Editorial Open Access
Copyright ©2010 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Stem Cells. Aug 26, 2010; 2(4): 61-66
Published online Aug 26, 2010. doi: 10.4252/wjsc.v2.i4.61
Host tissue response in stem cell therapy
Techung Lee, Department of Biochemistry and Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, NY 14214, United States
Author contributions: Lee T wrote this paper.
Supported by NIH HL84590; New York State Stem Cell Board, United States
Correspondence to: Techung Lee, PhD, Department of Biochemistry and Center for Research in Cardiovascular Medicine, University at Buffalo, 140 Farber Hall, 3435 Main Street, Buffalo, NY 14214, United States. chunglee@buffalo.edu
Telephone: +1-716-8293106 Fax: +1-716-8293106
Received: February 21, 2010
Revised: July 22, 2010
Accepted: July 29, 2010
Published online: August 26, 2010

Abstract

Preclinical and clinical trials of stem cell therapy have been carried out for treating a broad spectrum of diseases using several types of adult stem cells. While encouraging therapeutic results have been obtained, much remains to be investigated regarding the best cell type to use, cell dosage, delivery route, long-term safety, clinical feasibility, and ultimately treatment cost. Logistic aspects of stem cell therapeutics remain an area that requires urgent attention from the medical community. Recent cardiovascular trial studies have demonstrated that growth factors and cytokines derived from the injected stem cells and host tissue appear to contribute largely to the observed therapeutic benefits, indicating that trophic actions rather than the multilineage potential (or stemness) of the administered stem cells may provide the underlying tissue healing power. However, the capacity for trophic factor production can be aberrantly downregulated as seen in human heart disease. Skeletal muscle is a dynamic tissue with an impressive ability to continuously respond to environmental stimuli. Indeed, a relation exists between active skeletal muscle and low cardiovascular risk, highlighting the critical link between the skeletal muscle and cardiovascular systems. Adding to this notion are recent studies showing that stem cells injected into skeletal muscle can rescue the failing rodent heart through activation of the muscle trophic factor network and mobilization of bone marrow multilineage progenitor cells. However, aging and disease can adversely affect the host tissue into which stem cells are injected. A better understanding of the host tissue response in stem cell therapy is necessary to advance the field and bridge the gap between preclinical and clinical findings.

Key Words: Stem cell therapy, Skeletal muscle, Heart, Growth factor, Cytokine



INTRODUCTION

Stem cell therapy has entered the realm of clinical trials evaluating several types of adult stem cells and targeting a broad spectrum of diseases (http://www.clinicaltrials.gov). These investigations have generally demonstrated the safety of stem cell administration. However, consistent and reproducible beneficial effects of stem cells, as might be inferred from various animal studies, have not been demonstrated. In cardiac repair, mixed results have been reported without a clear consensus on the best cell for tissue regeneration[1,2]. Interestingly, although the multilineage differentiation potential (stemness) of stem cells was originally thought to mediate their cardiovascular therapeutic attributes, it has now become clear that the secretion of multiple growth factors and cytokines (trophic action) by the injected stem cells is primarily responsible for many of the observed therapeutic benefits[3-8]. These recent findings have necessitated a revised view on the action of the exogenously delivered stem cells, and prompted us to adopt a more integrative approach in optimizing stem cell therapeutics. Specifically, a better understanding of the cross-talk mechanism, mediated by the injected stem cells and host tissue, may prove insightful in transitioning toward future routine clinical use of adult stem cells.

