Skip to main content
International Journal of Clinical and Experimental Medicine logoLink to International Journal of Clinical and Experimental Medicine
. 2011 Feb 6;4(1):74–80.

Chemokines and inflammation in heart disease: adaptive or maladaptive?

Sima T Tarzami 1
PMCID: PMC3048986  PMID: 21394288

Abstract

Heart disease is not only the leading cause of death, disability, and healthcare expense in the US, but also the leading cause of death worldwide. Therefore, treatments to lessen ischemia-related cardiac damage could affect a broad swath of the population and have significant health and fiscal impacts. Cardiac dysfunction has been associated with elevated circulating chemokine levels, both in animals and humans. Most studies in this area have focused on chemokine expression as a prominent feature of the post-infarction inflammatory response. Such studies have investigated the role of chemokines in inflammatory leukocyte recruitment. Other work on this topic has focused on stem-cell therapy or factors e.g. chemokines mobilizing bone marrow progenitor cells as possible avenues for improving contractile dysfunction. Findings from numerous preclinical studies and several initial clinical trials support the feasibility of promoting the recruitment of bone marrow-derived cells to the infarcted heart and increased homing following injury, supporting the notion that cell therapy might have therapeutic potential. They have not, however, addressed the possibility of an autocrine/paracrine effect wherein the chemokine receptors, present on the cardiac myocyte surface, modulate functional responses to stress in which can be adaptive or maladaptive in nature.

Keywords: Chemokines, stem-cell therapy, inflammation, myocardial infarction, congestive heart failure

Introduction

Coronary artery disease (CAD) is a leading cause of death in the United States. Acute myocardial infarction (MI) is a catastrophic manifestation of CAD that strikes nearly one million Americans each year. One fourth of afflicted patients die and many survivors develop impaired functional status, anginal symptoms, and diminished quality of life [1, 2]. The mainstay of current therapy in acute MI is the restoration of blood flow (reperfusion) to the affected area via thrombolytic therapy or an-gioplasty. Many studies have demonstrated clinical benefit from early reperfusion [3, 4]. However, reperfusion is itself associated with an inflammatory response and cardiac myocyte damage, called ischemia-reperfusion (I/R) injury. Thus, while the net result of early reperfusion is beneficial, an in-depth understanding of the complexity of the myocardial inflammatory response, including a determination of the cell type involved, is required to optimize current treatment strategies and to develop better ones. In addition, cardiac myocytes themselves may play a key role in the reperfusion process, as suggested by recent studies. For example, some work suggests that cardiac myocytes can both generate and respond to inflammatory responses, in an autocrine or paracrine manner, by modulating their contractile responses [5, 6] . We will describe the documented or hypothesized role of cytokines, in particular, chemokines, and their potential therapeutic role in the various patho-genesis steps of heart failure.

Cytokines, inflammation, and ischemic reperfusion injury

MI is associated with an inflammatory reaction, which leads to healing and scar formation [7]. Although inflammation can increase myocardial injury, an inflammatory response is a prerequisite for cardiac healing and scar formation.

However, chronic inflammation can lead to additional cell death and myocardial remodeling. Therefore, the extent of the inflammatory response may, in turn, be an important determinant of the host's outcome [8,9]. One of the major therapeutic goals is to design strategies aimed at minimizing myocardial necrosis and optimizing cardiac repair following myocardial infarction. However, attempts to completely limit inflammatory response, for example with the use steroids, have failed to reverse cardiac dysfunction [10]. Cell-specific anti-inflammatory therapies have been more promising. Several studies have shown the benefits of neutrophil inhibition [11, 12], presumably through inhibiting reactive oxygen species (ROS) and endothe-lial activation [13]. Mast cells have also been implicated as a source of cytokines, notably an immunologically active pre-formed tumor necrosis factor (TNF)-α [14]. In clinical studies, there have been discrepancies as to the relationship between serum cytokine levels in patients with MI and clinical outcome. Several small studies have demonstrated a positive correlation between serum cytokine levels, such as TNF [15], and interleukin (IL)-6 [16-18] and the size and severity of the MI. Other studies have not supported a significant correlation between cytokine levels and MI severity [19, 20].

