Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Vasc Endovasc Surg ; 57(2): 259-266, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30343000

RESUMO

OBJECTIVES: One third of infrainguinal vein bypasses may fail within the first 1.5 years. Pro- and anti-inflammatory mechanisms are thought to be involved in these graft stenoses and occlusions. In previous studies, low levels of anti-phosphorylcholine IgM (anti-PC IgM, an innate anti-inflammatory IgM) have been associated with increased cardiovascular events. In this study, the peri-operative dynamics of anti-PC IgM levels were established during leg bypass surgery, and associations assessed between anti-PC IgM levels and primary graft patency. DESIGN AND METHODS: This was a prospective, observational cohort study of infrainguinal autogenous vein bypass for peripheral arterial occlusive disease involving four university affiliated hospitals. Plasma cytokine and anti-PC IgM levels were measured pre- and post-operatively. The outcome of interest was loss of primary graft patency because of occlusion or intervention for graft stenosis. RESULTS: One hundred and forty-two consecutive patients were enrolled: mean age 66 (46-91); 91% white race and male; 72.5% critical limb ischaemia (Fontaine III or IV). Median pre-operative anti-PC IgM levels were 49 units/mL (IQR 32.3-107.7, mean 89.8 + 101 sd). During follow up of an average of 1.8 years (1 month-7.4 years), 50 (35.2%) grafts lost primary patency. Pre-operative levels of interleukin 6 or C-reactive protein did not predict graft failure. Patients with pre-operative anti-PC IgM values in the lowest quartile had a twofold increased risk of graft failure (multivariable Cox proportional hazard, p = .03, HR 2.11, 95% CI 1.09-4.07), even after accounting for the other significant factors of conduit diameter, distal anastomosis, smoking, and the severity of leg ischaemia. CONCLUSIONS: Low levels of anti-PC IgM are associated with vein bypass graft failure. This biological mediator may be a useful marker to identify patients at higher risk, and offers the potential for novel, directed therapies for vascular inflammation and its consequences.


Assuntos
Oclusão de Enxerto Vascular/cirurgia , Rejeição de Enxerto/diagnóstico , Imunoglobulina M/metabolismo , Doença Arterial Periférica/cirurgia , Fosforilcolina/imunologia , Enxerto Vascular/métodos , Idoso , Idoso de 80 Anos ou mais , Autoenxertos , Feminino , Oclusão de Enxerto Vascular/imunologia , Rejeição de Enxerto/imunologia , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Doença Arterial Periférica/imunologia , Estudos Prospectivos , Veia Safena/cirurgia , Resultado do Tratamento , Grau de Desobstrução Vascular
2.
J Vasc Surg ; 67(1): 309-317.e7, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526559

RESUMO

BACKGROUND: Cyclin-dependent kinase inhibitor 1B (p27Kip1) is a cell-cycle inhibitor whose -838C>A single nucleotide polymorphism (rs36228499; hereafter called p27 SNP) has been associated with the clinical failure of peripheral vein grafts, but the functional effects of this SNP have not been demonstrated. METHODS: Human saphenous vein adventitial cells and intimal/medial smooth muscle cells (SMCs) were derived from explants obtained at the time of lower extremity bypass operations. We determined the following in adventitial cells and SMCs as a function of the p27 SNP genotype: (1) p27 promoter activity, (2) p27 messenger (m)RNA and protein levels, and (3) growth and collagen gel contraction. Deoxyribonuclease I footprinting was also performed in adventitial cells and SMCs. RESULTS: p27 promoter activity, deoxyribonuclease I footprinting, p27 mRNA levels, and p27 protein levels demonstrated that the p27 SNP is functional in adventitial cells and SMCs. Both cell types with the graft failure protective AA genotype had more p27 mRNA and protein. As predicted because of higher levels of p27 protein, adventitial cells with the AA genotype grew slower than those of the CC genotype. Unexpectedly, SMCs did not show this genotype-dependent growth response. CONCLUSIONS: These results support the functionality of the p27 SNP in venous SMCs and adventitial cells, but an effect of the SNP on cell proliferation is limited to only adventitial cells. These data point to a potential role for adventitial cells in human vein graft failure and also suggest that SMCs express factors that interfere with the activity of p27.


