Sie sind auf Seite 1von 23

Review

This Review is in a thematic series on Inflammation, which includes the following articles:

The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis
Resolution of Acute Inflammation and the Role of Resolvins in Immunity, Thrombosis, and Vascular Biology
Circadian Influence on Metabolism and Inflammation in Atherosclerosis
Inflammatory Disequilibrium in Stroke
Chronic Heart Failure and Inflammation: What Do We Really Know?
Guest Editors: Nikolaos Frangogiannis and Matthias Nahrendorf

The Biological Basis for Cardiac Repair After


Myocardial Infarction
From Inflammation to Fibrosis
Sumanth D. Prabhu, Nikolaos G. Frangogiannis

Abstract: In adult mammals, massive sudden loss of cardiomyocytes after infarction overwhelms the limited
regenerative capacity of the myocardium, resulting in the formation of a collagen-based scar. Necrotic cells release
danger signals, activating innate immune pathways and triggering an intense inflammatory response. Stimulation
of toll-like receptor signaling and complement activation induces expression of proinflammatory cytokines (such
as interleukin-1 and tumor necrosis factor-α) and chemokines (such as monocyte chemoattractant protein-1/
chemokine (C-C motif) ligand 2 [CCL2]). Inflammatory signals promote adhesive interactions between leukocytes
and endothelial cells, leading to extravasation of neutrophils and monocytes. As infiltrating leukocytes clear the
infarct from dead cells, mediators repressing inflammation are released, and anti-inflammatory mononuclear cell
subsets predominate. Suppression of the inflammatory response is associated with activation of reparative cells.
Fibroblasts proliferate, undergo myofibroblast transdifferentiation, and deposit large amounts of extracellular
matrix proteins maintaining the structural integrity of the infarcted ventricle. The renin–angiotensin–aldosterone
system and members of the transforming growth factor-β family play an important role in activation of infarct
myofibroblasts. Maturation of the scar follows, as a network of cross-linked collagenous matrix is formed and
granulation tissue cells become apoptotic. This review discusses the cellular effectors and molecular signals regulating
the inflammatory and reparative response after myocardial infarction. Dysregulation of immune pathways,
impaired suppression of postinfarction inflammation, perturbed spatial containment of the inflammatory response,
and overactive fibrosis may cause adverse remodeling in patients with infarction contributing to the pathogenesis
of heart failure. Therapeutic modulation of the inflammatory and reparative response may hold promise for the
prevention of postinfarction heart failure.   (Circ Res. 2016;119:91-112. DOI: 10.1161/CIRCRESAHA.116.303577.)
Key Words: chemokines ■ cytokines ■ fibrosis ■ immune cells ■ inflammation ■ myocytes, cardiac
■ myocardial infarction

A dverse left ventricular (LV) remodeling after myocardial


infarction (MI) constitutes the structural basis for isch-
emic heart failure (HF) and comprised complex short- and
and molecular composition.1,2 Although multiple pathophysi-
ological factors converge to remodel the heart after MI, the
fundamental determinants of this process (and its progression
long-term changes in LV size, shape, function, and cellular to clinical HF) are the extent of the initial infarction and the

Original received February 6, 2016; revision received April 14, 2016; accepted April 15, 2016.
From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department
of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.).
Correspondence to Sumanth D. Prabhu, MD, Professor and Director, Division of Cardiovascular Disease, University of Alabama at Birmingham, 311
Tinsley Harrison Tower, 1900 University Blvd, Birmingham, AL 35294-0006. E-mail sprabhu@uab.edu; or Nikolaos G. Frangogiannis, MD, The Wilf
Family Cardiovascular Research Institute, Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Forchheimer G46B,
Bronx, NY 10461. E-mail nikolaos.frangogiannis@einstein.yu.edu
© 2016 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.116.303577

Downloaded from http://circres.ahajournals.org/ at91


California Institute of Technology on June 26, 2016
92  Circulation Research  June 24, 2016

Nonstandard Abbreviations and Acronyms

DAMP danger-associated molecular pattern


DC dendritic cells
HF heart failure
HMGB-1 high mobility group box-1
I/R ischemia/reperfusion
IL interleukin
LV left ventricular
MAPK mitogen-activated protein kinase
MI myocardial infarction
MyD88 myeloid differentiation factor 88
NF-κB nuclear factor-κB
NLR nucleotide-binding oligomerization domain–like receptor
PDGF platelet-derived growth factor
PRR pattern recognition receptor
RAGE receptor for advanced glycation end-products
ROS reactive oxygen species
TGF transforming growth factor
TIR Toll/IL-1R Figure 1. Biphasic nature of cardiac repair after myocardial
TLR/IL-1R toll-like receptor/IL-1 receptor infarction (MI). Early after MI, tissue injury and necrosis
initiates the inflammatory phase, consisting of intense sterile
TNF tumor necrosis factor-α inflammation, and the dynamic recruitment of several immune
TRP transient receptor potential cell subtypes including neutrophils, monocyte/macrophages,
dendritic cells, and lymphocytes. After ≈4 d in murine models,
this transitions to a reparative and proliferative phase, with shift
of immune cell polarity toward immunomodulation and resolution,
sufficiency of the post-MI reparative process. In clinical prac- myofibroblast proliferation, collagen deposition and scar
tice, limiting infarction extent is routinely addressed by timely formation, and neovascularization, thereby resulting in wound
coronary reperfusion. In contrast, therapeutic manipulation of healing. Neurohormonal activation and mechanical stress are
the ensuing repair process, which is driven principally by ro- other factors that influence this healing process. ROS indicates
reactive oxygen species.
bust tissue inflammation and subsequently by its active sup-
pression and resolution, has proved much more challenging
and elusive. Nonetheless, recent studies have suggested a large no large-scale immunomodulatory or anti-inflammatory thera-
number of potential therapeutic targets that may favorably in- peutic strategy post MI that has been successfully translated into
fluence cardiac wound healing and repair. In this review, we clinical practice, no doubt a reflection of both the exquisite com-
will broadly consider the multiplicity of cellular and molecular plexity and our incomplete understanding of the healing process.
factors that influence post-MI repair, highlighting the transla-
tional implications for these events in the amelioration of ad- Inflammatory Phase
verse remodeling and the development of ischemic HF. Molecular Cascades Implicated in the
Postinfarction Inflammatory Response
Phases of Cardiac Repair After MI Hypoxia during ischemia impairs vascular endothelial cell in-
Cardiac repair after MI results from a finely orchestrated and tegrity and its barrier function thereby augmenting vessel per-
complex series of events, initiated by intense sterile inflamma- meability, facilitating leukocyte infiltration.7 If the ischemic
tion and immune cell infiltration (inflammatory phase) that serve period is sufficiently prolonged, parenchymal and cardiomyo-
to digest and clear damaged cells and extracellular matrix tissue cyte cell death programs are activated, primarily because of
(≈3–4 d in mice), followed by a reparative phase with resolution cell necrosis, secondarily because of apoptosis and autopha-
of inflammation, (myo)fibroblast proliferation, scar formation, gic mechanisms.3,7 Restoration of blood flow may further aug-
and neovascularization over the next several days (Figure 1).3,4 ment tissue damage via reperfusion injury because of abrupt
Early inflammatory activation is a necessary event for the transi- reoxygenation, reactive oxygen species (ROS) generation,
tion to later reparative and proliferative programs. Appropriate and activation of the complement pathway.3,5,7–9 Necrotic and
and timely containment and resolution of inflammation are fur- stressed/injured cells, and the damaged extracellular matrix,
ther determinants of the quality of wound healing; a proper physi- release substances that act as danger signals, termed danger-
ological balance needs to be achieved between these 2 phases associated molecular patterns (DAMPs). DAMPs bind to
for optimal repair.5,6 An inflammatory phase that is dispropor- cognate pattern recognition receptors (PRRs) of the innate
tionately prolonged, of excessive magnitude, or insufficiently immune system on surviving parenchymal cells and infiltrat-
suppressed can lead to sustained tissue damage and improper ing leukocytes (and also activate the complement pathway)
healing, defective scar formation, and heightened cell loss and to robustly activate a cascade of inflammatory mediators, in-
contractile dysfunction, thereby promoting infarct expansion, ad- cluding inflammatory cytokines, chemokines, and cell adhe-
verse remodeling, and chamber dilatation. To date, there has been sion molecules.8,10–14 In addition to being passively released
Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016
Prabhu and Frangogiannis   Repair After Myocardial Infarction   93

Table.  Select DAMPs and Corresponding Pattern inflammation resolution and wound healing and is mediated, in
Recognition Receptors part, by macrophages expressing the myeloid-epithelial-repro-
DAMP Receptor(s) Reference(s)
ductive tyrosine kinase (Mertk).23

HMGB1 TLR2, TLR4, TLR9, 8, 10–12, 14, 15, Danger-Associated Molecular Patterns
RAGE 25–40, 83 HMGB1: HMGB1, a loosely associated chromatin protein
S100A8/S100A9 TLR4, NLRP3, RAGE 11, 14, 42–47, 83 involved in DNA stabilization and gene control,24 is a po-
tent mediator of inflammation after tissue injury.25 HMGB1
S100A1 Endolysosomal TLR4 48
is passively released from necrotic cells (but not from apop-
Fibronectin-EDA TLR2, TLR4 8, 10, 11, 51–53 totic cells),25 actively secreted by stimulated monocytes and
IL-1α IL-1R 14, 54, 83 macrophages,26 and induced by peroxynitrite and oxidative
HSP-60, HSP-70 TLR2, TLR4/6 8, 10, 11, 83
stress in ischemic cardiomyocytes.27 HGMB1 engages and
activates several TLRs (including TLR2, TLR4, and TLR9)
LMW hyaluronic acid TLR2, TLR4, NLRP3 8, 10, 11, 18, 83
and RAGE,8,15,28,29 to induce NF-κB nuclear translocation and
ATP P2X7/NLRP3 11, 14, 18, 83, 87, 88 proinflammatory signaling. HMGB1 also promotes monocyte
Uric acid TLR2, TLR4, NLRP3 11, 14, 18, 83 recruitment in a TLR- and RAGE-independent manner via di-
rect binding to CXCL12 (stromal cell–derived factor-1) and
Mitochondrial DNA TLR9, NLRP3 8, 10, 11, 14, 83
the formation of HMGB1-CXCL12 heterocomplexes that syn-
dsRNA TLR3 8, 14, 83 ergistically enhance CXCR4 signaling in inflammatory cells.30
ssRNA TLR7, TLR8 8, 10, 11, 14 In humans with acute MI, serum HMGB1 levels are ele-
Some references are in-depth review articles. DAMP indicates danger- vated and predictive of subsequent mortality, LV dysfunction,
associated molecular pattern; ds, double stranded; EDA, extra domain A; and effort intolerance.31–33 In rodents with reperfused15,34 and
HMGB, high mobility group box; HSP, heat shock protein; IL, interleukin; LMW, nonreperfused33 MI, serum levels and myocardial HMGB1 ex-
low molecular weight; NLR, nucleotide-binding oligomerization domain–like pression increase early after injury. In reperfused MI, HMGB1
receptor; P2X7, purinergic P2X7 receptor; RAGE, receptor for advanced plays a pivotal role in the activation of MAPK and NF-κB
glycation end-products; ss, single-stranded; and TLR, toll-like receptor.
pathways, increasing leukocyte infiltration, and augmenting
tissue injury, apoptosis, and infarct size, in part, via RAGE-
on cell necrosis or matrix damage, select DAMPs may also
dependent signaling.15,34 Interestingly and in contrast, howev-
be upregulated and secreted by stressed cardiomyocytes and
er, in acute and chronic nonreperfused MI models, augmenting
fibroblasts, and by activated leukocytes.8,11,12,15–17 HMGB1 via either exogenous administration35–37 or cardio-
Several DAMPs can trigger the inflammatory response myocyte-specific overexpression38,39 improved LV remodeling
during MI (Table). These including high mobility group box- and function with augmented c-kit+ progenitor cell infiltration,
1 (HMGB1), S100 proteins, fibronectin extra domain A, in- reduced dendritic cell (DC) accumulation, less collagen de-
terleukin (IL)-1α, heat shock proteins, low molecular weight position, and better tissue angiogenesis. Exogenous admin-
hyaluronic acid, ATP, uric acid, mitochondrial DNA, double- istration of HMGB1 before ischemia/reperfusion (I/R) also
stranded RNA, single-stranded RNA, and complement, among induces preconditioning and cardioprotection.40 Interestingly,
others.7,8,11,12,14 The PRRs are primarily the membrane-bound antibody-mediated HMGB1 neutralization in nonreperfused
toll-like receptor/IL-1 receptors (TLR/IL-1Rs), as well as cyto- MI reduced tissue inflammatory cytokine expression and mac-
solic nucleotide-binding oligomerization domain–like receptors rophage infiltration at day 3, but induced scar thinning and
(NLRs) and the cell-surface receptor for advanced glycation more pronounced LV remodeling, underscoring the concept
end-products (RAGE). The signaling pathways downstream that inflammation, properly regulated and controlled, is es-
of these PRRs have been comprehensively detailed in recent sential for optimal wound healing in the heart.33 Hence, these
reviews13,14,18,19 and briefly considered below. In the context results indicate that although HMGB1 is an inflammatory me-
of MI, downstream signaling converges on the activation of diator, the ultimate effects of HMGB1 modulation depend on
mitogen-activated protein kinases (MAPKs) and nuclear fac- the underlying disease, its temporal context, and the degree
tor (NF)-κB. These pathways (NF-κB in particular) drive the of inflammatory response referable to the pathophysiology.
expression of a large panel of proinflammatory genes including Compared with nonreperfused MI hearts, I/R hearts exhibit
inflammatory cytokines (eg, tumor necrosis factor-α [TNF], IL- greater magnitude but shorter duration of inflammatory cell
1β, IL-6, and IL-18); CXC chemokines containing the glutamic infiltration.9,41 This profile of augmented inflammation may
acid-leucine-arginine motif that act predominantly as neutro- explain the beneficial results with HMGB1 inhibition in reper-
phil chemoattractants; CC chemokines that attract monocytes fused MI, and the divergence in response from nonreperfused
and T-lymphocytes; cell adhesion molecules (eg, vascular cell MI in which angiogenic effects may predominate.
adhesion molecule, intercellular adhesion molecule, selectins), Other DAMPs: S100A8 (calgranulin A) and S100A9 (cal-
and complement factor B.14,20,21 Subsequent leukocyte recruit- granulin B) are members of the calcium-binding S100 family
ment further amplifies the inflammatory response, augments expressed in phagocytic cells.42 S100A8 and S100A9 rapidly
the production of DAMPs, and promotes both efferocytosis associate to form the S100A8/S100A9 heterodimer. S100A8/
of dying cells and tissue digestion via the release of proteases S100A9 complexes function as DAMPs secreted by neutro-
and oxidases.3,22 Efficient efferocytosis of apoptotic cardiomyo- phils and monocytes/macrophages during inflammatory condi-
cytes is particularly important for transitioning to the phase of tions and signal via RAGE and TLR4 receptors.43,44 Humans

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


94  Circulation Research  June 24, 2016

with acute MI exhibit elevated serum S100A8/A9 levels that domain-containing adaptor protein to trigger receptor complex
correlate with circulating neutrophil counts and the risk of car- interactions with IL-1R–associated kinases 4, 1, and 2, TNF
diovascular death and subsequent MI.45,46 In mice with reper- receptor–associated factor 6, and the MAPKKK transforming
fused MI, S100A8/S100A9 is rapidly expressed and released growth factor (TGF) activated kinase-1. As fully reviewed else-
after ischemia, primarily by inflammatory cells and fibroblasts, where,13,14 these signaling cascades ultimately activate NF-κB
and induces proinflammatory signaling, leukocyte infiltration, (p65 and p50) and MAPK pathways to upregulate a broad ar-
and cardiac dysfunction in a RAGE-dependent manner,47 sug- ray of proinflammatory mediators (Figure 2). TLR4 is also en-
gesting that these DAMPs are central to post-MI inflammation. docytosed after ligand binding; endolysosomal TLR4 signals
Interestingly, S100A1, the S100 protein most abundant in car- in a MyD88-independent manner via the cytoplasmic adaptor
diomyocytes, is also released from damaged cardiomyocytes TIR domain-containing adaptor inducing interferon-β and the
in both humans and mice with acute MI.48 However, rather than bridging adaptor TRIF-related adaptor molecule (TIR domain-
promoting generalized inflammation, S100A1 is taken up by containing adaptor inducing interferon-β–related adaptor mol-
endocytosis in adjacent cardiac fibroblasts to transiently acti- ecule). This pathway results in NF-κB nuclear translocation,
vate TLR4-endolysosomal signaling, resulting in an immuno- and the induction of type I interferon via activation of TANK-
modulatory and antifibrotic phenotype with beneficial effects binding kinase and the transcription factor interferon-regulato-
on post-MI LV remodeling in vivo.48 This suggests that specific ry factor 3. In the heart, the most highly expressed TLRs are
DAMPs have unique cell targets and functional roles in the car- TLR4, TLR2, TLR3, and TLR5,13 with TLR4 and TLR2 the
diac repair process that can be either pro- or anti-inflammatory. most studied in the context of myocardial injury.
In this regard, the β-galactoside–binding lectin galectin-1 is Augmented TLR4 activation, and increased expression of
expressed by hypoxic cardiomyocytes and infiltrating leuko- proinflammatory mediators downstream of TLR4 signaling,
cytes after MI and also imparts anti-inflammatory and cardio- has been demonstrated in circulating leukocytes from humans
protective effects in the remodeling heart.49 with acute MI,55–57 and correlated with the development of HF.56
Fibronectin is an extracellular matrix protein secreted by Similar finding has also been reported for TLR2 in circulating
fibroblasts in response to tissue injury and proinflammatory monocytes.58 Moreover, cardiac TLR4 expression increases both
cytokines,50 and includes an alternatively spliced exon coding after acute MI59 and in chronic HF.60,61 Mice with genetic disrup-
type III repeat extra domain A that binds to TLR-4 to activate tion or deficiency of TLR4,62,63 TLR2,64,65 MyD88,66 or TLR367
mast cells and leukocytes.51,52 Mice with parenchymal myocar- exhibit reduced infarct size after I/R and amelioration of patho-
dium-localized fibronectin-extra domain A deficiency exhibited logical remodeling after nonreperfused MI.59,67–70 Moreover, pre-
improved LV remodeling and function, less monocyte recruit- treatment with eritoran, a specific TLR4 antagonist, in mice,71
ment, and reduced remote zone fibrosis after nonreperfused MI or with an anti-TLR2 antibody in mice64 and pigs72 reduced
as compared with wild-type mice, indicating a critical role for infarct size after myocardial I/R. In both MI models, sustained
fibronectin-extra domain A in tissue inflammation and remodel- TLR-mediated signaling generally augmented cell apoptosis,
ing.53 Conversely, recent studies have demonstrated that necrot- inflammation, interstitial fibrosis, oxidative stress, and leuko-
ic cardiomyocytes (but not fibroblasts) release IL-1α as danger cyte recruitment, indicating maladaptive responses triggered by
signal that activates proinflammatory MAPK and NF-κB signal- TLR2, TLR4, and TLR3 after MI. Interestingly, and in contrast,
ing in cardiac fibroblasts in a myeloid differentiation factor 88 beneficial effects were referable to TLR5 after I/R, as TLR5 de-
(MyD88)–dependent but NLRP3- and TLR-independent man- ficiency increased infarct size, oxidant stress, inflammation, and
ner, via the activation of the IL-1R pathway.54 Hence, multiple LV dysfunction after reperfused MI.73
danger signals act in a concerted fashion on parenchymal and Interestingly, short-term TLR2,74,75 TLR4,13,76,77 and
inflammatory cells in the infarcted heart to drive and modulate TLR978 activation before I/R induces preconditioning and car-
inflammation. For a further discussion of DAMPs, the reader is dioprotection (with reductions in infarct size), in part, via TIR
referred to several comprehensive reviews.8,10–12 domain-containing adaptor protein- and PI3K/Akt-dependent
TLRs, NLRs, and RAGE mechanisms. Analogous short-term protective effects in car-
TLRs: The TLRs comprise the major PRRs on mammalian diac myocytes have similarly been observed for several innate
cells.14 Expressed most prominently on leukocytes, TLRs are immune signaling mediators, including NF-κB,79 inflamma-
also expressed by parenchymal cells, including cardiomyo- tory cytokines,80 and chemokines.81,82 These observations
cytes, fibroblasts, and endothelial cells. To date, 13 functional suggest a paradigm whereby short-term activation of innate
mammalian TLRs have been identified (10 in humans, TLRs immune pathways, primarily localized to cardiomyocytes,
1–10)13,14, that recognize a variety of pathogen-associated mo- yields cytoprotective and prosurvival effects via mitochon-
lecular patterns14 and DAMPs11,12 to trigger innate immune re- drial stabilization, whereas activation that is more prolonged
sponses. Of these, TLRs 1, 2, 4, 5, 6, and 11 are cell-surface or of greater magnitude and involving immune cells results
receptors, whereas TLR3 and TLRs 7 to 10 are expressed in in more robust inflammatory responses and leukocyte recruit-
endolysosomes.13,14 Signal transduction by TLRs and IL-1Rs ment that induce tissue injury.13,79,80,83 In this regard, in vivo
occurs through a conserved cytoplasmic Toll/IL-1R (TIR) do- chimeric mouse models64 and ex vivo isolated perfused heart
main that serves as the docking site for TIR-containing cyto- studies (in which circulating leukocytes are eliminated)84
plasmic adaptor proteins (Figure 2). Except for TLR3, all TLRs have demonstrated that after I/R, leukocyte-localized TLR2
(and IL-1Rs) engage with the adaptor MyD88 either directly is responsible for inducing myocardial injury and determines
or, for TLR2 and TLR4, in combination with the adaptor TIR infarct size, whereas parenchymal TLR2 signaling induces

