You are on page 1of 6

Journal of the American College of Cardiology Vol. 58, No.

23, 2011
© 2011 by the American College of Cardiology Foundation ISSN 0735-1097/$36.00
Published by Elsevier Inc. doi:10.1016/j.jacc.2011.08.034

STATE-OF-THE-ART PAPER

Repair After Myocardial Infarction,


Between Fantasy and Reality
The Role of Chemokines

Elisa A. Liehn, MD, PHD,*† Otilia Postea, PHD,* Adelina Curaj, MD,*‡§ Nikolaus Marx, MD†
Aachen, Germany; and Bucharest, Romania

Despite considerable progress over the last decades, acute myocardial infarction continues to remain the major
cause of morbidity and mortality worldwide. The present therapies include only cause-dependent interventions,
which are not able to reduce myocardial necrosis and optimize cardiac repair following infarction. This review
highlights the cellular and molecular processes after myocardial injury and focuses on chemokines, the main
modulators of the inflammatory and reparatory events, as the most valuable drug targets. (J Am Coll Cardiol
2011;58:2357–62) © 2011 by the American College of Cardiology Foundation

Modern cardiology has made considerable advances in the followed by mitochondria and genetic material transfer (6),
diagnosis and management of acute myocardial infarction discussion regarding this subject is purely speculative.
(MI), but there still remains the need to design strategies to However, it is generally accepted that Lin⫺/c-kit⫹ car-
regain the affected tissue and to re-establish the organ diac progenitor cells must exist (7) and could be localized
function entirely. beneath epicardium, or near the origin of the coronary
To accomplish this, sustained effort must be made to arteries and aorta, in a so-called cardiac stem cell niche (8).
understand the cellular and molecular mechanisms follow- On the other hand, mesenchymal stem cells (MSCs) were
ing MI (1). It is known that hypoxia induces death of also described as cardiac precursors and are mostly found in
cardiomyocytes and triggers an inflammatory response (1,2). bone marrow (9). We do not know their role in physiolog-
The recruited inflammatory cells clean the wound of tissue ical cardiac renewal; however, they have an important role in
debris, whereas fibroblasts and endothelial cells infiltrate, cardiac regeneration after injury (10) and were broadly used
proliferate, remodel the extracellular matrix, and determine as a source for stem cell therapy after MI (9).
scar formation (1). Recently, a network of interstitial cells with exceptionally
Beyond their crucial role in coordinating leukocyte re- long cellular processes, called telocytes, which assure the
cruitment (3), chemokines interfere with all phases of MI long-distance signaling in myocardium, has been described
(4) (Fig. 1, Online Appendix, Online Table 1), contributing (8). These cells seem to play a significant role in cardiac
to inflammatory and reparatory processes. development, physiology, renewal, and repair (11).
Heart: the “motor” of life. Myocardium is built from long A dramatic scenario occurs when, for various reasons, the
chains of striated muscle, and because of its increased blood supply to the heart cells is interrupted. Without
mitochondrial content, exhibits remarkable resistance to oxygen, most of the cardiomyocytes will die. Therefore, a
fatigue, sustaining all vital functions of the organism. rapid repair and renewal of the injured area is of critical
Despite what was hitherto believed, new evidence dem- importance.
onstrates the regeneration of heart muscle cells after birth Inflammatory phase: danger or necessity. Despite the
(5). But how is that possible? Although several studies have cardioprotective effects of some secreted cytokines (TNF␣, HIF)
reported a possible fusion of stem cells with cardiomyocytes and chemokines (CCL2, CXCL12, macrophage inhibitory
factor) (12–17), along with permanent damaging of the
From the *Institute for Molecular Cardiovascular Research (IMCAR), RWTH
cardiomyocyte, the local repair capacity of the heart is
Aachen University, Aachen, Germany; †Department of Cardiology, Pneumology, limited, and prompt external cellular help at this moment
Angiology and Internal Medicine Intensive Care (Internal Medicine I), University becomes essential.
Hospital, RWTH Aachen University, Aachen, Germany; ‡Department of Experi-
mental Molecular Imaging, RWTH Aachen University, Aachen, Germany; and the The initial inflammation is modulated through several
§Victor Babes National Institute of Pathology, Bucharest, Romania. All authors have molecular steps involving selectins, integrins, cell adhesion
reported that they have no relationships relevant to the contents of this paper to molecules (1), and various endogenous ligands, called danger-
disclose.
Manuscript received April 20, 2011; revised manuscript received July 26, 2011, associated molecular patterns (DAMPs) or “danger signals”
accepted August 2, 2011. (18). Moreover, several Toll-like receptors (TLRs) recog-
2358 Liehn et al. JACC Vol. 58, No. 23, 2011
Chemokines in Cardiac Repair November 29, 2011:2357–62

