Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Am Heart Assoc ; 6(7)2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28679560

ABSTRACT

BACKGROUND: Cardiac mesenchymal cell (CMC) administration improves cardiac function in animal models of heart failure. Although the precise mechanisms remain unclear, transdifferentiation and paracrine signaling are suggested to underlie their cardiac reparative effects. We have shown that histone deacetylase 1 (HDAC1) inhibition enhances CMC cardiomyogenic lineage commitment. Here, we investigated the impact of HDAC1 on CMC cytokine secretion and associated paracrine-mediated activities on endothelial cell function. METHODS AND RESULTS: CMCs were transduced with shRNA constructs targeting HDAC1 (shHDAC1) or nontarget (shNT) control. Cytokine arrays were used to assess the expression of secreted proteins in conditioned medium (CM) from shHDAC1 or shNT-transduced CMCs. In vitro functional assays for cell proliferation, protection from oxidative stress, cell migration, and tube formation were performed on human endothelial cells incubated with CM from the various treatment conditions. CM from shHDAC1-transduced CMCs contained more cytokines involved in cell growth/differentiation and more efficiently promoted endothelial cell proliferation and tube formation compared with CM from shNT. After evaluating key cytokines previously implicated in cell-therapy-mediated cardiac repair, we found that basic fibroblast growth factor was significantly upregulated in shHDAC1-transduced CMCs. Furthermore, shRNA-mediated knockdown of basic fibroblast growth factor in HDAC1-depleted CMCs inhibited the effects of shHDAC1 CM in promoting endothelial proliferation and tube formation-indicating that HDAC1 depletion activates CMC proangiogenic paracrine signaling in a basic fibroblast growth factor-dependent manner. CONCLUSIONS: These results reveal a hitherto unknown role for HDAC1 in the modulation of CMC cytokine secretion and implicate the targeted inhibition of HDAC1 in CMCs as a means to enhance paracrine-mediated neovascularization in cardiac cell therapy applications.


Subject(s)
Angiogenic Proteins/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Heart , Histone Deacetylase 1/deficiency , Human Umbilical Vein Endothelial Cells/metabolism , Mesenchymal Stem Cells/enzymology , Myocytes, Cardiac/enzymology , Neovascularization, Physiologic , Paracrine Communication , Angiogenic Proteins/metabolism , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/metabolism , Cytokines/metabolism , Enzyme Repression , Fibroblast Growth Factor 2/metabolism , Heart/metabolism , Histone Deacetylase 1/genetics , Humans , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress , Signal Transduction , Time Factors , Transduction, Genetic , Transfection
2.
Stem Cells ; 34(12): 2916-2929, 2016 12.
Article in English | MEDLINE | ID: mdl-27501845

ABSTRACT

Histone deacetylase (HDAC) regulation is an essential process in myogenic differentiation. Inhibitors targeting the activity of specific HDAC family members have been shown to enhance the cardiogenic differentiation capacity of discrete progenitor cell types; a key property of donor cell populations contributing to their afforded benefits in cardiac cell therapy applications. The influence of HDAC inhibition on cardiac-derived mesenchymal stromal cell (CMC) transdifferentiation or the role of specific HDAC family members in dictating cardiovascular cell lineage specification has not been investigated. In the current study, the consequences of HDAC inhibition on patient-derived CMC proliferation, cardiogenic program activation, and cardiovascular differentiation/cell lineage specification were investigated using pharmacologic and genetic targeting approaches. Here, CMCs exposed to the pan-HDAC inhibitor sodium butyrate exhibited induction of a cardiogenic transcriptional program and heightened expression of myocyte and endothelial lineage-specific markers when coaxed to differentiate in vitro. Further, shRNA knockdown screens revealed CMCs depleted of HDAC1 to promote the induction of a cardiogenic transcriptional program characterized by enhanced expression of cardiomyogenic- and vasculogenic-specific markers, a finding which depended on and correlated with enhanced acetylation and stabilization of p53. Cardiogenic gene activation and elevated p53 expression levels observed in HDAC1-depleted CMCs were associated with improved aptitude to assume a cardiomyogenic/vasculogenic cell-like fate in vitro. These results suggest that HDAC1 depletion-induced p53 expression alters CMC cell fate decisions and identify HDAC1 as a potential exploitable target to facilitate CMC-mediated myocardial repair in ischemic cardiomyopathy. Stem Cells 2016;34:2916-2929.


