Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 303
Filter
1.
Cardiovasc Res ; 120(2): 152-163, 2024 03 13.
Article in English | MEDLINE | ID: mdl-38175760

ABSTRACT

AIMS: Gene therapies to induce cardiomyocyte (CM) cell cycle re-entry have shown a potential to treat subacute ischaemic heart failure (IHF) but have not been tested in the more relevant setting of chronic IHF. Our group recently showed that polycistronic non-integrating lentivirus encoding Cdk1/CyclinB1 and Cdk4/CyclinD1 (TNNT2-4Fpolycistronic-NIL) is effective in inducing CM cell cycle re-entry and ameliorating subacute IHF models and preventing the subsequent IHF-induced congestions in the liver, kidneys, and lungs in rats and pigs. Here, we aim to test the long-term efficacy of TNNT2-4Fpolycistronic-NIL in a rat model of chronic IHF, a setting that differs pathophysiologically from subacute IHF and has greater clinical relevance. METHODS AND RESULTS: Rats were subjected to a 2-h coronary occlusion followed by reperfusion; 4 weeks later, rats were injected intramyocardially with either TNNT2-4Fpolycistronic-NIL or LacZ-NIL. Four months post-viral injection, TNNT2-4Fpolycistronic-NIL-treated rats showed a significant reduction in scar size and a significant improvement in left ventricular (LV) systolic cardiac function but not in the LV dilatation associated with chronic IHF. A mitosis reporter system developed in our lab showed significant induction of CM mitotic activity in TNNT2-4Fpolycistronic-NIL-treated rats. CONCLUSION: This study demonstrates, for the first time, that TNNT2-4Fpolycistronic-NIL gene therapy induces CM cell cycle re-entry in chronic IHF and improves LV function, and that this salubrious effect is sustained for at least 4 months. Given the high prevalence of chronic IHF, these results have significant clinical implications for developing a novel treatment for this deadly disease.


Subject(s)
Heart Failure , Myocardial Ischemia , Rats , Animals , Swine , Myocytes, Cardiac , Chronic Disease , Genetic Therapy , Cell Cycle
3.
Mol Cell Biochem ; 478(11): 2405-2407, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37776408
4.
Stem Cell Rev Rep ; 19(7): 2429-2446, 2023 10.
Article in English | MEDLINE | ID: mdl-37500831

ABSTRACT

BACKGROUND: Although cell therapy provides benefits for outcomes of heart failure, the most optimal cell type to be used clinically remains unknown. Most of the cell products used for therapy in humans require in vitro expansion to obtain a suitable number of cells for treatment; however, the clinical background of the donor and limited starting material may result in the impaired proliferative and reparative capacity of the cells expanded in vitro. Wharton's jelly mesenchymal cells (WJ MSCs) provide a multitude of advantages over adult tissue-derived cell products for therapy. These include large starting tissue material, superior proliferative capacity, and disease-free donors. Thus, WJ MSC if effective would be the most optimal cell source for clinical use. OBJECTIVES: This study evaluated the therapeutic efficacy of Wharton's jelly (WJ) and bone marrow (BM) mesenchymal stromal cells (MSCs) in chronic ischemic cardiomyopathy in rats. METHODS: Human WJ MSCs and BM MSCs were expanded in vitro, characterized, and evaluated for therapeutic efficacy in a immunodeficient rat model of ischemic cardiomyopathy. Cardiac function was evaluated with hemodynamics and echocardiography. The extent of cardiac fibrosis, hypertrophy, and inflammation was assessed with histological analysis. RESULTS: In vitro analysis revealed that WJ MSCs and BM MSCs are morphologically and immunophenotypically indistinguishable. Nevertheless, the functional analysis showed that WJ MSCs have a superior proliferative capacity, less senescent phenotype, and distinct transcriptomic profile compared to BM MSC. WJ MSCs and BM MSC injected in rat hearts chronically after MI produced a small, but not significant improvement in heart structure and function. Histological analysis showed no difference in the scar size, collagen content, cardiomyocyte cross-sectional area, and immune cell count. CONCLUSIONS: Human WJ and BM MSC have a small but not significant effect on cardiac structure and function when injected intramyocardially in immunodeficient rats chronically after MI.


