Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 16.
Published in final edited form as: Circ Res. 2013 Aug 16;113(5):10.1161/CIRCRESAHA.113.302083. doi: 10.1161/CIRCRESAHA.113.302083

Cortical Bone-Derived Stem Cells: A Novel Class of Cells for Myocardial Protection

Sang-Ging Ong 1,2, Joseph C Wu 1,2
PMCID: PMC3855861  NIHMSID: NIHMS517471  PMID: 23948579

Acute myocardial infarction (MI) remains a significant cause of mortality and morbidity worldwide, despite steady advances in our understanding and treatment of coronary heart disease.1 Following MI, lost myocardium is replaced with scar tissue, leading to left ventricular (LV) remodeling and ultimately culminating with ischemic cardiomyopathy and congestive heart failure. We now know that the adult heart is not a post-mitotic organ lacking capacity for self-renewal following injury. This paradigm shift occurred following identification of stem cell niches in the adult heart and isolation of cardiac progenitor cells, including c-kit+ cardiac stem cells (CSCs), side population cells, and cardiospheres.2-7 However, the endogenous proliferation of cardiac progenitor cells after MI is inadequate for full replacement of the large number of depleted cells.8 To overcome this problem, extensive efforts have been made in developing cell-based therapies to promote tissue repair by introduction of exogenous cells such as bone marrow-derived mononuclear cells (BM-MNCs), bone marrow-derived mesenchymal stem cells (BM-MSCs), adipose tissue-derived mesenchymal stem cells (AT-MSCs), CD34+ stem cells, c-kit+ CSCs, and cardiosphere-derived cells, as evidenced by recent clinical trials.9-16

For now, the BM-MNCs are the most widely used source of cells in cardiovascular regenerative therapy trials. However, despite some successful early-stage clinical trials, several recent studies involving the use of BM-MNCs have not been overwhelmingly positive. For example, in the placebo-controlled TIME trial in which investigators assessed whether differential timing of BM-MNCs delivery affected LVEF following acute MI (3-7 days), no significant differences between the BM-MNCs and placebo groups were observed for either primary or secondary end-points.17 In fact, the TIME trial was the third study in a series of studies (Late-TIME,18 FOCUS-CCTRN,16 and TIME) on BM-MNCs by the NIH-sponsored Cardiovascular Cell Therapy Research Network (CCTRN) that have all met with negative results. These findings were independently confirmed by a subsequent study, Swiss-AMI, in which intracoronary delivery of BM-MNCs in the presence of an acute MI did not improve LV ejection fraction (LVEF) when cells were delivered at 5-7 days or 3-4 weeks post-MI.19

By contrast, in the POSEIDON trial, patients with ischemic cardiomyopathy after previous MIs (years earlier) who were given either allogeneic or autologous BM-MSC sat different doses demonstrated modest improvements in terms of reversed LV remodeling, though this change did not translate into an improvement in LVEF.20 In a separate trial called C-CURE, investigators found that exposing BM-MSCs to a cardiogenic cocktail promoted the nuclear expression of myocytes-specific enhancer factor 2C (MEF2C), which led to improved LVEF and composite clinical score.21 More recently, two recent clinical trials which employed amplification of cardiac progenitor cells obtained from biopsied heart tissues followed by readministration to patients were performed. In the SCIPIO trial which used c-kit+ CSCs, patients had improved LVEF and smaller MI size at 1 year follow-up.10 The use of cardiosphere-derived cells in CADUCEUS was also shown to reduce infarct size in patients, although this was not accompanied by an improvement in LVEF.11 In addition to these trials, Vrtovec et al. reported that intracoronary CD34+ cell infusion in nonischemic dilated cardiomyopathy patients led to improved LVEF, exercise tolerance, and long-term survival.15 By labeling these CD34+ cells with 99mTc-hexamethylpropylenamine oxine and performing single photon emission computed tomography (SPECT) imaging, the authors demonstrated a positive correlation between early imaging of CD34+ cell retention and late assessment of cardiac functional outcome.

The inconsistencies in clinical outcome using various stem cell types demonstrate the need for a more thorough reevaluation of cell therapy strategies to eliminate any red herrings during preclinical stages. In addition, several other variables are crucial to determine the success of any cell therapy approach. First, the preparation and characterization of the various cell types have been shown to exert important effects on myocardial repair. For example, Seeger et al. reported that heparin, typically used as an anticoagulant for intracoronary infusion of BM-MNCs, impairs the functional capacity of these cells by blocking SDF-1/CXCR4 signaling, although this has been recently refuted.22, 23 Second, the dose, timing, and manner of cell delivery are also crucial, and they are in turn complicated by the diseased state, the functional capacity of the cells, and the complex interactions between the cells and host myocardial microenvironment. To solve these problems, a better understanding of the basic biological mechanisms underlying beneficial effects of cell therapy is needed, because knowledge of the mechanisms involved can offer a dynamic perspective of cardiac homeostasis and regenerative biology, eventually leading to improved cell therapy. Some of the hypothesized mechanisms include the following: secretion of paracrine factors; stimulation of neovascularization; activation of cardiac regeneration through recruitment of stem cells or proliferation of preformed cardiomyocytes; and transdifferentiation of transplanted stem cells.24-27

In this issue of Circulation Research, Duran et al. present an extensive study describing cortical bone-derived stem cells (CBSCs).28 The authors show that cells isolated from the cortical bone tissue (rather than the bone marrow)29 are in a more primitive state, negative for most markers of the hematopoietic lineage, but are still capable of differentiating into osteoblasts, chondrocytes, and adipocytes in vitro as seen with MSCs. The authors also compared the potency of these CBSCs to c-kit+/Sca1+ cardiac-derived stem cells (CDCs).10 In vitro, these two cell types were similar in terms of the type and amount of paracrine factors secreted (Ang-1, bFGF, HGF, IGF-1, PDGF, SCF, SDF-1, and VEGF) and were capable of differentiating into cells expressing cardiac-specific proteins.28 In vivo, wild-type C57BL/6 mice receiving CBSCs had better survival over a 6-week period (77%) compared to those receiving sham saline (50%) or CDCs (66%). Although mice receiving either CBSCs or CDCs showed improved cardiac function and attenuated adverse cardiac remodeling, these changes were more pronounced in the CBSC-treated group. There was no significant difference in the areas at risk (AAR) or ischemic size (IS) at 24 hours post-MI for both groups compared to the sham saline condition. However, at 6 weeks post-MI, chronic infarct size was significantly reduced in both groups, especially in the CBSC cohort.

To gain more insight into how these cells function in vivo as opposed to in vitro, the authors next performed immunostaining of the aforementioned 8 paracrine factors in both CBSC- and CDC-treated groups. Because CBSCs and CDCs were isolated from EGFP+ mice, co-staining analysis of both GFP and individual paracrine factors revealed that CBSCs secreted only two factors (βFGF and VEGF) upon transplantation, with the VEGF expression persisting as long as 2 weeks post-MI. By contrast, CDCs secreted three factors (βFGF, VEGF, and Ang-1) for a shorter period of time, only up to 24 hours. Interestingly, upregulation of VEGF, Ang-1, βFGF, and several other pro-angiogenic and anti-apoptotic cytokines was also reported in a recent study involving transplantation of induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) using microfluidic single-cell PCR analysis at 6 days following MI.30 An increase in neovascularization was observed in both CBSC- or CDC-treated hearts compared to saline-treated controls, although it should be noted that the majority of these blood vessels did not contain EGFP+ cells, indicating that they were derived via endogenous repair and not from the transplanted EGFP+ stem cells. Importantly, histological analysis demonstrated the presence of EGFP+ stem cell-derived adult cardiac myocytes that had normally striated α-sarcomeric actin networks and connexin43+ gap junctions throughout the border zone in CBSC-treated mice. By contrast, no EGFP+ cells with organized sarcomeres were detected in CDC-treated mice. Finally, when isolated for ex vivo analysis, these transplanted EGFP+ myocytes (CBSC group) displayed contractions and Ca2+ transients that were indistinguishable from their endogenous EGFP myocytes, indicating that they had acquired a mature phenotype.

Concluding from their data, the authors suggest that CBSCs represent a bone-derived c-kit+/Sca-1+ stem cell population that can support the injured heart through direct transdifferentiation into adult cardiomyocytes and, to a lesser extent, vascular cells, as well as through paracrine factors. Moreover, other aspects of this study may provide important clues into future applications of these CBSCs. For instance, only eight paracrine factors were selected for testing as pro-angiogenic factors, and these CBSCs have been shown to possess immunomodulatory properties.29, 31 Thus, it would be interesting to study the efficacy of these CBSCs when delivered in an allogeneic setting, which would model the recent POSEIDON trial.20 It is also interesting to note that although both CBSCs and CDCs secreted paracrine factors in the initial 24 hours, AAR and IS in these groups were not significantly different compared to the control saline group (which received no cell injection and hence no additional paracrine factors released at 24 hours), potentially casting doubt on the importance of paracrine molecules at least in this study. This was further highlighted by the fact that although CDCs stopped secreting any of those measured paracrine factor 24 hours later, as compared to the continued secretion of VEGF in CBSCs for up to 2 weeks, the improvement in LVEF and fractional shortening (FS) was similar in both groups at 1 week post-MI. This suggests that the 8 measured paracrine factors might be negligible at least in the early stages of MI. Of note, most of the molecules tested were targets of hypoxia-inducible factor-1 (HIF-1), and previous gene therapy study has demonstrated that upregulation of HIF-1 leads to a sustained elevation of pro-angiogenic factors (coincidentally including βFGF and VEGF measured in this study) at least for 2 weeks in a similar model.32 Thus, it may be worthwhile to consider combining gene and cell therapy to obtain greater synergistic effects.

Another particularly interesting aspect of this study was how the starting number of injected EGFP+ cells used (40,000 cells/heart) increased more than 3 times (∼130,000 cells/heart), as measured by the estimated final number of EGFP+ mature/non-mature cardiomyocytes harvested 6 weeks post-MI. The final number did not include other population of EGFP+ cells such as vascular smooth muscle cells, vascular endothelial cells, and EGFP+ cells, which are smaller and lack either cardiac or vascular cell proteins. Significantly improved cardiac function was seen as early as 1 week post-MI, which was impressive considering that the starting number of injected cells was lower than in other studies.33, 34 These EGFP+ CBSCs were delivered into the hostile inflammatory microenvironment of acute MI, and yet managed to survive, engraft, and proliferate robustly. By contrast, several other studies using longitudinal imaging analysis within the same transplanted animals have shown that most of the injected BM-MNCs, BM-MSCs, Sca1+ CSCs, and AT-MSCs in similar settings die within 6-8 weeks after transplantation.35-40 The aforementioned 300% increase in CBSC-derived new myocytes suggests that CBSCs either have a very high rate of retention following injection into the heart, or may possess a remarkable proliferative and cardiac differentiation capacity, which have not been observed in several other adult stem cell types.

One of the proposed mechanisms conferring protection was partially attributed to the formation and maturation of CBSC-derived new myocytes, which is in accordance with the high proliferative/differentiation capacity described above. At 1 and 2 weeks post-MI, EGFP+ cells at intermediate stages of myocyte differentiation were detected in all hearts analyzed (6/6). At 6 weeks post-MI, mature EGFP+ cardiomyocytes were only detected in 5 out of 8 hearts analyzed (∼60%), despite a significant improvement in cardiac function compared to saline or CDC-treated groups. Hence further investigation is warranted to explain the significant improvement in cardiac function despite finding mature EGFP+ myocyte in only around half of the hearts. At the same time, the findings here showing that CBSCs are superior to CDCs in forming mature cardiomyocytes are intriguing. For example, Oskouei et al reported that human c-kit+ CSCs were superior compared to MSCs in terms of improvement of cardiac function, cell engraftment, and also differentiation.41

As with all good research that generates more intriguing questions from its results, the Duran et al. study has brought a new class of stem cells to the forefront for potential use in cardiac regenerative medicine, but much more research needs to be conducted to fully understand the biology of these cells. One fascinating approach would be to perform transcriptomic profiling on these CBSCs that would provide a thorough understanding of the genetic signature of these cells, which can then be added to existing resources. For example, a recent study by Dey et al. identified molecular pathways specific for different cardiogenic stem and progenitor cells by comparing CSCs (c-kit+, Sca1+, and side population) and BM-derived progenitor cells (BM-c-kit+ and BM-MSCs).42 Inclusion of these CBSCs would be invaluable for comparison. Elucidation of the molecular pathways behind this cohort of cells which possess the secretory potential typically seen with MSCs along with myogenic potential typically associated with cells of cardiac origin offers the possibility of including CBSCs as an exciting new player in the field of cardiac regeneration.

Figure.

Figure

Sources of stem cells for cardiac regeneration and potential reparative mechanisms. Cortical bone stem cells which have been shown to transdifferentiate into adult cardiomyoctes while secreting paracrine factors might be an attractive new class of stem cells. MSCs: mesenchymal stromal cells; BM-MNCs: bone marrow-derived mononuclear cells; AT-MSCs: adipose tissue-derived mesenchymal stem cells, HSCs: hematopoietic stem cells; ESC-ECs: embryonic stem cell-derived endothelial cells; iPSC-ECs: induced pluripotent stem cell-derived endothelial cells; ESC-CMs: embryonic stem cell-derived cardiomyocytes; iPSC-CMs: induced pluripotent stem cell-derived cardiomyocytes. Please note that although cardiospheres contain a mixed population of cells other than c-kit+ cells, they are included here as stem cells for brevity.

Acknowledgments

Sources of Funding: We are grateful for the funding support by National Institute of Health U01 HL099776, R01 EB009689, American Heart Association Established Investigator Award, and Burroughs Wellcome Foundation Career Award for Medical Scientists (JCW).

Footnotes

Disclosures: None

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Magid D, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER, Moy CS, Mussolino ME, Nichol G, Paynter NP, Schreiner PJ, Sorlie PD, Stein J, Turan TN, Virani SS, Wong ND, Woo D, Turner MB. Heart disease and stroke statistics--2013 update: A report from the american heart association. Circulation. 2013;127:e6–e245. doi: 10.1161/CIR.0b013e31828124ad. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.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]
  • 3.Smith RR, Barile L, Cho HC, Leppo MK, Hare JM, Messina E, Giacomello A, Abraham MR, Marban E. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007;115:896–908. doi: 10.1161/CIRCULATIONAHA.106.655209. [DOI] [PubMed] [Google Scholar]
  • 4.Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso F, Salio M, Battaglia M, Latronico MVG, Coletta M, Vivarelli E, Frati L, Cossu G, Giacomello A. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circulation Research. 2004;95:911–921. doi: 10.1161/01.RES.0000147315.71699.51. [DOI] [PubMed] [Google Scholar]
  • 5.Martin CM, Meeson AP, Robertson SM, Hawke TJ, Richardson JA, Bates S, Goetsch SC, Gallardo TD, Garry DJ. Persistent expression of the atp-binding cassette transporter, abcg2, identifies cardiac sp cells in the developing and adult heart. Dev Biol. 2004;265:262–275. doi: 10.1016/j.ydbio.2003.09.028. [DOI] [PubMed] [Google Scholar]
  • 6.Scharenberg CW, Harkey MA, Torok-Storb B. The abcg2 transporter is an efficient hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood. 2002;99:507–512. doi: 10.1182/blood.v99.2.507. [DOI] [PubMed] [Google Scholar]
  • 7.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]
  • 8.Laflamme MA, Murry CE. Heart regeneration. Nature. 2011;473:326–335. doi: 10.1038/nature10147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB, Jr, Reisman MA, Schaer GL, Sherman W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54:2277–2286. doi: 10.1016/j.jacc.2009.06.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bolli R, Chugh AR, D'Amario D, Loughran JH, Stoddard MF, Ikram S, Beache GM, Wagner SG, Leri A, Hosoda T, Sanada F, Elmore JB, Goichberg P, Cappetta D, Solankhi NK, Fahsah I, Rokosh DG, Slaughter MS, Kajstura J, Anversa P. Cardiac stem cells in patients with ischaemic cardiomyopathy (scipio): Initial results of a randomised phase 1 trial. Lancet. 2011;378:1847–1857. doi: 10.1016/S0140-6736(11)61590-0. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 11.Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, Czer LS, Marban L, Mendizabal A, Johnston PV, Russell SD, Schuleri KH, Lardo AC, Gerstenblith G, Marban E. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (caduceus): A prospective, randomised phase 1 trial. Lancet. 2012;379:895–904. doi: 10.1016/S0140-6736(12)60195-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Valina C, Pinkernell K, Song YH, Bai X, Sadat S, Campeau RJ, Le Jemtel TH, Alt E. Intracoronary administration of autologous adipose tissue-derived stem cells improves left ventricular function, perfusion, and remodelling after acute myocardial infarction. Eur Heart J. 2007;28:2667–2677. doi: 10.1093/eurheartj/ehm426. [DOI] [PubMed] [Google Scholar]
  • 13.Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Egeland T, Endresen K, Ilebekk A, Mangschau A, Fjeld JG, Smith HJ, Taraldsrud E, Grogaard HK, Bjornerheim R, Brekke M, Muller C, Hopp E, Ragnarsson A, Brinchmann JE, Forfang K. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N Engl J Med. 2006;355:1199–1209. doi: 10.1056/NEJMoa055706. [DOI] [PubMed] [Google Scholar]
  • 14.Vrtovec B, Poglajen G, Sever M, Lezaic L, Domanovic D, Cernelc P, Haddad F, Torre-Amione G. Effects of intracoronary stem cell transplantation in patients with dilated cardiomyopathy. J Card Fail. 2011;17:272–281. doi: 10.1016/j.cardfail.2010.11.007. [DOI] [PubMed] [Google Scholar]
  • 15.Vrtovec B, Poglajen G, Lezaic L, Sever M, Domanovic D, Cernelc P, Socan A, Schrepfer S, Torre-Amione G, Haddad F, Wu JC. Effects of intracoronary cd34+ stem cell transplantation in nonischemic dilated cardiomyopathy patients: 5-year follow-up. Circ Res. 2013;112:165–173. doi: 10.1161/CIRCRESAHA.112.276519. [DOI] [PubMed] [Google Scholar]
  • 16.Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Silva GV, Lai D, Thomas JD, Kronenberg MW, Martin AD, Anderson RD, Traverse JH, Penn MS, Anwaruddin S, Hatzopoulos AK, Gee AP, Taylor DA, Cogle CR, Smith D, Westbrook L, Chen J, Handberg E, Olson RE, Geither C, Bowman S, Francescon J, Baraniuk S, Piller LB, Simpson LM, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD. Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: The focus-cctrn trial. JAMA. 2012;307:1717–1726. doi: 10.1001/jama.2012.418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Traverse Jh, H TD, P CJ, et al. Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: The time randomized trial. JAMA. 2012;308:2380–2389. doi: 10.1001/jama.2012.28726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, Forder JR, Byrne BJ, Hatzopoulos AK, Penn MS, Perin EC, Baran KW, Chambers J, Lambert C, Raveendran G, Simon DI, Vaughan DE, Simpson LM, Gee AP, Taylor DA, Cogle CR, Thomas JD, Silva GV, Jorgenson BC, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Smith DX, Baraniuk S, Piller LB, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: The latetime randomized trial. JAMA. 2011;306:2110–2119. doi: 10.1001/jama.2011.1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Surder D, Manka R, Lo Cicero V, Moccetti T, Rufibach K, Soncin S, Turchetto L, Radrizzani M, Astori G, Schwitter J, Erne P, Zuber M, Auf der Maur C, Jamshidi P, Gaemperli O, Windecker S, Moschovitis A, Wahl A, Buhler I, Wyss C, Kozerke S, Landmesser U, Luscher TF, Corti R. Intracoronary injection of bone marrow-derived mononuclear cells early or late after acute myocardial infarction: Effects on global left ventricular function. Circulation. 2013;127:1968–1979. doi: 10.1161/CIRCULATIONAHA.112.001035. [DOI] [PubMed] [Google Scholar]
  • 20.Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The poseidon randomized trial. JAMA. 2012;308:2369–2379. doi: 10.1001/jama.2012.25321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, El Nakadi B, Banovic M, Beleslin B, Vrolix M, Legrand V, Vrints C, Vanoverschelde JL, Crespo-Diaz R, Homsy C, Tendera M, Waldman S, Wijns W, Terzic A. Cardiopoietic stem cell therapy in heart failure: The c-cure (cardiopoietic stem cell therapy in heart failure) multicenter randomized trial with lineage-specified biologics. J Am Coll Cardiol. 2013;61:2329–2338. doi: 10.1016/j.jacc.2013.02.071. [DOI] [PubMed] [Google Scholar]
  • 22.Seeger FH, Rasper T, Fischer A, Muhly-Reinholz M, Hergenreider E, Leistner DM, Sommer K, Manavski Y, Henschler R, Chavakis E, Assmus B, Zeiher AM, Dimmeler S. Heparin disrupts the cxcr4/sdf-1 axis and impairs the functional capacity of bone marrow-derived mononuclear cells used for cardiovascular repair. Circ Res. 2012;111:854–862. doi: 10.1161/CIRCRESAHA.112.265678. [DOI] [PubMed] [Google Scholar]
  • 23.Moye LA, Simari RD, Skarlatos SI Cardiovascular Cell Therapy Research Network f. Bone marrow-derived cell therapy after myocardial infarction. JAMA. 2013;309:1459–1460. doi: 10.1001/jama.2013.2601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Beltrami AP, Urbanek K, Kajstura J, Yan SM, Finato N, Bussani R, Nadal-Ginard B, Silvestri F, Leri A, Beltrami CA, Anversa P. Evidence that human cardiac myocytes divide after myocardial infarction. New England Journal of Medicine. 2001;344:1750–1757. doi: 10.1056/NEJM200106073442303. [DOI] [PubMed] [Google Scholar]
  • 25.Wang X, Jameel MN, Li Q, Mansoor A, Qiang X, Swingen C, Panetta C, Zhang J. Stem cells for myocardial repair with use of a transarterial catheter. Circulation. 2009;120:S238–246. doi: 10.1161/CIRCULATIONAHA.109.885236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, Noiseux N, Zhang L, Pratt RE, Ingwall JS, Dzau VJ. Paracrine action accounts for marked protection of ischemic heart by akt-modified mesenchymal stem cells. Nat Med. 2005;11:367–368. doi: 10.1038/nm0405-367. [DOI] [PubMed] [Google Scholar]
  • 27.Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103:1204–1219. doi: 10.1161/CIRCRESAHA.108.176826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Duran JM, Makarewich CA, Sharp TE, Starosta T, Zhu F, Hoffman NE, Chiba Y, Madesh M, Berretta RM, Kubo H, Houser SR. Bone-derived stem cells repair the heart after myocardial infarction through transdifferentiation and paracrine signaling mechanisms. Circ Res. 2013;113:xxx–xxx. doi: 10.1161/CIRCRESAHA.113.301202. in this issue. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhu H, Guo ZK, Jiang XX, Li H, Wang XY, Yao HY, Zhang Y, Mao N. A protocol for isolation and culture of mesenchymal stem cells from mouse compact bone. Nat Protoc. 2010;5:550–560. doi: 10.1038/nprot.2009.238. [DOI] [PubMed] [Google Scholar]
  • 30.Gu M, Nguyen PK, Lee AS, Xu D, Hu S, Plews JR, Han L, Huber BC, Lee WH, Gong Y, de Almeida PE, Lyons J, Ikeno F, Pacharinsak C, Connolly AJ, Gambhir SS, Robbins RC, Longaker MT, Wu JC. Microfluidic single-cell analysis shows that porcine induced pluripotent stem cell-derived endothelial cells improve myocardial function by paracrine activation. Circ Res. 2012;111:882–893. doi: 10.1161/CIRCRESAHA.112.269001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Guo Z, Li H, Li X, Yu X, Wang H, Tang P, Mao N. In vitro characteristics and in vivo immunosuppressive activity of compact bone-derived murine mesenchymal progenitor cells. Stem Cells. 2006;24:992–1000. doi: 10.1634/stemcells.2005-0224. [DOI] [PubMed] [Google Scholar]
  • 32.Huang M, Nguyen P, Jia F, Hu S, Gong Y, de Almeida PE, Wang L, Nag D, Kay MA, Giaccia AJ, Robbins RC, Wu JC. Double knockdown of prolyl hydroxylase and factor-inhibiting hypoxia-inducible factor with nonviral minicircle gene therapy enhances stem cell mobilization and angiogenesis after myocardial infarction. Circulation. 2011;124:S46–54. doi: 10.1161/CIRCULATIONAHA.110.014019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Li TS, Cheng K, Malliaras K, Smith RR, Zhang Y, Sun B, Matsushita N, Blusztajn A, Terrovitis J, Kusuoka H, Marban L, Marban E. Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells. J Am Coll Cardiol. 2012;59:942–953. doi: 10.1016/j.jacc.2011.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tang XL, Rokosh G, Sanganalmath SK, Yuan F, Sato H, Mu J, Dai S, Li C, Chen N, Peng Y, Dawn B, Hunt G, Leri A, Kajstura J, Tiwari S, Shirk G, Anversa P, Bolli R. Intracoronary administration of cardiac progenitor cells alleviates left ventricular dysfunction in rats with a 30-day-old infarction. Circulation. 2010;121:293–305. doi: 10.1161/CIRCULATIONAHA.109.871905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sheikh AY, Huber BC, Narsinh KH, Spin JM, van der Bogt K, de Almeida PE, Ransohoff KJ, Kraft DL, Fajardo G, Ardigo D, Ransohoff J, Bernstein D, Fischbein MP, Robbins RC, Wu JC. In vivo functional and transcriptional profiling of bone marrow stem cells after transplantation into ischemic myocardium. Arterioscler Thromb Vasc Biol. 2012;32:92–102. doi: 10.1161/ATVBAHA.111.238618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Li Z, Lee A, Huang M, Chun H, Chung J, Chu P, Hoyt G, Yang P, Rosenberg J, Robbins RC, Wu JC. Imaging survival and function of transplanted cardiac resident stem cells. J Am Coll Cardiol. 2009;53:1229–1240. doi: 10.1016/j.jacc.2008.12.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.van der Bogt KE, Schrepfer S, Yu J, Sheikh AY, Hoyt G, Govaert JA, Velotta JB, Contag CH, Robbins RC, Wu JC. Comparison of transplantation of adipose tissue- and bone marrow-derived mesenchymal stem cells in the infarcted heart. Transplantation. 2009;87:642–652. doi: 10.1097/TP.0b013e31819609d9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.van der Bogt KE, Sheikh AY, Schrepfer S, Hoyt G, Cao F, Ransohoff KJ, Swijnenburg RJ, Pearl J, Lee A, Fischbein M, Contag CH, Robbins RC, Wu JC. Comparison of different adult stem cell types for treatment of myocardial ischemia. Circulation. 2008;118:S121–129. doi: 10.1161/CIRCULATIONAHA.107.759480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Swijnenburg RJ, Govaert JA, van der Bogt KE, Pearl JI, Huang M, Stein W, Hoyt G, Vogel H, Contag CH, Robbins RC, Wu JC. Timing of bone marrow cell delivery has minimal effects on cell viability and cardiac recovery after myocardial infarction. Circ Cardiovasc Imaging. 2010;3:77–85. doi: 10.1161/CIRCIMAGING.109.872085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Haider H, Ashraf M. Strategies to promote donor cell survival: Combining preconditioning approach with stem cell transplantation. J Mol Cell Cardiol. 2008;45:554–566. doi: 10.1016/j.yjmcc.2008.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Oskouei BN, Lamirault G, Joseph C, Treuer AV, Landa S, Da Silva J, Hatzistergos K, Dauer M, Balkan W, McNiece I, Hare JM. Increased potency of cardiac stem cells compared with bone marrow mesenchymal stem cells in cardiac repair. Stem Cells Transl Med. 2012;1:116–124. doi: 10.5966/sctm.2011-0015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dey D, Han L, Bauer M, Sanada F, Oikonomopoulos A, Hosoda T, Unno K, De Almeida P, Leri A, Wu JC. Dissecting the molecular relationship among various cardiogenic progenitor cells. Circ Res. 2013;112:1253–1262. doi: 10.1161/CIRCRESAHA.112.300779. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES