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.
Biores Open Access ; 9(1): 258-268, 2020.
Article in English | MEDLINE | ID: mdl-33376632

ABSTRACT

The myeloid cells infiltrating the heart early after acute myocardial infarction elaborate a secretome that largely orchestrates subsequent ventricular wall repair. Regulating this innate immune response could be a means to improve infarct healing. To pilot this concept, we utilized (ß1,3-d-) glucan-encapsulated small interfering RNA (siRNA)-containing particles (GeRPs), targeting mononuclear phagocytes, delivered to mice as a one-time intramyocardial injection immediately after acute infarction. Findings demonstrated that cardiac macrophages phagocytosed GeRPs in vivo and had little systemic dissemination, thus providing a means to deliver local therapeutics. Acute infarcts were then injected in vivo with phosphate-buffered saline (PBS; vehicle) or GeRPs loaded with siRNA to Map4k4, and excised hearts were examined at 3 and 7 days by quantitative polymerase chain reaction, flow cytometry, and histology. Compared with infarcted PBS-treated hearts, hearts with intrainfarct injections of siRNA-loaded GeRPs exhibited 69-89% reductions in transcripts for Map4k4 (mitogen-activated protein kinase kinase kinase kinase 4), interleukin (IL)-1ß, and tumor necrosis factor α at 3 days. Expression of other factors relevant to matrix remodeling-monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinases, hyaluronan synthases, matricellular proteins, and profibrotic factors transforming growth factor beta (TGF-ß), and connective tissue growth factor (CTGF)-were also decreased. Most effects peaked at 3 days, but, in some instances (Map4k4, IL-1ß, TGF-ß, CTGF, versican, and periostin), suppression persisted to 7 days. Thus, direct intramyocardial GeRP injection could serve as a novel and clinically translatable platform for in vivo RNA delivery to intracardiac macrophages for local and selective immunomodulation of the infarct microenvironment.

2.
J Thorac Cardiovasc Surg ; 148(6): 3180-8.e1, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25227700

ABSTRACT

OBJECTIVE: Generating myocyte grafts that bridge across infarcts could maximize their functional impact and best utilize small numbers of stem cells. To date, however, graft survival within acute infarcts has not been feasible. To enhance intrainfarct graft viability, human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were pretreated before implantation with cobalt protoporphyrin (CoPP), a pharmacologic inducer of cytoprotective heme oxygenase-1. METHODS: After preculturing with CoPP (vs phosphate-buffered saline), hESC-CMs were injected intramyocardially into acutely infarcted rat hearts, using directed injections to span the infarct. A further group received CoPP-pretreated hESC-CMs plus 4 weekly doses of systemic CoPP to prolong exposure to cytoprotectants. Two control groups with infarcts received vehicle-only intramyocardial injections or weekly systemic CoPP without cell therapy. Postinfarct ventricular function was gauged by echocardiography and graft size quantified at 8 weeks by histomorphometry. RESULTS: CoPP-preconditioned hESC-CMs formed stable grafts deep within infarcted myocardium, while grafts without CoPP exposure survived mainly at the infarct periphery. Fractional shortening was improved at 4 and 8 weeks in all hearts receiving cell therapies (P < .01 vs vehicle-only injections). CoPP treatment of both graft hESC-CMs and recipient animals resulted in the largest grafts, highest fractional shortening, preserved wall thickness, and reduced infarct dimensions. CONCLUSIONS: Cellular therapy delivered acutely after infarction significantly improved postinfarct ventricular function at 1 and 2 months. CoPP pretreatment of cells resulted in stable hESC-CM grafts within infarcted myocardium. This design enables construction of directionally oriented, infarct-spanning bands of new cardiomyocytes that might further improve functional restoration as engrafted myocytes proliferate and mature.