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   95

Figure 2. Toll-like receptor (TLR) and nucleotide-binding oligomerization domain–like receptor (NLR) signaling. Danger-
associated molecular patterns (DAMPs) released from necrotic and damaged cells and extracellular matrix bind to the TLRs, which
comprised an extracellular leucine-rich repeat (LRR) domain, a transmembrane (TM) domain, and a conserved cytoplasmic Toll/
interleukin (IL)-1R (TIR) domain, which serves as docking site for other TIR-containing cytoplasmic adaptor proteins (top left). The
binding of DAMPs, and often coreceptors such as CD14 and MD2, to TLR4 engages the adaptor myeloid differentiation factor 88
(MyD88) via the adaptor TIR domain-containing adaptor protein (TIRAP). MyD88 recruits IL-1R–associated kinase (IRAK) 4 and the
IRAK1/2 and TNF receptor associated factor (TRAF) 6 complex, which activates transforming growth factor activated kinase 1 (TAK1).
TAK1, a mitogen-activated protein kinase (MAPK) KK, then activates the MAPK cascade and also phosphorylates IκB kinase (IKK),
that leads to nuclear factor-κB (NF-κB) p65 and p50 nuclear translocation and the transcription of a panel of inflammatory genes,
including cytokines, chemokines, cell adhesion molecules, and complement factor B. Furthermore, after endocytosis, TLR4 signals in
a MyD88-independent manner via the cytoplasmic adaptor TRIF, in turn recruited through the bridging adaptor TRAM. This pathway
results in noncanonical IKK and NF-κB activation, as well as the induction of type I interferon (IFN) via TANK-binding kinase (TBK) 1 and
the transcription factor IFN-regulatory factor (IRF) 3. Except for TLR3, the other TLRs also signal, directly or indirectly as depicted, via
MyD88. Endolysosomal TLR3 activates IRF3 signaling via TRIF and TBK1, whereas endolysosomal TLR7/8 and TLR9 also induce type
I IFN via TRAF3 and IRF7 in a MyD88-dependent manner (not pictured). In addition to TLRs, intracellular NLRs respond to a variety of
DAMPs via the formation of multiprotein inflammasomes, comprised an activated NLR protein, the adaptor protein apoptosis speck-like
protein containing a caspase-recruitment domain (ASC), and procaspase-1. One proposed model of NLRP3 inflammasome activation
results from stimulation of the purinergic P2X7 ion channel by extracellular ATP, resulting in K+ efflux, recruitment of the pannexin-1 pore,
DAMP entry into cytosol, and activation of NLRP3. This activates caspase-1, which then converts pro–IL-18 and pro–IL-1β to active IL-
18 and IL-1β. As discussed in the text, cell-type–specific innate immune signaling can result in distinctive, and sometimes divergent, in
vivo physiological responses during acute MI. Schema derived from Mann,13 Newton and Dixit,14 Schroder and Tschopp,18 Chao,76 and
Mann83 (Illustration credit: Ben Smith).

contractile dysfunction without affecting infarct size. In con- (nucleotide-binding oligomerization domain 1 and nucleotide-
trast, both parenchymal and leukocytic TLR2 are needed binding oligomerization domain 2) that activate NF-κB, and
to mediate endothelial injury and dysfunction after I/R.65 the NLRs (NLRP1, NLRP3/cryopyrin, NLRC4) that augment
Interestingly, divergent effects of leukocyte vis-à-vis cardio- IL-1β and IL-18 secretion via the formation of multiprotein sig-
myocyte TLR4 signaling on cardiomyocyte impairment have naling complexes termed inflammasomes. The NLRs contain
also been shown in a model of systemic sepsis.85 Hence, the a caspase activation and recruitment or pyrin domain at the N
effects of TLR signaling, and proinflammatory mediators in terminus, a central NACHT domain, and a C-terminal leucine-
general, are complex and graded, rather than all-or-none, and rich repeat region. The inflammasome consists of an activated
depend heavily on the cellular and disease context. NLR protein, the adaptor protein apoptosis speck-like protein
NLRs: The NLR family of intracellular PRRs responds containing a caspase-recruitment domain, and procaspase-1.
to a variety of DAMPs (eg, ATP and uric acid) and in- The complex facilitates caspase-1 activation, which converts
clude nucleotide-binding oligomerization domain receptors pro–IL-18 and pro–IL-1β (generated by MyD88-dependent

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


96  Circulation Research  June 24, 2016

TLR pathways) to active IL-18 and IL-1β (Figure 2). One c-Rel, p50, and p52—comprise the NF-κB family; these sub-
proposed model of activation of NLRP3 involves extracellular units form homo- or heterodimers to modulate gene transcrip-
ATP stimulation of the purinergic P2X7 ion channel; this trig- tion. Classically, p65/p50 heterodimers are bound to inhibitor
gers K+ efflux, recruitment of the pannexin-1 membrane pore, of κBα (IκBα) in the cytoplasm. On IκBα phosphorylation by
and entry of DAMPs into cytosol to access NLRP3 (Figure 2).18 IκB kinase, IκBα is rapidly ubiquitinated and degraded by the
The major components of the NLRP3 inflammasome 26S proteasome, allowing for subunit nuclear translocation.
(apoptosis speck-like protein containing a caspase-recruitment Activation can also occur via IκBα-independent mechanisms
domain, cryopyrin, caspase-1) are upregulated and activated to release p50/RelB or p52/RelB. Importantly, only p65, RelB,
early after MI in a variety of cell types in the heart, not only in- and c-Rel contain transactivation domains; hence, p50 and p52
filtrating leukocytes, fibroblasts, and endothelial cells but also homodimers can repress rather than activate gene transcription.
border zone cardiomyocytes.86–88 IL-1β and IL-18, the cytokine Therefore, subunit-dependent differences in target gene speci-
end-products of inflammasome activation, are also increased ficity impact the transcriptional response and may contribute to
early after MI.88–90 Inflammasome activation has been suggest- the spectrum of effects observed on NF-κB activation, ranging
ed to occur initially in cardiac fibroblasts during reperfused MI, from cardioprotection to injury and cell death.
in response to ROS production and K+ efflux.86 Global targeted In humans with acute MI, circulating leukocytes exhibit
genetic disruption of apoptosis speck-like protein containing a marked activation of NF-κB.57,93 In rodents, NF-κB is ac-
caspase-recruitment domain or caspase-1,86 as well as antibody tivated in the heart early after MI in the infarct zone,89,90,94
neutralization of IL-18,90 has been shown to reduce infarct size and later (after 24 hours) in the remote zone.95 Importantly,
in vivo after I/R in mice. Apoptosis speck-like protein contain- whereas there is nuclear translocation of both p65 and p50 in
ing a caspase-recruitment domain loss-of function also im- the first 24 hours after MI, the profile shifts near exclusively
proved post-MI cardiac remodeling and fibrosis and decreased to p65 at later time points.95 Studies on a cardioprotective vis-
leukocyte infiltration and the expression of proinflammatory à-vis detrimental role of NF-κB during MI have yielded con-
cytokines and chemokines. Studies using chimeric mice indi- flicting results.79 Acute NF-κB activation is essential for late
cate that inflammasome activation in leukocytes and resident cardioprotection induced by ischemic preconditioning.96,97
cardiac cells both contribute to ischemic injury.86 Similarly, However, mice with cardiomyocyte-restricted overexpression
isolated perfused NLRP3−/− murine hearts exhibit reduced of phosphorylation-resistant IκBα,98 cardiomyocyte-specific
infarct size and improved contractile function during ex vivo p65 deletion,99 IκBα overexpression via gene transfer,100 NF-
I/R,88 whereas gene silencing of cryopyrin ameliorates cardiac κB double-stranded decoy DNA transfection,101 and phar-
remodeling and dysfunction in vivo after nonreperfused MI in macological blockade of IκBα102 or IκB kinase β103 have
mice.87 Hence, the NLRP3 inflammasome is a key mediator of demonstrated that NF-κB inhibition (primarily in cardiomyo-
the post-MI inflammatory response and tissue injury. cytes and perhaps more related to p65) during myocardial I/R
RAGE: RAGE is a PRR expressed by a variety of immune decreases infarct size, reduces inflammatory responses in-
and parenchymal cell types that interacts with several DAMPs cluding leukocyte infiltration, and improves cardiac function.
such as HMGB1 and S100A8/S100A9. RAGE activation Studies of nonreperfused MI in mice with cardiomyocyte-
triggers many intracellular signaling pathways, including restricted overexpression of phosphorylation-resistant IκBα95
NF-κB– and MAPK-dependent inflammatory genes.19 RAGE- or A20 (NF-κB signaling inhibitor),104 and IκBα gene trans-
deficient mice exhibited reduced infarct size, reduced leuko- fer105 have similarly shown that blocking NF-κB (mainly p65
cyte infiltration and inflammatory cytokine expression, and based on time course studies95) attenuates long-term adverse
improved cardiac remodeling and function after reperfused cardiac remodeling, dysfunction, and inflammation.
MI.15,47 Studies of chimeric mice indicate that RAGE signal- The p50 subunit lacks a transactivation domain and thus
ing in infiltrating leukocytes, rather than resident cardiac cells, can inhibit NF-κB transcriptional activity. A study using car-
is primarily responsible for these adverse proinflammatory diac magnetic resonance imaging in p50−/− mice demonstrated
and remodeling responses post MI.47 Thus, inflammatory cell-
that leukocyte p50 expression imparts beneficial effects on re-
localized RAGE, in particular, amplifies and promotes tissue
modeling after nonreperfused MI, by improving scar stability
injury during MI.
and matrix remodeling, and attenuating leukocyte infiltration
Nuclear Factor-κB and cytokine expression.106 These results contrasted with prior
NF-κB is a central transcriptional effector of inflammatory sig- I/R and permanent ligation studies of p50−/− mice that reported
naling. NF-κB activation and its subsequent nuclear transloca- the opposite results,107,108 but were consistent with a more re-
tion after MI trigger transcription of a large portfolio of genes cent study showing no effects of myocardial p50 deficiency
including inflammatory cytokines, CXC and CC chemokines, on infarct size during ex vivo I/R in the isolated perfused heart
and adhesion molecules. The spatial and temporal expression when the influence of circulating leukocytes was removed.109
of these mediators in resident and infiltrating cells choreo- Taken together, although further study is clearly required, the
graphs events that further amplify the inflammatory response prevailing evidence underscores the importance of subunit-
(cytokines) and attract and recruit specific leukocyte popula- specificity and cellular localization as determinants of NF-
tions (chemokines and adhesion molecules) to injured myo- κB–mediated responses after MI. One possible scenario is that
cardium. The signaling pathways and outputs linked to NF-κB cardiomyocyte p65 activation imparts tissue injurious effects,
activation, as well as its effects in cardiac diseases, have been whereas leukocyte p50 may provide a counterbalance to tem-
reviewed extensively.14,79,91,92 Five subunits—p65 (RelA), RelB, per excessive inflammation.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   97

Cytokines and Chemokines of anti-inflammatory monocytes,123 impaired macrophage


A variety of proinflammatory cytokines and chemokines are activation and aggravated post-MI cardiac remodeling.124
upregulated after MI as a result of innate immune activa- Moreover, clinical studies of anticytokine (anti-TNF biolog-
tion. The effects of cytokines and chemokines in normal and ics and IL-1R antagonists)83 and antichemokine strategies110
injured hearts have been extensively reviewed3,79,80,83,110–112; in humans with MI or HF have not proven clinical benefit
hence, only a few points will be highlighted in this review. (and in some cases suggested harm), suggesting that indis-
Proinflammatory cytokines such as IL-1, TNF, IL-6, and IL- criminate cytokine blockade eliminates both the beneficial
18 are upregulated and secreted early after MI and are key and the detrimental effects of cytokines and chemokines,
regulators and propagators of the inflammatory response. thereby yielding neutral responses.
This occurs because of (1) downstream signaling that fur- Indeed, these and other studies indicate that proinflamma-
ther amplifies the initial inflammatory responses via MAPK- tory cytokines and chemokines induce effects in the infarcted
and NF-κB pathways, (2) spatial, paracrine extension of heart that are not simply gradations of “good” versus “bad,”
the inflammatory response to neighboring parenchymal and but rather are complex and variable, depending on such fac-
infiltrating cells that express cytokine receptors, and (3) re- tors as the temporal and disease context, the prevailing cellu-
cruitment of leukocytes via the upregulation of adhesion lar composition in the microenvironment, and accompanying
molecules in endothelial cells and chemokines in myocar- pleiotropic influences on multiple (immune and nonimmune)
dium.3,22 Chemokines are broadly divided into CC, CXC, and cell processes that include not only inflammatory responses
CX3C subtypes.110 In general, the CC chemokines are strong but also events such as growth, differentiation, apoptosis, oxi-
attractants for mononuclear cells, whereas glutamic acid- dative stress, and mitochondrial function. For example, stud-
leucine-arginine+ CXC chemokines are strong neutrophil ies in mice deficient for TNF receptor (TNFR) 1 or 2 have
chemoattractants.3,110 The chemokine expression portfolio shown that after nonreperfused MI, TNF induces divergent
then regulates the recruitment of leukocyte subpopulations to TNFR-specific effects on remodeling, hypertrophy, NF-κB
infarcted myocardium. activity and inflammation, border zone fibrosis, and apoptosis
The central importance of specific cytokines and chemo- such that TNFR1 exacerbates, whereas TNFR2 ameliorates,
kines in the inflammatory response after MI is supported by these events.125 This suggests that opposing receptor-specific
multiple studies, including findings (among others) that (1) myocardial responses in vivo95,125,126 may explain the nega-
TNF−/− mice (or wild-type mice treated with anti-TNF anti- tive clinical trial results with global TNF blockade. Also,
body) exhibit smaller infarcts, attenuated leukocyte infil- although much of the current research focus is on the heart-
tration, and lower expression of chemokines and adhesion localized effects of proinflammatory mediators, extracardiac
molecules after I/R,113 (2) IL-1R type I deficient mice exhibit effects may be of equal import to cardiac repair. In this re-
less LV dilatation and dysfunction after reperfused MI, and gard, a recent study has demonstrated that circulating IL-1β
similar reductions in cardiac neutrophil and macrophage infil- induces the proliferation of bone marrow hematopoietic stem
tration and chemokine/cytokine expression,114 (3) mutant mice cells after MI, thereby enhancing circulating leukocytes and
with augmented activation of gp130, the common receptor inflammation in the infarcted heart.127 As another example,
subunit for the IL-6 cytokine family, exhibit adverse remodel- the chemokine CXCL12/stromal cell–derived factor-1 may
ing and heightened myocardial inflammation,115 (4) wild-type facilitate cardiac repair after MI by promoting homing and
mice treated with a competitive monocyte chemoattractant survival of stem cells and neovascularization.128,129 Finally, it
protein-1/chemokine (C-C motif) ligand 2 (MCP-1/CCL2) in- is important to recognize that the inflammation is a required
hibitor exhibit reduced infarct size and monocyte infiltration event for effective tissue repair, and as such suppression of
after I/R,116 whereas MCP-1–deficient mice have amelioration inflammatory activation that is not dysregulated or excessive
of adverse post-MI remodeling,117 and (5) mice deficient in may not necessarily result in salubrious effects on cardiac
CCR2, the cognate receptor for MCP-1, also exhibit reduced remodeling after MI. Moreover, there is evidence that loss-
infarct size and macrophage infiltration after I/R,118 and ame- of-function of select proinflammatory mediators (eg, MCP-
lioration of post-MI remodeling.119 1,117 IL-1,114 and myeloperoxidase130) attenuate inflammation
Nonetheless, despite preclinical data suggesting that and adverse cardiac remodeling but do not impact cell death
cytokines and chemokines serve to aggravate ischemic in- and infarct size during I/R. Hence, suppression of the inflam-
jury and remodeling, it should be recognized that several matory cascade during MI need not be accompanied by car-
studies conflict with this paradigm, showing that these me- diomyocyte salvage. These caveats are important to consider
diators engender cardioprotective responses and pleiotropic when designing immunomodulatory therapeutic strategies to
cellular effects on both immune and myocardial resident enhance post-MI cardiac repair.
cells.22,80,110,111 In mice, for example, dual TNF receptor de-
ficiency exacerbated ischemic injury and myocyte apoptosis Cellular Effectors of the Inflammatory Response
during I/R,120 IL-6 deficiency did not impact either infarct Cardiomyocytes, immune cells, vascular cells, and fibroblasts
size or post-MI remodeling,121 activation of signal transducer have been implicated as cellular effectors of the inflammatory
and activator of transcription 3 (signal transducer and activa- reaction in the healing infarct; their relative role in activation
tor of transcription 3, a signaling molecule downstream of of specific inflammatory cascades remains unclear. Resident
gp130), either via IL-11 or constitutively, attenuated post-MI myocardial cells sense tissue necrosis and trigger the postin-
remodeling and fibrosis and improved neovascularization,122 farction inflammatory reaction leading to recruitment of circu-
and CCR5 deficiency resulted in attenuated recruitment lating leukocyte subpopulations.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


98  Circulation Research  June 24, 2016

Cardiomyocytes may increase in size in the inflamed myocardium, thus serv-


Necrotic cardiomyocytes provide the main stimulus for the ing as exit points for extravasating neutrophils.146 Neutrophil
postinfarction inflammatory reaction, by releasing DAMPs extravasation across the microvasculature requires binding of
in the infarcted area. Surviving cardiomyocytes in the infarct leukocyte integrins to endothelial adhesion molecules147 and
border zone may also trigger inflammatory activation, by pro- subsequent interactions between endothelial integrin ligands
ducing and secreting cytokines in response to activation with and junctional proteins. Emigrated neutrophils release proteo-
IL-1, TLR ligands, or ROS. Immunohistochemical studies and lytic enzymes and contribute to the clearance of the wound
in situ hybridization experiments have suggested that viable from dead cells and matrix debris. Infiltrating neutrophils may
cardiomyocytes in the infarct border zone express intercellu- also amplify the immune response.148 Although both in vitro
lar adhesion molecule-1131 and may synthesize cytokines16 and and in vivo experiments have suggested that neutrophils may
chemokines.132 The relative contribution of cardiomyocyte- exert direct cytotoxic actions on viable cardiomyocytes ex-
derived inflammatory mediators in progression and extension tending ischemic injury,149,150 the significance of such effects
of postinfarction inflammation remains unknown. in the clinical context remains controversial.151