Abbreviations nize pathogen-associated molec- reaction and pave the way for monocytes. However, the
and Acronyms ular patterns (PAMPs), the basis explosion of inflammatory mediators leads to a self-
of the innate immune system (19), maintaining cycle, increasing myocardial damage. There-
EPC ⴝ endothelial
progenitor cell but are also endogenous markers fore, specific reduction of neutrophil infiltration could have
MI ⴝ myocardial infarction
of tissue damage (20). a favorable clinical impact on the evolution of an MI.
The first cells recruited within Besides the cellular external help, the inflammatory re-
MSC ⴝ mesenchymal stem
cell
hours for a short time to the in- sponse can also have another important task: preparing the
jured site are neutrophils. When surrounding unaffected cardiomyocytes for stress conditions.
arriving, they release proteolytic It is known that chemokines and cytokines are synthesized,
enzymes and reactive oxygen species, and directly injure not only in infarcted tissue, but also in unaffected regions
surrounding cells (1,21). Neutrophil depletion in animals (4), triggering initiation of defending mechanisms, such as
undergoing MI led to a marked decrease in infarct size activating HIF (39) and NF-kB (40), and place cardio-
(22,23). All successful experimental interventions of block- myocytes in a “pre-conditioning” state, increasing resistance
ing complement (24), reactive oxygen species (25), to a new hypoxic event. HIF induces specific synthesis of
or NF-kB pathway (26) reduce neutrophil infiltration. some chemokines, such as CXCL12 (41) and MIF (42),
CXCR2-binding chemokines, including CXCL8, CXCL2, with demonstrated cardioprotective effects. Impairing HIF
CXCL1 and CXCL6, as well as CCL3 and CCL5 (23,27), activation by hypoxic condition, as in diabetic hearts,
are also critical for neutrophil recruitment (4). Blocking interferes with ATP production and induces metabolic,
these chemokines and their receptors dramatically reduces energetic abnormalities, disturbing vessel development (43).
infarction size and preserves cardiac function. In conclusion, the inflammatory response after MI de-
On the other hand, neutrophils release chemotactic serves special attention and may provide therapeutic key
factors (28) and induce accumulation of splenic monocytes targets to control the entire wound healing and scar forma-
in injured tissue (29). Blocking this splenic monocytes tion (33).
exodus by angiotensin-converting enzyme inhibition has a Proliferation phase: when it starts and why it stops. The
beneficial effect on cardiac function (30). Two monocyte transition between the inflammatory and proliferation
populations have been identified, the inflammatory mono- phases is decisive. The heart wall, cleaned of dead cardio-
cytes: Gr1(⫹)CCR2(⫹)CX3CR1(lo)(Gr1high), and the myocytes and matrix debris, without new supporting struc-
resident monocytes: Gr1(⫺)CCR2(⫺)CX3CR1(hi)(Gr1low) tures, will inevitably break under the high blood pressure
(31). The first population of murine monocytes shares the and high mechanical force. The incidence of experimental
same characteristics with the classical human CD14hiCD16⫺ heart rupture occurs at a later time, after a hypoxic insult
monocytes, dominates the early phase of MI, and exhibits (44), and coincides with the passage between the 2 phases.
phagocytic, proteolytic, and inflammatory functions. They As soon as the necrotic cells and matrix debris are removed,
digest damaged tissue and clear the wound of cellular debris. the rapid inhibition of inflammation by TGF-␤ and IL10 (45)
The murine resident monocytes correspond to human is essential for the optimal infarct healing. Inflammatory cells
CD14loCD16⫹ nonclassical monocytes (32) and dominate the are replaced by resident or recruited Gr1low monocytes, lym-
later phase, have attenuated inflammatory properties, and phocytes, and mast cells that initiate healing.
promote healing via myofibroblast accumulation, angiogenesis, Resident monocytes, a heterogenic group of cells, patrol
and deposition of collagen (33). Neutrophils seem to sustain between the cardiomyocytes using CX3CR1 receptors (34),
the extravasation of inflammatory monocytes, in a CCR2- maturate into macrophages, and maintain the homeostasis
dependent manner, but not the recruitment of resident mono- of the normal tissue. After MI, they are recruited to the site
cytes (34). of injury by CX3CR1-dependent (46) or CCR5-dependent
Recent data have shown that Ly-6C(hi) monocytosis (47) mechanisms. Recent studies demonstrate the existence
(35), as well as CD14hiCD16⫺, but not resident mono- of 2 distinct types of macrophages, M1 and M2, with
cytes, negatively correlated with preservation of left ventric- similar behavior as Gr1high and Gr1low monocytes, respec-
ular function (36). Indeed, inhibiting CCL2 with a neutral- tively (48). However, it is not known whether the differen-
izing antibody or competitive peptide leads to pathological tiation is determined by monocyte maturation or later
defects in infarcted mice, such as decreased macrophage macrophage polarization.
infiltration, delayed replacement of dead cardiomyocytes Because the appropriate blood supply is crucial for heart
with granulation tissue, and decreased myofibroblasts accu- survival and function, angiogenesis is considered an optimal
mulation (37,38). However, delayed phagocytosis of injured therapeutic target. VEGF and CXCL12 control and mod-
cardiomyocytes may increase the arrhythmogenic potential ulate the recruitment of endothelial progenitor cells (EPCs)
or predisposition to mechanical complications, such as heart and proper formation of blood vessels (49). Although this
rupture (37). Therefore, the potential use of CCL2 as a occurs continuously, EPCs are not continuously recruited.
clinical therapeutic target should be carefully scrutinized. Earlier transplantation of EPCs seems to have maximal
In conclusion, until proven otherwise, short and limited beneficial effects, whereas later systemic transplantation of
presence of neutrophils is needed to initiate inflammatory EPCs failed to influence cardiac remodeling and function.
JACC Vol. 58, No. 23, 2011 Liehn et al. 2359
November 29, 2011:2357–62 Chemokines in Cardiac Repair

