Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Circ Res ; 122(7): 994-1005, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29367212

ABSTRACT

RATIONALE: Intracoronary administration of cardiosphere-derived cells (CDCs) in patients with single ventricles resulted in a short-term improvement in cardiac function. OBJECTIVE: To test the hypothesis that CDC infusion is associated with improved cardiac function and reduced mortality in patients with heart failure. METHODS AND RESULTS: We evaluated the effectiveness of CDCs using an integrated cohort study in 101 patients with single ventricles, including 41 patients who received CDC infusion and 60 controls treated with staged palliation alone. Heart failure with preserved ejection fraction (EF) or reduced EF was stratified by the cardiac function after surgical reconstruction. The main outcome measure was to evaluate the magnitude of improvement in cardiac function and all-cause mortality at 2 years. Animal studies were conducted to clarify the underlying mechanisms of heart failure with preserved EF and heart failure with reduced EF phenotypes. At 2 years, CDC infusion increased ventricular function (stage 2: +8.4±10.0% versus +1.6±6.4%, P=0.03; stage 3: +7.9±7.5% versus -1.1±5.5%, P<0.001) compared with controls. In all available follow-up data, survival did not differ between the 2 groups (log-rank P=0.225), whereas overall patients treated by CDCs had lower incidences of late failure (P=0.022), adverse events (P=0.013), and catheter intervention (P=0.005) compared with controls. CDC infusion was associated with a lower risk of adverse events (hazard ratio, 0.411; 95% CI, 0.179-0.942; P=0.036). Notably, CDC infusion reduced mortality (P=0.038) and late complications (P<0.05) in patients with heart failure with reduced EF but not with heart failure with preserved EF. CDC-treated rats significantly reversed myocardial fibrosis with differential collagen deposition and inflammatory responses between the heart failure phenotypes. CONCLUSIONS: CDC administration in patients with single ventricles showed favorable effects on ventricular function and was associated with reduced late complications except for all-cause mortality after staged procedures. Patients with heart failure with reduced EF but not heart failure with preserved EF treated by CDCs resulted in significant improvement in clinical outcome. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifiers: NCT01273857 and NCT01829750.


Subject(s)
Heart Defects, Congenital/therapy , Heart Failure/therapy , Heart Ventricles/abnormalities , Myoblasts, Cardiac/transplantation , Stem Cell Transplantation/methods , Child, Preschool , Female , Heart Defects, Congenital/complications , Heart Failure/etiology , Heart Failure/prevention & control , Hemodynamics , Humans , Infant , Male , Stem Cell Transplantation/adverse effects , Ventricular Function
3.
Circ Res ; 123(5): 579-589, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29921651

ABSTRACT

RATIONALE: Allogeneic cardiac stem cells (AlloCSC-01) have shown protective, immunoregulatory, and regenerative properties with a robust safety profile in large animal models of heart disease. OBJECTIVE: To investigate the safety and feasibility of early administration of AlloCSC-01 in patients with ST-segment-elevation myocardial infarction. METHODS AND RESULTS: CAREMI (Safety and Efficacy of Intracoronary Infusion of Allogeneic Human Cardiac Stem Cells in Patients With STEMI and Left Ventricular Dysfunction) was a phase I/II multicenter, randomized, double-blind, placebo-controlled trial in patients with ST-segment-elevation myocardial infarction, left ventricular ejection fraction ≤45%, and infarct size ≥25% of left ventricular mass by cardiac magnetic resonance, who were randomized (2:1) to receive AlloCSC-01 or placebo through the intracoronary route at days 5 to 7. The primary end point was safety and included all-cause death and major adverse cardiac events at 30 days (all-cause death, reinfarction, hospitalization because of heart failure, sustained ventricular tachycardia, ventricular fibrillation, and stroke). Secondary safety end points included major adverse cardiac events at 6 and 12 months, adverse events, and immunologic surveillance. Secondary exploratory efficacy end points were changes in infarct size (percentage of left ventricular mass) and indices of ventricular remodeling by magnetic resonance at 12 months. Forty-nine patients were included (92% male, 55±11 years), 33 randomized to AlloCSC-01 and 16 to placebo. No deaths or major adverse cardiac events were reported at 12 months. One severe adverse events in each group was considered possibly related to study treatment (allergic dermatitis and rash). AlloCSC-01 elicited low levels of donor-specific antibodies in 2 patients. No immune-related adverse events were found, and no differences between groups were observed in magnetic resonance-based efficacy parameters at 12 months. The estimated treatment effect of AlloCSC-01 on the absolute change from baseline in infarct size was -2.3% (95% confidence interval, -6.5% to 1.9%). CONCLUSIONS: AlloCSC-01 can be safely administered in ST-segment-elevation myocardial infarction patients with left ventricular dysfunction early after revascularization. Low immunogenicity and absence of immune-mediated events will facilitate adequately powered studies to demonstrate their clinical efficacy in this setting. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov . Unique identifier: NCT02439398.


Subject(s)
Myoblasts, Cardiac/transplantation , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Ventricular Dysfunction, Left/therapy , Aged , Female , Humans , Infusions, Intra-Arterial , Male , Middle Aged , Myoblasts, Cardiac/cytology , Myocardial Infarction/complications , Stem Cell Transplantation/adverse effects , Transplantation, Homologous , Ventricular Dysfunction, Left/complications
4.
Circ Res ; 122(7): 958-969, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29343525

ABSTRACT

RATIONALE: Human pluripotent stem cell-derived cardiovascular progenitor cells (hPSC-CVPCs) should be thoroughly investigated in large animal studies before testing in clinical trials. OBJECTIVE: The main of this study is to clarify whether hPSC-CVPCs can engraft for long time in the heart of primates after myocardial infarction (MI) and compare the effectiveness and safety of immunosuppression with cyclosporine alone or multiple-drug regimen (MDR) containing cyclosporine, methylprednisolone, and basiliximab in cynomolgus monkeys that had received intramyocardial injections of 1×107 EGFP (enhanced green fluorescent protein)-expressing hPSC-CVPCs after MI. A third group of animals received the immunosuppression MDR but without cell therapy after MI (MI+MDR group). METHODS AND RESULTS: Measurements of EGFP gene levels and EGFP immunofluorescence staining indicated that the hPSC-CVPC engraftment rate was greater in the MI+MDR+CVPC group than that in the MI+cyclosporine+CVPC group. However, even in the MI+MDR+CVPC group, no transplanted cells could be detected at 140 days after transplantation. Concomitantly, immunofluorescent analysis of CD3, CD4, and CD8 expression indicated that T-lymphocyte infiltration in the CVPC-transplanted hearts was less in the MDR-treated animals than in the cyclosporine-alone-treated animals. The recovery of left ventricular function on day 28 post-MI in the MI+MDR+CVPC group was better than that in the MI+MDR group. Apoptotic cardiac cells were also less common in the MI+MDR+CVPC group than in the MI+MDR group, although both immunosuppression regimens were associated with transient hepatic dysfunction. CONCLUSIONS: This is the largest study of hPSCs in nonhuman primates in cardiovascular field to date (n=32). Compared with cyclosporine alone, MDR attenuates immune rejection and improves survival of hPSC-CVPCs in primates; this is associated with less apoptosis of native cardiac cells and better recovery of left ventricular function at 28 days. However, even with MDR, transplanted hPSC-CVPCs do not engraft and do not survive at 140 days after transplantation, thereby excluding remuscularization as a mechanism for the functional effect.


Subject(s)
Human Embryonic Stem Cells/cytology , Muscle Development , Myoblasts, Cardiac/transplantation , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Animals , Cell Line , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Humans , Immunosuppression Therapy/adverse effects , Immunosuppression Therapy/methods , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Macaca fascicularis , Male , Myoblasts, Cardiac/cytology , Stem Cell Transplantation/adverse effects
5.
Circ J ; 79(7): 1422-30, 2015.
Article in English | MEDLINE | ID: mdl-26073608

ABSTRACT

Over the past 2 decades, cardiac regeneration has evolved from an exotic fringe of cardiovascular biology to the forefront of molecular, genetic, epigenetic, translational, and clinical investigations. The unmet patient need is the paucity of self-repair following infarction. Robust regeneration seen in models such as zebrafish and newborn mice has inspired the field, along with encouragement from modern methods that make even low levels of restorative growth discernible, changing the scientific and technical landscape for effective counter-measures. Approaches under study to augment cardiac repair complement each other, and encompass grafting cells of diverse kinds, restarting the cell cycle in post-mitotic ventricular myocytes, reprogramming non-myocytes, and exploiting the dormant progenitor/stem cells that lurk within the adult heart. The latter are the emphasis of the present review. Cardiac-resident stem cells (CSC) can be harvested from heart tissue, expanded, and delivered to the myocardium as a therapeutic product, whose benefits may be hoped to surpass those achieved in human trials of bone marrow. However, important questions are prompted by such cells' discovery. How do they benefit recipient hearts? Do they contribute, measurably, as an endogenous population, to self-repair? Even if "no," might CSCs be targets for activation in situ by growth factors and other developmental catalysts? And, what combination of distinguishing markers best demarcates the cells with robust clonal growth and cardiogenic potential?