Subject(s)
Mesenchymal Stem Cells , Myocardial Infarction , Myocardial Ischemia , Wharton Jelly , Adult , Rats , Humans , Animals , Bone Marrow , Myocardial Ischemia/therapy , Myocardial Infarction/metabolism
6.
Mol Cell Biochem ; 478(6): 1245-1250, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36282351

ABSTRACT

The loss of cardiomyocytes after myocardial infarction (MI) leads to heart failure. Recently, we demonstrated that transient overexpression of 4 cell cycle factors (4F), using a polycistronic non-integrating lentivirus (TNNT2-4F-NIL) resulted in significant improvement in cardiac function in a rat model of MI. Yet, it is crucial to demonstrate the reversal of the heart failure-related pathophysiological manifestations, such as renin-angiotensin-aldosterone system activation (RAAS). To assess that, Fisher 344 rats were randomized to receive TNNT2-4F-NIL or control virus seven days after coronary occlusion for 2 h followed by reperfusion. 4 months after treatment, N-terminal pro-brain natriuretic peptide, plasma renin activity, and aldosterone levels returned to the normal levels in rats treated with TNNT2-4F-NIL but not in vehicle-treated rats. Furthermore, the TNNT2-4F-NIL-treated group showed significantly less liver and kidney congestion than vehicle-treated rats. Thus, we conclude that in rat models of MI, TNNT2-4F-NIL reverses RAAS activation and subsequent systemic congestion.


Subject(s)
Heart Failure , Myocardial Infarction , Animals , Rats , Aldosterone/metabolism , Cell Cycle , Heart Failure/genetics , Heart Failure/therapy , Heart Failure/metabolism , Kidney/metabolism , Myocardial Infarction/metabolism , Renin/genetics , Renin/metabolism , Renin-Angiotensin System
7.
Mol Cell Biochem ; 478(4): 927-937, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36114991

ABSTRACT

Activated cardiac fibroblasts are involved in both reparative wound healing and maladaptive cardiac fibrosis after myocardial infarction (MI). Recent evidence suggests that PU.1 inhibition can enable reprogramming of profibrotic fibroblasts to quiescent fibroblasts, leading to attenuation of pathologic fibrosis in several fibrosis models. The role of PU.1 in acute MI has not been tested. We designed a randomized, blinded study to evaluate whether DB1976, a PU.1 inhibitor, attenuates cardiac function deterioration and fibrosis in a murine model of MI. A total of 44 Ai9 periostin-Cre transgenic mice were subjected to 60 min of coronary occlusion followed by reperfusion. At 7 days after MI, 37 mice were randomly assigned to control (vehicle) or DB1976 treatment and followed for 2 weeks. Left ventricular ejection fraction (EF), assessed by echocardiography, did not differ between the two groups before or after treatment (final EF, 33.3 ± 1.0% in control group and 31.2 ± 1.3% in DB1976 group). Subgroup analysis of female and male mice showed the same results. There were no differences in cardiac scar (trichrome stain) and fibrosis (interstitial/perivascular collagen; picrosirius stain) between groups. Results from the per-protocol dataset (including mice with pre-treatment EF < 35% only) were consistent with the full dataset. In conclusion, this randomized, blinded study demonstrates that DB1976, a PU.1 inhibitor, does not attenuate cardiac functional deterioration or cardiac fibrosis in a mouse model of MI caused by coronary occlusion/reperfusion.