Figure 1 Cellular and Molecular Events After MI

The healing after myocardial infarction involves 3 overlapping phases: inflammatory, proliferative, and healing phases. Each is characterized by specific events (red),
implicating different cells, controlled by specific chemokines (blue). Extracellular matrix (ECM) evolves to mature scar (dark blue), which ensures the stability and the
function of the heart. HIF ⫽ hypoxic inducible factor; IL ⫽ interleukin; MCP ⫽ monocyte chemotactic protein; MI ⫽ myocardial infarction; MIF ⫽ macrophage inhibitory
factor; MSC ⫽ mesenchymal stem cell; NF-kB ⫽ nuclear factor kappa B; SDF ⫽ stromal-derived factor; TGF ⫽ transforming growth factor; TNF ⫽ tumor necrosis factor.

Therefore, angiogenesis should be supported by the survival After infarction, angiostatic factors, such as CXCL10, are
and proliferation of EPCs shortly recruited after infarction up-regulated, inhibiting angiogenesis until the myocardium
and of local stem cells, differentiated towards an endothelial is cleaned of cell debris and a provisional matrix is formed
phenotype. A recent study highlights the important role of (1,51). Along with suppressing cytokines and chemokines of
MI-induced epicardium-derived cells in angiogenesis by the inflammatory phase, TGF-␤ suppresses the expression
secreting paracrine factors (50). However, these cells are not of CXCL10 and allows angiogenic factors, such as
able to differentiate into endothelial cells or cardiomyocytes, CXCL12, CXCL1 (52), CXCL2 (53), MIF (54,55), and
but into MSCs, and seem to offer cardiac protection. even CCL2 (56), to exert their function (1,51).
2360 Liehn et al. JACC Vol. 58, No. 23, 2011
Chemokines in Cardiac Repair November 29, 2011:2357–62