Yet with all the experimental evidentiary support for the importance of leukocyte infiltration in the initiation of the inflammatory cascade that results in myocardial ischemic injury, clinical data is lacking. There are large numbers of animal studies with various approaches to blocking neutrophils using anti-CD18, a key signaling receptor in lymphocyte adhesion and a very efficient neutrophils paralyzing agent, has showed benefit in animal models of reperfusion injury. However, Genentech's phase II clinical trials to treat acute MI using anti-CD18 monoclonal antibodies, have failed to yield clinical benefits [21]. This is similar to what was observed in the heart failure trials that used “targeted” approaches to neutralize cytokines such as TNF in patients with heart failure [22]. Two trial programmes (the RENAISSANCE and RECOVER trials) testing an anti-cytokine medication in chronic heart failure (CHF) were halted because these targeted approaches resulted in worsening clinical outcomes, thereby raising important questions about what role, if any, proinflamma-tory cytokines play in the pathogenesis of heart failure [23, 24]. Recent studies have implicated a previously unexplored class of receptors, namely chemokine receptors, as direct mediators of myocardial function and potentiators of the cardiac myocytes response to ischemic stress [5, 6]. Chemokines are member of the cytokines family; however, given the substantial structural and functional differences between cytokine receptors and chemokine receptors, it is likely that modulation of chemokine receptors will have different effects than those seen with cytokines and their receptors.

Chemokines and chemokine receptors

Chemokines are small glycoproteins that stimulate leukocyte migration to sites of an inflammatory or immune response. Four chemokine subfamilies (CXC, CC, C and CX3C) are classified based on their primary amino acid sequences [25]. Chemokines function as ligands for G protein-coupled receptors (GPCR), which are seven transmembrane-spanning receptor proteins. Chemokine receptors differ both structurally and functionally from their cytokine receptor cousins such as TNFR and IL-2R. Cytokine receptors typically act via “horizontal” receptor signal transduction, wherein receptor molecules diffuse within the membrane plane. Binding of cytokines alters the association between receptor molecules that often cross the membrane only once in a single alpha-helical segment; consequently, the association of protein domains inside the membrane is altered as well. Signals thus are transduced by changes in oligomeric state of the receptors. These receptors, as a rule, exist in an “off-state” before activation [26].

In contrast, chemokine receptors function as “vertical” receptors [27] wherein the initial agonist-receptor interaction changes the receptor conformation transmitted vertically through the membrane. These receptors often respond to small ligands that cause a transient and reversible change in membrane potential or in cellular metabolism. The preassembled and generally unimolecular nature of these receptors allows them to respond rapidly. The different nature at many levels of cytokine and chemokine receptors suggests strongly that their functions in the cardiac response to inflammation are likely to be quite different. Therefore, the investigation of cardiomyocytes chemokine receptors is likely to yield novel insights into the myocardial inflammatory response.

Chemokines and heart disease

Cardiac dysfunction as been associated with elevated circulating chemokine levels in both animals and humans [28-30]. One of the first chemokines to be detected in the myocardium in response to I/R injury was Interleukin-8 (IL-8), a CXC chemokine, (CXCL8) [31]. It was hypothesized that IL-8 participates in the neutrophil-induced myocardial injury. Several other chemokines have been detected early in the hearts in animal models of injury [32]. These include CCL2 (Chemokine (C-C motif) ligand 2; also known as monocyte chemoattractant protein (MCP-1)), and CXCL10 (also known as Inter-feron-γ inducible Protein-10 (IP-10)). Almost all such chemokine studies have focused on their expression as a prominent feature of the post-infarction inflammatory response and therefore considered just the chemokines’ role in inflammatory leukocyte recruitment [8]. These studies have not addressed possible autocrine/ paracrine effects wherein the ligand-activated chemokine receptors, present on the cardiac myocyte surface, modulate functional responses to stress in the myocytes themselves.

The identification of functional chemokine receptors on cardiac mycoytes opened up a new line of research bridging the areas of immunology (i.e. chemokines) and cardiovascular biology. Once thought to be present only on inflammatory cells, chemokine receptors are now recognized biological effectors on cardiac myocytes as well. Among the chemokine receptors found on cardiac myocytes are macrophage inflammatory protein-2 (MIP-2) receptor, CXCR2, monocyte chemeoattractant protein -1 (MCP-1) receptor, CCR2, and most notably stromal derived factor-1 (SDF-1 also known as CXCL12) recep-tor,CXCR4 [6, 33]. They are for the most part constitutively continuously expressed but are upregulated following oxidative stress, both in vivo and in vitro. Furthermore, MIP-2 protein appears to induce the expression of MCP-1, a putative protector cardiac myocytes from hy-poxia-induced apoptosis in vitro. It is noteworthy that in an animal model of I/R injury, the protective effect of MCP-1 was masked as a result of the recruitment of inflammatory cells in this model [33-35]. This is an example of how direct actions of chemokines on cardiac cells have been missed by the recruitment of inflammatory cells as seen in this model.

