Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Basic Res Cardiol ; 103(6): 572-81, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18604625

RESUMO

OBJECTIVE: Recent studies indicate that platelets influence endothelial progenitor cell (EPC) recruitment to sites of vascular injury and promote their differentiation to an endothelial phenotype. Patients with cardiovascular risk factors (CVRF) demonstrate a reduced number and impaired function of EPC, as well as platelet hyper-reactivity. Therefore, we investigated the interaction of platelets and EPC from patients with CVRF. METHODS AND RESULTS: Co-incubation of platelets and peripheral blood mononuclear cells, both from healthy volunteers, dose-dependently increased the number of adherent EPC. In contrast, patient-derived platelets failed to augment the number of adherent and migrating healthy and patient-derived EPC. However, co-incubation of platelets from healthy donors with mononuclear cells from patients with CVRF significantly enhanced the number of EPC, indicating that platelets from healthy volunteers are able to partially rescue the impairment of patient-derived EPC formation. Likewise, healthy donor-derived platelets augmented the impaired migration and clonal capacity of patient-derived EPC. Analysis of individual CVRF of platelet donors revealed that only diabetes mellitus inversely correlated with EPC number, colony formation and migration. The platelet supernatants from healthy volunteers that significantly increased EPC number contained IL-6, SDF-1, sCD40L and PDGF. While sCD40L and PDGF levels were comparable in platelet supernatants from healthy volunteers and patients with CVRF, the release of IL-6 and SDF-1 by patient-derived platelets was rather increased, thus, indicating that these soluble factors are not mediating the effect of platelet supernatants. CONCLUSION: Healthy volunteer-derived platelets provide a source of soluble factors to improve the number and function of EPC from patients with cardiovascular risk factors, particularly diabetes mellitus.


Assuntos
Plaquetas/patologia , Doenças Cardiovasculares/epidemiologia , Comunicação Celular/fisiologia , Diabetes Mellitus/patologia , Diabetes Mellitus/fisiopatologia , Endotélio Vascular/patologia , Células-Tronco Mesenquimais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Plaquetas/metabolismo , Estudos de Casos e Controles , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12/metabolismo , Diabetes Mellitus/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Interleucina-6/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Fatores de Risco
2.
J Mol Cell Cardiol ; 45(3): 429-36, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18619973

RESUMO

Endothelial progenitor cells (EPC) significantly contribute to neovascularization and endothelial regeneration. Risk factors for coronary artery disease, particularly diabetes mellitus, reduce the number and functional activity of EPC. As we have recently shown, expression and activity of the matrix degrading cysteine protease cathepsin L in EPC is required for tissue invasion and EPC-mediated improvement of neovascularization. Therefore, we investigated the effect of high glucose and diabetes mellitus on EPC invasion and cathepsin L activity. Incubation of EPC with high levels of glucose (10-30 mM) dose-dependently decreased cathepsin L activity (glucose 20 mM: 67+/-4% compared to control; p<0.05) and protein expression (48+/-5% of control, p<0.05). In contrast, other proteases of the cathepsin family such as cathepsins D and O, and the matrix metalloproteinases MMP-2 and MMP-9 were not altered with high glucose. Cathepsin L mRNA was not affected suggesting that a posttranscriptional mechanism is responsible for cathepsin L down-regulation. As a functional consequence, high glucose significantly reduced the gelatinolytic activity and invasion of EPC (50+/-5% of control). Importantly, EPC of patients with type 2 diabetes revealed profoundly decreased cathepsin L expression and activity as compared to EPC derived from healthy controls. Taken together, high glucose significantly reduces the protein expression and activity of cathepsin L, which is involved in matrix degradation and required for invasion of EPC into the ischemic tissue, and, thereby, may limit the functional capacity of EPC to improve neovascularization in diabetics.


