Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Biomaterials ; 97: 51-61, 2016 08.
Article in English | MEDLINE | ID: mdl-27162074

ABSTRACT

A major goal of tissue engineering is the creation of pre-vascularized tissues that have a high density of organized microvessels that can be rapidly perfused following implantation. This is especially critical for highly metabolic tissues like myocardium, where a thick myocardial engineered tissue would require rapid perfusion within the first several days to survive transplantation. In the present work, tissue patches containing human microvessels that were either randomly oriented or aligned were placed acutely on rat hearts post-infarction and for each case it was determined whether rapid inosculation could occur and perfusion of the patch could be maintained for 6 days in an infarct environment. Patches containing self-assembled microvessels were formed by co-entrapment of human blood outgrowth endothelial cells and human pericytes in fibrin gel. Cell-induced gel contraction was mechanically-constrained resulting in samples with high densities of microvessels that were either randomly oriented (with 420 ± 140 lumens/mm(2)) or uniaxially aligned (with 940 ± 240 lumens/mm(2)) at the time of implantation. These patches were sutured onto the epicardial surface of the hearts of athymic rats following permanent ligation of the left anterior descending artery. In both aligned and randomly oriented microvessel patches, inosculation occurred and perfusion of the transplanted human microvessels was maintained, proving the in vivo vascularization potential of these engineered tissues. No difference was found in the number of human microvessels that were perfused in the randomly oriented (111 ± 75 perfused lumens/mm(2)) and aligned (173 ± 97 perfused lumens/mm(2)) patches. Our results demonstrate that tissue patches containing a high density of either aligned or randomly oriented human pre-formed microvessels achieve rapid perfusion in the myocardial infarct environment - a necessary first-step toward the creation of a thick, perfusable heart patch.


Subject(s)
Blood Vessels/physiology , Myocardial Infarction/therapy , Neovascularization, Physiologic , Perfusion , Tissue Engineering , Animals , Cells, Cultured , Heart Function Tests , Humans , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats, Nude
2.
Stem Cells Transl Med ; 4(11): 1324-32, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26371342

ABSTRACT

UNLABELLED: A tissue-engineered cardiac patch provides a method to deliver cardiomyoctes to the injured myocardium with high cell retention and large, controlled infarct coverage, enhancing the ability of cells to limit remodeling after infarction. The patch environment can also yield increased survival. In the present study, we sought to assess the efficacy of a cardiac patch made from human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to engraft and limit left ventricular (LV) remodeling acutely after infarction. Cardiac patches were created from hiPSC-CMs and human pericytes (PCs) entrapped in a fibrin gel and implanted acutely onto athymic rat hearts. hiPSC-CMs not only remained viable after in vivo culture, but also increased in number by as much as twofold, consistent with colocalization of human nuclear antigen, cardiac troponin T, and Ki-67 staining. CM+PC patches led to reduced infarct sizes compared with myocardial infarction-only controls at week 4, and CM+PC patch recipient hearts exhibited greater fractional shortening over all groups at both 1 and 4 weeks after transplantation. However, a decline occurred in fractional shortening for all groups over 4 weeks, and LV thinning was not mitigated. CM+PC patches became vascularized in vivo, and microvessels were more abundant in the host myocardium border zone, suggesting a paracrine mechanism for the improved cardiac function. PCs in a PC-only control patch did not survive 4 weeks in vivo. Our results indicate that cardiac patches containing hiPSC-CMs engraft onto acute infarcts, and the hiPSC-CMs survive, proliferate, and contribute to a reduction in infarct size and improvements in cardiac function. SIGNIFICANCE: In the present study, a cardiac patch was created from human induced pluripotent stem cell-derived cardiomyocytes and human pericytes entrapped in a fibrin gel, and it was transplanted onto infarcted rat myocardium. It was found that a patch that contained both cardiomyocytes and pericytes survived transplantation and resulted in improved cardiac function and a reduced infarct size compared with controls.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Myocardial Infarction/surgery , Myocardium , Myocytes, Cardiac/transplantation , Animals , Disease Models, Animal , Heterografts , Humans , Induced Pluripotent Stem Cells/pathology , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Nude
3.
Cell Stem Cell ; 15(6): 750-61, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25479750

