Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Vet Comp Orthop Traumatol ; 24(2): 113-21, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21225086

RESUMO

INTRODUCTION: Synthetic void-fillers offer an alternative to autograft or allograft bone in the repair of segmental defects. However, the reparative process is delayed as only osteoconductive elements are present. The inclusion of pluripotential cells may resolve this limitation, and the use of allogeneic tissue provides the opportunity for an off-the-shelf remedy. The current study evaluated the utilisation of mesenchymal precursor cells (MPC) for the repair of an ovine critical-size tibial segmental defect. METHODS: Twenty-four, mature female sheep underwent surgery for the creation of a 3 cm tibial diaphyseal defect. In one group of 12 sheep the scaffold was used alone, and in the second group the scaffold was seeded with MPC. The defect was stabilised using a locking intramedullary nail and allowed to heal over a nine-month-period. Outcome assessments of healing included radiology of callus formation, computed tomography, assessment of new-bone volume, mechanical attributes, and histological evaluation of linear bone apposition rate and tissue response. RESULTS: The MPC-treated group displayed a significantly greater level of callus formation and rate of bone apposition in the defect. DISCUSSION: The incorporation of allogeneic MPC to a synthetic void filler stimulated early repair of critical-size diaphyseal segmental defects and holds potential as an off-the-shelf therapy for augmenting bone regeneration.


Assuntos
Diáfises/cirurgia , Procedimentos Ortopédicos/métodos , Carneiro Doméstico/cirurgia , Cirurgia Veterinária/métodos , Fraturas da Tíbia/cirurgia , Transplante Homólogo/veterinária , Animais , Materiais Biocompatíveis , Calo Ósseo/diagnóstico por imagem , Calo Ósseo/crescimento & desenvolvimento , Diáfises/diagnóstico por imagem , Diáfises/patologia , Feminino , Transplante de Células-Tronco Mesenquimais/métodos , Transplante de Células-Tronco Mesenquimais/veterinária , Radiografia , Fraturas da Tíbia/diagnóstico por imagem , Fraturas da Tíbia/patologia , Engenharia Tecidual/métodos , Transplante Homólogo/métodos
2.
J Cell Physiol ; 223(2): 530-40, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20162565

RESUMO

The cardiovascular therapeutic potential of bone marrow mesenchymal stromal/stem cells (MSC) is largely mediated by paracrine effects. Traditional preparation of MSC has involved plastic adherence-isolation. In contrast, prospective immunoselection aims to improve cell isolation by enriching for mesenchymal precursor cells (MPC) at higher purity. This study compared the biological characteristics and cardiovascular trophic activity of plastic adherence-isolated MSC (PA-MSC) and MPC prepared from the same human donors by immunoselection for stromal precursor antigen-1 (STRO-1). Compared to PA-MSC, STRO-1-MPC displayed greater (1) clonogenicity, (2) proliferative capacity, (3) multilineage differentiation potential, and (4) mRNA expression of mesenchymal stem cell-related transcripts. In vitro assays demonstrated that conditioned medium from STRO-1-MPC had greater paracrine activity than PA-MSC, with respect to cardiac cell proliferation and migration and endothelial cell migration and tube formation. In keeping with this, STRO-1-MPC exhibited higher gene and protein expression of CXCL12 and HGF. Inhibition of these cytokines attenuated endothelial tube formation and cardiac cell proliferation, respectively. Paracrine responses were enhanced by using supernatant from STRO-1(Bright) MPC and diminished with STRO-1(Dim) conditioned medium. Together, these findings indicate that prospective isolation gives rise to mesenchymal progeny that maintain a higher proportion of immature precursor cells compared to traditional plastic adherence-isolation. Enrichment for STRO-1 is also accompanied by increased expression of cardiovascular-relevant cytokines and enhanced trophic activity. Immunoselection thus provides a strategy for improving the cardiovascular reparative potential of mesenchymal cells.


Assuntos
Antígenos de Superfície/metabolismo , Células da Medula Óssea/metabolismo , Doenças Cardiovasculares/terapia , Diferenciação Celular/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina/fisiologia , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Biomarcadores/análise , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Adesão Celular/imunologia , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Separação Imunomagnética/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/imunologia , Células-Tronco Multipotentes/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Plásticos/química , RNA Mensageiro/metabolismo , Regeneração/fisiologia
3.
J Thorac Cardiovasc Surg ; 136(4): 1044-53, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18954648

RESUMO

OBJECTIVE: Hematopoietic progenitor cells are able to induce neovascularization of ischemic myocardium, inhibit apoptosis, and prevent heart failure. They express functional CC chemokine-binding receptor 3 (CCR3) and CXC chemokine-binding receptor 4 (CXCR4); however, the role of those receptors in migration of progenitor cells into the ischemic myocardium is unknown. METHODS: Myocardial infarction was surgically induced in athymic nude rats, and human bone marrow-derived CD34+ cells or saline was injected into the tail vein. Cell chemotaxis was studied in vitro using chemotaxis chambers with or without concomitant stimulation with eotaxin or stromal cell-derived factor-1. Cell migration into ischemic myocardium was evaluated by immunohistochemistry. CCR3 and CXCR4 antibodies or local injections of stromal cell-derived factor-1 were used to investigate the role of chemokine expression in the migration capacity of the injected cells. Morphologic analysis included evaluation of apoptosis and capillary density in the ischemic myocardium. RESULTS: Ischemic rat myocardium demonstrated induced messenger RNA expression for the CCR3-binding chemokines eotaxin, RANTES (regulated on activation, normal T expressed and secreted), and monocyte chemotactic protein-3, but not the CXCR4-binding chemokine stromal cell-derived factor-1. Migration of human angioblasts to ischemic rat myocardium was inhibited by a blocking anti-CCR3 monoclonal antibody, but not by a blocking anti-CXCR4 monoclonal antibody, which instead inhibited migration to bone marrow. Finally, intramyocardial injection of stromal cell-derived factor-1 redirected migration of human angioblasts to ischemic rat hearts, resulting in augmented neovascularization, enhanced cardiomyocyte survival, and functional cardiac recovery. CONCLUSIONS: CCR3-dependent chemokine interactions regulate endogenous migration of CD34+ progenitors from bone marrow to ischemic but not to normal myocardium. Manipulating CXCR4-dependent interactions could enhance the efficacy of cell therapy after myocardial infarction.


Assuntos
Antígenos CD34/metabolismo , Apoptose/fisiologia , Infarto do Miocárdio/patologia , Neovascularização Fisiológica/fisiologia , Receptores CCR3/metabolismo , Receptores CXCR4/metabolismo , Animais , Antígenos CD34/imunologia , Apoptose/efeitos dos fármacos , Biópsia por Agulha , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Quimiocina CCL5/metabolismo , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/fisiologia , Modelos Animais de Doenças , Células-Tronco Hematopoéticas , Humanos , Imuno-Histoquímica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Probabilidade , RNA Mensageiro/análise , Distribuição Aleatória , Ratos , Ratos Nus , Receptores CCR3/imunologia , Receptores CXCR4/imunologia , Sensibilidade e Especificidade , Técnicas de Cultura de Tecidos
4.
J Cell Physiol ; 214(2): 413-21, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17654479

RESUMO

Mesenchymal stem-like cells identified in different tissues reside in a perivascular niche. In the present study, we investigated the putative niche of adipose-derived stromal/stem cells (ASCs) using markers, associated with mesenchymal and perivascular cells, including STRO-1, CD146, and 3G5. Immunofluorescence staining of human adipose tissue sections, revealed that STRO-1 and 3G5 co-localized with CD146 to the perivascular regions of blood vessels. FACS was used to determine the capacity of the CD146, 3G5, and STRO-1 specific monoclonal antibodies to isolate clonogenic ASCs from disassociated human adipose tissue. Clonogenic fibroblastic colonies (CFU-F) were found to be enriched in those cell fractions selected with either STRO-1, CD146, or 3G5. Flow cytometric analysis revealed that cultured ASCs exhibited similar phenotypic profiles in relation to their expression of cell surface markers associated with stromal cells (CD44, CD90, CD105, CD106, CD146, CD166, STRO-1, alkaline phosphatase), endothelial cells (CD31, CD105, CD106, CD146, CD166), haematopoietic cells (CD14, CD31, CD45), and perivascular cells (3G5, STRO-1, CD146). The immunoselected ASCs populations maintained their characteristic multipotential properties as shown by their capacity to form Alizarin Red positive mineralized deposits, Oil Red O positive lipid droplets, and Alcian Blue positive proteoglycan-rich matrix in vitro. Furthermore, ASCs cultures established from either STRO-1, 3G5, or CD146 selected cell populations, were all capable of forming ectopic bone when transplanted subcutaneously into NOD/SCID mice. The findings presented here, describe a multipotential stem cell population within adult human adipose tissue, which appear to be intimately associated with perivascular cells surrounding the blood vessels.


Assuntos
Tecido Adiposo/citologia , Fenótipo , Células-Tronco Pluripotentes/citologia , Células Estromais/citologia , Adipogenia , Adulto , Animais , Anticorpos Monoclonais/metabolismo , Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Diferenciação Celular , Separação Celular/métodos , Células Cultivadas , Condrogênese , DNA Complementar/biossíntese , Feminino , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Camundongos , Camundongos SCID , Osteogênese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/transplante , Transplante Heterólogo
5.
Transpl Immunol ; 18(1): 13-21, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17584597

RESUMO

The interleukin-2 receptor alpha chain (IL-2Ra, CD25) plays a major part in shaping the dynamics of T cell populations following immune activation, due to its role in T cell proliferation and survival. Strategies to blunt the effector responses in transplantation have been developed by devising pharmaceutical agents to block the IL-2 pathways. However, such strategies could adversely affect the CD25(+)FOXP3(+)T regulatory (T reg) populations which also rely on intereukin-2 signaling for survival. The present study shows that a cohort of heart allograft recipients treated with Daclizumab (a humanized anti-CD25 antibody) display FOXP3 expression patterns consistent with functional T regulatory cell populations. High levels of FOXP3 were observed to correlate with lower incidence of and recovery from acute rejection, as well as lower levels of anti-donor HLA antibody production. Therefore, T reg populations appear fully functional in patients treated with Daclizumab, even when 5 doses were administered. By comparison, patients treated with fewer doses or no Daclizumab had a higher incidence of acute rejection, antibody production and graft failure. Therefore, our data indicates that Daclizumab treatment does not interfere with the generation of regulatory T cells and has a beneficial effect on heart allograft survival.


Assuntos
Fatores de Transcrição Forkhead/análise , Transplante de Coração/imunologia , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Linfócitos T Reguladores/imunologia , Adolescente , Adulto , Idoso , Feminino , Antígenos HLA/imunologia , Humanos , Masculino , Pessoa de Meia-Idade
6.
J Mol Cell Cardiol ; 40(4): 455-64, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16438981

RESUMO

In the adult, new blood vessel formation can occur either through angiogenesis from pre-existing mature endothelium or vasculogenesis mediated by bone marrow-derived endothelial precursors. We recently isolated endothelial progenitor cells, or angioblasts, in human adult bone marrow which have selective migratory properties for ischemic tissues, including myocardium, to where they home and induce vasculogenesis. Here we show that myocardial production of the IL-8/Gro-alpha CXC chemokine family is significantly increased after acute ischemia, and that this provides a chemoattractant gradient for bone marrow-derived endothelial progenitors, or angioblasts. This chemokine-mediated homing of bone marrow angioblasts to the ischemic heart regulates their ability to induce myocardial neovascularization, protection against cardiomyocyte apoptosis, and functional cardiac recovery. Together, our results indicate that CXC chemokines play a central role in regulating vasculogenesis in the adult, and suggest that manipulation of interactions between chemokines and their receptors on autologous human bone marrow-derived angioblasts could augment neovascularization of ischemic myocardial tissue.


Assuntos
Células da Medula Óssea/metabolismo , Movimento Celular , Quimiocinas CXC/metabolismo , Interleucina-8/metabolismo , Miocárdio/metabolismo , Neovascularização Patológica/metabolismo , Células-Tronco/metabolismo , Animais , Apoptose , Células da Medula Óssea/patologia , Quimiocina CXCL1 , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia , Ratos , Ratos Nus , Recuperação de Função Fisiológica , Transplante de Células-Tronco , Células-Tronco/patologia
7.
Am J Physiol Heart Circ Physiol ; 287(2): H525-32, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277196

RESUMO

The primary cardiac response to ischemic insult is cardiomyocyte hypertrophy, which initiates a genetic program culminating in apoptotic myocyte loss, progressive collagen replacement, and heart failure, a process termed cardiac remodeling. Although a few cardiomyocytes at the peri-infarct region can proliferate and regenerate after injury, no approaches are known to effectively induce endogenous cardiomyocytes to enter the cell cycle. We recently isolated, in human adult bone marrow, endothelial progenitor cells, or angioblasts, that migrate to ischemic myocardium, where they induce neovascularization and prevent myocardial remodeling. Here we show that increasing the number of angioblasts trafficking to the infarct zone results in dose-dependent neovascularization with development of progressively larger-sized capillaries. This results in sustained improvement in cardiac function by mechanisms involving protection against apoptosis and, strikingly, induction of proliferation/regeneration of endogenous cardiomyocytes. Our results suggest that agents that increase myocardial homing of bone marrow angioblasts could effectively induce endogenous cardiomyocytes to enter the cell cycle and improve functional cardiac recovery.


Assuntos
Células da Medula Óssea/citologia , Transplante de Células , Vasos Coronários/fisiopatologia , Endotélio Vascular/citologia , Infarto do Miocárdio/cirurgia , Neovascularização Fisiológica , Células-Tronco/citologia , Animais , Apoptose , Contagem de Células , Diferenciação Celular , Divisão Celular , Movimento Celular , Sobrevivência Celular , Fibrose , Coração/fisiopatologia , Humanos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ratos , Ratos Nus , Transplante Heterólogo
9.
Genes Immun ; 3(3): 123-35, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12070776

RESUMO

The receptor for advanced glycation end products (RAGE) and its proinflammatory S100/calgranulin ligands are enriched in joints of subjects with rheumatoid arthritis (RA) and amplify the immune/inflammatory response. In a model of inflammatory arthritis, blockade of RAGE in mice immunized and challenged with bovine type II collagen suppressed clinical and histologic evidence of arthritis, in parallel with diminished levels of TNF-alpha, IL-6, and matrix metalloproteinases (MMP) 3, 9 and 13 in affected tissues. Allelic variation within key domains of RAGE may influence these proinflammatory mechanisms, thereby predisposing individuals to heightened inflammatory responses. A polymorphism of the RAGE gene within the ligand-binding domain of the receptor has been identified, consisting of a glycine to serine change at position 82. Cells bearing the RAGE 82S allele displayed enhanced binding and cytokine/MMP generation following ligation by a prototypic S100/calgranulin compared with cells expressing the RAGE 82G allele. In human subjects, a case-control study demonstrated an increased prevalence of the 82S allele in patients with RA compared with control subjects. These data suggest that RAGE 82S upregulates the inflammatory response upon engagement of S100/calgranulins, and, thereby, may contribute to enhanced proinflammatory mechanisms in immune/inflammatory diseases.


Assuntos
Artrite Reumatoide/genética , Polimorfismo Genético , Receptores Imunológicos/genética , Alelos , Animais , Artrite Experimental/imunologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células CHO , Cricetinae , Humanos , Complexo Antígeno L1 Leucocitário/genética , Complexo Antígeno L1 Leucocitário/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo
10.
Ann Hematol ; 81 Suppl 2: S21-5, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12611063

RESUMO

In the pre-natal period, hemangioblasts derived from the human ventral aorta give rise to cellular elements involved in both hematopoiesis and vasculogenesis, resulting in formation of the primitive capillary network. Endothelial precursors with phenotypic and functional characteristics of embryonic hemangioblasts are also present in human adult bone marrow, and can be used to induce infarct bed vasculogenesis and angiogenesis after experimental myocardial infarction. The neovascularization results in decreased apoptosis of hypertrophied myocytes in the peri-infarct region, long-term salvage and survival of viable myocardium, reduction in collagen deposition, and sustained improvement in cardiac function. Autologous angioblasts may also be useful in cellular therapy strategies aiming to regenerate myocardial tissue after established heart failure. It is likely that protocols using cardiomyocyte/mesenchymal stem cells will require balanced co-administration of angioblasts to provide vascular structures for supply of oxygen and nutrients to both the chronically ischemic, endogenous myocardium and to the newly-implanted cardiomyocytes. Future studies will need to address the timing, relative concentrations, source and route of delivery of each of these cellular populations in animal models of acute and chronic myocardial ischemia.


Assuntos
Células da Medula Óssea , Vasos Coronários/citologia , Endotélio Vascular/citologia , Miocárdio , Neovascularização Fisiológica , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Divisão Celular , Vasos Coronários/fisiopatologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/terapia , Humanos , Infarto do Miocárdio , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia
11.
Circulation ; 104(12 Suppl 1): I177-83, 2001 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-11568052

RESUMO

BACKGROUND: The influence of sex on alloreactivity and graft outcome after heart transplantation was evaluated. METHODS AND RESULTS: A retrospective review of 520 consecutive recipients of a primary cardiac allograft between 1992 and 2000 at a single center was performed. The influence of sex on alloreactivity, acute rejection, transplant-related coronary artery disease, and survival was determined. Statistical methods included logistic regression analysis and Kaplan-Meier actuarial survival analysis. Female recipients had an increased prevalence before transplant of idiopathic cardiomyopathy, antinuclear antibodies, and HLA-B8, DR3 haplotypes. After transplant, female sex predicted shorter duration to a first rejection, higher cumulative rejection frequency, and earlier posttransplant production of anti-HLA antibodies. Female recipients had higher early mortality rates (<6 months) that were due to infection. Fatal infections correlated with 2-fold higher cyclosporine levels in female recipients. However, the incidence of transplant-related coronary artery disease developing beyond 1 year after transplant was lower in female than in male recipients. CONCLUSIONS: Females undergoing cardiac transplantation are more likely to manifest features of an underlying autoimmune state. This may predispose to a higher posttransplant risk of allograft rejection and requirement for increased immunosuppression. Earlier diagnosis and management of alloreactivity in female recipients before development of acute rejection and the use of more focused and less globally immunosuppressive agents during established rejections may have a significant effect on the clinical outcome of female cardiac allograft recipients.


Assuntos
Doenças Autoimunes/epidemiologia , Doença das Coronárias/epidemiologia , Rejeição de Enxerto/epidemiologia , Transplante de Coração , Autoanticorpos/sangue , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/imunologia , Causas de Morte , Estudos de Coortes , Comorbidade , Doença das Coronárias/diagnóstico , Doença das Coronárias/imunologia , Demografia , Intervalo Livre de Doença , Feminino , Rejeição de Enxerto/imunologia , Antígenos HLA/imunologia , Transplante de Coração/imunologia , Transplante de Coração/mortalidade , Humanos , Incidência , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Infecções Oportunistas/epidemiologia , Infecções Oportunistas/mortalidade , Prevalência , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Fatores Sexuais , Análise de Sobrevida , Taxa de Sobrevida
12.
Clin Exp Immunol ; 125(1): 142-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11472437