Assuntos
Túnica Adventícia/fisiologia , Proliferação de Células/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Rejeição de Enxerto/genética , Miócitos de Músculo Liso/fisiologia , Veia Safena/transplante , Enxerto Vascular/efeitos adversos , Túnica Adventícia/citologia , Idoso , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/metabolismo , Polimorfismo de Nucleotídeo Único , Cultura Primária de Células , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Veia Safena/citologia , Túnica Íntima/citologia , Túnica Íntima/fisiologia
3.
Drug Des Devel Ther ; 9: 5203-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26392754

RESUMO

OBJECTIVE: Thrombin has multiple proatherogenic effects including platelet activation and the induction of inflammatory processes. Recently, the cytokine oncostatin M has been shown to have proinflammatory effects. This study was designed to investigate the effects of thrombin inhibition on the initiation and progression of atherosclerosis and on the expression of oncostatin M. METHODS: Apolipoprotein E-deficient mice at different ages were fed the thrombin inhibitor dabigatran etexilate. The mean lesion area was measured in the aortic sinus and in the innominate artery. CD45-positive cells within the aortic tissue were measured by flow cytometry. Oncostatin M expression was measured in the tissue sections by immunocytochemistry. RESULTS: Treatment with dabigatran etexilate resulted in a significant reduction of the mean area of atherosclerotic lesions in the aortic sinus in both the young mice (11,176±1,500 µm(2) (control) versus 3,822±836 µm(2) (dabigatran etexilate), P<0.05) and selectively in the older mice at 28 weeks (234,099±13,500 µm(2) (control) versus 175,226±16,132 µm(2) (dabigatran etexilate), P<0.05). There were also fewer CD45-positive cells within the aortas of the dabigatran-treated mice and enhanced NO production in endothelial cells pretreated with dabigatran. In addition, the expression of oncostatin M was reduced in the lesions of dabigatran etexilate-treated mice. CONCLUSION: Inhibition of thrombin by dabigatran retards the development of early lesions and the progression of some established lesions in ApoE-/- mice. It improves endothelial function and retards macrophage accumulation within the vascular wall. Dabigatran also inhibits the expression of oncostatin M, and this suggests that oncostatin M may play a role in the initiation and progression of atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/tratamento farmacológico , Dabigatrana/farmacologia , Oncostatina M/metabolismo , Fatores Etários , Animais , Antitrombinas/farmacologia , Aterosclerose/patologia , Modelos Animais de Doenças , Progressão da Doença , Citometria de Fluxo , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Seio Aórtico/patologia
4.
Pathog Dis ; 72(1): 61-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24833344

RESUMO

Two hallmarks of advanced atherosclerosis are calcification and fibrosis. We hypothesized that Chlamydia pneumoniae infection may contribute to atherosclerosis by inducing the conversion of vascular smooth muscle cells to calcifying cells or by converting mesenchymal stem cells to osteochondrocytic or fibroblastic phenotypes. In this study, direct infection of bovine aortic smooth muscle cells (BSMCs) did not induce the expression of alkaline phosphatase or the deposition of extracellular calcium phosphate. However, conditioned media from C. pneumoniae-infected macrophages accelerated conversion of BSMCs to a calcifying phenotype. Treatment of the conditioned media with an anti-TNF-alpha blocking antibody abrogated this stimulatory effect. Treatment of perivascular Sca-1+, CD31-, CD45- cells from apoE-/- mouse aortas with the conditioned media from infected macrophages induced the Sca-1+ cells to produce collagen II, an additional marker of an osteochondrocytic phenotype. Treatment of mouse coronary perivascular Sca-1+, CD31-, CD45- cells with the supernatant from homogenates of C. pneumoniae-infected mouse lungs as compared to noninfected lungs induced expression of the Collagen 1α1 gene and deposition of collagen. Therefore, an increase in plasma cytokines or other factors in response to respiratory infection with C. pneumoniae or infection of macrophages within the blood vessel could contribute to both calcification and fibrosis of advanced atherosclerotic lesions.


Assuntos
Infecções por Chlamydia/patologia , Chlamydophila pneumoniae/fisiologia , Fibrose , Pulmão/microbiologia , Macrófagos/microbiologia , Células-Tronco Mesenquimais/patologia , Miócitos de Músculo Liso/patologia , Calcificação Vascular , Animais , Bovinos , Células Cultivadas , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Pulmão/patologia , Macrófagos/imunologia , Camundongos Endogâmicos C57BL
5.
Atherosclerosis ; 234(1): 169-75, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24657387