Assuntos
Catepsinas/antagonistas & inibidores , Movimento Celular/fisiologia , Endotélio Vascular/metabolismo , Glucose/efeitos adversos , Glucose/fisiologia , Células-Tronco/metabolismo , Adulto , Idoso , Catepsina L , Catepsinas/biossíntese , Catepsinas/metabolismo , Catepsinas/fisiologia , Células Cultivadas , Cisteína Endopeptidases/biossíntese , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/fisiologia , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Células-Tronco/enzimologia , Células-Tronco/patologia
3.
J Mol Cell Cardiol ; 39(5): 733-42, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16199052

RESUMO

Circulating endothelial progenitor cells (EPC) are incorporated into newly formed capillaries, enhance neovascularization after hind limb ischemia and improve cardiac function after ischemic injury. Incorporated progenitor cells may also promote neovascularization and cardiac regeneration by releasing factors, which act in a paracrine manner to support local angiogenesis and mobilize tissue residing progenitor cells. Therefore, we analyzed the expression profile of cytokines in human peripheral blood-derived EPC as opposed to human umbilical vein endothelial cells (HUVEC), human microvascular endothelial cells (HMVEC), and CD14(+) monocytes by microarray technology. A gene tree analysis revealed a distinct expression pattern of angiogenic growth factors in EPC, mature endothelial cells, and CD14(+) monocytes. VEGF-A, VEGF-B, SDF-1, and IGF-1 mRNA levels were higher in EPC as compared to HUVEC or HMVEC. The enhanced mRNA expression was paralleled by a significant release of VEGF, SDF-1, and IGF-1 protein into the cell culture supernatant of EPC. Moreover, immunohistological analysis of ischemic limbs from nude rats revealed that VEGF is also released from recruited human EPC in vivo. As a functional consequence, conditioned medium of EPC induced a strong migratory response of mature endothelial cells, which was significantly inhibited by VEGF and SDF-1 neutralizing antibodies. Finally, conditioned medium of EPC significantly stimulated the migration of cardiac resident c-kit(+) progenitor cells in vitro. Taken together, EPC exhibit a high expression of angiogenic growth factors, which enhanced migration of mature endothelial cells and tissue resident cardiac progenitor cells. In addition to the physical contribution of EPC to newly formed vessels, the enhanced expression of cytokines may be a supportive mechanism to improve blood vessel formation and cardiac regeneration after cell therapy.


Assuntos
Movimento Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Miocárdio/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Angiogênicas/genética , Animais , Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Perfilação da Expressão Gênica , Substâncias de Crescimento/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Solubilidade
4.
J Exp Med ; 201(11): 1825-35, 2005 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-15928198

RESUMO

The regulation of acetylation is central for the epigenetic control of lineage-specific gene expression and determines cell fate decisions. We provide evidence that the inhibition of histone deacetylases (HDACs) blocks the endothelial differentiation of adult progenitor cells. To define the mechanisms by which HDAC inhibition prevents endothelial differentiation, we determined the expression of homeobox transcription factors and demonstrated that HoxA9 expression is down-regulated by HDAC inhibitors. The causal involvement of HoxA9 in the endothelial differentiation of adult progenitor cells is supported by the finding that HoxA9 overexpression partially rescued the endothelial differentiation blockade induced by HDAC inhibitors. Knockdown and overexpression studies revealed that HoxA9 acts as a master switch to regulate the expression of prototypical endothelial-committed genes such as endothelial nitric oxide synthase, VEGF-R2, and VE-cadherin, and mediates the shear stress-induced maturation of endothelial cells. Consistently, HoxA9-deficient mice exhibited lower numbers of endothelial progenitor cells and showed an impaired postnatal neovascularization capacity after the induction of ischemia. Thus, HoxA9 is regulated by HDACs and is critical for postnatal neovascularization.