More recently, several groups have studied the effects of SDF-1-CXCR4 [36-40] in injury models. CXCR4 over-expression in vivo worsened cardiac function in an animal model of I/R injury. This deleterious effect of CXCR4 over-expression in vivo following I/R may result in part from the observed increase in production of its ligand (SDF-1). Over-expression of myocardial CXCR4 also leads to enhanced recruitment of inflammatory cells, increased TNF-α production and cell death/apoptosis [5]. These studies suggest that under conditions of stress, the paracrine and/or autocrine activation of CXCR4 by its ligand may trigger intracellular signaling pathways that exacerbate contractile dysfunction. One such pathway that may be affected is the beta-adrenergic signaling system. LaRocca et al. recently showed a clear interaction between CXCR4 and Beta-2 adrenergic receptor and that CXCR4 activation can interfere with Beta-adrenergic receptor downstream signaling [41]. Beta-adrenergic signaling plays a central role in the neuro-humoral regulation of the heart and the progression of heart failure. Given the central role of Beta-adrenergic signaling in progression of heart failure, further studies to unmask the potentials of CXCR4 in modulating disease pathophysiology are underway.

Chemokines and cell therapy

One potentially promising strategy for repairing damaged cardiac tissue following MI involves the use of cell therapy [42-46] to prevent or reverse heart failure [47]. Efforts in this area began with studies using already committed cells such as skeletal myoblasts [48], but recently a diverse range of cell types have been tested, including bone marrow (BM)-derived hematopoietic cells [49, 50], EPCs [44], mesen-chymal stem cells (MSCs) [51], resident cardiac stem cells [43, 52], and embryonic stem cells. (ESCs) [53]. A related strategy for cardiac repair involves the testing of cell mobilization with factors such as cytokines—e.g., with granulocyte-colony stimulating factor (G-CSF) (the MAGIC cell randomized clinical trial) [54]. Several laboratories have shown that the cells can be mobilized and can home to areas of injury, in part in response to SDF-1/CXCR4 interaction [55] .

Given SDF-1's essential role in BM homing and recruitment [45,56], SDF-1 has been tested in gene therapy as a recruiter of BM-derived SC to the myocardium, in a murine infarct model [57-59]. SDF-1, acting through, CXCR4, did indeed recruit BM-derived cells to the injury, supporting the notion that this chemokine might have therapeutic potential. Several clinical trials in this area have been recently been completed or are still ongoing. For example, a human trial -the REGENT trial - was designed to assess the value of infusing either selected CXCR4-positive stem cells or non-selected BM mononuclear cells (BM MNC) in myocardial regeneration [60]. The objective of REGENT was to determine the effect of intracoronary infusion of selected population of autologous BM-derivedCD34+CXCR4+ progenitor cells compared with non-selected BM MNC on LVfunction in patients with acute ST-segment elevation MI and reduced left ventricular ejection fraction (LVEF). In patients with acute MI who developed impairment of the LVEF despite timely and successful treatment with primary percutaneous coronary intervention (PCI), treatment with BMCs (neither selected nor non-selected) did not led to a significant improvement of LVEF. Another phase 1 trial, which is currently underway, is examining the effects of SDF-1 on infarc-ted hearts. In this study, the ligand is injected directly into the heart and then monitoring homing of BMCs into the myocardium of patients with ischemic heart disease. Also noteworthy is an open-label dose-escalation study to evaluate the safety of a single escalating dose of SDF-1, administered by endomyocardial injection to cohorts of adults with ischemic heart failure, which is currently enrolling patients. In this study, naked DNA that encodes SDF-1 is being injected directly into the myocardium as a single dose at multiple sites through a percutaneous, left ventricular approach, using a needle injection catheter (ClinicalTrials.gov identifier: NCT01082094). These on going trials will demonstrate the safety and efficacy of using chemokines e.g. SDF-1 in SC therapies to treat heart failure in subjects with ischemic cardio-myopathy.