Infection or injury typically triggers an inflammatory response in the host, and resolution of the inflamed state is an actively executed program. Similarly, implantation of large numbers of stem cells, whether autologous or allogeneic, is expected to elicit some host tissue immune response at least during the acute phase, and the duration and extent of this response may dictate the outcome of the cell therapy. Even in the case of implantation of medical devices possessing an inert and nonimmunogenic surface, a sequence of host inflammatory events can lead to fibrous capsule development, which can greatly compromise the device performance[9]. Although this consideration is particularly relevant for ex vivo-expanded stem cells, which may harbor xenoantigens resulting from the use of animal components (such as fetal bovine serum) in the culture medium, the topic has been discussed previously[10], and thus will not be covered here. Implanted stem cells are metabolically and functionally active, capable of interacting with the host tissue microenvironment and producing bioactive trophic factors, some of which may intervene in the host immune cascade. As elaborated below, these paracrine mediators can exert profound effects on the well-being of the host through modulation of tissue response. The focus of this editorial is to dissect this molecular cross-talk between the host tissue network and implanted bone marrow mesenchymal stem cells (MSCs) in the context of cardiovascular therapy.

TROPHIC ACTIONS OF MESENCHYMAL STEM CELLS

Bone marrow-derived MSCs have been extensively used in preclinical and clinical studies primarily because of the ease of cell isolation in large scale and their inherent immune privileged status[3,10,11]. Although MSCs can be used for therapeutic tissue engineering by virtue of their ability to differentiate into specialized cells, the beneficial effects of MSCs in treating cardiomyopathy, stroke, and osteoarthritis have been attributed mainly to their trophic activities[5,12-16]. We have used quantitative polymerase chain reaction (qPCR), flow cytometry, enzyme-linked immunosorbent assay, and Western blotting to show that bone marrow MSCs produce a diverse array of growth factors and cytokines such as angiopoietin-1, BDNF, BMP-7, FGF-1, FGF-2, FGF-5, FGF-7, FGF-9, G/M-CSF, GDF-9, HGF, IGF-1, IGF-2, IL-6, IL-11, LIF, MCP-1, NGF-β, SCF, SDF-1, TGF-β1, and VEGF[13,17,18]. While various single growth factor therapeutic regimens have been attempted with FGF, HGF, IGF, and VEGF, demonstrating impressive beneficial results in cardiac regeneration[19-22], the MSC therapy is unique in its engagement of functionally synergistic trophic factors[5,23], which may be required for efficient activation of the endogenous stem cell repair mechanism and a more sustained therapeutic effect. Therapeutically, the repertoire of MSC trophic factors can act in synergy to (1) inhibit apoptosis and limit tissue injury; (2) attenuate pathologic fibrotic remodeling; (3) promote angiogenesis and vasculogenesis; (4) activate resident tissue stem cells; and (5) modulate host immune response and reduce inflammatory oxidative stress[3].

Previous studies have shown that cells that are directly injected into the skeletal muscle bed are largely trapped in the local musculature[24]. To demonstrate that the trophic actions of MSCs underlie their cardiovascular therapeutic effects, we injected MSCs into the hamstrings of cardiomyopathic hamsters away from the myocardium, and confirmed that the intramuscularly injected MSCs were retained in the local muscular bed[25]. qPCR and histological cell-tracking assays revealed little, if any, cell migration from the injected site to other tissues, indicating that the vast majority of the injected MSCs were trapped in the musculature as expected. Despite this finding, the intramuscular MSCs significantly improved cardiac function by promoting heart cell regeneration[12]. Further, MSC-conditioned medium upon intramuscular injections was also found to be therapeutically effective for treating hamster heart failure, thus providing the ultimate proof for the critical role of trophic factors in stem cell therapy[12,13].

PARACRINE RESPONSE OF MUSCLE TISSUE

Skeletal muscle is a dynamic tissue with an adaptive capacity to continuously respond to environmental stimuli[26]. Its impressive ability to regenerate after injury or ischemic insult is coupled with the ability to produce many cardioprotective growth factors and cytokines. Indeed, the ability of skeletal muscle to function as a trophic factor-producing organ has increasingly been recognized[27-30]. Cytokines and growth factors produced and released by skeletal muscle, collectively designated as myokines, can potentially exert numerous trophic actions on other organs. Using both in vivo muscle injection and in vitro C2C12 skeletal myocyte culture, we have demonstrated muscle expression of several trophic factors such as HGF, IGF-2, NGF, and VEGF in response to trophic factor injections[31].In addition, implanted stem cells have been found to stimulate host muscle cells to produce angiogenic factors, resulting in neovascularization[32,33].

