You are on page 1of 11

REVIEWS

The inflammatory response in myocardial


injury, repair, and remodelling
Nikolaos G. Frangogiannis
Abstract | Myocardial infarction triggers an intense inflammatory response that is essential for cardiac repair,
but which is also implicated in the pathogenesis of postinfarction remodelling and heart failure. Signals in the
infarcted myocardium activate toll-like receptor signalling, while complement activation and generation of reactive
oxygen species induce cytokine and chemokine upregulation. Leukocytes recruited to the infarcted area, remove
dead cells and matrix debris by phagocytosis, while preparing the area for scar formation. Timely repression of
the inflammatory response is critical for effective healing, and is followed by activation of myofibroblasts that
secrete matrix proteins in the infarcted area. Members of the transforming growth factor family are critically
involved in suppression of inflammation and activation of a profibrotic programme. Translation of these concepts
to the clinic requires an understanding of the pathophysiological complexity and heterogeneity of postinfarction
remodelling in patients with myocardial infarction. Individuals with an overactive and prolonged postinfarction
inflammatory response might exhibit left ventricular dilatation and systolic dysfunction and might benefit from
targeted anti-IL1 or anti-chemokine therapies, whereas patients with an exaggerated fibrogenic reaction can
develop heart failure with preserved ejection fraction and might require inhibition of the Smad3 (mothers against
decapentaplegic homolog3) cascade. Biomarker-based approaches are needed to identify patients with distinct
pathophysiologic responses and to rationally implement inflammation-modulating strategies.
Frangogiannis, N.G. Nat. Rev. Cardiol. 11, 255265 (2014); published online 25 March 2014; doi:10.1038/nrcardio.2014.28

Introduction
More than 70years ago, cardiac pathologists noted that the critical role of the inflammatory cascade in response
myocardial infarction triggers an intense inflammatory to cardiac injury, and the involvement of inflammatory
reaction characterized by infiltration of leukocytes into the mediators in repair and remodelling of the infarcted heart,
infarcted heart.1 In the following decades, recognition of the reduced interest in this therapeutic direction was
the injurious properties of leukocytes and their close asso- unfortunate. The pathogenesis of heart failure after myo-
ciation with cardiomyocytes in the viable border zone of cardial infarction is intricately linked with the develop-
an infarct suggested that subpopulations of blood-derived ment of postinfarction ventricular remodelling. Structural,
cells can adhere to viable cardiomyocytes and might exert functional, and geometric alterations that involve both the
cytotoxic effects extending ischaemic injury (Figure1).2 infarcted and noninfarcted myocardial segments and lead
In the 1980s and 1990s, experimental studies demon- to chamber dilatation, increase sphericity of the ventricle
strated that targeting leukocyte-mediated inflammation and cardiac dysfunction.7 Cardiac remodelling is asso-
in reperfused myocardial infarction markedly reduced the ciated with the progression of heart failure, increased
size of the infarct, and thereby prevented an extension of incidence of arrhythmias, and poor prognosis in patients
ischaemic cardiomyocyte injury.36 Approaches targeting surviving a myocardial infarction.8 The extent of post
molecules involved in leukocyte activation, adhesion, and infarction remodelling is dependent on the infarct size
extravasation (such as integrins, selectins, and components and quality of cardiac repair.9 Experimental studies have
of the complement cascade) were successful in animal called into question the notion that inflammatorysignals
studies in attenuating ischaemic injury, leading to con- can extend ischaemic injury,10,11 but inflammatory path-
siderable enthusiasm for their potential use in humans.35 ways are undoubtedly critically involved in dilative and
Unfortunately, despite promising data from animal studies, fibrotic remodelling of the infarcted heart and, therefore,
translation of leukocyte-focused treatment into therapy drive key events in the pathogenesis of postinfarction
for human populations with myocardial infarction was heart failure.
The Wilf Family
Cardiovascular
unsuccessful, and several anti-inflammatory approaches In this Review, I discuss the role of inflammatory signals
Research Institute, failed to reduce infarct size in clinical investigations.6 in regulating repair and remodelling of the infarcted heart,
Albert Einstein College The disappointment from these early negative clinical and the attempts to identify therapeutic targets. From
of Medicine, 1300
Morris Park Avenue, results had lasting consequences in the field. Considering advances in the understanding of pathophysiology of
Forchheimer G46B, cardiac remodelling, I will attempt to provide a guide for
Bronx, NY 10461, USA.
nikolaos.frangogiannis@ Competing interests development of anti-inflammatory treatments for patients
einstein.yu.edu The author declares no competing interests. who survive a myocardial infarction.

NATURE REVIEWS | CARDIOLOGY VOLUME 11 | MAY 2014 | 255


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points cells. Themyofibroblasts secrete large amounts of extra


cellular matrix proteins, thereby preserving the structural
In the infarcted myocardium, cardiomyocyte death and degradation of the
integrity of the left ventricle. Apoptosis of themajority of
cardiac extracellular matrix releases signals that activate innate immune
pathways and trigger an intense inflammatory reaction
reparative cells marks the end of the proliferative phase, as
The role of postinfarction inflammation in extending ischaemic cardiomyocyte the infarct matures and a scar comprised of crosslinked
injury is controversial; however, inflammatory mediators are implicated in collagen is formed.12
dilative remodelling and in the pathogenesis of postinfarction heart failure
Early stimulation of inflammatory signalling is important for clearance of dead Necrosis and innate immune signals
cells from the infarcted area and for tissue repair Tissue injury generates endogenous signals that acti-
Timely repression of proinflammatory mediators protects the heart from vate the innate immune system; these molecules belong
excessive inflammatory injury
to a large family of mediators that warn the body of
Patients who survive a large myocardial infarction exhibit pathophysiological
heterogeneity, as subpopulations with progressive dilative remodelling or
injury and are known as danger-associated molecular
predominant diastolic dysfunction are identified patterns (DAMPs).13,14 The term alarmins describes a
Biomarker-based approaches are needed to identify patients with overactive group of structurally diverse endogenous signals that,
proinflammatory signalling or excessive fibrosis who might benefit from when released after tissue necrosis, promote activation
targeted therapies of innate immune cells by binding to pattern recogni-
tion receptors.15 The best characterized alarminhigh
mobility group proteinB1 (HMGB1)is a key initiator
Postinfarction inflammatory response of inflammatory injury following myocardial ischaemia
The adult mammalian heart has little regenerative capa through actions that might involve toll-like receptors
city, therefore, healing of the infarcted myoc ardium (TLRs), a family of transmembrane receptors that activate
is dependent on an orchestrated sequence of cellular downstream proinflammatory cascades, and the recep-
events that lead to the formation of a collagen-based scar. tor for advanced glycation end products (RAGE). 16,17
Repair of the infarcted myocardium can be described in Considering the fundamental role of alarmin-mediated
three overlapping phases: the inflammatory phase,the signalling in inflammation and repair, the fact that
proliferative phase, and the maturation phase.12 Acute both detrimental16,17 and beneficial18 effects of HMGB1
sudden death of cardiomyocytes in the infarcted heart have been reported in the infarcted myocardium is
rapidly activates innate immune pathways that trigger notsurprising.
an intense, but transient, inflammatory reaction. This Other intracellular constituents released by necrotic
response clears the infarcted area of dead cells and cells, such as heat shock proteins and ATP, might also
extracellular matrix debris, and is then actively repressed activate an immune response in the infarcted heart.13,1921
to prepare the infarct for the proliferative phase of The damaged extracellular matrix might also transduce
healing. During theproliferative phase, mononuclear key signals for activation of innate immune cells in the
cell and macrophage subpopulations secrete growth infarcted heart. Low molecular weight hyaluronan and
factors that recruit and activate mesenchymal repara- fibronectin fragments can activate TLRs and function as
tive cellspredominantly myofibroblasts and vascular important initiators of proinflammatory signalling.22,23

a b c Capture Activation
Rolling
Arrest

Neutrophil Transmigration

Endothelial cells
Adhesion

Proteases
Cardiomyocyte Cytotoxic ROS
injury Phagocytosis
Figure 1 | Cytotoxic inflammatory injury after myocardial infarction. Myocardial infarction is associated with an intense
inflammatory reaction and infiltration of the infarct with abundant leukocytes. a | Canine infarct (1h coronary occlusion/24h
reperfusion) stained with MAC387 (red), a marker for newly recruited myeloid cells (neutrophils and monocytes), and an
antimacrophage antibody (black). Abundant, newly recruited leukocytes closely associated with viable cardiomyocytes.
b | The same canine infarct after 7days of reperfusion. The density of MAC387 positive cells is markedly reduced. However,
mature macrophages are still abundant (black) reflecting repression of the acute inflammatory reaction. c | The close spatial
association between leukocytes and viable cardiomyocytes in the border zone and the injurious potential of subsets of blood-
derived cells generated the concept of leukocyte-mediated cardiomyocyte injury. Neutrophils interact with endothelial cells,
roll along the endothelial surface, decelerate to a firm arrest, transmigrate across the vascular wall, infiltrate the infarct, and
adhere to viable cardiomyocytes exerting cytotoxic effects and extending ischaemic injury. Infiltrating leukocytes are also
important for infarct repair by releasing proteases and ROS, thereby clearing the wound from dead cells and debris.
Abbreviation: ROS, reactive oxygen species.

