Sei sulla pagina 1di 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.

Toronto Medical Discovery


Tower, 101 College Street,
TMDT 3903 Toronto, Ontario,
M5G 1L7, Canada.
2
University of Ottawa Heart
Institute, 40 Ruskin Street,
Ottawa, Ontario, K1Y 4W7,
Canada.
3
Center for Cardiovascular
Research, Division of
Cardiology, Department of
Medicine, Washington
University School of Medicine,
St. Louis, Missouri 63110,
USA.
Correspondence to S.E.
e-mail: slava.epelman@
uhnresearch.ca
doi:10.1038/nri3800
1

The cardiovascular system evolved ~600 million years


ago as a means to transport nutrients and cells within
multicellular organisms1. Primitive organisms such as
Drosophila possess a single chamber that functions as
both a pumping tube and a simple vascular system2. More
complex organisms have compartmentalized cardio
vascular systems, comprising venous and arterial vascular
systems that are connected to a multi-chamber muscular
myocardium that continually receives and ejects blood
components. Despite the more complex nature of the
mammalian cardiovascular system, its primary functions
remain the same, and its importance to health and dis
ease is highlighted by the fact that cardiovascular disease
is the leading cause of death worldwide, with an increasing
burden over the past decade3. Therefore, understanding
both how cardiac tissue is injured and how cardiac tissue
regenerates is of prime importance to globalhealth.
The immune system evolved to enable host defence
against invading pathogens but also to promote tissue
growth and repair during development or following sterile
tissue injury, such as that which can occur within the
myocardium (BOX1). In this Review, we detail the roles of
individual immune cell subsets and immune signalling
pathways in both sterile and infectious cardiac injury.
We also use examples from the cardiac system to suggest
the idea that the immune system evolved to promote
tissue homeostasis, but that this beneficial activity comes
at a cost of increased bystander damage when the
immune system overreacts to internalinjury.

Immune anatomy of the resting heart


As in most tissues, the primary immune cells that
reside in the heart are macrophages, which are typically
observed near endothelial cells or within the interstitial
space, while very few, if any, monocytes are found within
cardiac tissue47 (FIG.1). Sparse populations of dendritic
cells (DCs) have been found within cardiac tissue, some
of which are localized in cardiac valves, where they
presumably sample antigens5,8. Mast cells are found in
resting cardiac tissue, and are thought to be important
early triggers of immune responses9. A small number of
Bcells and regulatory T cell subsets are also present in
cardiac tissue in resting conditions, whereas neutrophils
are typically not found within non-inflamed cardiac tis
sue5,10,11. Whether from sterile or infectious triggers,
cardiac tissue injury initiates a dynamic cellular cascade
that initially activates resident immune cells, and over
time this evolves in a coordinated manner that leads to
the recruitment of diverse leukocyte populations into
inflamed tissue.
Mechanisms of cardiac injury
The myocardium can be injured by various pathophysio
logical processes, which can be grouped broadly into
ischaemic and non-ischaemic aetiologies. In terms of
global disease burden, ischaemic injury is the primary
pathophysiological mechanism of injury 3,12. Occlusion
of a coronary vessel after acute plaque rupture can lead
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
A careful examination of examples from the natural world reveals temporal and
phylogenetic characteristics that predict the ability of tissues to regenerate in diverse
organisms. More primitive organisms such as invertebrates, reptiles and amphibians
have a striking regenerative potential when compared with mammals. For example,
both the zebrafish and newt heart can fully regrow after experiencing substantial
injury, and the salamander can fully regrow its limbs after amputation functions that
are not possessed by adult mammals141143. Very young mammals also retain this
regenerative capacity. The neonatal heart can fully regenerate after apical resection
of the left ventricle, myocardial infarction or necrotic injury, but this capacity is lost
after the first weeks of life8385. One important similarity between more primitive
organisms and very young mammals is a more limited (primitive) immune system144.
The macrophage is a specialized mononuclear phagocyte that resides in all tissues
from the earliest stages of development57,145. Loss of macrophages leads to
abnormalities in growth of complex vascular and neuronal networks, increased
mortality and stunted growth146152. Beyond supporting growth, macrophages also have
an important and more generalized role in the clearance of senescent cells during
embryonic development87,88. Importantly, non-selective depletion of all macrophages
impairs the ability of primitive organisms and young mammals to regenerate,
highlighting the crucial role these cells have in tissue growth and repair8385. Although
macrophages possess important regenerative functions, they can also mediate
pathology. Excessive expansion of macrophage populations during ischaemic injury
impairs tissue healing, indicating that certain macrophage subsets can interfere with
the regenerative process78. Understanding when macrophages do or do not promote
tissue repair is a crucial first step if we are to understand why the adult human heart has
only a limited regenerative capacity.

infarction occurs if blood flow is not restored, result


ing in permanent anoxic and low-nutrient tissue injury.
If blood flow is re-established to ischaemic (yet viable)
tissue, inflammatory reperfusion injury can also occur
(discussed below). Non-ischaemic cardiomyopathy is
a composite diagnosis that includes myocarditis that
occurs after viral or bacterial infections or toxin admin
istration. In addition, there are cardiomyopathies that
develop secondary to chronic hypertension. All these
forms of injury are influenced by genetic predisposition,
which can itself lead to early onset cardiac dysfunction13.
Whether through acute ischaemic injury or through
the gradual impairment of cardiac function following a
variety of clinical pathologies, irreversible heart failure
often develops. As we are coming to understand, the
immune system can contribute both during the initial
insult and during the chronic phase of cardiac injury
and despite the investment of substantial research
into understanding the contribution of immune cells
to cardiac injury and repair, much remains unknown.

Innate sensing of cardiac injury


Mammalian hearts use both innate and adaptive
immunity to respond to tissue injury resulting from
pathogens or environmental injury (for example,
ischaemia or haemodynamic overloading). Resident
cardiac immune cells are triggered by the detection
of pathogen-associated molecular patterns (PAMPs)
or damage-associated molecular patterns (DAMPs) by
a fixed number of germline encoded pattern recogni
tion receptors (PRRs). Classic examples of pathogenassociated molecular patterns include the lipopolysac
charides (LPSs) of Gram-negative microorganisms,
the teichoic acids of Gram-positive microorganisms,

the zymosans of yeast, the glycolipids of mycobacterium,


or the double-stranded RNAs of viruses (FIG.2). More
recently it has become clear that cardiac PRRs also rec
ognize the molecular patterns of endogenous material
released by dying or injured myocardial cells. Cells that
die by accidental necrosis, regulated necrosis (that is,
necroptosis) and/or secondary apoptosis release their
cytosolic contents into the extracellular space, thereby
initiating a brisk inflammatory response through the
engagement of an ensemble of extracellular or intra
cellular PRRs14. The time course of the inflamma
tory response that ensues following tissue injury is
remarkably consistent, irrespective of the specific cause
of the injury, and it is associated with the rapid influx of
neutrophils, and subsequently monocytes, into the area
of tissue injury. This inflammatory response has been
referred to as sterile inflammation, insofar as the inflam
mation following tissue injury occurs in the absence of
a known pathogenic infection15,16.
Many PRRs encountering PAMPs and DAMPs trig
ger signalling cascades that activate nuclear factorB
(NFB), activator protein 1 (AP-1), and interferon
regulatory factor (IRF) transcription factors, that in
turn regulate target genes that encode pro-inflammatory
cytokines and interferons (IFNs) in the heart 17. Another
subset of PRRs in the heart trigger a distinct proinflammatory mechanism that requires assembly of
cytosolic protein complexes called inflammasomes18.
Canonical inflammasomes convert pro-caspase 1 into
the catalytically active caspase 1 protease that is respon
sible the production of interleukin1 (IL1) and
IL18 cytokines that are sufficient to trigger
inflammatory responses in the heart 18.
PRRs can be subdivided into two major classes
based on their subcellular localization. Toll-like recep
tors (TLRs) and Ctype lectin receptors are found on
plasma membranes or endosomes, where they can
detect the presence of PAMPs or DAMPs. A second
class of PRRs resides in intracellular compartments,
and includes retinoic-acid inducible gene I (RIG-I)like
receptors (RLRs), NOD-like receptors (NLRs) and
absent-inmelanoma 2 (AIM2) receptors19,20.
Little is known of how TLR expression is regulated in
the heart. However, TLR4 seems to be upregulated in the
failing human heart and on circulating monocytes at
the time of myocardial infarction2123. In animal studies,
loss of TLR4 in haematopoietic cells is protective
in the setting of sepsis-induced cardiac dysfunction and
the loss of TLR2 in haematopoietic cells is protective
during ischaemic injury 2426. The loss of TLR4 is also
protective following ischaemic injury, although it is not
known whether this is attributable to the absence of
TLR4 activity in haematopoietic or other cell types2528.
During haemodynamic stress, mitochondria are typi
cally damaged; if the degradation of mitochondrial
DNA is inhibited in this setting, a TLR9dependent
inflammation-induced cardiomyopathy develops29.
NLRs function as cytosolic sensors of intracellular
DAMPs and PAMPs. In humans, the NLR family is
composed of 22 intracellular PRRs that share a central
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
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
pro-inflammatory cytokines and chemokines that are
Macrophage
responsible for recruiting inflammatory leukocytes
from the blood into the area of tissue injury 35. The initial
Blood
DC
vessel
inflammatory phase is followed by a proliferative phase
that is characterized by the expansion of neutrophil
Endothelial
TReg cell
and macrophage populations that are responsible for
cell
Mast cell
removing dead cells and matrix debris, as well as releas
ing cytokines and growth factors that lead to the forma
Neutrophil
tion of a highly vascularized granulation tissue, which is
B cell
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
tant initiators of inflammation a process that is ampli
Figure 1 | Immune cells in the resting heart. The majority of immune cells in the resting
fied following coronary reperfusion. Immediately after
heart are macrophages, which are found primarily surrounding
endothelial
but are
Nature
Reviewscells
| Immunology
blood flow is restored, resident cardiac mast cells release
also seen in the interstitium amongst cardiomyocytes. Mast cells, dendritic cells (DCs),
pre-formed pro-inflammatory mediators (for example,
Bcells and regulatory T (TReg) cells are found sparsely in cardiac tissue, while neutrophils
tumour necrosis factor (TNF), histamine and mast cell
and monocytes are not observed within myocardial tissue, but can be observed as
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.
30
repeat region . Analysis of human tissue has revealed that Timely restoration of blood flow to viable tissue is cru
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
ischaemiareperfusion 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 RIGI, melanoma CD11bCD18 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 deficits3941.
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 immunecells.
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
Ctype 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 tissueinjury.
Patients deficient in neutrophils or neutrophil function
One important issue that has been only partially suffer from devastating disseminated bacterial infec
addressed is the delineation of cell-type-specific roles tions, indicating a clear requirement for this cell type to
Granulation tissue
for PRRs. Beyond the few examples given here, it is not prevent expansion of otherwise harmless pathogens43.
This is tissue that arises
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
during the process of cardiac tissue injury and repair, are the best example of a cell type that promotes overall
and extracellular matrix occurs
which represents an important avenue for further longevity, but in the setting of sterile injury, they have no
in order to maintain the
integrity of the myocardial wall.
investigations.
known protectiverole.
Monocyte