Subject(s)
Adult Stem Cells/cytology , Myoblasts, Cardiac/cytology , Adult , Adult Stem Cells/physiology , Adult Stem Cells/transplantation , Animals , Biomarkers , Cell Differentiation , Cell Lineage , Cell- and Tissue-Based Therapy , Gene Expression Profiling , Heart/physiology , Humans , Mice , Models, Animal , Models, Cardiovascular , Myoblasts, Cardiac/physiology , Myoblasts, Cardiac/transplantation , Regeneration/physiology , Zebrafish
6.
Bull Exp Biol Med ; 157(1): 143-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24909728

ABSTRACT

The use of triple systemic transplantation of cardiomyoblasts raised from the culture of allogenic bone marrow mesenchymal stem cells of a healthy donor according to the new medical technology licensed by Federal Service on Surveillance in Healthcare in the therapy of a patient with late radiation cardiomyopathy and radiation exudative pericarditis developed 45 years after radiation therapy for Hodgkin lymphoma. High efficiency of systemic transplantation of mesenchymal stem cells partially differentiated towards cardiomyocytes was demonstrated. The therapeutic effect persists for more than 2 years. Possible mechanisms of the therapeutic effect of this type of stem cells and the prospects of using cell therapy in the treatment of late radiation injuries of vital organs and tissues are discussed.


Subject(s)
Cardiomyopathies/therapy , Mesenchymal Stem Cell Transplantation , Myoblasts, Cardiac/transplantation , Pericarditis/therapy , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cell Differentiation , Gamma Rays/adverse effects , Hodgkin Disease/complications , Hodgkin Disease/diagnostic imaging , Hodgkin Disease/pathology , Hodgkin Disease/radiotherapy , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Middle Aged , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/physiology , Pericarditis/diagnostic imaging , Pericarditis/etiology , Pericarditis/pathology , Transplantation, Homologous , Treatment Outcome , Ultrasonography
7.
Proc Natl Acad Sci U S A ; 107(8): 3329-34, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-19846783

ABSTRACT

Myocardial infarction resulting in irreversible loss of cardiomyocytes (CMs) remains a leading cause of heart failure. Although cell transplantation has modestly improved cardiac function, major challenges including increasing cell survival, engraftment, and functional integration with host tissue, remain. Embryonic stem cells (ESCs), which can be differentiated into cardiac progenitors (CPs) and CMs, represent a candidate cell source for cardiac cell therapy. However, it is not known what specific cell type or condition is the most appropriate for transplantation. This problem is exasperated by the lack of efficient and predictive strategies to screen the large numbers of parameters that may impact cell transplantation. We used a cardiac tissue model, engineered heart tissue (EHT), and quantitative molecular and electrophysiological analyses, to test transplantation conditions and specific cell populations for their potential to functionally integrate with the host tissue. In this study, we validated our analytical platform using contractile mouse neonatal CMs (nCMs) and noncontractile cardiac fibroblasts (cFBs), and screened for the integration potential of ESC-derived CMs and CPs (ESC-CMs and -CPs). Consistent with previous in vivo studies, cFB injection interfered with electrical signal propagation, whereas injected nCMs improved tissue function. Purified bioreactor-generated ESC-CMs exhibited a diminished capacity for electrophysiological integration; a result correlated with lower (compared with nCMs) connexin 43 expression. ESC-CPs, however, appeared able to appropriately mature and integrate into EHT, enhancing the amplitude of tissue contraction. Our results support the use of EHT as a model system to accelerate development of cardiac cell therapy strategies.


Subject(s)
Heart Failure/surgery , Myoblasts, Cardiac/physiology , Myocardial Contraction , Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Animals , Bioreactors , Cell Differentiation , Connexin 43/biosynthesis , Electrophysiological Phenomena , Fibroblasts/physiology , Heart Failure/etiology , Mice , Mice, Transgenic , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/transplantation , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Myocardial Infarction/surgery , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Rats, Sprague-Dawley
8.
Circulation ; 123(4): 364-73, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21242485

