51
Views
1
CrossRef citations to date
0
Altmetric
Review

Potential and clinical utility of stem cells in cardiovascular disease

, , &
Pages 49-56 | Published online: 26 Mar 2010

Abstract

The recent identification of bone marrow-derived adult stem cells and other types of stem cells that could improve heart function after transplantation have raised high expectations. The basic mechanisms have been studied mostly in murine models. However, these experiments revealed controversial results on transdifferentiation vs transfusion of adult stem cells vs paracrine effects of these cells, which is still being debated. Moreover, the reproducibility of these results in precisely translated large animal models is still less well investigated. Despite these weaknesses results of several clinical trials including several hundreds of patients with ischemic heart disease have been published. However, there are no solid data showing that any of these approaches can regenerate human myocardium. Even the effectiveness of cell therapy in these approaches is doubtful. In future we need in this important field of regenerative medicine: i) more experimental data in large animals that are closer to the anatomy and physiology of humans, including data on dose effects, comparison of different cell types and different delivery routes; ii) a better understanding of the molecular mechanisms involved in the fate of transplanted cells; iii) more intensive research on genuine regenerative medicine, applying genetic regulation and cell engineering.

Introduction

In some vertebrates such as zebrafish and newts cardiac regeneration was documented after severe injury based on division of cardiomyocytes.Citation1,Citation2 However, there is not yet proof-of-concept that stem cells can regenerate substantially damaged hearts in humans. Reviewing the literature on the clinical utility of human stem cells in cardiovascular diseases demands controversial considerations. First, clinicians have started studies on bone marrow-derived mononuclear cells (BMC) and myoblasts. Second, clinicians have obviously been influenced by publications in 2001 demonstrating that BMC can repair rodent hearts after experimental myocardial infarction.Citation3,Citation4 Neither the intramyocardial injection technique nor the cell type used in these preclinical studies was employed in the first clinical study on BMC therapy in humans with myocardial infarction.Citation5 Furthermore although the paper was entitled “Repair of infarcted myocardium”, the study was a non-randomized feasibility and safety study, and naturally the data could not provide any evidence for anatomical repair or regeneration influenced by the injected cells. Research on clinical use of stem cell therapy in cardiology was started with inadequate study design and misleading terms and definitions. Many questions have been raised on the validity of stem cell therapy research in cardiology. Even the proof-of-concept of regeneration of damaged hearts is highly controversial, since transdifferentiation of BMC into myocytesCitation3,Citation4 could not be reproduced by other groups, and transfusion of myocytes with labeled BMC has been demonstrated, raising the question of misinterpretation of cell transdifferentiation reports.Citation6,Citation7 Scientific progress in cell therapy or regenerative medicine is therefore in danger of being delayed. Do basic researchers and clinicians make the wrong studies? It is questionable that the first clinical trials on cell therapy in cardiology started without exact translation of large animal experiments.Citation5,Citation8 It would be not the first time that an important therapeutic approach in medical science has been delayed in the beginning, because clinical utility has been demanded too eagerly, as occurred, for example, in the first bone marrow transplantation, trials of gene therapy, and the first human heart transplantation.

In the large field of tissue engineering including heart valve engineering, artificial myocardial tissue, and engineered heart tissue, which so far are based mostly on neonatal rat cardiomyocytes, we like to refer on reviews focusing on this important preclinical work.Citation9,Citation10

Embryonic stem cells

In 1998 human embryonic stem cells (hESC) were first isolated.Citation11 These cells have the capacity for unlimited growth and per se transdifferentiation to most of all types of body cells. Mouse ESC had been isolated 17 years before however, there is still no proof that implantation of ESC in humans can cure a single chronic disease.Citation12 Despite the controversial interests between religious and political groups there is also concern for scientific reasons. Immunological reactions and the capability of forming teratomas after injection of undifferentiated ESC into the heart have been described.Citation9,Citation13Citation15 Although others have not observed these findings, and there is no obvious reason why these experiments did not show teratomas, it would be irresponsible to use undifferentiated ESC in humans. On the other hand, genetic selection of ESC as applied by the Field’s groupCitation16 is capable of creating ES-derived myocytes that have been successfully engrafted in the host heart. Engrafting of these ESCs was documented by immunostaining and alignment and apposition with host myocardial cells. Zandstra et alCitation17 selected myosin + cardiomyocytes out of mouse ESC before transfecting with a fusion gene consisting of cardiac myosin and neomycin resistance genes and neomycin treatment afterwards. Retinoic acid seemed to promote cardiac differentiation. Other genetic selection methods include fluorescent protein expression driven by different cardiac promotors.Citation15,Citation18Citation20 Also, other groups showed improvement of left ventricular ejection fraction by echocardiography after mouse ESC implementation.Citation21Citation23 Engineered ESC has not been shown to form teratomas in these reports.