Delivery of MSCs by intramuscular injection offers a relatively noninvasive strategy as skeletal muscle, being the most abundant tissue in the body, is amenable to repeated injection of large numbers of stem cells. This approach if validated clinically is expected to facilitate future stem cell therapy. The intramuscular injection regimen for heart failure treatment draws significant comparison to the relation between active skeletal muscle and low cardiovascular risk, and highlights the critical link between the skeletal muscle and cardiovascular systems. Exercise training can promote muscle production of trophic factors including HGF, IGF, IL-6, and VEGF[27,28,34,35], some of which have been used in pre-clinical or clinical studies for cardiovascular therapy[19,20,36]. These effects of exercise have been shown to induce angiogenesis, mobilize bone marrow progenitor cells, and protect myocardium after infarction. Indeed, we found that the skeletal muscle in response to MSC injections produced many trophic factors in a more sustained fashion. The hallmark of this unique and relatively noninvasive cardiac repair regimen thus lies in the trophic cross-talk mechanisms mediated initially by the exogenous short-lived MSC-derived trophic mediators and subsequently by the myriad of endogenous muscle-derived trophic factors. These trophic factors collectively activate several signaling transduction pathways well known for their regulatory roles in cell growth, differentiation, and survival, e.g. those mediated by PI3K/AKT, ERK1/2, and JAK/STAT3. These data together indicate that skeletal muscle actively produces growth factors, and the trophic capacity can be further boosted in response to stem cell signaling in vitro and in vivo.

ACTIVATION OF MUSCLE STEM CELLS

Whereas most tissue-specific stem cells are difficult to identify, muscle satellite (stem) cells can be readily identified based on their unique location between the plasma membrane and the ensheathing basal lamina[37,38]. In undamaged skeletal muscle, the majority of satellite cells are quiescent. Activated satellite cells caused by injury or exercise are able to proliferate, differentiate, and fuse to augment existing muscle fibers and to form new fibers[37,38]. Critical roles of stem cell niches have also been established for hematopoietic and intestinal crypt stem cells that can be recruited for tissue repair when required[39]. Stem cell activity is controlled by supporting extracellular matrix and cells in the immediate vicinity[40]. These influences can be mediated by direct cell contact or secretion of soluble products. Among the multiple trophic factors produced by MSCs, FGF-2, HGF, IGF-1, SDF-1, and VEGF have been shown to activate muscle satellite cells[41-45]. In this context, we have demonstrated that MSC injections causes activation of muscle satellite cells, which mediates effective formation of new myofibers and capillaries in both injected and non-injected muscles[25]. In addition, cell tracking studies revealed that the injected MSCs could directly participate in de novo capillary formation, although the extent to which this process might take place in vivo is probably limited[46]. Trophic factor actions further lead to the expansion of myocardial progenitor cells expressing c-kit, CD31, or CD133 markers[12,31]. Expansion of these cardiac progenitor cells in the diseased myocardium are thought to critically contribute to de novo cardiomyogenesis and angiogenesis necessary for cardiac regeneration[47].

MOBILIZATION OF BONE MARROW PROGENITOR CELLS

Mobilization of bone marrow progenitor cells plays an important role in tissue repair[48]. The MSCs used here have been shown to express trophic factors such as HGF, LIF, G/M-CSF, SDF-1, and VEGF[13,17,18], which are capable of mobilizing bone marrow progenitor cells. It should be noted that administration of G-CSF has been proposed as a potential new therapy for myocardial infarction[49], and intramuscular injection of LIF plasmid DNA has been found to be cardioprotective[50]. Indeed, we detected elevated levels of circulating HGF, LIF, and M-CSF along with increased circulating c-kit+, CD31+, and CD133+ bone marrow progenitor cells after MSC therapy[12]. The mobilized progenitor cells subsequently repopulate the diseased myocardium, and participate in endogenous cardiac repair mechanisms. Notably, we have obtained evidence that this cell mobilization mechanism becomes impaired in the old cardiomyopathic hamster, which may explain at least in part why the MSC therapeutic regimen fails to rescue the aging heart (see below). The molecular cross-talk between the injected MSCs and the bone marrow compartment illustrates the dynamic and functionally relevant signaling cascade involved in stem cell repair. The signaling cascade depicted here further activates myocardial expression of growth factor genes, highlighting an additional cross-talk mechanism between the injected MSCs and myocardium.