256 | MAY 2014 | VOLUME 11  www.nature.com/nrcardio


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Innate immune cells identify danger signals via engage- infiltrating the infarcted myocardium and understand-
ment of TLRs. Of the 13 members of the mammalian ing of the mechanisms mediating their recruitment has
TLR family, genetic loss-of-function studies indicate important therapeutic implications.
thatTLR2 and TLR4, both localized on the cell surface,
are important mediators of the postinfarction inflam The role of proinflammatory cytokines
matory reaction.2426 Activation of the complement system Expression of the proinflammatory cytokines TNF,
is also involved in transducing the immune response in IL1, and IL6 is markedly and consistently increased
the infarcted heart, and complement inhibition attenu- in experimental models of myocardial infarction.36,37
ates leukocyte recruitment after myocardial infarction.27 However, the pleiotropic properties of these cytokines
Ischaemia-mediated generation of reactive oxygen species and their multifunctional effects on all cell types involved
(ROS) is also important for the activation of inflammatory in cardiac injury and repair have hampered our under-
signals in the infarcted myocardium. ROS promote leuko standing of their functional roles in the infarcted and
cyte infiltration into the healing infarct by activating all remodelling heart. TNF is released following myocardial
steps of inflammatory cell recruitment.2830 ROS contrib- infarction,38 and can promote inflammatory injury indu
ute to generation of chemotactic gradients in three ways: cing chemokine and adhesion molecule synthesis in the
by inducing chemokine and cytokine expression,28 by pro- infarcted myocardium.39 As a highly pleiotropic mediator,
moting leukocyte integrin activation,29 and by inducing TNF can also protect cardiomyocytes from apoptosis.40
adhesion molecule synthesis.30 Divergent effects transduced through TNF receptors1
and 2 might regulate remodelling of the infarcted heart.41
Chemokines and cytokines The failure of anti-TNF strategies in patients with chronic
Activation of alarmin-mediated signalling induces a heart failure42 might reflect the pleiotropic actions of the
molecular programme that leads to the recruitmentof cytokine, and has discouraged the design of approaches
inflammatory cells in the healing infarct. Induction targeting the pathway in myocardial infarction. IL1
ofproinflammatory chemokines in the infarcted heart is also markedly induced in the infarct and mediates
generates chemotactic gradients that recruit leukocyte inflammatory leukocyte recruitment and activation,43,44
subpopulations via interactions with the corresponding while delaying myofibroblast activation.44 No protective
chemokine receptors. Upregulation of proinflammatory effects of IL1 on cardiomyocytes have been reported,
cytokines (such as tumor necrosis factor [TNF], IL1 and and because IL1 neutralization can attenuate cardio
members of the IL6 family) induce endothelial cell adhe- myocyte apoptosis invitro and invivo,45 inhibition of
sion molecule synthesis and activate leukocyte integrins, IL1 following myocardial infarction might have no major
mediating strong adhesive interactions that ultimately lead detrimental consequences.46
to extravasation of inflammatory cells into the infarct.9 IL6 is also upregulated in the infarcted myocardium,
and might modulate the inflammatory and reparative
The role of chemokines response signalling through IL6 receptor subunit beta
Expression of both major chemokine subfamilies (the and activating the JAK/STAT (janus tyrosine kinase/
CCand CXC chemokines) is increased in the infarcted signal transducer and activator of transcription) cascade.47
heart and mediates recruitment of inflammatory leuko Blocking IL6 function has been effective in the treat-
cytes. 31 CXC chemokines containing the tripeptide ment of rheumatic disorders.48 However, the pleiotropic
sequence GluLeuArg (known as the ELR motif, and also effectsof IL6 in the healing infarct and the induction of
found in IL8 in placental or eutherian mammals andthe other IL6 receptor family members that might compen-
corresponding CXCR2 ligands in mice),32 are rapidly sate for the loss of this cytokine, raise concerns about its
induced in the infarcted myocardium and mediate recruit- potential role as a therapeutic target in patients with myo-
ment of neutrophils.33 Trafficking of mononuclear cell cardial infarction. Experimental studies in IL6-knockout
subsets involves CC chemokine signalling; distinct inter mice demonstrated that the loss of IL6 does not affect
actions between chemokines and their receptors might be cardiac function and remodelling in a model of non
responsible for recruitment of different subpopulations of reperfused infarction.49 Other IL6 family cytokines might
mononuclear cells. CC motif chemokine2 (also known compensate for the loss of IL6 by activating JAK/STAT
asmonocyte chemoattractant protein1 [MCP1]) is signalling and maintaining STAT3 phosphorylation.
rapidly upregulated in the infarcted myocardium and Conversely, treatment with an anti-IL6 receptor anti-
mediates recruitment of proinflammatory phagocytotic body attenuated adverse remodelling in a mouse model
monocytes that clear the area of dead cells and matrix of nonreperfused infarction.50 Timely downregulation of
debris.34 Chemokines might also recruit inhibitory and the IL6 response might be important for infarct healing.
reparative mononuclear cell subsets into the infarct; In a mouse model of myocardial infarction, impaired sup-
however, the specific chemokinechemokine-receptor pression of IL6 receptor/STAT3 signalling was associated
pairs mediating these cellular events remain poorly under- with prolonged and enhanced inflammation increasing
stood. Interactions involving the CC chemokine receptor the incidence of cardiac rupture.51
type5 might be involved in recruitment of mononuclear
cell subpopulations with anti-inflammatory proper- Effectors of postinfarct inflammation
ties, such as inhibitory monocyte subsets and regulatory The inflammatory response in the infarcted myo-
Tcells.35 Systematic characterization of monocyte subsets cardium involves cells that are normally found in the

NATURE REVIEWS | CARDIOLOGY VOLUME 11 | MAY 2014 | 257


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Haematopoietic cells Platelets Lymphocyte

1 Monocyte

2 3

4
5

Neutrophil
6
PG

Endothelial cell Matrix


fragments
TLR
Cardiomyocyte
DAMPs

ROS
Fibroblast

Mast
C5a cell

Resident myocardial cells

Figure 2 | The postinfarction inflammatory response. In the infarcted myocardium, dying cardiomyocytes and damaged matrix
release DAMPs that activate TLR signalling in myocardial cells, triggering an inflammatory reaction. Activation of the
complement cascade and ROS generation also help initiate the inflammatory reaction. Dying and surviving cardiomyocytes,
endothelial cells, resident cardiac fibroblasts, resident mast cells and newly recruited neutrophils monocytes and platelets
participate in the post-infarction inflammatory response. However, their relative contributions remain unclear. Leukocytes are
recruited through activation of a multistep adhesion cascade.57 Capture (1) of circulating leukocytes by activated endothelial
cells is followed by rolling (2), mediated through interactions involving the selectins. Rolling leukocytes are activated (3) by
chemokines bound to PG on the endothelial surface. Activated leukocytes express integrins and adhere to endothelial cells
(4). Strengthening of the adhesive interaction (5) between leukocytes and endothelial cells is followed by transmigration of
the cells into the infarcted area (6). Abbreviations: C5a, complement factor 5a; DAMP, danger-associated molecular pattern;
PG, proteoglycan; ROS, reactive oxygen species; TLR, Toll-like receptor.