Induced pluripotent stem cells (iPS)

Most recently, adult human somatic cells were reprogrammed to pluripotent stem cells by transduction of four transcription factors: Oct3/4, Sox2, Klf4, and c-Myc.Citation24 This raises great hope for future regenerative therapy.Citation25 Thus, the problems of immunological rejection and embryo destruction can be overcome. The era of real cell replacement therapy could start with the onset of clinical trials after basic researchers and clinicians take account of the lessons learned from cell therapy trials performed. The successful establishment of multipotent adult germline stem cells (maGSCs) from mouse testis has opened another interesting route for true regenerative medicine in cardiovascular diseases. These authors found that maGSCs transplanted into normal hearts of mice were able to proliferate and differentiate. No tumor formation was detected up to 1 month after cell transplantation in these experiments.Citation26,Citation27

Cell cycle activity reinduction in cardiomyocytes could be another promising approach.Citation28,Citation29

Bone marrow-derived cells

Starting with the pioneer works of Kocher et al and Orlic et al,Citation3,Citation4 since 2001 intramyocardial delivery of bone marrow-derived cells have been shown to improve left ventricular function in the ischemically damaged heart.Citation30 Several clinical reports indicate the benefit of bone marrow-derived cells (BMC) grafting in patients with acute myocardial infarction (AMI), including CD133+ and CD34+Citation5,Citation31Citation37 bone marrow-derived mononuclear stem cells (BMC) and mesenchymal stem cells. Recent reports on intracoronary application of BMC in humans exhibit controversial results with regard to the improvement of left ventricular function (LVEF). Whereas the TOPCARE-AMI, REPAIR and IACT studies, and others revealed an increase in LVEFCitation37Citation40 (approx. 3% increase of LVEF) the studies by Janssens et alCitation41 and LundeCitation42 et al could not reproduce the results.

However, it is difficult to compare the results, because most groups that detected an improvement of LVEF in the cell groups showed impairment of LVEF in the control groups and vice versa. In the BOOST trial the placebo group even increased the LVEF more than the cell group after 18 months. Moreover, different imaging methods (MRI, left ventricular angiography, echocardiography) were applied in the different studies. The date of cell injection in patients after acute myocardial infarction ranged from 3 to 12 days, which makes a huge difference, since the stunning phase after revascularization takes place particularly during that time and discrimination between cell effects and discontinuation of the stunning phenomenon is impossible.

Moreover, the clinical studies are not comparable to each other, because different cell types, different isolation and selection techniques, different injection techniques, and different time intervals of cell administration after myocardial infarction are applied in the study protocols. Nonetheless, a meta-analysis of 10 controlled trials enrolling 698 patients after acute myocardial infarction treated with BMC suggest a short-term improvement of LVEF (2.97%).Citation43

Another problem is the shortening of telomeres of BMC which is responsible for decreased replicative capacity.Citation44 This phenomenon is also described in endothelial progenitor cells. Moreover, risk factors for cardiovascular diseases correlate with higher rated of in vitro senescence.Citation45 This was also observed for cardiac stem cells.Citation46,Citation47

Mesenchymal Stem Cells (MSC)

MSC were discovered and described as bone marrow stromal cells with multipotent potential.Citation47,Citation48 Cloned MSCs can differentiate into osteoblasts, adipocytes and chondrocytes.Citation48 Even cardiomyocytes were generated from MSCCitation49in vitro by demethylization with 5-azacytidine. Most studies used undifferentiated MCSs for injection in the damaged heart without differentiation into cardiomyocytes and lack of electromechanical junctions with host cells.Citation50 But attenuation of pathological left ventricular remodeling was observed frequently.Citation50Citation53 This is in accordance with other studies using allogeneic MSC.Citation54,Citation55 Moreover, MSC are described to induce tolerance which could be responsible for the reduction of graft-versus-host disease, rejection, and modulation of inflammation.Citation56,Citation57 These results make MSC interesting cell types for future research, especially for pharmaceutical companies, eg, Osiris Therapeutics.

New ongoing trials like the C-CURE (Cardio3 BioSciences) trial will show new results of intramyocardial delivery of MSC in different dosages in chronic ischemic heart disease. But certainly they will also raise questions about the fate and effectiveness of autologous MSCs.

CD133+ progenitor cells

CD133+ progenitor cells are examples for further cell therapy approaches. They can be extracted from the bone marrow and from peripheral blood after leukapheresis and have been shown to contribute to neoangiogenesis.Citation58 These progenitor cells seem to have similar positive effects on LVEF as BMC in clinical pilot trials.Citation36,Citation59,Citation60 and were used with very few preclinical experimental investigations before.Citation61 Large animal experiments are still lacking.

Myoblasts

Mouse skeletal muscle have been shown to contain a population of precursor cells that can differentiate into beating cells that express cardiomyocyte features.Citation62 In humans satellite cells (myoblasts) have been identified which can differentiate into myotubes and improve left ventricular function in animal experiments.Citation63 But there is no evidence that these cells can differentiate into cardiomyocytes.Citation13

Despite early research on skeletal myoblasts injection in the injured heartCitation63 there are several publications that showed that myoblasts can induce proarrhythmogenic effects and are therefore less suitable for clinical trials (paragraph stem cells and arrhythmogeneity). Intramyocardial myoblast injection therapy studies in patients with ischemic cardiomyopathy have been performed mostly during bypass surgery.Citation8,Citation64 These trials were conducted with simultaneous coronary reperfusion, so that precise discrimination of the cell injection effect and reperfusion therapy is problematic. This MAGIC trial was stopped because of lack of efficacy.