Subject(s)
Coronary Occlusion , Myocardial Infarction , Mice , Male , Female , Animals , Stroke Volume , Coronary Occlusion/pathology , Disease Models, Animal , Ventricular Function, Left , Myocardial Infarction/pathology , Mice, Transgenic , Fibrosis , Myocardium/pathology , Mice, Inbred C57BL , Ventricular Remodeling
9.
Commun Biol ; 5(1): 934, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36085302

ABSTRACT

There is need for a reliable in vitro system that can accurately replicate the cardiac physiological environment for drug testing. The limited availability of human heart tissue culture systems has led to inaccurate interpretations of cardiac-related drug effects. Here, we developed a cardiac tissue culture model (CTCM) that can electro-mechanically stimulate heart slices with physiological stretches in systole and diastole during the cardiac cycle. After 12 days in culture, this approach partially improved the viability of heart slices but did not completely maintain their structural integrity. Therefore, following small molecule screening, we found that the incorporation of 100 nM tri-iodothyronine (T3) and 1 µM dexamethasone (Dex) into our culture media preserved the microscopic structure of the slices for 12 days. When combined with T3/Dex treatment, the CTCM system maintained the transcriptional profile, viability, metabolic activity, and structural integrity for 12 days at the same levels as the fresh heart tissue. Furthermore, overstretching the cardiac tissue induced cardiac hypertrophic signaling in culture, which provides a proof of concept for the ability of the CTCM to emulate cardiac stretch-induced hypertrophic conditions. In conclusion, CTCM can emulate cardiac physiology and pathophysiology in culture for an extended time, thereby enabling reliable drug screening.


Subject(s)
Biomimetics , Heart , Cardiomegaly , Culture Media , Humans , Systole
10.
iScience ; 25(8): 104656, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35847554

ABSTRACT

Successful cell therapy requires cells to resist the hostile ischemic myocardium, be retained to continue secreting cardioprotective growth factors/exosomes, and resist immunological host responses. Clinically relevant stem/progenitor cells in a rodent model of acute myocardial infarction (MI) demonstrated that neonatal cardiac mesenchymal stromal cells (nMSCs) provide the most robust cardiac functional recovery. Transplanted nMSCs significantly increased the number of tissue reparative macrophages and regulatory T-cells and decreased monocyte-derived inflammatory macrophages and neutrophils in the host myocardium. mRNA microarray and single-cell analyses combined with targeted depletion studies established CD47 in nMSCs as a key molecule responsible for cell retention in the myocardium through an antiphagocytic mechanism regulated by miR34a-5p. Gain and loss-of-function studies demonstrated that miR34a-5p also regulated the production of exosomes and cardioprotective paracrine factors in the nMSC secretome. In conclusion, miR34a-5p and CD47 play an important role in determining the composition of nMSCs' secretome and immune evasion, respectively.

11.
J Transl Med ; 20(1): 323, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864544

ABSTRACT

BACKGROUND: Despite promising results in clinical studies, the mechanism for the beneficial effects of allogenic cell-based therapies remains unclear. Macrophages are not only critical mediators of inflammation but also critical players in cardiac remodeling. We hypothesized that transplanted allogenic rat cardiac progenitor cells (rCPCs) augment T-regulatory cells which ultimately promote proliferation of M2 like macrophages by an as-yet undefined mechanism. METHODS AND RESULTS: To test this hypothesis, we used crossover rat strains for exploring the mechanism of myocardial repair by allogenic CPCs. Human CPCs (hCPCs) were isolated from adult patients undergoing coronary artery bypass grafting, and rat CPCs (rCPCs) were isolated from male Wistar-Kyoto (WKY) rat hearts. Allogenic rCPCs suppressed the proliferation of T-cells observed in mixed lymphocyte reactions in vitro. Transplanted syngeneic or allogeneic rCPCs significantly increased cardiac function in a rat myocardial infarct (MI) model, whereas xenogeneic CPCs did not. Allogeneic rCPCs stimulated immunomodulatory responses by specifically increasing T-regulatory cells and M2 polarization, while maintaining their cardiac recovery potential and safety profile. Mechanistically, we confirmed the inactivation of NF-kB in Treg cells and increased M2 macrophages in the myocardium after MI by transplanted CPCs derived GDF15 and it's uptake by CD48 receptor on immune cells. CONCLUSION: Collectively, these findings strongly support the active immunomodulatory properties and robust therapeutic potential of allogenic CPCs in post-MI cardiac dysfunction.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myocardial Infarction , Adult , Animals , Growth Differentiation Factor 15 , Humans , Male , Multipotent Stem Cells , Myocardial Infarction/therapy , Myocardium , Myocytes, Cardiac , Rats , Rats, Inbred WKY , Stem Cell Transplantation
12.
Curr Opin Cardiol ; 37(3): 193-200, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35612934

