. K., Evans, S. & Barger, P. M. survival and repair after myocardial infarction. behavior. Science 317, 666–670 (2007).
Innate immunity in the adult mammalian heart: for Proc. Natl Acad. Sci. USA 103, 2304–2309 (2006). 66. Carlin, L. M. et al. Nr4a1‑dependent Ly6Clow
whom the cell tolls. Trans. Am. Clin. Climatol. Assoc. 41. Waskow, C., Paul, S., Haller, C., Gassmann, M. & monocytes monitor endothelial cells and orchestrate
121, 34–50 (2010). Rodewald, H. R. Viable c‑Kit(W/W) mutants reveal their disposal. Cell 153, 362–375 (2013).
18. Mezzaroma, E. et al. The inflammasome promotes pivotal role for c‑kit in the maintenance of 67. Frangogiannis, N. G. et al. Critical role of monocyte
adverse cardiac remodeling following acute myocardial lymphopoiesis. Immunity 17, 277–288 (2002). chemoattractant protein‑1/CC chemokine ligand 2
infarction in the mouse. Proc. Natl Acad. Sci. USA 42. Dreyer, W. J. et al. Kinetics of C5a release in cardiac in the pathogenesis of ischemic cardiomyopathy.
108, 19725–19730 (2011). lymph of dogs experiencing coronary artery ischemia- Circulation 115, 584–592 (2007).
19. Takeuchi, O. & Akira, S. Pattern recognition receptors reperfusion injury. Circ. Res. 71, 1518–1524 (1992). An important early study showing that CCR2
and inflammation. Cell 140, 805–820 (2010). 43. Newburger, P. E. & Dale, D. C. Evaluation and deficiency limits cardiac injury. Later studies
20. Rathinam, V. A. et al. The AIM2 inflammasome is management of patients with isolated neutropenia. would suggest this was due to a lack of blood
essential for host defense against cytosolic bacteria Semin. Hematol. 50, 198–206 (2013). monocytes.
and DNA viruses. Nature Immunol. 11, 395–402 44. Singh, M. & Saini, H. K. Resident cardiac mast cells 68. Hilgendorf, I. et al. Ly‑6Chigh monocytes depend on
(2010). and ischemia-reperfusion injury. J. Cardiovasc. Nr4a1 to balance both inflammatory and reparative
21. Frantz, S. et al. Toll4 (TLR4) expression in cardiac Pharmacol. Ther. 8, 135–148 (2003). phases in the infarcted myocardium. Circ. Res.114,
myocytes in normal and failing myocardium. 45. McDonald, B. et al. Intravascular danger signals 1611–1622 (2014).
J. Clin. Invest. 104, 271–280 (1999). guide neutrophils to sites of sterile inflammation. 69. Leuschner, F. et al. Angiotensin-converting enzyme
22. Birks, E. J. et al. Increased toll-like receptor 4 in the Science 330, 362–366 (2010). inhibition prevents the release of monocytes from
myocardium of patients requiring left ventricular assist 46. Li, W. et al. Intravital 2‑photon imaging of leukocyte their splenic reservoir in mice with myocardial
devices. J. Heart Lung Transplant. 23, 228–235 trafficking in beating heart. J. Clin. Invest. 122, infarction. Circ. Res. 107, 1364–1373 (2010).
(2004). 2499–2508 (2012). 70. Swirski, F. K. et al. Identification of splenic reservoir
23. Kashiwagi, M. et al. Differential expression of Toll-like 47. Gwechenberger, M. et al. Cardiac myocytes produce monocytes and their deployment to inflammatory
receptor 4 and human monocyte subsets in acute interleukin‑6 in culture and in viable border zone of sites. Science 325, 612–616 (2009).
myocardial infarction. Atherosclerosis 221, 249–253 reperfused infarctions. Circulation 99, 546–551 This is the first study to show that the spleen can
(2012). (1999). be a source of monocytes following cardiac injury.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 127

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

71. Leuschner, F. et al. Rapid monocyte kinetics in acute 93. Hofmann, U. et al. Activation of CD4+ T lymphocytes 117. Haudek, S. B. et al. Monocytic fibroblast precursors
myocardial infarction are sustained by extramedullary improves wound healing and survival after mediate fibrosis in angiotensin-II‑induced cardiac
monocytopoiesis. J. Exp. Med. 209, 123–137 experimental myocardial infarction in mice. hypertrophy. J. Mol. Cell Cardiol. 49, 499–507
(2012). Circulation 125, 1652–1663 (2012). (2010).