Adipose derived stem cells

Adipocyte-derived stem cells are multilineage cells within the stroma vascular fraction of subcutaneous adipose tissue. These cells express surface markers Sca-1 and CD44, but not CD34, CD31, c-kit and CD45.Citation65 They have angiogenic potential by secreting of VEGF and HGF.Citation65,Citation66 Even cardiomyocyte-like transdifferentiation, in vitro expansion and cardioprotective effects in a mouse-infarction model has been decribed.Citation67

Resident myocardial progenitor cells

Recently, the myocardium – formerly known as a postmitotic tissue – was shown to contain cells with regenerative capacity like other tissues, eg, the skin, bone marrow, intestine.Citation68Citation70 These cardiac stem cells show a reentry into the cell cycle, are clonogenic, self-renewing and multipotentCitation69 and capable to regenerate the ischemic myocardium after injection into the border zone of myocardial infarction. Only a small number of these cells are distributed in the atria, the ventricles, and the epicardium. Although, resident cardiac stem cells have been shown to have the potential to differentiate into cardiomyocytes, endothelial, and smooth muscle cells,Citation69,Citation71 this occurs at a very low rate.Citation72 However, these cardiac stem cells could be isolated and amplified in vitro and therefore are interesting candidates for induction of myocardial regeneration.Citation46,Citation73Citation76 Recently, evidence of renewal of human cardiomyocytes (1% annually at the age of 25) was suggested by analysis of carbon-14 integration in human cardiomyocyte DNA, but whether this renewal derives from residential stem cells or cardiomyocyte duplication is not clear.Citation77

Paracrine effects of stem cells

Recently some experimental data have confirmed paracrine effects of stem cells, including angiogenesis and antiapoptotic effects.Citation7,Citation78Citation81 Particularly MSC were investigated and some studies provide evidence for this hypothesis.Citation80,Citation81 Dai et alCitation54 and Tang et alCitation79 reported on the enhanced expression of bFGF, SDF1-alpha, and vascular endothelial growth factor (VEGF) accompanied by a downregulation of proapoptotic protein Bax in ischemic myocardium after MSC implantation. Cultured MSC were shown to secret large amounts of angiogenic, anti-apoptotic, hepatocyte growth factor and insulin-like growth factor-1.Citation82 It seems that the beneficial effects of MSC are mediated by inhibition of myocardial fibrosis and an increase of angiogenesis and not by transdifferentiation.

Paracrine effects of myoblasts have also been described more recently.Citation83 These effects include proangiogenic (PDF), anti-apoptotic (BAG-1, BCL-2) and extracellular matrix remodeling (MMP-2, MMP-7) genes.

It can be assumed that paracrine and angiogenic effects are responsible for the cardioprotective effects on heart function as described in other studies.Citation55,Citation79,Citation80,Citation84,Citation85

However, the term “regenerative medicine”, if applied specifically to stem cell engraftment and forming new differentiated myocytes, would then be misleading.

Stem cells and arrhythmogenicity

Safety concerns on the injection of skeletal myoblasts in the chronic ischemic myocardium in clinical studies were raised after documentation of ventricular tachycardias.Citation8,Citation86 With regard to the implantation of myoblasts in clinical studies, most ventricular arrhythmic events occurred in the first 4 weeks after cell implantation. In 4 out of 10 patients, ventricular tachycardias occurred between 11 and 22 days;Citation8 in similar studies 2 patients out of 21 developed ventricular tachycardias 1 day after coronary artery bypass surgery.Citation64,Citation87

Mechanisms of arrhythmia include re-entry, ectopy, and automaticity and potentially the heterogeneity of action potentials of implanted cells. Particularly, implantation of myoblasts has been shown to create an arrhythmogenic substrate, because the implanted cells do not build junctions to the surrounding myocytes.Citation88 In these studies myoblasts labeled with Green fluorescent protein were transplanted into rat infarcted myocardium, and differentiated into peculiar “hyperexcitable myotubes with a contractile activity fully independent of neighbouring cardiomyocytes”Citation88 or were located in cell clusters without connexin expression.Citation89 Unfortunately, this analysis was published after performance of clinical trials.

Arrhythmogenicity can be induced through re-entry, automaticity, or triggered activity. All these phenomena can be investigated in large animal models, but publications on this topic in these models are rare.