ABSTRACT

PURPOSE OF REVIEW: Clinical trials of adult cell therapy for chronic heart failure are often misrepresented in an unfairly negative light. Results are claimed to be 'negative', 'incremental', or 'modest'. This common misconception is detrimental to medical progress and needs to be dispelled. RECENT FINDINGS: Contrary to the false narrative of scientific and lay media, the outcome of recent trials of cell therapy for heart failure has been encouraging and even exciting. Specifically, with the exception of ALLSTAR, in the past 2 years several Phase II-III double-blind, randomized trials have yielded impressive results, demonstrating not just safety but also salubrious effects on cardiac function (MSC-HF) or clinical events (MSC-HF, CONCERT-HF, and DREAM-HF) for at least 1 year after a single administration of cells. Such outcomes were neither incremental nor minor, nor achievable with one dose of any other nondevice therapy for heart failure. SUMMARY: The oft-repeated assertion that cell therapy does not benefit patients with chronic heart failure is based on a misrepresentation of the literature and is contrary to the available scientific evidence. Although the mechanism of action of cell therapy is unclear, research on its use in heart failure should continue, as only rigorous, well designed, Phase III trials can definitely confirm or refute its efficacy.


Subject(s)
Heart Failure , Adult , Cell- and Tissue-Based Therapy , Chronic Disease , Double-Blind Method , Humans , Randomized Controlled Trials as Topic , Treatment Outcome
14.
Circulation ; 145(17): 1339-1355, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35061545

ABSTRACT

BACKGROUND: The regenerative capacity of the heart after myocardial infarction is limited. Our previous study showed that ectopic introduction of 4 cell cycle factors (4F; CDK1 [cyclin-dependent kinase 1], CDK4 [cyclin-dependent kinase 4], CCNB [cyclin B1], and CCND [cyclin D1]) promotes cardiomyocyte proliferation in 15% to 20% of infected cardiomyocytes in vitro and in vivo and improves cardiac function after myocardial infarction in mice. METHODS: Using temporal single-cell RNA sequencing, we aimed to identify the necessary reprogramming stages during the forced cardiomyocyte proliferation with 4F on a single cell basis. Using rat and pig models of ischemic heart failure, we aimed to start the first preclinical testing to introduce 4F gene therapy as a candidate for the treatment of ischemia-induced heart failure. RESULTS: Temporal bulk and single-cell RNA sequencing and further biochemical validations of mature human induced pluripotent stem cell-derived cardiomyocytes treated with either LacZ or 4F adenoviruses revealed full cell cycle reprogramming in 15% of the cardiomyocyte population at 48 hours after infection with 4F, which was associated mainly with sarcomere disassembly and metabolic reprogramming (n=3/time point/group). Transient overexpression of 4F, specifically in cardiomyocytes, was achieved using a polycistronic nonintegrating lentivirus (NIL) encoding 4F; each is driven by a TNNT2 (cardiac troponin T isoform 2) promoter (TNNT2-4Fpolycistronic-NIL). TNNT2-4Fpolycistronic-NIL or control virus was injected intramyocardially 1 week after myocardial infarction in rats (n=10/group) or pigs (n=6-7/group). Four weeks after injection, TNNT2-4Fpolycistronic-NIL-treated animals showed significant improvement in left ventricular ejection fraction and scar size compared with the control virus-treated animals. At 4 months after treatment, rats that received TNNT2-4Fpolycistronic-NIL still showed a sustained improvement in cardiac function and no obvious development of cardiac arrhythmias or systemic tumorigenesis (n=10/group). CONCLUSIONS: This study provides mechanistic insights into the process of forced cardiomyocyte proliferation and advances the clinical feasibility of this approach by minimizing the oncogenic potential of the cell cycle factors owing to the use of a novel transient and cardiomyocyte-specific viral construct.