Endothelial Cells Monocyte Subpopulations


The heart is a highly vascular organ; in adult mammals, en- Two distinct waves of monocyte recruitment have been iden-
dothelial cells are the most abundant noncardiomyocytes.133 tified in healing myocardial infarcts.152 Early recruitment of
Extravasation of leukocytes into the infarcted area requires proinflammatory Ly6Chi monocytes is mediated through the ac-
endothelial activation. Endothelial-specific activation of the tivation of the MCP-1/CCR2 axis.117 At a later stage, anti-inflam-
transcription factor forkhead box O4 after infarction has been matory monocyte subpopulations are selectively recruited and
demonstrated to promote neutrophil infiltration in the infarct- may participate in resolution of the postinfarction inflammatory
ed heart.134 DAMPs released by dying cardiomyocytes induce response. During the first few hours after infarction, high levels
rapid upregulation of endothelial adhesion molecules, trigger- of IL-1 in the infarct may stimulate a proinflammatory program
ing adhesive interactions with activated leukocytes. Preformed in infarct monocytes. Monocytes infiltrating the infarcted myo-
P-selectin is rapidly mobilized from Weibel-Palade bodies,135 cardium originate not only from the bone marrow but also from
and E-selectin is upregulated in the ischemic endothelium. the spleen, which may serve as a large reservoir of mononuclear
Once expressed on the endothelial surface, selectins bind cells that can be rapidly deployed to sites of inflammation.153
to their leukocyte ligands, capturing neutrophils and mono- Lymphocytes
cytes and mediating rolling along the venular endothelium.136 Early infiltration of the infarcted heart with lymphocyte sub-
Moreover, activated endothelial cells in the infarct zone serve sets has been extensively documented in both large animal154
as an important source of cytokines and chemokines.137,138 and in rodent models41 of MI. Experiments in a rat model of
Neutrophils MI suggested that cytotoxic T lymphocytes are activated after
Neutrophils are the first immune cell type to infiltrate the in- infarction; in vitro studies suggested that these cells may exert
farcted myocardium3,41 in response to such factors as DAMPs, cytotoxic actions on healthy cardiomyocytes.155 Whether infil-
cytokines and chemokines, endogenous lipid mediators (eg, trating T cells extend ischemic injury in vivo remains unknown.
prostaglandin E2, leukotriene B4), histamine, and complement Emerging evidence suggests that lymphocyte subpopulations
components.139–141 Infiltration of the infarct with neutrophils may play an important role as orchestrators of the inflammatory
is predominantly localized in the border zone and is acceler- response. Using both genetic and antibody-mediated depletion
ated and accentuated by reperfusion. Neutrophil extravasation strategies, Zouggari et al156 demonstrated that B cells promote
in the infarcted heart is dependent on activation of adhesive mobilization of proinflammatory Ly6Chi monocytes, thus play-
interactions between the leukocytes and the endothelial cells ing an important role in activation of the inflammatory cascade.
(Figure 3). Circulating neutrophils expressing selectin ligands Fibroblasts
are captured by the activated endothelium and roll along The adult mammalian myocardium contains abundant cardiac
the endothelial layer. Rolling neutrophils sense chemokines fibroblasts157; in the absence of injury, these cells remain qui-
bound to glycosaminoglycans on the endothelial surface. escent and may play a role in maintaining the extracellular
Interactions between CXC chemokines and the CXCR2 re- matrix network. However, when stimulated with DAMPs,
ceptor expressed by the neutrophils induce conformational fibroblasts are capable of secreting large amounts of inflam-
changes of leukocyte integrins142,143 strengthening the adhe- matory cytokines and chemokines.158 In the infarcted myocar-
sive interaction, and resulting in arrest and adhesion of the dium, fibroblasts may respond to stimulation with ROS and
neutrophil to the endothelial surface. Extensive experimental IL-1, acquiring a proinflammatory phenotype, and serving as
evidence suggests that binding of neutrophil integrins, such an important source of chemokines and cytokines.159 Because
as lymphocyte function-associated antigen 1 and macro- several other cell types are capable of proinflammatory acti-
phage-1 antigen (Mac1), with endothelial intercellular adhe- vation during the early phase of infarct healing, the relative
sion molecules is essential for firm adhesion.144 Neutrophil contribution of fibroblasts remains unknown. Activation of
transmigration follows, as leukocytes actively crawl toward IL-1 signaling in cardiac fibroblasts during the inflammatory
endothelial junctions, then migrate through basement mem- phase of cardiac repair inhibits α-smooth muscle actin expres-
brane regions with low levels of matrix protein expression.145 sion and delays myofibroblast conversion, promoting a ma-
These regions may overlap with gaps between pericytes that trix-degrading phenotype (Figure 4).159 Thus, cytokine-driven

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   99

Figure 3. Neutrophil extravasation in the infarcted myocardium is dependent on activation of adhesive interactions between
leukocytes and endothelial cells (EC). Proinflammatory mediators induce expression of selectins on the endothelial surface, leading
to tethering and rolling of circulating neutrophils. Rolling neutrophils sense chemokines bound to glycosaminoglycans on the endothelial
surface and exhibit integrin activation. Interactions between leukocyte integrins and their endothelial ligands result in firm adhesion of
the neutrophils. Subsequently neutrophils crawl toward endothelial junctions and transmigrate between pericytes, through basement
membrane regions with low expression of matrix proteins. Extravasated neutrophils release proteases (both serine proteases and matrix
metallproteinases), and reactive oxygen species (ROS), thus contributing to clearance of the wound. Neutrophils may also modulate
inflammatory responses, both by secreting cytokines, and by regulating cytokine activity through release of proteases. Excessive or
prolonged neutrophil actions may promote matrix degradation. Because neutrophils are predominantly localized in the infarct border
zone, it has been suggested that they may adhere to viable cardiomyocytes, exerting cytotoxic effects. However, the significance of
leukocyte-mediated cardiomyocyte injury remains debated.

inflammatory activation of the fibroblasts may prevent prema- of the inflammatory cascade. During progression of the inflam-
ture acquisition of a synthetic myofibroblast phenotype, until matory cascade, recruitment of large numbers of monocytes and
the infarct is cleared of dead cells and matrix debris. proliferation of resident macrophage subsets results in marked
expansion of the cardiac macrophage population. The abundant,
Resident Mast Cells and Macrophages
dynamic, and highly plastic population of infarct macrophages
The heart contains resident populations of mast cells and mac-
plays an important role in regulation of the inflammatory and
rophages that may play an important role in the activation of
reparative response after MI.
the inflammatory cascade. Mast cells are strategically located
in perivascular areas and contain preformed stores of inflam- Extracellular Matrix
matory mediators, such as TNF, histamine, and tryptase.160,161 Both cardiomyocytes and noncardiomyocytes are enmeshed
Cytokine stimulation, adenosine, ROS, and activation of the in a network of extracellular matrix proteins. The cardiac
complement cascade induce mast cell degranulation. TNF and interstitial matrix does not only simply serve as a structural
histamine released by resident mast cells may play an impor- scaffold but also transduce molecular signals and play an ac-
tant role in triggering the postinfarction inflammatory response. tive role in regulation of inflammatory and reparative respons-
Recent studies have characterized the resident macrophage es.166,167 Fragmentation of the extracellular matrix provides a
population in mouse myocardium.162–165 Using flow cytometry key stimulus for activation of the inflammatory cascade after
and lineage tracing approaches, Epelman et al162 found signifi- infarction. Generation and release of collagen and fibronectin
cant heterogeneity in macrophage populations in adult mouse fragments have been implicated in activation of proinflamma-
hearts. At steady state, 2 distinct macrophage pools were identi- tory signaling.168 Hyaluronan degradation may result in release
fied: a CCR2-negative subset that represented an embryonically of high molecular weight fragments with potent proinflamma-
established lineage that originated from yolk sac macrophages tory properties, capable of inducing cytokine and chemokine
and fetal monocytes and a second (much smaller) pool derived synthesis by endothelial and immune cells.169
from circulating CCR2+ monocytes. The fate of these subpopu-
lations after infarction and their contribution in regulation of the
Reparative and Proliferative Phase
postinfarction inflammatory reaction remains unclear. Heidt et
al163 suggested that, at least in nonreperfused infarcts, resident Inhibition and Resolution of the Inflammatory
cardiac macrophages die and may be replaced by monocyte- Response
derived CCR2-expressing cells with potent proinflammatory Cell Types Involved in Suppression of the Inflammatory
properties. Reperfusion may protect resident macrophage sub- Response
populations in the infarcted area; thus, in models of reperfused Neutrophils: The transition from the inflammatory to the re-
infarction, these cells may play an important role in activation parative and proliferative phase after MI is driven by changes

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


100  Circulation Research  June 24, 2016

Figure 4. Cellular effectors and molecular signals that repress and resolve inflammation after myocardial infarction leading
to the transition from the inflammatory to the proliferative phase of cardiac repair. Recruitment of anti-inflammatory monocyte
(Mo) subsets (1), T-cell subpopulations, such as regulatory T cells (Tregs) (2) and invariant Natural Killer T cells (iNKT; 3) contributes to
repression of the postinfarction inflammatory response. Moreover, members of the transforming growth factor (TGF)-β family (such as
TGF-β1 and growth differentiation factor (GDF)-15 inhibit neutrophil transmigration by attenuating expression of adhesion molecules
by endothelial cells (ECs) (4). Recruitment of pericytes (P) by microvascular ECs is mediated through platelet-derived growth factor
(PDGF) receptor-β actions and may also contribute to suppression of postinfarction inflammation (5). Macrophages (Ma) acquire an anti-
inflammatory phenotype, secreting TGF-β, interleukin (IL)-10, and proresolving lipid mediators, on ingestion of apoptotic neutrophils
(aN) (6). Dendritic cells (DCs) are also activated after infarction and secrete anti-inflammatory cytokines (7). Cardiomyocytes (CM) in the
border zone may contribute to suppression and spatial containment of the postinfarction inflammatory response by secreting mediators
that promote an anti-inflammatory macrophage phenotype (such as regenerating islet-derived-3β [Reg-3β]; 8). Fibroblasts (F) also exhibit
dynamic phenotypic alterations that mark the transition from the inflammatory to the proliferative phase (9). During the inflammatory
phase, inflammatory cytokines (such as IL-1β and TNF-α) and activation of toll-like receptor (TLR)–dependent signaling by matrix
fragments may activate a proinflammatory fibroblast phenotype. Stimulation of fibroblasts with IL-1β induces matrix metallproteinase
expression and chemokine synthesis, while reducing α-smooth muscle actin levels. During the proliferative phase, activation of TGF-
β–dependent cascades stimulates a matrix-preserving myofibroblast (MF) phenotype.

in the cardiac microenvironment. Although their survival can in the infarcted heart. A recent study demonstrated that neu-
be prolonged by DAMPs, proinflammatory cytokines, hypox- trophils, via secreted neutrophil gelatinase-associated lipo-
ia, and acidosis, neutrophils are short-lived cells that rapidly calin, polarize macrophages toward a reparative phenotype,
undergo cell death, primarily by apoptosis but also secondarily thereby orchestrating tissue healing (Figure 4).171
necrosis.139,170 In various models of acute inflammation, late- Monocytes, macrophages, and DCs: After the early ap-
stage and apoptotic neutrophils are critical for ushering inflam- pearance of neutrophils, monocytes and macrophages (Mo/
mation resolution by several mechanisms: (1) the release of Mϕ) comprise the most abundant cells in the infarcted
mediators that promote inflammation resolution such as pro- heart.3,4,41 Seminal studies by Nahrendorf, Swirski, and co-
resolving lipid mediators (eg, lipoxins and resolvins), annexin workers4,152,172–174 demonstrated that Mo/Mϕ display phasic
A1, and lactoferrin that dampen neutrophil transmigration and functional heterogeneity that serve to guide proper wound
entry, and promote neutrophil apoptosis and the phagocytic healing. The initial phase (peak day ≈3–4 post MI) promotes
uptake of apoptotic neutrophils by macrophages,139–141 (2) the tissue digestion and is characterized by Ly6Chi monocytes and
expression of decoy and scavenging chemokine and cytokine M1 macrophages that are proteolytic, with augmented expres-
receptors on apoptotic neutrophils that results in tissue deple- sion of proteinases (eg, cathepsins and matrix metallprotein-
tion of these mediators,139,141 and (3) the expression of eat-me ases), and proinflammatory, with augmented TNF expression.
signals (eg, phosphatidylserine) that facilitate the ingestion of The second phase (peak day ≈7 post MI) promotes tissue re-
apoptotic neutrophils by macrophages.139,141 The subsequent pair, with a predominance of Ly6Clo monocytes and M2-like
phagocytic clearance of these apoptotic cells induces a pro- macrophages with augmented expression of anti-inflammato-
resolving M2 phenotype in macrophages and secretion of ry, profibrotic, and angiogenic factors (eg, IL-10, TGF-β, and
anti-inflammatory and profibrotic cytokines such as IL-10 and vascular endothelial growth factor). It should be emphasized
TGF-β that suppress inflammation and promote tissue repair. that in the healing infarct, macrophages cannot be simply cat-
It should be noted that although these are fundamental aspects egorized as polarized M1/M2 cells, but exhibit a wide range
of inflammatory cell biology, they have not been widely tested of nuanced phenotypes. Moreover, the repertoire of reparative

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   101

monocytes/macrophages is not limited to traditional cytokines the inflammatory response. In contrast, CD4−γδT-cells seem
and growth factors. A recent investigation identified a novel to promote neutrophil and macrophage infiltration and impart
secreted protein called myeloid-derived growth factor, as an detrimental effects on post-MI remodeling.182
essential monocyte-derived mediator that may promote repair Vascular cells: Tissue neovascularization is essential for
of the infarcted heart.175 supplying the healing infarct with nutrients and oxygen. A ro-
Initial studies4,152 suggested that the 2 phases resulted from bust angiogenic response occurs after MI with rapid upregu-
separate waves of circulating monocyte infiltration—early re- lation of VEGF in viable border zone cardiomyocytes, and
cruitment of Ly6ChiCCR2+CX3CR1lo monocytes in response upregulation of vascular endothelial growth factor receptors in
to augmented myocardial CCL-2/MCP-1 expression, and later border zone vasculature and in new vessels extending into the
recruitment of Ly6CloCCR2− CX3CR1hi monocytes in response infarct zone.3,184 Initially, the neovessels are enlarged and lack
to augmented myocardial expression of fractalkine, the ligand a pericyte and smooth muscle cell mural coating; these fea-
for CX3CR1. However, more recent work173 using chimeric tures promote vessel hyperpermeability and inflammatory cell
mice deficient for hematopoietic cell Nr4a1 (an orphan nucle- extravasation into the infarct tissue.185,186 During later phases
ar hormone receptor essential for patrolling Ly6Clo monocyte of infarct healing, however, these neovessels mature and be-
development176) indicated that both phases derive from proin- come invested with a mural coat. This process is dependent
flammatory Ly6Chi monocytes, and that during the reparative on endothelial cell platelet-derived growth factor (PDGF) and
phase, recruited Ly6Chi monocytes switch their phenotype to PDGF receptor-β signaling in pericytes and smooth muscle
Ly-6Clo anti-inflammatory macrophages that proliferate lo- cells.186 Defects in the formation of the mural coat result in
cally to effect inflammation resolution and wound healing. prolonged inflammatory cell infiltration in the infarct zone
Although molecular regulators such as Nr4a1173 and interfer- and reduced collagen deposition, suggesting that PDGF-β–
on-regulatory factor 5172 may serve as important modulators mediated pericyte investment of neovessels is critical for the
of inflammatory vis-à-vis reparative polarity in macrophages, proper resolution of inflammation post MI. The importance
the specific microenvironmental cues that induce this switch of pericytes is further supported by the findings that in vivo
remain poorly defined. human pericyte transplantation into the peri-infarct zone of
In addition to Mo/Mϕ, CD11c+ DCs infiltrate the infarct- mice attenuates vascular permeability, reduces tissue leuko-
ed heart, predominantly during the reparative phase.41,177 DCs cyte infiltration, augments angiogenesis, and improves cardiac
are essential for proper inflammation resolution, scar forma- remodeling.187,188
tion, and angiogenesis post-MI, as DC ablation resulted in Cardiomyocytes: It is tempting to hypothesize that vi-
persistent cardiac accumulation of Ly6Chigh monocytes and able border zone cardiomyocytes may secrete mediators
CD206− macrophages, sustained proinflammatory cytokine that limit extension of the inflammatory response, protecting
expression, reduced endothelial cell proliferation, and dete- noninfarcted myocardium from the unwanted effects of unre-
rioration of LV function post-MI.177 strained inflammation.3,189 However, information on specific
T-lymphocytes: T-lymphocytes, including CD4+ and CD8+ cardiomyocyte-derived signals that may contribute to con-
T-cells, Foxp3+ regulatory cells, invariant natural killer T tainment of inflammation after infarction remains limited.
cells, and γδT-cells, infiltrate the heart after MI, most robustly Recent evidence suggested that, in the infarcted myocardium,
during the reparative phase.41,178–182 Studies using CD4+ T-cell– cardiomyocytes may secrete regenerating islet-derived-3β, a
deficient mice, and OT-II mice that exhibit defective T-cell an- mediator that regulates macrophage recruitment and inhibits
tigen recognition, have demonstrated that CD4+ helper T cells inflammatory activation, preventing cardiac rupture and ex-
are activated after MI likely in response to released cardiac pansion of injury.190
autoantigens, and that these T cells promote wound healing, Molecular Signals Implicated in Resolution of
resolution of inflammation and proinflammatory monocyte Postinfarction Inflammation
infiltration, and proper collagen matrix formation and scar Timely suppression and spatial containment of the postinfarc-
formation, thereby limiting adverse remodeling.178 However, tion inflammatory reaction is dependent on release of secreted
the specific identity of these autoantigens remain unclear. anti-inflammatory mediators (such as IL-10, members of the
Notably, in a hindlimb ischemia model, CD4+ T cells were TGF-β family, and lipid-derived proresolving mediators) and
also shown to be essential for the recruitment of proangiogenic on activation of intracellular STOP signals that inhibit the in-
macrophages and collateral artery formation.183 A recent study nate immune response. Defects in the molecular pathways
using both genetic and antibody-based approaches to modu- responsible for suppression and resolution of the inflamma-
late regulatory T cells early post MI showed that CD4+Foxp3+ tory response may be involved in the pathogenesis of adverse
regulatory T cells are also essential for favorable wound heal- remodeling and HF after MI.191
ing, scar formation, and inflammation resolution after MI, in IL-10: IL-10 exerts potent anti-inflammatory actions,
part, by modulating macrophage differentiation toward an suppressing synthesis of proinflammatory cytokines and che-
M2-like phenotype.181 Moreover, the activation of invariant mokines in macrophages192 through the activation of signal
natural killer cells after reperfused179 or nonreperfused MI180 transducer and activator of transcription 3 signaling.193 IL-10
has been shown to reduce leukocyte infiltration, myocardial upregulation has been documented in both rodent and large
injury, and adverse remodeling, in part, by enhancing the ex- animal models of MI.154,194 Although the late timing of IL-10
pression of anti-inflammatory cytokines such as IL-10. Hence, upregulation is consistent with a possible role in suppres-
multiple T-lymphocyte subsets contribute to suppression of sion of proinflammatory signaling, experiments using IL-10