heart and newly recruited cells (Figure2); however, the chemokine-driven mobilization of proinflammatory
relative contribution of the different cell types remains monocytes in the infarct.60 Reparative monocytes follow
unclear. In the absence of injury, the adult mammalian the proinflammatory cells, but the signals involved their
myocardium contains relatively small populations of recruitment remain poorly understood. Macrophage
macrophages, mast cells,52 and dendritic cells.53 Cardiac subsets with proinflammatory properties also infiltrate
resident mast cells contain preformed proinflammatory the infarct and might sustain a proinflammatory environ
cytokines, and can be rapidly activated following myo- ment in the infarcted myocardium.61 The concept sug-
cardial ischaemia releasing their granular content and gesting recruitment of two polarized populations of
triggering the inflammatory cascade.38 ROS generation, monocytes or macrophages in the infarct represents an
adenosine, and complement factorC5a might stimu- oversimplification, because several subpopulations of cells
late mast cell degranulation.5456 The function of TLR with distinct functional properties (and perhaps also with
ligands in this context is less convincingly established. varying potential for differentiation and activation) are
In the early hours after myocardial infarction, leukocytes probably recruited in the infarcted myocardium.
(neutrophils and mononuclear cells) rapidly infiltrate the As the most abundant noncardiomyocyte population
infarct. Circulating neutrophils are recruited through acti- in the mammalian heart, fibroblasts might also contrib-
vation of both chemokine-dependent 28 and chemokine- ute to the initiation of the inflammatory reaction in the
independent pathways.57,58 Monocyte subpopulations infarcted myocardium. Activation of the inflammasome
infiltrate the myocardium sequentiallyfirst by pro cascade (the molecular platform that triggers activation
inflammatory monocytes that are rapidly mobilized from of inflammatory caspases and processes pro-IL1) has
the bone marrow and the splenic reservoir.59 Recruitment been demonstrated in infarct fibroblasts,62 which might
of inflammatory monocytes into the infarcted heart reflect an important role for these versatile cells in pro-
is the result of marked upregulation of the MCP1.34 inflammatory signalling. During the early stages of the
Experiments in a mouse model of nonreperfused infarc postinfarction response, fibroblasts acquire a proinflam-
tion suggested that B lymphocytes might mediate matory and matrix-degrading phenotype; local release of

258 | MAY 2014 | VOLUME 11  www.nature.com/nrcardio


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

IL1 might inhibit their conversion into matrix-synthetic syndrome have increased mortality in the absence of an
myofibroblasts44 until the infarct microenvironment increase in new coronary events.71 Adverse prognosis
is cleared from dead cells and matrix debris and can in these patients might reflect increased remodelling
support deposition of a new collagen-based matrix. and accentuated injury in individuals with defective
The heart is abundant with blood vessels, endothelial activation of anti-inflammatory pathways.72
cells might, therefore, be important in the synthesis and
release of proinflammatory cytokines and chemokines.31 Anti-inflammatory signalling
Descriptive studies in canine models have identified All cell types involved in cardiac repair likely participate
venular endothelial cells as an important source of chemo in repression and resolution of the postinfarction inflam-
kines in the infarcted myocardium.63,64 ROS generation matory reaction; however, the key cellular effectors that
and activation of TLR signalling by alarmins released drive inhibition of inflammation remain unknown.
by dying cells and matrix debris might mediate inflam- Through their unique cytokine expression profile, and
matory activation of the infarct endothelium. Platelets their potential for regulated recruitment and activation in
also accumulate within the infarcted myocardium and response to local stimuli, inhibitory subsets of monocytes
might be important for the inflammatory reaction, both and lymphocytes and anti-inflammatory macrophages are
through direct release of cytokines and chemokines and ideally suited to suppress inflammation in the infarcted
by modulating phenotype of other celltypes.65 heart. Experimental studies have demonstrated dynamic
Dying cardiomyocytes are crucial for triggering inflam- changes in macrophage phenotype in the infarcted heart,
matory pathway activation through release of DAMPs; which suggests a transition from early infiltration with
however, the potential role of viable border zone cardio- proinflammatory M1 cells tothe late predominance of
myocytes as a source of inflammatory mediators remains reparative M2 macrophages.73 Thesignals leading to these
unclear. In a canine model of reperfused myocardial infarc- phenotypic changes of infarct macrophages remain poorly
tion, border zone cardiomyocytes have been identified as understood, and two questions remain: are macrophage
a source of IL6.66 However, noncardiomyocytes (includ- subsets derived from distinct monocyte subpopulations,
ing leukocytes, vascular cells, and fibroblasts) can produce and do dynamic changes in the infarct microenvironment
large amounts of cytokines and chemokines;38,64,67 there- mediate acquisition of an inhibitory profile?
fore, the relative importance of cardiomyocytederived Evidence suggests that the phagocytotic activity of
inflammatory mediators isunknown. macrophages can be important for modulating their
phenotype and in repression of the inflammatory reac-
Effective repair and inflammation tion. Efficient clearance of apoptotic cells by phagocytes
Repair of injured tissues is dependent on timely suppres- (known as efferocytosis) activates proresolving signals
sion and containment of inflammation. This process is that might aid the transition from inflammation to repair.
accompanied by activation of mesenchymal cells that The induction of tyrosine-protein kinase Mer in macro
restore tissue integrity. Extensive experimental work phages seems to be important for cardiomyocyte effero-
suggests that repression of proinflammatory signalling cytosis and subsequent suppression of the post-infarction
is not a passive process, but requires induction of inhibi- inflammatory reaction.74 Inhibitory lymphoc yte sub-
tory molecules and activation of suppressive pathways.68 populations, such as regulatory Tcells, can participate
In injured tissues, overactive, prolonged, or spatially in suppression of the postinfarction inflammatory
expanded inflammatory reactions lead to accentu- response.35 Moreover, fibroblasts and vascular cells are
ated damage and dysfunction. Myocardial function is abundant in the healing infarct and might also contribute
intricately linked with preservation of structural integ- to suppression of inflammatory signalling. Acquisition
rity, impaired suppression or defective containment of of a pericyte coat by angiogenic vessels in the infarcted
inflammation in the injured heart can, therefore, have heart might suppress inflammatory activity stabilizing the
catastrophic consequences. Prolonging inflammatory microvasculature and preventing prolonged recruitment
signalling in the infarcted heart might have many con- ofleukocytes.75
sequences, including loss of cardiomyocytes, suppres-
sion of systolic function, enhanced matrix-degrading Molecular stop signals
processes leading to chamber dilation, increased tissue Negative regulation of proinflammatory signalling
breakdown causing loss of ventricular wall integrity and pathways is essential to maintain tissue homeostasis
cardiac rupture, and extended fibrotic changes beyond and activate the reparative response after the clearance
the initial infarct. Extensive experimental evidence, of dead cells. Both intracellular molecules and soluble
derived from mouse models with impaired repression mediators have been implicated in the inhibition of the
or resolution of the inflammatory response, suggests that inflammatory reaction following myocardial infarc-
overactive inflammatory signalling leads to increased tion. In our own work, we have identified IL1 receptor-
left ventricular dilatation following myocardial infarc- associated kinase3 (IRAK3; also known as IRAKM),
tion.11,35,69,70 Whether such defects lead to dilative remod- a member of the IRAK family. IRAK3 does not activate
elling in humans has not been established. However, inflammation, but functions as an inhibitor of innate
evidence from clinical studies suggests that patients with immune signalling 76 and as an essential intracellular
persistent elevation of serum inflammatory biomark- molecule for repression of macrophage-driven inflam-
ers (such as MCP1) 1month after an acute coronary mation and fibroblast-mediated matrix degradation

NATURE REVIEWS | CARDIOLOGY VOLUME 11 | MAY 2014 | 259


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Transforming growth factor


Activation of TGF signalling cascades is a key molecular
Endothelial cell
TREG cell link between the inflammatory and reparative response
(Figure3). Latent TGF is stored in the myocardium
and can be rapidly activated following injury. Generation
TGF- of ROS, induction of matricellular proteins (such as
Cardiomyocyte thrombospondin1), and activation of proteases contribute
Regulatory
macrophage to activation of preformed TGF in the infarcted area.92,93
Moreover, platelets, leukocytes and fibroblasts infiltrat-
Fibroblast ing the infarcted heart synthesize and release denovo
Myofibroblast
TGF, further increasing its levels.67 TGF bioactivity
Figure 3 | TGF is a key mediator in postinfarction is increased during the early hours after infarction;94
remodelling. TGF exerts anti-inflammatory actions, however, the abundance of proinflammatory mediators
inducing a regulatory macrophage phenotype, promoting at this stage might reduce cellular responsiveness to
regulatory Treg cell activation and reducing adhesion
TGF, delaying myofibroblast transdifferentiation and
molecule synthesis by endothelial cells. TGF is also
critically involved in fibroblast to myofibroblast conversion
matrix deposition until the infarct is cleared from dead
by activating a profibrotic signalling cascade. Abbreviations: cells and matrix debris.44 As proinflammatory signalling
TGF, transforming growth factor; Treg, regulatory Tcell. is repressed, TGF signalling promotes myofibroblast
transdifferentiation and activates a matrix-preserving
molecular programme, inducing expression of collagens
following myocardial infarction.11 IRAK3 is expressed and fibronectin, while upregulating synthesis of protease
in fibroblasts and a subset of infarct macrophages, and inhibitors (such as metalloproteinase inhibitor1).95 TGF
promotes an anti-inflammatory phenotype that inhibits signals through activation of intracellular effectors, the
cytokine expression. In addition to induction of intra- Smads, and through Smad-independent pathways (such
cellular signals that make cells less responsive to proin- as, mitogen-activated protein kinases). Experimental
flammatory activation, expression ofdecoy cytokine and evidence suggests that the profibrotic, matrix-preserving
chemokine receptors, and releaseof soluble inhibitory actions of TGF in fibroblasts are predominantly medi-
mediators might be important additional mechanisms ated through activation of Smad3 signalling;95,96 the poten-
involved in suppression of the inflammatory reaction. tial involvement of smad-independent cascades remains
Members of the transforming growth factor (TGF) poorly understood. The TGF signalling cascade inter-
family,7779 IL10,80 and proinflammatory-resolving lipid acts with several other important pathways that regulate
mediators81 have been identified as secreted media- the fibrogenic response in the remodelling myocardium.
tors that might act as inhibitors of the postinfarction Neurohumoral mediators, such as angiotensinII97 and
inflammatory reaction. aldosterone98 are important for fibroblast activation;
their effects might be mediated in part through activa-
From inflammation to fibrosis tion of TGF signalling.99 Moreover, the Notch path-
Repression of inflammation in the infarcted heart is waya signalling cascade critically involved in cardiac
associated with activation of mesenchymal cells that fibrotic responsesnegatively regulates the TGF/Smad
deposit extracellular matrix proteins, thereby preserv- response.100,101 In addition to the critical effects of angio-
ing the structural integrity of the infarcted heart. The tensinII and of the TGF/Smad cascade, other fibrogenic
adult mammalian heart contains an abundant popula- growth factors (such as platelet-derived growth factors)
tion of interstitial and perivascular fibroblasts;82,83 these might modulate fibroblast phenotype regulating their
cells can transdifferentiate into myofibroblasts, cells that proliferation, synthetic profile, and migratory activity.
express contractile proteins (such as smooth muscle
actin) and are key for repair of the infarcted myocar- Targeting the infarcted heart
dium by secreting matrix proteins.8486 In addition to Lessons from the past
the resident cardiac fibroblasts, bone marrow-derived Unfortunately, translation of early evidence that anti-
fibroblast progenitors, endothelial cells undergoing inflammatory strategies might reduce infarct size into a
transdifferentiation into mesenchymal cells, smooth clinical context has been disappointing. Although anti-
muscle cells, and pericytes might contribute to the CD11/CD18 integrin approaches were very effective at
infarct myofibroblast population. 8791 Conversion of reducing infarct size in experimental models,102104 three
fibroblasts into myofibroblasts requires the co-operation small clinical trials targeting 2integrins in patients with
of several microenvironmental factors: activation of myocardial infarction did not demonstrate beneficial
TGF, a key mediator in induction of contractile pro- effects.105107 A large clinical trial targeting the comple-
teins in mesenchymal cells; expression and deposition of ment system, a pathway critical in activation of the post
specialized matrix proteins, such as EDA fibronectin and infarction inflammatory reaction, also showed no benefit
matricellular proteins;92 increased mechanical stress trig- in patients undergoing percutaneous interventions
gered by the disruption of the normal matrix network; and for acute myocardial infarction.108 These failures had a
removal of proinflammatory mediators (such as IL1) lasting influence and reduced enthusiasm for the poten-
that inhibit myofibroblast conversion. tial clinical usefulness of anti-inflammatory approaches

260 | MAY 2014 | VOLUME 11  www.nature.com/nrcardio


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

in patients with myocardial infarction. Moreover, such relevant information for myocardial infarction, specifi-
failures question the usefulness of animal models in pre- cally, is the pathophysiological complexity and hetero-
dicting the success of therapeutic approaches.109 What is geneity of the condition in humans. Optimally executed
the reason for the apparent disconnect between findings investigations in animal models are designed to eliminate
in animals and clinical investigations? Why is translating variability, test a hypothesis, and provide insights into a
promising approaches to the clinical context so difficult, molecular pathway or cellular process. In a typical loss-
despite abundant evidence in experimental models? of-function study to examine a specific mediator, the
The academic community and public are often overly goal is to compare responses of age-matched and sex-
optimistic about new and promising therapeutic strat matched animals with identical genetic backgrounds that
egies. Studies with impressive positive results generate only differ in the presence or absence of the mediator of
great enthusiasm and are more likely to be published. interest. This strategy is optimal for understanding the
Laboratories reporting these observations are more likely pathophysiology of disease, but unfortunately limits our
to attract funding and have a better chance of completing ability to make translational predictions. Patients with
their work. During the early stages after the introduction myocardial infarction differ in a wide range of factors
of a new concept, therefore, the literature often reflects a that affect outcome. Genetic profile, age, sex, the pres-
publication bias that favours positive findings and results ence of comorbid conditions (such as hypertension,
in overly optimistic appraisal of the therapeutic potential hyperlipidaemia and diabetes mellitus), treatment with
of a strategy. Only after a concept is established in the pharmacological agents, and the pattern of the disease,
scientific community does the publication of negative are some of the important clinical variables that can
studies becomes attractive; by which time the published profoundly affect the response to myocardial infarction.
work might better reflect the collective experience of the Considering the complexity of the human pathophysiol-
scientific community. In the field of cardiac injury and ogy, attempts to introduce these variables and generate
repair, the abundant early reports suggesting that infarct an animal model of high predictive value are impractical.
size could be reduced using anti-inflammatory strategies An illustration of the profound effects of one of these
were later challenged by studies in genetically targeted factors on the postinfarction inflammatory and repara-
animals showing that postinfarction inflammation does tive response is provided by our experience in senescent
not extend ischaemic cardiomyocyte injury.10,34,43 animals. In a model of reperfused myocardial infarction,
Animal models are great tools for dissection of patho- aging was associated with a marked suppression (and
physiological concepts. However, these results cannot modest prolongation) of the inflammatory reaction fol-
directly predict effectiveness in a clinical context, owing lowing reperfused myocardial infarction.111 On the basis
to limitations of the animal model itself and the complex of these observations, one reason that explains the lack of
pathophysiology of human diseases. Animal models of effectiveness of targeted anti-inflammatory strategies in
myocardial infarction cannot, therefore, fully recapitu- humans might be the advanced age of many patients with
late the clinical outcome observed in humans. In clini- myocardial infarction. Young animals have been used in
cal trials, mortality is the most important end point; by all studies showing beneficial effects of anti-inflammatory
contrast, mortality data in animal investigations are often strategies after myocardial infarction. The failure of these
difficult to interpret and do not always provide evidence therapies in humans might, therefore, reflect a less robust
that can be easily translated to humans. Cardiac rupture inflammatory reaction in older individuals. The dysregu-
is the most-common cause of death in mouse models of lated immune responses associated with senescence com-
nonreperfused myocardial infarction,49,69 but is uncom- plicate efforts to design therapeutic strategies targeting
mon in humans and its incidence has declined over the the postinfarction inflammatory reaction.
past 30years, owing to the introduction of reperfusion
strategies and advances in medical care.110 Conversely, The future: modulating inflammation
ventricular arrhythmias are a common causes of death in Successful clinical translation requires both pathophysio
patients with acute myocardial infarction, but in mouse logical insights and an understanding of the clinical
models the incidence of fatal arrhythmias is low and the context. Implementation of this simple principle is of
mechanisms of arrhythmogenesis might differ owing to paramount importance in myocardial infarction. Over
the small size of the mouse heart and its rapid beating the past 30years, experimental studies have revealed
rate. Moreover, animal models of surgical coronary important mechanisms in the reparative and remodel-
occlusion do not provide information on the incidence ling responses that occur after myocardial infarction.
of recurrent coronary events, and the severity of post Experiments using animal models have highlighted
infarction heart failure as a clinical syndrome cannot be the complexity of inflammatory pathwayscytokines
reliably assessed in mice. Conclusions about outcomes in and growth factors are highly pleiotropic mediators
animal models of myocardial infarction are often based that exert multiple effects on all cell types involved in
on extrapolation from data reflecting specific functional cardiac injury and repair. Understanding the temporal
end points. Although these conclusions might provide and spatial regulation of inflammatory signals is criti-
important and accurate pathophysiological insights, cal to the design effective therapies. For example, early
direct relevance to clinical outcome is limited. activation of cytokine and chemokine pathways might be
Perhaps the most-important reason for the challenges important in clearing the infarct of dead cells and debris,
in translating experimental findings into clinically and for stimulation of downstream reparative cascades.

NATURE REVIEWS | CARDIOLOGY VOLUME 11 | MAY 2014 | 261


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

function are dependent on the balance between matrix-


degrading and matrix-preserving signals. Overactive
Anti-IL-1, anti-MCP-1
matrix-degrading processes caused by local activation
Dilatation, of matrixmetalloproteases by proinflammatory media-
Inflammatory systolic
biomarkers dysfunction tors, such as IL1 and MCP-1, are generally associated
with decreased tensile strength, leading to left ventricular
dilatation and systolic dysfunction. Degradation of the
interstitial matrix is also associated with cardiomyocyte
slippage and might lead to cardiomyocyte death owing
to deprivation of key prosurvival signals transduced by
the matrix.23 Conversely, overactive matrix-preserving
Fibrosis,
Profibrotic
diastolic responses, possibly associated with accentuated TGF
biomarkers
dysfunction signalling cascades, promote fibrosis and might cause
Anti-smad3 diastolic dysfunction. Patients with myocardial infarc-
tion exhibit very different remodelling responses, which
are at least in part independent of the size of the infarct.
Figure 4 | Biomarker-based approaches to target the inflammatory response The molecular determinants of geometric remodel-
inpatients with acute myocardial infarction. Patients surviving a myocardial ling in patients with myocardial infarction remain
infarctionexhibit pathophysiologically heterogeneous responses, which are partly unknown; however, one could speculate that exagger-
independent of the size of the infarct. Distinct pathophysiological responses might ated left ventricular dilatation might reflect overactive
be related to differences in genetic background and to the presence of conditions, proinflammatory signalling in individuals with defective
such as diabetes mellitus or hypertension, which affect inflammatory and fibrogenic
downregulation of acute inflammation. The association
pathways. After myocardial infarction, some patients develop progressive dilatation
and systolic dysfunction, whereas others develop diastolic dysfunction.
between increased mortality and persistent elevation of
Dilatationmight reflect excessive inflammatory activity causing matrix degradation. proinflammatory chemokines in the serum of patients
Conversely, diastolic dysfunction might indicate overactive profibrotic signalling. with acute coronary syndromes might reflect the adverse
Wepropose the measurement of inflammatory biomarkers, such as serum cytokine consequences of prolonged inflammation on the remod-
and chemokine levels, and of profibrotic markers, including indicators of matrix elling myocardium.71 However, certain subpopulations of
synthesis and remodelling, to stratify patients into subpopulations based on the patient, such as those with diabetes, have postinfarction
predominant pathophysiology. Patients with overactive inflammation might benefit heart failure in the absence of clinically relevant dilata-
from targeted inhibition of inflammatory signals (anti-IL-1 or anti-MCP-1 strategies),
tion.118 In patients with diabetes, postinfarction heart
whereas patients with profibrotic responses might benefit from inhibition of the
TGF/smad cascade. Abbreviation: MCP-1, monocyte chemoattractant protein1 failure is often linked with diastolic dysfunction, 119
(also known as CC motif chemokine2). and might reflect excessive activation of the profibrotic
TGF/Smad axis.120 Different therapeutic strategies are,
therefore, needed for these pathophysiologically distinct
However, prolonged or excessive induction of proinflam- patient subpopulations.
matory signalling is associated with accentuated injury The development of strategies to identify subgroups
and increased adverse remodelling. Spatial containment of patients with distinct pathophysiological alterations
of inflammatory cascades is equally importanteffective represents an important step towards implementation of
repair is dependent on signals that prevent extension of effective therapy targeting the inflammatory and fibrotic
the inflammatory response into the viable myocardium, response in myocardial infarction. Biomarker-based
therefore, limiting fibrosis to the infarcted region. strategies are needed to identify individuals with over-
Proinflammatory mediators, such as TNF, often active inflammatory responses and patients withexces-
exert both detrimental and protective responses on the sive fibrosis (Figure4). Serum levels of inflammatory
same cell type mediated through distinct receptors cytokines and chemokines might provide useful infor-
dissection of the pathways involved in these effects mation on the underlying pathophysiology. However,
might lead to more specific and effective therapeutic such markers are influenced by a wide range of clini-
strategies. The growing interest in cardiac regeneration cal and pathological conditions, such as the extent of
through cell therapy 112114 has added a new perspec- atherosclerotic disease, diabetes, obesity, and meta-
tive to the potential role of inflammatory signals in bolic dysfunction, and might not reflect alterations in
cardiac repair, stressing the role of selected chemokines the myocardial inflammatory and reparative process.
(such as stromal cell-derived factor1 [also known as Molecular imaging modalities can reveal structural, cel-
CXC motif chemokine12]) 115 cytokines, and growth lular, and molecular alterations in the infarcted heart,
factors,116,117 in regulating trafficking, activation, dif- and might be particularly promising for identification of
ferentiation, and survival of progenitor cells. The effect patients with overactive inflammatory responses. Patients
of inflamm atory signalling in extending ischaemic with such a response might benefit from targeted anti-
cardiomyocyte injury remains controversial; however, inflammatory approaches, such as pharmacological
the involvement of inflammatory and fibrogenic signals interventions to inhibit MCP-1 or IL1. The crucial role
in cardiac remodelling and in the development of post- of the IL1 system in postinfarction inflammation,43 the
infarction heart failure is already well-established. In availability of effective IL1 inhibitors and neutralizing
theinfarcted myocardium, left ventricular geometry and antibodies,121 the safety of IL1 antagonists in patients

262 | MAY 2014 | VOLUME 11  www.nature.com/nrcardio


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

with rheumatic disease, and the promising results of with myocardial infarction. However, the complexity
treatment with the IL1 receptor antagonist anakinra in of the pathophysiological process in humans is a major
patients with ST-segment elevation myocardial infarc- challenge for clinical translation. Biomarker and imaging-
tion,122,123 explain the recent focus of the cardiovascular based strategies identifying patient subpopulations with
community on IL1-related targets. Conversely, biomark- overactive proinflammatory or fibrogenic signalling
ers that reflect excessive extracellular matrix protein might contribute to rational implementation of therapies
synthesis124 or overactive TGF responses might be to prevent postinfarction heart failure.
useful in identification of individuals with predominant
fibrotic remodelling. These patients might benefit from
inhibition of Smad3. With careful exclusion of patients Review criteria
vulnerable to aneurysmal dilation, the potential adverse The PubMed database was searched for English-
consequences of Smad inhibition in vascular remodel- language articles published between January 1970
ling might be avoided.125,126 Development of personal- and January 2014 using the following keywords:
myocardial infarction, cardiac remodeling,
ized, biomarker-based approaches are, therefore, needed
inflammation, fibrosis, cytokine, chemokine,
to effectively target inflammatory signalling in patients
leukocyte, neutrophil, monocytes, myofibroblast,
with myocardial infarction.127 interleukin1, interleukin6, TGF, infarct
healing and extracellular matrix. Abstracts were
Conclusions reviewed and manuscripts focusing on the role of
Inflammatory pathways are critically involved in the inflammatoryandreparative cascades in cardiac injury,
repair and adverse remodelling of the infarcted heart. repair, and remodeling were analyzed in detail. The
Therapeutic approaches targeting specific components reference sections of these articles were also consulted
to identify additional papers of potential interest.
of the inflammatory response are promising for patients