Subject(s)
Heart Failure , Induced Pluripotent Stem Cells , Myocardial Infarction , Animals , Cell Cycle , Heart Failure/complications , Heart Failure/genetics , Heart Failure/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Rats , Stroke Volume , Swine , Ventricular Function, Left
16.
Cardiovasc Res ; 118(4): 951-976, 2022 03 16.
Article in English | MEDLINE | ID: mdl-33871588

ABSTRACT

This review summarizes the results of clinical trials of cell therapy in patients with heart failure (HF). In contrast to acute myocardial infarction (where results have been consistently negative for more than a decade), in the setting of HF the results of Phase I-II trials are encouraging, both in ischaemic and non-ischaemic cardiomyopathy. Several well-designed Phase II studies have met their primary endpoint and demonstrated an efficacy signal, which is remarkable considering that only one dose of cells was used. That an efficacy signal was seen 6-12 months after a single treatment provides a rationale for larger, rigorous trials. Importantly, no safety concerns have emerged. Amongst the various cell types tested, mesenchymal stromal cells derived from bone marrow (BM), umbilical cord, or adipose tissue show the greatest promise. In contrast, embryonic stem cells are not likely to become a clinical therapy. Unfractionated BM cells and cardiosphere-derived cells have been abandoned. The cell products used for HF will most likely be allogeneic. New approaches, such as repeated cell treatment and intravenous delivery, may revolutionize the field. As is the case for most new therapies, the development of cell therapies for HF has been slow, plagued by multifarious problems, and punctuated by many setbacks; at present, the utility of cell therapy in HF remains to be determined. What the field needs is rigorous, well-designed Phase III trials. The most important things to move forward are to keep an open mind, avoid preconceived notions, and let ourselves be guided by the evidence.


Subject(s)
Heart Failure , Mesenchymal Stem Cell Transplantation , Myocardial Ischemia , Cell- and Tissue-Based Therapy/adverse effects , Heart Failure/diagnosis , Heart Failure/therapy , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Myocardial Ischemia/therapy , Treatment Outcome
17.
Cardiovasc Res ; 118(11): 2428-2436, 2022 08 24.
Article in English | MEDLINE | ID: mdl-34387303

ABSTRACT

Exogenous cell-based therapy has emerged as a promising new strategy to facilitate repair of hearts damaged by acute or chronic injury. However, the field of cell-based therapy is handicapped by the lack of standardized definitions and terminology, making comparisons across studies challenging. Even the term 'stem cell therapy' is misleading because only a small percentage of cells derived from adult bone marrow, peripheral blood, or adipose tissue meets the accepted haematopoietic or developmental definition of stem cells. Furthermore, cells (stem or otherwise) are dynamic biological products, meaning that their surface-marker expression, phenotypic and functional characteristics, and the products they secrete in response to their microenvironment can change. It is also important to point out that most surface markers are seldom specific for a cell type. In this article, we discuss the lack of consistency in the descriptive terminology used in cell-based therapies and offer guidelines aimed at standardizing nomenclature and definitions to improve communication among investigators and the general public.


Subject(s)
Adipose Tissue , Cell- and Tissue-Based Therapy , Adult , Humans , Lung , Stem Cell Transplantation
18.
Mol Cell Biochem ; 477(2): 431-444, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34783963