72. Molawi, K. et al. Progressive replacement of embryo- 94. Matsumoto, K. et al. Regulatory T lymphocytes 118. Sadoshima, J. & Izumo, S. Molecular characterization
derived cardiac macrophages with age. J. Exp. Med. attenuate myocardial infarction-induced ventricular of angiotensin II-induced hypertrophy of cardiac
211, 2151–2158 (2014). remodeling in mice. Int. Heart J. 52, 382–387 (2011). myocytes and hyperplasia of cardiac fibroblasts.
73. Bracey, N. A. et al. Mitochondrial NLRP3 protein 95. Dobaczewski, M., Xia, Y., Bujak, M., Critical role of the AT1 receptor subtype. Circ. Res.
induces reactive oxygen species to promote Smad Gonzalez-Quesada, C. & Frangogiannis, N. G. 73, 413–423 (1993).
protein signaling and fibrosis independent from the CCR5 signaling suppresses inflammation and reduces 119. Marko, L. et al. Interferon-γ signaling inhibition
inflammasome. J. Biol. Chem. 289, 19571–19584 adverse remodeling of the infarcted heart, mediating ameliorates angiotensin II‑induced cardiac damage.
(2014). recruitment of regulatory T cells. Am. J. Pathol. 176, Hypertension 60, 1430–1436 (2012).
74. Bracey, N. A. et al. The Nlrp3 inflammasome 2177–2187 (2010). 120. Han, Y. L. et al. Reciprocal interaction between
promotes myocardial dysfunction in structural 96. Yang, Z. et al. Myocardial infarct-sparing effect of macrophages and T cells stimulates IFN-γ and
cardiomyopathy through interleukin‑1β. adenosine A2A receptor activation is due to its action MCP‑1 production in Ang II‑induced cardiac
Exp. Physiol. 98, 462–472 (2013). on CD4+ T lymphocytes. Circulation 114, 2056–2064 inflammation and fibrosis. PLoS ONE. 7, e35506
75. Dunay, I. R. et al. Gr1+ inflammatory monocytes (2006). (2012).
are required for mucosal resistance to the pathogen 97. Clynes, R. Protective mechanisms of IVIG. 121. Crawford, J. R., Haudek, S. B., Cieslik, K. A., Trial, J. &
Toxoplasma gondii. Immunity 29, 306–317 Curr. Opin. Immunol. 19, 646–651 (2007). Entman, M. L. Origin of developmental precursors
(2008). 98. Curato, C. et al. Identification of noncytotoxic and dictates the pathophysiologic role of cardiac
76. Kim, Y. G. et al. The Nod2 sensor promotes intestinal IL‑10‑producing CD8+AT2R+ T cell population in fibroblasts. J. Cardiovasc. Transl. Res. 5, 749–759
pathogen eradication via the chemokine response to ischemic heart injury. J. Immunol. 185, (2012).
CCL2‑dependent recruitment of inflammatory 6286–6293 (2010). 122. Coura, J. R. & Borges-Pereira, J. Chagas disease:
monocytes. Immunity 34, 769–780 (2011). 99. Kaschina, E. et al. Angiotensin II type 2 receptor 100 years after its discovery. A systemic review.
77. van Amerongen, M. J., Harmsen, M. C., stimulation: a novel option of therapeutic interference Acta Trop. 115, 5–13 (2010).
van Rooijen, N., Petersen, A. H. & van Luyn, M. J. with the renin-angiotensin system in myocardial 123. Kindermann, I. et al. Update on myocarditis.
Macrophage depletion impairs wound healing and infarction? Circulation 118, 2523–2532 (2008). J. Am. Coll. Cardiol. 59, 779–792 (2012).
increases left ventricular remodeling after 100. Cohn, J. N. & Tognoni, G. A randomized trial of the 124. Neu, N. et al. Cardiac myosin induces myocarditis in
myocardial injury in mice. Am. J. Pathol. 170, angiotensin-receptor blocker valsartan in chronic heart genetically predisposed mice. J. Immunol. 139,
818–829 (2007). failure. N. Engl. J. Med. 345, 1667–1675 (2001). 3630–3636 (1987).