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


102  Circulation Research  June 24, 2016

null animals have produced conflicting results. In a mouse and may play an important role in resolution of the inflam-
model of coronary occlusion/reperfusion, genetic loss of matory infiltrate after infarction. Protective effects of ex-
IL-10 was associated with increased early mortality and ac- ogenous resolvin E1 and resolvin D1 administration have
centuated expression of proinflammatory genes.195 However, been demonstrated in rodent models of I/R and nonreper-
another study with a much higher sample size did not con- fused MI205,206; however, the potential role of endogenous
firm these observations, demonstrating comparable mortal- proresolving lipid mediators in suppression and resolution
ity and dilative remodeling in IL-10 null and WT animals of the postinfarction inflammatory response has not been
undergoing reperfused infarction protocols, and suggesting investigated.
that IL-10 loss is associated with relatively subtle alterations
Do Chemokine-Mediated Effects Suppress Inflammation by
(including increased myocardial levels of TNF and MCP-1).196 Recruiting Anti-Inflammatory Leukocytes?
Interestingly, in human patients, high plasma levels of IL-10 Although traditionally viewed as proinflammatory media-
predict adverse outcome in patients with acute coronary syn- tors, certain members of the chemokine family may suppress
dromes197 and ST-segment–elevation MI.198 This observation inflammation by recruiting anti-inflammatory monocyte and
may reflect a compensatory accentuation of anti-inflammatory lymphocyte subsets to the infarcted myocardium. In a mouse
cytokine synthesis in high-risk patients. model of reperfused MI, genetic disruption of the CC che-
Members of the TGF-β family: Several members of the mokine receptor CCR5 caused enhanced inflammation and
TGF-β family have been implicated in negative regulation of accentuated dilative remodeling, associated with decreased
the inflammatory reaction. Unfortunately, dissection of the infiltration of the infarct by regulatory T cells.123 Specific
role of these mediators in postinfarction inflammation and chemokine–chemokine receptor pairs may mediate recruit-
repair has been hampered by the complex biology of their ment of anti-inflammatory monocyte and lymphocyte subsets,
regulation and activation, by their pleiotropic and context- thus protecting the infarcted myocardium from unrestrained
dependent actions on all cell types involved in infarct heal- inflammation.
ing, and by the complexity of their downstream signaling
effectors. Considering its actions on immune and reparative Intracellular Pathways Involved in Negative Regulation of
cells and the time course of its upregulation after MI, TGF- the Inflammatory Cascade
β1 may serve as the master switch regulating the transition Activation of intracellular pathways that restrain the innate
from inflammation to fibrosis.199 Neutralization experiments immune response may also play a crucial role in timely sup-
using gene therapy with the extracellular domain of the type pression of the postinfarction inflammatory response, protect-
II TGF-β receptor in a model of MI suggested that early inhi- ing from adverse remodeling. Expression of IL-1R–associated
kinase-M, a negative regulator of the innate immune response,
bition may worsen dysfunction accentuating the inflammato-
is upregulated in infarct macrophages and fibroblasts and
ry response, whereas late disruption of TGF-β signaling may
inhibits macrophage-derived cytokine expression, while
protect from interstitial fibrosis and hypertrophic remodel-
promoting a matrix-preserving myofibroblast phenotype in
ing.200 A recent study suggested that, although broad inhibi-
cardiac fibroblasts.207,208
tion of TGF-β after infarction causes early mortality caused
by cardiac rupture, cardiomyocyte-specific disruption of the Fibroblast Activation and Formation of the Scar
TGF-β receptors was protective and stimulated a wide range
Myofibroblast Transdifferentiation and Acquisition of a
of anti-inflammatory and cytoprotective signals.201 Thus, Synthetic Phenotype
the detrimental actions of early TGF-β inhibition on the in- Expansion of the cardiac fibroblast population and conversion
farcted myocardium may not be because of direct actions on into synthetic myofibroblasts are hallmarks of the prolifera-
cardiomyocyte survival, but may reflect loss of anti-inflam- tive phase of cardiac repair.209–211 Myofibroblasts are pheno-
matory actions on inflammatory cells, endothelial cells, or typically modulated fibroblasts that develop stress fibers and
fibroblasts. express contractile proteins, such as α-smooth muscle actin
Growth differentiation factor-15, a member of the TGF- and the embryonal isoform of smooth muscle myosin.210,212
β family, is also implicated in suppression of the inflam- The origin of infarct myofibroblasts remains a debated is-
matory response after infarction. Growth differentiation sue. Experimental studies using bone marrow transplantation
factor-15 exerts potent anti-inflammatory actions by coun- strategies have produced conflicting results, suggesting that
teracting chemokine-triggered leukocyte integrin activation. either resident fibroblasts213 or circulating bone marrow pro-
Thus, growth differentiation factor-15 loss in mice is associ- genitors214 may be the main source of myofibroblasts in the
ated with accentuated postinfarction inflammation and fatal infarct. Endothelial cells undergoing mesenchymal transdif-
cardiac rupture after MI.202 In patients with ST-segment– ferentiation,215 epicardial epithelial cells, and pericytes may
elevation MI, elevated plasma growth differentiation fac- represent additional sources of myofibroblasts in the healing
tor-15 levels are associated with increased mortality,203 infarct.216 Recent studies using lineage tracing approaches
likely reflecting activation of a protective anti-inflammatory in a model of MI have demonstrated that epicardial-derived
pathway in patients with an accentuated postinfarction in- cells that colonize the adult mammalian cardiac interstitium
flammatory reaction. massively transdifferentiate into myofibroblasts in the in-
Lipid-derived proresolving mediators: Proresolving lip- farcted heart.217 Thus, after MI, interstitial fibroblasts that sur-
id mediators (including the lipoxins, resolvins, protectins, vive the ischemic insult, or cells recruited from neighboring
and maresins)204 have potent anti-inflammatory properties viable areas, may undergo myofibroblast transdifferentiation,

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   103

in response to increased levels of bioactive TGF-β and to the mediators, such as thePDGFs,235,236 members of the fibroblast
changes in the composition of the surrounding extracellular growth factor family,237 and the mast cell–derived proteases
matrix. Moreover, in the healing infarct, marked induction of tryptase238 and chymase239 are also released in the infarcted
chemokines in response to extensive cardiomyocyte necrosis heart and may activate infarct myofibroblasts.
may result in recruitment and activation of additional subsets
of reparative fibroblasts that may play an important role in Extracellular Matrix During the Proliferative Phase
scar formation. of Infarct Healing
The proliferative phase of infarct healing is characterized by
Mediators Involved in Myofibroblast Activation dynamic alterations in the extracellular matrix network that di-
Conversion of fibroblasts into myofibroblasts requires the rectly regulate cellular phenotype and activity. As matrix frag-
cooperation of both soluble mediators and specialized ma- ments in the infarct zone are phagocytosed, a provisional matrix
trix components. TGF-β is induced and activated in the is formed, composed predominantly of fibrin and fibronectin.240
infarcted myocardium and is critically involved in myofi- This highly plastic matrix network serves as a scaffold for mi-
broblast transdifferentiation. In vitro studies have suggest- grating and proliferating cells, facilitating the dynamic changes
ed that TGF-β1–induced myofibroblast conversion may be that occur in the healing wound. The infarct matrix is also en-
mediated through both canonical Smad-dependent218 and riched through deposition of several members of the matricellu-
Smad-independent signaling pathways.219,220 Modulation of lar protein family. These structurally unrelated macromolecules
the extracellular matrix also contributes to myofibroblast are not present in normal myocardium, but are markedly upreg-
conversion. TGF-β–mediated myofibroblast transdifferen- ulated in the infarcted and remodeling heart. Unlike structural
tiation requires activation of an outside-in signaling pathway matrix components (such as collagens and elastin), matricel-
transduced by polymerized fibronectin-extra domain A.53,221 lular proteins do not provide mechanical support, but may bind
Moreover, secretion and deposition of matricellular proteins, to matrix proteins and cell receptors transducing signaling
such as thrombospondin-1 and osteopontinfamily, may con- cascades. Thrombospondin-1,241 tenascin-C,242 secreted pro-
tribute to myofibroblast conversion, both by exerting direct tein acidic and cysteine-rich,243 periostin,244,245 osteopontin,246
actions on cellular phenotype and by accentuating growth osteoglycin,247 and members of the CCN family248 have been
factor-mediated responses.222–224 implicated in regulation of the inflammatory and reparative re-
A growing body of evidence suggests that members of sponse after MI. Because matricellular proteins are capable of
the transient receptor potential (TRP) family of ion channels modulating behavior and function of all cells involved in car-
play an essential role in regulating fibroblast to myofibroblast diac repair, remodeling and fibrosis, dissection of specific mo-
transition.225 TRP channels are ubiquitously expressed in all lecular mechanisms responsible for the observed in vivo effects
cell types, providing calcium entry pathways and regulating is particularly challenging. Modulation of growth factor activity
a wide range of Ca2+-dependent cell functions.226 Although and signaling241,243 or regulation of collagen fibrillogenesis and
in vitro studies have implicated TRPV4227 and TRPM7228 maturation246,247 has been suggested as potential mechanisms
channels in cardiac myofibroblast transdifferentiation, their for specific matricellular actions.
in vivo role is unclear. On the contrary, both in vitro and in
vivo studies documented a critical role for TRPC6 in infarct Maturation Phase
myofibroblast conversion, demonstrating that TRPC6 absence The proliferative phase of cardiac repair is followed by scar
is associated with impaired fibroblast function and increased maturation, as the extracellular matrix becomes cross-linked,
mortality caused by cardiac rupture.229 and reparative cells are deactivated, and may undergo apoptosis.
Acquisition of a myofibroblast phenotype by infarct fi- The molecular signals implicated in quiescence of infarct myo-
broblasts is associated with increased proliferative activity fibroblasts remain unknown. Withdrawal of fibrogenic growth
and stimulation of a matrix-preserving program. Extensive factors, activation of inhibitory STOP signals that terminate
experimental evidence suggests that the activation of the re- TGF-β and angiotensin II signaling, and clearance of matricel-
nin–angiotensin–aldosterone system promotes myofibroblast lular proteins, may suppress proliferation and reduce the matrix-
proliferation and stimulates matrix synthesis. In addition to synthetic activity of the fibroblasts. Induction and secretion of
the well-described effects of circulating angiotensin II and al- antifibrotic mediators may also contribute to termination of the
dosterone, local generation of angiotensin II in the infarct has matrix-synthetic response. The CXC chemokine interferon-
also been implicated in fibroblast activation.230 Angiotensin γ–inducible protein-10/CXCL10 inhibits fibroblast migration
II accentuates proliferative activity in cardiac fibroblasts and through proteoglycan-mediated actions, is upregulated in the in-
induces synthesis of extracellular matrix proteins and inte- farcted heart, and contributes to spatial containment of the fibro-
grins,231 through effects that predominantly involve the AT1 genic response in the infarcted region.249,250 However, because
receptor.232 Aldosterone also promotes cardiac fibroblast of the early timing of its induction in the infarcted myocardium,
proliferation by stimulating Kirsten Ras-A (Ki-RasA) and interferon-γ–inducible protein-10 is an unlikely candidate for
MAPK1/2 signaling.233 Some actions of the renin–angioten- a role in scar maturation. Although reduction in myofibroblast
sin–aldosterone system in cardiac fibroblasts may be medi- density during infarct maturation has been well document-
ated through activation of the TGF-β cascade. In addition to ed,210,251 the fate of these cells remains unknown. Apoptotic death
its critical effects on myofibroblast conversion, TGF-β also may mediate fibroblast loss in the scar and in the infarct border
activates a matrix-preserving program in cardiac fibroblasts zone;252 the mediators responsible for fibroblast-specific activa-
through Smad-dependent signaling.218,234 Other fibrogenic tion of proapoptotic signaling are unknown.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


104  Circulation Research  June 24, 2016

Inflammatory Cells in the Late Phase facilitation of active resolution may prevent persistent inflam-
of Post-MI Cardiac Remodeling mation and ameliorate late HF.
As detailed above and summarized in Figure 5, the 2 phases of Alternatively, chronic inflammation, and tissue inflam-
healing after MI are acute inflammation with intense cellular matory cell infiltration, in ischemic HF may represent late
infiltration, lasting up to 4 days in mice, followed by resolu- recrudescence of immune activation toward the heart in re-
tion and repair with active resolution of inflammation, quies- sponse to as-of-yet poorly identified factors or antigens.256 In
cence of cell activity, and scar stabilization and maturation support of this idea, Ismahil et al257 recently demonstrated that
>14 days in mice. Although the time frame of these events is in chronic ischemic HF, there is local (and systemic) expan-
well defined in the murine model, larger animal models ex- sion of proinflammatory Mo/Mϕ, DCs, and T cells, together
hibit comparatively greater persistence of cellular infiltration with heightened splenic expression of alarmins and proinflam-
and slower formation of granulation tissue after MI.194 After matory mediators, and structural splenic remodeling consis-
healing, in both humans and animal models with MI, a subset tent with heightened antigen processing. Moreover, activated
of those afflicted will exhibit late progressive ventricular dila- mononuclear splenocytes trafficked to the failing heart to
tation and HF, a state characterized by chronic inflammation promote apoptosis, fibrosis, and dysfunction, suggesting that
(Figure 5).80 Among other variables, both larger infarcts253 and adverse LV remodeling was in part immune mediated, pos-
greater initial inflammatory activation254 are strong predic- sibly in response to cardiac-derived alarmins. The role of par-
tors of late cardiac remodeling and HF in humans. Moreover, ticular immune cell populations in the pathogenesis of chronic
histopathologic studies of human failing hearts with chronic remodeling and inflammation in HF requires further study,
ischemic cardiomyopathy have demonstrated augmented tis- along with their organ-specific derivation (eg, local prolifera-
sue macrophages and T-lymphocytes, and increased adhe- tion vis-à-vis infiltration from remote sites) and the potential
sion molecule expression in endothelial cells.255 One potential antigens and molecular pathways responsible for their activa-
explanation for these findings is that late cardiac remodeling tion. As discussed above, these mechanisms may be separate
may be driven, in part, by incomplete or impaired resolution and distinct from those that promote myocardial salvage after
of myocardial inflammation with larger degrees of injury acute MI.
post-MI that amplifies over time. Indeed, the exogenous ad-
ministration of the proresolving lipid mediator resolvin D1 Targeting the Inflammatory Response in MI
after nonreperfused MI hastened inflammation resolution and Over the past 30 years, experimental work has revealed
improved post-MI ventricular remodeling,206 suggesting that a crucial role for the inflammatory cascade in cardiac re-
pair, remodeling, and fibrosis after MI. Unfortunately, this
new knowledge has not yet translated into effective therapy.
Because of the close link between inflammation and repair,
nonselective inhibition of inflammation after MI (using corti-
costeroids or nonsteroidal anti-inflammatory drugs) may have
detrimental effects on scar formation, promoting rupture and
accentuating adverse remodeling.258 Thus, selective targeting
of injurious proinflammatory signals is needed. Unfortunately,
in clinical studies, approaches targeting several specific pro-
inflammatory pathways have produced disappointing results.
Despite promising experimental data in large animal models,
CD11/CD18 integrin inhibition failed to reduce the size of the
infarct in patients with MI.259 In a large clinical trial, comple-
ment inhibition did not reduce mortality and major adverse
events in patients with ST-segment–elevation MI.260 P-selectin
inhibition in patients with acute coronary syndromes seemed
to attenuate cardiomyocyte necrosis, but was associated with
trends toward worse clinical outcome.261,262 What are the
causes for these translational failures?
Figure 5. Early and late inflammation after myocardial First, inflammatory mediators are notoriously pleiotropic,
infarction (MI). The early inflammatory phase after MI (≈4 d in exerting a wide range of actions on many different cell types.
mice) is characterized by robust innate and adaptive immune
cell infiltration and tissue digestion. This is subsequently Thus, targeting a specific cytokine, growth factor, or inflam-
followed a phase of resolution, myofibroblast proliferation, and matory pathway may modulate several cellular responses;
wound repair (lasting ≈10–14 d in mice), during which immune the clinical implications of these actions cannot always be
cells are polarized toward an anti-inflammatory state. However, predicted by investigating animal models. Second, temporal
larger infarcts with more pronounced inflammatory activation
exhibit progressive ventricular dilatation and heart failure (HF) and spatial considerations are critical determinants of the ef-
over the long term, together with persistent inflammation fectiveness of an intervention, as described above. The re-
and tissue immune cell infiltration. Chronic inflammation parative response after MI is a highly dynamic process; the
may represent incomplete inflammation resolution during the
reparative phase and subsequent amplification with time, or
window of therapeutic opportunity is brief. Moreover, con-
a second wave of resurgent immune activation in response to sidering the spatial heterogeneity of the cellular environment
poorly defined factors. in the remodeling infarcted heart, therapeutic interventions

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   105

may have distinct effects on the infarct, the border zone, and Sources of Funding
the remote remodeling myocardium. Thus, a successful strat- This study was supported by National Institutes of Health (NIH)
egy requires careful design, optimally exploiting knowledge R01 HL76246 and R01 HL85440 (to Dr Frangogiannis); NIH R01
on the time course and topographical characteristics of the HL125735 and VA Merit I01 BX002706 (to Dr Prabhu).
inflammatory response. Third, the pathophysiologic hetero-
geneity of postinfarction remodeling in humans complicates Disclosures
implementation of therapeutic strategies. The severity and None.
characteristics of the remodeling response after infarction are
not dependent only on the size of the acute infarct. Genetic References
background, concomitant conditions (such as hypertension 1. Prabhu SD. Post-infarction ventricular remodeling: an array of mo-
lecular events. J Mol Cell Cardiol. 2005;38:547–550. doi: 10.1016/j.
or diabetes mellitus), age, and sex have major impact on the yjmcc.2005.01.014.
inflammatory response and profoundly affect post-MI remod- 2. Sutton MG, Sharpe N. Left ventricular remodeling after myocardial in-
eling and clinical outcome. farction: pathophysiology and therapy. Circulation. 2000;101:2981–2988.
3. Frangogiannis NG. Regulation of the inflammatory response
The pathophysiologic heterogeneity of patients with MI
in cardiac repair. Circ Res. 2012;110:159–173. doi: 10.1161/
has important therapeutic implications. Certain patient sub- CIRCRESAHA.111.243162.
populations exhibit prolonged accentuation of proinflamma- 4. Nahrendorf M, Pittet MJ, Swirski FK. Monocytes: protagonists
tory signaling after infarction that may be associated with of infarct inflammation and repair after myocardial infarction.
Circulation. 2010;121:2437–2445. doi: 10.1161/CIRCULATIONAHA.
dilative remodeling and systolic dysfunction. These patients 109.916346.
may benefit from interventions targeting the inflammatory cas- 5. Kain V, Prabhu SD, Halade GV. Inflammation revisited: inflammation ver-
cade, such as IL-1 inhibition.263 Other patients do not exhibit sus resolution of inflammation following myocardial infarction. Basic Res
significant postinfarction ventricular dilation, but develop a Cardiol. 2014;109:444. doi: 10.1007/s00395-014-0444-7.
6. Panizzi P, Swirski FK, Figueiredo JL, Waterman P, Sosnovik DE, Aikawa
pronounced hypertrophic and fibrotic response, associated E, Libby P, Pittet M, Weissleder R, Nahrendorf M. Impaired infarct
with diastolic dysfunction. This type of adverse remodeling is healing in atherosclerotic mice with Ly-6C(hi) monocytosis. J Am Coll
particularly common in diabetics and may be associated with Cardiol. 2010;55:1629–1638. doi: 10.1016/j.jacc.2009.08.089.
7. Eltzschig HK, Eckle T. Ischemia and reperfusion–from mechanism to
an overactive TGF-β system.264–266 Identification of patient translation. Nat Med. 2011;17:1391–1401. doi: 10.1038/nm.2507.
subpopulations with distinct pathophysiologic perturbations, 8. Timmers L, Pasterkamp G, de Hoog VC, Arslan F, Appelman Y, de Kleijn
using suitable biomarkers or molecular imaging modalities, DP. The innate immune response in reperfused myocardium. Cardiovasc
is needed to design effective therapies to attenuate adverse re- Res. 2012;94:276–283. doi: 10.1093/cvr/cvs018.
9. Vandervelde S, van Amerongen MJ, Tio RA, Petersen AH, van Luyn MJ,
modeling in patients with MI. Harmsen MC. Increased inflammatory response and neovascularization in
The potential involvement of inflammatory signals in tis- reperfused vs. non-reperfused murine myocardial infarction. Cardiovasc
sue regeneration adds a new perspective to the therapeutic po- Pathol. 2006;15:83–90. doi: 10.1016/j.carpath.2005.10.006.
10. Arslan F, de Kleijn DP, Pasterkamp G. Innate immune signaling in

tential of strategies targeting the postinfarction inflammatory
cardiac ischemia. Nat Rev Cardiol. 2011;8:292–300. doi: 10.1038/
response. Recently, neonatal macrophage subsets have been nrcardio.2011.38.
suggested to activate a regenerative program in the infarcted 11. de Haan JJ, Smeets MB, Pasterkamp G, Arslan F. Danger signals in
myocardium.267 Moreover, certain chemokines (such as stro- the initiation of the inflammatory response after myocardial infarc-
tion. Mediators Inflamm. 2013;2013:206039. doi: 10.1155/2013/
mal cell–derived factor-1/CXCL12), cytokines, and growth 206039.
factors268–270 may regulate trafficking, activation, differentia- 12. Ghigo A, Franco I, Morello F, Hirsch E. Myocyte signalling in leuco-
tion, and survival of progenitor cells. Clearly, this field is in cyte recruitment to the heart. Cardiovasc Res. 2014;102:270–280. doi:
its infancy. Whether modulation of the inflammatory response 10.1093/cvr/cvu030.
13. Mann DL. The emerging role of innate immunity in the heart and vascu-
can contribute to activation of a regenerative program in adult lar system: for whom the cell tolls. Circ Res. 2011;108:1133–1145. doi:
mammalian myocardium is unknown. 10.1161/CIRCRESAHA.110.226936.
14. Newton K, Dixit VM. Signaling in innate immunity and inflammation.
Cold Spring Harb Perspect Biol. 2012;4:. doi: 10.1101/cshperspect.
Concluding Remarks a006049.
Our growing knowledge on the role of inflammatory cas- 15. Andrassy M, Volz HC, Igwe JC, et al. High-mobility group box-1 in isch-
cades in injury, repair, and remodeling of the infarcted heart emia-reperfusion injury of the heart. Circulation. 2008;117:3216–3226.
has revealed new therapeutic targets for patients surviving doi: 10.1161/CIRCULATIONAHA.108.769331.
16. Gwechenberger M, Mendoza LH, Youker KA, Frangogiannis NG, Smith
acute MI. Unfortunately, the pleiotropic actions of inflamma- CW, Michael LH, Entman ML. Cardiac myocytes produce interleukin-6
tory mediators, and the remarkable pathophysiologic hetero- in culture and in viable border zone of reperfused infarctions. Circulation.
geneity of cardiac remodeling in human patients, pose major 1999;99:546–551.
17. Yamauchi-Takihara K, Ihara Y, Ogata A, Yoshizaki K, Azuma J,

challenges for clinical implementation of strategies targeting Kishimoto T. Hypoxic stress induces cardiac myocyte-derived interleu-
the inflammatory response. A concerted effort is needed to kin-6. Circulation. 1995;91:1520–1524.
dissect the cellular actions and molecular signals activated 18. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832.
by inflammatory mediators, and to identify the pathophysi- doi: 10.1016/j.cell.2010.01.040.
19. Xie J, Méndez JD, Méndez-Valenzuela V, Aguilar-Hernández MM. Cellular
ologic perturbations that may be responsible for adverse re- signalling of the receptor for advanced glycation end products (RAGE).
modeling in vulnerable patients. Strategies modulating the Cell Signal. 2013;25:2185–2197. doi: 10.1016/j.cellsig.2013.06.013.
inflammatory cascade may not only hold promise as protec- 20. Kobayashi Y. Neutrophil infiltration and chemokines. Crit Rev Immunol.
2006;26:307–316.
tive measures to prevent remodeling and HF but also may
21. White GE, Iqbal AJ, Greaves DR. CC chemokine receptors and chronic
prove crucial in realizing the visionary goal of myocardial inflammation–therapeutic opportunities and pharmacological challenges.
regeneration.271 Pharmacol Rev. 2013;65:47–89. doi: 10.1124/pr.111.005074.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