Open questions

Our understanding of the role of the SDF-1/CXCR4 receptor system in the heart failure has been hampered, however, by the lackof information about the cardiac myocytes expression/function of CXCR4 and SDF-1 during the various pathogenesis steps of heart failure. The cellular expression patterns of SDF-1 and the CXCR4 receptor can both generate inflammatory responses, in an autocrine or paracrine manner, and effect the cardiac myocytes adaptive signaling mechanisms e.g. contractile responses after ischemia. However, mostif not all of these approaches have additional direct effects on the homing of BMCs into the myocardium of patients with ischemic heart disease and it is difficult to determine the beneficial contribution of chemokines cell mobilization from those associated with inflammation and/or contractile dysfunction. Based on the possible pathogenic role of chemokines in the development and progression of heart failure, many investigators have focused on myocardial gene expression and cellular localization of chemokines and their corresponding receptors in various stages of heart failure.

In summary, chemokine receptors such as CXCR4 belong to the GPCR family, and like other members of this family, are accessible targets for drug development [61,62]. CXCR4, like many GPCRs, exhibits heterogeneity of expression in different tissues and cell types, thus providing some selectivity (in some cases, specificity) in the targeting of these receptors for the activation or blockade of physiological events [63]. As a class, GPCRs have been the most successful molecular drug targets in clinical medicine. Agonists and antagonists of GPCRs are used to treat diseases of every major organ system including the CNS and cardiovascular systems. Additionally, the discovery of functional chemokine receptors on cardiac myocyte has broad implications for optimization of preexisting chemokine/chemokine receptor-based SC therapies for ischemic cardiomyopathy.

Acknowledgments

The author apologizes to many colleagues whose work could not be cited because of space limitations. This publication was supported in part by National Institutes of Health grant 1K02HL102163, and American Heart Association grant 10GRNT4180006.