Subject(s)
Epigenesis, Genetic , Histone Deacetylase 1/metabolism , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Acetylation , Biomarkers/metabolism , Butyric Acid/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Proliferation/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Neovascularization, Physiologic/drug effects , Organ Specificity/drug effects , Organ Specificity/genetics , Transcription, Genetic/drug effects
3.
PLoS One ; 10(10): e0140798, 2015.
Article in English | MEDLINE | ID: mdl-26474484

ABSTRACT

A recent phase I clinical trial (SCIPIO) has shown that autologous c-kit+ cardiac progenitor cells (CPCs) improve cardiac function and quality of life when transplanted into patients with ischemic heart disease. Although c-kit is widely used as a marker of resident CPCs, its role in the regulation of the cellular characteristics of CPCs remains unknown. We hypothesized that c-kit plays a role in the survival, growth, and migration of CPCs. To test this hypothesis, human CPCs were grown under stress conditions in the presence or absence of SCF, and the effects of SCF-mediated activation of c-kit on CPC survival/growth and migration were measured. SCF treatment led to a significant increase in cell survival and a reduction in cell death under serum depletion conditions. In addition, SCF significantly promoted CPC migration in vitro. Furthermore, the pro-survival and pro-migratory effects of SCF were augmented by c-kit overexpression and abrogated by c-kit inhibition with imatinib. Mechanistically, c-kit activation in CPCs led to activation of the PI3K and the MAPK pathways. With the use of specific inhibitors, we confirmed that the SCF/c-kit-dependent survival and chemotaxis of CPCs are dependent on both pathways. Taken together, our findings suggest that c-kit promotes the survival/growth and migration of human CPCs cultured ex vivo via the activation of PI3K and MAPK pathways. These results imply that the efficiency of CPC homing to the injury site as well as their survival after transplantation may be improved by modulating the activity of c-kit.


Subject(s)
Chemotaxis , MAP Kinase Signaling System , Myocardium/cytology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/cytology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemotaxis/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Factor/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism
4.
Basic Res Cardiol ; 110(5): 503, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26150250

ABSTRACT

It is commonly thought that the optimal method for intracoronary administration of cells is to stop coronary flow during cell infusion, in order to prolong cell/vascular wall contact, enhance adhesion, and promote extravasation of cells into the interstitial space. However, occlusion of a coronary artery with a balloon involves serious risks of vascular damage and/or dissection, particularly in non-stented segments such as those commonly found in patients with heart failure. It remains unknown whether the use of the stop-flow technique results in improved donor cell retention. Acute myocardial infarction was produced in 14 pigs. One to two months later, pigs received 10 million indium-111 oxyquinoline (oxine)-labeled c-kit(pos) human cardiac stem cells (hCSCs) via intracoronary infusion with (n = 7) or without (n = 7) balloon inflation. Pigs received cyclosporine to prevent acute graft rejection. Animals were euthanized 24 h later and hearts harvested for radioactivity measurements. With the stop-flow technique, the retention of hCSCs at 24 h was 5.41 ± 0.80 % of the injected dose (n = 7), compared with 4.87 ± 0.62 % without coronary occlusion (n = 7), (P = 0.60). When cells are delivered intracoronarily in a clinically relevant porcine model of chronic ischemic cardiomyopathy, the use of the stop-flow technique does not result in greater myocardial cell retention at 24 h compared with non-occlusive infusion. These results have practical implications for the design of cell therapy trials. Our observations suggest that the increased risk of complications secondary to coronary manipulation and occlusion is not warranted.


Subject(s)
Myocardial Ischemia/surgery , Myocytes, Cardiac/transplantation , Stem Cell Transplantation/methods , Animals , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Humans , Proto-Oncogene Proteins c-kit , Sus scrofa
5.
Circ Heart Fail ; 8(4): 757-65, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25995227

ABSTRACT

BACKGROUND: Although c-kit(pos) cardiac stem cells (CSCs) preserve left ventricular (LV) function and structure after myocardial infarction, CSC doses have been chosen arbitrarily, and the dose-effect relationship is unknown. METHODS AND RESULTS: Rats underwent a 90-minute coronary occlusion followed by 35 days of reperfusion. Vehicle or CSCs at 5 escalating doses (0.3×10(6), 0.75×10(6), 1.5×10(6), 3.0×10(6), and 6.0×10(6) cells/heart) were given intracoronarily 4 h after reperfusion. The lowest dose (0.3×10(6)) had no effect on LV function and morphology, whereas 0.75, 1.5, and 3.0×10(6) significantly improved regional and global LV function (echocardiography and hemodynamic studies). These 3 doses had similar effects on echocardiographic parameters (infarct wall thickening fraction, LV end-systolic and end-diastolic volumes, LV ejection fraction) and hemodynamic variables (LV end-diastolic pressure, LV dP/dtmax, preload adjusted maximal power, end-systolic elastance, preload recruitable stroke work) and produced similar reductions in apoptosis, scar size, infarct wall thinning, and LV expansion index and similar increases in viable myocardium in the risk region (morphometry). Infusion of 6.0×10(6) CSCs markedly increased postprocedural mortality. Green fluorescent protein and 5-bromo-2'-deoxyuridine staining indicated that persistence of donor cells and formation of new myocytes were negligible with all doses. CONCLUSIONS: Surprisingly, in this rat model of acute myocardial infarction, the dose-response relationship for intracoronary CSCs is flat. A minimal dose between 0.3 and 0.75×10(6) is necessary for efficacy; above this threshold, a 4-fold increase in cell number does not produce greater improvement in LV function or structure. Further increases in cell dose are harmful.