78. Panizzi, P. et al. Impaired infarct healing in 101. Dickstein, K. & Kjekshus, J. Effects of losartan and 125. Sagar, S., Liu, P. P. & Cooper, L. T. Jr. Myocarditis.
atherosclerotic mice with Ly‑6C(hi) monocytosis. captopril on mortality and morbidity in high-risk Lancet 379, 738–747 (2012).
J. Am. Coll. Cardiol. 55, 1629–1638 (2010). patients after acute myocardial infarction: the 126. Kuhl, U. et al. High prevalence of viral genomes and
79. Dewald, O. et al. CCL2/monocyte chemoattractant OPTIMAAL randomised trial. Optimal Trial in multiple viral infections in the myocardium of adults
protein‑1 regulates inflammatory responses critical to Myocardial Infarction with Angiotensin II Antagonist with “idiopathic” left ventricular dysfunction.
healing myocardial infarcts. Circ. Res. 96, 881–889 Losartan. Lancet 360, 752–760 (2002). Circulation 111, 887–893 (2005).
(2005). 102. Mehta, P. K. & Griendling, K. K. Angiotensin II cell 127. Martino, T. A. et al. The coxsackie-adenovirus receptor
80. Leuschner, F. et al. Therapeutic siRNA silencing in signaling: physiological and pathological effects in the (CAR) is used by reference strains and clinical isolates
inflammatory monocytes in mice. Nature Biotech. 29, cardiovascular system. Am. J. Physiol. Cell Physiol. representing all six serotypes of coxsackievirus group
1005–1010 (2011). 292, C82–C97 (2007). B and by swine vesicular disease virus. Virology 271,
81. Serbina, N. V. & Pamer, E. G. Monocyte emigration 103. Bouchentouf, M. et al. Induction of cardiac 99–108 (2000).
from bone marrow during bacterial infection requires angiogenesis requires killer cell lectin-like receptor 1 128. Coyne, C. B. & Bergelson, J. M. Virus-induced Abl
signals mediated by chemokine receptor CCR2. and α4β7 integrin expression by NK cells. J. Immunol. and Fyn kinase signals permit coxsackievirus entry
Nature Immunol. 7, 311–317 (2006). 185, 7014–7025 (2010). through epithelial tight junctions. Cell 124, 119–131
82. Zhou, L. et al. Monocyte chemoattractant protein‑1 104. Fadok, V. A. et al. Macrophages that have ingested (2006).
induces a novel transcription factor that causes apoptotic cells in vitro inhibit proinflammatory This was the first study to show how cardiotropic
cardiac myocyte apoptosis and ventricular cytokine production through autocrine/paracrine viruses penetrate host epithelial barriers.
dysfunction. Circ. Res. 98, 1177–1185 (2006). mechanisms involving TGF-β, PGE2, and PAF. 129. Liu, P. P. & Opavsky, M. A. Viral myocarditis:
83. Aurora, A. B. et al. Macrophages are required for J. Clin. Invest. 101, 890–898 (1998). receptors that bridge the cardiovascular with the
neonatal heart regeneration. J. Clin. Invest. 124, 105. Voll, R. E. et al. Immunosuppressive effects of immune system? Circ. Res. 86, 253–254 (2000).
1382–1392 (2014). apoptotic cells. Nature 390, 350–351 (1997). 130. Kallewaard, N. L. et al. Tissue-specific deletion
84. Porrello, E. R. et al. Transient regenerative potential of 106. Xue, J. et al. Transcriptome-based network analysis of the coxsackievirus and adenovirus receptor
the neonatal mouse heart. Science 331, 1078–1080 reveals a spectrum model of human macrophage protects mice from virus-induced pancreatitis
(2011). activation. Immunity 40, 274–288 (2014). and myocarditis. Cell Host Microbe 6, 91–98
This is the first study to show that the mammalian 107. Stark, M. A. et al. Phagocytosis of apoptotic (2009).
neonatal heart can regenerate fully, similarly to neutrophils regulates granulopoiesis via IL‑23 and 131. Liu, P. et al. The tyrosine kinase p56lck is essential
what is observed in more primitive organisms. IL‑17. Immunity 22, 285–294 (2005). in coxsackievirus B3‑mediated heart disease.