References

  • 1.Manuel DG, Wilson S, Maaten S. The 2006 canadian dyslipidemia guidelines will prevent more deaths while treating fewer people–but should they be further modified? The Canadian journal of cardiology. 2008;24:617–620. doi: 10.1016/s0828-282x(08)70648-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Spertus JA, Radford MJ, Every NR, Ellerbeck EF, Peterson ED, Krumholz HM. Challenges and opportunities in quantifying the quality of care for acute myocardial infarction: Summary from the acute myocardial infarction working group of the american heart association/american college of cardiology first scientific forum on quality of care and outcomes research in cardiovascular disease and stroke. Circulation. 2003;107:1681–1691. doi: 10.1161/01.CIR.0000062026.90014.63. [DOI] [PubMed] [Google Scholar]
  • 3.Ryan TJ, Anderson JL, Antman EM, Braniff BA, Brooks NH, Califf RM, Hillis LD, Hiratzka LF, Rapaport E, Riegel BJ, Russell RO, Smith EE, Jr, Weaver WD. Acc/aha guidelines for the management of patients with acute myocardial infarction. A report of the american college of cardiology/american heart association task force on practice guidelines (committee on management of acute myocardial infarction) J Am Coll Cardiol. 1996;28:1328–1428. doi: 10.1016/s0735-1097(96)00392-0. [DOI] [PubMed] [Google Scholar]
  • 4.De Luca G, Suryapranata H, Marino P. Reperfusion strategies in acute st-elevation myocardial infarction: An overview of current status. Progress in cardiovascular diseases. 2008;50:352–382. doi: 10.1016/j.pcad.2007.11.004. [DOI] [PubMed] [Google Scholar]
  • 5.Chen J, Chemaly E, Liang L, Kho C, Lee A, Park J, Altman P, Schecter AD, Hajjar RJ, Tarzami ST. Effects of cxcr4 gene transfer on cardiac function after ischemia-reperfusion injury. Am J Pathol. 176:1705–1715. doi: 10.2353/ajpath.2010.090451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pyo RT, Sui J, Dhume A, Palomeque J, Blaxall BC, Diaz G, Tunstead J, Logothetis DE, Hajjar RJ, Schecter AD. Cxcr4 modulates contractility in adult cardiac myocytes. J Mol Cell Cardiol. 2006;41:834–844. doi: 10.1016/j.yjmcc.2006.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Entman ML, Smith CW. Postreperfusion inflammation: A model for reaction to injury in cardiovascular disease. Cardiovasc Res. 1994;28:1301–1311. doi: 10.1093/cvr/28.9.1301. [DOI] [PubMed] [Google Scholar]
  • 8.Frangogiannis NG, Entman ML. Targeting the chemokines in myocardial inflammation. Circulation. 2004;110:1341–1342. doi: 10.1161/01.CIR.0000141560.18364.63. [DOI] [PubMed] [Google Scholar]
  • 9.Balabanian K, Harriague J, Decrion C, Lagane B, Shorte S, Baleux F, Virelizier JL, Arenzana-Seisdedos F, Chakrabarti LA. Cxcr4-tropic hiv-1 envelope glycoprotein functions as a viral chemokine in unstimulated primary cd4+ t lymphocytes. J Immunol. 2004;173:7150–7160. doi: 10.4049/jimmunol.173.12.7150. [DOI] [PubMed] [Google Scholar]
  • 10.Roberts R, DeMello V, Sobel BE. Deleterious effects of methylprednisolone in patients with myocardial infarction. Circulation. 1976;53:I204–206. [PubMed] [Google Scholar]
  • 11.Jolly SR, Kane WJ, Hook BG, Abrams GD, Kunkel SL, Lucchesi BR. Reduction of myocardial infarct size by neutrophil depletion: Effect of duration of occlusion. American heart journal. 1986;112:682–690. doi: 10.1016/0002-8703(86)90461-8. [DOI] [PubMed] [Google Scholar]
  • 12.Litt MR, Jeremy RW, Weisman HF, Winkelstein JA, Becker LC. Neutrophil depletion limited to reperfusion reduces myocardial infarct size after 90 minutes of ischemia. Evidence for neutrophil-mediated reperfusion injury. Circulation. 1989;80:1816–1827. doi: 10.1161/01.cir.80.6.1816. [DOI] [PubMed] [Google Scholar]
  • 13.Frangogiannis NG, Smith CW, Entman ML. The inflammatory response in myocardial infarction. Cardiovasc Res. 2002;53:31–47. doi: 10.1016/s0008-6363(01)00434-5. [DOI] [PubMed] [Google Scholar]
  • 14.Gordon JR, Galli SJ. Mast cells as a source of both preformed and immunologically inducible tnf-alpha/cachectin. Nature. 1990;346:274–276. doi: 10.1038/346274a0. [DOI] [PubMed] [Google Scholar]
  • 15.Maury CP, Teppo AM. Circulating tumour necrosis factor-alpha (cachectin) in myocardial infarction. Journal of internal medicine. 1989;225:333–336. doi: 10.1111/j.1365-2796.1989.tb00090.x. [DOI] [PubMed] [Google Scholar]
  • 16.Ikeda U, Ohkawa F, Seino Y, Yamamoto K, Hidaka Y, Kasahara T, Kawai T, Shimada K. Serum interleukin 6 levels become elevated in acute myocardial infarction. J Mol Cell Cardiol. 1992;24:579–584. doi: 10.1016/0022-2828(92)91042-4. [DOI] [PubMed] [Google Scholar]
  • 17.Cruickshank AM, Oldroyd KG, Cobbe SM. Serum interleukin-6 in suspected myocardial infarction. Lancet. 1994;343:974. doi: 10.1016/s0140-6736(94)90096-5. [DOI] [PubMed] [Google Scholar]
  • 18.Miyao Y, Yasue H, Ogawa H, Misumi I, Masuda T, Sakamoto T, Morita E. Elevated plasma interleukin-6 levels in patients with acute myocardial infarction. American heart journal. 1993;126:1299–1304. doi: 10.1016/0002-8703(93)90526-f. [DOI] [PubMed] [Google Scholar]
  • 19.Basaran Y, Basaran MM, Babacan KF, Ener B, Okay T, Gok H, Ozdemir M. Serum tumor necrosis factor levels in acute myocardial infarction and unstable angina pectoris. Angiology. 1993;44:332–337. doi: 10.1177/000331979304400411. [DOI] [PubMed] [Google Scholar]
  • 20.Deten A, Volz HC, Briest W, Zimmer HG. Cardiac cytokine expression is upregulated in the acute phase after myocardial infarction. Experimental studies in rats. Cardiovasc Res. 2002;55:329–340. doi: 10.1016/s0008-6363(02)00413-3. [DOI] [PubMed] [Google Scholar]
  • 21.Dove A. Cd18 trials disappoint again. Nature biotechnology. 2000;18:817–818. doi: 10.1038/78412. [DOI] [PubMed] [Google Scholar]
  • 22.Anker SD, Coats AJ. How to recover from renaissance? The significance of the results of recover, renaissance, renewal and attach. International journal of cardiology. 2002;86:123–130. doi: 10.1016/s0167-5273(02)00470-9. [DOI] [PubMed] [Google Scholar]
  • 23.Anker SD, von Haehling S. Inflammatory mediators in chronic heart failure: An overview. Heart. 2004;90:464–470. doi: 10.1136/hrt.2002.007005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mann DL. Inflammatory mediators and the failing heart: Past, present, and the foreseeable future. Circ Res. 2002;91:988–998. doi: 10.1161/01.res.0000043825.01705.1b. [DOI] [PubMed] [Google Scholar]
  • 25.Rollins BJ. Chemokines. Blood. 1997;90:909–928. [PubMed] [Google Scholar]
  • 26.Stroud RM, Wells JA. Mechanistic diversity of cytokine receptor signaling across cell membranes. Sci STKE. 2004;2004:re7. doi: 10.1126/stke.2312004re7. [DOI] [PubMed] [Google Scholar]
  • 27.Horuk R. The interleukin-8-receptor family: From chemokines to malaria. Immunol Today. 1994;15:169–174. doi: 10.1016/0167-5699(94)90314-X. [DOI] [PubMed] [Google Scholar]
  • 28.Seino Y, Ikeda U, Sekiguchi H, Morita M, Konishi K, Kasahara T, Shimada K. Expression of leukocyte chemotactic cytokines in myocardial tissue. Cytokine. 1995;7:301–304. doi: 10.1006/cyto.1995.0037. [DOI] [PubMed] [Google Scholar]
  • 29.Shioi T, Matsumori A, Kihara Y, Inoko M, Ono K, Iwanaga Y, Yamada T, Iwasaki A, Matsushima K, Sasayama S. Increased expression of inter-leukin-1 beta and monocyte chemotactic and activating factor/monocyte chemoattractant protein-1 in the hypertrophied and failing heart with pressure overload. Circ Res. 1997;81:664–671. doi: 10.1161/01.res.81.5.664. [DOI] [PubMed] [Google Scholar]
  • 30.Behr TM, Wang X, Aiyar N, Coatney RW, Li X, Koster P, Angermann CE, Ohlstein E, Feuerstein GZ, Winaver J. Monocyte chemoattractant protein-1 is upregulated in rats with volume-overload congestive heart failure. Circulation. 2000;102:1315–1322. doi: 10.1161/01.cir.102.11.1315. [DOI] [PubMed] [Google Scholar]
  • 31.Kukielka GL, Youker KA, Michael LH, Kumar AG, Ballantyne CM, Smith CW, Entman ML. Role of early reperfusion in the induction of adhesion molecules and cytokines in previously ischemic myocardium. Mol Cell Biochem. 1995;147:5–12. doi: 10.1007/BF00944777. [DOI] [PubMed] [Google Scholar]
  • 32.Frangogiannis NG, Mendoza LH, Lewallen M, Michael LH, Smith CW, Entman ML. Induction and suppression of interferon-inducible protein 10 in reperfused myocardial infarcts may regulate angiogenesis. Faseb J. 2001;15:1428–1430. doi: 10.1096/fj.00-0745fje. [DOI] [PubMed] [Google Scholar]
  • 33.Tarzami ST, Cheng R, Miao W, Kitsis RN, Berman JW. Chemokine expression in myocardial ischemia: Mip-2 dependent mcp-1 expression protects cardiomyocytes from cell death. J Mol Cell Cardiol. 2002;34:209–221. doi: 10.1006/jmcc.2001.1503. [DOI] [PubMed] [Google Scholar]