Only few studies have addressed the electrophysiological properties of transplanted mesenchymal stem cells (MSC), although at present these cells are being studied in several cardiological clinical studies.Citation90

Some authors have described that mixtures of MSC and neonatal rat cardiomyocytes exhibit an arrhythmogenic substrate with decreased conduction velocity and easily inducible sustained re-entrant tachycardia, suggesting a proarrhythmic substrate induced by MSC.Citation91,Citation92 Chen et al reported that MSC prolonged local activation time and increase the activation time dispersion in a rabbit heart failure model.Citation93 Thus, the therapeutic potential of mesenchymal stem cells for myocardial regeneration may be limited by proarrhythmic effects. Moreover, MSC have been described to alter electrophysiological properties in a large animal model.Citation94 The authors described a significantly shorter epicardial effective refractory period at all pacing sites 3 months after pigs received MSC intravenously, suggesting a proarrhythmic potential.

Our group evaluated the electrophysiological effects of intramyocardial MSC injection based on a three-dimensional electromechanical mapping technique in vivo. We found no change in conduction velocity and no evidence of a substrate modification towards a higher risk for re-entry tachycardias in the area of injected cells in a post-infarction porcine model.Citation51 The results of the study by Mills et al revealed that intravenous MSC infusion (contrary to our study using intramyocardial injection) in a rat acute infarction model even tended to reduce arrhythmia inducibility.Citation95 In that study MSC enhanced electrical viability and preserved impulse propagation in the infarct border zone as demonstrated by an optical mapping system. Histologically MSC shows a diffuse engraftment in the host myocardium and expressed connexin.

In clinical studies implanting BMC intramyocardially in patients with chronic ischemic heart disease the incidence of ventricular arrhythmia do not increase. These clinical studies were performed using the NOGA mapping system in patients with myocardial ischemia and no revascularisation optionCitation96Citation98 showing improvement of regional wall motion and perfusion, and partly of ejection fraction. In the study by Perin et al, 1 patient died 14 weeks after BMC implantation, presumably of sudden cardiac death.Citation98 In 20 patients treated with BMC Beeres et al could not find an increased incidence of ventricular arrhythmias in holter monitoring. Corresponding to our preclinical results,Citation51 BMC did not alter electrophysiological properties evaluated with electromechanical mapping before and 3 months after cell injection.Citation99 Programmed ventricular stimulation was performed only in 1 randomized clinical study which found no differences between the BMC group and the control group.Citation32 This study included intracoronary infusion in patients with acute myocardial infarction. In clinical studies infusing BMC, no proarrhythmic effect has yet been described.

For the transplantation of ESC-derived myocytes, further studies should address the arrhythmogenic potential in large animal models. Zhang et al described an increase of arrhythmic potential of embryonic stem cell derived cardiomyocytes.Citation100 These cardiomyocytes showed spontaneous activity, prolonged potential duration of action, and easily inducible arrhythmias, and therefore the arrhythmogeneic potential should be carefully investigated.

Cell delivery techniques

Our group applied intramyocardial injection, because this is by far the most-used delivery technique in preclinical studies.Citation101 Moreover we tested efficacy and safety in a large animal model (swine) before starting a trial in humans.Citation52,Citation102 It is remarkable that most investigators prefer the intracoronary route, despite the fact that very few animal studies exist on this technique, and most studies have used rodents with intramyocardial cell injections. It is not necessary to use a different delivery route in humans. A simple explanation might be the fact that experimental studies on intracoronary cell injection are expensive due to the need for large animals. In contrast, in humans intracoronary cell delivery is cheaper and less time consuming for interventional cardiologists, because no left ventricular mapping is needed as for intramyocardial cell injection. Moreover the intramyocardial cell delivery technique is more evident, since preclinical experiments using radiolabeled cells indicate that intracoronary infusion results in a myocardial cell residency of 2.6% compared with 11.0% using the intramyocardial injection approach.Citation103 Therefore, intramyocardial injection may have a more pronounced effect on LV function.Citation104 Furthermore, intracoronary cell injection was associated with an increased remote organ engraftment compared with the intramyocardial injection in both an animal model as well as a clinical study.Citation103,Citation105

Challenges and future directions

The rapid translation of preclinical cell-based therapy to restore damaged myocardium has raised questions concerning the best cell type as well as the best delivery route, and the best time of cell injection into the myocardium. All these questions should be addressed and challenged by the Task Force of the European Society of Cardiology,Citation106 especially as several new clinical trials are in progress in the United States, Europe and in Asia without standardization of methods (cell harvest, isolation, preparation, delivery, dosage). Pharmacokinetics and pharmacodynamics should be assessed in new drugs as well as in cell therapy. In a review by Murry et al it was pointed out that cell dosages ranged by 6700 fold in the published trials,Citation30 which underline the need for standardization of cell products. Instead, the Task Force of the ESC stresses a “pragmatic approach to demonstrate clinical efficacy” and does not recommend testing all possible combinations of cell types, number of cells, and so on, in animal models, because it “would take the best part of the century”. Moreover, an extension to cardiomyopathy patients was demanded. In view of the controversies outlined in this review, these statements should be questioned.