RESUMO

OBJECTIVE: Smooth muscle cell (SMC) de-differentiation is a key step that leads to pathological narrowing of blood vessels. De-differentiation involves a reduction in the expression of the SMC contractile genes that are the hallmark of quiescent SMCs. While there is considerable evidence linking inflammation to vascular diseases, very little is known about the mechanisms by which inflammatory signals lead to SMC de-differentiation. Given that the Signal Transducers and Activators of Transcription (STAT) transcriptional factors are the key signaling molecules activated by many inflammatory cytokines and growth factors, the aim of the present study was to determine if STAT transcriptional factors play a role SMC de-differentiation. METHODS AND RESULTS: Using shRNA targeted to STAT-1 and STAT-3, we show by real time RT-PCR and Western immunoblots that STAT-1 significantly reduces SMC contractile gene expression. In contrast, STAT-3 promotes expression of SMC contractile genes. Over-expression studies of STAT-1 and STAT-3 confirmed our observation that STAT-1 down-regulates whereas STAT-3 promotes SMC contractile gene expression. Bioinformatics analysis shows that promoters of all SMC contractile genes contain STAT binding sites. Finally, using ChIP analysis, we show that both STAT-1 and STAT-3 associate with the calponin gene. CONCLUSION: These data indicate that the balance of STAT-1 and STAT-3 influences the differentiation status of SMCs. Increased levels of STAT-1 promote SMC de-differentiation, whereas high levels of STAT-3 drive SMC into a more mature phenotype. Thus, inhibition of STAT-1 may represent a novel target for therapeutic intervention in the control of vascular diseases such as atherosclerosis and restenosis.


Assuntos
Desdiferenciação Celular , Regulação da Expressão Gênica , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/fisiologia , Células Cultivadas , Humanos , Fenótipo
6.
Biochem Biophys Res Commun ; 428(1): 179-84, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23068100

RESUMO

Increased microvessel density in atherosclerotic plaques plays a major role in promoting plaque destabilization resulting in increased risk of stroke and myocardial infarction. Previously we have shown that expression of the inflammatory cytokine, Oncostatin-M (OSM), in human atherosclerotic plaques correlated with increased microvessel density, indicating a role for OSM in promoting plaque angiogenesis. The purpose of this study was to determine the mechanism by which OSM regulates Vascular Endothelial Growth Factor (VEGF) expression in human coronary artery smooth muscle cells. Using shRNA and overexpression studies, we have shown that the transcription factor, STAT-1 inhibited VEGF expression, while STAT-3 promoted the expression of VEGF. We further show that the mechanism by which STAT-1 and STAT-3 regulates VEGF expression is through modulation of Hypoxia Inducible Factor-1α (HIF-1α). STAT-1 suppresses HIF-1α expression, whereas STAT-3 positively regulates HIF-1α expression. These results provide evidence that activated STAT-1 and STAT-3 regulate VEGF expression indirectly, by modulating HIF-1α activity.


Assuntos
Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Oncostatina M/fisiologia , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Células Cultivadas , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Oncostatina M/farmacologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT3/genética , Fator A de Crescimento do Endotélio Vascular/genética
7.
Development ; 138(20): 4451-63, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21880786

RESUMO

Fibronectin (FN) is a major component of the extracellular matrix and functions in cell adhesion, cell spreading and cell migration. In the retina, FN is transiently expressed and assembled on astrocytes (ACs), which guide sprouting tip cells and deposit a provisional matrix for sprouting angiogenesis. The precise function of FN in retinal angiogenesis is largely unknown. Using genetic tools, we show that astrocytes are the major source of cellular FN during angiogenesis in the mouse retina. Deletion of astrocytic FN reduces radial endothelial migration during vascular plexus formation in a gene dose-dependent manner. This effect correlates with reduced VEGF receptor 2 and PI3K/AKT signalling, and can be mimicked by selectively inhibiting VEGF-A binding to FN through intraocular injection of blocking peptides. By contrast, AC-specific replacement of the integrin-binding RGD sequence with FN-RGE or endothelial deletion of itga5 shows little effect on migration and PI3K/AKT signalling, but impairs filopodial alignment along AC processes, suggesting that FN-integrin α5ß1 interaction is involved in filopodial adhesion to the astrocytic matrix. AC FN shares its VEGF-binding function and cell-surface distribution with heparan-sulfate (HS), and genetic deletion of both FN and HS together greatly enhances the migration defect, indicating a synergistic function of FN and HS in VEGF binding. We propose that in vivo the VEGF-binding properties of FN and HS promote directional tip cell migration, whereas FN integrin-binding functions to support filopodia adhesion to the astrocytic migration template.