85. Lavine, K. et al. Distinct macrophage lineages 108. Tan, W. et al. IL‑17F/IL‑17R interaction stimulates Nature Med. 6, 429–434 (2000).
contribute to disparate patterns of cardiac recovery granulopoiesis in mice. Exp. Hematol. 36, The first study to detail the mechanisms through
and remodeling in the neonatal and adult heart. 1417–1427 (2008). which cardiotropic viruses mediate intracellular
Proc. Natl Acad. Sci. 111, 16029–16034 (2014). 109. Schwarzenberger, P. et al. Requirement of signalling events and cell injury.
This is the first study to show that resident endogenous stem cell factor and granulocyte-colony- 132. Irie-Sasaki, J. et al. CD45 is a JAK phosphatase and
neonatal heart macrophages, not recruited stimulating factor for IL‑17‑mediated granulopoiesis. negatively regulates cytokine receptor signalling.
monocytes, have a key role in neonatal heart J. Immunol. 164, 4783–4789 (2000). Nature 409, 349–354 (2001).
regeneration. Similar findings were seen in 110. Yan, X. et al. Deleterious effect of the IL‑23/IL‑17A 133. Valaperti, A. et al. Innate immune interleukin‑1
reference 83. axis and γδT cells on left ventricular remodeling after receptor-associated kinase 4 exacerbates viral
86. Marodi, L. Neonatal innate immunity to infectious myocardial infarction. J. Am. Heart Assoc. 1, myocarditis by reducing CCR5+ CD11b+ monocyte
agents. Infect. Immun. 74, 1999–2006 (2006). e004408 (2012). migration and impairing interferon production.
87. Munoz-Espin, D. et al. Programmed cell senescence 111. Liao, Y. H. et al. Interleukin‑17A contributes to Circulation 128, 1542–1554 (2013).
during mammalian embryonic development. myocardial ischemia/reperfusion injury by regulating 134. Riad, A. et al. Myeloid differentiation factor‑88
Cell 155, 1104–1118 (2013). cardiomyocyte apoptosis and neutrophil infiltration. contributes to TLR9‑mediated modulation of acute
88. Storer, M. et al. Senescence is a developmental J. Am. Coll. Cardiol. 59, 420–429 (2012). coxsackievirus B3‑induced myocarditis in vivo.
mechanism that contributes to embryonic growth 112. Feng, W. et al. IL‑17 induces myocardial fibrosis and Am. J. Physiol. Heart Circ. Physiol. 298,
and patterning. Cell 155, 1119–1130 (2013). enhances RANKL/OPG and MMP/TIMP signaling in H2024–H2031 (2010).
89. Wan, E. et al. Enhanced efferocytosis of apoptotic isoproterenol-induced heart failure. Exp. Mol. Pathol. 135. Fuse, K. et al. Myeloid differentiation factor‑88 plays a
cardiomyocytes through myeloid-epithelial- 87, 212–218 (2009). crucial role in the pathogenesis of Coxsackievirus
reproductive tyrosine kinase links acute inflammation 113. Savvatis, K. et al. Interleukin‑23 deficiency leads to B3‑induced myocarditis and influences type I
resolution to cardiac repair after infarction. impaired wound healing and adverse prognosis after interferon production. Circulation 112, 2276–2285
Circ. Res. 113, 1004–1012 (2013). myocardial infarction. Circ. Heart Fail. 7, 161–171 (2005).
90. Gautier, E. L. et al. Gene-expression profiles and (2014). 136. Holm, G. H. et al. Interferon regulatory factor 3
transcriptional regulatory pathways that underlie the 114. Haudek, S. B. et al. Rho kinase‑1 mediates cardiac attenuates reovirus myocarditis and contributes to
identity and diversity of mouse tissue macrophages. fibrosis by regulating fibroblast precursor cell viral clearance. J. Virol. 84, 6900–6908 (2010).
Nature Immunol. 13, 1118–1128 (2012). differentiation. Cardiovasc. Res. 83, 511–518 (2009). 137. Opavsky, M. A. et al. Susceptibility to myocarditis is
91. Mounier, R. et al. AMPKα1 regulates macrophage 115. Lim, D. S. et al. Angiotensin II blockade reverses dependent on the response of αβ T lymphocytes to
skewing at the time of resolution of inflammation myocardial fibrosis in a transgenic mouse model of coxsackieviral infection. Circ. Res. 85, 551–558
during skeletal muscle regeneration. Cell. Metab. 18, human hypertrophic cardiomyopathy. Circulation 103, (1999).