Reviewing the literature on preclinical investigations, it is remarkable that nearly every cell type tested so far seems to be equipotent for positive effects on LV function independent of the timing after myocardial infarction, cell numbers and the methods used.Citation30 Therefore, experimental models to directly compare different cell types should be evaluatedCitation15,Citation51,Citation107 to reduce investigator-related bias.

Most reports use rodent models not recognizing that there are major differences between rodents and large mammals in mechanisms of myocardial contraction and ischemia.Citation108 Also, there is a wide spectrum of collateral flow between various mammalian species especially for the lack of pre-existing collaterals in the swine heart, but also from a metabolic standpoint in comparision with rodents.

Most clinical studies do not adapt the protocols from experimental studies, making comparsion with preclinical work difficult. Rodent hearts seem to be regenerative. However, the murine heart is more likely to regenerate with a billion cells than a human heart weighing approximately 300 to 500 g. Certainly less than 5 g of heart tissue would be regenerated with these cell counts applied in clinical studies. None of the clinically used imaging techniques available are sensitive enough to detect the anatomical and functional contribution of 5 g of myocardial tissue, even if all cell survived, nested and transdifferentiated. Approximately 1 billion cells should transdifferentiate into cardiomyocytes and contract synchronously with host myocardium to restore ischemia-induced cardiac damage.Citation30 Thus, even the possibility of self-renewing of the human heartCitation109 is naturally limited to induce relevant effects in damaged hearts.

What we need for new clinical trials are

  1. biologyWhat is the fate of injected cells in humans? Is there a long-term effect of injected cell in large animal experiments? What dosage of cells is optimal in large animal experiments? Which cells are responsible for which biological effect? We need a better understanding of the molecular mechanisms involved in the fate of transplanted cells.

  2. technical evaluationWhat delivery route is most effective? What is the best point of time to deliver cells? What are the dose effects?

  3. intensifying the research on new approaches for stem cell engineering and the research on genuine regenerative medicine applying genetic regulation.

Disclosure

KK has received educational honoraria from BDS, USA.