Assuntos
Astrócitos/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Animais , Movimento Celular , Matriz Extracelular/metabolismo , Fibronectinas/deficiência , Fibronectinas/genética , Heparitina Sulfato/metabolismo , Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oligopeptídeos/química , Fosfatidilinositol 3-Quinases/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasos Retinianos/inervação , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Atherosclerosis ; 216(2): 292-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21376322

RESUMO

OBJECTIVE: Chronic inflammation plays a pivotal role in the development and progression of atherosclerosis. The inflammatory response is mediated by cytokines. The aim of this study was to determine if Oncostatin M (OSM), a monocyte and T-lymphocyte specific cytokine is present in atherosclerotic lesions. We also investigated the roles of signal transducer and activator of transcription (STAT)-1 and STAT-3 in regulating OSM-induced smooth muscle cell (SMC) proliferation, migration and cellular fibronectin (cFN) synthesis. METHODS AND RESULTS: Immunostaining of atherosclerotic lesions from human carotid plaques demonstrated the expression of OSM antigen in both macrophages and SMCs. Explanted SMCs from human carotid plaques expressed OSM mRNA and protein as determined by RT-PCR and Western blotting. Using the chow-fed ApoE(-/-) mouse model of atherosclerosis, we observed that OSM was initially expressed in the intima at 20 weeks of age. By 30 weeks, OSM was expressed in both the intima and media. In vitro studies show that OSM promotes SMC proliferation, migration and cFN synthesis. Lentivirus mediated-inhibition of STAT-1 and STAT-3 prevented OSM-induced SMC proliferation, migration and cellular fibronectin synthesis. CONCLUSIONS: These findings demonstrate that OSM is expressed in atherosclerotic lesions and may contribute to the progression of atherosclerosis by promoting SMC proliferation, migration and extracellular matrix protein synthesis through the STAT pathway.


Assuntos
Aterosclerose/metabolismo , Oncostatina M/biossíntese , Oncostatina M/fisiologia , Animais , Aterosclerose/sangue , Movimento Celular , Proliferação de Células , Matriz Extracelular/metabolismo , Fibronectinas/sangue , Humanos , Imuno-Histoquímica/métodos , Inflamação , Camundongos , Camundongos Transgênicos , Miócitos de Músculo Liso/citologia , Fator de Transcrição STAT1/sangue , Fator de Transcrição STAT3/sangue , Linfócitos T/citologia
9.
J Biomed Mater Res A ; 95(2): 641-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20725965

RESUMO

Based on our discoveries of a unique, synergistic interplay between vascular endothelial growth factor (VEGF) and specific domains of the matrix protein fibronectin (FN), we used recombinant technology to create a new protein construct derived from the cell-binding and VEGF-binding domains of FN. We wished to test the hypothesis that this prototype recombinant FN (rFN) protein would enhance cellular and capillary ingrowth in vivo into expanded polytetrafluoroethylene (ePTFE) implants. ePTFE disks of high porosity (60 micron internodal distance) were embedded with fibrin gel and heparin, with/without mixtures of VEGF and rFN and were implanted subcutaneously in rats. Control implants embedded with fibrin glue and heparin alone showed an average of 8.5% (±0.51% standard error mean (SEM)) cellular ingrowth. The addition of either VEGF or rFN caused a modest but significant increase in cellular ingrowth (12.7 ± 1% and 11.8 ± 0.98%, respectively, p < 0.004). However, the combination of rFN/VEGF/heparin dramatically increased cellular ingrowth (27.6 ± 1.62%, p < 0.001), compared with all other treatments. Quantification of capillary ingrowth yielded the same pattern. These results suggest that the incorporation of such biological modulators into cardiovascular implants could offer new strategies for the design of a ready-made small diameter prosthetic graft with enhanced capacity for neovascularization and endothelialization.