251–264 (2013). 789–791 (2001). 138. Shi, Y. et al. Regulatory T cells protect mice against
92. Arnold, L. et al. Inflammatory monocytes recruited 116. Xu, J. et al. CCR2 mediates the uptake of bone coxsackievirus-induced myocarditis through the
after skeletal muscle injury switch into marrow-derived fibroblast precursors in angiotensin transforming growth factor β-coxsackie-adenovirus
antiinflammatory macrophages to support II‑induced cardiac fibrosis. Am. J. Physiol. Heart Circ. receptor pathway. Circulation 121, 2624–2634
myogenesis. J. Exp. Med. 204, 1057–1069 (2007). Physiol. 301, H538–H547 (2011). (2010).

128 | FEBRUARY 2015 | VOLUME 15 www.nature.com/reviews/immunol

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

139. Mason, J. W. et al. A clinical trial of immunosuppressive 144. Buchmann, K. Evolution of innate immunity: development, tissue injury and regeneration.
therapy for myocarditis. The Myocarditis Treatment clues from invertebrates via fish to mammals. Int. J. Dev. Biol. 55, 495–503 (2011).
Trial Investigators. N. Engl. J. Med. 333, 269–275 Front. Immunol. 5, 459 (2014). 150. Fantin, A. et al. Tissue macrophages act as cellular
(1995). 145. Epelman, S., Lavine, K. J. & Randolph, G. J. chaperones for vascular anastomosis downstream
140. Kuhl, U. et al. Interferon-β treatment eliminates Origin and functions of tissue macrophages. of VEGF-mediated endothelial tip cell induction.
cardiotropic viruses and improves left ventricular Immunity 41, 21–35 (2014). Blood 116, 829–840 (2010).
function in patients with myocardial persistence of 146. Dai, X. M. et al. Targeted disruption of the mouse 151. Arnold, T. & Betsholtz, C. The importance of microglia
viral genomes and left ventricular dysfunction. colony-stimulating factor 1 receptor gene results in in the development of the vasculature in the central
Circulation 107, 2793–2798 (2003). osteopetrosis, mononuclear phagocyte deficiency, nervous system. Vasc. Cell 5, 4 (2013).
141. Poss, K. D., Wilson, L. G. & Keating, M. T. increased primitive progenitor cell frequencies, and 152. Lobov, I. B. et al. WNT7b mediates macrophage-
Heart regeneration in zebrafish. Science 298, reproductive defects. Blood 99, 111–120 (2002). induced programmed cell death in patterning
2188–2190 (2002). 147. McKercher, S. R. et al. Targeted disruption of the of the vasculature. Nature 437, 417–421 (2005).
142. Laube, F., Heister, M., Scholz, C., Borchardt, T. & PU.1 gene results in multiple hematopoietic 153. Naqvi, N. et al. A proliferative burst during
Braun, T. Re‑programming of newt cardiomyocytes is abnormalities. EMBO J. 15, 5647–5658 (1996). preadolescence establishes the final cardiomyocyte
induced by tissue regeneration. J. Cell Sci. 119, 148. Wiktor-Jedrzejczak, W. et al. Total absence of colony- number. Cell 157, 795–807 (2014).
4719–4729 (2006). stimulating factor 1 in the macrophage-deficient 154. Bergmann, O. et al. Evidence for cardiomyocyte
143. Godwin, J. W., Pinto, A. R. & Rosenthal, N. A. osteopetrotic (op/op) mouse. Proc. Natl Acad. Sci. renewal in humans. Science 324, 98–102 (2009).
Macrophages are required for adult salamander USA 87, 4828–4832 (1990).
limb regeneration. Proc. Natl Acad. Sci. USA 110, 149. Nucera, S., Biziato, D. & De, P. M. The interplay Competing interests statement
9415–9420 (2013). between macrophages and angiogenesis in The authors declare no competing interests.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | FEBRUARY 2015 | 129

© 2015 Macmillan Publishers Limited. All rights reserved


Copyright of Nature Reviews Immunology is the property of Nature Publishing Group and its
content may not be copied or emailed to multiple sites or posted to a listserv without the
copyright holder's express written permission. However, users may print, download, or email
articles for individual use.

S-ar putea să vă placă și