1. Mallory, G.K., White, P.D. & Salcedo-Salgar, J. 12. Frangogiannis, N.G. Regulation of the 25. Timmers, L. etal. Toll-like receptor4 mediates
The speed of healing of myocardial infarction. inflammatory response in cardiac repair. maladaptive left ventricular remodeling and
Astudy of the pathologic anatomy in seventy-two Circ.Res. 110, 159173 (2012). impairs cardiac function after myocardial
cases. Am. Heart J. 18, 647671 (1939). 13. Timmers, L. etal. The innate immune response infarction. Circ. Res. 102, 257264 (2008).
2. Entman, M.L. etal. Neutrophil induced oxidative in reperfused myocardium. Cardiovasc. Res. 94, 26. Arslan, F. etal. Myocardial ischemia/reperfusion
injury of cardiac myocytes. A compartmented 276283 (2012). injury is mediated by leukocytic toll-like
system requiring CD11b/CD18ICAM1 14. Bianchi, M.E. DAMPs, PAMPs and alarmins: receptor2 and reduced by systemic
adherence. J. Clin. Invest. 90, 13351345 allwe need to know about danger. J. Leukoc. Biol. administration of a novel antitolllike receptor2
(1992). 81, 15 (2007). antibody. Circulation 121, 8090 (2010).
3. Yamazaki, T. etal. Expression of intercellular 15. Chan, J.K. etal. Alarmins: awaiting a clinical 27. Weisman, H.F. etal. Soluble human complement
adhesion molecule1 in rat heart with ischemia/ response. J. Clin. Invest. 122, 27112719 (2012). receptor type1: invivo inhibitor of complement
reperfusion and limitation of infarct size by 16. Andrassy, M. etal. High-mobility group box1 suppressing post-ischemic myocardial
treatment with antibodies against cell adhesion inischemia-reperfusion injury of the heart. inflammation and necrosis. Science 249, 146151
molecules. Am. J. Pathol. 143, 410418 (1993). Circulation 117, 32163226 (2008). (1990).
4. Simpson, P.J. etal. Reduction of experimental 17. Ding, H.S. etal. The HMGB1-TLR4 axis 28. Chandrasekar, B., Smith, J.B. & Freeman, G.L.
canine myocardial reperfusion injury by a contributes to myocardial ischemia/reperfusion Ischemia-reperfusion of rat myocardium activates
monoclonal antibody (anti-Mo1, anti-CD11b) that injury via regulation of cardiomyocyte apoptosis. nuclear factor-KappaB and induces neutrophil
inhibits leukocyte adhesion. J. Clin. Invest. 81, Gene 527, 389393 (2013). infiltration via lipopolysaccharide-induced CXC
624629 (1988). 18. Kitahara, T. etal. High-mobility group Box1 chemokine. Circulation 103, 22962302 (2001).
5. Tojo, S.J. etal. Reduction of rat myocardial restores cardiac function after myocardial 29. Mulligan, M.S. etal. Roles of 2 integrins of rat
ischemia and reperfusion injury by sialyl infarction in transgenic mice. Cardiovasc. Res. neutrophils in complement- and oxygen radical-
LewisXoligosaccharide and anti-rat Pselectin 80, 4046 (2008). mediated acute inflammatory injury. J. Immunol.
antibodies. Glycobiology 6, 463469 (1996). 19. Zou, N. etal. Critical role of extracellular heat 148, 18471857 (1992).
6. Christia, P. & Frangogiannis, N.G. Targeting shock cognate protein 70 in the myocardial 30. Fan, H. etal. Oxygen radicals trigger activation of
inflammatory pathways in myocardial infarction. inflammatory response and cardiac dysfunction NFkappaB and AP1 and upregulation of ICAM1
Eur. J. Clin. Invest. 43, 986995 (2013). after global ischemia-reperfusion. Am. J. Physiol. in reperfused canine heart. Am. J. Physiol. Heart
7. Cohn, J.N., Ferrari, R. & Sharpe, N. on behalf of Heart Circ. Physiol. 294, H2805H2813 Circ. Physiol. 282, H1778H1786 (2002).
an International Forum on Cardiac Remodeling. (2008). 31. Frangogiannis, N.G. Chemokines in ischemia
Cardiac remodelingconcepts and clinical 20. Arslan, F., de Kleijn, D.P. & Pasterkamp, G. and reperfusion. Thromb. Haemost. 97, 738747
implications: a consensus paper from an Innate immune signaling in cardiac ischemia. (2007).
international forum on cardiac remodeling. Nat. Rev. Cardiol. 8, 292300 (2011). 32. Clark-Lewis, I., Schumacher, C., Baggiolini, M.
J.Am. Coll. Cardiol. 35, 569582 (2000). 21. Mezzaroma, E. etal. The inflammasome &Moser, B. Structure-activity relationships of
8. White, H.D. etal. Left ventricular end-systolic promotes adverse cardiac remodeling following interleukin8 determined using chemically
volume as the major determinant of survival acute myocardial infarction in the mouse. Proc. synthesized analogs. Critical role of NH2-terminal
after recovery from myocardial infarction. Natl Acad. Sci. USA 108, 1972519730 (2011). residues and evidence for uncoupling of neutrophil
Circulation 76, 4451 (1987). 22. Huebener, P. etal. CD44 is critically involved in chemotaxis, exocytosis, and receptor binding
9. Frangogiannis, N.G. The immune system and infarct healing by regulating the inflammatory and activities. J. Biol. Chem. 266, 2312823134
cardiac repair. Pharmacol. Res. 58, 88111 fibrotic response. J. Immunol. 180, 26252633 (1991).
(2008). (2008). 33. Ivey, C.L., Williams, F.M., Collins, P.D.,
10. Briaud, S.A. etal. Leukocyte trafficking 23. Dobaczewski, M., Gonzalez-Quesada, C. & Jose,P.J. & Williams, T.J. Neutrophil
andmyocardial reperfusion injury in Frangogiannis, N.G. The extracellular matrix as chemoattractants generated in two phases
ICAM1/Pselectinknockout mice. Am. J. Physiol. a modulator of the inflammatory and reparative during reperfusion of ischemic myocardium in
Heart Circ. Physiol. 280, H60H67 (2001). response following myocardial infarction. J. Mol. the rabbit. Evidence for a role for C5a and
11. Chen, W. etal. Endogenous IRAKM attenuates Cell Cardiol. 48, 504511 (2010). interleukin8. J. Clin. Invest. 95, 27202728
postinfarction remodeling through effects on 24. Oyama, J. etal. Reduced myocardial ischemia- (1995).
macrophages and fibroblasts. Arterioscler. reperfusion injury in toll-like receptor4deficient 34. Dewald, O. etal. CCL2/Monocyte Chemoattractant
Thromb. Vasc. Biol. 32, 25982608 (2012). mice. Circulation 109, 784789 (2004). protein1 regulates inflammatory responses