Assuntos
Indutores da Angiogênese/metabolismo , Capilares/fisiologia , Fibronectinas/metabolismo , Implantes Experimentais , Neovascularização Fisiológica , Politetrafluoretileno/química , Proteínas Recombinantes/metabolismo , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Capilares/citologia , Células Cultivadas , Adesivo Tecidual de Fibrina/metabolismo , Fibronectinas/genética , Heparina/metabolismo , Humanos , Teste de Materiais , Ratos , Ratos Long-Evans , Proteínas Recombinantes/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
J Cell Biochem ; 111(2): 461-8, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20524207

RESUMO

Heparin and heparin-like molecules are known to modulate the cellular responses to vascular endothelial growth factor-A (VEGF-A). In this study, we investigated the likely mechanisms for heparin's influence on the biological activity of VEGF-A. Previous studies have shown that exogenous heparin's effects on the biological activity of VEGF-A are many and varied, in part due to the endogenous cell-surface heparan sulfates. To circumvent this problem, we used mutant endothelial cells lacking cell-surface heparan sulfates. We showed that VEGF-induced cellular responses are dependent in part on the presence of the heparan sulfates, and that exogenous heparin significantly augments VEGF's cellular effects especially when endogenous heparan sulfates are absent. Exogenous heparin was also found to play a cross-bridging role between VEGF-A(165) and putative heparin-binding sites within its cognate receptor, VEGFR2 when they were examined in isolation. The cross-bridging appears to be more dependent on molecular weight than on a specific heparin structure. This was confirmed by surface plasmon resonance binding studies using sugar chips immobilized with defined oligosaccharide structures, which showed that VEGF-A(165) binds to a relatively broad range of sulfated glycosaminoglycan structures. Finally, studies of the far-UV circular dichroism spectra of VEGF-A(165) showed that heparin can also modulate the conformation and secondary structure of the protein.


Assuntos
Células Endoteliais/metabolismo , Heparina/farmacologia , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Animais , Sítios de Ligação , Glicosaminoglicanos , Heparitina Sulfato , Humanos , Peso Molecular , Conformação Proteica/efeitos dos fármacos , Ressonância de Plasmônio de Superfície , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Future Cardiol ; 4(4): 409-26, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19804321

RESUMO

Neovacularization is an important biological process whereby new blood vessels develop in both health and disease. During development, blood vessels are formed from mesodermal cells in a process called vasculogenesis. The vascular network then expands by the sprouting of new vessel networks from pre-established vessels in a process known as angiogenesis. However, in adult life, undesirable neovascularization is associated with tumor development and a growing list of 'angiogenesis-dependent' diseases, including cardiovascular complications. Furthermore, diseases characterized by ischemia-induced tissue damage cause a neovascularization response to facilitate tissue repair. Recent research has identified novel molecular and cellular mediators of neovascularization that, in adult life, recapitulate angiogenic processes observed during embryonic development. The discovery of vascular progenitor cells and new molecules that display selective functions in modulating endothelial cell fate, migration and patterning, vessel morphogenesis and the amplification of angiogenic signaling by regulating the master signal VEGF, opens the door to new clinical strategies that target angiogenesis-dependent diseases or that can promote therapeutic neovascularization.

12.
Circ Res ; 99(8): 853-60, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-17008606

RESUMO

We describe extracellular interactions between fibronectin (Fn) and vascular endothelial growth factor (VEGF) that influence integrin-growth factor receptor crosstalk and cellular responses. In previous work, we found that VEGF bound specifically to fibronectin (Fn) but not vitronectin or collagens. Herein we report that VEGF binds to the heparin-II domain of Fn and that the cell-binding and VEGF-binding domains of Fn, when physically linked, are necessary and sufficient to promote VEGF-induced endothelial cell proliferation, migration, and Erk activation. Using recombinant Fn domains, the C-terminal heparin-II domain of Fn (type III repeats 13 to 14) was identified as a key VEGF-binding site. Mutation of the heparin-binding residues on FnIII(13-14) abolished VEGF binding, and peptides corresponding to the heparin-binding sequences in FnIII(13-14) inhibited VEGF binding to Fn. Fn fragments containing both the alpha5beta1 integrin-binding domain (III 9 to 10) and the VEGF-binding domain (III 13 to 14) significantly enhanced VEGF-induced EC migration and proliferation and induced strong phosphorylation of the VEGF receptor and Erk. Neither the cell-binding or VEGF-binding fragment of Fn alone had comparable VEGF-promoting effects. These results suggest that the mechanism of VEGF/Fn synergism is mediated extracellularly by the formation of a novel VEGF/Fn complex requiring both the cell-binding and VEGF-binding domains linked in a single molecular unit. These data also highlight a new function for the Fn C-terminal heparin-binding domain that may have important implications for angiogenesis and tumor growth.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/fisiologia , Fibronectinas/fisiologia , Heparina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Sítios de Ligação , Ligação Competitiva , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Sinergismo Farmacológico , Proteínas da Matriz Extracelular/fisiologia , Fibronectinas/genética , Fibronectinas/isolamento & purificação , Fibronectinas/metabolismo , Humanos , Peptídeos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
13.
J Biol Chem ; 281(49): 37686-96, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-16982624