  • 34.Tarzami ST, Miao W, Mani K, Lopez L, Factor SM, Berman JW, Kitsis RN. Opposing effects mediated by the chemokine receptor cxcr2 on myocardial ischemia-reperfusion injury: Recruitment of potentially damaging neutrophils and direct myocardial protection. Circulation. 2003;108:2387–2392. doi: 10.1161/01.CIR.0000093192.72099.9A. [DOI] [PubMed] [Google Scholar]
  • 35.Tarzami ST, Calderon TM, Deguzman A, Lopez L, Kitsis RN, Berman JW. Mcp-1/ccl2 protects cardiac myocytes from hypoxia-induced apop-tosis by a g(alphai)-independent pathway. Biochem Biophys Res Commun. 2005;335:1008–1016. doi: 10.1016/j.bbrc.2005.07.168. [DOI] [PubMed] [Google Scholar]
  • 36.Khan MZ, Brandimarti R, Patel JP, Huynh N, Wang J, Huang Z, Fatatis A, Meucci O. Apoptotic and antiapoptotic effects of cxcr4: Is it a matter of intrinsic efficacy? Implications for hiv neuro-pathogenesis. AIDS research and human retro-viruses. 2004;20:1063–1071. doi: 10.1089/aid.2004.20.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hu X, Dai S, Wu WJ, Tan W, Zhu X, Mu J, Guo Y, Bolli R, Rokosh G. Stromal cell derived factor-1 alpha confers protection against myocardial ischemia/reperfusion injury: Role of the cardiac stromal cell derived factor-1 alpha cxcr4 axis. Circulation. 2007;116:654–663. doi: 10.1161/CIRCULATIONAHA.106.672451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Penn MS, Zhang M, Deglurkar I, Topol EJ. Role of stem cell homing in myocardial regeneration. International journal of cardiology. 2004;95(Suppl 1):S23–25. doi: 10.1016/s0167-5273(04)90007-1. [DOI] [PubMed] [Google Scholar]
  • 39.Colamussi ML, Secchiero P, Zella D, Curreli S, Mirandola P, Capitani S, Zauli G. Stromal derived factor-1alpha induces apoptosis in activated primary cd4+ t cells. AIDS (London, England) 2000;14:748–750. doi: 10.1097/00002030-200004140-00017. [DOI] [PubMed] [Google Scholar]
  • 40.Ilhan A, Nabokikh A, Maj M, Vidakovic M, Nielsen JH, Prikoszovich T, Niederle B, Base W, Luger A, Wagner L. Cxcl12/sdf-1 over-expression in human insulinomas and its biological relevance. Molecular and cellular endocrinology. 2009;298:1–10. doi: 10.1016/j.mce.2008.10.015. [DOI] [PubMed] [Google Scholar]
  • 41.LaRocca TJ, Schwarzkopf M, Altman P, Zhang S, Gupta A, Gomes I, Alvin Z, Champion HC, Haddad G, Hajjar RJ, Devi LA, Schecter AD, Tarzami ST. Beta2-adrenergic receptor signaling in the cardiac myocyte is modulated by interactions with cxcr4. J Cardiovasc Pharmacol. doi: 10.1097/FJC.0b013e3181f713fe. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Limbourg FP, Ringes-Lichtenberg S, Schaefer A, Jacoby C, Mehraein Y, Jager MD, Limbourg A, Fuchs M, Klein G, Ballmaier M, Schlitt HJ, Schrader J, Hilfiker-Kleiner D, Drexler H. Haematopoietic stem cells improve cardiac function after infarction without permanent cardiac engraftment. Eur J Heart Fail. 2005;7:722–729. doi: 10.1016/j.ejheart.2005.06.005. [DOI] [PubMed] [Google Scholar]
  • 43.Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–776. doi: 10.1016/s0092-8674(03)00687-1. [DOI] [PubMed] [Google Scholar]
  • 44.Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7:430–436. doi: 10.1038/86498. [DOI] [PubMed] [Google Scholar]
  • 45.Hughes S. Cardiac stem cells. The Journal of pathology. 2002;197:468–478. doi: 10.1002/path.1159. [DOI] [PubMed] [Google Scholar]
  • 46.Britten MB, Abolmaali ND, Assmus B, Lehmann R, Honold J, Schmitt J, Vogl TJ, Martin H, Schachinger V, Dimmeler S, Zeiher AM. Infarct remodeling after intracoronary progenitor cell treatment in patients with acute myocardial infarction (topcare-ami): Mechanistic insights from serial contrast-enhanced magnetic resonance imaging. Circulation. 2003;108:2212–2218. doi: 10.1161/01.CIR.0000095788.78169.AF. [DOI] [PubMed] [Google Scholar]
  • 47.Laflamme MA, Zbinden S, Epstein SE, Murry CE. Cell-based therapy for myocardial ischemia and infarction: Pathophysiological mechanisms. Annu Rev Pathol. 2007;2:307–339. doi: 10.1146/annurev.pathol.2.010506.092038. [DOI] [PubMed] [Google Scholar]
  • 48.Jain M, DerSimonian H, Brenner DA, Ngoy S, Teller P, Edge AS, Zawadzka A, Wetzel K, Sawyer DB, Colucci WS, Apstein CS, Liao R. Cell therapy attenuates deleterious ventricular remodeling and improves cardiac performance after myocardial infarction. Circulation. 2001;103:1920–1927. doi: 10.1161/01.cir.103.14.1920. [DOI] [PubMed] [Google Scholar]