NATURE REVIEWS | IMMUNOLOGY

VOLUME 15 | FEBRUARY 2015 | 119


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Heart

Cell necrosis

DAMPs
dsRNA
CpG
Uric acid
crystal
HMGB1
ATP
HSP
IL-1

Receptors
TLR3
TLR9
NLRP3
TLR2TLR4RAGE
P2Y11 and P2X7
TLR4
IL-1R

ECM
degradation

DAMPs
Hyaluronan
fragment
Heparan
sulphate

Receptors
CD44TLR4
TLR4

Thrombus

Ischaemic area

Immune cell recruitment


and activation

Figure 2 | Cardiac injury and sensing damaged tissue. The figure shows a coronary
artery occlusion (black) that leads to ischaemic tissue injuryNature
(grey zone).
From
within the
Reviews
| Immunology
ischaemic area, cell necrosis, extracellular matrix (ECM) degradation and recruitment
of immune cells all lead to the production of specific damage-associated molecular
patterns (DAMPs), which are recognized by pattern recognition receptors. This leads
to the generation of inflammatory responses to internal injury signals. CpG, CpG
dinucleotides; dsRNA, double-stranded RNA; HMGB1, high-mobility group box1;
HSP, heat shock protein; IL, interleukin; IL-1R, IL-1 receptor; NLRP3, NOD-, LRRand pyrin domain-containing 3; P2Y, P2Y purinoceptor; P2X, P2X purinoceptor;
RAGE, receptor for advanced glycation end-products; TLR, Toll-like receptor.

Neutrophil recruitment is mediated in two phases


(FIG.3a); the first phase is peripheral activation prior to
infiltration. Mitochondria in all cell types, including
cardiomyocytes, contain formylated peptides and mito
chondrial DNA, both of which are structurally similar
to bacterial components. In an analogous system (skel
etal muscle necrosis), these mitochondrial DAMPs are
released. Formylated peptides and mitochondrial DNA
are sensed by formyl peptide receptor 1 (FPR1) and
TLR9, respectively, and the triggering of these receptors
promotes neutrophil activation and their recruitment to
inflamed tissues29. It is important to confirm whether
similar mechanisms are involved in neutrophil activation
following cardiomyocyte necrosis. The ability to sense
mitochondrial motifs is not surprising as mitochondria
are endosymbionts and related to microorganisms in
many ways. However, the exact role of mitochondrial
motifs in cardiac injury remains to be defined.
The second phase of neutrophil activation depends on
cardiac endothelial cells. Pro-inflammatory mediators,
such as TNF, IL1 and histamine, activate endothelium
and induce the upregulation of adhesion molecules that
enable neutrophil transmigration between and through
endothelial cells to reach the site of tissue injury 9,44.
Detailed imaging studies in other organ systems have
aided our understanding of how neutrophils enter
damaged tissue. For example, following necrotic tissue
injury in the liver, neutrophils adhere to more remote

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
convoluted path is that necrotic tissue is usually non- (or
under) perfused, and transendothelial migration at the
site of injury is not possible. Unfortunately, such detailed
imaging of migrating neutrophils has yet to be done in
the injured beating heart, but techniques to achieve this
are being developed46. Such studies will be highly inform
ative for understanding the spatial dynamics of resident
macrophages, recruited neutrophils and monocytes
during the progression of ischaemic cardiacinjury.
An important early mediator of tissue injury seems
to be IL6, which is produced in an autocrine fashion by
cardiomyocytes and recruited myeloid cells (both neutro
phils and macrophages)47,48. IL6 upregulates intracellular
adhesion molecule 1 (ICAM1) expression on cardio
myocytes, inducing neutrophil binding and stimulating
cytotoxic activity 4951. The role of neutrophils following
cardiac injury may be more pronounced when major
areas of the myocardium are at risk of cell death (but have
not yet died), such as following ischaemiareperfusion
injury. Neutrophils produce numerous proteases that
contribute to injury, and blockade of neutrophil recruit
ment seems to be most effective during more limited
episodes of ischaemia, rather than during longer episodes
in which cardiomyocyte death is largely secondary to
anoxia and nutrient deprivation5254.

Cardiac macrophages
The dominant view for the past half century has been
that bone marrow-derived haematopoietic stem cells
(HSCs) give rise to circulating blood monocytes, which
enter into tissues and become tissue macrophages55.
However, in the past few years a series of more definitive
publications have drastically revised our understand
ing of monocyte and macrophage origin by showing
that many tissue-resident macrophage populations are
established during embryonic development, and they
are subsequently maintained by self-renewal, rather than
through blood monocyte input 5,5661. Given the impor
tance of monocytes and macrophages to cardiovascular
disease, we review recent insights into the origin of
functions of these subsets and how they contribute to
cardiac tissue injury andrepair.
There are two main subsets of circulating mono
cytes in mice, LY6Chi monocytes and LY6Clow monocytes.
LY6C+ monocyte progenitors give rise to LY6Chi mono
cytes and, through a nuclear receptor subfamily 4
group A member 1 (NR4A1)-dependent transcrip
tional programme, LY6C hi monocytes differentiate
into LY6Clow monocytes58,62,63. Global transcriptional
profiling has shown these monocyte subsets are con
served in humans64 and they have very different roles
invivo. LY6Clow monocytes adhere to and move along
the endothelium, both clearing damaged cells and
triggering inflammatory responses without entering
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
cardiomyocyte

Endothelial
cell activation
Histamine,
TNF and IL-1

DAMPs

Infarct
zone

Mast cell
degranulation

C5

Peripheral
neutrophil
activation

C5a

Proteinases and
collagenases

Phagocytosis of
necrotic cells

Neutrophil
recruitment
Ischaemia and anoxia

b 34 days post ischaemic injury

IL-17A promotes
neutrophil production
in bone marrow

IL-17A induces
apoptosis
Apoptotic
cardiomyocyte
Apoptotic
neutrophil

IL-10
and TGF

IL-17A

IL-17A

IL-10 IL-10 suppresses


monocyte activation

T cell
IL-23

Activated
monocyte

Endothelial cell
CCL7

Monocytes produce
pro-inammatory
cytokines and
chemokines
TGF

Monocytes ingest
apoptotic cells and
release anti-inammatory
cytokines

TReg cell

CCR2-dependent
monocyte recruitment

Innate
B cell
Innate B cells activated
in response to DAMPs

Fibroblast
TGF induces
brosis
Nature Reviews | Immunology

Figure 3 | Immune response to ischaemic injury in adult animals. a | Early after ischaemic injury (within 24hours),
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 interleukin1 (IL1)), 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 (34days), there is recruitment of LY6Chi
monocytes from the blood into ischaemic cardiac tissue. Some of these monocytes originate from reservoirs in the
spleen. Innate Bcells that express immunoglobulins IgM and IgD are also recruited into the myocardium and produce
CCchemokine ligand 7 (CCL7), which promotes further monocyte recruitment10. Innate Bcell 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- (TGF) and IL10, and thereby decrease leukocyte
recruitment. Monocytes produce IL23, which drives the production of IL17A by Tcells. IL17A 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 IL23 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
ing ischaemic injury or hypertensive stress; whereas
LY6Clow monocytes do not seem to be directly recruited
into the myocardium5,59,61,67,68. Monocytes recruited into
ischaemic myocardium are found in the blood, but the
spleen is also a monocyte reservoir that can be used
when blood and bone marrow stores are insufficient69,70.
Splenic monocytes also seem to have a protective role, as
splenectomy leads to impaired infarct healing 71. Monocyte
recruitment is in part dependent on innateBcells, which
are also recruited to the ischaemic myocardium, and
these B cells drive monocyte population expansion
through a CCchemokine ligand 7 (CCL7)-dependent
process10 (FIG.3b). Once monocytes enter the tissue they
begin to differentiate into macrophages. For many years,
it has been very challenging to separate recruited mono
cytes from resident macrophages and, as a result, they
have been analysed as a single population. However,
as we describe below, these populations have different
ontological lineages, and this has important functional
implications.