106  Circulation Research  June 24, 2016

22. Nian M, Lee P, Khaper N, Liu P. Inflammatory cytokines and post- 40. Hu X, Jiang H, Cui B, Xu C, Lu Z, He B. Preconditioning with high
myocardial infarction remodeling. Circ Res. 2004;94:1543–1553. doi: mobility group box 1 protein protects against myocardial ischemia-
10.1161/01.RES.0000130526.20854.fa. reperfusion injury. Int J Cardiol. 2010;145:111–112. doi: 10.1016/j.
23. Wan E, Yeap XY, Dehn S, et al. Enhanced efferocytosis of apop- ijcard.2009.05.057.
totic cardiomyocytes through myeloid-epithelial-reproductive ty- 41. Yan X, Anzai A, Katsumata Y, Matsuhashi T, Ito K, Endo J, Yamamoto
rosine kinase links acute inflammation resolution to cardiac repair T, Takeshima A, Shinmura K, Shen W, Fukuda K, Sano M. Temporal
after infarction. Circ Res. 2013;113:1004–1012. doi: 10.1161/ dynamics of cardiac immune cell accumulation following acute myo-
CIRCRESAHA.113.301198. cardial infarction. J Mol Cell Cardiol. 2013;62:24–35. doi: 10.1016/j.
24. Bianchi ME, Beltrame M. Upwardly mobile proteins. Workshop: the role yjmcc.2013.04.023.
of HMG proteins in chromatin structure, gene expression and neoplasia. 42. Foell D, Wittkowski H, Vogl T, Roth J. S100 proteins expressed in phago-
EMBO Rep. 2000;1:109–114. doi: 10.1093/embo-reports/kvd030. cytes: a novel group of damage-associated molecular pattern molecules. J
25. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 Leukoc Biol. 2007;81:28–37. doi: 10.1189/jlb.0306170.
by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 43. Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA,
10.1038/nature00858. Nacken W, Foell D, van der Poll T, Sorg C, Roth J. Mrp8 and Mrp14 are
26. Wang H, Bloom O, Zhang M, et al. HMG-1 as a late mediator of endotoxin endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-
lethality in mice. Science. 1999;285:248–251. induced shock. Nat Med. 2007;13:1042–1049. doi: 10.1038/nm1638.
27. Loukili N, Rosenblatt-Velin N, Li J, Clerc S, Pacher P, Feihl F, Waeber 44. Hofmann MA, Drury S, Fu C, et al. RAGE mediates a novel proinflamma-
B, Liaudet L. Peroxynitrite induces HMGB1 release by cardiac cells in tory axis: a central cell surface receptor for S100/calgranulin polypeptides.
vitro and HMGB1 upregulation in the infarcted myocardium in vivo. Cell. 1999;97:889–901.
Cardiovasc Res. 2011;89:586–594. doi: 10.1093/cvr/cvq373. 45. Katashima T, Naruko T, Terasaki F, Fujita M, Otsuka K, Murakami S, Sato
28. Park JS, Svetkauskaite D, He Q, Kim JY, Strassheim D, Ishizaka A, A, Hiroe M, Ikura Y, Ueda M, Ikemoto M, Kitaura Y. Enhanced expression
Abraham E. Involvement of toll-like receptors 2 and 4 in cellular activa- of the S100A8/A9 complex in acute myocardial infarction patients. Circ J.
tion by high mobility group box 1 protein. J Biol Chem. 2004;279:7370– 2010;74:741–748.
7377. doi: 10.1074/jbc.M306793200. 46. Morrow DA, Wang Y, Croce K, Sakuma M, Sabatine MS, Gao H, Pradhan
29. Tian J, Avalos AM, Mao SY, et al. Toll-like receptor 9-dependent activa- AD, Healy AM, Buros J, McCabe CH, Libby P, Cannon CP, Braunwald
tion by DNA-containing immune complexes is mediated by HMGB1 and E, Simon DI. Myeloid-related protein 8/14 and the risk of cardiovascu-
RAGE. Nat Immunol. 2007;8:487–496. doi: 10.1038/ni1457. lar death or myocardial infarction after an acute coronary syndrome
30. Schiraldi M, Raucci A, Muñoz LM, et al. HMGB1 promotes recruitment in the Pravastatin or Atorvastatin Evaluation and Infection Therapy:
of inflammatory cells to damaged tissues by forming a complex with Thrombolysis in Myocardial Infarction (PROVE IT-TIMI 22) trial. Am
CXCL12 and signaling via CXCR4. J Exp Med. 2012;209:551–563. doi: Heart J. 2008;155:49–55. doi: 10.1016/j.ahj.2007.08.018.
10.1084/jem.20111739. 47. Volz HC, Laohachewin D, Seidel C, Lasitschka F, Keilbach K, Wienbrandt
31. Cirillo P, Giallauria F, Pacileo M, Petrillo G, D’Agostino M, Vigorito C, AR, Andrassy J, Bierhaus A, Kaya Z, Katus HA, Andrassy M. S100A8/
Chiariello M. Increased high mobility group box-1 protein levels are as- A9 aggravates post-ischemic heart failure through activation of RAGE-
sociated with impaired cardiopulmonary and echocardiographic findings dependent NF-κB signaling. Basic Res Cardiol. 2012;107:250. doi:
after acute myocardial infarction. J Card Fail. 2009;15:362–367. doi: 10.1007/s00395-012-0250-z.
10.1016/j.cardfail.2008.11.010. 48. Rohde D, Schön C, Boerries M, et al. S100A1 is released from ischemic
32. Sørensen MV, Pedersen S, Møgelvang R, Skov-Jensen J, Flyvbjerg
cardiomyocytes and signals myocardial damage via Toll-like receptor 4.
A. Plasma high-mobility group box 1 levels predict mortality after ST- EMBO Mol Med. 2014;6:778–794. doi: 10.15252/emmm.201303498.
segment elevation myocardial infarction. JACC Cardiovasc Interv. 49. Seropian IM, Cerliani JP, Toldo S, et al. Galectin-1 controls cardiac in-
2011;4:281–286. doi: 10.1016/j.jcin.2010.10.015. flammation and ventricular remodeling during acute myocardial infarc-
33. Kohno T, Anzai T, Naito K, Miyasho T, Okamoto M, Yokota H, Yamada tion. Am J Pathol. 2013;182:29–40. doi: 10.1016/j.ajpath.2012.09.022.
S, Maekawa Y, Takahashi T, Yoshikawa T, Ishizaka A, Ogawa S. Role 50. Reddy VS, Harskamp RE, van Ginkel MW, Calhoon J, Baisden CE, Kim
of high-mobility group box 1 protein in post-infarction healing process IS, Valente AJ, Chandrasekar B. Interleukin-18 stimulates fibronectin ex-
and left ventricular remodelling. Cardiovasc Res. 2009;81:565–573. doi: pression in primary human cardiac fibroblasts via PI3K-Akt-dependent
10.1093/cvr/cvn291. NF-kappaB activation. J Cell Physiol. 2008;215:697–707. doi: 10.1002/
34. Xu H, Yao Y, Su Z, Yang Y, Kao R, Martin CM, Rui T. Endogenous jcp.21348.
HMGB1 contributes to ischemia-reperfusion-induced myocar- 51. Gondokaryono SP, Ushio H, Niyonsaba F, Hara M, Takenaka H,

dial apoptosis by potentiating the effect of TNF-α/JNK. Am J Jayawardana ST, Ikeda S, Okumura K, Ogawa H. The extra domain A of
Physiol Heart Circ Physiol. 2011;300:H913–H921. doi: 10.1152/ fibronectin stimulates murine mast cells via toll-like receptor 4. J Leukoc
ajpheart.00703.2010. Biol. 2007;82:657–665. doi: 10.1189/jlb.1206730.
35. Limana F, Germani A, Zacheo A, Kajstura J, Di Carlo A, Borsellino 52. Schoneveld AH, Hoefer I, Sluijter JP, Laman JD, de Kleijn DP,

G, Leoni O, Palumbo R, Battistini L, Rastaldo R, Müller S, Pompilio Pasterkamp G. Atherosclerotic lesion development and Toll like receptor
G, Anversa P, Bianchi ME, Capogrossi MC. Exogenous high-mobility 2 and 4 responsiveness. Atherosclerosis. 2008;197:95–104. doi: 10.1016/j.
group box 1 protein induces myocardial regeneration after infarction via atherosclerosis.2007.08.004.
enhanced cardiac C-kit+ cell proliferation and differentiation. Circ Res. 53. Arslan F, Smeets MB, Riem Vis PW, Karper JC, Quax PH, Bongartz LG,
2005;97:e73–e83. doi: 10.1161/01.RES.0000186276.06104.04. Peters JH, Hoefer IE, Doevendans PA, Pasterkamp G, de Kleijn DP. Lack
36. Takahashi K, Fukushima S, Yamahara K, Yashiro K, Shintani Y, Coppen of fibronectin-EDA promotes survival and prevents adverse remodeling
SR, Salem HK, Brouilette SW, Yacoub MH, Suzuki K. Modulated inflam- and heart function deterioration after myocardial infarction. Circ Res.
mation by injection of high-mobility group box 1 recovers post-infarction 2011;108:582–592. doi: 10.1161/CIRCRESAHA.110.224428.
chronically failing heart. Circulation. 2008;118:S106–S114. doi: 10.1161/ 54. Lugrin J, Parapanov R, Rosenblatt-Velin N, Rignault-Clerc S, Feihl F,
CIRCULATIONAHA.107.757443. Waeber B, Müller O, Vergely C, Zeller M, Tardivel A, Schneider P, Pacher
37. Limana F, Esposito G, D’Arcangelo D, Di Carlo A, Romani S, Melillo P, Liaudet L. Cutting edge: IL-1α is a crucial danger signal triggering
G, Mangoni A, Bertolami C, Pompilio G, Germani A, Capogrossi MC. acute myocardial inflammation during myocardial infarction. J Immunol.
HMGB1 attenuates cardiac remodelling in the failing heart via enhanced 2015;194:499–503. doi: 10.4049/jimmunol.1401948.
cardiac regeneration and miR-206-mediated inhibition of TIMP-3. PLoS 55. Methe H, Kim JO, Kofler S, Weis M, Nabauer M, Koglin J. Expansion of
One. 2011;6:e19845. doi: 10.1371/journal.pone.0019845. circulating Toll-like receptor 4-positive monocytes in patients with acute
38. Kitahara T, Takeishi Y, Harada M, Niizeki T, Suzuki S, Sasaki T, Ishino M, coronary syndrome. Circulation. 2005;111:2654–2661. doi: 10.1161/
Bilim O, Nakajima O, Kubota I. High-mobility group box 1 restores car- CIRCULATIONAHA.104.498865.
diac function after myocardial infarction in transgenic mice. Cardiovasc 56. Satoh M, Shimoda Y, Maesawa C, Akatsu T, Ishikawa Y, Minami Y,
Res. 2008;80:40–46. doi: 10.1093/cvr/cvn163. Hiramori K, Nakamura M. Activated toll-like receptor 4 in monocytes
39. Nakamura Y, Suzuki S, Shimizu T, Miyata M, Shishido T, Ikeda K, Saitoh is associated with heart failure after acute myocardial infarction. Int J
S, Kubota I, Takeishi Y. High mobility group box 1 promotes angiogen- Cardiol. 2006;109:226–234. doi: 10.1016/j.ijcard.2005.06.023.
esis from bone marrow-derived endothelial progenitor cells after myocar- 57. van der Pouw Kraan TC, Bernink FJ, Yildirim C, Koolwijk P, Baggen
dial infarction. J Atheroscler Thromb. 2015;22:570–581. doi: 10.5551/ JM, Timmers L, Beek AM, Diamant M, Chen WJ, van Rossum AC, van
jat.27235. Royen N, Horrevoets AJ, Appelman YE. Systemic toll-like receptor and

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   107

interleukin-18 pathway activation in patients with acute ST elevation myo- ischaemia-reperfusion in the mouse. Clin Sci (Lond). 2015;129:187–198.
cardial infarction. J Mol Cell Cardiol. 2014;67:94–102. doi: 10.1016/j. doi: 10.1042/CS20140444.
yjmcc.2013.12.021. 74. Ha T, Hu Y, Liu L, Lu C, McMullen JR, Kelley J, Kao RL, Williams DL,
58. Selejan S, Pöss J, Walter F, Hohl M, Kaiser R, Kazakov A, Böhm M, Link Gao X, Li C. TLR2 ligands induce cardioprotection against ischaemia/
A. Ischaemia-induced up-regulation of Toll-like receptor 2 in circulating reperfusion injury through a PI3K/Akt-dependent mechanism. Cardiovasc
monocytes in cardiogenic shock. Eur Heart J. 2012;33:1085–1094. doi: Res. 2010;87:694–703. doi: 10.1093/cvr/cvq116.
10.1093/eurheartj/ehr377. 75. Dong JW, Vallejo JG, Tzeng HP, Thomas JA, Mann DL. Innate immu-
59. Timmers L, Sluijter JP, van Keulen JK, Hoefer IE, Nederhoff MG,
nity mediates myocardial preconditioning through Toll-like receptor 2 and
Goumans MJ, Doevendans PA, van Echteld CJ, Joles JA, Quax PH, TIRAP-dependent signaling pathways. Am J Physiol Heart Circ Physiol.
Piek JJ, Pasterkamp G, de Kleijn DP. Toll-like receptor 4 mediates mal- 2010;298:H1079–H1087. doi: 10.1152/ajpheart.00306.2009.
adaptive left ventricular remodeling and impairs cardiac function af- 76. Chao W. Toll-like receptor signaling: a critical modulator of cell sur-
ter myocardial infarction. Circ Res. 2008;102:257–264. doi: 10.1161/ vival and ischemic injury in the heart. Am J Physiol Heart Circ Physiol.
CIRCRESAHA.107.158220. 2009;296:H1–12. doi: 10.1152/ajpheart.00995.2008.
60. Frantz S, Kobzik L, Kim YD, Fukazawa R, Medzhitov R, Lee RT, Kelly 77. Ha T, Hua F, Liu X, Ma J, McMullen JR, Shioi T, Izumo S, Kelley J,
RA. Toll4 (TLR4) expression in cardiac myocytes in normal and failing Gao X, Browder W, Williams DL, Kao RL, Li C. Lipopolysaccharide-
myocardium. J Clin Invest. 1999;104:271–280. doi: 10.1172/JCI6709. induced myocardial protection against ischaemia/reperfusion injury is
61. Liu L, Wang Y, Cao ZY, Wang MM, Liu XM, Gao T, Hu QK, Yuan WJ and mediated through a PI3K/Akt-dependent mechanism. Cardiovasc Res.
Lin L. Up-regulated TLR4 in cardiomyocytes exacerbates heart failure af- 2008;78:546–553. doi: 10.1093/cvr/cvn037.
ter long-term myocardial infarction. J Cell Mol Med. 2015;19(12):2728– 78. Cao Z, Ren D, Ha T, Liu L, Wang X, Kalbfleisch J, Gao X, Kao R,
2740. doi: 10.1111/jcmm.12659. Williams D, Li C. CpG-ODN, the TLR9 agonist, attenuates myocar-
62. Oyama J, Blais C Jr, Liu X, Pu M, Kobzik L, Kelly RA, Bourcier T. dial ischemia/reperfusion injury: involving activation of PI3K/Akt
Reduced myocardial ischemia-reperfusion injury in toll-like recep- signaling. Biochim Biophys Acta. 2013;1832:96–104. doi: 10.1016/j.
tor 4-deficient mice. Circulation. 2004;109:784–789. doi: 10.1161/01. bbadis.2012.08.008.
CIR.0000112575.66565.84. 79. Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-κB in
63. Kim SC, Ghanem A, Stapel H, Tiemann K, Knuefermann P, Hoeft A, the heart: to be or not to NF-κB. Circ Res. 2011;108:1122–1132. doi:
Meyer R, Grohé C, Knowlton AA, Baumgarten G. Toll-like receptor 4 de- 10.1161/CIRCRESAHA.110.226928.
ficiency: smaller infarcts, but no gain in function. BMC Physiol. 2007;7:5. 80. Mann DL. Inflammatory mediators and the failing heart: past, present, and
doi: 10.1186/1472-6793-7-5. the foreseeable future. Circ Res. 2002;91:988–998.
64. Arslan F, Smeets MB, O’Neill LA, Keogh B, McGuirk P, Timmers L, 81. Davidson SM, Selvaraj P, He D, Boi-Doku C, Yellon RL, Vicencio

Tersteeg C, Hoefer IE, Doevendans PA, Pasterkamp G, de Kleijn DP. JM, Yellon DM. Remote ischaemic preconditioning involves signal-
Myocardial ischemia/reperfusion injury is mediated by leukocytic toll- ling through the SDF-1α/CXCR4 signalling axis. Basic Res Cardiol.
like receptor-2 and reduced by systemic administration of a novel anti- 2013;108:377. doi: 10.1007/s00395-013-0377-6.
toll-like receptor-2 antibody. Circulation. 2010;121:80–90. doi: 10.1161/ 82. Morimoto H, Hirose M, Takahashi M, Kawaguchi M, Ise H, Kolattukudy
CIRCULATIONAHA.109.880187. PE, Yamada M, Ikeda U. MCP-1 induces cardioprotection against isch-
65. Favre J, Musette P, Douin-Echinard V, Laude K, Henry JP, Arnal aemia/reperfusion injury: role of reactive oxygen species. Cardiovasc Res.
JF, Thuillez C, Richard V. Toll-like receptors 2-deficient mice are 2008;78:554–562. doi: 10.1093/cvr/cvn035.
protected against postischemic coronary endothelial dysfunction. 83. Mann DL. Innate immunity and the failing heart: the cytokine hy-

Arterioscler Thromb Vasc Biol. 2007;27:1064–1071. doi: 10.1161/ pothesis revisited. Circ Res. 2015;116:1254–1268. doi: 10.1161/
ATVBAHA.107.140723. CIRCRESAHA.116.302317.
66. Feng Y, Zhao H, Xu X, Buys ES, Raher MJ, Bopassa JC, Thibault H, 84. Sakata Y, Dong JW, Vallejo JG, Huang CH, Baker JS, Tracey KJ, Tacheuchi
Scherrer-Crosbie M, Schmidt U, Chao W. Innate immune adaptor MyD88 O, Akira S, Mann DL. Toll-like receptor 2 modulates left ventricular func-
mediates neutrophil recruitment and myocardial injury after ischemia- tion following ischemia-reperfusion injury. Am J Physiol Heart Circ
reperfusion in mice. Am J Physiol Heart Circ Physiol. 2008;295:H1311– Physiol. 2007;292:H503–H509. doi: 10.1152/ajpheart.00642.2006.
H1318. doi: 10.1152/ajpheart.00119.2008. 85. Tavener SA, Long EM, Robbins SM, McRae KM, Van Remmen H, Kubes
67. Lu C, Ren D, Wang X, Ha T, Liu L, Lee EJ, Hu J, Kalbfleisch J, Gao P. Immune cell Toll-like receptor 4 is required for cardiac myocyte impair-
X, Kao R, Williams D, Li C. Toll-like receptor 3 plays a role in myo- ment during endotoxemia. Circ Res. 2004;95:700–707. doi: 10.1161/01.
cardial infarction and ischemia/reperfusion injury. Biochim Biophys Acta. RES.0000144175.70140.8c.
2014;1842:22–31. doi: 10.1016/j.bbadis.2013.10.006. 86. Kawaguchi M, Takahashi M, Hata T, et al. Inflammasome activa-