References

  • PossKDGetting to the heart of regeneration in zebrafishSemin Cell Dev Biol200718364517178459
  • BorchardtTBraunTCardiovascular regeneration in non-mammalian model systems: what are the differences between newts and man?Thromb Haemost20079831131817721612
  • OrlicDKajsturaJChimentiSBone marrow cells regenerate infarcted myocardiumNature200141070170511287958
  • KocherAASchusterMDSzabolcsMJNeovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac functionNat Med2001743043611283669
  • StrauerBEBrehmMZeusTRepair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humansCirculation20021061913191812370212
  • MurryCESoonpaaMHReineckeHHaematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarctsNature200442866466815034593
  • ZiegelhoefferTFernandezBKostinSBone marrow-derived cells do not incorporate into the adult growing vasculatureCirc Res20049423023814656934
  • MenaschePHagegeAAVilquinJTAutologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunctionJ Am Coll Cardiol2003411078108312679204
  • ErdoFBuhrleCBlunkJHost-dependent tumorigenesis of embryonic stem cell transplantation in experimental strokeJ Cereb Blood Flow Metab20032378078512843782
  • ZimmermannWHDidieMDokerSHeart muscle engineering: an update on cardiac muscle replacement therapyCardiovasc Res20067141942916697358
  • ThomsonJAItskovitz-EldorJShapiroSSEmbryonic stem cell lines derived from human blastocystsScience1998282114511479804556
  • EvansMJKaufmanMHEstablishment in culture of pluripotential cells from mouse embryosNature19812921541567242681
  • LaflammeMAMurryCERegenerating the heartNat Biotechnol20052384585616003373
  • RiessPMolcanyiMBentzKEmbryonic stem cell transplantation after experimental traumatic brain injury dramatically improves neurological outcome, but may cause tumorsJ Neurotrauma20072421622517263685
  • KolossovEBostaniTRoellWEngraftment of engineered ES cell-derived cardiomyocytes but not BM cells restores contractile function to the infarcted myocardiumJ Exp Med20062032315232716954371
  • KlugMGSoonpaaMHKohGYFieldLJGenetically selected cardiomyocytes from differentiating embronic stem cells form stable intracardiac graftsJ Clin Invest1996982162248690796
  • ZandstraPWBauwensCYinTScalable production of embryonic stem cell-derived cardiomyocytesTissue Eng2003976777813678453
  • HidakaKLeeJKKimHSChamber-specific differentiation of Nkx2.5-positive cardiac precursor cells from murine embryonic stem cellsFaseb J20031774074212594186
  • KolossovELuZDrobinskayaIIdentification and characterization of embryonic stem cell-derived pacemaker and atrial cardiomyocytesFaseb J20051957757915659535
  • MeyerNJaconiMLandopoulouAFortPPuceatMA fluorescent reporter gene as a marker for ventricular specification in ES-derived cardiac cellsFEBS Lett200047815115810922488
  • MinJYYangYConversoKLTransplantation of embryonic stem cells improves cardiac function in postinfarcted ratsJ Appl Physiol20029228829611744672
  • MinJYYangYSullivanMFLong-term improvement of cardiac function in rats after infarction by transplantation of embryonic stem cellsJ Thorac Cardiovasc Surg200312536136912579106
  • HodgsonDMBehfarAZingmanLVStable benefit of embryonic stem cell therapy in myocardial infarctionAm J Physiol Heart Circ Physiol2004287H471H47915277190
  • TakahashiKTanabeKOhnukiMInduction of pluripotent stem cells from adult human fibroblasts by defined factorsCell200713186187218035408
  • TanakaTTohyamaSMurataMIn vitro pharmacologic testing using human induced pluripotent stem cell-derived cardiomyocytesBiochem Biophys Res Commun200938549750219464263
  • GuanKWagnerSUnsoldBGeneration of functional cardiomyocytes from adult mouse spermatogonial stem cellsCirc Res20071001615162517478732
  • GuanKNayerniaKMaierLSWagnerSPluripotency of spermatogonial stem cells from adult mouse testisNature20064401199120316565704
  • FieldLJModulation of the cardiomyocyte cell cycle in genetically altered animalsAnn N Y Acad Sci2004101516017015201157
  • CampaVMGutierrez-LanzaRCerignoliFNotch activates cell cycle reentry and progression in quiescent cardiomyocytesJ Cell Biol200818312914118838555
  • MurryCEReineckeHPabonLMRegeneration gaps: observations on stem cells and cardiac repairJ Am Coll Cardiol2006471777178516682301
  • AssmusBHonoldJSchachingerVTranscoronary transplantation of progenitor cells after myocardial infarctionN Engl J Med20063551222123216990385
  • MeyerGPWollertKCLotzJIntracoronary bone marrow cell transfer after myocardial infarction: eighteen months’ follow-up data from the randomized, controlled BOOST (BOne marrOw transfer to enhance ST-elevation infarct regeneration) trialCirculation20061131287129416520413
  • ChenSLFangWWYeFEffect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarctionAm J Cardiol200494929515219514
  • SchachingerVErbsSElsasserAImproved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial infarction: final 1-year results of the REPAIR-AMI trialEur Heart J2006272775278317098754
  • AssmusBWalterDHLehmannRIntracoronary infusion of progenitor cells is not associated with aggravated restenosis development or atherosclerotic disease progression in patients with acute myocardial infarctionEur Heart J20062729852995
  • BartunekJVanderheydenMVandekerckhoveBIntracoronary injection of CD133-positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safetyCirculation2005112178183
  • SchachingerVAssmusBBrittenMBTransplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI TrialJ Am Coll Cardiol2004441690169915489105
  • AssmusBSchachingerVTeupeCTransplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI)Circulation20021063009301712473544
  • Fernandez-AvilesFSan RomanJAGarcia-FradeJExperimental and clinical regenerative capability of human bone marrow cells after myocardial infarctionCirc Res20049574274815358665