RESUMO

Disintegrins are a family of potent inhibitors of cell-cell and cell-matrix adhesion. In this study we have identified a region of the disintegrin elegantin, termed the "linker domain" (amino acids 38-47), with inhibitory activity toward alpha(5)beta(1)-mediated cell adhesion on fibronectin (Fn). Using a chimeric structure-function approach in which sequences of the functionally distinct disintegrin kistrin were introduced into the elegantin template at targeted sites, a loss of inhibitory function toward alpha(5)beta(1)-mediated adhesion on Fn was observed when the elegantin linker domain was substituted. Subsequent analysis comparing the inhibitory efficacies of the panel of elegantin-kistrin chimeras toward CHO alpha(5) cell adhesion on recombinant Fn III(6-10) fragments showed that the loss of inhibitory activity associated with the disruption of the elegantin linker domain was dependent upon the presence of a functional Fn III(9) synergy site within the Fn III(6-10) substrate. This suggested that the elegantin linker domain inhibits primarily the activity of the Fn synergy domain in promoting alpha(5)beta(1) integrin-mediated cell adhesion. Construction of a cyclic peptide corresponding to the entire region of the elegantin linker domain showed that this domain has intrinsic alpha(5)beta(1) inhibitory activity comparable with the activity of the RGDS peptide. These data demonstrate a novel biological function for a disintegrin domain that antagonizes integrin-mediated cell adhesion.


Assuntos
Adesão Celular/fisiologia , Fibronectinas/metabolismo , Integrina alfa5beta1/antagonistas & inibidores , Peptídeos/química , Peptídeos/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Técnicas In Vitro , Integrina alfa5beta1/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
14.
J Biomed Mater Res B Appl Biomater ; 77(2): 389-400, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16278848

RESUMO

A polyester (PET) reinforced fibrin-FN-VEGF-TGFbeta vascular graft, formed by a four-step preclotting technique of a porous PET arterial graft, shows the overlapping inflammation, proliferation, and remodeling steps of normal wound healing when implanted in the descending thoracic aorta (DTA) position in the dog, forming a surface layer of endothelial cells. While the DTA grafts readily healed (i.e., endothelialized), similar grafts implanted in the carotid-femoral artery position did not fully heal. Since the initial phases of healing were shown to be dependent upon the transport of blood-borne constituents to the graft surface, the extent of healing appears to be dependent on the fluid dynamics present in the artery-graft-artery construct. The length of the noncompliant graft, the construction of the anastomoses, bends in the construct, graft diameter, and graft compliance can affect the fluid dynamics in the implant, and thus the healing of the graft. This has clinical relevance for the testing and development of new vascular graft materials.


Assuntos
Prótese Vascular , Hemorreologia , Engenharia Tecidual , Cicatrização , Anastomose Cirúrgica , Animais , Aorta Torácica , Artérias Carótidas , Cães , Células Endoteliais/citologia , Artéria Femoral , Poliésteres/uso terapêutico
15.
BMC Cell Biol ; 6(1): 8, 2005 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-15717924

RESUMO

BACKGROUND: The growth of new blood vessels in adult life requires the initiation of endothelial cell migration and proliferation from pre-existing vessels in addition to the recruitment and differentiation of circulating endothelial progenitor cells. Signals emanating from growth factors and the extracellular matrix are important in regulating these processes. RESULTS: Here we report that fibronectin (FN) and vitronectin (VN) modulate the responses of endothelial cells to HGF (Scatter Factor), an important pro-angiogenic mediator. Novel binding sites for HGF were identified on both FN and VN that generate molecular complexes with enhanced biological activity and these were identified in the supernatants of degranulated platelet suspensions implicating their release and formation in vivo. In the absence of co-stimulation with an ECM glycoprotein, HGF could not promote endothelial cell migration but retained the capacity to induce a proliferative response utilising the Map kinase pathway. Through promoting Met-Integrin association, HGF-FN and HGF-VN complexes coordinated and enhanced endothelial cell migration through activation of the PI-3 kinase pathway involving a Ras-dependent mechanism whereas a Ras-independent and attenuated migratory response was promoted by co-stimulation of cells with HGF and a non-binding partner ECM glycoprotein such as collagen-1. CONCLUSIONS: These studies identify a novel mechanism and pathway of HGF signalling in endothelial cells involving cooperation between Met and integrins in a Ras dependent manner. These findings have implications for the regulation of neovascularization in both health and disease.