ABSTRACT

Mounting evidence shows that cell therapy provides therapeutic benefits in experimental and clinical settings of chronic heart failure. However, direct cardiac delivery of cells via transendocardial injection is logistically complex, expensive, entails risks, and is not amenable to multiple dosing. Intravenous administration would be a more convenient and clinically applicable route for cell therapy. Thus, we determined whether intravenous infusion of three widely used cell types improves left ventricular (LV) function and structure and compared their efficacy. Rats with a 30-day-old myocardial infarction (MI) received intravenous infusion of vehicle (PBS) or 1 of 3 types of cells: bone marrow mesenchymal stromal cells (MSCs), cardiac mesenchymal cells (CMCs), and c-kit-positive cardiac cells (CPCs), at a dose of 12 × 106 cells. Rats were followed for 35 days after treatment to determine LV functional status by serial echocardiography and hemodynamic studies. Blood samples were collected for Hemavet analysis to determine inflammatory cell profile. LV ejection fraction (EF) dropped ≥ 20 points in all hearts at 30 days after MI and deteriorated further at 35-day follow-up in the vehicle-treated group. In contrast, deterioration of EF was halted in rats that received MSCs and attenuated in those that received CMCs or CPCs. None of the 3 types of cells significantly altered scar size, myocardial content of collagen or CD45-positive cells, or Hemavet profile. This study demonstrates that a single intravenous administration of 3 types of cells in rats with chronic ischemic cardiomyopathy is effective in attenuating the progressive deterioration in LV function. The extent of LV functional improvement was greatest with CPCs, intermediate with CMCs, and least with MSCs.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/therapy , Administration, Intravenous , Allografts , Animals , Male , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Rats , Rats, Inbred F344
19.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34948245

ABSTRACT

Heme oxygenase-1 (HO-1) is one of the most powerful cytoprotective proteins known. The goal of this study was to explore the effects of HO-1 in c-kit-positive cardiac cells (CPCs). LinNEG/c-kitPOS CPCs were isolated and expanded from wild-type (WT), HO-1 transgenic (TG), or HO-1 knockout (KO) mouse hearts. Compared with WT CPCs, cell proliferation was significantly increased in HO-1TG CPCs and decreased in HO-1KO CPCs. HO-1TG CPCs also exhibited a marked increase in new DNA synthesis during the S-phase of cell division, not only under normoxia (21% O2) but after severe hypoxia (1% O2 for 16 h). These properties of HO-1TG CPCs were associated with nuclear translocation (and thus activation) of Nrf2, a key transcription factor that regulates antioxidant genes, and increased protein expression of Ec-SOD, the only extracellular antioxidant enzyme. These data demonstrate that HO-1 upregulates Ec-SOD in CPCs and suggest that this occurs via activation of Nrf2, which thus is potentially involved in the crosstalk between two antioxidants, HO-1 in cytoplasm and Ec-SOD in extracellular matrix. Overexpression of HO-1 in CPCs may improve the survival and reparative ability of CPCs after transplantation and thus may have potential clinical application to increase efficacy of cell therapy.


Subject(s)
Heme Oxygenase-1/metabolism , Heme Oxygenase-1/physiology , Myocytes, Cardiac/metabolism , Animals , Antioxidants/pharmacology , Cell Proliferation , DNA Replication/drug effects , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Heart , Heme Oxygenase (Decyclizing)/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/physiology , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-kit/metabolism , S Phase , Superoxide Dismutase/metabolism
20.
J Am Coll Cardiol ; 78(21): 2092-2105, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34794691

ABSTRACT

This paper aims to provide an important update on the recent preclinical and clinical trials using cell therapy strategies and engineered heart tissues for the treatment of postinfarction left ventricular remodeling and heart failure. In addition to the authors' own works and opinions on the roadblocks of the field, they discuss novel approaches for cardiac remuscularization via the activation of proliferative mechanisms in resident cardiomyocytes or direct reprogramming of somatic cells into cardiomyocytes. This paper's main mindset is to present current and future strategies in light of their implications for the design of future patient trials with the ultimate objective of facilitating the translation of discoveries in regenerative myocardial therapies to the clinic.


Subject(s)
Heart Failure/therapy , Myocardial Infarction/therapy , Regeneration/physiology , Regenerative Medicine/methods , Translational Research, Biomedical/methods , Ventricular Remodeling/physiology , Animals , Blood Vessel Prosthesis/trends , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Heart Failure/physiopathology , Humans , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Myocytes, Cardiac/transplantation , Regenerative Medicine/trends , Review Literature as Topic , Translational Research, Biomedical/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...