ABSTRACT

BACKGROUND: Human cardiac progenitor cells (hCPCs) may promote myocardial regeneration in adult ischemic myocardium. The regenerative capacity of hCPCs in young patients with nonischemic congenital heart defects for potential use in congenital heart defect repair warrants exploration. METHODS AND RESULTS: Human right atrial specimens were obtained during routine congenital cardiac surgery across 3 groups: neonates (age, <30 days), infants (age, 1 month to 2 years), and children (age, >2 to ≤13 years). C-kit(+) hCPCs were 3-fold higher in neonates than in children >2 years of age. hCPC proliferation was greatest during the neonatal period as evidenced by c-kit(+) Ki67(+) expression but decreased with age. hCPC differentiation capacity was also greatest in neonatal right atrium as evidenced by c-kit(+), NKX2-5(+), NOTCH1(+), and NUMB(+) expression. Despite the age-dependent decline in resident hCPCs, we isolated and expanded right atrium-derived CPCs from all patients (n=103) across all ages and diagnoses using the cardiosphere method. Intact cardiospheres contained a mix of heart-derived cell subpopulations that included cardiac progenitor cells expressing c-kit(+), Islet-1, and supporting cells. The number of c-kit(+)-expressing cells was highest in human cardiosphere-derived cells (hCDCs) grown from neonatal and infant right atrium. Furthermore, hCDCs could differentiate into diverse cardiovascular lineages by in vitro differentiation assays. Transplanted hCDCs promoted greater myocardial regeneration and functional improvement in infarcted myocardium than transplanted cardiac fibroblasts. CONCLUSIONS: Resident hCPCs are most abundant in the neonatal period and rapidly decrease over time. hCDCs can be reproducibly isolated and expanded from young human myocardial samples regardless of age or diagnosis. hCPCs are functional and have potential in congenital cardiac repair.


Subject(s)
Heart Defects, Congenital/surgery , Myoblasts, Cardiac/physiology , Myoblasts, Cardiac/transplantation , Adolescent , Age Factors , Animals , Cell Differentiation , Cell Proliferation , Child , Child, Preschool , Clinical Trials as Topic , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , Infant , Infant, Newborn , Ki-67 Antigen/metabolism , Male , Membrane Proteins/metabolism , Myoblasts, Cardiac/cytology , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Nude , Receptor, Notch1/metabolism , Transcription Factors/metabolism
9.
Biochem Biophys Res Commun ; 425(4): 859-65, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22898045

ABSTRACT

Transplantation of human cardiomyoblast-like cells (hCLCs) from human adipose tissue-derived multi-lineage progenitor cells improved left ventricular function and survival of rats with myocardial infarction. Here we examined the effect of intracoronary artery transplantation of human CLCs in a swine model of chronic heart failure. Twenty-four pigs underwent balloon-occlusion of the first diagonal branch followed by reperfusion, with a second balloon-occlusion of the left ascending coronary artery 1 week later followed by reperfusion. Four weeks after the second occlusion/reperfusion, 17 of the 18 surviving animals with severe chronic MI (ejection fraction <35% by echocardiography) were immunosuppressed then randomly assigned to receive either intracoronary artery transplantation of hCLCs hADMPCs or placebo lactic Ringer's solution with heparin. Intracoronary artery transplantation was followed by the distribution of DiI-stained hCLCs into the scarred myocardial milieu. Echocardiography at post-transplant days 4 and 8 weeks showed rescue and maintenance of cardiac function in the hCLCs transplanted group, but not in the control animals, indicating myocardial functional recovery by hCLCs intracoronary transplantation. At 8 week post-transplantation, 7 of 8 hCLCs transplanted animals were still alive compared with only 1 of the 5 control (p=0.0147). Histological studies at week 12 post-transplantation demonstrated engraftment of the pre DiI-stained hCLCs into the scarred myocardium and their expression of human specific alpha-cardiac actin. Human alpha cardiac actin-positive cells also expressed cardiac nuclear factors; nkx2.5 and GATA-4. Our results suggest that intracoronary artery transplantation of hCLCs is a potentially effective therapeutic strategy for future cardiac tissue regeneration.


Subject(s)
Adipose Tissue/cytology , Coronary Vessels/surgery , Myoblasts, Cardiac/transplantation , Myocardial Infarction/surgery , Ventricular Dysfunction, Left/surgery , Animals , Cell Lineage , Chronic Disease , Disease Models, Animal , Humans , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Stem Cell Transplantation/methods , Swine , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
10.
J Mol Cell Cardiol ; 50(2): 258-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20600097

ABSTRACT

Cardiac cell therapy has now been in clinical use since 10 years. Both autologous skeletal myoblasts and bone marrow-derived different cell subsets (mononuclear cells, hematopoietic progenitors, mesenchymal stem cells) have been investigated in different settings (acute myocardial infarction, refractory angina and chronic heart failure). Despite the huge variability in cell processing techniques, dosing, timing of delivery and route for cell transfer, some lessons can yet be drawn, primarily from randomized controlled trials and summarized as follows: Techniques used for cell preparation are reasonably well controlled although better standardization and improvement in scale-up procedures remain necessary; cell therapy is overall safe, with the caveat of ventricular arrhythmias which still require careful scrutinization; the cell type needs to be tailored to the primary clinical indication, whereas the paracrine effects of bone marrow cells may be therapeutically efficacious for limitation of remodelling or relief of angina, only cells endowed with a true cardiomyogenic differentiation potential are likely to effect regeneration of chronic scars; autologous cells are primarily limited by their variable and unpredictable functionality, thereby calling attention to banked, consistent and readily available allogeneic cell products provided the immunological issues inherent in their use can be satisfactorily addressed; regardless of the cell type, a meaningful and sustained therapeutic benefit is unlikely to occur until cell transfer and survival techniques are improved to allow greater engraftment rates; and trial end points probably need to be reassessed to focus on mechanistic issues or hard end points depending on whether new or already extensively used cells are investigated. Hopefully, these lessons may serve as a building block whose incorporation in the design of second-generation trials will help making them more clinically successful. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Subject(s)
Cell- and Tissue-Based Therapy/standards , Clinical Trials as Topic , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/transplantation , Heart Diseases/therapy , Humans , Stem Cell Transplantation/methods
11.
Crit Care Med ; 39(5): 1082-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21242796

ABSTRACT

OBJECTIVES: Arrhythmia is of concern after cardiac stem cell transplantation in repairing infarcted myocardium. However, whether transplantation improved the ventricular fibrillation threshold and whether severe malignant ventricular arrhythmia is induced in the myocardial infarction model are still unclear. We sought to investigate the electrophysiologic characteristics and ventricular fibrillation threshold in rats with myocardial infarction by treatment with allogeneic cardiac stem cells. DESIGN: Prospective, randomized, controlled study. SETTING: University-affiliated hospital. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Myocardial infarction was induced in 20 male Sprague-Dawley rats. Two weeks later, animals were randomized to receive 5 × 10(6) cardiac stem cells labeled with PKH26 in phosphate buffer solution or a phosphate buffer solution-alone injection into the infarcted anterior ventricular-free wall. MEASUREMENTS AND MAIN RESULTS: Six weeks after the cardiac stem cell or phosphate buffer solution injection, electrophysiologic characteristics and ventricular fibrillation threshold were measured at the infarct area, infarct marginal zone, and noninfarct zone. Labeled cardiac stem cells were observed in 5-µm cryostat sections from each harvested heart. The unipolar electrogram activation recovery time dispersions were shorter in the cardiac stem cell group compared with those at the phosphate buffer solution group (15.5 ± 4.4 vs. 38.6 ± 14.9 msecs, p = .000177). Malignant ventricular arrhythmias were significantly (p = .00108) less inducible in the cardiac stem cell group (one of ten) than the phosphate buffer solution group (nine of ten). The ventricular fibrillation thresholds were greatly improved in the cardiac stem cell group compared with the phosphate buffer solution group. Labeled cardiac stem cells were identified in the infarct zone and infarct marginal zone and expressed Connexin-43, von Willebrand factor, α-smooth muscle actin, and α-sarcomeric actin. CONCLUSIONS: Cardiac stem cells may modulate the electrophysiologic abnormality and improve the ventricular fibrillation threshold in rats with myocardial infarction treated with allogeneic cardiac stem cells and cardiac stem cell express markers that suggest muscle, endothelium, and vascular smooth muscle phenotypes in vivo.


Subject(s)
Electrocardiography , Myoblasts, Cardiac/transplantation , Myocardial Infarction/surgery , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Electrophysiologic Techniques, Cardiac , Immunohistochemistry , Male , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Postoperative Complications/diagnosis , Postoperative Complications/mortality , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Risk Assessment , Survival Rate , Transplantation, Homologous , Treatment Outcome , Ventricular Fibrillation/physiopathology , Ventricular Fibrillation/prevention & control , Ventricular Remodeling/physiology
12.
Mol Pharm ; 8(5): 1573-81, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21542647