Innate Bcells
These cells are thought to
become rapidly activated in
the absence of classical
Tcell-dependent antigen
presentation mechanisms.
They are mobilized by other
cell types and/or inflammatory
triggers.

ApoE-deficient mice
These mice are used to model
atherosclerosis. They have
increased total plasma
cholesterol levels and
increased bone marrow
production of monocytes and
heightened inflammatory
responses.

Cardiac macrophage subsets. Recently, studies using


genetic fate mapping, parabiosis and adoptive transfer
techniques have defined resident cardiac macrophage
populations in much more detail. Rather than being a
homogenous population, resident cardiac macrophages
comprise three discrete subsets that have different ori
gins and functions5 (FIG.4a). These three macrophage
subsets are defined by their differential expression
of MHC classII and CC chemokine receptor 2 (CCR2).
MHC classIIhi and MHC classIIlow cardiac macrophages
are both CCR2 and are, numerically, the dominant
subsets in the heart 5. These macrophage subsets are
primarily derived from embryonic progenitors, with
a substantial number arising from embryonic yolk sac
precursors, and they renew through insitu proliferation,
rather than through monocyte input. After birth, there
is some dilution of embryonically derived macrophages
by recruited monocyte-derived macrophages in the
heart, but in adult mice the majority of resident cardiac
macrophages remain of embryonic origin5,72.
The third cardiac macrophage subset is made up
of CCR2+ macrophages, which are derived from, and
slowly replenished by, circulating blood monocytes.
Detailed studies during hypertensive stress suggest that
embryonically derived macrophage populations expand
solely through insitu proliferation, whereas monocytederived macrophages require monocyte input prior to
proliferative expansion in the tissue5. CCR2+ macro
phages express high levels of pro-inflammatory genes,
including those associated with the NLPR3 inflamma
some, which is required to process and deliver IL1 to
the heart during cardiac stress5. Inflammasome acti
vation promotes adverse cardiac remodelling follow
ing ischaemic injury, genetic cardiac hypertrophy and
hypertensive cardiac disease18,73,74; it is likely that CCR2+
cardiac macrophages are involved in inflammasome acti
vation in each of these settings. The robust pro-inflam
matory gene signature of CCR2+ cardiac macrophages
suggests that they are important for immune protection

against invading pathogens, but their inappropriate


activation in the setting of sterile inflammation may
lead to unwanted pathology 75,76. Prior studies have used
dichotomous expression of two classical myeloid cell
markers (F4/80 and CD11b) in order discern embry
onic versus adult monocyte origin57. However, differ
ent cardiac macrophage lineages (embryonic and adult)
cannot be distinguished by these cell surface markers
alone and, as such, genetic fate mapping remains the
most accurate method for lineage discrimination of
cardiac macrophages in adult mice5,72.
Functions of cardiac macrophages. One of the interest
ing challenges in the setting of cardiac tissue injury is to
define the roles of recruited monocytes and monocytederived macrophages. Non-selective depletion strategies
have shown that in the absence of both monocytes and
macrophages, scar formation is impaired after cardiac
ischaemic injury, with decreased collagen production,
decreased angiogenesis and increased mortality due
to myocardial rupture6,77. However, experiments using
ApoEdeficient mice (apolipoprotein-E-deficient mice) sug
gest that excessive expansion of macrophage populations
can have a detrimental effect on infarct healing, leading to
excessive inflammation and impaired cardiac function78.
Furthermore, studies of cardiac injury in Ccr2/ mice,
which lack circulating monocytes, indicate that monocyte
recruitment and the associated inflammatory response
leads to increased cardiac pathology 7981. The loss of
LY6Chi monocytes also prevents hypertension-induced
cardiac fibrosis and improves left ventricle function after
myocardial infarction. Taken together, these data suggest
that recruited monocytes have a pathological role in the
setting of sterile cardiac injury, but that resident embry
onically derived macrophages are likely to have important
roles in the tissue-repair response6,67,82.
Interestingly, as already mentioned, the neonatal heart
has a remarkable capacity to regenerate in response to
multiple forms of tissue injury, but the regenerative pro
cess is lost if resident embryonically derived macrophages
are eliminated8385. In fact, the neonatal heart supports
the expansion of tissue-resident embryonically derived
macrophage populations following injury, as opposed
to the recruitment of monocytes, and this seems to be a
fundamental difference between the neonatal and adult
heart. Neonatal cardiac macrophages promote endothelial
cell activation and cardiomyocyte growth, and they gener
ate minimal inflammation after stimulation through TLR
and inflammasome pathways85 (FIG.4b). These data suggest
that that the neonatal heart avoids excessive inflamma
tory responses, which may be a crucial factor that aids the
regenerative process, although this benefit may come at
the cost of being more susceptible to infections86.
Macrophage subsets that are primarily derived from
embryonic precursors are more efficient at internaliz
ing debris and engulfing apoptotic cardiomyocytes, sug
gesting they have important homeostatic roles. Indeed,
these functions are reminiscent of embryonic macrophage
functions during development and could suggest that
some of these functions are hard-wired into the adult
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

MHC class IIlow yolk sacCardiomyocyte derived macrophage

Later in embryonic development

Adult heart
MHC class IIhi
macrophage MHC class IIlow
macrophage

IIlow fetal

MHC class
monocyte-derived macrophage

Monocyte

Blood
vessel

Developing heart seeded by


yolk sac-derived macrophages

Embryonically
derived macrophages
proliferate in situ
MHC class IIhi macrophages develop
from MHC class IIlow populations

Heart seeded by macrophages


derived from fetal monocytes

Adult monocytes give rise to MHC class IIhi


and MHC class IIlow macrophages and
short-lived CCR2+ macrophages

Neonatal heart

Adult heart
Receptive
neonatal
cardiomyocyte
Quiescent
adult
cardiomyocyte

Tissue injury

Expansion
of resident
macrophage
populations

Angiogenesis
Cardiomyocyte proliferation
Inammation

Tissue injury

Regeneration of
cardiac tissue

Angiogenesis
Cardiomyocyte proliferation
Inammation
Impeded regeneration
of cardiac tissue
Scar formation

Figure 4 | Role of embryonically derived macrophages during tissue injury


and repair. a | During embryonic development, extra-embryonic yolk-sac
derived macrophages (shown in blue) initially seed the developing heart. The
cells express low levels of MHC classII molecules. Later during development,
fetal monocyte-derived macrophages (shown in orange) also infiltrate the
heart, and both of these embryonic macrophage populations expand further
during development, and after birth, by insitu proliferation. MHC classIIhi
macrophages develop from these populations of MHC classIIlow macrophages
after weaning5,72. During this time, haematopoietic stem cell-derived
monocytes infiltrate the heart and also differentiate into MHC classIIhi
macrophages (dark green) and MHC classIIlow macrophages (light green).
MHC classIIhi and MHC classIIlow macrophages persist in the tissue as an
ontologically mixed population, primarily composed of embryonically derived
macrophages5. Monocytes that infiltrate the heart can also become short-lived
CC-chemokine receptor 2 (CCR2)+ macrophages, which are entirely derived
from blood monocytes5. b | The figure shows a comparison of the macrophage
response to tissue injury in neonatal animals (which regenerate cardiac tissue)

and adult animals (where there is minimal tissue regeneration). In the neonate,
Nature Reviews | Immunology
resident embryonically derived macrophage populations expand without
notable monocyte input. Embryonically derived macrophages promote
angiogenesis, cardiomyocyte proliferation and drive minimal inflammation
when stimulated by damage-associated molecular patterns (DAMPs). These
properties of neonatal macrophage populations facilitate cardiac
regeneration85. Following cardiac injury in the adult, there is marked expansion
of CCR2 + monocyte and macrophage populations. Notably, these
macrophages possess a limited ability to promote angiogenesis and
cardiomyocyte population expansion, but have notable capacity to drive
inflammatory responses, which impedes tissue regeneration5,85. Importantly,
neonatal cardiomyocytes are primed to divide and therefore are receptive to
regenerative signals84,153,154. Adult cardiomyocytes are much less receptive
to regenerative signals, and growth signals from embryonically derived cardiac
macrophages are one of many important components of cardiac regeneration
promotion in adult tissues. A key therapeutic goal would be to recapture the
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.
Expression of the MER tyrosine kinase (MERTK) a
phagocytic receptor that is a highly specific marker
of tissue macrophages is upregulated on cardiac
macrophages after myocardial infarction, and loss of
MERTK expression leads to the accumulation of apop
totic cardiomyocytes, increased neutrophil persistence
and decreased levels of the anti-inflammatory cytokine
IL10 in the myocardium89,90. This ongoing inflamma
tion ultimately results in decreased cardiac function89,90.
Phagocytosis of apoptotic cells (efferocytosis) in skeletal
muscle results in the transition of macrophages to a more
anti-inflammatory state through the upregulation of the
intracellular signalling kinase AMPK, which is associated
with downregulation of pro-inflammatory genes (such as
Tnf) and the upregulation of anti-inflammatory genes,
such as Il10 (REFS91,92). Together, these data suggest that
strategies that target newly recruited monocytes while
sparing resident macrophages may yield therapeutic
benefit during cardiacinjury.

Adaptive immune cells and heart injury


Lymphocytes have been reported to have diverse roles
during cardiac tissue injury and repair. CD4deficient
mice, MHC classIIdeficient mice and mice that express
a single transgenic Tcell receptor show impaired wound
healing and increased monocyte population expansion
after ischaemic injury. This suggests that CD4+ Tcells
have protective functions during cardiac injury, and
that auto-antigens may be presented to CD4+ Tcells by
MHC classIIexpressing cells (such as macrophages and
DCs), which then drives immunosuppressive responses
in the myocardium93. Interestingly, it may be that regu
latory Tcells are the key CD4+ Tcell subset involved
here, and regulatory T cells have been shown to promote
myocardial recovery through an IL10dependent path
way 94,95. Another study found that recombination acti
vating gene (RAG)-deficient mice (which lack Tcells
and Bcells) have decreased infarct sizes after ischaemic
injury, and that the transfer of CD4+ Tcells into the RAGdeficient mice had a negative effect on infarct healing.
Therefore, in certain situations CD4+ Tcells may have
pathogenic roles following ischaemic injury96. In addi
tion, RAG-deficient mice have decreased levels of circu
lating IgG antibodies, and these antibodies are known to
have immunosuppressive effects (as reviewed in REF.97).
CD8+ Tcells have also been shown to have regula
tory roles following ischaemic injury. CD8+ Tcells
express type2 angiotensin II receptor (AGTR2), which
is known to drive anti-inflammatory responses98,99.
Angiotensin II is part of the reninangiotensin system,
which has long been known to be a crucial pathological
pathway in heart disease100,101. Angiotensin II is thought
to primarily induce cardiomyocyte hypertrophy and
increase vascular tone by signalling through AGTR1,
in a process that is amplified by blockade of AGTR2
(as reviewed in REF.102). After ischaemiareperfusion
injury AGTR2expressing CD8+ Tcells, inhibit inflam
mation and decrease infarct size by producing IL10 in an
angiotensin IIdependent fashion, and this subsequently
dampens the immune response98.

In addition to these roles of Tcells, other lympho


cytes have been shown to shape inflammatory responses
in cardiac tissue. Natural killer (NK) cells were reported
to infiltrate the myocardium and have protective effects
following ischaemic injury 103. The adoptive transfer of
activated NK cells into mice was shown to protect against
left ventricle dysfunction following ischaemia by pre
venting fibrosis and enhancing angiogenesis through a
contact-dependent interaction with endothelial cells.

Transition from acute inflammation to fibrosis


One of the first important steps that limits tissue injury
and promotes the transition to tissue healing is the
resolution of neutrophil recruitment. Macrophages
are believed to initiate the process through two key
mechanisms that have been linked to their phagocytic
capacity. First, it has been well described invitro and
invivo that macrophages that have ingested apoptotic
cells increase their production of anti-inflammatory
and pro-fibrotic cytokines, such as IL10 and TGF,
while they decrease production of pro-inflammatory
cytokines, such as IL1 and TNF 104,105. Following
ischaemic injury, if all cardiac macrophages are absent,
or they if they lack the phagocytic receptor MERTK,
there is neutrophil persistence within infarcted myo
cardial tissue and ongoing inflammation. This suggests
that the activation of cardiac macrophage phagocytic
pathways limits inflammation, and that the loss of the
phagocytic macrophages results in an accumulation of
apoptotic material in the infarct zone5,6,89.The transi
tion from inflammatory to pro-fibrotic macrophages
has been classically thought to involve polarization
changes (from an M1 to an M2 macrophage pheno
type); however, it is becoming clear that macrophages
are very heterogeneous and fit along a broad spectrum
of possibilities, rather than into clearly defined subsets.
Further work is required to define where individual
cardiac macrophage subsets fit into the M1 and M2
framework106.
The second mechanism involves a complex cytokine
cascade that depends on Tcells and their production
of IL23 and IL17A. The production of IL17A leads to
neutrophil production and release from bone marrow
stores107. Interestingly, IL17A is regulated by secretion
of IL23 by macrophages, and the ingestion of apoptotic
neutrophils decreases IL23 production, which leads to
decreased IL17A levels and decreased neutrophil pro
duction in the bone marrow 107109. Following ischaemic
cardiac injury, Tcells and neutrophils themselves seem
to be important sources of IL17A production within the
myocardium, and mice that lack IL23 have reduced
IL17A levels in cardiac tissue, reduced neutrophil accu
mulation, increased Tcell population expansion and
improved infarct healing 110. Indeed, IL17A seems to also
be directly toxic to cardiomyocytes, and neutralization
of IL17A improves infarct healing 111 (FIG.3b).
Additional work has shown an important link between
IL17A production and activation of fibrotic pathways
within the heart that lead to collagen deposition, both fol
lowing ischaemia and haemodynamic strain112,113. In this
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

TLR2
or TLR4
MYD88

CAR

DAF
Cardiomyocyte

FYN
or ABL

LCK
Endosome

Viral entry

Viral nucleic
acids
MDA5

RIG-I

Activation of
cytosolic PRRs
IRAK4

NF-B

TLR7

TLR3
TRIF

Activation of
endosomal PRRs

IRF3

p50 p65
NF-B activation

IRF activation

Type I IFN
production

Activation of immunopathological response,


tissue damage

Protective
immune response

Figure 5 | Interaction of coxsackievirus B with the host innate and acquired immune
system. Coxsackievirus B (CVB) engages the internalizingNature
receptor
coxsackievirus
and
Reviews
| Immunology
adenovirus receptor (CAR), and the receptor-associated tyrosine kinases FYN and ABL
can facilitate viral remodelling of the cell cytoskeleton to promote viral cell entry. This
process is further aided by the coreceptor decay accelerating factor (DAF) and its
associated tyrosine kinase LCK. Viral components can be detected by the intracellular
pattern recognition receptors (PRRs) melanoma differentiation-associated gene 5
(MDA5) and retinoic acid-inducible gene I (RIGI) leading to the activation of nuclear
factorB (NFB)dependent inflammatory pathways. NF-Bdependent responses can
also be activated following the engagement of CVB with cell surface-expressed Toll-like
receptors (TLRs), which signal through the myeloid differentiation primary response
protein 88 (MYD88) adaptor and downstream signal intermediates such as IL1-receptorassociated kinase 4 (IRAK4) and TNF receptor-associated factor 6 (TRAF6). These
pathways facilitate viral proliferation and host immune-mediated tissue damage. By
contrast, activation of interferon regulatory factor 3 (IRF3) following the triggering of
the TIR-domain-containing adapter-inducing IFN (TRIF) pathway by CVB leads to the
production of protective typeI interferons (IFNs). There is mutual counter-regulation of
the MYD88 IRAK4 and TRIFIRF3 pathways. The subsequent maturation of CD4+ and
CD8+ Tcell subsets is also detrimental for the host, whereas regulatory Tcells are
host-protective. Cross-talk between acquired and innate immune signalling pathways
also modulates the host response to CVB infection.

loss of IL23 was demonstrated to impair infarct healing,


which was mechanistically linked to a failure to activate
fibroblasts, leading to impaired scar formation and myo
cardial rupture113. These data are reminiscent of the rela
tionship between monocyte influx and tissue healing after

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
are yet-to-be determined, context-dependent factors
that modulate whether a particular cell type or pathway
mediates cardiac tissue repair or damage.
The activation of cardiac fibroblasts and subse
quent extracellular matrix deposition is an important
component of cardiac ischaemic injury and repair.
Interestingly, cardiac fibroblasts can also become acti
vated in more subtle forms of cardiac stress that also
lead to impaired cardiac function owing to increased
myocardial stiffness (such as hypertension caused
by infusion of angiotensin II). In this setting there is
rapid recruitment of neutrophils and monocytes into
the myocardium and also expansion of resident car
diac macrophage populations5,114118. 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+ Tcells
through an IFN-dependent pathway 119,120 (reviewed
in detail in REF.121).

