11 results on '"Beltrami, AP"'
Search Results
2. Role of TPBG (Trophoblast Glycoprotein) Antigen in Human Pericyte Migratory and Angiogenic Activity.
- Author
-
Spencer HL, Jover E, Cathery W, Avolio E, Rodriguez-Arabaolaza I, Thomas AC, Alvino VV, Sala-Newby G, Dang Z, Fagnano M, Reni C, Rowlinson J, Vono R, Spinetti G, Beltrami AP, Gargioli C, Caporali A, Angelini G, and Madeddu P
- Subjects
- Animals, Antigens, Surface genetics, Antigens, Surface metabolism, Cells, Cultured, Chemokine CXCL12 genetics, Chemokine CXCL12 metabolism, Disease Models, Animal, Extracellular Signal-Regulated MAP Kinases metabolism, Hindlimb, Humans, Ischemia genetics, Ischemia metabolism, Ischemia physiopathology, Ischemia surgery, Male, Membrane Glycoproteins genetics, Mice, Inbred C57BL, Mice, Nude, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Pericytes transplantation, Phosphorylation, Receptors, CXCR genetics, Receptors, CXCR metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Signal Transduction, Trans-Activators genetics, Trans-Activators metabolism, Cell Movement, Membrane Glycoproteins metabolism, Muscle, Skeletal blood supply, Neovascularization, Physiologic, Pericytes metabolism, Saphenous Vein metabolism
- Abstract
Objective- To determine the role of the oncofetal protein TPBG (trophoblast glycoprotein) in normal vascular function and reparative vascularization. Approach and Results- Immunohistochemistry of human veins was used to show TPBG expression in vascular smooth muscle cells and adventitial pericyte-like cells (APCs). ELISA, Western blot, immunocytochemistry, and proximity ligation assays evidenced a hypoxia-dependent upregulation of TPBG in APCs not found in vascular smooth muscle cells or endothelial cells. This involves the transcriptional modulator CITED2 (Atypical chemokine receptor 3 CBP/p300-interacting transactivator with glutamic acid (E)/aspartic acid (D)-rich tail) and downstream activation of CXCL12 (chemokine [C-X-C motif] ligand-12) signaling through the CXCR7 (C-X-C chemokine receptor type 7) receptor and ERK1/2 (extracellular signal-regulated kinases 1/2). TPBG silencing by siRNA transfection downregulated CXCL12, CXCR7, and pERK (phospho Thr202/Tyr204 ERK1/2) and reduced the APC migratory and proangiogenic capacities. TPBG forced expression induced opposite effects, which were associated with the formation of CXCR7/CXCR4 (C-X-C chemokine receptor type 4) heterodimers and could be contrasted by CXCL12 and CXCR7 neutralization. In vivo Matrigel plug assays using APCs with or without TPBG silencing evidenced TPBG is essential for angiogenesis. Finally, in immunosuppressed mice with limb ischemia, intramuscular injection of TPBG-overexpressing APCs surpassed naïve APCs in enhancing perfusion recovery and reducing the rate of toe necrosis. Conclusions- TPBG orchestrates the migratory and angiogenic activities of pericytes through the activation of the CXCL12/CXCR7/pERK axis. This novel mechanism could be a relevant target for therapeutic improvement of reparative angiogenesis.
- Published
- 2019
- Full Text
- View/download PDF
3. Combined intramyocardial delivery of human pericytes and cardiac stem cells additively improves the healing of mouse infarcted hearts through stimulation of vascular and muscular repair.
- Author
-
Avolio E, Meloni M, Spencer HL, Riu F, Katare R, Mangialardi G, Oikawa A, Rodriguez-Arabaolaza I, Dang Z, Mitchell K, Reni C, Alvino VV, Rowlinson J, Livi U, Cesselli D, Angelini G, Emanueli C, Beltrami AP, and Madeddu P
- Subjects
- Angiogenic Proteins metabolism, Animals, Cell Differentiation, Cell Proliferation, Cell Survival, Cells, Cultured, Coculture Techniques, Disease Models, Animal, Fibrosis, Hemodynamics, Humans, Mice, SCID, Myocardial Contraction, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardium metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Paracrine Communication, Pericytes metabolism, Phenotype, Recovery of Function, Saphenous Vein cytology, Time Factors, Ventricular Remodeling, Myocardial Infarction surgery, Myocardium pathology, Myocytes, Cardiac transplantation, Neovascularization, Physiologic, Pericytes transplantation, Regeneration, Stem Cell Transplantation
- Abstract
Rationale: Optimization of cell therapy for cardiac repair may require the association of different cell populations with complementary activities., Objective: Compare the reparative potential of saphenous vein-derived pericytes (SVPs) with that of cardiac stem cells (CSCs) in a model of myocardial infarction, and investigate whether combined cell transplantation provides further improvements., Methods and Results: SVPs and CSCs were isolated from vein leftovers of coronary artery bypass graft surgery and discarded atrial specimens of transplanted hearts, respectively. Single or dual cell therapy (300 000 cells of each type per heart) was tested in infarcted SCID (severe combined immunodeficiency)-Beige mice. SVPs and CSCs alone improved cardiac contractility as assessed by echocardiography at 14 days post myocardial infarction. The effect was maintained, although attenuated at 42 days. At histological level, SVPs and CSCs similarly inhibited infarct size and interstitial fibrosis, SVPs were superior in inducing angiogenesis and CSCs in promoting cardiomyocyte proliferation and recruitment of endogenous stem cells. The combination of cells additively reduced the infarct size and promoted vascular proliferation and arteriogenesis, but did not surpass single therapies with regard to contractility indexes. SVPs and CSCs secrete similar amounts of hepatocyte growth factor, vascular endothelial growth factor, fibroblast growth factor, stem cell factor, and stromal cell-derived factor-1, whereas SVPs release higher quantities of angiopoietins and microRNA-132. Coculture of the 2 cell populations results in competitive as well as enhancing paracrine activities. In particular, the release of stromal cell-derived factor-1 was synergistically augmented along with downregulation of stromal cell-derived factor-1-degrading enzyme dipeptidyl peptidase 4., Conclusions: Combinatory therapy with SVPs and CSCs may complementarily help the repair of infarcted hearts., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
4. Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway.
- Author
-
Spinetti G, Cordella D, Fortunato O, Sangalli E, Losa S, Gotti A, Carnelli F, Rosa F, Riboldi S, Sessa F, Avolio E, Beltrami AP, Emanueli C, and Madeddu P
- Subjects
- Adipose Tissue metabolism, Adipose Tissue pathology, Adult, Aged, Aged, 80 and over, Antigens, CD34 metabolism, Apoptosis, Biomarkers metabolism, Bone Marrow Cells immunology, Bone Marrow Cells pathology, Bone Marrow Examination, Case-Control Studies, Cell Lineage, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 pathology, Diabetic Angiopathies genetics, Diabetic Angiopathies pathology, Endothelial Cells metabolism, Endothelial Cells pathology, Female, Flow Cytometry, Forkhead Box Protein O3, Forkhead Transcription Factors genetics, Gene Expression Regulation, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells pathology, Humans, Immunohistochemistry, Ischemia genetics, Ischemia metabolism, Ischemia pathology, Male, MicroRNAs genetics, Microvessels immunology, Microvessels pathology, Middle Aged, Peripheral Arterial Disease genetics, Peripheral Arterial Disease metabolism, Peripheral Arterial Disease pathology, Transfection, Bone Marrow Cells metabolism, Diabetes Mellitus, Type 2 metabolism, Diabetic Angiopathies metabolism, Forkhead Transcription Factors metabolism, Hematopoietic Stem Cells metabolism, MicroRNAs metabolism, Microvessels metabolism, Signal Transduction, Stem Cell Niche
- Abstract
Rationale: The impact of diabetes mellitus on bone marrow (BM) structure is incompletely understood., Objective: Investigate the effect of type-2 diabetes mellitus (T2DM) on BM microvascular and hematopoietic cell composition in patients without vascular complications., Methods and Results: Bone samples were obtained from T2DM patients and nondiabetic controls (C) during hip replacement surgery and from T2DM patients undergoing amputation for critical limb ischemia. BM composition was assessed by histomorphometry, immunostaining, and flow cytometry. Expressional studies were performed on CD34(pos) immunosorted BM progenitor cells (PCs). Diabetes mellitus causes a reduction of hematopoietic tissue, fat deposition, and microvascular rarefaction, especially when associated with critical limb ischemia. Immunohistochemistry documented increased apoptosis and reduced abundance of CD34(pos)-PCs in diabetic groups. Likewise, flow cytometry showed scarcity of BM PCs in T2DM and T2DM+critical limb ischemia compared with C, but similar levels of mature hematopoietic cells. Activation of apoptosis in CD34(pos)-PCs was associated with upregulation and nuclear localization of the proapoptotic factor FOXO3a and induction of FOXO3a targets, p21 and p27(kip1). Moreover, microRNA-155, which regulates cell survival through inhibition of FOXO3a, was downregulated in diabetic CD34(pos)-PCs and inversely correlated with FOXO3a levels. The effect of diabetes mellitus on anatomic and molecular end points was confirmed when considering background covariates. Furthermore, exposure of healthy CD34(pos)-PCs to high glucose reproduced the transcriptional changes induced by diabetes mellitus, with this effect being reversed by forced expression of microRNA-155., Conclusions: We provide new anatomic and molecular evidence for the damaging effect of diabetes mellitus on human BM, comprising microvascular rarefaction and shortage of PCs attributable to activation of proapoptotic pathway.
- Published
- 2013
- Full Text
- View/download PDF
5. Human unrestricted somatic stem cells: how far from clinics?
- Author
-
Baccarani U and Beltrami AP
- Subjects
- Animals, Humans, Cord Blood Stem Cell Transplantation, Liver Neoplasms, Experimental surgery, Liver Regeneration, Multipotent Stem Cells transplantation
- Published
- 2012
- Full Text
- View/download PDF
6. Transplantation of human pericyte progenitor cells improves the repair of infarcted heart through activation of an angiogenic program involving micro-RNA-132.
- Author
-
Katare R, Riu F, Mitchell K, Gubernator M, Campagnolo P, Cui Y, Fortunato O, Avolio E, Cesselli D, Beltrami AP, Angelini G, Emanueli C, and Madeddu P
- Subjects
- Animals, Cells, Cultured, Humans, Male, Mice, Mice, Nude, MicroRNAs metabolism, Myocardial Infarction metabolism, Myocardial Infarction pathology, Pericytes metabolism, Rats, Mesenchymal Stem Cell Transplantation methods, MicroRNAs biosynthesis, Myocardial Infarction surgery, Neovascularization, Physiologic physiology, Pericytes transplantation, Stem Cells metabolism
- Abstract
Rationale: Pericytes are key regulators of vascular maturation, but their value for cardiac repair remains unknown., Objective: We investigated the therapeutic activity and mechanistic targets of saphenous vein-derived pericyte progenitor cells (SVPs) in a mouse myocardial infarction (MI) model., Methods and Results: SVPs have a low immunogenic profile and are resistant to hypoxia/starvation (H/S). Transplantation of SVPs into the peri-infarct zone of immunodeficient CD1/Foxn-1(nu/nu) or immunocompetent CD1 mice attenuated left ventricular dilatation and improved ejection fraction compared to vehicle. Moreover, SVPs reduced myocardial scar, cardiomyocyte apoptosis and interstitial fibrosis, improved myocardial blood flow and neovascularization, and attenuated vascular permeability. SVPs secrete vascular endothelial growth factor A, angiopoietin-1, and chemokines and induce an endogenous angiocrine response by the host, through recruitment of vascular endothelial growth factor B expressing monocytes. The association of donor- and recipient-derived stimuli activates the proangiogenic and prosurvival Akt/eNOS/Bcl-2 signaling pathway. Moreover, microRNA-132 (miR-132) was constitutively expressed and secreted by SVPs and remarkably upregulated, together with its transcriptional activator cyclic AMP response element-binding protein, on stimulation by H/S or vascular endothelial growth factor B. We next investigated if SVP-secreted miR-132 acts as a paracrine activator of cardiac healing. In vitro studies showed that SVP conditioned medium stimulates endothelial tube formation and reduces myofibroblast differentiation, through inhibition of Ras-GTPase activating protein and methyl-CpG-binding protein 2, which are validated miR-132 targets. Furthermore, miR-132 inhibition by antimiR-132 decreased SVP capacity to improve contractility, reparative angiogenesis, and interstitial fibrosis in infarcted hearts., Conclusion: SVP transplantation produces long-term improvement of cardiac function through a novel paracrine mechanism involving the secretion of miR-132 and inhibition of its target genes.
- Published
- 2011
- Full Text
- View/download PDF
7. Intravenous gene therapy with PIM-1 via a cardiotropic viral vector halts the progression of diabetic cardiomyopathy through promotion of prosurvival signaling.
- Author
-
Katare R, Caporali A, Zentilin L, Avolio E, Sala-Newby G, Oikawa A, Cesselli D, Beltrami AP, Giacca M, Emanueli C, and Madeddu P
- Subjects
- Animals, Apoptosis genetics, Cell Survival physiology, Cells, Cultured, Diabetic Cardiomyopathies pathology, Genetic Vectors administration & dosage, Glucose administration & dosage, Humans, Injections, Intravenous, Male, Mice, Plasmids administration & dosage, Rats, Rats, Wistar, Diabetic Cardiomyopathies genetics, Diabetic Cardiomyopathies prevention & control, Disease Progression, Genetic Therapy methods, Genetic Vectors genetics, Proto-Oncogene Proteins c-pim-1 administration & dosage, Proto-Oncogene Proteins c-pim-1 genetics, Signal Transduction genetics
- Abstract
Rationale: Studies in transgenic mice showed the key role of (Pim-1) (proviral integration site for Moloney murine leukemia virus-1) in the control of cardiomyocyte function and viability., Objective: We investigated whether Pim-1 represents a novel mechanistic target for the cure of diabetic cardiomyopathy, a steadily increasing cause of nonischemic heart failure., Methods and Results: In streptozotocin-induced type 1 diabetic mice, Pim-1 protein levels declined during progression of cardiomyopathy, along with upregulation of Pim-1 inhibitors, protein phosphatase 2A, and microRNA-1. Moreover, diabetic hearts showed low levels of antiapoptotic B-cell lymphoma-2 (Bcl-2) protein and increased proapoptotic caspase-3 activity. Studies on adult rat cardiomyocytes and murine cardiac progenitor cells challenged with high glucose confirmed the in vivo expressional changes. In rescue studies, anti-microRNA-1 boosted Pim-1 and Bcl-2 expression and promoted cardiomyocyte and cardiac progenitor cell survival under high glucose conditions. Similarly, transfection with Pim-1 plasmid prevented high glucose-induced cardiomyocyte and cardiac progenitor cell apoptosis. Finally, a single intravenous injection of human PIM-1 via cardiotropic serotype-9 adeno-associated virus (1 × 10(10) or 5 × 10(10) plaque-forming units per animal) at 4 weeks after diabetes induction led to sustained cardiac overexpression of Pim-1 and improved diastolic function and prevented left ventricular dilation and failure. Histological examination showed reduced cardiomyocyte apoptosis and fibrosis in association with increased c-kit(+) cells and cardiomyocyte proliferation, whereas molecular analysis confirmed activation of the prosurvival pathway and conservation of sarcoendoplasmic reticulum Ca(2+)-ATPase and α-myosin heavy chain in Pim-1-treated hearts., Conclusions: Pim-1 downregulation contributes in the pathogenesis of diabetic cardiomyopathy. Systemic delivery of human PIM-1 via cardiotropic adeno-associated virus serotype-9 represents a novel and effective approach to treat diabetic cardiomyopathy.
- Published
- 2011
- Full Text
- View/download PDF
8. Myocyte turnover in the aging human heart.
- Author
-
Kajstura J, Gurusamy N, Ogórek B, Goichberg P, Clavo-Rondon C, Hosoda T, D'Amario D, Bardelli S, Beltrami AP, Cesselli D, Bussani R, del Monte F, Quaini F, Rota M, Beltrami CA, Buchholz BA, Leri A, and Anversa P
- Subjects
- Adult, Aged, Aged, 80 and over, Apoptosis physiology, Cell Death physiology, Cells, Cultured, Female, Heart anatomy & histology, Humans, Male, Middle Aged, Sex Characteristics, Young Adult, Adult Stem Cells cytology, Adult Stem Cells physiology, Cell Differentiation physiology, Cellular Senescence physiology, Heart physiology, Myocytes, Cardiac cytology, Myocytes, Cardiac physiology
- Abstract
Rationale: The turnover of cardiomyocytes in the aging female and male heart is currently unknown, emphasizing the need to define human myocardial biology., Objective: The effects of age and gender on the magnitude of myocyte regeneration and the origin of newly formed cardiomyocytes were determined., Methods and Results: The interaction of myocyte replacement, cellular senescence, growth inhibition, and apoptosis was measured in normal female (n=32) and male (n=42) human hearts collected from patients 19 to 104 years of age who died from causes other than cardiovascular diseases. A progressive loss of telomeric DNA in human cardiac stem cells (hCSCs) occurs with aging and the newly formed cardiomyocytes inherit short telomeres and rapidly reach the senescent phenotype. Our data provide novel information on the superior ability of the female heart to sustain the multiple variables associated with the development of the senescent myopathy. At all ages, the female heart is equipped with a larger pool of functionally competent hCSCs and younger myocytes than the male myocardium. The replicative potential is higher and telomeres are longer in female hCSCs than in male hCSCs. In the female heart, myocyte turnover occurs at a rate of 10%, 14%, and 40% per year at 20, 60, and 100 years of age, respectively. Corresponding values in the male heart are 7%, 12%, and 32% per year, documenting that cardiomyogenesis involves a large and progressively increasing number of parenchymal cells with aging. From 20 to 100 years of age, the myocyte compartment is replaced 15 times in women and 11 times in men., Conclusions: The human heart is a highly dynamic organ regulated by a pool of resident hCSCs that modulate cardiac homeostasis and condition organ aging.
- Published
- 2010
- Full Text
- View/download PDF
9. Human adult vena saphena contains perivascular progenitor cells endowed with clonogenic and proangiogenic potential.
- Author
-
Campagnolo P, Cesselli D, Al Haj Zen A, Beltrami AP, Kränkel N, Katare R, Angelini G, Emanueli C, and Madeddu P
- Subjects
- Adult, Adult Stem Cells metabolism, Animals, Antigens, CD34 metabolism, Cell Separation methods, Cells, Cultured, Clone Cells cytology, Coronary Artery Bypass, Disease Models, Animal, Flow Cytometry, Graft Survival, Hindlimb blood supply, Humans, Injections, Intramuscular, Ischemia pathology, Male, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Mice, Mice, Mutant Strains, Mice, Nude, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Adult Stem Cells cytology, Hematopoietic Stem Cell Transplantation methods, Ischemia therapy, Neovascularization, Physiologic physiology, Saphenous Vein cytology
- Abstract
Background: Clinical trials in ischemic patients showed the safety and benefit of autologous bone marrow progenitor cell transplantation. Non-bone marrow progenitor cells with proangiogenic capacities have been described, yet they remain clinically unexploited owing to their scarcity, difficulty of access, and low ex vivo expansibility. We investigated the presence, antigenic profile, expansion capacity, and proangiogenic potential of progenitor cells from the saphenous vein of patients undergoing coronary artery bypass surgery., Methods and Results: CD34-positive cells, negative for the endothelial marker von Willebrand factor, were localized around adventitial vasa vasorum. After dissection of the vein from surrounding tissues and enzymatic digestion, CD34-positive/CD31-negative cells were isolated by selective culture, immunomagnetic beads, or fluorescence-assisted cell sorting. In the presence of serum, CD34-positive/CD31-negative cells gave rise to a highly proliferative population that expressed pericyte/mesenchymal antigens together with the stem cell marker Sox2 and showed clonogenic and multilineage differentiation capacities. We called this population "saphenous vein-derived progenitor cells" (SVPs). In culture, SVPs integrated into networks formed by endothelial cells and supported angiogenesis through paracrine mechanisms. Reciprocally, endothelial cell-released factors facilitated SVP migration. These interactive responses were inhibited by Tie-2 or platelet-derived growth factor-BB blockade. Intramuscular injection of SVPs in ischemic limbs of immunodeficient mice improved neovascularization and blood flow recovery. At 14 days after transplantation, proliferating SVPs were still detectable in the recipient muscles, where they established N-cadherin-mediated physical contact with the capillary endothelium., Conclusions: SVPs generated from human vein CD34-positive/CD31-negative progenitor cells might represent a new therapeutic tool for angiogenic therapy in ischemic patients.
- Published
- 2010
- Full Text
- View/download PDF
10. Multipotent progenitor cells are present in human peripheral blood.
- Author
-
Cesselli D, Beltrami AP, Rigo S, Bergamin N, D'Aurizio F, Verardo R, Piazza S, Klaric E, Fanin R, Toffoletto B, Marzinotto S, Mariuzzi L, Finato N, Pandolfi M, Leri A, Schneider C, Beltrami CA, and Anversa P
- Subjects
- Blood Cells drug effects, Blood Cells metabolism, Cell Differentiation drug effects, Cells, Cultured, Gene Expression Profiling, Granulocyte Colony-Stimulating Factor pharmacology, Humans, Kruppel-Like Factor 4, Leukapheresis, Multipotent Stem Cells drug effects, Multipotent Stem Cells metabolism, Blood Cells cytology, Multipotent Stem Cells cytology
- Abstract
To determine whether the peripheral blood in humans contains a population of multipotent progenitor cells (MPCs), products of leukapheresis were obtained from healthy donor volunteers following the administration of granulocyte colony-stimulating factor. Small clusters of adherent proliferating cells were collected, and these cells continued to divide up to 40 population doublings without reaching replicative senescence and growth arrest. MPCs were positive for the transcription factors Nanog, Oct3/4, Sox2, c-Myc, and Klf4 and expressed several antigens characteristic of mesenchymal stem cells. However, they were negative for markers of hematopoietic stem/progenitor cells and bone marrow cell lineages. MPCs had a cloning efficiency of approximately 3%, and following their expansion, retained a highly immature phenotype. Under permissive culture conditions, MPCs differentiated into neurons, glial cells, hepatocytes, cardiomyocytes, endothelial cells, and osteoblasts. Moreover, the gene expression profile of MPCs partially overlapped with that of neural and embryonic stem cells, further demonstrating their primitive, uncommitted phenotype. Following subcutaneous transplantation in nonimmunosuppressed mice, MPCs migrated to distant organs and integrated structurally and functionally within the new tissue, acquiring the identity of resident parenchymal cells. In conclusion, undifferentiated cells with properties of embryonic stem cells can be isolated and expanded from human peripheral blood after granulocyte colony-stimulating factor administration. This cell pool may constitute a unique source of autologous cells with critical clinical import.
- Published
- 2009
- Full Text
- View/download PDF
11. Oxidative stress-mediated cardiac cell death is a major determinant of ventricular dysfunction and failure in dog dilated cardiomyopathy.
- Author
-
Cesselli D, Jakoniuk I, Barlucchi L, Beltrami AP, Hintze TH, Nadal-Ginard B, Kajstura J, Leri A, and Anversa P
- Subjects
- Animals, Blotting, Western, Cardiac Pacing, Artificial, Cardiomyopathy, Dilated pathology, Caspase 3, Caspase 9, Caspases metabolism, Cytochrome c Group metabolism, Disease Models, Animal, Dogs, Enzyme Activation physiology, Hemodynamics, Immunohistochemistry, In Situ Nick-End Labeling, Myocardium metabolism, Myocardium pathology, Protein Biosynthesis, Reactive Oxygen Species metabolism, Shc Signaling Adaptor Proteins, Tumor Suppressor Protein p53 metabolism, Tyrosine metabolism, Ventricular Dysfunction pathology, Adaptor Proteins, Signal Transducing, Adaptor Proteins, Vesicular Transport, Apoptosis, Cardiomyopathy, Dilated physiopathology, Oxidative Stress, Tyrosine analogs & derivatives, Ventricular Dysfunction etiology, Ventricular Dysfunction physiopathology
- Abstract
Cell death has been questioned as a mechanism of ventricular failure. In this report, we tested the hypothesis that apoptotic death of myocytes, endothelial cells, and fibroblasts is implicated in the development of the dilated myopathy induced by ventricular pacing. Accumulation of reactive oxygen products such as nitrotyrosine, potentiation of the oxidative stress response by p66(shc) expression, formation of p53 fragments, release of cytochrome c, and caspase activation were examined to establish whether these events were coupled with apoptotic cell death in the paced dog heart. Myocyte, endothelial cell, and fibroblast apoptosis was detected before indices of severe impairment of cardiac function became apparent. Cell death increased with the duration of pacing, and myocyte death exceeded endothelial cell and fibroblast death throughout. Nitrotyrosine formation and p66(shc) levels progressively increased with pacing and were associated with cell apoptosis. Similarly, p50 (DeltaN) fragments augmented paralleling the degree of cell death in the failing heart. Moreover, cytochrome c release and activation of caspase-9 and -3 increased from 1 to 4 weeks of pacing. In conclusion, cardiac cell death precedes ventricular decompensation and correlates with the time-dependent deterioration of function in this model. Oxidative stress may be critical for activation of apoptosis in the overloaded heart.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.