NATURE REVIEWS | CARDIOLOGY VOLUME 11 | MAY 2014 | 263


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

critical to healing myocardial infarcts. Circ. Res. 52. Gersch, C. etal. Mast cells and macrophages acute myocardial infarction. Am. J. Pathol. 182,
96, 881889 (2005). innormal C57/BL/6 mice. Histochem. Cell Biol. 2940 (2013).
35. Dobaczewski, M., Xia, Y., Bujak, M., 118, 4149 (2002). 71. de Lemos, J.A. etal. Serial measurement of
GonzalezQuesada, C. & Frangogiannis, N.G. 53. Darden, A.G., Forbes, R.D., Darden, P.M. monocyte chemoattractant protein1 after acute
CCR5 signaling suppresses inflammation and &Guttmann, R.D. The effects of genetics and coronary syndromes: results from the A to Z.
reduces adverse remodeling of the infarcted age on expression of MHC class II and CD4 trial. J. Am. Coll. Cardiol. 50, 21172124 (2007).
heart, mediating recruitment of regulatory antigens on rat cardiac interstitial dendritic 72. Frangogiannis, N.G. The prognostic value of
Tcells. Am. J. Pathol. 176, 21772187 (2010). cells. Cell. Immunol. 126, 322330 (1990). monocyte chemoattractant protein1/CCL2 in
36. Herskowitz, A., Choi, S., Ansari, A.A. 54. Ito, B.R., Engler, R.L. & del Balzo, U. Role of acute coronary syndromes. J. Am. Coll. Cardiol.
&Wesselingh, S. Cytokine mRNA expression cardiac mast cells in complement C5a-induced 50, 21252127 (2007).
inpostischemic/reperfused myocardium. myocardial ischemia. Am. J. Physiol. 264, 73. Yan, X. etal. Temporal dynamics of cardiac
Am.J.Pathol. 146, 419428 (1995). H1346H1354 (1993). immune cell accumulation following acute
37. Dewald, O. etal. Of mice and dogs: species- 55. Linden, J. Molecular approach to adenosine myocardial infarction. J. Mol. Cell Cardiol. 62,
specific differences in the inflammatory receptors: receptor-mediated mechanisms of 2435 (2013).
response following myocardial infarction. tissue protection. Annu. Rev. Pharmacol. Toxicol. 74. Wan, E. etal. Enhanced efferocytosis of
Am.J.Pathol. 164, 665677 (2004). 41, 775787 (2001). apoptotic cardiomyocytes through myeloid-
38. Frangogiannis, N.G. etal. Resident cardiac mast 56. Gilles, S., Zahler, S., Welsch, U., Sommerhoff,C.P. epithelialreproductive tyrosine kinase links
cells degranulate and release preformed TNF-, & Becker, B.F. Release of TNF- during acute inflammation resolution to cardiac repair
initiating the cytokine cascade in experimental myocardial reperfusion depends on oxidative after infarction. Circ. Res. 113, 10041012
canine myocardial ischemia/reperfusion. stress and is prevented by mast cell stabilizers. (2013).
Circulation 98, 699710 (1998). Cardiovasc. Res. 60, 608616 (2003). 75. Zymek, P. etal. The role of platelet-derived
39. Maekawa, N. etal. Improved myocardial 57. Ley, K., Laudanna, C., Cybulsky, M.I. & growth factor signaling in healing myocardial
ischemia/reperfusion injury in mice lacking Nourshargh, S. Getting to the site of infarcts. J.Am. Coll. Cardiol 48, 23152323
tumor necrosis factor-. J. Am. Coll. Cardiol. 39, inflammation: the leukocyte adhesion cascade (2006).
12291235 (2002). updated. Nat. Rev. Immunol. 7, 678689 (2007). 76. Kobayashi, K. etal. IRAKM is a negative
40. Kurrelmeyer, K.M. etal. Endogenous tumor 58. Hoshida, S. etal. Attenuation of neutrophil regulator of Toll-like receptor signaling. Cell 110,
necrosis factor protects the adult cardiac function by inhibitors of arachidonate 191202 (2002).
myocyte against ischemic-induced apoptosis in metabolism reduces the extent of canine 77. Kempf, T. etal. GDF15 is an inhibitor of
a murine model of acute myocardial infarction. myocardial infarction. Am. J. Cardiol. 63, 24E28E leukocyte integrin activation required for survival
Proc. Natl Acad. Sci. USA 97, 54565461 (1989). after myocardial infarction in mice. Nat. Med. 17,
(2000). 59. Swirski, F.K. etal. Identification of splenic 581588 (2011).
41. Hamid, T. etal. Divergent tumor necrosis factor reservoir monocytes and their deployment to 78. Ikeuchi, M. etal. Inhibition of TGF- signaling
receptor-related remodeling responses in inflammatory sites. Science 325, 612616 exacerbates early cardiac dysfunction but
heartfailure: role of nuclear factor-kappaB (2009). prevents late remodeling after infarction.
andinflammatory activation. Circulation 119, 60. Zouggari, Y. etal. Blymphocytes trigger Cardiovasc. Res. 64, 526535 (2004).
13861397 (2009). monocyte mobilization and impair heart function 79. Dobaczewski, M., Chen, W. & Frangogiannis,N.G.
42. Mann, D.L. etal. Targeted anticytokine therapy after acute myocardial infarction. Nat. Med. 19, Transforming growth factor (TGF)- signaling
in patients with chronic heart failure: results 12731280 (2013). incardiac remodeling. J. Mol. Cell Cardiol. 51,
ofthe Randomized Etanercept Worldwide 61. Nahrendorf, M. & Swirski, F.K. Monocyte 600606 (2011).
Evaluation (RENEWAL). Circulation 109, andmacrophage heterogeneity in the heart. 80. Frangogiannis, N.G. etal. IL10 is induced in the
15941602 (2004). Circ.Res. 112, 16241633 (2013). reperfused myocardium and may modulate the
43. Bujak, M. etal. Interleukin1 receptor typeI 62. Kawaguchi, M. etal. Inflammasome activation reaction to injury. J. Immunol. 165, 27982808
signaling critically regulates infarct healing and ofcardiac fibroblasts is essential for myocardial (2000).
cardiac remodeling. Am. J. Pathol. 173, 5767 ischemia/reperfusion injury. Circulation 123, 81. Keyes, K.T. etal. Resolvin E1 protects the rat
(2008). 594604 (2011). heart against reperfusion injury. Am. J. Physiol.
44. Saxena, A. etal. IL1 induces proinflammatory 63. Frangogiannis, N.G. etal. Induction and Heart Circ. Physiol. 299, H153H164 (2010).
leukocyte infiltration and regulates fibroblast suppression of interferon-inducible protein10 82. Souders, C.A., Bowers, S.L. & Baudino, T.A.
phenotype in the infarcted myocardium. inreperfused myocardial infarcts may regulate Cardiac fibroblast: the renaissance cell.
J.Immunol. 191, 48384848 (2013). angiogenesis. FASEB J. 15, 14281430 (2001). Circ.Res. 105, 11641176 (2009).
45. Abbate, A. etal. Anakinra, a recombinant human 64. Kumar, A.G. etal. Induction of monocyte 83. Chen, W. & Frangogiannis, N.G. Fibroblasts in
interleukin1 receptor antagonist, inhibits chemoattractant protein1 in the small veins post-infarction inflammation and cardiac repair.
apoptosis in experimental acute myocardial ofthe ischemic and reperfused canine Biochim. Biophys. Acta 1833, 945953 (2013).
infarction. Circulation 117, 26702683 (2008). myocardium. Circulation 95, 693700 (1997). 84. Willems, I.E., Havenith, M.G., De Mey, J.G.
46. Van Tassell, B.W., Toldo, S., Mezzaroma, E. 65. Liu, Y. etal. Novel role of platelets in mediating &Daemen, M.J. The -smooth muscle actin-
&Abbate, A. Targeting interleukin1 in heart inflammatory responses and ventricular rupture positive cells in healing human myocardial
disease. Circulation 128, 19101923 (2013). or remodeling following myocardial infarction. scars. Am. J. Pathol. 145, 868875 (1994).
47. Fischer, P. & Hilfiker-Kleiner, D. Role of gp130- Arterioscler. Thromb. Vasc. Biol. 31, 834841 85. Cleutjens, J.P., Verluyten, M.J., Smiths, J.F.
mediated signalling pathways in the heart and (2011). &Daemen, M.J. Collagen remodeling after
its impact on potential therapeutic aspects. 66. Gwechenberger, M. etal. Cardiac myocytes myocardial infarction in the rat heart. Am. J.
Br.J. Pharmacol. 153 (Suppl 1), S414S427 produce interleukin6 in culture and in viable Pathol. 147, 325338 (1995).
(2008). border zone of reperfused infarctions. Circulation 86. Frangogiannis, N.G., Michael, L.H. &
48. Mima, T. & Nishimoto, N. Clinical value of 99, 546551 (1999). Entman,M.L. Myofibroblasts in reperfused
blocking IL6 receptor. Curr. Opin. Rheumatol. 21, 67. Christia, P. etal. Systematic characterization of myocardial infarcts express the embryonic form
224230 (2009). myocardial inflammation, repair, and remodeling of smooth muscle myosin heavy chain (SMemb).
49. Fuchs, M. etal. Role of interleukin6 for LV in a mouse model of reperfused myocardial Cardiovasc. Res. 48, 89100 (2000).
remodeling and survival after experimental infarction. J. Histochem. Cytochem. 61, 555570 87. Zeisberg, E.M. etal. Endothelialtomesenchymal
myocardial infarction. FASEB J. 17, 21182120 (2013). transition contributes to cardiac fibrosis.
(2003). 68. Fullerton, J.N., OBrien, A.J. & Gilroy, D.W. Nat.Med. 13, 952961 (2007).
50. Kobara, M. etal. Antibody against interleukin6 Pathways mediating resolution of inflammation: 88. Mllmann, H. etal. Bone marrow-derived cells
receptor attenuates left ventricular remodelling when enough is too much. J. Pathol. 231, 820 contribute to infarct remodelling. Cardiovasc.
after myocardial infarction in mice. Cardiovasc. (2013). Res. 71, 661671 (2006).
Res. 87, 424430 (2010). 69. Cochain, C. etal. The chemokine decoy 89. Hinz, B. etal. The myofibroblast: one function,
51. Hilfiker-Kleiner, D. etal. Continuous receptorD6 prevents excessive inflammation multiple origins. Am. J. Pathol. 170, 18071816
glycoprotein130mediated signal transducer and adverse ventricular remodeling after (2007).
and activator of transcription3 activation myocardial infarction. Arterioscler Thromb. Vasc. 90. Davis, J. & Molkentin, J.D. Myofibroblasts:
promotes inflammation, left ventricular rupture, Biol. 32, 22062213 (2012). Trustyour heart and let fate decide. J. Mol.
and adverse outcome in subacute myocardial 70. Seropian, I.M. etal. Galectin1 controls cardiac CellCardiol. http://dx.doi.org/10.1016/
infarction. Circulation 122, 145155 (2010). inflammation and ventricular remodeling during j.yjmcc.2013.10.019.