Myocarditis as a model of cardiac inflammation


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
mechanisms that occur following tissue injury deter
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
tious pathogens can also be involved, such as bacteria or
the protozoan parasite Trypanosoma cruzii, which is the
cause of Chagas disease, the commonest cause of heart
failure in South America122,123. Myocarditis can also
result from non-infectious triggers of DAMP release,
which activate the innate immune response leading to
sterile inflammation, such as allergic reactions to drugs
or chemicals124.
Myocarditis accounts for about one in nine cases
of heart failure and remains one of the most common
reasons for heart transplantation worldwide, as spe
cific treatments are lacking 125. The most common viral
triggers of myocarditis include enteroviruses, such as
coxsackievirus B3 (CVB3) and CVB4, and adenovi
ruses. Virus-induced myocarditis shows a periodicity
in its prevalence in human populations, which is pos
sibly related to herd immunity. European studies also
show a substantial contribution of parvovirus B19,
which shows tropism for endothelial cells and the bone
marrow 126. Coxsackieviruses and adenoviruses target
host tissues, including the immune, cardiovascular and
neurological systems, through their binding and inter
nalization by coxsackievirus and adenovirus receptor
(CAR) which is an immune-regulated tight junc
tion protein127,128. Internalization of the virus is assisted
by cobinding to decay accelerating factor (DAF; also
known as CD55), a ubiquitously expressed host pro
tein that inhibits complement activation129 (FIG.5). Loss
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,
suggesting that entry into cardiomyocytes is required
for tissue damage130. However, although viral entry and
subsequent viral proliferation trigger disease, they are
not necessarily the primary determinants of disease out
comes. Both the innate and adaptive immune responses
have crucial roles in the progression of viral myocar
ditis, as shown by the fact that genetic deletion of the
TLR-signalling proteins myeloid differentiation pri
mary response protein 88 (MYD88) or IL1R-associated
kinase 4 (IRAK4), or deletion of the T cell receptorassociated tyrosine kinase LCK, ameliorates myocardial
inflammation and improves survival in mice, despite
increased levels of viral proliferation131134.
Following viral entry into the target cell, such as an
immune cell or cardiomyocyte, the virus can engage
intracellular NLRs, including RIGI and MDA5. The
endosomal degradation of the virus can also lead to the
activation of TLR3 and TLR732,133. The signalling cas
cades induced by such PRR activation can have detri
mental consequences for the host. As stated above, the
MYD88IRAK4 pathway, showed a very marked protec
tive benefit in mice during CVB3 infection. At least four
different mechanisms are thought to mediate protection
in this setting. First, the loss of MYD88IRAK4 signal
ling decreases downstream activation of TNF receptorassociated factor6 (TRAF6) and nuclear translocation of
the NFB complexes, resulting in reduced pro-inflam
matory cytokine production and Tcell activation135.
Second, increased IRF3 or IRF5 homodimerization and
phosphorylation of signal transducers and activators of
transcription 1 (STAT1) and STAT5 occurs in the absence
of MYD88 or IRAK4, leading to increased production of
protective antiviral typeI IFNs133135. Third, IRAK4 dele
tion facilitates the mobilization of protective CCR5+
monocytes from the bone marrow into the myocar
dium133. Fourth, the absence of MYD88 or IRAK4 leads
to downregulation of CAR expression and decreased
viral proliferation135. However, the genetic deletion of
IRF3, which is activated by the TIR-domain-containing
adaptor-inducing IFN (TRIF)-dependent TLR path
way, leads to much worse outcome during virus-induced
myocarditis, with mice showing increased mortality and
greater levels of viral proliferation136. Mechanistically, this
is associated with decreased typeI interferon production
and increased activation and translocation of NF-B136.
These observations have several important implica
tions. The first is that increased expression of viral entry
receptors and enhanced viral proliferation are, paradoxi
cally, facilitated by the host innate immune response
probably an evolutionarily selected advantage for the
pathogenic virus at the expense of the host. The second
is that intracellular immune signalling pathways seem to
induce functional responses that regulate trafficking of
inflammatory cells, such as CCR5+ monocyte-derived
macrophages, from the bone marrow. Indeed this is also
the case in sterile inflammation post myocardial infarction,
in which IRAK4 signalling seems to regulate the traffick
ing and maturation of dendritic cells into the myocardium,
with subsequent orchestration of host inflammatory
responses that dictate tissue remodelling and host survival.

The activation of innate immune signalling path


ways also sets the stage for subsequent Tcell maturation
and participation in the host inflammatory response.
Genetic deletion of LCK, which impairs Tcell matura
tion, results in almost complete protection of the host
against CVB3 infection131. A similar outcome is observed
following CD4+ or CD8+ Tcell subset deletion, or fol
lowing deletion of the leukocyte tyrosine phosphatase
CD45, which is associated with upregulation of typeI
IFNs132,137. Adoptive transfer of regulatory T cells into
CVB3infected hosts also had a marked protective
effect, again associated with a decrease in viral prolif
eration and upregulation of typeI IFNs138. Surprisingly,
there was a general downregulation of molecules asso
ciated with TLR signalling pathways and even TLR4
itself. Moreover, beyond their role during infections,
regulatory T cells also promote recovery through an
IL10dependent pathway following ischaemic injury 95.
These data suggest that there is close cross-talk between
the regulatory T cell subsets and the innate immune sig
nalling pathways that operate during both infectious and
sterileinjury.
The above progress in understanding myocarditic
processes has moved the field forward from the ear
lier failed attempts to use immunosuppression to treat
patients with myocarditis139. Subsequently, a PhaseII
trial has demonstrated that in patients with persistent
viral proliferation, intravenous delivery of IFN clears
the virus and improves symptoms of myocarditis140.
Targeted immunosuppressive therapy may be indi
cated for patients with persistent immune activation, in
which the viral proliferation phase has already passed.
Nevertheless, there are still large gaps in our knowledge,
including why there are periodic enteroviral outbreaks
in the population. Furthermore, why do some patients
develop severe viral myocarditis that requires transplan
tation, while others are able to rebalance the immune
response and promptly recover from viral mycocarditis?
Are there biomarkers to permit one to predict an indi
vidual patients susceptibility and, for those at high risk,
could vaccination be a viable option?

Future directions
The ultimate goal in terms of understanding how the
immune system directs inflammatory and reparative
programmes following cardiac injury is the develop
ment of therapeutic strategies that promote tissue
regeneration. Evolutionary pressures drove both the
development of primitive phagocytic cells that promote
tissue growth and wound healing, but also drove the
development of innate and adaptive immune mecha
nisms that promote survival of the host in the face
of infectious threats. Adaptations that both enhance
pathogen clearance and promote tissue repair may
result in a zero-sum game, whereby the very adapta
tions that enhance host defence result in a loss of repar
ative potential owing to excessive inflammation. The
ability to selectively suppress excessive inflammation
while preserving reparative functions is a novel thera
peutic avenue that can enhance recovery in patients
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
Ma,X., Cong,P., Hou,X., Edgecombe,G.D. &
Strausfeld,N.J. An exceptionally preserved arthropod
cardiovascular system from the early Cambrian.
Nature Commun. 5, 3560 (2014).
2. Bier,E. & Bodmer,R. Drosophila, an emerging model
for cardiac disease. Gene 342, 111 (2004).
3. Lozano,R. etal. Global and regional mortality from
235 causes of death for 20 age groups in 1990 and
2010: a systematic analysis for the Global Burden of
Disease Study 2010. Lancet 380, 20952128
(2012).
4. Pinto,A.R. etal. An abundant tissue macrophage
population in the adult murine heart with a distinct
alternatively-activated macrophage profile.
PLoS ONE. 7, e36814 (2012).
5. Epelman,S. etal. Embryonic and adult-derived
resident cardiac macrophages are maintained through
distinct mechanisms at steady state and during
inflammation. Immunity 40, 91104 (2014).
This is the first study to show that cardiac
macrophages are not a single population but are
composed of distinct subsets, with different origins
and functions.
6. Nahrendorf,M. etal. The healing myocardium
sequentially mobilizes two monocyte subsets with
divergent and complementary functions. J.Exp. Med.
204, 30373047 (2007).
This report shows the initial characterization of
cardiac macrophages in the resting heart and after
ischaemic injury.
7. Swirski,F.K. & Nahrendorf,M. Leukocyte behavior in
atherosclerosis, myocardial infarction, and heart
failure. Science 339, 161166 (2013).
8. Choi,J.H. etal. Identification of antigen-presenting
dendritic cells in mouse aorta and cardiac valves.
J.Exp. Med. 206, 497505 (2009).
9. Frangogiannis,N.G. etal. Resident cardiac mast cells
degranulate and release preformed TNF-, initiating
the cytokine cascade in experimental canine
myocardial ischemia/reperfusion. Circulation 98,
699710 (1998).
10. Zouggari,Y. etal. B lymphocytes trigger monocyte
mobilization and impair heart function after acute
myocardial infarction. Nature Med. 19, 12731280
(2013).
11. Saxena,A. etal. Regulatory Tcells are recruited in
the infarcted mouse myocardium and may modulate
fibroblast phenotype and function. Am. J.Physiol.
Heart Circ. Physiol. 307, H1233H1242 (2014).
12. Roger,V.L. etal. Heart disease and stroke
statistics2012 update: a report from the American
Heart Association. Circulation 125, e2e220
(2012).
13. Jacoby,D. & McKenna,W.J. Genetics of inherited
cardiomyopathy. Eur. Heart J. 33, 296304 (2012).
14. Sangiuliano,B., Perez,N.M., Moreira,D.F. &
Belizario,J.E. Cell death-associated molecularpattern molecules: inflammatory signaling and
control. Mediators Inflamm. 2014, 821043
(2014).
15. Matzinger,P. Tolerance, danger, and the extended
family. Annu. Rev. Immunol. 12, 9911045 (1994).
16. Janeway,C. A. Jr. Approaching the asymptote?
Evolution and revolution in immunology.
Cold Spring Harb. Symp. Quant. Biol. 54Pt. 1, 113
(1989).
17. Mann,D.L., Topkara,V.K., Evans,S. & Barger,P.M.
Innate immunity in the adult mammalian heart: for
whom the cell tolls. Trans. Am. Clin. Climatol. Assoc.
121, 3450 (2010).
18. Mezzaroma,E. etal. The inflammasome promotes
adverse cardiac remodeling following acute myocardial
infarction in the mouse. Proc. Natl Acad. Sci. USA
108, 1972519730 (2011).
19. Takeuchi,O. & Akira,S. Pattern recognition receptors
and inflammation. Cell 140, 805820 (2010).
20. Rathinam,V.A. etal. The AIM2 inflammasome is
essential for host defense against cytosolic bacteria
and DNA viruses. Nature Immunol. 11, 395402
(2010).
21. Frantz,S. etal. Toll4 (TLR4) expression in cardiac
myocytes in normal and failing myocardium.
J.Clin. Invest. 104, 271280 (1999).
22. Birks,E.J. etal. Increased toll-like receptor 4 in the
myocardium of patients requiring left ventricular assist
devices. J.Heart Lung Transplant. 23, 228235
(2004).
23. Kashiwagi,M. etal. Differential expression of Toll-like
receptor 4 and human monocyte subsets in acute
myocardial infarction. Atherosclerosis 221, 249253
(2012).
1.