The effect of MSC administration on mobilization of bone marrow progenitor cells can be mimicked by administration of statins[51], which are a class of HMG-CoA reductase inhibitors currently used clinically to lower cholesterol levels, retard the progression of atherosclerosis, and reduce death from cardiovascular disease[52]. Both MSCs and statins appear to mobilize bone marrow cells harboring the CD133 and/or c-kit surface markers, which are also expressed by some cardiac stem cells[12,51]. These circulating progenitor cells are thus likely to play a major role in contributing to myocardial regeneration, although detailed cellular and molecular mechanisms underlying this tissue repair process remain to be elucidated. It would be of interest to determine whether a combined MSC and statin therapy may more potently recruit bone marrow progenitor cells, resulting in a more effective cardiac therapeutic regimen.

HOST TISSUE AS A MAJOR COMPETENCE FACTOR IN STEM CELL THERAPY

Aging and disease can greatly affect the environment in which stem cells are injected. Genomic, cellular, and structural damage elicited by reactive oxygen species increase with age and translate into impaired tissue function, and oxidative stress triggered by inflammation has been implicated in the pathogenesis of many diseases[53,54]. A major challenge encountered in stem cell therapy is rapid loss of most of the injected cells after implantation[25]. This is presumably caused in part by hostile diseased tissue environments usually infiltrated with inflammatory, fibrotic, fatty, and calcified components, making it difficult for the exogenously delivered stem cells to engraft and survive. Studies have suggested that old brains are less able to support the expansion and differentiation of neuronal progenitor cells[55]. Although it is not clear if this is due to an intrinsic age-related deficit in neuronal progenitor cells, studies of endothelial progenitor cells have revealed disease- and age-associated functional impairment[56,57]. Interestingly, the degree of regeneration of the bone marrow stroma is inversely related to chronological age[58], and while the basal hematopoietic capacity is maintained throughout life, the ability of hematopoietic stem cells to respond to stress and differentiation cues appears to decrease with age[59,60].

Satellite cells of aging muscle exhibit a markedly impaired ability to produce myoblasts, which is associated with insufficient up-regulation of the Notch ligand Delta[61]. Increased Wnt signaling during aging further diverts satellite cells toward a fibrogenic lineage, contributing to increased tissue fibrosis with age[62]. Although the adult heart contains resident cardiac stem cells capable of supporting limited myocardial regeneration[47], age-associated senescence of cardiac stem cells leads to a decreased number of cardiomyocytes and heart failure[63]. Consistent with these demonstrations, aged tissue has been found to be more refractory to stem cell therapy[64], which may be associated with inadequate cell-matrix interaction and the presence of inhibitory elements. For instance, the extra lamina caused by the deposition of collagen in older dystrophic muscle[65] can potentially impede stem cell engraftment and survival. The presence of degraded fibronectin and elastin products can cause necrotic cell death[66]. Since the Notch signaling cascade in muscle satellite cells is mediated by myofiber expression of Delta, which is impaired in aged muscle[61], changes in aged myofibers are expected to impact significantly on the efficacy of the intramuscularly implanted MSCs. Given that stem cell therapy primarily targets age-associated tissue dysfunction and degeneration, host tissue competence will need to be taken into consideration in future cell therapeutics.

PERSPECTIVE

Stem cell therapeutics is entering into the clinical realm with much enthusiasm and optimism. Recent cardiovascular trial studies have demonstrated that growth factors and cytokines derived from the injected stem cells and host tissue appear to contribute largely to the observed therapeutic benefits. Thus, trophic actions rather than multilineage potentials (or stemness) of the administered stem cells are taking the center stage. Also emerging from these studies is that host tissue competence can greatly influence the outcome of stem cell therapy. Logistic aspects of stem cell therapeutics remain an area that requires urgent attention from the medical community. In cardiac repair for instance, intracoronary infusion or intramyocardial injection are mostly used for cell delivery. These delivery methods are invasive, often clinically unsuitable, and can introduce harmful scar tissue, arrhythmia, calcification, or microinfarction in the heart[67-70]. As demonstrated here with intramuscular injections of MSCs, stepping outside of the heart and taking an integrative therapeutic approach can offer an innovative cardiac repair regimen, which, if validated clinically, could minimize many perceived side effects of stem cell therapy and reduce the treatment cost. Skeletal muscle can be a major source of therapeutic trophic factors due to the large body mass of the tissue, and activation of the skeletal muscle trophic factor network can potentially be an attractive therapeutic strategy for regenerative medicine. A better understanding of host tissue response in stem cell therapy is thus likely to provide additional insights for formulating logistically sound and therapeutically effective stem cell therapeutics.

Footnotes

Peer reviewer: Roberta Morosetti, MD, Department of Neurosciences, Catholic University, Largo A. Gemelli, 8, 00168 Rome, Italy

S- Editor Wang JL L- Editor Hughes D E- Editor Yang C

References
1.  Burt RK, Loh Y, Pearce W, Beohar N, Barr WG, Craig R, Wen Y, Rapp JA, Kessler J. Clinical applications of blood-derived and marrow-derived stem cells for nonmalignant diseases. JAMA. 2008;299:925-936.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008;451:937-942.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Caplan AI. Why are MSCs therapeutic? New data: new insight. J Pathol. 2009;217:318-324.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Prockop DJ. "Stemness" does not explain the repair of many tissues by mesenchymal stem/multipotent stromal cells (MSCs). Clin Pharmacol Ther. 2007;82:241-243.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103:1204-1219.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, Semprun-Prieto L, Delafontaine P, Prockop DJ. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5:54-63.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Perez-Ilzarbe M, Agbulut O, Pelacho B, Ciorba C, San Jose-Eneriz E, Desnos M, Hagège AA, Aranda P, Andreu EJ, Menasché P. Characterization of the paracrine effects of human skeletal myoblasts transplanted in infarcted myocardium. Eur J Heart Fail. 2008;10:1065-1072.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Hinkel R, El-Aouni C, Olson T, Horstkotte J, Mayer S, Müller S, Willhauck M, Spitzweg C, Gildehaus FJ, Münzing W. Thymosin beta4 is an essential paracrine factor of embryonic endothelial progenitor cell-mediated cardioprotection. Circulation. 2008;117:2232-2240.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Kamath S, Bhattacharyya D, Padukudru C, Timmons RB, Tang L. Surface chemistry influences implant-mediated host tissue responses. J Biomed Mater Res A. 2008;86:617-626.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Tyndall A, Walker UA, Cope A, Dazzi F, De Bari C, Fibbe W, Guiducci S, Jones S, Jorgensen C, Le Blanc K. Immunomodulatory properties of mesenchymal stem cells: a review based on an interdisciplinary meeting held at the Kennedy Institute of Rheumatology Division, London, UK, 31 October 2005. Arthritis Res Ther. 2007;9:301.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Giordano A, Galderisi U, Marino IR. From the laboratory bench to the patient's bedside: an update on clinical trials with mesenchymal stem cells. J Cell Physiol. 2007;211:27-35.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure therapy mediated by the trophic activities of bone marrow mesenchymal stem cells: a noninvasive therapeutic regimen. Am J Physiol Heart Circ Physiol. 2009;296:H1888-H1897.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Zisa D, Shabbir A, Suzuki G, Lee T. Vascular endothelial growth factor (VEGF) as a key therapeutic trophic factor in bone marrow mesenchymal stem cell-mediated cardiac repair. Biochem Biophys Res Commun. 2009;390:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48:3464-3474.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Li Y, Chen J, Chen XG, Wang L, Gautam SC, Xu YX, Katakowski M, Zhang LJ, Lu M, Janakiraman N. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology. 2002;59:514-523.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Mahmood A, Lu D, Lu M, Chopp M. Treatment of traumatic brain injury in adult rats with intravenous administration of human bone marrow stromal cells. Neurosurgery. 2003;53:697-702; discussion 702-703.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Vacanti V, Kong E, Suzuki G, Sato K, Canty JM, Lee T. Phenotypic changes of adult porcine mesenchymal stem cells induced by prolonged passaging in culture. J Cell Physiol. 2005;205:194-201.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Lin H, Shabbir A, Molnar M, Yang J, Marion S, Canty JM Jr, Lee T. Adenoviral expression of vascular endothelial growth factor splice variants differentially regulate bone marrow-derived mesenchymal stem cells. J Cell Physiol. 2008;216:458-468.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Suzuki G, Lee TC, Fallavollita JA, Canty JM Jr. Adenoviral gene transfer of FGF-5 to hibernating myocardium improves function and stimulates myocytes to hypertrophy and reenter the cell cycle. Circ Res. 2005;96:767-775.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Nakamura T, Matsumoto K, Mizuno S, Sawa Y, Matsuda H, Nakamura T. Hepatocyte growth factor prevents tissue fibrosis, remodeling, and dysfunction in cardiomyopathic hamster hearts. Am J Physiol Heart Circ Physiol. 2005;288:H2131-H2139.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Serose A, Prudhon B, Salmon A, Doyennette MA, Fiszman MY, Fromes Y. Administration of insulin-like growth factor-1 (IGF-1) improves both structure and function of delta-sarcoglycan deficient cardiac muscle in the hamster. Basic Res Cardiol. 2005;100:161-170.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Losordo DW, Vale PR, Symes JF, Dunnington CH, Esakof DD, Maysky M, Ashare AB, Lathi K, Isner JM. Gene therapy for myocardial angiogenesis: initial clinical results with direct myocardial injection of phVEGF165 as sole therapy for myocardial ischemia. Circulation. 1998;98:2800-2804.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98:1076-1084.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Dezawa M, Ishikawa H, Itokazu Y, Yoshihara T, Hoshino M, Takeda S, Ide C, Nabeshima Y. Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science. 2005;309:314-317.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Shabbir A, Zisa D, Leiker M, Johnston C, Lin H, Lee T. Muscular dystrophy therapy by nonautologous mesenchymal stem cells: muscle regeneration without immunosuppression and inflammation. Transplantation. 2009;87:1275-1282.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Ambrosio F, Kadi F, Lexell J, Fitzgerald GK, Boninger ML, Huard J. The effect of muscle loading on skeletal muscle regenerative potential: an update of current research findings relating to aging and neuromuscular pathology. Am J Phys Med Rehabil. 2009;88:145-155.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Pedersen BK, Febbraio MA. Muscle as an endocrine organ: focus on muscle-derived interleukin-6. Physiol Rev. 2008;88:1379-1406.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Walsh K. Adipokines, myokines and cardiovascular disease. Circ J. 2009;73:13-18.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Hughes RA, Sendtner M, Goldfarb M, Lindholm D, Thoenen H. Evidence that fibroblast growth factor 5 is a major muscle-derived survival factor for cultured spinal motoneurons. Neuron. 1993;10:369-377.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Oppenheim RW, Wiese S, Prevette D, Armanini M, Wang S, Houenou LJ, Holtmann B, Gotz R, Pennica D, Sendtner M. Cardiotrophin-1, a muscle-derived cytokine, is required for the survival of subpopulations of developing motoneurons. J Neurosci. 2001;21:1283-1291.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Zisa D, Shabbir A, Mastri M, Suzuki G, Lee T. Intramuscular VEGF repairs the failing heart: role of host-derived growth factors and mobilization of progenitor cells. Am J Physiol Regul Integr Comp Physiol. 2009;297:R1503-R1515.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Tateno K, Minamino T, Toko H, Akazawa H, Shimizu N, Takeda S, Kunieda T, Miyauchi H, Oyama T, Matsuura K. Critical roles of muscle-secreted angiogenic factors in therapeutic neovascularization. Circ Res. 2006;98:1194-1202.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Cho HJ, Lee N, Lee JY, Choi YJ, Ii M, Wecker A, Jeong JO, Curry C, Qin G, Yoon YS. Role of host tissues for sustained humoral effects after endothelial progenitor cell transplantation into the ischemic heart. J Exp Med. 2007;204:3257-3269.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Wu G, Rana JS, Wykrzykowska J, Du Z, Ke Q, Kang P, Li J, Laham RJ. Exercise-induced expression of VEGF and salvation of myocardium in the early stage of myocardial infarction. Am J Physiol Heart Circ Physiol. 2009;296:H389-H395.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Trenerry MK, Carey KA, Ward AC, Cameron-Smith D. STAT3 signaling is activated in human skeletal muscle following acute resistance exercise. J Appl Physiol. 2007;102:1483-1489.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Testa U, Pannitteri G, Condorelli GL. Vascular endothelial growth factors in cardiovascular medicine. J Cardiovasc Med (Hagerstown). 2008;9:1190-1221.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Kuang S, Gillespie MA, Rudnicki MA. Niche regulation of muscle satellite cell self-renewal and differentiation. Cell Stem Cell. 2008;2:22-31.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Cornelison DD. Context matters: in vivo and in vitro influences on muscle satellite cell activity. J Cell Biochem. 2008;105:663-669.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Scadden DT. The stem-cell niche as an entity of action. Nature. 2006;441:1075-1079.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Jones DL, Wagers AJ. No place like home: anatomy and function of the stem cell niche. Nat Rev Mol Cell Biol. 2008;9:11-21.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Tatsumi R, Anderson JE, Nevoret CJ, Halevy O, Allen RE. HGF/SF is present in normal adult skeletal muscle and is capable of activating satellite cells. Dev Biol. 1998;194:114-128.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  DiMario J, Buffinger N, Yamada S, Strohman RC. Fibroblast growth factor in the extracellular matrix of dystrophic (mdx) mouse muscle. Science. 1989;244:688-690.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Machida S, Booth FW. Insulin-like growth factor 1 and muscle growth: implication for satellite cell proliferation. Proc Nutr Soc. 2004;63:337-340.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Sherwood RI, Christensen JL, Conboy IM, Conboy MJ, Rando TA, Weissman IL, Wagers AJ. Isolation of adult mouse myogenic progenitors: functional heterogeneity of cells within and engrafting skeletal muscle. Cell. 2004;119:543-554.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Arsic N, Zacchigna S, Zentilin L, Ramirez-Correa G, Pattarini L, Salvi A, Sinagra G, Giacca M. Vascular endothelial growth factor stimulates skeletal muscle regeneration in vivo. Mol Ther. 2004;10:844-854.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Leiker M, Suzuki G, Iyer VS, Canty JM Jr, Lee T. Assessment of a nuclear affinity labeling method for tracking implanted mesenchymal stem cells. Cell Transplant. 2008;17:911-922.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763-776.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  LaBarge MA, Blau HM. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell. 2002;111:589-601.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Ince H, Petzsch M, Kleine HD, Schmidt H, Rehders T, Körber T, Schümichen C, Freund M, Nienaber CA. Preservation from left ventricular remodeling by front-integrated revascularization and stem cell liberation in evolving acute myocardial infarction by use of granulocyte-colony-stimulating factor (FIRSTLINE-AMI). Circulation. 2005;112:3097-3106.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Zou Y, Takano H, Mizukami M, Akazawa H, Qin Y, Toko H, Sakamoto M, Minamino T, Nagai T, Komuro I. Leukemia inhibitory factor enhances survival of cardiomyocytes and induces regeneration of myocardium after myocardial infarction. Circulation. 2003;108:748-753.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Suzuki G, Iyer V, Cimato T, Canty JM Jr. Pravastatin improves function in hibernating myocardium by mobilizing CD133+ and cKit+ bone marrow progenitor cells and promoting myocytes to reenter the growth phase of the cardiac cell cycle. Circ Res. 2009;104:255-264, 10p following 264.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Ramasubbu K, Estep J, White DL, Deswal A, Mann DL. Experimental and clinical basis for the use of statins in patients with ischemic and nonischemic cardiomyopathy. J Am Coll Cardiol. 2008;51:415-426.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Hekimi S, Guarente L. Genetics and the specificity of the aging process. Science. 2003;299:1351-1354.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Hasty P, Campisi J, Hoeijmakers J, van Steeg H, Vijg J. Aging and genome maintenance: lessons from the mouse? Science. 2003;299:1355-1359.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Zitnik G, Martin GM. Age-related decline in neurogenesis: old cells or old environment? J Neurosci Res. 2002;70:258-263.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Masuda H, Asahara T. Post-natal endothelial progenitor cells for neovascularization in tissue regeneration. Cardiovasc Res. 2003;58:390-398.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, Finkel T. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348:593-600.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Wolf NS, Arora RK. Depletion of reserve in the hemopoietic system: I. Self-replication by stromal cells related to chronologic age. Mech Ageing Dev. 1982;20:127-140.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Schlessinger D, Van Zant G. Does functional depletion of stem cells drive aging? Mech Ageing Dev. 2001;122:1537-1553.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Van Zant G. Genetic control of stem cells: implications for aging. Int J Hematol. 2003;77:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Conboy IM, Conboy MJ, Smythe GM, Rando TA. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302:1575-1577.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, Rando TA. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science. 2007;317:807-810.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Torella D, Rota M, Nurzynska D, Musso E, Monsen A, Shiraishi I, Zias E, Walsh K, Rosenzweig A, Sussman MA. Cardiac stem cell and myocyte aging, heart failure, and insulin-like growth factor-1 overexpression. Circ Res. 2004;94:514-524.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Lehrke S, Mazhari R, Durand DJ, Zheng M, Bedja D, Zimmet JM, Schuleri KH, Chi AS, Gabrielson KL, Hare JM. Aging impairs the beneficial effect of granulocyte colony-stimulating factor and stem cell factor on post-myocardial infarction remodeling. Circ Res. 2006;99:553-560.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Alexakis C, Partridge T, Bou-Gharios G. Implication of the satellite cell in dystrophic muscle fibrosis: a self-perpetuating mechanism of collagen overproduction. Am J Physiol Cell Physiol. 2007;293:C661-C669.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Robert L, Labat-Robert J. Aging of connective tissues: from genetic to epigenetic mechanisms. Biogerontology. 2000;1:123-131.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Breitbach M, Bostani T, Roell W, Xia Y, Dewald O, Nygren JM, Fries JW, Tiemann K, Bohlen H, Hescheler J. Potential risks of bone marrow cell transplantation into infarcted hearts. Blood. 2007;110:1362-1369.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Yoon YS, Park JS, Tkebuchava T, Luedeman C, Losordo DW. Unexpected severe calcification after transplantation of bone marrow cells in acute myocardial infarction. Circulation. 2004;109:3154-3157.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Widimsky P, Penicka M. Complications after intracoronary stem cell transplantation in idiopathic dilated cardiomyopathy. Int J Cardiol. 2006;111:178-179.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Vulliet PR, Greeley M, Halloran SM, MacDonald KA, Kittleson MD. Intra-coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. Lancet. 2004;363:783-784.  [PubMed]  [DOI]  [Cited in This Article: ]