Another player is recruited lymphocytes, which synthe- decreased ventricular function (69), inhibition of myofibro-
size IL-10 and tissue inhibitor of metalloproteinase (TIMP)-1, blast is considered an appropriate therapeutic goal. Our data
modulating the mononuclear cell phenotype and T helper 2 have shown that increased collagen content even supports
polarization, in a CCL2-dependent manner (57). Recently, contractility and preserves heart function (23,70). However,
it was shown that CCR5-mediated regulatory T cell recruit- using noninvasive methods to visualize the interstitial
ment reduced post-infarction inflammation, preventing ex- changes in vivo (71) should enable us to monitor the
cessive matrix degradation and attenuating adverse remod- remodeling and signal specific changes.
eling (58). Although lymphocytes seem to have important To date, we know that mature scar is a dynamic tissue,
roles during healing after MI, there are still many unknowns vascularized and metabolically active. Experimental cell
that have to be clarified. transplantation in mature scar induces cell-specific changes,
Fibroblasts produce extracellular matrix and provide me- improving ventricular function (65,70,72,73), the same
chanical support for the newly formed structures (1,51). effect being observed also by biologically inactive glass
Resident, as well as recruited, fibroblast progenitors (59) microsphere transplantation. This leads to the conclusion
activate and differentiate into myofibroblasts under TGF-␤ that the underlying mechanisms might be based on inflam-
(60) and CCL2 (61) stimulation, developing morphological matory processes induced by transplantation (73). This
and contractile characteristics of smooth muscle cells. The could represent a potential therapeutic strategy in patients
synthesized collagen together with proteoglycans and glyco- with extended scar tissue, for whom all current clinical
proteins constitute the extracellular matrix (62). CD44, treatment alternatives have been exhausted.
expressed on all implicated cells, is critically involved in the Strategies for clinical settings of the new therapies. Be-
regulation of the fibroblast function (63). side cell therapy, manipulation of different biological pathways
Although the original cardiac matrix network is degraded in the heart by gene therapy, antibodies, peptides, silencing
by metalloproteinases and cleared together with cellular RNA, or microRNA seems to represent other promising
debris, extravasation of plasma proteins results in a complex approaches to heart failure treatment. However, despite the
and dynamic matrix network based on fibrin and fibronec- evident effects in experimental models, the clinical application
tin, serving as a scaffold for migration and proliferation of requires extensive research and intensive discussion.
the cells (64). Once the inflammatory phase is repressed, Since the systemic application of drugs would have
and myofibroblasts proliferate, this fibrin-based provisional uncontrolled adverse effects, local delivery is necessary. For
matrix is gradually replaced by collagen. Specific proteins, example, the intracoronary transfer of viral vectors (74) or
such as thrombospondin-1 and -2, osteopontin, tenascin C, Hemagglutinating Virus of Japan liposome (75) carrying a
periostin, and SPARC, regulate the cellular interaction, gene proved to be very simple and less invasive. Likewise,
induce cross-linking, and organize the mature matrix (64). intracoronary gene-containing microbubbles followed by
Another important aspect of the proliferative phase is ultrasonic destruction at the desired position may represent
cardiomyocyte regeneration after injury. The major goal of a feasible clinic setting (76). The same strategies can be
cellular therapies is to restore the initial integrity of the heart applied when using stimulating or blocking peptides, anti-
tissue. Transplanted fetal cardiomyocytes integrate between bodies, or other control tools for gene transcription, such as
the existent cells and improve contractile heart function silencing RNA or microRNA. On the other hand, due to
(65). However, it would be a real challenge, not only the unpredictable progress of the events, it is not possible to
methodically, but also ethically, to obtain these cells for a implement these strategies in any clinical settings.
future clinical application. Therefore, other cells, such as Supreme control: fine-tuning the chemokine. Despite
MSCs, are broadly used as a source for stem cell therapy extensive research, our current knowledge regarding the
after MI (9,66). It seems that CXCL12 (67) and CCL7 control of molecular and cellular events after MI is still
(68) chemokines control myocardial homing of MSC after limited. Therefore, to be able to create efficient therapies,
injury, which engraft and differentiate into cardiomyocytes, we need to understand how all these processes are modu-
or fuse partially/permanently with neighbor cells, transfer- lated and controlled. Chemokines and chemokine receptors
ring genetic and mitochondrial material (6). Unfortunately, have the most precise functions in leukocyte trafficking,
despite extensive research in the last few years, the cell- angiogenesis, collagen synthesis, and cardiac repairing. Ma-
mediated regenerative mechanisms remain elusive, making nipulating them, by interfering with receptor–ligand inter-
their clinical implementation difficult. action, chemokine– glycosaminoglycan interaction, hetero-
Healing phase: “dead” or “living” scar? In the last stage, merization, or signaling pathways induced upon receptor
the scar contains a dense collagen-based extracellular matrix, activation (77), should enable us to control and modify
myofibroblasts, stem cells, and newly formed vessels. Un- molecular and cellular events (Table 1, Online Table 1).
known mechanisms activate apoptotic pathways in myofi-
broblasts and vascular cells, and determine the loss of most Conclusions
cellular components (64). The mature infarction scar was for
a long time considered inert, incapable of biological func- Understanding the basic mechanisms involved in MI is crucial
tion. Since increased collagen content was associated with for the development of new strategies to reduce injury and
JACC Vol. 58, No. 23, 2011 Liehn et al. 2361
November 29, 2011:2357–62 Chemokines in Cardiac Repair

ischemic-induced apoptosis in a murine model of acute myocardial


Potential
Disadvantages
Potential Advantages Advantages
of Targeting
and and
Chemokines infarction. Proc Natl Acad Sci U S A 2000;97:5456 – 61.
Table 1
Disadvantages of Targeting Chemokines 13. Saxena A, Fish JE, White MD, et al. Stromal cell-derived factor-
1alpha is cardioprotective after myocardial infarction. Circulation
Advantages Disadvantages
2008;117:2224 –31.
Precise and differentiated actions on 14. Miller EJ, Li J, Leng L, et al. Macrophage migration inhibitory factor
leukocyte subsets and stem cells stimulates AMP-activated protein kinase in the ischaemic heart.
Precise and differentiated actions on Concomitant actions are Nature 2008;451:578 – 82.
angiogenesis, collagen synthesis, poorly studied 15. Tarzami ST, Cheng R, Miao W, et al. Chemokine expression in
and cardiac remodeling myocardial ischemia: MIP-2 dependent MCP-1 expression protects
Specific and selective agonists and Price cardiomyocytes from cell death. J Mol Cell Cardiol 2002;34:209 –21.
antagonists available 16. Loor G, Schumacker PT. Role of hypoxia-inducible factor in cell
Local or systemic applications Interference with other organs survival during myocardial ischemia-reperfusion. Cell Death Differ
by systemic applications 2008;15:686 –90.
(partly disadvantageous) 17. Shohet RV, Garcia JA. Keeping the engine primed: HIF factors as key
regulators of cardiac metabolism and angiogenesis during ischemia. J
Very short action: side effects rapidly reversible Sometimes repetitive
Mol Med 2007;85:1309 –15.
application is needed
18. Arslan F, de Kleijn DP, Pasterkamp G. Innate immune signaling in
Easy combination with other carrying systems cardiac ischemia. Nat Rev Cardiol 2011;8:292–300.
(liposomes, microbubbles, nanoparticles) 19. Erridge C. The roles of pathogen-associated molecular patterns in
atherosclerosis. Trends Cardiovasc Med 2008;18:52– 6.
20. McNeill E, Channon KM, Greaves DR. Inflammatory cell recruit-
promote repair, which together with the classical interventions ment in cardiovascular disease: murine models and potential clinical
should improve the left ventricular remodeling, the function of applications. Clin Sci (Lond) 2010;118:641–55.
21. Jaeschke H, Smith CW. Mechanisms of neutrophil-induced paren-
the heart, and the prognosis of a patient after MI. chymal cell injury. J Leukoc Biol 1997;61:647–53.
22. Jolly SR, Kane WJ, Hook BG, et al. Reduction of myocardial infarct
Acknowledgment size by neutrophil depletion: effect of duration of occlusion. Am
Heart J 1986;112:682–90.
The authors want to thank Dr. O. Bucur for critical review 23. Liehn EA, Merx MW, Postea O, et al. Ccr1 deficiency reduces
of the manuscript. inflammatory remodelling and preserves left ventricular function after
myocardial infarction. J Cell Mol Med 2008;12:496 –506.
24. Griselli M, Herbert J, Hutchinson WL, et al. C-reactive protein and
Reprint requests and correspondence: Dr. Elisa A. Liehn, complement are important mediators of tissue damage in acute
Department of Cardiology, Pneumology, Angiology and Internal myocardial infarction. J Exp Med 1999;190:1733– 40.
Medicine Intensive Care (Internal Medicine I), University Hos- 25. Chen Z, Siu B, Ho YS, et al. Overexpression of MnSOD protects
pital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074 against myocardial ischemia/reperfusion injury in transgenic mice. J
Mol Cell Cardiol 1998;30:2281–9.
Aachen, Germany. E-mail: eliehn@ukaachen.de. 26. Izumi T, Saito Y, Kishimoto I, et al. Blockade of the natriuretic
peptide receptor guanylyl cyclase-A inhibits NF-kappaB activation
and alleviates myocardial ischemia/reperfusion injury. J Clin Invest