  • 49.Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410:701–705. doi: 10.1038/35070587. [DOI] [PubMed] [Google Scholar]
  • 50.Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004;428:664–668. doi: 10.1038/nature02446. [DOI] [PubMed] [Google Scholar]
  • 51.Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105:93–98. doi: 10.1161/hc0102.101442. [DOI] [PubMed] [Google Scholar]
  • 52.Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, Pocius J, Michael LH, Behringer RR, Garry DJ, Entman ML, Schneider MD. Cardiac progenitor cells from adult myocardium: Homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. 2003;100:12313–12318. doi: 10.1073/pnas.2132126100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mummery C, Ward-van Oostwaard D, Doevendans P, Spijker R, van den Brink S, Hassink R, van der Heyden M, Opthof T, Pera M, de la Riviere AB, Passier R, Tertoolen L. Differentiation of human embryonic stem cells to cardiomyocytes: Role of coculture with visceral endoderm- like cells. Circulation. 2003;107:2733–2740. doi: 10.1161/01.CIR.0000068356.38592.68. [DOI] [PubMed] [Google Scholar]
  • 54.Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK, Kim YJ, Soo Lee D, Sohn DW, Han KS, Oh BH, Lee MM, Park YB. Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: The magic cell randomised clinical trial. Lancet. 2004;363:751–756. doi: 10.1016/S0140-6736(04)15689-4. [DOI] [PubMed] [Google Scholar]
  • 55.Dimmeler S. Regulation of bone marrowderived vascular progenitor cell mobilization and maintenance. Arteriosclerosis, thrombosis, and vascular biology. 30:1088–1093. doi: 10.1161/ATVBAHA.109.191668. [DOI] [PubMed] [Google Scholar]
  • 56.Jo DY, Rafii S, Hamada T, Moore MA. Chemotaxis of primitive hematopoietic cells in response to stromal cell-derived factor-1. J Clin Invest. 2000;105:101–111. doi: 10.1172/JCI7954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Moore MA, Hattori K, Heissig B, Shieh JH, Dias S, Crystal RG, Rafii S. Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovector-mediated elevation of serum levels of sdf-1, vegf, and angiopoietin-1. Annals of the New York Academy of Sciences. 2001;938:36–45. doi: 10.1111/j.1749-6632.2001.tb03572.x. discussion 45-37. [DOI] [PubMed] [Google Scholar]
  • 58.Hiasa K, Ishibashi M, Ohtani K, Inoue S, Zhao Q, Kitamoto S, Sata M, Ichiki T, Takeshita A, Egashira K. Gene transfer of stromal cellderived factor-1alpha enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthase-related pathway: Nextgeneration chemokine therapy for therapeutic neovascularization. Circulation. 2004;109:2454–2461. doi: 10.1161/01.CIR.0000128213.96779.61. [DOI] [PubMed] [Google Scholar]
  • 59.Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, Giordano FJ. Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation. 2004;110:3300–3305. doi: 10.1161/01.CIR.0000147780.30124.CF. [DOI] [PubMed] [Google Scholar]
  • 60.Tendera M, Wojakowski W, Ruzyllo W, Chojnowska L, Kepka C, Tracz W, Musialek P, Piwowarska W, Nessler J, Buszman P, Grajek S, Breborowicz P, Majka M, Ratajczak MZ. Intracoronary infusion of bone marrow-derived selected cd34+cxcr4+ cells and non-selected mononuclear cells in patients with acute stemi and reduced left ventricular ejection fraction: Results of randomized, multicentre myocardial regeneration by intracoronary infusion of selected population of stem cells in acute myocardial infarction (regent) trial. Eur Heart J. 2009;30:1313–1321. doi: 10.1093/eurheartj/ehp073. [DOI] [PubMed] [Google Scholar]
  • 61.Onuffer JJ, Horuk R. Chemokines, chemokine receptors and small-molecule antagonists: Recent developments. Trends in pharmacological sciences. 2002;23:459–467. doi: 10.1016/s0165-6147(02)02064-3. [DOI] [PubMed] [Google Scholar]
  • 62.Chen C, Li J, Bot G, Szabo I, Rogers TJ, Liu-Chen LY. Heterodimerization and crossdesensitization between the mu-opioid receptor and the chemokine ccr5 receptor. European journal of pharmacology. 2004;483:175–186. doi: 10.1016/j.ejphar.2003.10.033. [DOI] [PubMed] [Google Scholar]
  • 63.Insel PA, Tang CM, Hahntow I, Michel MC. Impact of gpcrs in clinical medicine: Monogenic diseases, genetic variants and drug targets. Biochimica et biophysica acta. 2007;1768:994–1005. doi: 10.1016/j.bbamem.2006.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Medicine are provided here courtesy of e-Century Publishing Corporation

RESOURCES