Assuntos
Células Endoteliais/metabolismo , Fibronectinas/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Integrinas/metabolismo , Transdução de Sinais , Vitronectina/metabolismo , Sítios de Ligação , Células Endoteliais/citologia , Endotélio Vascular/citologia , Matriz Extracelular , Substâncias de Crescimento , Humanos , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas , Proteínas Proto-Oncogênicas c-met , Receptores de Fatores de Crescimento
16.
J Biomed Mater Res B Appl Biomater ; 70(2): 223-32, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15264304

RESUMO

A bigraft, composed of a 30-microm internodal distance expanded polytetrafluoroethylene (ePTFE) arterial graft segment and a preclotted polyethylene terephthalate arterial graft segment, was used to study the healing process of two different materials in the same dog. Healing was followed by FTIR-ATR spectroscopy and correlated with ELISA analyses of selected growth factors and matrix proteins. The FTIR analyses of the ePTFE explants from 1 and 3 h; 3, 5, 7, 10, and 14 days; and 4 and 8 weeks showed that the ePTFE grafts did not heal (endothelialize), but appeared to remain in overlapping inflammation and proliferation stages of wound healing. Although FN was found in the fibrin layer coating the luminal surface of the ePTFE, VEGF was not present. The inability of VEGF to complex with FN may be the result of FN binding sites being blocked or hidden by the conformation of the fibrin clot that forms on ePTFE. Also, TGF-beta was not present beyond the initial clot formation at 3 h postimplantation. The absence of these two growth factors may be an important factor in the lack of healing of 30-microm ePTFE grafts, because both were found to be present during the early stages of healing for the preclotted polyester grafts that did endothelialize.


Assuntos
Prótese Vascular , Polietilenotereftalatos , Politetrafluoretileno , Cicatrização , Animais , Sangue , Coagulação Sanguínea , Cães , Ensaio de Imunoadsorção Enzimática , Fibronectinas/metabolismo , Teste de Materiais , Espectroscopia de Infravermelho com Transformada de Fourier , Artérias Torácicas/cirurgia , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
J Vasc Surg ; 39(3): 655-60, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14981463

RESUMO

BACKGROUND: Adult endothelial progenitor cells (EPC) may be a useful source for engineering the endothelialization of vascular grafts. However, the optimal factors that promote differentiation of EPCs into endothelium remain to be elucidated. The goal of this current report was to determine which extracellular matrix (ECM) protein might modulate or enhance the effects of EPCs on differentiation into mature endothelium. METHODS: Human EPCs (CD34(+) cells) were cultured in ECM-coated six-well plates in MCDB-131 medium containing vascular endothelial growth factor (VEGF), insulin-like growth factor-1, and basic fibroblast growth factor. After 21 days, differentiated endothelial colonies were confirmed by immunofluorescence for von Willebrand factor (vWF) and vascular-endothelial (VE)-cadherin and mRNA expression of the endothelial markers Flk-1, vWF, and VE-cadherin. Cell migration toward the VEGF-matrix protein combinations was also measured. RESULTS: As judged by positive staining for endothelial markers vWF and VE-cadherin, the combination of VEGF with fibronectin (FN) produced significantly more endothelial colonies (P <.05) than did collagens I or IV or vitronectin. Defined fragments of FN did not enhance VEGF-mediated effects. Fibrinogen produced intermediate stimulation of differentiation. FN also enhanced VEGF-mediated CD34(+) cell migration. Blockade of alpha5beta1, but not alphavbeta3 or alphavbeta5, inhibited both VEGF-mediated CD34(+) cell differentiation and migration. CONCLUSIONS: VEGF and FN together significantly promote the migration and differentiation of CD34(+) cells. This synergism is specific to FN and the alpha5beta1 integrin. Combinations of VEGF and FN may be useful in promoting differentiation of circulating endothelial progenitors into endothelial cells for tissue engineering. Clinical relevance Treatment of injured or diseased tissues with adult stem cells is a promising approach. In particular, bone marrow derived circulating endothelial progenitors (CEP's) have been shown to differentiate into endothelial cells in vitro and promote tissue revascularization of ischemic limbs and myocardium in vivo. Because of the relative ease of obtaining CEP's and as well as its high proliferative rate, CEP's may have clinical potential for endothelialization of prosthetic vascular grafts and revascularization of injured myocardium. However, there is a need to better understand the molecular pathways involved in the proliferation and differentiation of CEP's to take full advantage of its clinical potential.


Assuntos
Antígenos CD34/imunologia , Diferenciação Celular/efeitos dos fármacos , Fibronectinas/farmacologia , Células-Tronco/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Adulto , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/fisiologia , Matriz Extracelular/fisiologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Integrina alfa5beta1/imunologia , Integrina alfa5beta1/fisiologia , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/fisiologia , Engenharia Tecidual/métodos
18.
Cell Commun Adhes ; 10(2): 59-67, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14681057

RESUMO

Heparin is known to influence the growth, proliferation, and migration of vascular cells, but the precise mechanisms are unknown. We previously demonstrated that unfractionated heparin (UH) binds to the platelet integrin alpha(IIb)beta(3), and enhances ligand binding. To help define the specificity and site(s) of heparin-integrin interactions, we employed the erythroleukemic K562 cell line, transfected to express specific integrins (alpha(v)beta(3), alpha(v)beta(5), and alpha(IIb)beta(3)). By comparing K562 cells expressing a common alpha subunit (Kalpha(v)beta(3), Kalpha(v)beta(5)) with cells expressing a common beta subunit (Kalpha(v)beta(3), Kalpha(IIb)beta(3)), we observed that heparin differentially modulated integrin-mediated adhesion to vitronectin. UH at 0.5-7.5 microg/ml consistently enhanced the adhesion of beta(3) expressing cells (Kalpha(v)beta(3),Kalpha(IIb)beta(3)). In contrast, UH at 0.5-7.5 microg/ml inhibited Kalpha(v)beta(5) adhesion. Experiments using integrin-blocking antibodies, appropriate control ligands, and nontransfected native K562 cells revealed that heparin's actions were mediated by the specific integrins under study. Preincubation of heparin with Kalpha(v)beta(3) cells enhanced adhesion, while preincubation of heparin with the adhesive substrate (vitronectin) had minimal effect. There was a structural specificity to heparin's effect, in that a low molecular weight heparin and chondroitin sulfate showed significantly less enhancement of adhesion. These findings suggest that heparin's modulation of integrin-ligand interactions occurs through its action on the integrin. The inhibitory or stimulatory effects of heparin depend on the beta subunit type, and the potency is dictated by structural characteristics of the glycosaminoglycan.


Assuntos
Células Sanguíneas/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Heparina/farmacologia , Cadeias alfa de Integrinas/efeitos dos fármacos , Cadeias beta de Integrinas/efeitos dos fármacos , Anticorpos/farmacologia , Anticoagulantes/farmacologia , Células Sanguíneas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sulfatos de Condroitina/farmacologia , Relação Dose-Resposta a Droga , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/metabolismo , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Peso Molecular , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transfecção , Vitronectina/metabolismo
19.
Circ Res ; 91(1): 25-31, 2002 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-12114318

RESUMO

Interactions between integrins and growth factor receptors play a critical role in the development and healing of the vasculature. This study mapped two binding domains on fibronectin (FN) that modulate the activity of the angiogenic factor, vascular endothelial growth factor (VEGF). Using solid-phase assays and surface plasmon resonance analysis, we identified two novel VEGF binding domains within the N- and C-terminus of the FN molecule. Native FN bound to VEGF enhanced endothelial cell migration and mitogen-activated protein (MAP) kinase activity, but FN that is devoid of the VEGF binding domains failed to do so. Coprecipitation studies confirmed a direct physical association between VEGF receptor-2 (Flk-1) and the FN integrin, alpha5beta1, which required intact FN because FN fragments lacking the VEGF binding domains failed to support receptor association. Thrombin-activated platelets released intact VEGF/FN complexes, which stimulated endothelial cell migration and could be inhibited by soluble high affinity VEGF receptor 1 and antibodies to alpha5beta1 integrin. This study demonstrates that FN is potentially a physiological cofactor for VEGF and provides insights into mechanisms by which growth factor receptors and integrins cooperate to influence cellular behavior.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/metabolismo , Fibronectinas/metabolismo , Linfocinas/metabolismo , Sítios de Ligação , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Fatores de Crescimento Endotelial/química , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Ativação Enzimática , Proteínas da Matriz Extracelular/farmacologia , Fibronectinas/química , Humanos , Linfocinas/química , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fragmentos de Peptídeos/farmacologia , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fibronectina/metabolismo , Receptores de Vitronectina/metabolismo , Trombina/farmacologia , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Vitronectina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...