REFERENCES 2001;108:203–13.
27. Braunersreuther V, Pellieux C, Pelli G, et al. Chemokine CCL5/
1. Frangogiannis NG, Smith CW, Entman ML. The inflammatory RANTES inhibition reduces myocardial reperfusion injury in athero-
response in myocardial infarction. Cardiovasc Res 2002;53:31– 47. sclerotic mice. J Mol Cell Cardiol 2010;48:789 –98.
2. Marx N, Neumann FJ, Ott I, et al. Induction of cytokine expression in
28. Soehnlein O, Zernecke A, Weber C. Neutrophils launch monocyte
leukocytes in acute myocardial infarction. J Am Coll Cardiol 1997;30:165–70.
extravasation by release of granule proteins. Thromb Haemost 2009;
3. Liehn EA, Zernecke A, Postea O, Weber C. Chemokines: inflammatory
102:198 –205.
mediators of atherosclerosis. Arch Physiol Biochem 2006;112:229–38.
29. Swirski FK, Nahrendorf M, Etzrodt M, et al. Identification of splenic
4. Frangogiannis NG. Chemokines in ischemia and reperfusion. Thromb
reservoir monocytes and their deployment to inflammatory sites.
Haemost 2007;97:738 – 47.
Science 2009;325:612– 6.
5. Bergmann O, Bhardwaj RD, Bernard S, et al. Evidence for cardio-
myocyte renewal in humans. Science 2009;324:98 –102. 30. Leuschner F, Panizzi P, Chico-Calero I, et al. Angiotensin-converting
6. Acquistapace A, Bru T, Lesault PF, et al. Human mesenchymal stem enzyme inhibition prevents the release of monocytes from their splenic
cells reprogram adult cardiomyocytes towards a progenitor-like state reservoir in mice with myocardial infarction. Circ Res 2010;107:1364–73.
through partial cell fusion and mitochondria transfer. Stem Cells 31. Nahrendorf M, Swirski FK, Aikawa E, et al. The healing myocardium
2011;29:812–24. sequentially mobilizes two monocyte subsets with divergent and
7. Tallini YN, Greene KS, Craven M, et al. c-kit expression identifies complementary functions. J Exp Med 2007;204:3037– 47.
cardiovascular precursors in the neonatal heart. Proc Natl Acad Sci 32. Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations
U S A 2009;106:1808 –13. and their differentiation patterns during infection. J Leukoc Biol
8. Popescu LM, Faussone-Pellegrini MS. TELOCYTES—a case of 2007;82:244 –52.
serendipity: the winding way from Interstitial Cells of Cajal (ICC), via 33. Nahrendorf M, Pittet MJ, Swirski FK. Monocytes: protagonists of
Interstitial Cajal-Like Cells (ICLC) to TELOCYTES. J Cell Mol infarct inflammation and repair after myocardial infarction. Circula-
Med, 14:729 – 40. tion 2010;121:2437– 45.
9. Choi YH, Kurtz A, Stamm C. Mesenchymal stem cells for cardiac cell 34. Soehnlein O, Zernecke A, Eriksson EE, et al. Neutrophil secretion
therapy. Hum Gene Ther 2011;22:3–17. products pave the way for inflammatory monocytes. Blood 2008;112:
10. Phinney DG, Prockop DJ. Concise review: mesenchymal stem/ 1461–71.
multipotent stromal cells: the state of transdifferentiation and modes of 35. Panizzi P, Swirski FK, Figueiredo JL, et al. Impaired infarct healing in
tissue repair— current views. Stem Cells 2007;25:2896 –902. atherosclerotic mice with Ly-6C(hi) monocytosis. J Am Coll Cardiol
11. Bani D, Formigli L, Gherghiceanu M, Faussone-Pellegrini MS. 2010;55:1629 –38.
Telocytes as supporting cells for myocardial tissue organization in 36. Tsujioka H, Imanishi T, Ikejima H, et al. Impact of heterogeneity of
developing and adult heart. J Cell Mol Med;14:2531– 8. human peripheral blood monocyte subsets on myocardial salvage in
12. Kurrelmeyer KM, Michael LH, Baumgarten G, et al. Endogenous patients with primary acute myocardial infarction. J Am Coll Cardiol
tumor necrosis factor protects the adult cardiac myocyte against 2009;54:130 – 8.
2362 Liehn et al. JACC Vol. 58, No. 23, 2011
Chemokines in Cardiac Repair November 29, 2011:2357–62

37. Dewald O, Zymek P, Winkelmann K, et al. CCL2/Monocyte 59. Fujita J, Mori M, Kawada H, et al. Administration of granulocyte
Chemoattractant Protein-1 regulates inflammatory responses critical colony-stimulating factor after myocardial infarction enhances the
to healing myocardial infarcts. Circ Res 2005;96:881–9. recruitment of hematopoietic stem cell-derived myofibroblasts and
38. Liehn EA, Piccinini AM, Koenen RR, et al. A new monocyte contributes to cardiac repair. Stem Cells 2007;25:2750 –9.
chemotactic protein-1/chemokine CC motif ligand-2 competitor lim- 60. Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. Transforming
iting neointima formation and myocardial ischemia/reperfusion injury growth factor-beta 1 induces alpha-smooth muscle actin expression in
in mice. J Am Coll Cardiol 2010;56:1847–57. granulation tissue myofibroblasts and in quiescent and growing cul-
39. Tekin D, Dursun AD, Xi L. Hypoxia inducible factor 1 (HIF-1) and tured fibroblasts. J Cell Biol 1993;122:103–11.
cardioprotection. Acta Pharmacol Sin 2010;31:1085–94. 61. Morimoto H, Takahashi M, Izawa A, et al. Cardiac overexpression of
40. Lenardo MJ, Baltimore D. NF-kappa B: a pleiotropic mediator of monocyte chemoattractant protein-1 in transgenic mice prevents
inducible and tissue-specific gene control. Cell 1989;58:227–9. cardiac dysfunction and remodeling after myocardial infarction. Circ
41. Loh SA, Chang EI, Galvez MG, et al. SDF-1 alpha expression during Res 2006;99:891–9.
wound healing in the aged is HIF dependent. Plast Reconstr Surg 62. Lindsey ML, Mann DL, Entman ML, Spinale FG. Extracellular matrix
2009;123:65S–75S. remodeling following myocardial injury. Ann Med 2003;35:316–26.
42. Fu H, Luo F, Yang L, et al. Hypoxia stimulates the expression of 63. Huebener P, Abou-Khamis T, Zymek P, et al. CD44 is critically
macrophage migration inhibitory factor in human vascular smooth involved in infarct healing by regulating the inflammatory and fibrotic
muscle cells via HIF-1alpha dependent pathway. BMC Cell Biol response. J Immunol 2008;180:2625–33.
2010;11:66. 64. Dobaczewski M, Gonzalez-Quesada C, Frangogiannis NG. The
43. Heather LC, Clarke K. Metabolism, hypoxia and the diabetic heart. J extracellular matrix as a modulator of the inflammatory and reparative
Mol Cell Cardiol 2011;50:598 – 605. response following myocardial infarction. J Mol Cell Cardiol;48:504–11.
44. Gao XM, Xu Q, Kiriazis H, et al. Mouse model of post-infarct 65. Skobel E, Schuh A, Schwarz ER, et al. Transplantation of fetal
ventricular rupture: time course, strain- and gender-dependency, cardiomyocytes into infarcted rat hearts results in long-term functional
tensile strength, and histopathology. Cardiovasc Res 2005;65:469 –77. improvement. Tissue Eng 2004;10:849 – 64.
45. Zymek P, Nah DY, Bujak M, et al. Interleukin-10 is not a critical 66. Hatzistergos KE, Quevedo H, Oskouei BN, et al. Bone marrow
regulator of infarct healing and left ventricular remodeling. Cardiovasc mesenchymal stem cells stimulate cardiac stem cell proliferation and
Res 2007;74:313–22. differentiation. Circ Res 2010;107:913–22.
46. Auffray C, Fogg D, Garfa M, et al. Monitoring of blood vessels and 67. Zhang M, Mal N, Kiedrowski M, et al. SDF-1 expression by
tissues by a population of monocytes with patrolling behavior. Science mesenchymal stem cells results in trophic support of cardiac myocytes
2007;317:666 –70. after myocardial infarction. FASEB J 2007;21:3197–207.
47. Tacke F, Alvarez D, Kaplan TJ, et al. Monocyte subsets differentially 68. Schenk S, Mal N, Finan A, et al. Monocyte chemotactic protein-3 is
employ CCR2, CCR5, and CX3CR1 to accumulate within athero- a myocardial mesenchymal stem cell homing factor. Stem Cells
sclerotic plaques. J Clin Invest 2007;117:185–94. 2007;25:245–51.
48. Geissmann F, Auffray C, Palframan R, et al. Blood monocytes: 69. Frangogiannis NG, Dewald O, Xia Y, et al. Critical role of monocyte
distinct subsets, how they relate to dendritic cells, and their possible chemoattractant protein-1/CC chemokine ligand 2 in the pathogen-