Subject(s)
Embryonic Stem Cells/drug effects , Embryonic Stem Cells/transplantation , Myocardial Infarction/surgery , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/transplantation , Protoporphyrins/pharmacology , Ventricular Function/drug effects , Animals , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoprotection , Disease Models, Animal , Embryonic Stem Cells/metabolism , Graft Survival/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Humans , Molecular Targeted Therapy , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Nude , Signal Transduction/drug effects , Time Factors , Up-Regulation
3.
Stem Cells Transl Med ; 3(6): 734-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24736402

ABSTRACT

Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) can regenerate infarcted myocardium. However, when implanted into acutely infarcted hearts, few cells survive the first week postimplant. To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). When hESC-CMs were challenged with an in vitro hypoxia/reoxygenation injury, mimicking cell transplantation into an ischemic site, survival was significantly greater among cells pretreated with CoPP versus phosphate-buffered saline (PBS)-pretreated controls. Compared with PBS-pretreated cells, CoPP-pretreated hESC-CM preparations exhibited higher levels of HO-1 expression, Akt phosphorylation, and vascular endothelial growth factor production, with reduced apoptosis, and a 30% decrease in intracellular reactive oxygen species. For in vivo translation, 1 × 10(7) hESC-CMs were pretreated ex vivo with CoPP or PBS and then injected intramyocardially into rat hearts immediately following acute infarction (permanent coronary ligation). At 1 week, hESC-CM content, assessed by quantitative polymerase chain reaction for human Alu sequences, was 17-fold higher in hearts receiving CoPP- than PBS-pretreated cells. On histomorphometry, cardiomyocyte graft size was 2.6-fold larger in hearts receiving CoPP- than PBS-pretreated cells, occupying up to 12% of the ventricular area. Vascular density of host-perfused human-derived capillaries was significantly greater in grafts composed of CoPP- than PBS-pretreated cells. Taken together, these experiments demonstrate that ex vivo pretreatment of hESC-CMs with a single dose of CoPP before intramyocardial implantation more than doubled resulting graft size and improved early graft vascularization in acutely infarcted hearts. These findings open the door for delivery of these, or other, stem cells during acute interventional therapy following myocardial infarction or ischemia.


Subject(s)
Embryonic Stem Cells/drug effects , Embryonic Stem Cells/transplantation , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/transplantation , Protoporphyrins/pharmacology , Regeneration , Animals , Apoptosis/drug effects , Cell Differentiation , Cell Line , Cell Proliferation , Disease Models, Animal , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/pathology , Enzyme Induction , Female , Graft Survival , Heme Oxygenase-1/biosynthesis , Humans , Male , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Nude , Reactive Oxygen Species/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism
4.
Circulation ; 120(16): 1585-97, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19805649

ABSTRACT

BACKGROUND: The progressive shift from a young to an aged heart is characterized by alterations in the cardiac matrix. The present study investigated whether the matricellular protein thrombospondin-2 (TSP-2) may affect cardiac dimensions and function with physiological aging of the heart. METHODS AND RESULTS: TSP-2 knockout (KO) and wild-type mice were followed up to an age of 60 weeks. Survival rate, cardiac function, and morphology did not differ at a young age in TSP-2 KO compared with wild-type mice. However, >55% of the TSP-2 KO mice died between 24 and 60 weeks of age, whereas <10% of the wild-type mice died. In the absence of TSP-2, older mice displayed a severe dilated cardiomyopathy with impaired systolic function, increased cardiac dilatation, and fibrosis. Ultrastructural analysis revealed progressive myocyte stress and death, accompanied by an inflammatory response and replacement fibrosis, in aging TSP-2 KO animals, whereas capillary or coronary morphology or density was not affected. Importantly, adeno-associated virus-9 gene-mediated transfer of TSP-2 in 7-week-old TSP-2 KO mice normalized their survival and prevented dilated cardiomyopathy. In TSP-2 KO animals, age-related cardiomyopathy was accompanied by increased matrix metalloproteinase-2 and decreased tissue transglutaminase-2 activity, together with impaired collagen cross-linking. At the cardiomyocyte level, TSP-2 deficiency in vivo and its knockdown in vitro decreased the activation of the Akt survival pathway in cardiomyocytes. CONCLUSIONS: TSP-2 expression in the heart protects against age-dependent dilated cardiomyopathy.


Subject(s)
Aging , Cardiomyopathy, Dilated/etiology , Myocardium/metabolism , Thrombospondins/deficiency , Animals , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Cell Death , Enzyme Activation , Female , Fibrosis , Gene Transfer Techniques , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Knockout , Myocarditis/etiology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Proto-Oncogene Proteins c-akt/metabolism , Thrombospondins/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...