68. Riad A, Jäger S, Sobirey M, Escher F, Yaulema-Riss A, Westermann tion of cardiac fibroblasts is essential for myocardial ischemia/re-
D, Karatas A, Heimesaat MM, Bereswill S, Dragun D, Pauschinger M, perfusion injury. Circulation. 2011;123:594–604. doi: 10.1161/
Schultheiss HP, Tschöpe C. Toll-like receptor-4 modulates survival by in- CIRCULATIONAHA.110.982777.
duction of left ventricular remodeling after myocardial infarction in mice. 87. Mezzaroma E, Toldo S, Farkas D, Seropian IM, Van Tassell BW, Salloum
J Immunol. 2008;180:6954–6961. FN, Kannan HR, Menna AC, Voelkel NF, Abbate A. The inflammasome
69. Shishido T, Nozaki N, Yamaguchi S, Shibata Y, Nitobe J, Miyamoto T, promotes adverse cardiac remodeling following acute myocardial infarc-
Takahashi H, Arimoto T, Maeda K, Yamakawa M, Takeuchi O, Akira S, tion in the mouse. Proc Natl Acad Sci U S A. 2011;108:19725–19730. doi:
Takeishi Y, Kubota I. Toll-like receptor-2 modulates ventricular remod- 10.1073/pnas.1108586108.
eling after myocardial infarction. Circulation. 2003;108:2905–2910. doi: 88. Sandanger Ø, Ranheim T, Vinge LE, et al. The NLRP3 inflammasome is
10.1161/01.CIR.0000101921.93016.1C. up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-
70. Singh MV, Swaminathan PD, Luczak ED, Kutschke W, Weiss RM,
reperfusion injury. Cardiovasc Res. 2013;99:164–174. doi: 10.1093/cvr/
Anderson ME. MyD88 mediated inflammatory signaling leads to CaMKII cvt091.
oxidation, cardiac hypertrophy and death after myocardial infarction. J 89. Chandrasekar B, Smith JB, Freeman GL. Ischemia-reperfusion of rat
Mol Cell Cardiol. 2012;52:1135–1144. myocardium activates nuclear factor-KappaB and induces neutrophil in-
71. Shimamoto A, Chong AJ, Yada M, Shomura S, Takayama H, Fleisig AJ, filtration via lipopolysaccharide-induced CXC chemokine. Circulation.
Agnew ML, Hampton CR, Rothnie CL, Spring DJ, Pohlman TH, Shimpo 2001;103:2296–2302.
H, Verrier ED. Inhibition of Toll-like receptor 4 with eritoran attenuates 90. Venkatachalam K, Prabhu SD, Reddy VS, Boylston WH, Valente AJ,
myocardial ischemia-reperfusion injury. Circulation. 2006;114:I270–I274. Chandrasekar B. Neutralization of interleukin-18 ameliorates ischemia/re-
72. Arslan F, Houtgraaf JH, Keogh B, Kazemi K, de Jong R, McCormack perfusion-induced myocardial injury. J Biol Chem. 2009;284:7853–7865.
WJ, O’Neill LA, McGuirk P, Timmers L, Smeets MB, Akeroyd L, Reilly doi: 10.1074/jbc.M808824200.
M, Pasterkamp G, de Kleijn DP. Treatment with OPN-305, a humanized 91. Chen LF, Greene WC. Shaping the nuclear action of NF-kappaB. Nat Rev
anti-Toll-Like receptor-2 antibody, reduces myocardial ischemia/reperfu- Mol Cell Biol. 2004;5:392–401. doi: 10.1038/nrm1368.
sion injury in pigs. Circ Cardiovasc Interv. 2012;5:279–287. doi: 10.1161/ 92. Hayden MS, Ghosh S. Signaling to NF-kappaB. Genes Dev. 2004;18:2195–
CIRCINTERVENTIONS.111.967596. 2224. doi: 10.1101/gad.1228704.
73. Parapanov R, Lugrin J, Rosenblatt-Velin N, Feihl F, Waeber B, Milano 93. Ritchie ME. Nuclear factor-kappaB is selectively and markedly

G, Vergely C, Li N, Pacher P, Liaudet L. Toll-like receptor 5 deficien- activated in humans with unstable angina pectoris. Circulation.
cy exacerbates cardiac injury and inflammation induced by myocardial 1998;98:1707–1713.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


108  Circulation Research  June 24, 2016

94. Chandrasekar B, Freeman GL. Induction of nuclear factor kappaB myocardium. Pharmacol Ther. 2014;143:305–315. doi: 10.1016/j.
and activation protein 1 in postischemic myocardium. FEBS Lett. pharmthera.2014.03.009.
1997;401:30–34. 113. Maekawa N, Wada H, Kanda T, Niwa T, Yamada Y, Saito K, Fujiwara
95. Hamid T, Guo SZ, Kingery JR, Xiang X, Dawn B, Prabhu SD. H, Sekikawa K, Seishima M. Improved myocardial ischemia/reperfusion
Cardiomyocyte NF-κB p65 promotes adverse remodelling, apopto- injury in mice lacking tumor necrosis factor-alpha. J Am Coll Cardiol.
sis, and endoplasmic reticulum stress in heart failure. Cardiovasc Res. 2002;39:1229–1235.
2011;89:129–138. doi: 10.1093/cvr/cvq274. 114. Bujak M, Dobaczewski M, Chatila K, Mendoza LH, Li N, Reddy A,
96. Xuan YT, Tang XL, Banerjee S, Takano H, Li RC, Han H, Qiu Y, Li Frangogiannis NG. Interleukin-1 receptor type I signaling critically regu-
JJ, Bolli R. Nuclear factor-kappaB plays an essential role in the late lates infarct healing and cardiac remodeling. Am J Pathol. 2008;173:57–
phase of ischemic preconditioning in conscious rabbits. Circ Res. 67. doi: 10.2353/ajpath.2008.070974.
1999;84:1095–1109. 115. Hilfiker-Kleiner D, Shukla P, Klein G, Schaefer A, Stapel B, Hoch M,
97. Tranter M, Ren X, Forde T, Wilhide ME, Chen J, Sartor MA, Medvedovic Müller W, Scherr M, Theilmeier G, Ernst M, Hilfiker A, Drexler H.
M, Jones WK. NF-kappaB driven cardioprotective gene programs; Continuous glycoprotein-130-mediated signal transducer and activator
Hsp70.3 and cardioprotection after late ischemic preconditioning. J Mol of transcription-3 activation promotes inflammation, left ventricular rup-
Cell Cardiol. 2010;49:664–672. doi: 10.1016/j.yjmcc.2010.07.001. ture, and adverse outcome in subacute myocardial infarction. Circulation.
98. Brown M, McGuinness M, Wright T, Ren X, Wang Y, Boivin GP, Hahn 2010;122:145–155. doi: 10.1161/CIRCULATIONAHA.109.933127.
H, Feldman AM, Jones WK. Cardiac-specific blockade of NF-kappaB in 116. Liehn EA, Piccinini AM, Koenen RR, Soehnlein O, Adage T, Fatu R,
cardiac pathophysiology: differences between acute and chronic stimuli Curaj A, Popescu A, Zernecke A, Kungl AJ, Weber C. A new mono-
in vivo. Am J Physiol Heart Circ Physiol. 2005;289:H466–H476. doi: cyte chemotactic protein-1/chemokine CC motif ligand-2 competitor
10.1152/ajpheart.00170.2004. limiting neointima formation and myocardial ischemia/reperfusion in-
99. Zhang XQ, Tang R, Li L, Szucsik A, Javan H, Saegusa N, Spitzer KW, jury in mice. J Am Coll Cardiol. 2010;56:1847–1857. doi: 10.1016/j.
Selzman CH. Cardiomyocyte-specific p65 NF-κB deletion protects the in- jacc.2010.04.066.
jured heart by preservation of calcium handling. Am J Physiol Heart Circ 117. Dewald O, Zymek P, Winkelmann K, Koerting A, Ren G, Abou-Khamis
Physiol. 2013;305:H1089–H1097. doi: 10.1152/ajpheart.00067.2013. T, Michael LH, Rollins BJ, Entman ML, Frangogiannis NG. CCL2/
100. Squadrito F, Deodato B, Squadrito G, Seminara P, Passaniti M, Venuti Monocyte Chemoattractant Protein-1 regulates inflammatory responses
FS, Giacca M, Minutoli L, Adamo EB, Bellomo M, Marini R, Galeano critical to healing myocardial infarcts. Circ Res. 2005;96:881–889. doi:
M, Marini H, Altavilla D. Gene transfer of IkappaBalpha limits infarct 10.1161/01.RES.0000163017.13772.3a.
size in a mouse model of myocardial ischemia-reperfusion injury. Lab 118. Hayasaki T, Kaikita K, Okuma T, Yamamoto E, Kuziel WA, Ogawa H,
Invest. 2003;83:1097–1104. Takeya M. CC chemokine receptor-2 deficiency attenuates oxidative
101. Morishita R, Sugimoto T, Aoki M, Kida I, Tomita N, Moriguchi A, stress and infarct size caused by myocardial ischemia-reperfusion in
Maeda K, Sawa Y, Kaneda Y, Higaki J, Ogihara T. In vivo transfection of mice. Circ J. 2006;70:342–351.
cis element “decoy” against nuclear factor-kappaB binding site prevents 119. Kaikita K, Hayasaki T, Okuma T, Kuziel WA, Ogawa H, Takeya M.
myocardial infarction. Nat Med. 1997;3:894–899. Targeted deletion of CC chemokine receptor 2 attenuates left ventricu-
102. Onai Y, Suzuki J, Kakuta T, Maejima Y, Haraguchi G, Fukasawa H, Muto lar remodeling after experimental myocardial infarction. Am J Pathol.
S, Itai A, Isobe M. Inhibition of IkappaB phosphorylation in cardiomyo- 2004;165:439–447. doi: 10.1016/S0002-9440(10)63309-3.
cytes attenuates myocardial ischemia/reperfusion injury. Cardiovasc 120. Kurrelmeyer KM, Michael LH, Baumgarten G, Taffet GE, Peschon
Res. 2004;63:51–59. doi: 10.1016/j.cardiores.2004.03.002. JJ, Sivasubramanian N, Entman ML, Mann DL. Endogenous tumor
103. Moss NC, Stansfield WE, Willis MS, Tang RH, Selzman CH. IKKbeta necrosis factor protects the adult cardiac myocyte against ischemic-
inhibition attenuates myocardial injury and dysfunction following induced apoptosis in a murine model of acute myocardial infarc-
acute ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. tion. Proc Natl Acad Sci U S A. 2000;97:5456–5461. doi: 10.1073/
2007;293:H2248–H2253. doi: 10.1152/ajpheart.00776.2007. pnas.070036297.
104. Li HL, Zhuo ML, Wang D, Wang AB, Cai H, Sun LH, Yang Q, Huang 121. Fuchs M, Hilfiker A, Kaminski K, Hilfiker-Kleiner D, Guener Z, Klein
Y, Wei YS, Liu PP, Liu DP, Liang CC. Targeted cardiac overexpression G, Podewski E, Schieffer B, Rose-John S, Drexler H. Role of interleu-
of A20 improves left ventricular performance and reduces compensatory kin-6 for LV remodeling and survival after experimental myocardial in-
hypertrophy after myocardial infarction. Circulation. 2007;115:1885– farction. FASEB J. 2003;17:2118–2120. doi: 10.1096/fj.03-0331fje.
1894. doi: 10.1161/CIRCULATIONAHA.106.656835. 122. Obana M, Maeda M, Takeda K, Hayama A, Mohri T, Yamashita

105. Trescher K, Bernecker O, Fellner B, Gyöngyösi M, Schäfer R,
T, Nakaoka Y, Komuro I, Takeda K, Matsumiya G, Azuma J, Fujio
Aharinejad S, DeMartin R, Wolner E, Podesser BK. Inflammation and Y. Therapeutic activation of signal transducer and activator of tran-
postinfarct remodeling: overexpression of IkappaB prevents ventricular scription 3 by interleukin-11 ameliorates cardiac fibrosis after myo-
dilation via increasing TIMP levels. Cardiovasc Res. 2006;69:746–754. cardial infarction. Circulation. 2010;121:684–691. doi: 10.1161/
doi: 10.1016/j.cardiores.2005.11.027. CIRCULATIONAHA.109.893677.
106. Timmers L, van Keulen JK, Hoefer IE, Meijs MF, van Middelaar B, den 123. Dobaczewski M, Xia Y, Bujak M, Gonzalez-Quesada C, Frangogiannis
Ouden K, van Echteld CJ, Pasterkamp G, de Kleijn DP. Targeted deletion NG. CCR5 signaling suppresses inflammation and reduces ad-
of nuclear factor kappaB p50 enhances cardiac remodeling and dysfunc- verse remodeling of the infarcted heart, mediating recruitment of
tion following myocardial infarction. Circ Res. 2009;104:699–706. doi: regulatory T cells. Am J Pathol. 2010;176:2177–2187. doi: 10.2353/
10.1161/CIRCRESAHA.108.189746. ajpath.2010.090759.
107. Frantz S, Tillmanns J, Kuhlencordt PJ, Schmidt I, Adamek A, Dienesch 124. Zamilpa R, Kanakia R, Cigarroa J IV, Dai Q, Escobar GP, Martinez
C, Thum T, Gerondakis S, Ertl G, Bauersachs J. Tissue-specific effects H, Jimenez F, Ahuja SS, Lindsey ML. CC chemokine receptor 5 dele-
of the nuclear factor kappaB subunit p50 on myocardial ischemia- tion impairs macrophage activation and induces adverse remodeling
reperfusion injury. Am J Pathol. 2007;171:507–512. doi: 10.2353/ following myocardial infarction. Am J Physiol Heart Circ Physiol.
ajpath.2007.061042. 2011;300:H1418–H1426. doi: 10.1152/ajpheart.01002.2010.
108. Frantz S, Hu K, Bayer B, Gerondakis S, Strotmann J, Adamek A, Ertl G, 125. Hamid T, Gu Y, Ortines RV, Bhattacharya C, Wang G, Xuan YT, Prabhu
Bauersachs J. Absence of NF-kappaB subunit p50 improves heart failure SD. Divergent tumor necrosis factor receptor-related remodeling re-
after myocardial infarction. FASEB J. 2006;20:1918–1920. doi: 10.1096/ sponses in heart failure: role of nuclear factor-kappaB and inflam-
fj.05-5133fje. matory activation. Circulation. 2009;119:1386–1397. doi: 10.1161/
109. Zhang LX, Zhao Y, Cheng G, Guo TL, Chin YE, Liu PY, Zhao TC. CIRCULATIONAHA.108.802918.
Targeted deletion of NF-kappaB p50 diminishes the cardioprotection 126. Garlie JB, Hamid T, Gu Y, Ismahil MA, Chandrasekar B, Prabhu

of histone deacetylase inhibition. Am J Physiol Heart Circ Physiol. SD. Tumor necrosis factor receptor 2 signaling limits β-adrenergic
2010;298:H2154–H2163. doi: 10.1152/ajpheart.01015.2009. receptor-mediated cardiac hypertrophy in vivo. Basic Res Cardiol.
110. Cavalera M, Frangogiannis NG. Targeting the chemokines in cardiac re- 2011;106:1193–1205. doi: 10.1007/s00395-011-0196-6.
pair. Curr Pharm Des. 2014;20:1971–1979. 127. Sager HB, Heidt T, Hulsmans M, Dutta P, Courties G, Sebas M,

111. Prabhu SD. Cytokine-induced modulation of cardiac function. Circ Res. Wojtkiewicz GR, Tricot B, Iwamoto Y, Sun Y, Weissleder R, Libby
2004;95:1140–1153. doi: 10.1161/01.RES.0000150734.79804.92. P, Swirski FK, Nahrendorf M. Targeting interleukin-1β reduces leu-
112. Bromage DI, Davidson SM, Yellon DM. Stromal derived fac-
kocyte production after acute myocardial infarction. Circulation.
tor 1α: a chemokine that delivers a two-pronged defence of the 2015;132:1880–1890. doi: 10.1161/CIRCULATIONAHA.115.016160.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   109

128. Guo Y, Hangoc G, Bian H, Pelus LM, Broxmeyer HE. SDF-1/CXCL12 147. Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW.

enhances survival and chemotaxis of murine embryonic stem cells Emerging mechanisms of neutrophil recruitment across endothelium.
and production of primitive and definitive hematopoietic progeni- Trends Immunol. 2011;32:461–469. doi: 10.1016/j.it.2011.06.009.
tor cells. Stem Cells. 2005;23:1324–1332. doi: 10.1634/stemcells. 148. Boufenzer A, Lemarié J, Simon T, et al. TREM-1 Mediates inflammatory
2005-0085. injury and cardiac remodeling following myocardial infarction. Circ Res.
129. Saxena A, Fish JE, White MD, Yu S, Smyth JW, Shaw RM, DiMaio JM, 2015;116:1772–1782. doi: 10.1161/CIRCRESAHA.116.305628.
Srivastava D. Stromal cell-derived factor-1alpha is cardioprotective after 149. Simpson PJ, Todd RF III, Fantone JC, Mickelson JK, Griffin JD, Lucchesi
myocardial infarction. Circulation. 2008;117:2224–2231. doi: 10.1161/ BR. Reduction of experimental canine myocardial reperfusion injury by
CIRCULATIONAHA.107.694992. a monoclonal antibody (anti-Mo1, anti-CD11b) that inhibits leukocyte
130. Vasilyev N, Williams T, Brennan ML, Unzek S, Zhou X, Heinecke JW, adhesion. J Clin Invest. 1988;81:624–629. doi: 10.1172/JCI113364.
Spitz DR, Topol EJ, Hazen SL, Penn MS. Myeloperoxidase-generated 150. Entman ML, Youker K, Shoji T, Kukielka G, Shappell SB, Taylor AA,
oxidants modulate left ventricular remodeling but not infarct size after Smith CW. Neutrophil induced oxidative injury of cardiac myocytes. A
myocardial infarction. Circulation. 2005;112:2812–2820. doi: 10.1161/ compartmented system requiring CD11b/CD18-ICAM-1 adherence. J
CIRCULATIONAHA.105.542340. Clin Invest. 1992;90:1335–1345. doi: 10.1172/JCI115999.
131. Kukielka GL, Hawkins HK, Michael L, Manning AM, Youker K, 151. Christia P, Frangogiannis NG. Targeting inflammatory pathways in myo-
Lane C, Entman ML, Smith CW, Anderson DC. Regulation of inter- cardial infarction. Eur J Clin Invest. 2013;43:986–995. doi: 10.1111/
cellular adhesion molecule-1 (ICAM-1) in ischemic and reperfused eci.12118.
canine myocardium. J Clin Invest. 1993;92:1504–1516. doi: 10.1172/ 152. Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger

JCI116729. T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ. The healing myo-
132. Tarzami ST, Cheng R, Miao W, Kitsis RN, Berman JW. Chemokine cardium sequentially mobilizes two monocyte subsets with divergent
expression in myocardial ischemia: MIP-2 dependent MCP-1 expres- and complementary functions. J Exp Med. 2007;204:3037–3047. doi:
sion protects cardiomyocytes from cell death. J Mol Cell Cardiol. 10.1084/jem.20070885.
2002;34:209–221. doi: 10.1006/jmcc.2001.1503. 153. Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo
133. Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’Antoni ML, Debuque R, V, Panizzi P, Figueiredo JL, Kohler RH, Chudnovskiy A, Waterman P,
Chandran A, Wang L, Arora K, Rosenthal NA, Tallquist MD. Revisiting Aikawa E, Mempel TR, Libby P, Weissleder R, Pittet MJ. Identification
Cardiac Cellular Composition. Circ Res. 2016;118:400–409. doi: of splenic reservoir monocytes and their deployment to inflammatory
10.1161/CIRCRESAHA.115.307778. sites. Science. 2009;325:612–616. doi: 10.1126/science.1175202.
134. Zhu M, Goetsch SC, Wang Z, Luo R, Hill JA, Schneider J, Morris SM 154. Frangogiannis NG, Mendoza LH, Lindsey ML, Ballantyne CM,

Jr, Liu ZP. FoxO4 promotes early inflammatory response upon myocar- Michael LH, Smith CW, Entman ML. IL-10 is induced in the reper-
dial infarction via endothelial Arg1. Circ Res. 2015;117:967–977. doi: fused myocardium and may modulate the reaction to injury. J Immunol.
10.1161/CIRCRESAHA.115.306919. 2000;165:2798–2808.
135. Weyrich AS, Ma XY, Lefer DJ, Albertine KH, Lefer AM. In vivo neutral- 155. Varda-Bloom N, Leor J, Ohad DG, Hasin Y, Amar M, Fixler R, Battler A,
ization of P-selectin protects feline heart and endothelium in myocardial Eldar M, Hasin D. Cytotoxic T lymphocytes are activated following myo-
ischemia and reperfusion injury. J Clin Invest. 1993;91:2620–2629. doi: cardial infarction and can recognize and kill healthy myocytes in vitro. J
10.1172/JCI116501. Mol Cell Cardiol. 2000;32:2141–2149. doi: 10.1006/jmcc.2000.1261.
136. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health 156. Zouggari Y, Ait-Oufella H, Bonnin P, et al. B lymphocytes trigger mono-
and inflammation. Nat Rev Immunol. 2013;13:159–175. doi: 10.1038/ cyte mobilization and impair heart function after acute myocardial in-
nri3399. farction. Nat Med. 2013;19:1273–1280. doi: 10.1038/nm.3284.
137. Kumar AG, Ballantyne CM, Michael LH, Kukielka GL, Youker KA, 157. Souders CA, Bowers SL, Baudino TA. Cardiac fibroblast: the re-

Lindsey ML, Hawkins HK, Birdsall HH, MacKay CR, LaRosa GJ, naissance cell. Circ Res. 2009;105:1164–1176. doi: 10.1161/
Rossen RD, Smith CW, Entman ML. Induction of monocyte chemoat- CIRCRESAHA.109.209809.
tractant protein-1 in the small veins of the ischemic and reperfused ca- 158. Shinde AV, Frangogiannis NG. Fibroblasts in myocardial infarction: a
nine myocardium. Circulation. 1997;95:693–700. role in inflammation and repair. J Mol Cell Cardiol. 2014;70:74–82. doi:
138. Frangogiannis NG, Mendoza LH, Lewallen M, Michael LH, Smith CW, 10.1016/j.yjmcc.2013.11.015.
Entman ML. Induction and suppression of interferon-inducible protein 159. Saxena A, Chen W, Su Y, Rai V, Uche OU, Li N, Frangogiannis NG. IL-1
10 in reperfused myocardial infarcts may regulate angiogenesis. FASEB induces proinflammatory leukocyte infiltration and regulates fibroblast
J. 2001;15:1428–1430. phenotype in the infarcted myocardium. J Immunol. 2013;191:4838–
139. Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in 4848. doi: 10.4049/jimmunol.1300725.
the activation and regulation of innate and adaptive immunity. Nat Rev 160. Frangogiannis NG, Lindsey ML, Michael LH, Youker KA, Bressler RB,
Immunol. 2011;11:519–531. doi: 10.1038/nri3024. Mendoza LH, Spengler RN, Smith CW, Entman ML. Resident cardiac
140. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual mast cells degranulate and release preformed TNF-alpha, initiating the
anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. cytokine cascade in experimental canine myocardial ischemia/reperfu-
2008;8:349–361. doi: 10.1038/nri2294. sion. Circulation. 1998;98:699–710.
141. Soehnlein O, Lindbom L. Phagocyte partnership during the onset and 161. Somasundaram P, Ren G, Nagar H, Kraemer D, Mendoza L, Michael
resolution of inflammation. Nat Rev Immunol. 2010;10:427–439. doi: LH, Caughey GH, Entman ML, Frangogiannis NG. Mast cell tryptase
10.1038/nri2779. may modulate endothelial cell phenotype in healing myocardial infarcts.
142. Detmers PA, Lo SK, Olsen-Egbert E, Walz A, Baggiolini M, Cohn ZA. J Pathol. 2005;205:102–111. doi: 10.1002/path.1690.
Neutrophil-activating protein 1/interleukin 8 stimulates the binding ac- 162. Epelman S, Lavine KJ, Beaudin AE, et al. Embryonic and adult-derived
tivity of the leukocyte adhesion receptor CD11b/CD18 on human neutro- resident cardiac macrophages are maintained through distinct mecha-
phils. J Exp Med. 1990;171:1155–1162. nisms at steady state and during inflammation. Immunity. 2014;40:91–
143. Herter J, Zarbock A. Integrin Regulation during Leukocyte Recruitment. 104. doi: 10.1016/j.immuni.2013.11.019.
J Immunol. 2013;190:4451–4457. doi: 10.4049/jimmunol.1203179. 163. Heidt T, Courties G, Dutta P, Sager HB, Sebas M, Iwamoto Y, Sun Y, Da
144. Phillipson M, Heit B, Colarusso P, Liu L, Ballantyne CM, Kubes P. Silva N, Panizzi P, van der Laan AM, van der Lahn AM, Swirski FK,
Intraluminal crawling of neutrophils to emigration sites: a molecularly Weissleder R, Nahrendorf M. Differential contribution of monocytes to
distinct process from adhesion in the recruitment cascade. J Exp Med. heart macrophages in steady-state and after myocardial infarction. Circ
2006;203:2569–2575. doi: 10.1084/jem.20060925. Res. 2014;115:284–295. doi: 10.1161/CIRCRESAHA.115.303567.
145. Wang S, Voisin MB, Larbi KY, Dangerfield J, Scheiermann C, Tran M, 164. Pinto AR, Paolicelli R, Salimova E, Gospocic J, Slonimsky E, Bilbao-
Maxwell PH, Sorokin L, Nourshargh S. Venular basement membranes Cortes D, Godwin JW, Rosenthal NA. An abundant tissue macrophage
contain specific matrix protein low expression regions that act as exit population in the adult murine heart with a distinct alternatively-activated
points for emigrating neutrophils. J Exp Med. 2006;203:1519–1532. doi: macrophage profile. PLoS One. 2012;7:e36814. doi: 10.1371/journal.
10.1084/jem.20051210. pone.0036814.
146. Proebstl D, Voisin MB, Woodfin A, Whiteford J, D’Acquisto F, Jones 165. Mylonas KJ, Jenkins SJ, Castellan RF, Ruckerl D, McGregor K,

GE, Rowe D, Nourshargh S. Pericytes support neutrophil subendothe- Phythian-Adams AT, Hewitson JP, Campbell SM, MacDonald AS, Allen
lial cell crawling and breaching of venular walls in vivo. J Exp Med. JE, Gray GA. The adult murine heart has a sparse, phagocytically active
2012;209:1219–1234. doi: 10.1084/jem.20111622. macrophage population that expands through monocyte recruitment and

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


110  Circulation Research  June 24, 2016

adopts an ‘M2’ phenotype in response to Th2 immunologic challenge. 184. Li J, Brown LF, Hibberd MG, Grossman JD, Morgan JP, Simons M.
Immunobiology. 2015;220:924–933. doi: 10.1016/j.imbio.2015.01.013. VEGF, flk-1, and flt-1 expression in a rat myocardial infarction model of
166. Dobaczewski M, Gonzalez-Quesada C, Frangogiannis NG. The extracel- angiogenesis. Am J Physiol. 1996;270:H1803–H1811.
lular matrix as a modulator of the inflammatory and reparative response 185. Ren G, Michael LH, Entman ML, Frangogiannis NG. Morphological
following myocardial infarction. J Mol Cell Cardiol. 2010;48:504–511. characteristics of the microvasculature in healing myocardial infarcts.
doi: 10.1016/j.yjmcc.2009.07.015. J Histochem Cytochem. 2002;50:71–79.
167. Rienks M, Papageorgiou AP, Frangogiannis NG, Heymans S. Myocardial 186. Zymek P, Bujak M, Chatila K, Cieslak A, Thakker G, Entman ML,
extracellular matrix: an ever-changing and diverse entity. Circ Res. Frangogiannis NG. The role of platelet-derived growth factor signaling
2014;114:872–888. doi: 10.1161/CIRCRESAHA.114.302533. in healing myocardial infarcts. J Am Coll Cardiol. 2006;48:2315–2323.
168. Trial J, Baughn RE, Wygant JN, McIntyre BW, Birdsall HH, Youker doi: 10.1016/j.jacc.2006.07.060.
KA, Evans A, Entman ML, Rossen RD. Fibronectin fragments modu- 187. Chen CW, Okada M, Proto JD, Gao X, Sekiya N, Beckman SA, Corselli
late monocyte VLA-5 expression and monocyte migration. J Clin Invest. M, Crisan M, Saparov A, Tobita K, Péault B, Huard J. Human pericytes
1999;104:419–430. doi: 10.1172/JCI4824. for ischemic heart repair. Stem Cells. 2013;31:305–316. doi: 10.1002/
169. Huebener P, Abou-Khamis T, Zymek P, Bujak M, Ying X, Chatila K, stem.1285.
Haudek S, Thakker G, Frangogiannis NG. CD44 is critically involved 188. Katare R, Riu F, Mitchell K, Gubernator M, Campagnolo P, Cui Y,
in infarct healing by regulating the inflammatory and fibrotic response. J Fortunato O, Avolio E, Cesselli D, Beltrami AP, Angelini G, Emanueli C,
Immunol. 2008;180:2625–2633. Madeddu P. Transplantation of human pericyte progenitor cells improves
170. Geering B, Stoeckle C, Conus S, Simon HU. Living and dying for in- the repair of infarcted heart through activation of an angiogenic program
flammation: neutrophils, eosinophils, basophils. Trends Immunol. involving micro-RNA-132. Circ Res. 2011;109:894–906. doi: 10.1161/
2013;34:398–409. doi: 10.1016/j.it.2013.04.002. CIRCRESAHA.111.251546.
171. Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, 189. Frangogiannis NG. The reparative function of cardiomyocytes in the
Weber C, Soehnlein O, Steffens S. Neutrophils orchestrate post-myocardial infarcted myocardium. Cell Metab. 2015;21:797–798. doi: 10.1016/j.
infarction healing by polarizing macrophages towards a reparative pheno- cmet.2015.05.015.
type [published online ahead of print February 2, 2016]. Eur Heart J. doi: 190. Lörchner H, Pöling J, Gajawada P, Hou Y, Polyakova V, Kostin S, Adrian-
http://dx.doi.org/10.1093/eurheartj/ehw002. http://eurheartj.oxfordjournals. Segarra JM, Boettger T, Wietelmann A, Warnecke H, Richter M, Kubin
org/content/early/2016/02/01/eurheartj.ehw002. T, Braun T. Myocardial healing requires Reg3β-dependent accumulation
172. Courties G, Heidt T, Sebas M, et al. In vivo silencing of the transcrip- of macrophages in the ischemic heart. Nat Med. 2015;21:353–362. doi:
tion factor IRF5 reprograms the macrophage phenotype and improves 10.1038/nm.3816.
infarct healing. J Am Coll Cardiol. 2014;63:1556–1566. doi: 10.1016/j. 191. Frangogiannis NG. The inflammatory response in myocardial injury, re-
jacc.2013.11.023. pair, and remodelling. Nat Rev Cardiol. 2014;11:255–265. doi: 10.1038/
173. Hilgendorf I, Gerhardt LM, Tan TC, Winter C, Holderried TA,
nrcardio.2014.28.
Chousterman BG, Iwamoto Y, Liao R, Zirlik A, Scherer-Crosbie M, 192. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. IL-
Hedrick CC, Libby P, Nahrendorf M, Weissleder R, Swirski FK. Ly- 10 inhibits cytokine production by activated macrophages. J Immunol.
6Chigh monocytes depend on Nr4a1 to balance both inflammatory and 1991;147:3815–3822.
reparative phases in the infarcted myocardium. Circ Res. 2014;114:1611– 193. Matsukawa A, Kudo S, Maeda T, Numata K, Watanabe H, Takeda K,
1622. doi: 10.1161/CIRCRESAHA.114.303204. Akira S, Ito T. Stat3 in resident macrophages as a repressor protein of
174. Leuschner F, Rauch PJ, Ueno T, et al. Rapid monocyte kinetics in acute inflammatory response. J Immunol. 2005;175:3354–3359.
myocardial infarction are sustained by extramedullary monocytopoiesis. 194. Dewald O, Ren G, Duerr GD, Zoerlein M, Klemm C, Gersch C, Tincey
J Exp Med. 2012;209:123–137. doi: 10.1084/jem.20111009. S, Michael LH, Entman ML, Frangogiannis NG. Of mice and dogs:
175. Korf-Klingebiel M, Reboll MR, Klede S, et al. Myeloid-derived growth species-specific differences in the inflammatory response following
factor (C19orf10) mediates cardiac repair following myocardial infarc- myocardial infarction. Am J Pathol. 2004;164:665–677. doi: 10.1016/
tion. Nat Med. 2015;21:140–149. doi: 10.1038/nm.3778. S0002-9440(10)63154-9.
176. Hanna RN, Carlin LM, Hubbeling HG, Nackiewicz D, Green AM, Punt 195. Yang Z, Zingarelli B, Szabó C. Crucial role of endogenous interleukin-10
JA, Geissmann F, Hedrick CC. The transcription factor NR4A1 (Nur77) production in myocardial ischemia/reperfusion injury. Circulation.
controls bone marrow differentiation and the survival of Ly6C- mono- 2000;101:1019–1026.
cytes. Nat Immunol. 2011;12:778–785. doi: 10.1038/ni.2063. 196. Zymek P, Nah DY, Bujak M, Ren G, Koerting A, Leucker T, Huebener P,
177. Anzai A, Anzai T, Nagai S, et al. Regulatory role of dendritic
Taffet G, Entman M, Frangogiannis NG. Interleukin-10 is not a critical
cells in postinfarction healing and left ventricular remodeling. Circulation. regulator of infarct healing and left ventricular remodeling. Cardiovasc
2012;125:1234–1245. doi: 10.1161/CIRCULATIONAHA.111.052126. Res. 2007;74:313–322. doi: 10.1016/j.cardiores.2006.11.028.
178. Hofmann U, Beyersdorf N, Weirather J, Podolskaya A, Bauersachs
197. Cavusoglu E, Marmur JD, Hojjati MR, Chopra V, Butala M, Subnani R,
J, Ertl G, Kerkau T, Frantz S. Activation of CD4+ T lymphocytes im- Huda MS, Yanamadala S, Ruwende C, Eng C, Pinsky DJ. Plasma inter-
proves wound healing and survival after experimental myocardial leukin-10 levels and adverse outcomes in acute coronary syndrome. Am
infarction in mice. Circulation. 2012;125:1652–1663. doi: 10.1161/ J Med. 2011;124:724–730. doi: 10.1016/j.amjmed.2011.02.040.
CIRCULATIONAHA.111.044164. 198. Ammirati E, Cannistraci CV, Cristell NA, et al. Identification and

179. Homma T, Kinugawa S, Takahashi M, et al. Activation of invariant natu- predictive value of interleukin-6+ interleukin-10+ and interleu-
ral killer T cells by α-galactosylceramide ameliorates myocardial isch- kin-6- interleukin-10+ cytokine patterns in ST-elevation acute myo-
emia/reperfusion injury in mice. J Mol Cell Cardiol. 2013;62:179–188. cardial infarction. Circ Res. 2012;111:1336–1348. doi: 10.1161/
doi: 10.1016/j.yjmcc.2013.06.004. CIRCRESAHA.111.262477.
180. Sobirin MA, Kinugawa S, Takahashi M, Fukushima A, Homma T, Ono 199. Dobaczewski M, Chen W, Frangogiannis NG. Transforming growth

T, Hirabayashi K, Suga T, Azalia P, Takada S, Taniguchi M, Nakayama factor (TGF)-β signaling in cardiac remodeling. J Mol Cell Cardiol.
T, Ishimori N, Iwabuchi K, Tsutsui H. Activation of natural killer T cells 2011;51:600–606. doi: 10.1016/j.yjmcc.2010.10.033.
ameliorates postinfarct cardiac remodeling and failure in mice. Circ Res. 200. Ikeuchi M, Tsutsui H, Shiomi T, Matsusaka H, Matsushima S, Wen J,
2012;111:1037–1047. doi: 10.1161/CIRCRESAHA.112.270132. Kubota T, Takeshita A. Inhibition of TGF-beta signaling exacerbates
181. Weirather J, Hofmann UD, Beyersdorf N, Ramos GC, Vogel B,
early cardiac dysfunction but prevents late remodeling after infarction.
Frey A, Ertl G, Kerkau T, Frantz S. Foxp3+ CD4+ T cells improve Cardiovasc Res. 2004;64:526–535. doi: 10.1016/j.cardiores.2004.07.017.
healing after myocardial infarction by modulating monocyte/mac- 201. Rainer PP, Hao S, Vanhoutte D, Lee DI, Koitabashi N, Molkentin JD,
rophage differentiation. Circ Res. 2014;115:55–67. doi: 10.1161/ Kass DA. Cardiomyocyte-specific transforming growth factor β suppres-
CIRCRESAHA.115.303895. sion blocks neutrophil infiltration, augments multiple cytoprotective cas-
182. Yan X, Shichita T, Katsumata Y, et al. Deleterious effect of the IL-23/IL-17A cades, and reduces early mortality after myocardial infarction. Circ Res.
axis and γδT cells on left ventricular remodeling after myocardial infarc- 2014;114:1246–1257. doi: 10.1161/CIRCRESAHA.114.302653.
tion. J Am Heart Assoc. 2012;1:e004408. doi: 10.1161/JAHA.112.004408. 202. Kempf T, Zarbock A, Widera C, et al. GDF-15 is an inhibitor of leuko-
183. Stabile E, Burnett MS, Watkins C, Kinnaird T, Bachis A, la Sala A, cyte integrin activation required for survival after myocardial infarction
Miller JM, Shou M, Epstein SE, Fuchs S. Impaired arteriogenic re- in mice. Nat Med. 2011;17:581–588. doi: 10.1038/nm.2354.
sponse to acute hindlimb ischemia in CD4-knockout mice. Circulation. 203. Kempf T, Björklund E, Olofsson S, Lindahl B, Allhoff T, Peter T, Tongers
2003;108:205–210. doi: 10.1161/01.CIR.0000079225.50817.71. J, Wollert KC, Wallentin L. Growth-differentiation factor-15 improves

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


Prabhu and Frangogiannis   Repair After Myocardial Infarction   111

risk stratification in ST-segment elevation myocardial infarction. Eur 222. Lenga Y, Koh A, Perera AS, McCulloch CA, Sodek J, Zohar R.

Heart J. 2007;28:2858–2865. doi: 10.1093/eurheartj/ehm465. Osteopontin expression is required for myofibroblast differentiation.
204. Serhan CN. Pro-resolving lipid mediators are leads for resolution physi- Circ Res. 2008;102:319–327. doi: 10.1161/CIRCRESAHA.107.160408.
ology. Nature. 2014;510:92–101. doi: 10.1038/nature13479. 223. Xia Y, Dobaczewski M, Gonzalez-Quesada C, Chen W, Biernacka

205. Keyes KT, Ye Y, Lin Y, Zhang C, Perez-Polo JR, Gjorstrup P, Birnbaum A, Li N, Lee DW, Frangogiannis NG. Endogenous thrombospondin 1
Y. Resolvin E1 protects the rat heart against reperfusion injury. Am J protects the pressure-overloaded myocardium by modulating fibroblast
Physiol Heart Circ Physiol. 2010;299:H153–H164. doi: 10.1152/ phenotype and matrix metabolism. Hypertension. 2011;58:902–911. doi:
ajpheart.01057.2009. 10.1161/HYPERTENSIONAHA.111.175323.
206. Kain V, Ingle KA, Colas RA, Dalli J, Prabhu SD, Serhan CN, Joshi M, 224. Frangogiannis NG. Matricellular proteins in cardiac adapta-

Halade GV. Resolvin D1 activates the inflammation resolving response tion and disease. Physiol Rev. 2012;92:635–688. doi: 10.1152/
at splenic and ventricular site following myocardial infarction leading to physrev.00008.2011.
improved ventricular function. J Mol Cell Cardiol. 2015;84:24–35. doi: 225. Thodeti CK, Paruchuri S and Meszaros JG. A TRP to cardiac fibroblast
10.1016/j.yjmcc.2015.04.003. differentiation. Channels. 2013;7:211–4.
207. Chen W, Saxena A, Li N, Sun J, Gupta A, Lee DW, Tian Q, Dobaczewski 226. Nilius B. TRP channels in disease. Biochim Biophys Acta.

M, Frangogiannis NG. Endogenous IRAK-M attenuates postinfarc- 2007;1772:805–812. doi: 10.1016/j.bbadis.2007.02.002.
tion remodeling through effects on macrophages and fibroblasts. 227. Adapala RK, Thoppil RJ, Luther DJ, Paruchuri S, Meszaros JG, Chilian
Arterioscler Thromb Vasc Biol. 2012;32:2598–2608. doi: 10.1161/ WM, Thodeti CK. TRPV4 channels mediate cardiac fibroblast differen-
ATVBAHA.112.300310. tiation by integrating mechanical and soluble signals. J Mol Cell Cardiol.
208. Saxena A, Shinde AV, Haque Z, Wu YJ, Chen W, Su Y, Frangogiannis 2013;54:45–52. doi: 10.1016/j.yjmcc.2012.10.016.
NG. The role of Interleukin Receptor Associated Kinase (IRAK)-M in 228. Du J, Xie J, Zhang Z, Tsujikawa H, Fusco D, Silverman D, Liang B,
regulation of myofibroblast phenotype in vitro, and in an experimental Yue L. TRPM7-mediated Ca2+ signals confer fibrogenesis in hu-
model of non-reperfused myocardial infarction. J Mol Cell Cardiol. man atrial fibrillation. Circ Res. 2010;106:992–1003. doi: 10.1161/
2015;89:223–231. doi: 10.1016/j.yjmcc.2015.11.001. CIRCRESAHA.109.206771.
209. Willems IE, Havenith MG, De Mey JG, Daemen MJ. The alpha-smooth 229. Davis J, Burr AR, Davis GF, Birnbaumer L, Molkentin JD. A TRPC6-
muscle actin-positive cells in healing human myocardial scars. Am J dependent pathway for myofibroblast transdifferentiation and
Pathol. 1994;145:868–875. wound healing in vivo. Dev Cell. 2012;23:705–715. doi: 10.1016/j.
210. Frangogiannis NG, Michael LH, Entman ML. Myofibroblasts in reper- devcel.2012.08.017.
fused myocardial infarcts express the embryonic form of smooth muscle 230. Sun Y, Weber KT. Angiotensin converting enzyme and myofibroblasts
myosin heavy chain (SMemb). Cardiovasc Res. 2000;48:89–100. during tissue repair in the rat heart. J Mol Cell Cardiol. 1996;28:851–
211. Turner NA, Porter KE. Function and fate of myofibroblasts after
858. doi: 10.1006/jmcc.1996.0080.
myocardial infarction. Fibrogenesis Tissue Repair. 2013;6:5. doi: 231. Booz GW, Baker KM. Molecular signalling mechanisms control-

10.1186/1755-1536-6-5. ling growth and function of cardiac fibroblasts. Cardiovasc Res.
212. Santiago JJ, Dangerfield AL, Rattan SG, Bathe KL, Cunnington RH, 1995;30:537–543.
Raizman JE, Bedosky KM, Freed DH, Kardami E, Dixon IM. Cardiac 232. McEwan PE, Gray GA, Sherry L, Webb DJ, Kenyon CJ. Differential ef-
fibroblast to myofibroblast differentiation in vivo and in vitro: expression fects of angiotensin II on cardiac cell proliferation and intramyocardial
of focal adhesion components in neonatal and adult rat ventricular myo- perivascular fibrosis in vivo. Circulation. 1998;98:2765–2773.
fibroblasts. Dev Dyn. 2010;239:1573–1584. doi: 10.1002/dvdy.22280. 233. Stockand JD, Meszaros JG. Aldosterone stimulates proliferation of car-
213. Yano T, Miura T, Ikeda Y, Matsuda E, Saito K, Miki T, Kobayashi H, diac fibroblasts by activating Ki-RasA and MAPK1/2 signaling. Am
Nishino Y, Ohtani S, Shimamoto K. Intracardiac fibroblasts, but not J Physiol Heart Circ Physiol. 2003;284:H176–H184. doi: 10.1152/
bone marrow derived cells, are the origin of myofibroblasts in myocar- ajpheart.00421.2002.
dial infarct repair. Cardiovasc Pathol. 2005;14:241–246. doi: 10.1016/j. 234. Bujak M, Ren G, Kweon HJ, Dobaczewski M, Reddy A, Taffet G, Wang
carpath.2005.05.004. XF, Frangogiannis NG. Essential role of Smad3 in infarct healing and
214. Möllmann H, Nef HM, Kostin S, von Kalle C, Pilz I, Weber M, Schaper in the pathogenesis of cardiac remodeling. Circulation. 2007;116:2127–
J, Hamm CW, Elsässer A. Bone marrow-derived cells contribute to in- 2138. doi: 10.1161/CIRCULATIONAHA.107.704197.
farct remodelling. Cardiovasc Res. 2006;71:661–671. doi: 10.1016/j. 235. Zymek P, Cieslak A, Tincey S, Ren G, Michael L, Entman ML and
cardiores.2006.06.013. Frangogiannis N. Platelet derived growth factor receptor (PDGFR)-beta
215. Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, signaling regulates vascular maturation in healing myocardial infarcts. J
Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Am Coll Cardiol. 2005;45:5A.
Sayegh MH, Izumo S, Kalluri R. Endothelial-to-mesenchymal transition 236. Zhao W, Zhao T, Huang V, Chen Y, Ahokas RA, Sun Y. Platelet-

contributes to cardiac fibrosis. Nat Med. 2007;13:952–961. doi: 10.1038/ derived growth factor involvement in myocardial remodeling follow-
nm1613. ing infarction. J Mol Cell Cardiol. 2011;51:830–838. doi: 10.1016/j.
216. Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML,
yjmcc.2011.06.023.
Gabbiani G. The myofibroblast: one function, multiple origins. Am J 237. Santiago JJ, McNaughton LJ, Koleini N, Ma X, Bestvater B, Nickel BE,
Pathol. 2007;170:1807–1816. doi: 10.2353/ajpath.2007.070112. Fandrich RR, Wigle JT, Freed DH, Arora RC, Kardami E. High molecu-
217. Ruiz-Villalba A, Simón AM, Pogontke C, Castillo MI, Abizanda G, lar weight fibroblast growth factor-2 in the human heart is a potential
Pelacho B, Sánchez-Domínguez R, Segovia JC, Prósper F, Pérez- target for prevention of cardiac remodeling. PLoS One. 2014;9:e97281.
Pomares JM. Interacting resident epicardium-derived fibroblasts and doi: 10.1371/journal.pone.0097281.
recruited bone marrow cells form myocardial infarction scar. J Am Coll 238. McLarty JL, Meléndez GC, Brower GL, Janicki JS, Levick SP.

Cardiol. 2015;65:2057–2066. doi: 10.1016/j.jacc.2015.03.520. Tryptase/Protease-activated receptor 2 interactions induce selective
218. Dobaczewski M, Bujak M, Li N, Gonzalez-Quesada C, Mendoza LH, mitogen-activated protein kinase signaling and collagen synthesis by
Wang XF, Frangogiannis NG. Smad3 signaling critically regulates fibro- cardiac fibroblasts. Hypertension. 2011;58:264–270. doi: 10.1161/
blast phenotype and function in healing myocardial infarction. Circ Res. HYPERTENSIONAHA.111.169417.
2010;107:418–428. doi: 10.1161/CIRCRESAHA.109.216101. 239. Zhao XY, Zhao LY, Zheng QS, Su JL, Guan H, Shang FJ, Niu XL, He
219. Wang S, Wilkes MC, Leof EB, Hirschberg R. Imatinib mesylate blocks YP, Lu XL. Chymase induces profibrotic response via transforming
a non-Smad TGF-beta pathway and reduces renal fibrogenesis in vivo. growth factor-beta 1/Smad activation in rat cardiac fibroblasts. Mol Cell
FASEB J. 2005;19:1–11. doi: 10.1096/fj.04-2370com. Biochem. 2008;310:159–166. doi: 10.1007/s11010-007-9676-2.
220. Hashimoto S, Gon Y, Takeshita I, Matsumoto K, Maruoka S, Horie T. 240. Dobaczewski M, Bujak M, Zymek P, Ren G, Entman ML, Frangogiannis
Transforming growth Factor-beta1 induces phenotypic modulation of NG. Extracellular matrix remodeling in canine and mouse myo-
human lung fibroblasts to myofibroblast through a c-Jun-NH2-terminal cardial infarcts. Cell Tissue Res. 2006;324:475–488. doi: 10.1007/
kinase-dependent pathway. Am J Respir Crit Care Med. 2001;163:152– s00441-005-0144-6.
157. doi: 10.1164/ajrccm.163.1.2005069. 241. Frangogiannis NG, Ren G, Dewald O, Zymek P, Haudek S, Koerting
221. Serini G, Bochaton-Piallat ML, Ropraz P, Geinoz A, Borsi L, Zardi L, A, Winkelmann K, Michael LH, Lawler J, Entman ML. Critical role
Gabbiani G. The fibronectin domain ED-A is crucial for myofibroblastic of endogenous thrombospondin-1 in preventing expansion of healing
phenotype induction by transforming growth factor-beta1. J Cell Biol. myocardial infarcts. Circulation. 2005;111:2935–2942. doi: 10.1161/
1998;142:873–881. CIRCULATIONAHA.104.510354.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


112  Circulation Research  June 24, 2016

242. Nishioka T, Onishi K, Shimojo N, Nagano Y, Matsusaka H, Ikeuchi M, of the cardiosplenic axis. Circ Res. 2014;114:266–282. doi: 10.1161/
Ide T, Tsutsui H, Hiroe M, Yoshida T, Imanaka-Yoshida K. Tenascin-C CIRCRESAHA.113.301720.
may aggravate left ventricular remodeling and function after myocardial 258. Saxena A, Russo I, Frangogiannis NG. Inflammation as a therapeu-
infarction in mice. Am J Physiol Heart Circ Physiol. 2010;298:H1072– tic target in myocardial infarction: learning from past failures to meet
H1078. doi: 10.1152/ajpheart.00255.2009. future challenges. Transl Res. 2016;167:152–166. doi: 10.1016/j.
243. Schellings MW, Vanhoutte D, Swinnen M, Cleutjens JP, Debets J, van trsl.2015.07.002.
Leeuwen RE, d’Hooge J, Van de Werf F, Carmeliet P, Pinto YM, Sage 259. Baran KW, Nguyen M, McKendall GR, Lambrew CT, Dykstra G,

EH, Heymans S. Absence of SPARC results in increased cardiac rup- Palmeri ST, Gibbons RJ, Borzak S, Sobel BE, Gourlay SG, Rundle
ture and dysfunction after acute myocardial infarction. J Exp Med. AC, Gibson CM, Barron HV; Limitation of Myocardial Infarction
2009;206:113–123. doi: 10.1084/jem.20081244. Following Thrombolysis in Acute Myocardial Infarction (LIMIT AMI)
244. Oka T, Xu J, Kaiser RA, Melendez J, Hambleton M, Sargent MA, Lorts Study Group. Double-blind, randomized trial of an anti-CD18 anti-
A, Brunskill EW, Dorn GW 2nd, Conway SJ, Aronow BJ, Robbins J, body in conjunction with recombinant tissue plasminogen activator for
Molkentin JD. Genetic manipulation of periostin expression reveals acute myocardial infarction: limitation of myocardial infarction follow-
a role in cardiac hypertrophy and ventricular remodeling. Circ Res. ing thrombolysis in acute myocardial infarction (LIMIT AMI) study.
2007;101:313–321. doi: 10.1161/CIRCRESAHA.107.149047. Circulation. 2001;104:2778–2783.
245. Shimazaki M, Nakamura K, Kii I, Kashima T, Amizuka N, Li M, Saito 260. APEX AMI Investigators; Armstrong PW, Granger CB, Adams PX,
M, Fukuda K, Nishiyama T, Kitajima S, Saga Y, Fukayama M, Sata M, Hamm C, Holmes D, Jr., O’Neill WW, Todaro TG, Vahanian A, Van de
Kudo A. Periostin is essential for cardiac healing after acute myocardial Werf F. Pexelizumab for acute ST-elevation myocardial infarction in
infarction. J Exp Med. 2008;205:295–303. doi: 10.1084/jem.20071297. patients undergoing primary percutaneous coronary intervention: a ran-
246. Trueblood NA, Xie Z, Communal C, Sam F, Ngoy S, Liaw L, Jenkins domized controlled trial. JAMA. 2007;297:43–51.
AW, Wang J, Sawyer DB, Bing OH, Apstein CS, Colucci WS, Singh 261. Tardif JC, Tanguay JF, Wright SR, Duchatelle V, Petroni T, Grégoire JC,
K. Exaggerated left ventricular dilation and reduced collagen deposi- Ibrahim R, Heinonen TM, Robb S, Bertrand OF, Cournoyer D, Johnson
tion after myocardial infarction in mice lacking osteopontin. Circ Res. D, Mann J, Guertin MC, L’Allier PL. Effects of the P-selectin antago-
2001;88:1080–1087. nist inclacumab on myocardial damage after percutaneous coronary in-
247. Van Aelst LN, Voss S, Carai P, et al. Osteoglycin prevents cardiac tervention for non-ST-segment elevation myocardial infarction: results
dilatation and dysfunction after myocardial infarction through infarct of the SELECT-ACS trial. J Am Coll Cardiol. 2013;61:2048–2055. doi:
collagen strengthening. Circ Res. 2015;116:425–436. doi: 10.1161/ 10.1016/j.jacc.2013.03.003.
CIRCRESAHA.116.304599. 262. Seropian IM, Toldo S, Van Tassell BW, Abbate A. Anti-inflammatory
248. Ahmed MS, Gravning J, Martinov VN, von Lueder TG, Edvardsen T, strategies for ventricular remodeling following ST-segment elevation
Czibik G, Moe IT, Vinge LE, Øie E, Valen G, Attramadal H. Mechanisms acute myocardial infarction. J Am Coll Cardiol. 2014;63:1593–1603.
of novel cardioprotective functions of CCN2/CTGF in myocar- doi: 10.1016/j.jacc.2014.01.014.
dial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 263. Abbate A, Kontos MC, Grizzard JD, Biondi-Zoccai GG, Van Tassell
2011;300:H1291–H1302. doi: 10.1152/ajpheart.00604.2010. BW, Robati R, Roach LM, Arena RA, Roberts CS, Varma A, Gelwix
249. Bujak M, Dobaczewski M, Gonzalez-Quesada C, Xia Y, Leucker
CC, Salloum FN, Hastillo A, Dinarello CA, Vetrovec GW; VCU-ART
T, Zymek P, Veeranna V, Tager AM, Luster AD, Frangogiannis NG. Investigators. Interleukin-1 blockade with anakinra to prevent ad-
Induction of the CXC chemokine interferon-gamma-inducible protein 10 verse cardiac remodeling after acute myocardial infarction (Virginia
regulates the reparative response following myocardial infarction. Circ Commonwealth University Anakinra Remodeling Trial [VCU-ART]
Res. 2009;105:973–983. doi: 10.1161/CIRCRESAHA.109.199471. Pilot study). Am J Cardiol. 2010;105:1371–1377.e1. doi: 10.1016/j.
250. Saxena A, Bujak M, Frunza O, Dobaczewski M, Gonzalez-Quesada C, amjcard.2009.12.059.
Lu B, Gerard C, Frangogiannis NG. CXCR3-independent actions of the 264. Carrabba N, Valenti R, Parodi G, Santoro GM, Antoniucci D. Left

CXC chemokine CXCL10 in the infarcted myocardium and in isolated ventricular remodeling and heart failure in diabetic patients treated
cardiac fibroblasts are mediated through proteoglycans. Cardiovasc Res. with primary angioplasty for acute myocardial infarction. Circulation.
2014;103:217–227. doi: 10.1093/cvr/cvu138. 2004;110:1974–1979. doi: 10.1161/01.CIR.0000143376.64970.4A.
251. Christia P, Bujak M, Gonzalez-Quesada C, Chen W, Dobaczewski M, 265. Aronson D, Musallam A, Lessick J, Dabbah S, Carasso S, Hammerman
Reddy A, Frangogiannis NG. Systematic characterization of myocardial H, Reisner S, Agmon Y, Mutlak D. Impact of diastolic dysfunction on
inflammation, repair, and remodeling in a mouse model of reperfused the development of heart failure in diabetic patients after acute myo-
myocardial infarction. J Histochem Cytochem. 2013;61:555–570. doi: cardial infarction. Circ Heart Fail. 2010;3:125–131. doi: 10.1161/
10.1369/0022155413493912. CIRCHEARTFAILURE.109.877340.
252. Takemura G, Ohno M, Hayakawa Y, Misao J, Kanoh M, Ohno A, Uno Y, 266. Biernacka A, Cavalera M, Wang J, Russo I, Shinde A, Kong P, Gonzalez-
Minatoguchi S, Fujiwara T, Fujiwara H. Role of apoptosis in the disap- Quesada C, Rai V, Dobaczewski M, Lee DW, Wang XF, Frangogiannis
pearance of infiltrated and proliferated interstitial cells after myocardial NG. Smad3 Signaling Promotes Fibrosis While Preserving Cardiac and
infarction. Circ Res. 1998;82:1130–1138. Aortic Geometry in Obese Diabetic Mice. Circ Heart Fail. 2015;8:788–
253. Larose E, Rodés-Cabau J, Pibarot P, et al. Predicting late myocardial 798. doi: 10.1161/CIRCHEARTFAILURE.114.001963.
recovery and outcomes in the early hours of ST-segment elevation myo- 267. Aurora AB, Porrello ER, Tan W, Mahmoud AI, Hill JA, Bassel-Duby
cardial infarction traditional measures compared with microvascular R, Sadek HA, Olson EN. Macrophages are required for neonatal
obstruction, salvaged myocardium, and necrosis characteristics by heart regeneration. J Clin Invest. 2014;124:1382–1392. doi: 10.1172/
cardiovascular magnetic resonance. J Am Coll Cardiol. 2010;55:2459– JCI72181.
2469. doi: 10.1016/j.jacc.2010.02.033. 268. Penn MS, Pastore J, Miller T, Aras R. SDF-1 in myocardial repair. Gene
254. van Diepen S, Newby LK, Lopes RD, et al.; APEX AMI Investigators. Ther. 2012;19:583–587. doi: 10.1038/gt.2012.32.
Prognostic relevance of baseline pro- and anti-inflammatory markers in 269. Beohar N, Rapp J, Pandya S, Losordo DW. Rebuilding the damaged
STEMI: an APEX AMI substudy. Int J Cardiol. 2013;168:2127–2133. heart: the potential of cytokines and growth factors in the treatment of
doi: 10.1016/j.ijcard.2013.01.004. ischemic heart disease. J Am Coll Cardiol. 2010;56:1287–1297. doi:
255. Devaux B, Scholz D, Hirche A, Klövekorn WP, Schaper J. Upregulation 10.1016/j.jacc.2010.05.039.
of cell adhesion molecules and the presence of low grade inflammation 270. Xiang FL, Lu X, Hammoud L, Zhu P, Chidiac P, Robbins J, Feng Q.
in human chronic heart failure. Eur Heart J. 1997;18:470–479. Cardiomyocyte-specific overexpression of human stem cell factor im-
256. Prabhu SD. It takes two to tango: monocyte and macrophage dual- proves cardiac function and survival after myocardial infarction in mice.
ity in the infarcted heart. Circ Res. 2014;114:1558–1560. doi: 10.1161/ Circulation. 2009;120:1065–74, 9 p following 1074. doi: 10.1161/
CIRCRESAHA.114.303933. CIRCULATIONAHA.108.839068.
257. Ismahil MA, Hamid T, Bansal SS, Patel B, Kingery JR, Prabhu SD. 271. Frangogiannis NG. Inflammation in cardiac injury, repair and re-

Remodeling of the mononuclear phagocyte network underlies chronic generation. Curr Opin Cardiol. 2015;30:240–245. doi: 10.1097/
inflammation and disease progression in heart failure: critical importance HCO.0000000000000158.

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016


The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation
to Fibrosis
Sumanth D. Prabhu and Nikolaos G. Frangogiannis

Circ Res. 2016;119:91-112


doi: 10.1161/CIRCRESAHA.116.303577
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2016 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/119/1/91

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation Research is online at:


http://circres.ahajournals.org//subscriptions/

Downloaded from http://circres.ahajournals.org/ at California Institute of Technology on June 26, 2016

Das könnte Ihnen auch gefallen