roles in the regulation of T-cell responses. Immunol Cell Biol esis of ischemic cardiomyopathy. Circulation 2007;115:584 –92.
2008;86:398 – 408. 70. Schuh A, Liehn EA, Sasse A, et al. Transplantation of endothelial
49. Abbott JD, Huang Y, Liu D, et al. Stromal cell-derived factor-1alpha progenitor cells improves neovascularization and left ventricular func-
plays a critical role in stem cell recruitment to the heart after tion after myocardial infarction in a rat model. Basic Res Cardiol
myocardial infarction but is not sufficient to induce homing in the 2008;103:69 –77.
absence of injury. Circulation 2004;110:3300 –5. 71. van den Borne SW, Isobe S, Verjans JW, et al. Molecular imaging of
50. Zhou B, Honor LB, He H, et al. Adult mouse epicardium modulates interstitial alterations in remodeling myocardium after myocardial
myocardial injury by secreting paracrine factors. J Clin Invest 2011; infarction. J Am Coll Cardiol 2008;52:2017–28.
121:1894 –904. 72. Merx MW, Zernecke A, Liehn EA, et al. Transplantation of human
51. Frangogiannis NG, Mendoza LH, Smith CW, et al. Induction of the umbilical vein endothelial cells improves left ventricular function in a
synthesis of the C-X-C chemokine interferon-gamma-inducible rat model of myocardial infarction. Basic Res Cardiol 2005;100:208–16.
protein-10 in experimental canine endotoxemia. Cell Tissue Res 73. Schuh A, Liehn EA, Sasse A, et al. Improved left ventricular function
2000;302:365–76. after transplantation of microspheres and fibroblasts in a rat model of
52. Liehn EA, Schober A, Weber C. Blockade of keratinocyte-derived myocardial infarction. Basic Res Cardiol 2009;104:403–11.
chemokine inhibits endothelial recovery and enhances plaque forma- 74. Ishikawa K, Ladage D, Tilemann L, Fish K, Kawase Y, Hajjar RJ.
tion after arterial injury in ApoE-deficient mice. Arterioscler Thromb Gene transfer for ischemic heart failure in a preclinical model. J Vis
Vasc Biol 2004;24:1891– 6. Exp 2011;(51). pii: 2778.
53. Strieter RM, Polverini PJ, Kunkel SL, et al. The functional role of the 75. Aoki M, Morishita R, Muraishi A, et al. Efficient in vivo gene transfer
ELR motif in CXC chemokine-mediated angiogenesis. J Biol Chem into the heart in the rat myocardial infarction model using the HVJ
1995;270:27348 –57. (Hemagglutinating Virus of Japan)—liposome method. J Mol Cell
54. Simons D, Grieb G, Hristov M, et al. Hypoxia-induced endothelial Cardiol 1997;29:949 –59.
secretion of macrophage migration inhibitory factor and role in 76. Kondo I, Ohmori K, Oshita A, et al. Treatment of acute myocardial
endothelial progenitor cell recruitment. J Cell Mol Med 2010;15:668 –78. infarction by hepatocyte growth factor gene transfer: the first demon-
55. Weber C, Kraemer S, Drechsler M, et al. Structural determinants of stration of myocardial transfer of a ”functional” gene using ultrasonic
MIF functions in CXCR2-mediated inflammatory and atherogenic microbubble destruction. J Am Coll Cardiol 2004;44:644 –53.
leukocyte recruitment. Proc Natl Acad Sci U S A 2008;105:16278 – 83. 77. Proudfoot AE, Power CA, Rommel C, Wells TN. Strategies for chemokine
56. Weber KS, Nelson PJ, Grone HJ, Weber C. Expression of CCR2 by antagonists as therapeutics. Semin Immunol 2003;15:57–65.
endothelial cells : implications for MCP-1 mediated wound injury
repair and In vivo inflammatory activation of endothelium. Arterio- Key Words: chemokine y inflammation y myocardial infarction y
scler Thromb Vasc Biol 1999;19:2085–93. remodeling.
57. Gu L, Tseng S, Horner RM, et al. Control of TH2 polarization by the
chemokine monocyte chemoattractant protein-1. Nature 2000;404:407–11. APPENDIX
58. Dobaczewski M, Xia Y, Bujak M, et al. CCR5 signaling suppresses
inflammation and reduces adverse remodeling of the infarcted heart, For supplementary material and a table,
mediating recruitment of regulatory T cells. Am J Pathol;176:2177–87. please see the online version of this paper.

You might also like