Subject(s)
Myocardial Infarction/surgery , Myocardium/pathology , Myocytes, Cardiac/transplantation , Regeneration , Stem Cell Transplantation , Ventricular Function, Left , Animals , Apoptosis , Biomarkers/metabolism , Capillaries/physiopathology , Cardiac Output , Cell Differentiation , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Male , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Proto-Oncogene Proteins c-kit/metabolism , Rats, Inbred F344 , Recovery of Function , Stem Cell Transplantation/adverse effects , Stem Cells/metabolism , Time Factors , Tissue Survival , Ultrasonography , Ventricular Pressure
6.
PLoS One ; 10(4): e0124227, 2015.
Article in English | MEDLINE | ID: mdl-25905721

ABSTRACT

BACKGROUND: There is mounting interest in using c-kit positive human cardiac stem cells (c-kit(pos) hCSCs) to repair infarcted myocardium in patients with ischemic cardiomyopathy. A recent phase I clinical trial (SCIPIO) has shown that intracoronary infusion of 1 million hCSCs is safe. Higher doses of CSCs may provide superior reparative ability; however, it is unknown if doses >1 million cells are safe. To address this issue, we examined the effects of 20 million hCSCs in pigs. METHODS: Right atrial appendage samples were obtained from patients undergoing cardiac surgery. The tissue was processed by an established protocol with eventual immunomagnetic sorting to obtain in vitro expanded hCSCs. A cumulative dose of 20 million cells was given intracoronarily to pigs without stop flow. Safety was assessed by measurement of serial biomarkers (cardiac: troponin I and CK-MB, renal: creatinine and BUN, and hepatic: AST, ALT, and alkaline phosphatase) and echocardiography pre- and post-infusion. hCSC retention 30 days after infusion was quantified by PCR for human genomic DNA. All personnel were blinded as to group assignment. RESULTS: Compared with vehicle-treated controls (n=5), pigs that received 20 million hCSCs (n=9) showed no significant change in cardiac function or end organ damage (assessed by organ specific biomarkers) that could be attributed to hCSCs (P>0.05 in all cases). No hCSCs could be detected in left ventricular samples 30 days after infusion. CONCLUSIONS: Intracoronary infusion of 20 million c-kit positive hCSCs in pigs (equivalent to ~40 million hCSCs in humans) does not cause acute cardiac injury, impairment of cardiac function, or liver and renal injury. These results have immediate translational value and lay the groundwork for using doses of CSCs >1 million in future clinical trials. Further studies are needed to ascertain whether administration of >1 million hCSCs is associated with greater efficacy in patients with ischemic cardiomyopathy.


Subject(s)
Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Transplantation , Animals , Female , Humans , Immunomagnetic Separation , Myocytes, Cardiac/cytology , Swine
7.
Circ Res ; 116(7): 1216-30, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25814683

ABSTRACT

Although numerous preclinical investigations have consistently demonstrated salubrious effects of c-kit(pos) cardiac cells administered after myocardial infarction, the mechanism of action remains highly controversial. We and others have found little or no evidence that these cells differentiate into mature functional cardiomyocytes, suggesting paracrine effects. In this review, we propose a new paradigm predicated on a comprehensive analysis of the literature, including studies of cardiac development; we have (facetiously) dubbed this conceptual construct "string theory" of c-kit(pos) cardiac cells because it reconciles multifarious and sometimes apparently discrepant results. There is strong evidence that, during development, the c-kit receptor is expressed in different pools of cardiac progenitors (some capable of robust cardiomyogenesis and others with little or no contribution to myocytes). Accordingly, c-kit positivity, in itself, does not define the embryonic origins, lineage capabilities, or differentiation capacities of specific cardiac progenitors. C-kit(pos) cells derived from the first heart field exhibit cardiomyogenic potential during development, but these cells are likely depleted shortly before or after birth. The residual c-kit(pos) cells found in the adult heart are probably of proepicardial origin, possess a mesenchymal phenotype (resembling bone marrow mesenchymal stem/stromal cells), and are capable of contributing significantly only to nonmyocytic lineages (fibroblasts, smooth muscle cells, and endothelial cells). If these 2 populations (first heart field and proepicardium) express different levels of c-kit, the cardiomyogenic potential of first heart field progenitors might be reconciled with recent results of c-kit(pos) cell lineage tracing studies. The concept that c-kit expression in the adult heart identifies epicardium-derived, noncardiomyogenic precursors with a mesenchymal phenotype helps to explain the beneficial effects of c-kit(pos) cell administration to ischemically damaged hearts despite the observed paucity of cardiomyogenic differentiation of these cells.


Subject(s)
Cell Lineage , Models, Cardiovascular , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Proto-Oncogene Proteins c-kit , Adventitia/cytology , Blood Vessels/cytology , Blood Vessels/embryology , Cell Differentiation , Clinical Trials, Phase I as Topic , Cytokines/physiology , Endocardium/cytology , Endocardium/embryology , Epithelial-Mesenchymal Transition , Graft Survival , Heart/embryology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Muscle, Smooth/cytology , Myocytes, Cardiac/chemistry , Paracrine Communication , Pericardium/cytology , Pericardium/embryology , Stem Cells/chemistry , Stem Cells/classification , Stem Cells/cytology , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...