ABSTRACT

Cardiac myocyte differentiation reported thus far is from iPS cells generated from mouse and human fibroblasts. However, there is no article on the generation of iPS cells from cardiac ventricular specific cell types such as H9c2 cells. Therefore, whether transduced H9c2 cells, originally isolated from embryonic cardiac ventricular tissue, will be able to generate iPS cells and have the potential to repair and regenerate infarcted myocardium remains completely elusive. We transduced H9c2 cells with four stemness factors, Oct3/4, Sox2, Klf4, and c-Myc, and successfully reprogrammed them into iPS cells. These iPS cells were able to differentiate into beating cardiac myocytes and positively stained for cardiac specific sarcomeric α-actin and myosin heavy chain proteins. Following transplantation in the infarcted myocardium, there were newly differentiated cardiac myocytes and formation of gap junction proteins at 2 weeks post-myocardial infarction (MI), suggesting newly formed cardiac myocytes were integrated into the native myocardium. Furthermore, transplanted iPS cells significantly (p < 0.05) inhibited apoptosis and fibrosis and improved cardiac function compared with MI and MI+H9c2 cell groups. Moreover, our iPS cell derived cardiac myocyte differentiation in vitro and in vivo was comparable to embryonic stem cells in the present study. In conclusion we report for the first time that we have H9c2 cell-derived iPS cells which contain the potential to differentiate into cardiac myocytes in the cell culture system and repair and regenerate infarcted myocardium with improved cardiac function in vivo.


Subject(s)
Heart/physiology , Heart/physiopathology , Induced Pluripotent Stem Cells/transplantation , Myocardial Infarction/therapy , Regeneration , Animals , Apoptosis , Cell Differentiation , Cell Line , Connexins/metabolism , Female , Fibrosis/prevention & control , Heart Function Tests , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Myoblasts, Cardiac/transplantation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Rats , Transduction, Genetic
13.
Proc Natl Acad Sci U S A ; 105(5): 1668-73, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18216245

ABSTRACT

Coronary artery disease is the most common cause of cardiac failure in the Western world, and to date there is no alternative to bypass surgery for severe coronary atherosclerosis. We report that c-kit-positive cardiac progenitor cells (CPCs) activated with insulin-like growth factor 1 and hepatocyte growth factor before their injection in proximity of the site of occlusion of the left coronary artery in rats, engrafted within the host myocardium forming temporary niches. Subsequently, CPCs divided and differentiated into endothelial cells and smooth muscle cells and, to a lesser extent, into cardiomyocytes. The acquisition of vascular lineages appeared to be mediated by the up-regulation of hypoxia-inducible factor 1alpha, which promoted the synthesis and secretion of stromal-derived factor 1 from hypoxic coronary vessels. Stromal-derived factor 1 was critical in the conversion of CPCs to the vascular fate. CPCs formed conductive and intermediate-sized coronary arteries together with resistance arterioles and capillaries. The new vessels were connected with the primary coronary circulation, and this increase in vascularization more than doubled myocardial blood flow in the infarcted myocardium. This beneficial effect, together with myocardial regeneration attenuated postinfarction dilated myopathy, reduced infarct size and improved function. In conclusion, locally delivered activated CPCs generate de novo coronary vasculature and may be implemented clinically for restoration of blood supply to the ischemic myocardium.


Subject(s)
Coronary Vessels/physiology , Myoblasts, Cardiac/physiology , Neovascularization, Physiologic , Regeneration , Stem Cells/physiology , Animals , Cell Differentiation , Chemokine CXCL12/metabolism , Coronary Vessels/cytology , Endothelial Cells/cytology , Female , Hepatocyte Growth Factor/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin-Like Growth Factor I/pharmacology , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/transplantation , Myocardial Ischemia/metabolism , Myocytes, Smooth Muscle/cytology , Proto-Oncogene Proteins c-kit/analysis , Rats , Rats, Inbred F344 , Stem Cell Transplantation , Stem Cells/chemistry , Stem Cells/drug effects
14.
Nihon Rinsho ; 69(12): 2194-202, 2011 Dec.
Article in Japanese | MEDLINE | ID: mdl-22242319

ABSTRACT

The efficacy of regenerative cell therapy as strategy of cardiac functional recovery in patients with end-stage heart failure has been recently reported, and clinical applications of autologous skeletal myoblast cell transplantation has been already started in Europe and U.S. We developed cell-sheet technology with a temperature-responsive culture dish, which enabled cell-to-cell junction to retain, and started a clinical study of cardiac regenerative therapy using autologous skeletal myoblast cell sheet implantation.


Subject(s)
Myoblasts, Cardiac/transplantation , Myocardial Ischemia/therapy , Heart Transplantation , Heart-Assist Devices , Humans
15.
J Biomed Biotechnol ; 2010: 858094, 2010.
Article in English | MEDLINE | ID: mdl-21331169

ABSTRACT

The potential of genetically modified cardiomyoblasts in treating damaged myocardium is well known. However, efficient delivery of these cells is of major concern during treatment. The limiting factors are the massive cell death that occurs soon after their intramyocardial transplantation into the beating heart. To address these problems, we generated recombinant baculoviruses (BacMam viruses) which efficiently transduced cardiomyoblast cells under optimized conditions. These genetically modified cells were then protected in a new polymeric microcapsule using poly-ethylene-glycol (PEG), alginate, and poly-L-lysine (PLL) polymers for efficient delivery. Results showed that microcapsules maintain cell viability and support cell proliferation for at least 30 days. The capsules exhibit strong immunoprotective potential and have high mechanical and osmotic stability with more than 70% intact capsules. The encased transduced cells showed a rapid transgene expression inside the capsule for at least 15 days. However, preclinical studies are needed to further explore its long-term functional benefits.


Subject(s)
Alginates/administration & dosage , Baculoviridae/genetics , Cloning, Molecular/methods , Myoblasts, Cardiac/physiology , Myoblasts, Cardiac/transplantation , Polyethylene Glycols/administration & dosage , Polylysine/analogs & derivatives , Transduction, Genetic/methods , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transplantation/methods , Cloning, Molecular/drug effects , DNA, Recombinant/genetics , Drug Stability , Materials Testing , Microscopy, Fluorescence , Microspheres , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Polylysine/administration & dosage , Spodoptera/virology , Transgenes
17.
PLoS One ; 14(12): e0227283, 2019.
Article in English | MEDLINE | ID: mdl-31891633

ABSTRACT

Upon myocardial damage, the release of cardiac proteins induces a strong antibody-mediated immune response, which can lead to adverse cardiac remodeling and eventually heart failure (HF). Stem cell therapy using mesenchymal stromal cells (MSCs) or cardiomyocyte progenitor cells (CPCs) previously showed beneficial effects on cardiac function despite low engraftment in the heart. Paracrine mediators are likely of great importance, where, for example, MSC-derived extracellular vesicles (EVs) also show immunosuppressive properties in vitro. However, the limited capacity of MSCs to differentiate into cardiac cells and the sufficient scaling of MSC-derived EVs remain a challenge to clinical translation. Therefore, we investigated the immunosuppressive actions of endogenous CPCs and CPC-derived EVs on antibody production in vitro, using both healthy controls and end-stage HF patients. Both MSCs and CPCs strongly inhibit lymphocyte proliferation and antibody production in vitro. Furthermore, CPC-derived EVs significantly lowered the levels of IgG1, IgG4, and IgM, especially when administered for longer duration. In line with previous findings, plasma cells of end-stage HF patients showed high production of IgG3, which can be inhibited by MSCs in vitro. MSCs and CPCs inhibit in vitro antibody production of both healthy and end-stage HF-derived immune cells. CPC-derived paracrine factors, such as EVs, show similar effects, but do not provide the complete immunosuppressive capacity of CPCs. The strongest immunosuppressive effects were observed using MSCs, suggesting that MSCs might be the best candidates for therapeutic targeting of B-cell responses in HF.


Subject(s)
B-Lymphocytes/immunology , Heart Failure/therapy , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mesenchymal Stem Cell Transplantation , Myoblasts, Cardiac/transplantation , B-Lymphocytes/cytology , Cell Proliferation , Cells, Cultured , Extracellular Vesicles/immunology , Heart Failure/immunology , Humans
18.
Anatol J Cardiol ; 20(6): 318-329, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30504732

ABSTRACT

OBJECTIVE: To evaluate the effects of transplantation of hypoxia-inducible factor-1α (HIF-1α) gene-modified cardiac stem cells (CSCs) on the cardiac function of heart failure rats after myocardial infarction (MI). METHODS: Twenty-four Sprague-Dawley rats were randomly divided into three groups: HIF-1α-modified CSCs group, single CSCs group, and model group. The model of heart failure after MI was established by thoracotomy-left anterior descending coronary artery ligation, followed by injection of modified CSCs, single CSCs, and PBS, respectively, 2 weeks later. The results were observed 4 weeks later. RESULTS: CSCs were infected with recombinant adenovirus. HIF-1α mRNA level of HIF-1α-enhanced green fluorescent protein (EGFP)+CSCs group significantly surpassed those of EGFP+CSCs and CSCs groups (p<0.001). Left ventricular ejection fractions (LVEFs) of HIF-1α+CSCs+MI and CSCs+MI groups significantly increased compared with the model group (p<0.001). The difference between LVEFs before and after transplantation was positively correlated with the survival rate of CSCs in infarction border zone (r=0.867, p<0.001). The apoptosis rate of HIF1α+CSCs+MI group was significantly lower than that of CSCs+MI group (p=0.008). The expression of vascular endothelial growth factor protein in HIF-1α+CSCs+MI group significantly increased, compared with that of MI group (p<0.001). The capillary density of HIF-1α+CSCs+MI group significantly exceeded that of CSCs+MI group (p<0.001). CONCLUSION: Transplantation of either HIF-1α-modified CSCs or single CSCs reduced cardiomyocyte apoptosis in rats with heart failure after MI, promoted vascular regeneration in infarct area, and improved cardiac function. Particularly, HIF-1α-modified CSCs had more significant effects.


