Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 312(4): H711-H720, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28188215

RESUMO

Endothelial cystathionine γ-lyase (CSEγ) contributes to cardiovascular homeostasis, mainly through production of H2S. However, the molecular mechanisms that control CSEγ gene expression in the endothelium during cardiovascular diseases are unclear. The aim of the current study is to determine the role of specific histone deacetylases (HDACs) in the regulation of endothelial CSEγ. Reduced CSEγ mRNA expression and protein abundance were observed in human aortic endothelial cells (HAEC) exposed to oxidized LDL (OxLDL) and in aortas from atherogenic apolipoprotein E knockout (ApoE-/-) mice fed a high-fat diet compared with controls. Intact murine aortic rings exposed to OxLDL (50 µg/ml) for 24 h exhibited impaired endothelium-dependent vasorelaxation that was blocked by CSEγ overexpression or the H2S donor NaHS. CSEγ expression was upregulated by pan-HDAC inhibitors and by class II-specific HDAC inhibitors, but not by other class-specific inhibitors. The HDAC6 selective inhibitor tubacin and HDAC6-specific siRNA increased CSEγ expression and blocked OxLDL-mediated reductions in endothelial CSEγ expression and CSEγ promoter activity, indicating that HDAC6 is a specific regulator of CSEγ expression. Consistent with this finding, HDAC6 mRNA, protein expression, and activity were upregulated in OxLDL-exposed HAEC, but not in human aortic smooth muscle cells. HDAC6 protein levels in aortas from high-fat diet-fed ApoE-/- mice were comparable to those in controls, whereas HDAC6 activity was robustly upregulated. Together, our findings indicate that HDAC6 is upregulated by atherogenic stimuli via posttranslational modifications and is a critical regulator of CSEγ expression in vascular endothelium. Inhibition of HDAC6 activity may improve endothelial function and prevent or reverse the development of atherosclerosis.NEW & NOTEWORTHY Oxidative injury to endothelial cells by oxidized LDL reduced cystathionine γ-lyase (CSEγ) expression and H2S production, leading to endothelial dysfunction, which was prevented by histone deacetylase 6 (HDAC6) inhibition. Our data suggest HDAC6 as a novel therapeutic target to prevent the development of atherosclerosis.


Assuntos
Cistationina gama-Liase/metabolismo , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/genética , Animais , Apolipoproteínas E/genética , Cistationina gama-Liase/biossíntese , Cistationina gama-Liase/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Desacetilase 6 de Histona , Humanos , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Lipoproteínas LDL/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Vasodilatação/efeitos dos fármacos , Vasodilatação/genética
2.
J Vis Exp ; (94)2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25489842

RESUMO

Polycystic kidney disease (PKD) is one of the most common causes of end-stage kidney disease, a devastating disease for which there is no cure. The molecular mechanisms leading to cyst formation in PKD remain somewhat unclear, but many genes are thought to be involved. Wnt5a is a non-canonical glycoprotein that regulates a wide range of developmental processes. Wnt5a works through the planar cell polarity (PCP) pathway that regulates oriented cell division during renal tubular cell elongation. Defects of the PCP pathway have been found to cause kidney cyst formation. Our paper describes a method for developing a zebrafish cystic kidney disease model by knockdown of the wnt5a gene with wnt5a antisense morpholino (MO) oligonucleotides. Tg(wt1b:GFP) transgenic zebrafish were used to visualize kidney structure and kidney cysts following wnt5a knockdown. Two distinct antisense MOs (AUG - and splice-site) were used and both resulted in curly tail down phenotype and cyst formation after wnt5a knockdown. Injection of mouse Wnt5a mRNA, resistant to the MOs due to a difference in primary base pair structure, rescued the abnormal phenotype, demonstrating that the phenotype was not due to "off-target" effects of the morpholino. This work supports the validity of using a zebrafish model to study wnt5a function in the kidney.


Assuntos
Modelos Animais de Doenças , Doenças Renais Policísticas/genética , Proteínas Wnt/genética , Animais , Animais Geneticamente Modificados , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/genética , Oligonucleotídeos Antissenso/genética , Doenças Renais Policísticas/metabolismo , Proteínas Wnt/deficiência , Proteínas Wnt/metabolismo , Proteína Wnt-5a , Peixe-Zebra
3.
J Hypertens ; 31(10): 1939-43, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23881298