Assuntos
Diferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Histona Desacetilases/metabolismo , Proteínas de Homeodomínio/biossíntese , Animais , Antígenos CD , Caderinas/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Sangue Fetal/citologia , Sangue Fetal/fisiologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Humanos , Isquemia/metabolismo , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Estresse Mecânico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Blood ; 104(12): 3591-7, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15161665

RESUMO

Adult and embryonic stem cells hold great promise for regenerative medicine. Expression profiling of stem cells revealed a characteristic imprint of genes, so-called "stemness" genes, providing resistance to stress. Circulating progenitor cells with an endothelial phenotype (EPCs) can be isolated from peripheral blood and contribute to neovascularization and endothelial regeneration. We investigated whether EPCs are equipped with an antioxidative defense to provide resistance against oxidative stress. EPCs exhibited a significantly lower basal reactive oxygen species (ROS) concentration as compared with mature umbilical vein endothelial cells (HUVECs). Incubation with H(2)O(2) (500 microM) or the redox cycler LY-83583 (10 microM) profoundly increased the ROS concentration to 3- and 4-fold and induced apoptosis in HUVECs. In contrast, H(2)O(2) and LY-83583 induced only a minor increase in intracellular ROS levels and apoptosis in EPCs. Consistently, the expression of the intracellular antioxidative enzymes catalase, glutathione peroxidase and manganese superoxide dismutase (MnSOD), was significantly higher in EPCs versus HUVECs and human microvascular endothelial cells. In accordance, combined inhibition of these antioxidative enzymes increased ROS levels in EPCs and impaired EPC survival and migration. Taken together, EPCs reveal a higher expression of antioxidative enzymes and, thus, are exquisitely equipped to be protected against oxidative stress consistent with their progenitor cell character.


Assuntos
Endotélio Vascular/citologia , Estresse Oxidativo/genética , Células-Tronco/fisiologia , Apoptose/efeitos dos fármacos , Células Sanguíneas , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Endotélio Vascular/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Imunidade Inata/genética , Oxirredutases/análise , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Veias Umbilicais/citologia
6.
Shock ; 21(1): 13-6, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14676678

RESUMO

Ischemia in various organs and tissues takes place during and as a direct result of multiple trauma (MT). Bone marrow-derived endothelial progenitor cells (EPCs) are involved in neovascularization after ischemic incidences. Here, we report that serum derived from patients with MT stimulates differentiation of EPCs in vitro from peripheral blood mononuclear cells (PBMCs). EPCs were identified by DiL-Acetyl-LDL-uptake with concomitant UEA-I-lectin binding. A significant increase in EPC numbers was noted when PBMCs were cultivated for 72 h with the serum of MT patients (n = 25) obtained at 5 days. Furthermore, serum from MT patients enhanced the functional acting of EPCs to form prevascular structures in matrigel. Reverse transcription polymerase chain reaction analysis revealed gene expression of transforming growth factor (TGF)-beta1- and vascular endothelial growth factor (VEGF) receptors 1 and 2. Reverse transcription polymerase chain reaction analysis was based on further cultivated cell preparations, which contained at least 80% EPCs. Moreover, the addition of recombinant VEGF or low concentrations of TGF-beta increased EPC differentiation. In addition, neutralization of TGF-beta1 and of VEGF165 in MT serum using specific antibodies resulted in a significant decrease in EPC differentiation. Our data indicate that TGF-beta1 and VEGF165 play a pivotal role for EPC differentiation induced by serum of polytrauma patients.


Assuntos
Meios de Cultura , Endotélio/citologia , Soro/citologia , Células-Tronco/citologia , Western Blotting , Diferenciação Celular , Células Cultivadas , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Citometria de Fluxo , Humanos , Isquemia , Laminina/farmacologia , Lectinas/metabolismo , Leucócitos Mononucleares/metabolismo , Neovascularização Patológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteoglicanas/farmacologia , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Choque , Fatores de Tempo , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta1 , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ferimentos e Lesões , Fator de von Willebrand/metabolismo
7.
Circulation ; 108(20): 2511-6, 2003 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-14581410