  • StrauerBEBrehmMZeusTRegeneration of human infarcted heart muscle by intracoronary autologous bone marrow cell transplantation in chronic coronary artery disease: the IACT StudyJ Am Coll Cardiol2005461651165816256864
  • JanssensSDuboisCBogaertJAutologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trialLancet200636711312116413875
  • LundeKSolheimSAakhusSIntracoronary injection of mononuclear bone marrow cells in acute myocardial infarctionN Engl J Med20063551199120916990383
  • LipinskiMJBiondi-ZoccaiGGAbbateAImpact of intracoronary cell therapy on left ventricular function in the setting of acute myocardial infarction: a collaborative systematic review and meta-analysis of controlled clinical trialsJ Am Coll Cardiol2007501761176717964040
  • AnversaPKajsturaJLeriABolliRLife and death of cardiac stem cells: a paradigm shift in cardiac biologyCirculation20061131451146316549650
  • HillJMZalosGHalcoxJPCirculating endothelial progenitor cells, vascular function, and cardiovascular riskN Engl J Med200334859360012584367
  • UrbanekKTorellaDSheikhFMyocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failureProc Natl Acad Sci U S A20051028692869715932947
  • CaplanAIMesenchymal stem cellsJ Orthop Res199196416501870029
  • PittengerMFMackayAMBeckSCMultilineage potential of adult human mesenchymal stem cellsScience199928414314710102814
  • HakunoDFukudaKMakinoSBone marrow-derived regenerated cardiomyocytes (CMG Cells) express functional adrenergic and muscarinic receptorsCirculation200210538038611804996
  • ShakeJGGruberPJBaumgartnerWAMesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effectsAnn Thorac Surg20027319191925 discussion 192612078791
  • KrauseKSchneiderCLangeCEndocardial electrogram analysis after intramyocardial injection of mesenchymal stem cells in the chronic ischemic myocardium pacingClin Electrophysiol20093213191328
  • SchneiderCKrauseKJaquetKIntramyocardial transplantation of bone marrow-derived stem cells: ultrasonic strain rate imaging in a model of hibernating myocardiumJ Card Fail20081486187219041051
  • AmadoLCSaliarisAPSchuleriKHCardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarctionProc Natl Acad Sci U S A2005102114741147916061805
  • DaiWHaleSLMartinBJAllogeneic mesenchymal stem cell transplantation in postinfarcted rat myocardium: short- and long-term effectsCirculation200511221422315998673
  • KinnairdTStabileEBurnettMSLocal delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanismsCirculation20041091543154915023891
  • AggarwalSPittengerMFHuman mesenchymal stem cells modulate allogeneic immune cell responsesBlood20051051815182215494428
  • Le BlancKImmunomodulatory effects of fetal and adult mesenchymal stem cellsCytotherapy2003548548914660044
  • ReyesMDudekAJahagirdarBKoodieLMarkerPHVerfaillieCMOrigin of endothelial progenitors in human postnatal bone marrowJ Clin Invest200210933734611827993
  • ManginasAGoussetisEKoutelouMPilot study to evaluate the safety and feasibility of intracoronary CD133(+) and CD133(−) CD34(+) cell therapy in patients with nonviable anterior myocardial infarctionCatheter Cardiovasc Interv20076977378117394248
  • StammCWestphalBKleineHDAutologous bone-marrow stem-cell transplantation for myocardial regenerationLancet2003361454612517467
  • AgbulutOVanderveldeSAl AttarNComparison of human skeletal myoblasts and bone marrow-derived CD133+ progenitors for the repair of infarcted myocardiumJ Am Coll Cardiol20044445846315261948
  • WinitskySOGopalTVHassanzadehSAdult murine skeletal muscle contains cells that can differentiate into beating cardiomyocytes in vitroPLoS Biol20053e8715757365
  • DowellJDRubartMPasumarthiKBSoonpaaMHFieldLJMyocyte and myogenic stem cell transplantation in the heartCardiovasc Res20035833635012757868
  • HerrerosJProsperFPerezAAutologous intramyocardial injection of cultured skeletal muscle-derived stem cells in patients with non-acute myocardial infarctionEur Heart J2003242012202014613737
  • Planat-BenardVSilvestreJSCousinBPlasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectivesCirculation200410965666314734516
  • Planat-BenardVMenardCAndreMSpontaneous cardiomyocyte differentiation from adipose tissue stroma cellsCirc Res20049422322914656930
  • LeobonBRoncalliJJoffreCAdipose-derived cardiomyogenic cells: in vitro expansion and functional improvement in a mouse model of myocardial infarctionCardiovasc Res20098375776719505931
  • BeltramiAPUrbanekKKajsturaJEvidence that human cardiac myocytes divide after myocardial infarctionN Engl J Med20013441750175711396441
  • BeltramiAPBarlucchiLTorellaDAdult cardiac stem cells are multipotent and support myocardial regenerationCell200311476377614505575
  • WinterEMGraussRWHogersBPreservation of left ventricular function and attenuation of remodeling after transplantation of human epicardium-derived cells into the infarcted mouse heartCirculation200711691792717684151
  • OhHBradfuteSBGallardoTDCardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarctionProc Natl Acad Sci U S A2003100123131231814530411
  • HsiehPCSegersVFDavisMEEvidence from a genetic fate-mapping study that stem cells refresh adult mammalian cardiomyocytes after injuryNat Med20071397097417660827
  • MessinaEDe AngelisLFratiGIsolation and expansion of adult cardiac stem cells from human and murine heartCirc Res20049591192115472116
  • BarileLChimentiIGaetaniRCardiac stem cells: isolation, expansion and experimental use for myocardial regenerationNat Clin Pract Cardiovasc Med20074Suppl 1S9S1417230222
  • UrbanekKRotaMCascaperaSCardiac stem cells possess growth factor-receptor systems that after activation regenerate the infarcted myocardium, improving ventricular function and long-term survivalCirc Res20059766367316141414