RESUMO

Endothelial dysfunction plays a key role in the development and progression of cardiovascular disease. In patients with hypertension, endothelial dysfunction is characterized by a decrease of vasodilator factors release. Recent evidence highlights the involvement of regulatory T cell in the cardiovascular physiology and pathology. An increasing body of data suggest that an imbalance in the immune system triggers inflammation and compromises the cardiovascular homeostasis. In this mini-review, we will highlight the role of immune regulatory T cells in hypertension-induced vascular dysfunction.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/metabolismo , Hipertensão/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfócitos T Reguladores/imunologia , Doenças Vasculares/imunologia , Animais , Aterosclerose/complicações , Aterosclerose/imunologia , Pressão Sanguínea , Endotélio Vascular/patologia , Homeostase , Humanos , Hipertensão/complicações , Tolerância Imunológica , Inflamação , Camundongos , Microcirculação
4.
Circulation ; 121(9): 1104-12, 2010 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-20176991

RESUMO

BACKGROUND: Little is known about the role of endothelial progenitor cells (EPCs) in atherosclerosis. Accordingly, we performed a series of assessments with hypercholesterolemic (apolipoprotein E-null [ApoE(-/-)]) and wild-type (WT) mice to evaluate how cholesterol influences reendothelialization, atherosclerosis, and EPC function after arterial injury. METHODS AND RESULTS: Unexpectedly, reendothelialization (assessed by resistance to Evans blue staining) and circulating EPC counts (EPC culture assay) were greater in ApoE(-/-) mice than in WT mice, and transplantation of ApoE(-/-) bone marrow in WT mice accelerated endothelial recovery and increased recruitment of bone marrow-derived EPCs to the neoendothelium. Cholesterol concentration-dependently promoted the proliferation (MTS assay) of both ApoE(-/-) and WT EPCs, and the concentration dependence of EPC adhesion (to vitronectin-, collagen type I-, fibronectin-, and laminin-coated plates), migration (modified Boyden chamber assay), and antiapoptotic (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling stain) activity was biphasic. Cholesterol enhanced the messenger RNA expression (quantitative, real-time reverse-transcription polymerase chain reaction) of vascular endothelial growth factor and inhibited Notch1 messenger RNA expression in both ApoE(-/-) and WT EPCs, whereas endothelial nitric oxide synthase messenger RNA expression increased in ApoE(-/-) EPCs and declined in WT EPCs after cholesterol exposure. EPC activity was greater in Notch1(+/-) EPCs than in WT EPCs, and transplantation of Notch1(+/-) bone marrow accelerated endothelial recovery after arterial injury in WT mice. CONCLUSIONS: The results presented here provide novel insights into the role of EPCs during atherosclerosis and suggest that cholesterol and Notch1 may be involved in the regulation of EPC activity.


Assuntos
Lesões das Artérias Carótidas/patologia , Células Endoteliais/patologia , Hipercolesterolemia/patologia , Células-Tronco Mesenquimais/patologia , Receptor Notch1/fisiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Transplante de Medula Óssea , Lesões das Artérias Carótidas/complicações , Movimento Celular , Colesterol/sangue , Regulação da Expressão Gênica , Genótipo , Hipercolesterolemia/complicações , Hipercolesterolemia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/biossíntese , Óxido Nítrico Sintase Tipo III/genética , RNA Mensageiro/biossíntese , Quimera por Radiação , Receptor Notch1/biossíntese , Receptor Notch1/deficiência , Receptor Notch1/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
5.
Circulation ; 119(5): 699-708, 2009 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19171856

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) are known to promote neovascularization in ischemic diseases. Recent evidence suggested that diabetic neuropathy is causally related to impaired angiogenesis and deficient growth factors. Accordingly, we investigated whether diabetic neuropathy could be reversed by local transplantation of EPCs. METHODS AND RESULTS: We found that motor and sensory nerve conduction velocities, blood flow, and capillary density were reduced in sciatic nerves of streptozotocin-induced diabetic mice but recovered to normal levels after hind-limb injection of bone marrow-derived EPCs. Injected EPCs were preferentially and durably engrafted in the sciatic nerves. A portion of engrafted EPCs were uniquely localized in close proximity to vasa nervorum, and a smaller portion of these EPCs were colocalized with endothelial cells. Multiple angiogenic and neurotrophic factors were significantly increased in the EPC-injected nerves. These dual angiogenic and neurotrophic effects of EPCs were confirmed by higher proliferation of Schwann cells and endothelial cells cultured in EPC-conditioned media. CONCLUSIONS: We demonstrate for the first time that bone marrow-derived EPCs could reverse various manifestations of diabetic neuropathy. These therapeutic effects were mediated by direct augmentation of neovascularization in peripheral nerves through long-term and preferential engraftment of EPCs in nerves and particularly vasa nervorum and their paracrine effects. These findings suggest that EPC transplantation could represent an innovative therapeutic option for treating diabetic neuropathy.


Assuntos
Diabetes Mellitus Experimental/complicações , Neuropatias Diabéticas/terapia , Células Endoteliais/citologia , Transplante de Células-Tronco Hematopoéticas , Nervo Isquiático/irrigação sanguínea , Animais , Apoptose , Células da Medula Óssea/citologia , Divisão Celular , Células Cultivadas , Neuropatias Diabéticas/fisiopatologia , Células-Tronco Hematopoéticas/citologia , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Condução Nervosa , Células de Schwann/citologia , Nervo Isquiático/fisiologia
6.
J Exp Med ; 204(13): 3257-69, 2007 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-18070934

RESUMO

Noncellular differentiation effects have emerged as important mechanisms mediating therapeutic effects of stem or progenitor cell transplantation. Here, we investigated the expression patterns and sources of humoral factors and their regional and systemic biological effects after bone marrow (BM)-derived endothelial progenitor cell (EPC) transplantation into ischemic myocardium. Although most of the transplanted EPCs disappeared within a week, up-regulation of multiple humoral factors was sustained for longer than two weeks, which correlated well with the recovery of cardiac function. To determine the source of the humoral factors, we injected human EPCs into immunodeficient mice. Whereas the expression of human EPC (donor)-derived cytokines rapidly decreased to a nondetectable level within a week, up-regulation of mouse (recipient)-derived cytokines, including factors that could mobilize BM cells, was sustained. Histologically, we observed higher capillary density, a higher proliferation of myocardial cells, a lower cardiomyocyte apoptosis, and reduced infarct size. Furthermore, after EPC transplantation, BM-derived stem or progenitor cells were increased in the peripheral circulation and incorporated into the site of neovascularization and myocardial repair. These data indicate that myocardial EPC transplantation induces humoral effects, which are sustained by host tissues and play a crucial role in repairing myocardial injury.


Assuntos
Formação de Anticorpos , Transplante de Células/métodos , Células Endoteliais/citologia , Coração/fisiologia , Isquemia Miocárdica/patologia , Células-Tronco/citologia , Animais , Apoptose , Proliferação de Células , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Modelos Biológicos , Infarto do Miocárdio/patologia , Células-Tronco/metabolismo
7.
Circulation ; 114(21): 2261-70, 2006 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-17088460

RESUMO

BACKGROUND: Estradiol (E2) modulates the kinetics of circulating endothelial progenitor cells (EPCs) and favorably affects neovascularization after ischemic injury. However, the roles of estrogen receptors alpha (ER alpha) and beta (ER beta) in EPC biology are largely unknown. METHODS AND RESULTS: In response to E2, migration, tube formation, adhesion, and estrogen-responsive element-dependent gene transcription activities were severely impaired in EPCs obtained from ER alpha-knockout mice (ER alphaKO) and moderately impaired in ER betaKO EPCs. The number of ER alphaKO EPCs (42.4+/-1.5; P<0.001) and ER betaKO EPCs (55.4+/-1.8; P=0.03) incorporated into the ischemic border zone was reduced as compared with wild-type (WT) EPCs (72.5+/-1.3). In bone marrow transplantation (BMT) models, the number of mobilized endogenous EPCs in E2-treated mice was significantly reduced in ER alphaKO BMT (WT mice transplanted with ER alphaKO bone marrow) (2.03+/-0.18%; P=0.004 versus WT BMT) and ER betaKO BMT (2.62+/-0.07%; P=0.02 versus WT) compared with WT BMT (2.87+/-0.13%) (WT to WT BMT as control) mice. Capillary density at the border zone of ischemic myocardium also was significantly reduced in ER alphaKO BMT and ER betaKO BMT compared with WT mice (WT BMT, 1718+/-75/mm2; ER alphaKO BMT, 1107+/-48/mm2; ER betaKO BMT, 1567+/-50/mm2). ER alpha mRNA was expressed more abundantly on EPCs compared with ER beta. Moreover, vascular endothelial growth factor was significantly downregulated on ER alphaKO EPCs compared with WT EPCs both in vitro and in vivo. CONCLUSIONS: Both ER alpha and ER beta contribute to E2-mediated EPC activation and tissue incorporation and to preservation of cardiac function after myocardial infarction. ER alpha plays a more prominent role in this process. Moreover, ER alpha contributes to upregulation of vascular endothelial growth factor, revealing possible mechanisms of an effect of E2 on EPC biology. Finally, these data provide additional evidence of the importance of bone marrow-derived EPC phenotype in ischemic tissue repair.


Assuntos
Células da Medula Óssea/citologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Coração/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Transplante de Células-Tronco , Animais , Cardiotônicos/farmacologia , Diferenciação Celular , Células Cultivadas , Células Endoteliais/citologia , Estradiol/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/deficiência , Receptor beta de Estrogênio/deficiência , Feminino , Mobilização de Células-Tronco Hematopoéticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recuperação de Função Fisiológica , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Proc Natl Acad Sci U S A ; 103(29): 11015-20, 2006 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16835303

RESUMO

The transcription factor E2F1 is known to regulate cell proliferation and has been thought to modulate tumorigenesis via this mechanism alone. Here we show that mice deficient in E2F1 exhibit enhanced angiogenesis. The proangiogenic phenotype in E2F1 deficiency is the result of overproduction of vascular endothelial growth factor (VEGF) and is prevented by VEGF blockade. Under hypoxic conditions, E2F1 down-regulates the expression of VEGF promoter activity by associating with p53 and specifically down-regulating expression of VEGF but not other hypoxia-inducible genes, suggesting a promoter structure context-dependent regulation mechanism. We found that the minimum VEGF promoter mediating transcriptional repression by E2F1 features an E2F1- binding site with four Sp-1 sites in close proximity. These data disclose an unexpected function of endogenous E2F1: regulation of angiogenic activity via p53-dependent transcriptional control of VEGF expression.


Assuntos
Ciclo Celular , Fator de Transcrição E2F1/metabolismo , Transcrição Gênica/genética , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Células Cultivadas , Regulação para Baixo , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Supressora de Tumor p53/genética
10.
Circ Res ; 98(5): 697-704, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16484619

RESUMO

Delayed reendothelialization contributes to restenosis after angioplasty and stenting in diabetes. Prior data have shown that bone marrow (BM)-derived endothelial progenitor cells (EPCs) contribute to endothelial recovery after arterial injury. We investigated the hypothesis that the EPC contribution to reendothelialization may be impaired in diabetes, resulting in delayed reendothelialization. Reendothelialization was significantly reduced in diabetic mice compared with nondiabetic mice in a wire-induced carotid denudation model. The EPC contribution to neoendothelium was significantly reduced in Tie2/LacZ BM-transplanted diabetic versus nondiabetic mice. BM from diabetic and nondiabetic mice was transplanted into nondiabetic mice, revealing that reendothelialization was impaired in the recipients of diabetic BM. To examine the relative roles of denuded artery versus EPCs in diabetes, we injected diabetic and nondiabetic EPCs intravenously after arterial injury in diabetic and nondiabetic mice. Diabetic EPCs recruitment to the neoendothelium was significantly reduced, regardless of the diabetic status of the recipient mice. In vitro, diabetic EPCs exhibited decreased migration and adhesion activities. Vascular endothelial growth factor and endothelial NO synthase expressions were also significantly reduced in diabetic EPCs. Notably, thrombospondin-1 mRNA expression was significantly upregulated in diabetic EPCs, associating with the decreased EPC adhesion activity in vitro and in vivo. Reendothelialization is impaired by malfunctioning EPCs in diabetes. Diabetic EPCs have phenotypic differences involving thrombospondin-1 expression compared with nondiabetic EPCs, revealing potential novel mechanistic insights and therapeutic targets to improve reendothelialization and reduce restenosis in diabetes.


Assuntos
Diabetes Mellitus/fisiopatologia , Células Endoteliais/fisiologia , Células-Tronco/fisiologia , Trombospondina 1/fisiologia , Animais , Transplante de Medula Óssea , Adesão Celular , Movimento Celular , Células Cultivadas , Citocinas/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
11.
Nat Clin Pract Cardiovasc Med ; 3 Suppl 1: S123-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501618

RESUMO

Previous studies have shown that local angiogenic gene therapy acts, in part, by recruiting endothelial progenitor cells (EPCs) to ischemic tissue. Recent data indicate that patients with the most severe vascular disease may have insufficient or deficient EPCs and the poorest response to angiogenic therapy. Accordingly, we hypothesized that combining human CD34(+) cell implantation with local vascular endothelial growth factor 2 (phVEGF2) gene therapy might overcome these deficiencies. The addition of VEGF2 to EPC cultures resulted in significant and dose-dependent decreases in EPC apoptosis. Phosphorylated Akt (p-Akt) was increased in VEGF2-treated EPCs. In vivo, myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in 34 immunodeficient rats. The animals were then randomized to one of four treatment groups: cell therapy alone with human CD34(+) cells; VEGF2 gene therapy alone; combination therapy with CD34(+) cells plus phVEGF2; or CD34(-) cells and 50 microg empty plasmid. Four weeks after MI, animals treated with combination therapy showed improved fractional shortening, increased capillary density, and reduced infarct size compared with the other three groups. Combination therapy was also associated with an increased number of circulating EPCs 1 week after MI. Combined subtherapeutic doses of cell and gene therapy result in a significant therapeutic effect compared to monotherapy. This approach may overcome therapeutic failures (e.g. inability of certain patients to mobilize sufficient EPCs) and may also offer safety advantages by allowing lower dosing strategies.


Assuntos
Antígenos CD34/análise , Células Endoteliais/transplante , Infarto do Miocárdio/terapia , Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose , Células Cultivadas , Terapia Combinada , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Feminino , Terapia Genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Ratos , Ratos Nus , Transplante Autólogo , Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular/farmacologia
12.
J Exp Med ; 203(1): 153-63, 2006 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-16401693

RESUMO

The cell surface receptor alpha4 integrin plays a critical role in the homing, engraftment, and maintenance of hematopoietic progenitor cells (HPCs) in the bone marrow (BM). Down-regulation or functional blockade of alpha4 integrin or its ligand vascular cell adhesion molecule-1 mobilizes long-term HPCs. We investigated the role of alpha4 integrin in the mobilization and homing of BM endothelial progenitor cells (EPCs). EPCs with endothelial colony-forming activity in the BM are exclusively alpha4 integrin-expressing cells. In vivo, a single dose of anti-alpha4 integrin antibody resulted in increased circulating EPC counts for 3 d. In hindlimb ischemia and myocardial infarction, systemically administered anti-alpha4 integrin antibody increased recruitment and incorporation of BM EPCs in newly formed vasculature and improved functional blood flow recovery and tissue preservation. Interestingly, BM EPCs that had been preblocked with anti-alpha4 integrin ex vivo or collected from alpha4 integrin-deficient mice incorporated as well as control cells into the neovasculature in ischemic sites, suggesting that alpha4 integrin may be dispensable or play a redundant role in EPC homing to ischemic tissue. These data indicate that functional disruption of alpha4 integrin may represent a potential angiogenic therapy for ischemic disease by increasing the available circulating supply of EPCs.


Assuntos
Integrina alfa4/metabolismo , Isquemia Miocárdica/fisiopatologia , Neovascularização Fisiológica , Células-Tronco/fisiologia , Animais , Medula Óssea , Movimento Celular , Células Endoteliais , Integrina alfa4/genética , Masculino , Camundongos , Camundongos Knockout
13.
Regen Med ; 1(3): 337-45, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-17465787

RESUMO

Since the first experiments of cell transplantation into the heart were performed in the early 1990s, the identification of adult stem cells has triggered attempts to regenerate damaged heart tissue by cellular transplantation. Until recently, a multitude of adult stem or progenitor cells from various tissues have been proposed to meet this end. Bone marrow in particular has emerged as the most promising source for stem and progenitor cells because, besides being the organ of hematopoietic maintenance, it contains a complex assortment of stem and progenitor cells. A large body of provocative experimental evidence for vascular and myocardial regeneration by these cells has generated further enthusiasm for their use. However, many questions remain unanswered in this new field of research regarding the therapeutic potential and the mechanisms responsible for the observed therapeutic effects. In this review, the authors discuss the therapeutic capacity of currently available representative bone marrow-derived stem and progenitor cells for treating ischemic heart diseases.


Assuntos
Células da Medula Óssea/citologia , Isquemia Miocárdica/terapia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Adulto , Diferenciação Celular , Previsões , Humanos , Modelos Cardiovasculares , Infarto do Miocárdio/patologia , Miocárdio/citologia , Miócitos Cardíacos , Neovascularização Fisiológica , Regeneração , Células-Tronco/classificação , Células-Tronco/citologia , Doadores de Tecidos
14.
Vasc Med ; 10(1): 15-22, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15920995

RESUMO

Hyperhomocyst(e)inemia (HH) is an established independent risk factor for coronary, cerebral and peripheral vascular diseases. Recent studies have indicated that certain cardiovascular risk factors, including diabetes and hypercholesterolemia, impair expression of vascular endothelial growth factor (VEGF) and endogenous angiogenesis. In this study, we investigate the impact of moderate HH on angiogenesis and VEGF pathway in a mouse model of hindlimb ischemia. Upon induction of unilateral hindlimb ischemia, endogenous angiogenesis, expression of VEGF, and phosphorylation of the VEGF receptor Flk-1 were evaluated in mice heterozygous for a deletion of the cystathionine beta-synthase gene (CBS) and compared with those observed in CBS+/+ mice. CBS+/- mice exhibit moderate HH, as demonstrated by measuring plasma total homocyst(e)ine (tHcy) levels, which were significantly higher in these animals compared with CBS+/+ mice (4.77 +/- 0.82 vs 2.10 +/- 0.28, p < 0.01). Twenty-eight days after induction of ischemia, hindlimb blood flow was significantly reduced in CBS+/- mice compared with CBS+/+ animals (0.49 +/- 0.03, n = 12 vs 0.71 +/- 0.09, n = 10; p < 0.05). In addition, there was a significant negative correlation between plasma homocyst(e)ine levels and the laser Doppler perfusion ratio in CBS+/- mice (p = 0.0087, r = -0.7171). While VEGF expression and Flk-1 phosphorylation were not impaired in the ischemic muscles of CBS+/- mice, phosphorylation of the endothelial cell survival factor Akt was significantly inhibited by homocyst(e)ine in a dose-dependent manner in human umbilical vein endothelial cell (HUVECs) in vitro. In conclusion, our findings demonstrate that endogenous angiogenesis is inversely related to plasma levels of homocyst(e)ine in genetically engineered, heterozygous mice with moderate HH. This impairment, however, is not dependent on reduced expression of VEGF or impaired phosphorylation of its receptor Flk-1. In contrast, our data suggest that impaired Akt phosphorylation mediates the impairment of angiogenesis associated with HH.


Assuntos
Hiper-Homocisteinemia/fisiopatologia , Isquemia/sangue , Neovascularização Fisiológica/fisiologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos , Fosforilação , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Circulation ; 112(1): 93-102, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15983249

RESUMO

BACKGROUND: Peripheral neuropathy is a frequent and major complication of diabetes. METHODS AND RESULTS: Severe peripheral neuropathy developed in type II diabetic mice, characterized by significant slowing of motor and sensory nerve conduction velocities. Rosuvastatin restored nerve vascularity, including vessel size, and nerve function also recovered to the levels of nondiabetic mice. Neuronal nitric oxide synthase expression in sciatic nerves was reduced in diabetic mice but was preserved by rosuvastatin. Coadministration of a nitric oxide synthase inhibitor with rosuvastatin attenuated the beneficial effects of rosuvastatin on nerve function and limited the recovery of vasa nervorum and nerve function. In vitro, rosuvastatin inhibited downregulation of neuronal nitric oxide synthase expression induced by high-glucose conditions in cultured Schwann cells. Furthermore, Akt phosphorylation in Schwann cells, downregulated by high-glucose conditions, was also restored by rosuvastatin, consistent with the change of neuronal nitric oxide synthase expression. Akt inhibition independently reduced neuronal nitric oxide synthase expression in Schwann cells in low-glucose cultures. CONCLUSIONS: These data indicate that the HMG-CoA reductase inhibitor rosuvastatin has a favorable effect on diabetic neuropathy independent of its cholesterol-lowering effect. Our data provide evidence that this effect may be mediated in part via neuronal nitric oxide synthase/nitric oxide and phosphatidylinositol 3-kinase/Akt-signaling pathways and also suggest that restoration or preservation of the microcirculation of the sciatic nerve may be involved.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Óxido Nítrico Sintase Tipo I/fisiologia , Vasa Nervorum/efeitos dos fármacos , Animais , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Regulação para Baixo/efeitos dos fármacos , Fluorbenzenos/administração & dosagem , Fluorbenzenos/farmacologia , Glucose/farmacologia , Masculino , Camundongos , Camundongos Mutantes , Óxido Nítrico Sintase Tipo I/genética , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Rosuvastatina Cálcica , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Resultado do Tratamento
16.
Circulation ; 111(20): 2662-70, 2005 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15897348

RESUMO

BACKGROUND: Toxic neuropathies induced by cisplatin and other chemotherapeutic agents are important clinical problems because of their high incidence, their lack of effective treatment, and the fact that neuropathy represents a dose-limiting factor for these therapies. The pathogenic basis for toxic neuropathies induced by chemotherapeutic agents has not been completely elucidated. METHODS AND RESULTS: We investigated the hypothesis that experimental toxic neuropathy results from an antiangiogenic effect of these drugs, resulting in destruction of the vasa nervorum, and accordingly that the neuropathy could be prevented or reversed by locally administered VEGF gene transfer without augmenting tumor growth. In an animal model of cisplatin-induced neuropathy, nerve blood flow was markedly attenuated, and there was a profound reduction in the number of vasa nervorum associated with marked endothelial cell apoptosis, resulting in a severe peripheral neuropathy with focal axonal degeneration characteristic of ischemic neuropathy. After intramuscular gene transfer of naked plasmid DNA encoding VEGF-1 in animals with an established neuropathy, vascularity and blood flow returned to levels similar to those of control rats, peripheral nerve function was restored, and histological nerve architecture was normalized. Gene therapy administered in parallel with cisplatin chemotherapy completely attenuated endothelial cell apoptosis and inhibited destruction of nerve vasculature, deterioration of nerve function, and axonal degeneration. In a rat tumor model, VEGF gene transfer administered locally did not alter tumor growth or vascularity. CONCLUSIONS: These findings implicate microvascular damage as the basis for toxic neuropathy induced by cisplatin and suggest that local angiogenic gene therapy may constitute a novel prevention or treatment for this disorder without augmenting tumor growth or vascularization.


Assuntos
Cisplatino/efeitos adversos , Doenças do Sistema Nervoso Periférico/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Animais , Apoptose , Endotélio Vascular/citologia , Terapia Genética , Humanos , Masculino , Neoplasias/irrigação sanguínea , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/terapia , Ratos , Ratos Sprague-Dawley , Carga Tumoral/efeitos dos fármacos , Veias Umbilicais/citologia , Vasa Nervorum/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
17.
Circulation ; 111(9): 1114-20, 2005 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-15723985

RESUMO

BACKGROUND: The function of bone marrow-derived endothelial progenitor cells (EPCs) in repair of ischemic tissue has been the subject of intense scrutiny, and the capacity of these cells to contribute significantly to new blood vessels remains controversial. The possibility that EPCs could act as reservoirs of cytokines has been implied by several observations; however, a specific role for cytokine delivery has not been identified. METHODS AND RESULTS: We performed a series of experiments that revealed the rapid recruitment of EPCs to the myocardium by very short periods of ischemia, so-called ischemic preconditioning. The recruited EPCs express an array of potentially cardioprotective cytokines including nitric oxide synthase isoforms. Bone marrow transplantation studies, using donor marrow null for nitric oxide synthase isoforms, revealed that both endothelial and inducible nitric oxide synthase derived from bone marrow cells play essential roles in the cardioprotective effect that normally occurs after ischemic preconditioning. CONCLUSIONS: These findings provide novel insights about the role of bone marrow-derived cells in ischemic preconditioning and also reveal that distinct mechanisms regulate recovery after ischemia-reperfusion and chronic ischemic injury.


Assuntos
Transplante de Medula Óssea , Células Endoteliais/enzimologia , Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/patologia , Isquemia Miocárdica/fisiopatologia , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico/fisiologia , Células-Tronco/enzimologia , Animais , Células Sanguíneas/citologia , Células Sanguíneas/enzimologia , Células da Medula Óssea/citologia , Células da Medula Óssea/enzimologia , Capilares/patologia , Hipóxia Celular , Movimento Celular , Células Cultivadas/enzimologia , Circulação Coronária , Citocinas/fisiologia , Células Endoteliais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Miocárdio/patologia , Neovascularização Fisiológica , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Fosforilação , Processamento de Proteína Pós-Traducional , Quimera por Radiação , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
18.
J Clin Invest ; 115(2): 326-38, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15690083

RESUMO

We have identified a subpopulation of stem cells within adult human BM, isolated at the single-cell level, that self-renew without loss of multipotency for more than 140 population doublings and exhibit the capacity for differentiation into cells of all 3 germ layers. Based on surface marker expression, these clonally expanded human BM-derived multipotent stem cells (hBMSCs) do not appear to belong to any previously described BM-derived stem cell population. Intramyocardial transplantation of hBMSCs after myocardial infarction resulted in robust engraftment of transplanted cells, which exhibited colocalization with markers of cardiomyocyte (CMC), EC, and smooth muscle cell (SMC) identity, consistent with differentiation of hBMSCs into multiple lineages in vivo. Furthermore, upregulation of paracrine factors including angiogenic cytokines and antiapoptotic factors, and proliferation of host ECs and CMCs, were observed in the hBMSC-transplanted hearts. Coculture of hBMSCs with CMCs, ECs, or SMCs revealed that phenotypic changes of hBMSCs result from both differentiation and fusion. Collectively, the favorable effect of hBMSC transplantation after myocardial infarction appears to be due to augmentation of proliferation and preservation of host myocardial tissues as well as differentiation of hBMSCs for tissue regeneration and repair. To our knowledge, this is the first demonstration that a specific population of multipotent human BM-derived stem cells can induce both therapeutic neovascularization and endogenous and exogenous cardiomyogenesis.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Coração/fisiologia , Células-Tronco Multipotentes/transplante , Infarto do Miocárdio/terapia , Regeneração/fisiologia , Adulto , Animais , Células da Medula Óssea/citologia , Linhagem da Célula/fisiologia , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Masculino , Células-Tronco Multipotentes/fisiologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/fisiologia , Neovascularização Fisiológica/fisiologia , Ratos , Ratos Nus
19.
Circulation ; 110(1): 36-45, 2004 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-15210598

RESUMO

BACKGROUND: Drug-eluting stents represent a useful strategy for the prevention of restenosis using various antiproliferative drugs. These strategies share the liability of impairing endothelial recovery, thereby altering the natural biology of the vessel wall and increasing the associated risk of stent thrombosis. Accordingly, we tested the hypothesis that local delivery via gene-eluting stent of naked plasmid DNA encoding for human vascular endothelial growth factor (VEGF)-2 could achieve similar reductions in neointima formation while accelerating, rather than inhibiting, reendothelialization. METHODS AND RESULTS: phVEGF 2-plasmid (100 or 200 microg per stent)-coated BiodivYsio phosphorylcholine polymer stents versus uncoated stents were deployed in a randomized, blinded fashion in iliac arteries of 40 normocholesterolemic and 16 hypercholesterolemic rabbits. Reendothelialization was nearly complete in the VEGF stent group after 10 days and was significantly greater than in control stents (98.7+/-1% versus 79.0+/-6%, P<0.01). At 3 months, intravascular ultrasound analysis revealed that lumen cross-sectional area (4.2+/-0.4 versus 2.27+/-0.3 mm(2), P<0.001) was significantly greater and percent cross-sectional narrowing was significantly lower (23.4+/-6 versus 51.2+/-10, P<0.001) in VEGF stents compared with control stents implanted in hypercholesterolemic rabbits. Transgene expression was detectable in the vessel wall along with improved functional recovery of stented segments, resulting in a 2.4-fold increase in NO production. CONCLUSIONS: Acceleration of reendothelialization via VEGF-2 gene-eluting stents provides an alternative treatment strategy for the prevention of restenosis. VEGF-2 gene-eluting stents may be considered as a stand-alone or combination therapy.


Assuntos
Arteriopatias Oclusivas/prevenção & controle , Técnicas de Transferência de Genes , Terapia Genética/métodos , Stents , Fatores de Crescimento do Endotélio Vascular/genética , Animais , Arteriopatias Oclusivas/diagnóstico por imagem , Arteriopatias Oclusivas/patologia , Terapia Combinada , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Expressão Gênica , Masculino , Óxido Nítrico/biossíntese , Plasmídeos/administração & dosagem , Coelhos , Células-Tronco/citologia , Ultrassonografia , Fatores de Crescimento do Endotélio Vascular/análise
20.
J Clin Invest ; 111(5): 717-25, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12618526

RESUMO

Although lymphedema is a common clinical condition, treatment for this disabling condition remains limited and largely ineffective. Recently, it has been reported that overexpression of VEGF-C correlates with increased lymphatic vessel growth (lymphangiogenesis). However, the effect of VEGF-C-induced lymphangiogenesis on lymphedema has yet to be demonstrated. Here we investigated the impact of local transfer of naked plasmid DNA encoding human VEGF-C (phVEGF-C) on two animal models of lymphedema: one in the rabbit ear and the other in the mouse tail. In a rabbit model, following local phVEGF-C gene transfer, VEGFR-3 expression was significantly increased. This gene transfer led to a decrease in thickness and volume of lymphedema, improvement of lymphatic function demonstrated by serial lymphoscintigraphy, and finally, attenuation of the fibrofatty changes of the skin, the final consequences of lymphedema. The favorable effect of phVEGF-C on lymphedema was reconfirmed in a mouse tail model. Immunohistochemical analysis using lymphatic-specific markers: VEGFR-3, lymphatic endothelial hyaluronan receptor-1, together with the proliferation marker Ki-67 Ab revealed that phVEGF-C transfection potently induced new lymphatic vessel growth. This study, we believe for the first time, documents that gene transfer of phVEGF-C resolves lymphedema through direct augmentation of lymphangiogenesis. This novel therapeutic strategy may merit clinical investigation in patients with lymphedema.


Assuntos
Fatores de Crescimento Endotelial/genética , Terapia Genética , Sistema Linfático/fisiologia , Linfedema/terapia , Neovascularização Fisiológica/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Imuno-Histoquímica , Dados de Sequência Molecular , Fosforilação , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transgenes , Fator C de Crescimento do Endotélio Vascular , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...