RESUMO

BACKGROUND: Transplantation of ex vivo expanded circulating endothelial progenitor cells (EPCs) from peripheral blood mononuclear cells improves the neovascularization after critical ischemia. However, the origin of the endothelial progenitor lineage and its characteristics have not yet been clearly defined. Therefore, we investigated whether the phenotype and functional capacity of EPCs to improve neovascularization depend on their monocytic origin. METHODS AND RESULTS: Monocytic CD14+ cells were isolated from mononuclear cells and incubated on fibronectin-coated dishes in endothelial medium in the presence of vascular endothelial growth factor. After 4 days of cultivation, adherent cells deriving from CD14+ or CD14- mononuclear cells showed equal expression of endothelial marker proteins and capacity for clonal expansion as determined by measuring endothelial colony-forming units. In addition, transplanted EPCs (5x10(5) cells) deriving from CD14+ or CD14- cells were incorporated into vascular structures of nude mice after hind-limb ischemia and significantly improved neovascularization from 0.27+/-0.12 (no cells) to 0.66+/-0.12 and 0.65+/-0.17, respectively (P<0.001; laser Doppler-derived relative blood flow). In contrast, no functional improvement of neovascularization was detected when freshly isolated CD14+ mononuclear cells without ex vivo expansion were used (0.33+/-0.17). Moreover, macrophages or dendritic cells differentiated from isolated CD14+ cells were significantly less effective in improving neovascularization than EPCs cultivated from the same starting population (P<0.01). CONCLUSIONS: These data demonstrate that EPCs can be generated from nonmonocytic CD14- peripheral blood mononuclear cells and exhibit a unique functional activity to improve neovascularization after hind-limb ischemia.


Assuntos
Isquemia/terapia , Leucócitos Mononucleares/citologia , Neovascularização Fisiológica/fisiologia , Transplante de Células-Tronco , Animais , Antígenos de Diferenciação/biossíntese , Diferenciação Celular , Separação Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Modelos Animais de Doenças , Feminino , Membro Posterior/irrigação sanguínea , Humanos , Separação Imunomagnética , Isquemia/imunologia , Leucócitos Mononucleares/metabolismo , Receptores de Lipopolissacarídeos/biossíntese , Camundongos , Camundongos Nus , Neovascularização Fisiológica/imunologia , Células-Tronco/citologia , Células-Tronco/metabolismo
8.
Circulation ; 106(8): 981-6, 2002 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-12186804

RESUMO

BACKGROUND: The CD40/CD40 ligand system is involved in atherogenesis. Activated T lymphocytes and platelets, which express high amounts of CD40 ligand (CD40L) on their surface, contribute significantly to plaque instability with ensuing thrombus formation, leading to acute coronary syndromes. Because reendothelialization may play a pivotal role for plaque stabilization, we investigated a potential role of CD40L on endothelial cell (EC) migration. METHODS AND RESULTS: Stimulation of ECs with recombinant CD40L prevented vascular endothelial growth factor (VEGF)-induced EC migration, as determined by a "scratched wound assay." In addition, activated T lymphocytes and platelets significantly inhibited VEGF-induced EC migration and tube formation in vitro. Because the activation of endothelial nitric oxide (NO) synthase and the release of NO are required for EC migration and angiogenesis, we analyzed the effect of NO. Coincubation with the NO donor S-nitroso-N-acetyl-penicillamine (SNAP) did not reverse the inhibitory effect of CD40L on VEGF-induced EC migration and tube formation. In addition, EC migration induced by SNAP was completely inhibited by CD40L. CD40L, however, induced the production of reactive oxygen species and reduced endothelial NO bioavailability. This reactive oxygen species-dependent effect of CD40L stimulation was reversed with vitamin C or N-acetylcysteine. CONCLUSIONS: The activation of the CD40 receptor inhibits EC migration by increasing reactive oxygen species. The blockade of EC migration by CD40L may critically affect endothelial regeneration after plaque erosion and thereby may contribute to the increased risk for development of acute coronary events in patients with high circulating levels of CD40L.


Assuntos
Ligante de CD40/farmacologia , Movimento Celular , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/farmacologia , Plaquetas/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Fatores de Crescimento Endotelial/antagonistas & inibidores , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Humanos , Ativação Linfocitária , Linfocinas/antagonistas & inibidores , Óxido Nítrico/biossíntese , Estresse Oxidativo , Linfócitos T/imunologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
Circ Res ; 91(1): 38-45, 2002 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-12114320

RESUMO

Tyrosine kinase cascades may play a role in the hypoxic regulation of hypoxia-inducible factor (HIF)-1. We investigated the role of tyrosine kinase phosphorylation and of the Shc/Ras cascade on hypoxic HIF-1 stabilization. Exposure of human umbilical vein endothelial cells to hypoxia results in HIF protein stabilization as early as 10 minutes, with a maximum at 3 hours, and also in Shc tyrosine phosphorylation, with a maximum at 10 minutes. To test whether Shc directly mediates hypoxia-induced HIF stabilization, human umbilical vein endothelial cells were transfected with a dominant-negative Shc mutant (dnShc), resulting in significantly reduced HIF protein levels compared with control. Similar results were obtained with cells transfected with dominant-negative Ras, a known downstream effector of Shc. Hypoxia-induced Ras activity was significantly reduced in cells transfected with dnShc compared with control levels, indicating that Ras indeed acts downstream from Shc. Moreover, cells pretreated with a specific Raf-1 kinase inhibitor, a known downstream effector of Ras, exhibited reduced HIF protein levels. To examine the functional consequences of Shc in hypoxic signaling, HIF-1 ubiquitination, protein stabilization, and endothelial cell migration were assessed. Overexpression of dnShc increased ubiquitination of HIF-1 and reduced the half-life of the protein. Moreover, dnShc, dominant-negative Ras, or the Raf-1 kinase inhibitor significantly inhibited migration under hypoxia. Thus, Shc in concert with Ras and Raf-1 contributes to hypoxia-induced HIF-1alpha protein stabilization and endothelial cell migration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Fatores de Transcrição , Western Blotting , Hipóxia Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Cobalto/farmacologia , Cicloeximida/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mutação , Plasmídeos/genética , Proteínas/genética , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-raf/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Transfecção , Tirfostinas/farmacologia , Ubiquitina/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
10.
Circ Res ; 90(6): 737-44, 2002 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-11934843

RESUMO

3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) exert potent vasculoprotective effects. However, the potential contribution to angiogenesis is controversial. In the present study, we demonstrate that atorvastatin dose-dependently affects endothelial cell migration and angiogenesis. In vivo relevant concentrations of 0.01 to 0.1 micromol/L atorvastatin or mevastatin promote the migration of mature endothelial cells and tube formation. Moreover, atorvastatin also increases migration and the potency to form vessel structures of circulating endothelial progenitor cells, which may contribute to vasculogenesis. In contrast, higher concentrations (>0.1 micromol/L atorvastatin) block angiogenesis and migration by inducing endothelial cell apoptosis. The dose-dependent promigratory and proangiogenic effects of atorvastatin on mature endothelial cells are correlated with the activation of the phosphatidylinositol 3-kinase-Akt pathway, as determined by the phosphorylation of Akt and endothelial NO synthase (eNOS) at Ser1177. In addition, the stimulation of migration and tube formation was blocked by phosphatidylinositol 3-kinase inhibitors. In contrast, the well-established stabilization of eNOS mRNA was achieved only at higher concentrations, suggesting that posttranscriptional activation rather than an increase in eNOS expression mediates the proangiogenic effect of atorvastatin. Taken together, these data suggest that statins exert a double-edged role in angiogenesis signaling by promoting the migration of mature endothelial cells and endothelial progenitor cells at low concentrations, whereas the antiangiogenic effects were achieved only at high concentrations.


Assuntos
Proteínas de Bactérias , Movimento Celular/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Monócitos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Pirróis/farmacologia , Apoptose/efeitos dos fármacos , Atorvastatina , Células Cultivadas , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Enterotoxinas/metabolismo , Fibroblastos , Humanos , Monócitos/citologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
FASEB J ; 16(7): 706-8, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11978735

RESUMO

Endostatin is an anti-angiogenic factor that inhibits endothelial cell (EC) migration and induces EC apoptosis. Because nitric oxide (NO) plays a key role in vascular endothelial growth factor (VEGF)-induced angiogenesis, we hypothesized that endostatin interferes with the activation of the endothelial NO synthase (eNOS). Human recombinant endostatin significantly reduced VEGF-induced NO-release, which suggests that endostatin inhibits eNOS activation. Because the activation of eNOS by VEGF is associated with the Akt-dependent phosphorylation of eNOS at Ser1177, we investigated whether endostatin interferes with phosphorylation of eNOS. Endostatin reduced VEGF-induced phosphorylation of eNOS at Ser1177, whereas Akt phosphorylation was not affected. Coinciding with the inhibition of eNOS phosphorylation, endostatin completely blocked VEGF-induced EC migration. The NO-donor SNAP reversed the inhibitory effect of endostatin on EC migration. In addition, endostatin significantly inhibited VEGF-induced tube formation, whereas endostatin did not affect tube formation induced by NO. Finally, a non-dephosphorylatable constitutive active eNOS construct (S1177D), but not constitutive active Akt, abolished the inhibitory effect of endostatin on EC migration. Endostatin activated PP2A, which is known to directly dephosphorylate eNOS at Ser1177. Inhibition of PP2A prevented the inhibitory effect of endostatin. Thus, endostatin inhibits VEGF-induced EC migration and angiogenesis upstream of NO-synthesis via dephosphorylation of eNOS at Ser1177.


Assuntos
Inibidores da Angiogênese/farmacologia , Colágeno/farmacologia , Endotélio Vascular/enzimologia , Óxido Nítrico Sintase/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Serina-Treonina Quinases , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Endostatinas , Fatores de Crescimento Endotelial/antagonistas & inibidores , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Humanos , Linfocinas/antagonistas & inibidores , Modelos Biológicos , Óxido Nítrico Sintase/química , Óxido Nítrico Sintase Tipo III , Fosforilação , Fosfosserina/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
12.
Arterioscler Thromb Vasc Biol ; 22(1): 69-75, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11788463

RESUMO

Endothelial cell (EC) migration is required for angiogenesis, neovascularization, and reendothelialization. Integrins, known as alphabeta-heterodimeric cell-surface receptors, regulate cell migration and are essential for mechanotransduction of hemodynamic forces. Therefore, we investigated the effect of shear stress on EC migration and the contribution of the integrins and integrin-dependent signaling pathways in a scratched-wound assay. Laminar shear stress-induced EC migration was significantly reduced by integrin-receptor blocking with RGD peptides or with neutralizing antibodies against integrin subunits alpha(5) and beta(1), whereas antibodies against alpha(v)beta(3) or alpha(2)beta(1) had no effect. Cell-surface levels of the integrin alpha(5) and beta(1) were specifically upregulated in migrating ECs at the wound edges. Consistent with the important role of integrins for shear stress-increased cell migration, blockade of the integrin-associated adapter protein Shc by overexpression of dominant negative construct inhibited shear stress-stimulated EC migration. Moreover, pharmacological inhibition of the integrin downstream effector signaling molecules ERK1/2 or phosphatidyl-inositol-3-kinase prevented shear stress-induced EC migration. In contrast, inhibition of the NO synthase had no effect. Taken together, our results indicate that laminar shear stress enhances EC migration via the fibronectin receptor subunits alpha(5) and beta(1), which serve as central mechanotransducers in ECs. Shear stress-induced enhancement of EC migration might contribute importantly to accelerated reendothelialization of denuded arteries.


Assuntos
Movimento Celular/fisiologia , Endotélio Vascular/fisiologia , Hemorreologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores de Fibronectina/fisiologia , Receptores de Vitronectina/fisiologia , Células Cultivadas , Endotélio Vascular/citologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Fosforilação , Transdução de Sinais , Veias Umbilicais/citologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...