24. Arslan,F. etal. Myocardial ischemia/reperfusion injury


is mediated by leukocytic toll-like receptor2 and
reduced by systemic administration of a novel anti-tolllike receptor2 antibody. Circulation 121, 8090
(2010).
25. Fallach,R. etal. Cardiomyocyte Toll-like receptor 4 is
involved in heart dysfunction following septic shock
or myocardial ischemia. J.Mol. Cell Cardiol. 48,
12361244 (2010).
26. Binck,B.W. etal. Bone marrow-derived cells
contribute to contractile dysfunction in endotoxic
shock. Am. J.Physiol. Heart Circ. Physiol. 288,
H577H583 (2005).
27. Oyama,J. etal. Reduced myocardial ischemiareperfusion injury in toll-like receptor 4deficient mice.
Circulation 109, 784789 (2004).
28. Tavener,S.A. etal. Immune cell Toll-like receptor 4
is required for cardiac myocyte impairment during
endotoxemia. Circ. Res. 95, 700707 (2004).
29. Oka,T. etal. Mitochondrial DNA that escapes from
autophagy causes inflammation and heart failure.
Nature 485, 251255 (2012).
This is the first study to show that endogenous
DAMPs are released from the myocardium during
haemodynamic strain, a process that impairs
cardiac function.
30. Geddes,K., Magalhaes,J.G. & Girardin,S.E.
Unleashing the therapeutic potential of NOD-like
receptors. Nature Rev. Drug Discov. 8, 465479
(2009).
31. Kawaguchi,M. etal. Inflammasome activation of
cardiac fibroblasts is essential for myocardial
ischemia/reperfusion injury. Circulation 123,
594604 (2011).
32. McCartney,S.A. etal. RNA sensor-induced typeI IFN
prevents diabetes caused by a cell-tropic virus in
mice. J.Clin. Invest. 121, 14971507 (2011).
33. Philip,J., Xu,Z., Bowles,N.E. & Vallejo,J.G.
Cardiac-specific overexpression of melanoma
differentiation-associated gene5 protects mice from
lethal viral myocarditis. Circ. Heart Fail. 6, 326334
(2013).
34. Lech,M. etal. Quantitative expression of Ctype lectin
receptors in humans and mice. Int. J.Mol. Sci. 13,
1011310131 (2012).
35. Frangogiannis,N.G. & Entman,M.L. Chemokines in
myocardial ischemia. Trends Cardiovasc. Med. 15,
163169 (2005).
36. Frangogiannis,N.G. The mechanistic basis of infarct
healing. Antioxid. Redox. Signal. 8, 19071939 (2006).
37. Chakraborti,T., Mandal,A., Mandal,M., Das,S. &
Chakraborti,S. Complement activation in heart
diseases. Role of oxidants. Cell Signal. 12, 607617
(2000).
38. Foreman,K.E., Glovsky,M.M., Warner,R.L.,
Horvath,S.J. & Ward,P.A. Comparative effect of C3a
and C5a on adhesion molecule expression on
neutrophils and endothelial cells. Inflammation 20,
19 (1996).
39. Bhattacharya,K. etal. Mast cell deficient W/Wv mice
have lower serum IL6 and less cardiac tissue necrosis
than their normal littermates following myocardial
ischemia-reperfusion. Int. J.Immunopathol.
Pharmacol. 20, 6974 (2007).
40. Ayach,B.B. etal. Stem cell factor receptor induces
progenitor and natural killer cell-mediated cardiac
survival and repair after myocardial infarction.
Proc. Natl Acad. Sci. USA 103, 23042309 (2006).
41. Waskow,C., Paul,S., Haller,C., Gassmann,M. &
Rodewald,H.R. Viable cKit(W/W) mutants reveal
pivotal role for ckit in the maintenance of
lymphopoiesis. Immunity 17, 277288 (2002).
42. Dreyer,W.J. etal. Kinetics of C5a release in cardiac
lymph of dogs experiencing coronary artery ischemiareperfusion injury. Circ. Res. 71, 15181524 (1992).
43. Newburger,P.E. & Dale,D.C. Evaluation and
management of patients with isolated neutropenia.
Semin. Hematol. 50, 198206 (2013).
44. Singh,M. & Saini,H.K. Resident cardiac mast cells
and ischemia-reperfusion injury. J.Cardiovasc.
Pharmacol. Ther. 8, 135148 (2003).
45. McDonald,B. etal. Intravascular danger signals
guide neutrophils to sites of sterile inflammation.
Science 330, 362366 (2010).
46. Li,W. etal. Intravital 2photon imaging of leukocyte
trafficking in beating heart. J.Clin. Invest. 122,
24992508 (2012).
47. Gwechenberger,M. etal. Cardiac myocytes produce
interleukin6 in culture and in viable border zone of
reperfused infarctions. Circulation 99, 546551
(1999).

NATURE REVIEWS | IMMUNOLOGY

48. Youker,K. etal. Neutrophil adherence to isolated


adult cardiac myocytes. Induction by cardiac lymph
collected during ischemia and reperfusion.
J.Clin. Invest. 89, 602609 (1992).
This important early study demonstrated the role
of neutrophils in cardiomyocyte injury.
49. Entman,M.L. etal. Neutrophil induced oxidative
injury of cardiac myocytes. A compartmented system
requiring CD11b/CD18ICAM1 adherence.
J.Clin. Invest. 90, 13351345 (1992).
50. Entman,M.L. etal. Neutrophil adherence to isolated
adult canine myocytes. Evidence for a
CD18dependent mechanism. J.Clin. Invest. 85,
14971506 (1990).
51. Tyagi,S., Klickstein,L.B. & Nicholson-Weller,A.
C5astimulated human neutrophils use a subset of 2
integrins to support the adhesion-dependent phase of
superoxide production. J.Leukoc. Biol. 68, 679686
(2000).
52. Kawakami,R. etal. Overexpression of brain
natriuretic peptide facilitates neutrophil infiltration
and cardiac matrix metalloproteinase9 expression
after acute myocardial infarction. Circulation 110,
33063312 (2004).
53. Romson,J.L. etal. Reduction of the extent of
ischemic myocardial injury by neutrophil depletion in
the dog. Circulation 67, 10161023 (1983).
54. Jolly,S.R. etal. Reduction of myocardial infarct size
by neutrophil depletion: effect of duration of occlusion.
Am. Heart J. 112, 682690 (1986).
55. Van Furth,R. & Cohn,Z.A. The origin and kinetics
of mononuclear phagocytes. J.Exp. Med. 128,
415435 (1968).
56. Ginhoux,F. etal. Fate mapping analysis reveals that
adult microglia derive from primitive macrophages.
Science 330, 841845 (2010).
57. Schulz,C. etal. A lineage of myeloid cells independent
of Myb and hematopoietic stem cells. Science 336,
8690 (2012).
58. Yona,S. etal. Fate mapping reveals origins and
dynamics of monocytes and tissue macrophages
under homeostasis. Immunity. 38, 7991 (2013).
59. Hashimoto,D. etal. Tissue-resident macrophages
self-maintain locally throughout adult life with
minimal contribution from circulating monocytes.
Immunity. 38, 792804 (2013).
60. Guilliams,M. etal. Alveolar macrophages develop
from fetal monocytes that differentiate into long-lived
cells in the first week of life via GMCSF. J.Exp. Med.
210, 19771992 (2013).
61. Jakubzick,C. etal. Minimal differentiation of classical
monocytes as they survey steady-state tissues and
transport antigen to lymph nodes. Immunity. 39,
599610 (2013).
62. Hanna,R.N. etal. The transcription factor NR4A1
(Nur77) controls bone marrow differentiation and the
survival of Ly6C- monocytes. Nature Immunol. 12,
778785 (2011).
63. Hettinger,J. etal. Origin of monocytes and
macrophages in a committed progenitor.
Nature Immunol. 14, 821830 (2013).
64. Ingersoll,M.A. etal. Comparison of gene expression
profiles between human and mouse monocyte
subsets. Blood 115, e10e19 (2010).
65. Auffray,C. etal. Monitoring of blood vessels and
tissues by a population of monocytes with patrolling
behavior. Science 317, 666670 (2007).
66. Carlin,L.M. etal. Nr4a1dependent Ly6Clow
monocytes monitor endothelial cells and orchestrate
their disposal. Cell 153, 362375 (2013).
67. Frangogiannis,N.G. etal. Critical role of monocyte
chemoattractant protein1/CC chemokine ligand 2
in the pathogenesis of ischemic cardiomyopathy.
Circulation 115, 584592 (2007).
An important early study showing that CCR2
deficiency limits cardiac injury. Later studies
would suggest this was due to a lack of blood
monocytes.
68. Hilgendorf,I. etal. Ly6Chigh monocytes depend on
Nr4a1 to balance both inflammatory and reparative
phases in the infarcted myocardium. Circ. Res.114,
16111622 (2014).
69. Leuschner,F. etal. Angiotensin-converting enzyme
inhibition prevents the release of monocytes from
their splenic reservoir in mice with myocardial
infarction. Circ. Res. 107, 13641373 (2010).
70. Swirski,F.K. etal. Identification of splenic reservoir
monocytes and their deployment to inflammatory
sites. Science 325, 612616 (2009).
This is the first study to show that the spleen can
be a source of monocytes following cardiac injury.

VOLUME 15 | FEBRUARY 2015 | 127


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
71. Leuschner,F. etal. Rapid monocyte kinetics in acute
myocardial infarction are sustained by extramedullary
monocytopoiesis. J.Exp. Med. 209, 123137
(2012).
72. Molawi,K. etal. Progressive replacement of embryoderived cardiac macrophages with age. J.Exp. Med.
211, 21512158 (2014).
73. Bracey,N.A. etal. Mitochondrial NLRP3 protein
induces reactive oxygen species to promote Smad
protein signaling and fibrosis independent from the
inflammasome. J.Biol. Chem. 289, 1957119584
(2014).
74. Bracey,N.A. etal. The Nlrp3 inflammasome
promotes myocardial dysfunction in structural
cardiomyopathy through interleukin1.
Exp. Physiol. 98, 462472 (2013).
75. Dunay,I.R. etal. Gr1+ inflammatory monocytes
are required for mucosal resistance to the pathogen
Toxoplasma gondii. Immunity 29, 306317
(2008).
76. Kim,Y.G. etal. The Nod2 sensor promotes intestinal
pathogen eradication via the chemokine
CCL2dependent recruitment of inflammatory
monocytes. Immunity 34, 769780 (2011).
77. van Amerongen,M.J., Harmsen,M.C.,
van Rooijen,N., Petersen,A.H. & van Luyn,M.J.
Macrophage depletion impairs wound healing and
increases left ventricular remodeling after
myocardial injury in mice. Am. J.Pathol. 170,
818829 (2007).
78. Panizzi,P. etal. Impaired infarct healing in
atherosclerotic mice with Ly6C(hi) monocytosis.
J.Am. Coll. Cardiol. 55, 16291638 (2010).
79. Dewald,O. etal. CCL2/monocyte chemoattractant
protein1 regulates inflammatory responses critical to
healing myocardial infarcts. Circ. Res. 96, 881889
(2005).
80. Leuschner,F. etal. Therapeutic siRNA silencing in
inflammatory monocytes in mice. Nature Biotech. 29,
10051010 (2011).
81. Serbina,N.V. & Pamer,E.G. Monocyte emigration
from bone marrow during bacterial infection requires
signals mediated by chemokine receptor CCR2.
Nature Immunol. 7, 311317 (2006).
82. Zhou,L. etal. Monocyte chemoattractant protein1
induces a novel transcription factor that causes
cardiac myocyte apoptosis and ventricular
dysfunction. Circ. Res. 98, 11771185 (2006).
83. Aurora,A.B. etal. Macrophages are required for
neonatal heart regeneration. J.Clin. Invest. 124,
13821392 (2014).
84. Porrello,E.R. etal. Transient regenerative potential of
the neonatal mouse heart. Science 331, 10781080
(2011).
This is the first study to show that the mammalian
neonatal heart can regenerate fully, similarly to
what is observed in more primitive organisms.
85. Lavine,K. etal. Distinct macrophage lineages
contribute to disparate patterns of cardiac recovery
and remodeling in the neonatal and adult heart.
Proc. Natl Acad. Sci. 111, 1602916034 (2014).
This is the first study to show that resident
neonatal heart macrophages, not recruited
monocytes, have a key role in neonatal heart
regeneration. Similar findings were seen in
reference 83.
86. Marodi,L. Neonatal innate immunity to infectious
agents. Infect. Immun. 74, 19992006 (2006).
87. Munoz-Espin,D. etal. Programmed cell senescence
during mammalian embryonic development.
Cell 155, 11041118 (2013).
88. Storer,M. etal. Senescence is a developmental
mechanism that contributes to embryonic growth
and patterning. Cell 155, 11191130 (2013).
89. Wan,E. etal. Enhanced efferocytosis of apoptotic
cardiomyocytes through myeloid-epithelialreproductive tyrosine kinase links acute inflammation
resolution to cardiac repair after infarction.
Circ. Res. 113, 10041012 (2013).
90. Gautier,E.L. etal. Gene-expression profiles and
transcriptional regulatory pathways that underlie the
identity and diversity of mouse tissue macrophages.
Nature Immunol. 13, 11181128 (2012).
91. Mounier,R. etal. AMPK1 regulates macrophage
skewing at the time of resolution of inflammation
during skeletal muscle regeneration. Cell. Metab. 18,
251264 (2013).
92. Arnold,L. etal. Inflammatory monocytes recruited
after skeletal muscle injury switch into
antiinflammatory macrophages to support
myogenesis. J.Exp. Med. 204, 10571069 (2007).

93. Hofmann,U. etal. Activation of CD4+ T lymphocytes


improves wound healing and survival after
experimental myocardial infarction in mice.
Circulation 125, 16521663 (2012).
94. Matsumoto,K. etal. Regulatory T lymphocytes
attenuate myocardial infarction-induced ventricular
remodeling in mice. Int. Heart J. 52, 382387 (2011).
95. Dobaczewski,M., Xia,Y., Bujak,M.,
Gonzalez-Quesada,C. & Frangogiannis,N.G.
CCR5 signaling suppresses inflammation and reduces
adverse remodeling of the infarcted heart, mediating
recruitment of regulatory Tcells. Am. J.Pathol. 176,
21772187 (2010).
96. Yang,Z. etal. Myocardial infarct-sparing effect of
adenosine A2A receptor activation is due to its action
on CD4+ T lymphocytes. Circulation 114, 20562064
(2006).
97. Clynes,R. Protective mechanisms of IVIG.
Curr. Opin. Immunol. 19, 646651 (2007).
98. Curato,C. etal. Identification of noncytotoxic and
IL10producing CD8+AT2R+ Tcell population in
response to ischemic heart injury. J.Immunol. 185,
62866293 (2010).
99. Kaschina,E. etal. Angiotensin II type2 receptor
stimulation: a novel option of therapeutic interference
with the renin-angiotensin system in myocardial
infarction? Circulation 118, 25232532 (2008).
100. Cohn,J.N. & Tognoni,G. A randomized trial of the
angiotensin-receptor blocker valsartan in chronic heart
failure. N.Engl. J.Med. 345, 16671675 (2001).
101. Dickstein,K. & Kjekshus,J. Effects of losartan and
captopril on mortality and morbidity in high-risk
patients after acute myocardial infarction: the
OPTIMAAL randomised trial. Optimal Trial in
Myocardial Infarction with Angiotensin II Antagonist
Losartan. Lancet 360, 752760 (2002).
102. Mehta,P.K. & Griendling,K.K. Angiotensin II cell
signaling: physiological and pathological effects in the
cardiovascular system. Am. J.Physiol. Cell Physiol.
292, C82C97 (2007).
103. Bouchentouf,M. etal. Induction of cardiac
angiogenesis requires killer cell lectin-like receptor 1
and 47 integrin expression by NK cells. J.Immunol.
185, 70147025 (2010).
104. Fadok,V.A. etal. Macrophages that have ingested
apoptotic cells invitro inhibit proinflammatory
cytokine production through autocrine/paracrine
mechanisms involving TGF-, PGE2, and PAF.
J.Clin. Invest. 101, 890898 (1998).
105. Voll,R.E. etal. Immunosuppressive effects of
apoptotic cells. Nature 390, 350351 (1997).
106. Xue,J. etal. Transcriptome-based network analysis
reveals a spectrum model of human macrophage
activation. Immunity 40, 274288 (2014).
107. Stark,M.A. etal. Phagocytosis of apoptotic
neutrophils regulates granulopoiesis via IL23 and
IL17. Immunity 22, 285294 (2005).
108. Tan,W. etal. IL17F/IL17R interaction stimulates
granulopoiesis in mice. Exp. Hematol. 36,
14171427 (2008).
109. Schwarzenberger,P. etal. Requirement of
endogenous stem cell factor and granulocyte-colonystimulating factor for IL17mediated granulopoiesis.
J.Immunol. 164, 47834789 (2000).
110. Yan,X. etal. Deleterious effect of the IL23/IL17A
axis and T cells on left ventricular remodeling after
myocardial infarction. J.Am. Heart Assoc. 1,
e004408 (2012).
111. Liao,Y.H. etal. Interleukin17A contributes to
myocardial ischemia/reperfusion injury by regulating
cardiomyocyte apoptosis and neutrophil infiltration.
J.Am. Coll. Cardiol. 59, 420429 (2012).
112. Feng,W. etal. IL17 induces myocardial fibrosis and
enhances RANKL/OPG and MMP/TIMP signaling in
isoproterenol-induced heart failure. Exp. Mol. Pathol.
87, 212218 (2009).
113. Savvatis,K. etal. Interleukin23 deficiency leads to
impaired wound healing and adverse prognosis after
myocardial infarction. Circ. Heart Fail. 7, 161171
(2014).
114. Haudek,S.B. etal. Rho kinase1 mediates cardiac
fibrosis by regulating fibroblast precursor cell
differentiation. Cardiovasc. Res. 83, 511518 (2009).
115. Lim,D.S. etal. Angiotensin II blockade reverses
myocardial fibrosis in a transgenic mouse model of
human hypertrophic cardiomyopathy. Circulation 103,
789791 (2001).
116. Xu,J. etal. CCR2 mediates the uptake of bone
marrow-derived fibroblast precursors in angiotensin
IIinduced cardiac fibrosis. Am. J.Physiol. Heart Circ.
Physiol. 301, H538H547 (2011).

128 | FEBRUARY 2015 | VOLUME 15

117. Haudek,S.B. etal. Monocytic fibroblast precursors


mediate fibrosis in angiotensin-IIinduced cardiac
hypertrophy. J.Mol. Cell Cardiol. 49, 499507
(2010).
118. Sadoshima,J. & Izumo,S. Molecular characterization
of angiotensin II-induced hypertrophy of cardiac
myocytes and hyperplasia of cardiac fibroblasts.
Critical role of the AT1 receptor subtype. Circ. Res.
73, 413423 (1993).
119. Marko,L. etal. Interferon- signaling inhibition
ameliorates angiotensin IIinduced cardiac damage.
Hypertension 60, 14301436 (2012).
120. Han,Y.L. etal. Reciprocal interaction between
macrophages and Tcells stimulates IFN- and
MCP1 production in Ang IIinduced cardiac
inflammation and fibrosis. PLoS ONE. 7, e35506
(2012).
121. Crawford,J.R., Haudek,S.B., Cieslik,K.A., Trial,J. &
Entman,M.L. Origin of developmental precursors
dictates the pathophysiologic role of cardiac
fibroblasts. J.Cardiovasc. Transl. Res. 5, 749759
(2012).
122. Coura,J.R. & Borges-Pereira,J. Chagas disease:
100years after its discovery. A systemic review.
Acta Trop. 115, 513 (2010).
123. Kindermann,I. etal. Update on myocarditis.
J.Am. Coll. Cardiol. 59, 779792 (2012).
124. Neu,N. etal. Cardiac myosin induces myocarditis in
genetically predisposed mice. J.Immunol. 139,
36303636 (1987).
125. Sagar,S., Liu,P.P. & Cooper,L.T.Jr. Myocarditis.
Lancet 379, 738747 (2012).
126. Kuhl,U. etal. High prevalence of viral genomes and
multiple viral infections in the myocardium of adults
with idiopathic left ventricular dysfunction.
Circulation 111, 887893 (2005).
127. Martino,T.A. etal. The coxsackie-adenovirus receptor
(CAR) is used by reference strains and clinical isolates
representing all six serotypes of coxsackievirus group
B and by swine vesicular disease virus. Virology 271,
99108 (2000).
128. Coyne,C.B. & Bergelson,J.M. Virus-induced Abl
and Fyn kinase signals permit coxsackievirus entry
through epithelial tight junctions. Cell 124, 119131
(2006).
This was the first study to show how cardiotropic
viruses penetrate host epithelial barriers.
129. Liu,P.P. & Opavsky,M.A. Viral myocarditis:
receptors that bridge the cardiovascular with the
immune system? Circ. Res. 86, 253254 (2000).
130. Kallewaard,N.L. etal. Tissue-specific deletion
of the coxsackievirus and adenovirus receptor
protects mice from virus-induced pancreatitis
and myocarditis. Cell Host Microbe 6, 9198
(2009).
131. Liu,P. etal. The tyrosine kinase p56lck is essential
in coxsackievirus B3mediated heart disease.
Nature Med. 6, 429434 (2000).
The first study to detail the mechanisms through
which cardiotropic viruses mediate intracellular
signalling events and cell injury.
132. Irie-Sasaki,J. etal. CD45 is a JAK phosphatase and
negatively regulates cytokine receptor signalling.
Nature 409, 349354 (2001).
133. Valaperti,A. etal. Innate immune interleukin1
receptor-associated kinase 4 exacerbates viral
myocarditis by reducing CCR5+ CD11b+ monocyte
migration and impairing interferon production.
Circulation 128, 15421554 (2013).
134. Riad,A. etal. Myeloid differentiation factor88
contributes to TLR9mediated modulation of acute
coxsackievirus B3induced myocarditis invivo.
Am. J.Physiol. Heart Circ. Physiol. 298,
H2024H2031 (2010).
135. Fuse,K. etal. Myeloid differentiation factor88 plays a
crucial role in the pathogenesis of Coxsackievirus
B3induced myocarditis and influences typeI
interferon production. Circulation 112, 22762285
(2005).
136. Holm,G.H. etal. Interferon regulatory factor 3
attenuates reovirus myocarditis and contributes to
viral clearance. J.Virol. 84, 69006908 (2010).
137. Opavsky,M.A. etal. Susceptibility to myocarditis is
dependent on the response of T lymphocytes to
coxsackieviral infection. Circ. Res. 85, 551558
(1999).
138. Shi,Y. etal. Regulatory Tcells protect mice against
coxsackievirus-induced myocarditis through the
transforming growth factor -coxsackie-adenovirus
receptor pathway. Circulation 121, 26242634
(2010).

www.nature.com/reviews/immunol
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
139. Mason,J.W. etal. A clinical trial of immunosuppressive
therapy for myocarditis. The Myocarditis Treatment
Trial Investigators. N.Engl. J.Med. 333, 269275
(1995).
140. Kuhl,U. etal. Interferon- treatment eliminates
cardiotropic viruses and improves left ventricular
function in patients with myocardial persistence of
viral genomes and left ventricular dysfunction.
Circulation 107, 27932798 (2003).
141. Poss,K.D., Wilson,L.G. & Keating,M.T.
Heart regeneration in zebrafish. Science 298,
21882190 (2002).
142. Laube,F., Heister,M., Scholz,C., Borchardt,T. &
Braun,T. Reprogramming of newt cardiomyocytes is
induced by tissue regeneration. J.Cell Sci. 119,
47194729 (2006).
143. Godwin,J.W., Pinto,A.R. & Rosenthal,N.A.
Macrophages are required for adult salamander
limb regeneration. Proc. Natl Acad. Sci. USA 110,
94159420 (2013).

144. Buchmann,K. Evolution of innate immunity:


clues from invertebrates via fish to mammals.
Front. Immunol. 5, 459 (2014).
145. Epelman,S., Lavine,K.J. & Randolph,G.J.
Origin and functions of tissue macrophages.
Immunity 41, 2135 (2014).
146. Dai,X.M. etal. Targeted disruption of the mouse
colony-stimulating factor 1 receptor gene results in
osteopetrosis, mononuclear phagocyte deficiency,
increased primitive progenitor cell frequencies, and
reproductive defects. Blood 99, 111120 (2002).
147. McKercher,S.R. etal. Targeted disruption of the
PU.1 gene results in multiple hematopoietic
abnormalities. EMBO J. 15, 56475658 (1996).
148. Wiktor-Jedrzejczak,W. etal. Total absence of colonystimulating factor 1 in the macrophage-deficient
osteopetrotic (op/op) mouse. Proc. Natl Acad. Sci.
USA 87, 48284832 (1990).
149. Nucera,S., Biziato,D. & De,P.M. The interplay
between macrophages and angiogenesis in

NATURE REVIEWS | IMMUNOLOGY

development, tissue injury and regeneration.


Int. J.Dev. Biol. 55, 495503 (2011).
150. Fantin,A. etal. Tissue macrophages act as cellular
chaperones for vascular anastomosis downstream
of VEGF-mediated endothelial tip cell induction.
Blood 116, 829840 (2010).
151. Arnold,T. & Betsholtz,C. The importance of microglia
in the development of the vasculature in the central
nervous system. Vasc. Cell 5, 4 (2013).
152. Lobov,I.B. etal. WNT7b mediates macrophageinduced programmed cell death in patterning
of the vasculature. Nature 437, 417421 (2005).
153. Naqvi,N. etal. A proliferative burst during
preadolescence establishes the final cardiomyocyte
number. Cell 157, 795807 (2014).
154. Bergmann,O. etal. Evidence for cardiomyocyte
renewal in humans. Science 324, 98102 (2009).

Competing interests statement

The authors declare no competing interests.

VOLUME 15 | FEBRUARY 2015 | 129


2015 Macmillan Publishers Limited. All rights reserved

Reproduced with permission of the copyright owner. Further reproduction prohibited without
permission.

Potrebbero piacerti anche