RESUMO

Haemodialysis is a widespread option for end-stage renal disease (ESRD). Long-term success of dialysis is, however, limited by a high rate of serious bacterial and viral infections. We compared T cell functions in ESRD patients undergoing haemodialysis (n = 20), or were not dialysed and received conventional medical treatment (n = 20). Healthy volunteers (n = 15) served as controls. The T cell phenotype was examined by immunofluorescence using fluorochrome-labelled monoclonal antibodies and FACS analysis. The concentration of soluble CD95/Fas and of tumour necrosis factor-alpha receptor type 1 (sTNFR1) in the sera was quantified by ELISA. Activation-induced programmed T cell death was triggered by anti-CD3/CD28 antibodies and measured by 7-AAD staining. All immunological tests were performed at least 1 month after dialysis initiation. T cell proliferation in response to phytohaemagglutinin or anti-CD3 monoclonal antibodies was moderately diminished in non-dialysed patients and markedly reduced in haemodialysis patients compared to healthy controls (P < 0.01 and P < 0.001, respectively). In a mixed lymphocyte culture the proliferative response of T cells from dialysed patients was significantly diminished (P < 0.001). T cells of both non-dialysed and dialysed patients have augmented CD95/Fas and CD45RO expression, increased sCD95/Fas and sTNFR1 release and spontaneously undergo apoptosis. Culture of T cells from haemodialysis patients with anti-CD3/CD28 antibodies increased the proportion of CD4(+) T cells committing activation-induced cell death by a mean 7.5-fold compared to T-helper cells from non-dialysed patients (P < 0.001). Renal failure and initiation of haemodialysis results in a reduced proliferative T cell response, an aberrant state of T cell activation and heightened susceptibility of CD4(+) T cells to activation-induced cell death.


Assuntos
Antígenos CD/biossíntese , Apoptose , Falência Renal Crônica/imunologia , Receptores do Fator de Necrose Tumoral/biossíntese , Diálise Renal , Linfócitos T/imunologia , Receptor fas/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Divisão Celular , Feminino , Humanos , Imunofenotipagem , Falência Renal Crônica/sangue , Falência Renal Crônica/terapia , Antígenos Comuns de Leucócito/biossíntese , Masculino , Pessoa de Meia-Idade , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Receptores Tipo I de Fatores de Necrose Tumoral , Solubilidade , Linfócitos T/citologia
13.
Nat Med ; 7(4): 430-6, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11283669

RESUMO

Left ventricular remodeling is a major cause of progressive heart failure and death after myocardial infarction. Although neoangiogenesis within the infarcted tissue is an integral component of the remodeling process, the capillary network is unable to support the greater demands of the hypertrophied myocardium, resulting in progressive loss of viable tissue, infarct extension and fibrous replacement. Here we show that bone marrow from adult humans contains endothelial precursors with phenotypic and functional characteristics of embryonic hemangioblasts, and that these can be used to directly induce new blood vessel formation in the infarct-bed (vasculogenesis) and proliferation of preexisting vasculature (angiogenesis) after experimental myocardial infarction. The neoangiogenesis resulted in decreased apoptosis of hypertrophied myocytes in the peri-infarct region, long-term salvage and survival of viable myocardium, reduction in collagen deposition and sustained improvement in cardiac function. The use of cytokine-mobilized autologous human bone-marrow-derived angioblasts for revascularization of infarcted myocardium (alone or in conjunction with currently used therapies) has the potential to significantly reduce morbidity and mortality associated with left ventricular remodeling.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Isquemia Miocárdica/terapia , Revascularização Miocárdica/métodos , Adulto , Animais , Antígenos CD34/metabolismo , Apoptose , Vasos Sanguíneos/citologia , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos/farmacologia , Coração/fisiopatologia , Mobilização de Células-Tronco Hematopoéticas , Humanos , Hipertrofia , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , Neovascularização Fisiológica , Ratos , Ratos Nus , Remodelação Ventricular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...