  • TorellaDEllisonGMMendez-FerrerSIbanezBNadal-GinardBResident human cardiac stem cells: role in cardiac cellular homeostasis and potential for myocardial regenerationNat Clin Pract Cardiovasc Med20063Suppl 1S8S1316501638
  • BergmannOBhardwajRDBernardSEvidence for cardiomyocyte renewal in humansScience20093249810219342590
  • KubalCShethKNadal-GinardBGalinanesMBone marrow cells have a potent anti-ischemic effect against myocardial cell death in humansJ Thorac Cardiovasc Surg20061321112111817059931
  • TangYLZhaoQQinXParacrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarctionAnn Thorac Surg200580229236 discussion 236–22715975372
  • GnecchiMHeHLiangODParacrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cellsNat Med20051136736815812508
  • GnecchiMHeHNoiseuxNEvidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvementFaseb J20062066166916581974
  • NagayaNFujiiTIwaseTIntravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesisAm J Physiol Heart Circ Physiol2004287H2670H267615284059
  • Perez-IlzarbeMAgbulutOPelachoBCharacterization of the paracrine effects of human skeletal myoblasts transplanted in infarcted myocardiumEur J Heart Fail2008101065107218805052
  • KamihataHMatsubaraHNishiueTImplantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokinesCirculation20011041046105211524400
  • ZengLHuQWangXBioenergetic and functional consequences of bone marrow-derived multipotent progenitor cell transplantation in hearts with postinfarction left ventricular remodelingCirculation20071151866187517389266
  • MenaschePHagegeAScorsinMAutologous skeletal myoblast transplantation for cardiac insufficiency. First clinical caseArch Mal Coeur Vaiss20019418018211338251
  • SiminiakTBurchardtPKurpiszMPostinfarction heart failure: surgical and trans-coronary-venous transplantation of autologous myoblastsNat Clin Pract Cardiovasc Med20063Suppl 1S46S5116501631
  • LeobonBGarcinIMenaschePVilquinJTAudinatECharpakSMyoblasts transplanted into rat infarcted myocardium are functionally isolated from their hostProc Natl Acad Sci U S A20031007808781112805561
  • FoutsKFernandesBMalNLiuJLauritaKRElectrophysiological consequence of skeletal myoblast transplantation in normal and infarcted canine myocardiumHeart Rhythm2006345246116567294
  • KatritsisDGSotiropoulouPAKarvouniETranscoronary transplantation of autologous mesenchymal stem cells and endothelial progenitors into infarcted human myocardiumCatheter Cardiovasc Interv20056532132915954106
  • ChangMGTungLSekarRBProarrhythmic potential of mesenchymal stem cell transplantation revealed in an in vitro coculture modelCirculation20061131832184116606790
  • BeeresSLAtsmaDEvan der LaarseAHuman adult bone marrow mesenchymal stem cells repair experimental conduction block in rat cardiomyocyte culturesJ Am Coll Cardiol2005461943195216286184
  • ChenMFanZCLiuXJEffects of autologous stem cell transplantation on ventricular electrophysiology in doxorubicin-induced heart failureCell Biol Int20063057658216731012
  • PriceMJChouCCFrantzenMIntravenous mesenchymal stem cell therapy early after reperfused acute myocardial infarction improves left ventricular function and alters electrophysiologic propertiesInt J Cardiol200611123123916246440
  • MillsWRMalNKiedrowskiMJStem cell therapy enhances electrical viability in myocardial infarctionJ Mol Cell Cardiol20074230431417070540
  • FuchsSKornowskiRWeiszGSafety and feasibility of transendocardial autologous bone marrow cell transplantation in patients with advanced heart diseaseAm J Cardiol20069782382916516583
  • TseHFThambarSKwongYLSafety of catheter-based intramyocardial autologous bone marrow cells implantation for therapeutic angiogenesisAm J Cardiol200698606216784921
  • PerinECDohmannHFBorojevicRImproved exercise capacity and ischemia 6 and 12 months after transendocardial injection of autologous bone marrow mononuclear cells for ischemic cardiomyopathyCirculation2004110II213II21815364865
  • BeeresSLZeppenfeldKBaxJJElectrophysiological and arrhythmogenic effects of intramyocardial bone marrow cell injection in patients with chronic ischemic heart diseaseHeart Rhythm2007425726517341383
  • ZhangYMHartzellCNarlowMDudleySCJrStem cell-derived cardiomyocytes demonstrate arrhythmic potentialCirculation20021061294129912208808
  • KrauseKJaquetKSchneiderCPercutaneous intramyocardial stem cell injection in patients with acute myocardial infarction: first-in-man studyHeart2009951145115219336430
  • SchneiderCJaquetKGeidelSTransplantation of bone marrow-derived stem cells improves myocardial diastolic function: strain rate imaging in a model of hibernating myocardiumJ Am Soc Echocardiogr2009
  • HouDYoussefEABrintonTJRadiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: implications for current clinical trialsCirculation2005112I150I15616159808
  • PerinECSilvaGVAssadJAComparison of intracoronary and transendocardial delivery of allogeneic mesenchymal cells in a canine model of acute myocardial infarctionJ Mol Cell Cardiol20084448649518061611
  • HofmannMWollertKCMeyerGPMonitoring of bone marrow cell homing into the infarcted human myocardiumCirculation20051112198220215851598
  • BartunekJDimmelerSDrexlerHThe consensus of the task force of the European Society of Cardiology concerning the clinical investigation of the use of autologous adult stem cells for repair of the heartEur Heart J2006271338134016543252
  • van der BogtKESheikhAYSchrepferSComparison of different adult stem cell types for treatment of myocardial ischemiaCirculation2008118S121S12918824743
  • HughesGCPostMJSimonsMAnnexBHTranslational physiology: porcine models of human coronary artery disease: implications for preclinical trials of therapeutic angiogenesisJ Appl Physiol2003941689170112679343
  • QuainiFUrbanekKBeltramiAPChimerism of the transplanted heartN Engl J Med200234651511777997