264 | MAY 2014 | VOLUME 11  www.nature.com/nrcardio


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

91. Lajiness, J.D. & Conway, S.J. Origin, myocardial ischemia-reperfusion injury. 117. Xiang, F.L. etal. Cardiomyocyte-specific
development, and differentiation of cardiac Circulation 88, 17791787 (1993). overexpression of human stem cell factor
fibroblasts. J. Mol. Cell Cardiol http:// 105. Baran, K.W. etal. Double-blind, randomized trial improves cardiac function and survival after
dx.doi.org/10.1016/j.yjmcc.2013.11.003. of an anti-CD18 antibody in conjunction with myocardial infarction in mice. Circulation 120,
92. Frangogiannis, N.G. Matricellular proteins in recombinant tissue plasminogen activator for 10651074 (2009).
cardiac adaptation and disease. Physiol. Rev. acute myocardial infarction: limitation of 118. Carrabba, N., Valenti, R., Parodi, G.,
92,635688 (2012). myocardial infarction following thrombolysis in Santoro,G.M. & Antoniucci, D. Left ventricular
93. Frangogiannis, N.G. etal. The critical role of acute myocardial infarction (LIMITAMI) study. remodeling and heart failure in diabetic patients
endogenous thrombospondin1 in preventing Circulation 104, 27782783 (2001). treated with primary angioplasty for acute
expansion of healing myocardial infarcts. 106. Faxon, D.P., Gibbons, R.J., Chronos, N.A., myocardial infarction. Circulation 110, 19741979
Circulation 111, 29352942 (2005). Gurbel, P.A. & Sheehan, F. The effect of (2004).
94. Birdsall, H.H. etal. Complement C5a, TGF- 1, blockade of the CD11/CD18 integrin receptor 119. Aronson, D. etal. Impact of diastolic dysfunction
and MCP1, in sequence, induce migration of on infarct size in patients with acute myocardial on the development of heart failure in diabetic
monocytes into ischemic canine myocardium infarction treated with direct angioplasty: the patients after acute myocardial infarction.
within the first one to five hours after results of the HALT-MI study. J. Am. Coll. Cardiol. Circ.Heart Fail. 3, 125131 (2010).
reperfusion. Circulation 95, 684692 (1997). 40, 11991204 (2002). 120. Biernacka, A., Dobaczewski, M.
95. Bujak, M. etal. Essential role of smad3 in infarct 107. Rusnak, J.M. etal. An anti-CD11/CD18 &Frangogiannis,N.G. TGF- signaling in
healing and in the pathogenesis of cardiac monoclonal antibody in patients with acute fibrosis. Growth Factors 29, 196202 (2011).
remodeling. Circulation 116, 21272138 (2007). myocardial infarction having percutaneous 121. Ridker, P.M., Thuren, T., Zalewski, A. & Libby, P.
96. Dobaczewski, M. etal. Smad3 signaling critically transluminal coronary angioplasty (the FESTIVAL Interleukin-1 inhibition and the prevention of
regulates fibroblast phenotype and function in study). Am. J. Cardiol. 88, 482487 (2001). recurrent cardiovascular events: rationale and
healing myocardial infarction. Circ. Res. 107, 108. Armstrong, P.W. etal. Pexelizumab for acute design of the Canakinumab Anti-inflammatory
418428 (2010). STelevation myocardial infarction in patients Thrombosis Outcomes Study (CANTOS).
97. Weber, K.T., Sun, Y., Bhattacharya, S.K., undergoing primary percutaneous coronary Am.Heart J. 162, 597605 (2011).
Ahokas, R.A. & Gerling, I.C. Myofibroblast- intervention: a randomized controlled trial. JAMA 122. Abbate, A. etal. Interleukin1 blockade with
mediated mechanisms of pathological 297, 4351 (2007). anakinra to prevent adverse cardiac remodeling
remodelling of the heart. Nat. Rev. Cardiol. 10, 109. Dove, A. CD18 trials disappoint again. after acute myocardial infarction (Virginia
1526 (2012). Nat.Biotechnol. 18, 817818 (2000). Commonwealth University Anakinra Remodeling
98. Cohn, J.N. & Colucci, W. Cardiovascular effects 110. Figueras, J. etal. Changes in hospital mortality Trial [VCU-ART] pilot study). Am. J. Cardiol. 105,
of aldosterone and post-acute myocardial rates in 425 patients with acute STelevation 13711377 (2010).
infarction pathophysiology. Am. J. Cardiol 97, myocardial infarction and cardiac rupture over 123. Abbate, A. etal. Effects of interleukin1 blockade
4F12F (2006). a30-year period. Circulation 118, 27832789 with anakinra on adverse cardiac remodeling
99. Schultz Jel, J. etal. TGF-1 mediates the (2008). and heart failure after acute myocardial
hypertrophic cardiomyocyte growth induced by 111. Bujak, M. etal. Aging-related defects are infarction [from the Virginia Commonwealth
angiotensinII. J. Clin. Invest. 109, 787796 associated with adverse cardiac remodeling University-Anakinra Remodeling Trial (2)
(2002). inamouse model of reperfused myocardial (VCUART2) pilot study]. Am. J. Cardiol. 111,
100. Nemir, M. etal. The notch pathway controls infarction. J. Am. Coll. Cardiol. 51, 13841392 13941400 (2013).
fibrotic and regenerative repair in the adult (2008). 124. Lopez, B., Gonzalez, A. & Diez, J. Circulating
heart. Eur. Heart J. http://dx.doi.org/10.1093/ 112. Laflamme, M.A. & Murry, C.E. Regenerating biomarkers of collagen metabolism in cardiac
eurheartj/ehs269. theheart. Nat. Biotechnol. 23, 845856 (2005). diseases. Circulation 121, 16451654 (2010).
101. Sassoli, C. etal. Relaxin prevents cardiac 113. Bolli, R. etal. Cardiac stem cells in patients 125. Tan, C.K. etal. SMAD3 deficiency
fibroblast-myofibroblast transition via withischaemic cardiomyopathy (SCIPIO): initial promotesinflammatory aortic aneurysms
notch1mediated inhibition of TGF-/Smad3 results of a randomised phase1 trial. Lancet inangiotensinIIinfused mice via activation of
signaling. PLoS ONE 8, e63896 (2013). 378, 18471857 (2011). iNOS. J. Am. Heart Assoc. 2, e000269 (2013).
102. Arai, M. etal. An anti-CD18 antibody limits 114. Makkar, R.R. etal. Intracoronary 126. Li, J. etal. Blockade of endothelial-mesenchymal
infarct size and preserves left ventricular cardiospherederived cells for heart transition by a Smad3 inhibitor delays the early
function in dogs with ischemia and 48-hour regenerationafter myocardial infarction development of streptozotocin-induced diabetic
reperfusion. J. Am. Coll. Cardiol 27, 12781285 (CADUCEUS): a prospective, randomised nephropathy. Diabetes 59, 26122624 (2010).
(1996). phase1 trial. Lancet 379, 895904 (2012). 127. Frangogiannis, N.G. Biomarkers: hopes and
103. Aversano, T., Zhou, W., Nedelman, M., 115. Penn, M.S., Pastore, J., Miller, T. & Aras, R. challenges in the path from discovery to clinical
Nakada,M. & Weisman, H. A chimeric IgG4 SDF1 in myocardial repair. Gene Ther. 19, practice. Transl. Res. 159, 197204 (2012).
monoclonal antibody directed against CD18 583587 (2012).
reduces infarct size in a primate model of 116. Beohar, N., Rapp, J., Pandya, S. & Losordo, D.W. Acknowledgements
myocardial ischemia and reperfusion. J. Am. Coll. Rebuilding the damaged heart: the potential of Dr Frangogiannis laboratory is funded by NIH grants
Cardiol. 25, 781788 (1995). cytokines and growth factors in the treatment of R01 HL76246 and R01 HL85440 and by the Wilf
104. Lefer, D.J. etal. Cardioprotective actions of ischemic heart disease. J. Am. Coll. Cardiol. 56, Family Cardiovascular Research Institute, Albert
amonoclonal antibody against CD18 in 12871297 (2010). Einstein College of Medicine, New York, USA.

NATURE REVIEWS | CARDIOLOGY VOLUME 11 | MAY 2014 | 265


2014 Macmillan Publishers Limited. All rights reserved

You might also like