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) hold promise for myocardial repair following injury, but preclinical studies in large animal models are required to determine optimal cell preparation and delivery strategies to maximize functional benefits and to evaluate safety. Here, we utilized a porcine model of acute myocardial infarction (MI) to investigate the functional impact of intramyocardial transplantation of hiPSC-derived cardiomyocytes, endothelial cells, and smooth muscle cells, in combination with a 3D fibrin patch loaded with insulin growth factor (IGF)-encapsulated microspheres. hiPSC-derived cardiomyocytes integrated into host myocardium and generated organized sarcomeric structures, and endothelial and smooth muscle cells contributed to host vasculature. Trilineage cell transplantation significantly improved left ventricular function, myocardial metabolism, and arteriole density, while reducing infarct size, ventricular wall stress, and apoptosis without inducing ventricular arrhythmias. These findings in a large animal MI model highlight the potential of utilizing hiPSC-derived cells for cardiac repair.


Subject(s)
Endothelial Cells/transplantation , Heart Ventricles/metabolism , Induced Pluripotent Stem Cells/physiology , Myocardial Infarction/therapy , Myocardium/metabolism , Myocytes, Cardiac/transplantation , Myocytes, Smooth Muscle/transplantation , Stem Cell Transplantation , Acute Disease , Animals , Apoptosis , Cell Differentiation , Cell Lineage , Cells, Cultured , Disease Models, Animal , Endothelial Cells/physiology , Fibrin/administration & dosage , Heart Ventricles/pathology , Humans , Insulin-Like Growth Factor I/administration & dosage , Microspheres , Myocardial Infarction/pathology , Myocytes, Cardiac/physiology , Myocytes, Smooth Muscle/physiology , Recovery of Function , Swine
4.
Tissue Eng Part A ; 20(7-8): 1325-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24295499

ABSTRACT

Cell therapies have emerged as a promising treatment for the prevention of heart failure after myocardial infarction (MI). This study evaluated the capacity of an aligned, fibrin-based, stretch-conditioned cardiac patch consisting of either the native population or a cardiomyocyte (CM)-depleted population (i.e., CM+ or CM- patches) of neonatal rat heart cells to ameliorate left ventricular (LV) remodeling in the acute-phase postinfarction in syngeneic, immunocompetent rats. Patches were exposed to 7 days of static culture and 7 days of cyclic stretching prior to implantation. Within 1 week of implantation, both patches became vascularized, and non-CMs began migrating from CM+ patches. By week 4, patches had been remodeled into collagenous tissue, and live, elongated, donor CMs were found within grafted CM+ patches. Significant improvement in cardiac contractile function was seen with the administration of the CM+ patch (ejection fraction increased from 35.1% ± 4.0% for MI only to 58.8% ± 7.3% with a CM+ patch, p<0.05) associated with a 77% reduction in infarct size (61.3% ± 7.9% for MI only, 13.9% ± 10.8% for CM+ patch, p<0.05), and the elimination of LV free-wall thinning. Decreased infarct size and reduced wall thinning also occurred with the administration of the CM- patch (infarct size 36.9% ± 10.2%, LV wall thickness: 1058.2 ± 135.4 µm for CM- patch, 661.3 ± 37.4 µm for MI only, p<0.05), but without improvements in cardiac function. Approximately 36.5% of the transplanted CMs survived at 4 weeks; however, they remained separated and electrically uncoupled from the host myocardium by a layer of CM-free tissue, which suggests that the benefits of CM+ patch transplantation resulted from paracrine mechanisms originating from CMs. Collectively, these observations suggest that the transplantation of CM-containing engineered heart tissue patches can lead to dramatic improvements in cardiac function and remodeling after acute MI.


Subject(s)
Myocardial Infarction/therapy , Myocardium/pathology , Regenerative Medicine/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Disease Models, Animal , Electrocardiography , Extracellular Matrix/metabolism , Implants, Experimental , Indoles/metabolism , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Neovascularization, Physiologic , Prosthesis Implantation , Rats , Ultrasonography , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...