Subject(s)
Heart Failure/physiopathology , Heart Failure/therapy , Heart/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myoblasts, Cardiac/transplantation , Myocardial Infarction/complications , Adenoviridae , Animals , Apoptosis , Cell Culture Techniques , Genetic Engineering , Heart/diagnostic imaging , Heart Failure/etiology , Male , Neovascularization, Physiologic , Rats , Rats, Sprague-Dawley , Transfection , Ultrasonography , Vascular Endothelial Growth Factor A/metabolism
19.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282820

ABSTRACT

Sudden death is the most common mode of exodus in patients with heart failure and preserved ejection fraction (HFpEF). Cardiosphere-derived cells (CDCs) reduce inflammation and fibrosis in a rat model of HFpEF, improving diastolic function and prolonging survival. We tested the hypothesis that CDCs decrease ventricular arrhythmias (VAs) and thereby possibly contribute to prolonged survival. Dahl salt-sensitive rats were fed a high-salt diet to induce HFpEF. Allogeneic rat CDCs (or phosphate-buffered saline as placebo) were injected in rats with echo-verified HFpEF. CDC-injected HFpEF rats were less prone to VA induction by programmed electrical stimulation. Action potential duration (APD) was shortened, and APD homogeneity was increased by CDC injection. Transient outward potassium current density was upregulated in cardiomyocytes from CDC rats relative to placebo, as were the underlying transcript (Kcnd3) and protein (Kv4.3) levels. Fibrosis was attenuated in CDC-treated hearts, and survival was increased. Sudden death risk also trended down, albeit nonsignificantly. CDC therapy decreased VA in HFpEF rats by shortening APD, improving APD homogeneity, and decreasing fibrosis. Unlike other stem/progenitor cells, which often exacerbate arrhythmias, CDCs reverse electrical remodeling and suppress arrhythmogenesis in HFpEF.


Subject(s)
Action Potentials , Arrhythmias, Cardiac/prevention & control , Death, Sudden, Cardiac/prevention & control , Heart Failure/mortality , Myoblasts, Cardiac/transplantation , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/mortality , Death, Sudden, Cardiac/etiology , Disease Models, Animal , Echocardiography , Electrocardiography , Heart Failure/etiology , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Humans , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Inbred Dahl , Shal Potassium Channels/metabolism , Sodium, Dietary/adverse effects , Stroke Volume , Transplantation, Homologous , Ventricular Remodeling
20.
Cell Transplant ; 16(9): 951-61, 2007.
Article in English | MEDLINE | ID: mdl-18293894

ABSTRACT

Over the past decade, the concept that the heart could undergo cardiac regeneration has rapidly evolved. Studies have indicated that numerous sites in the body harbor stem or progenitor cells, prompting clinical trials of these potential therapeutic cell-based approaches. Most notable are the series of trials utilizing either skeletal myoblasts or autologous whole bone marrow. More recently the quest has focused on specific bone marrow constituents, most notably the mesenchymal stem cell, which has several unique advantages including immunoprivilege, immunosuppression, and the ability to home to areas of tissue injury. Most recently, cells have been identified within the heart itself that are capable of self-replication and differentiation. The discovery of cardiac stem cells offers not only a potential therapeutic approach but also provides a plausible target for endogenous activation as a therapeutic strategy. Together the new insights obtained from studies of cell-based cardiac therapy have ushered in new biological paradigms and enormous potential for novel therapeutic strategies for cardiac disease.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Myocardial Infarction/therapy , Myocardium/pathology , Stem Cell Transplantation , Stem Cells/physiology , Animals , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Clinical Trials as Topic , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Humans , Mesenchymal Stem Cells/physiology , Myoblasts, Cardiac/physiology , Myoblasts, Cardiac/transplantation , Myoblasts, Skeletal/physiology , Myoblasts, Skeletal/transplantation , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL