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.
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 ; 68(6S): 165S-176S.e6, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29914830

RESUMO

OBJECTIVE: When an autogenous vein is harvested and used for arterial bypass, it suffers physical and biologic injuries that may set in motion the cellular processes that lead to wall thickening, fibrosis, stenosis, and ultimately graft failure. Whereas the injurious effects of surgical preparation of the vein conduit have been extensively studied, little is known about the influence of the clinical environment of the donor leg from which the vein is obtained. METHODS: We studied the cellular responses of fresh saphenous vein samples obtained before implantation in 46 patients undergoing elective lower extremity bypass surgery. Using an ex vivo model of response to injury, we quantified the outgrowth of cells from explants of the adventitial and medial layers of the vein. We correlated this cellular outgrowth with the clinical characteristics of the patients, including the Wound, Ischemia, and foot Infection classification of the donor leg for ischemia, wounds, and infection as well as smoking and diabetes. RESULTS: Cellular outgrowth was significantly faster and more robust from the adventitial layer than from the medial layer. The factors of leg ischemia (P < .001), smoking (P = .042), and leg infection (P = .045) were associated with impaired overall outgrowth from the adventitial tissue on multivariable analysis. Only ischemia (P = .046) was associated with impaired outgrowth of smooth muscle cells (SMCs) from the medial tissue. Co-culture of adventitial cells and SMCs propagated from vein explants revealed that adventitial cells significantly inhibited the growth of SMCs, whereas SMCs promoted the growth of adventitial cells. The AA genotype of the -838C>A p27 polymorphism (previously associated with superior graft patency) enhanced these effects, whereas the factor of smoking attenuated adventitial cell inhibition of SMC growth. Comparing gene expression, the cells cultured from the media overexpress Kyoto Encyclopedia of Genes and Genomes pathways associated with inflammation and infection, whereas those from the adventitia overexpress gene families associated with development and stem/progenitor cell maintenance. CONCLUSIONS: The adverse clinical environment of the leg may influence the biologic behavior of the cells in the vein wall, especially the adventitial cells. Chronic ischemia was the most significant factor that retards adventitial cell outgrowth. The cells arising from the vein adventitia may be key players in determining a healthy adaptive or a pathologic response to the injuries associated with vein grafting.


Assuntos
Isquemia/cirurgia , Extremidade Inferior/irrigação sanguínea , Doença Arterial Periférica/cirurgia , Veia Safena/transplante , Coleta de Tecidos e Órgãos/métodos , Enxerto Vascular/métodos , Idoso , Autoenxertos , Proliferação de Células , Células Cultivadas , Microambiente Celular , Técnicas de Cocultura , Inibidor de Quinase Dependente de Ciclina p27/genética , Feminino , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Doença Arterial Periférica/genética , Doença Arterial Periférica/metabolismo , Doença Arterial Periférica/patologia , Polimorfismo Genético , Estudos Prospectivos , Fatores de Risco , Veia Safena/metabolismo , Veia Safena/patologia , Fumar/efeitos adversos , Fumar/metabolismo , Fumar/patologia , Técnicas de Cultura de Tecidos , Coleta de Tecidos e Órgãos/efeitos adversos , Enxerto Vascular/efeitos adversos , Grau de Desobstrução Vascular , Remodelação Vascular , Infecção dos Ferimentos/metabolismo , Infecção dos Ferimentos/patologia
3.
J Vasc Surg ; 67(5): 1556-1570.e9, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28647196

RESUMO

OBJECTIVE: Venous valves are essential but are prone to injury, thrombosis, and fibrosis. We compared the behavior and gene expression of smooth muscle cells (SMCs) in the valve sinus vs nonvalve sites to elucidate biologic differences associated with vein valves. METHODS: Tissue explants of fresh human saphenous veins were prepared, and the migration of SMCs from explants of valve sinus vs nonvalve sinus areas was measured. Proliferation and death of SMCs were determined by staining for Ki67 and terminal deoxynucleotidyl transferase dUTP nick end labeling. Proliferation and migration of passaged valve vs nonvalve SMCs were determined by cell counts and using microchemotaxis chambers. Global gene expression in valve vs nonvalve intima-media was determined by RNA sequencing. RESULTS: Valve SMCs demonstrated greater proliferation in tissue explants compared with nonvalve SMCs (19.3% ± 5.4% vs 6.8% ± 2.0% Ki67-positive nuclei at 4 days, respectively; mean ± standard error of the mean, five veins; P < .05). This was also true for migration (18.2 ± 2.7 vs 7.5 ± 3.0 migrated SMCs/explant at 6 days, respectively; 24 veins, 15 explants/vein; P < .0001). Cell death was not different (39.6% ± 16.1% vs 41.5% ± 16.0% terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells, respectively, at 4 days, five veins). Cultured valve SMCs also proliferated faster than nonvalve SMCs in response to platelet-derived growth factor subunit BB (2.9 ± 0.2-fold vs 2.1 ± 0.2-fold of control, respectively; P < .001; n = 5 pairs of cells). This was also true for migration (6.5 ± 1.2-fold vs 4.4 ± 0.8-fold of control, respectively; P < .001; n = 7 pairs of cells). Blockade of fibroblast growth factor 2 (FGF2) inhibited the increased responses of valve SMCs but had no effect on nonvalve SMCs. Exogenous FGF2 increased migration of valve but not of nonvalve SMCs. Unlike in the isolated, cultured cells, blockade of FGF2 in the tissue explants did not block migration of valve or nonvalve SMCs from the explants. Thirty-seven genes were differentially expressed by valve compared with nonvalve intimal-medial tissue (11 veins). Peptide-mediated inhibition of SEMA3A, one of the differentially expressed genes, increased the number of migrated SMCs of valve but not of nonvalve explants. CONCLUSIONS: Valve compared with nonvalve SMCs have greater rates of migration and proliferation, which may in part explain the propensity for pathologic lesion formation in valves. Whereas FGF2 mediates these effects in cultured SMCs, the mediators of these stimulatory effects in the valve wall tissue remain unclear but may be among the differentially expressed genes discovered in this study. One of these genes, SEMA3A, mediates a valve-specific inhibitory effect on the injury response of valve SMCs.


Assuntos
Movimento Celular , Proliferação de Células , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Lesões do Sistema Vascular/patologia , Válvulas Venosas/patologia , Becaplermina , Morte Celular , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neointima , Proteínas Proto-Oncogênicas c-sis/farmacologia , Veia Safena/lesões , Veia Safena/metabolismo , Veia Safena/patologia , Semaforina-3A/genética , Semaforina-3A/metabolismo , Fatores de Tempo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Válvulas Venosas/efeitos dos fármacos , Válvulas Venosas/lesões , Válvulas Venosas/metabolismo
4.
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
5.
J Vasc Surg ; 64(1): 202-209.e6, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-25935274

RESUMO

OBJECTIVE: Approximately 30% of autogenous vein grafts develop luminal narrowing and fail because of intimal hyperplasia or negative remodeling. We previously found that vein graft cells from patients who later develop stenosis proliferate more in vitro in response to growth factors than cells from patients who maintain patent grafts. To discover novel determinants of vein graft outcome, we have analyzed gene expression profiles of these cells using a systems biology approach to cluster the genes into modules by their coexpression patterns and to correlate the results with growth data from our prior study and with new studies of migration and matrix remodeling. METHODS: RNA from 4-hour serum- or platelet-derived growth factor (PDGF)-BB-stimulated human saphenous vein cells obtained from the outer vein wall (20 cell lines) was used for microarray analysis of gene expression, followed by weighted gene coexpression network analysis. Cell migration in microchemotaxis chambers in response to PDGF-BB and cell-mediated collagen gel contraction in response to serum were also determined. Gene function was determined using short-interfering RNA to inhibit gene expression before subjecting cells to growth or collagen gel contraction assays. These cells were derived from samples of the vein grafts obtained at surgery, and the long-term fate of these bypass grafts was known. RESULTS: Neither migration nor cell-mediated collagen gel contraction showed a correlation with graft outcome. Although 1188 and 1340 genes were differentially expressed in response to treatment with serum and PDGF, respectively, no single gene was differentially expressed in cells isolated from patients whose grafts stenosed compared with those that remained patent. Network analysis revealed four unique groups of genes, which we term modules, associated with PDGF responses, and 20 unique modules associated with serum responses. The "yellow" and "skyblue" modules, from PDGF and serum analyses, respectively, correlated with later graft stenosis (P = .005 and P = .02, respectively). In response to PDGF, yellow was also associated with increased cell growth. For serum, skyblue was also associated with inhibition of collagen gel contraction. The hub genes for yellow and skyblue (ie, the gene most connected to other genes in the module), scavenger receptor class A member 5 (SCARA5) and suprabasin (SBSN), respectively, were tested for effects on proliferation and collagen contraction. Knockdown of SCARA5 increased proliferation by 29.9% ± 7.8% (P < .01), whereas knockdown of SBSN had no effect. Knockdown of SBSN increased collagen gel contraction by 24.2% ± 8.6% (P < .05), whereas knockdown of SCARA5 had no effect. CONCLUSIONS: Using weighted gene coexpression network analysis of cultured vein graft cell gene expression, we have discovered two small gene modules, which comprise 42 genes, that are associated with vein graft failure. Further experiments are needed to delineate the venous cells that express these genes in vivo and the roles these genes play in vein graft healing, starting with the module hub genes SCARA5 and SBSN, which have been shown to have modest effects on cell proliferation or collagen gel contraction.


Assuntos
Antígenos de Diferenciação/genética , Oclusão de Enxerto Vascular/genética , Proteínas de Neoplasias/genética , Receptores Depuradores Classe A/genética , Enxerto Vascular/efeitos adversos , Grau de Desobstrução Vascular/genética , Veias/transplante , Becaplermina , Linhagem Celular , Movimento Celular , Proliferação de Células , Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Predisposição Genética para Doença , Oclusão de Enxerto Vascular/diagnóstico , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Hiperplasia , Neointima , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Interferência de RNA , Fatores de Risco , Biologia de Sistemas , Transfecção , Resultado do Tratamento , Veias/efeitos dos fármacos , Veias/metabolismo , Veias/fisiopatologia , Cicatrização
6.
J Vasc Surg ; 63(4): 1044-50, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25935273

RESUMO

OBJECTIVE: Markers containing dyes such as crystal violet (CAS 548-62-9) are routinely used on the adventitia of vein bypass grafts to avoid twisting during placement. Because little is known about how these dyes affect vein graft healing and function, we determined the effect of crystal violet on cell migration and proliferation, which are responses to injury after grafting. METHODS: Fresh human saphenous veins were obtained as residual specimens from leg bypass surgeries. Portions of the vein that had been surgically marked with crystal violet were analyzed separately from those that had no dye marking. In the laboratory, they were split into easily dissected inner and outer layers after removal of endothelium. This cleavage plane was within the circular muscle layer of the media. Cell migration from explants was measured daily as either (1) percentage of migration-positive explants, which exclusively measures migration, or (2) number of cells on the plastic surrounding each explant, which measures migration plus proliferation. Cell proliferation and apoptosis (Ki67 and TUNEL staining, respectively) were determined in dye-marked and unmarked areas of cultured vein rings. The dose-dependent effects of crystal violet were measured for cell migration from explants as well as for proliferation, migration, and death of cultured outer layer cells. Dye was extracted from explants with ethanol and quantified by spectrophotometry. RESULTS: There was significantly less cell migration from visibly blue compared with unstained outer layer explants by both methods. There was no significant difference in migration from inner layer explants adjacent to blue-stained or unstained sections of vein because dye did not penetrate to the inner layer. Ki67 staining of vein in organ culture, which is a measure of proliferation, progressively increased up to 6 days in nonblue outer layer and was abolished in the blue outer layer. Evidence of apoptosis (TUNEL staining) was present throughout the wall and not different in blue-stained and unstained vein wall segments. Blue outer layer explants had 65.9 ± 8.0 ng dye/explant compared with 2.1 ± 1.3 for nonblue outer layer explants. Dye applied in vitro to either outer or inner layer explants dose dependently inhibited migration (IC50∼10 ng/explant). The IC50s of crystal violet for outer layer cell proliferation and migration were 0.1 and 1.2 µg/mL, whereas the EC50 for death was between 1 and 10 µg/mL. CONCLUSIONS: Crystal violet inhibits venous cell migration and proliferation, indicating that alternative methods should be considered for marking vein grafts.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Corantes/toxicidade , Violeta Genciana/toxicidade , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Equipamentos Cirúrgicos , Cicatrização/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Relação Dose-Resposta a Droga , Desenho de Equipamento , Humanos , Antígeno Ki-67/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/cirurgia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Técnicas de Cultura de Órgãos , Veia Safena/efeitos dos fármacos , Veia Safena/metabolismo , Veia Safena/patologia , Fatores de Tempo
7.
J Vasc Surg ; 57(5): 1179-85.e1-2, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23312942

RESUMO

OBJECTIVE: Factors responsible for the variability in outcomes after lower extremity vein bypass grafting (LEVBG) are poorly understood. Recent evidence has suggested that a single nucleotide polymorphism (SNP) in the promoter region of the p27(Kip1) gene, a cell-cycle regulator, is associated with coronary in-stent restenosis. We hypothesized an association with vein graft patency. METHODS: This was a retrospective genetic association study nested within a prospective cohort of 204 patients from three referral centers undergoing LEVBG for claudication or critical ischemia. The main outcome measure was primary vein graft patency. RESULTS: All patients were followed up for a minimum of 1 year with duplex graft surveillance (median follow-up, 893 days; interquartile range, 539-1315). Genomic DNA was isolated and SNP analysis for the p27(Kip1)-838C>A variants was performed. Allele frequencies were correlated with graft outcome using survival analysis and Cox proportional hazards modeling. The p27(Kip1)-838C>A allele frequencies observed were CA, 53%; CC, 30%; and AA, 17%, satisfying Hardy-Weinberg equilibrium. Race (P = .025) and history of coronary artery disease (P = .027) were different across the genotypes; all other baseline variables were similar. Primary graft patency was greater among patients with the -838AA genotype (75% AA vs 55% CA/CC at 3 years; P = .029). In a Cox proportional hazards model including age, sex, race, diabetes, critical limb ischemia, redo (vs primary) bypass, vein type, and baseline C-reactive protein level, the p27(Kip1)-838AA genotype was significantly associated with higher graft patency (hazard ratio for failure, 0.4; 95% confidence interval, 0.17-0.93). Genotype was also associated with early (0-1 month) changes in graft lumen diameter by ultrasound imaging. CONCLUSIONS: These data suggest that the p27(Kip1)-838C>A SNP is associated with LEVBG patency and, together with previous reports, underscore a central role for p27(Kip1) in the generic response to vascular injury.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/genética , Oclusão de Enxerto Vascular/genética , Claudicação Intermitente/cirurgia , Isquemia/cirurgia , Extremidade Inferior/irrigação sanguínea , Doença Arterial Periférica/cirurgia , Polimorfismo de Nucleotídeo Único , Enxerto Vascular/efeitos adversos , Grau de Desobstrução Vascular/genética , Veias/transplante , Idoso , Estado Terminal , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Oclusão de Enxerto Vascular/diagnóstico por imagem , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Claudicação Intermitente/genética , Claudicação Intermitente/fisiopatologia , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Doença Arterial Periférica/genética , Doença Arterial Periférica/fisiopatologia , Fenótipo , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Ultrassonografia Doppler Dupla , Estados Unidos , Veias/diagnóstico por imagem , Veias/fisiopatologia
8.
PLoS One ; 7(8): e43701, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912899

RESUMO

Syndecans are receptors for soluble ligands, including heparin-binding growth factors, and matrix proteins. However, intracellular targets of syndecan-1 (Sdc-1)-mediated signaling are not fully understood. A yeast two-hybrid protein interaction screening of a mouse embryo library identified the ubiquitin and SUMO-1 E3 ligase, Topors, as a novel ligand of the Sdc-1 cytoplasmic domain (S1CD), a finding confirmed by ligand blotting and co-precipitation with Sdc-1 from cell lysates. Deletion mutagenesis identified an 18-amino acid sequence of Topors required for the interaction with the S1CD. By immunohistochemistry, Topors and Sdc-1 co-localized near the cell periphery in normal murine mammary gland (NMuMG) cells in vitro and in mouse embryonic epithelia in vivo. Finally, siRNA-mediated knockdown of Topors demonstrated that Topors is a growth promoter for murine arterial smooth muscle cells and is required for the inhibitory effect of Sdc-1 on cell growth and platelet-derived growth factor-B induction. These data suggest a novel mechanism for the inhibitory effects of Sdc-1 on cell growth that involves the interaction between the cytoplasmic domain of Sdc-1 and the SUMO-1 E3 ligase, Topors.


Assuntos
Proliferação de Células , Proteínas Proto-Oncogênicas c-sis/metabolismo , Sindecana-1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Células Cultivadas , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Células NIH 3T3 , Ligação Proteica , Proteínas Proto-Oncogênicas c-sis/genética , Interferência de RNA , Sindecana-1/genética , Trombina/farmacologia , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases/genética
9.
J Vasc Surg ; 54(1): 182-191.e24, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21493032

RESUMO

OBJECTIVE: High blood flow induces neointimal atrophy in polytetrafluoroethylene (PTFE) aortoiliac grafts and a tight external PTFE wrap of the iliac artery induces medial atrophy. In both nonhuman primate models, atrophy with loss of smooth muscle cells and extracellular matrix (ECM) begins at ≤4 days. We hypothesized that matrix loss would be linked to cell death, but the factors and mechanisms involved are not known. The purpose of this study was to determine commonly regulated genes in these two models, which we hypothesized would be a small set of genes that might be key regulators of vascular atrophy. METHODS: DNA microarray analysis (Sentrix Human Ref 8; Illumina, San Diego, Calif; ∼23,000 genes) was performed on arterial tissue from the wrap model (n = 9) and graft neointima from the graft model (n = 5) 1 day after wrapping or the switch to high flow, respectively. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) was also performed. Expression of this vascular atrophy gene set was also studied after Fas ligand-induced cell death in cultured smooth muscle cells and organ cultured arteries. RESULTS: Microarray analysis showed 15 genes were regulated in the same direction in both atrophy models: 9 upregulated and 6 downregulated. Seven of nine upregulated genes were confirmed by qRT-PCR in both models. Upregulated genes included the ECM-degrading enzymes ADAMTS4, tissue plasminogen activator (PLAT), and hyaluronidase 2; possible growth regulatory factors, including chromosome 8 open reading frame 4 and leucine-rich repeat family containing 8; a differentiation regulatory factor (musculoskeletal embryonic nuclear protein 1); a dead cell removal factor (ficolin 3); and a prostaglandin transporter (solute carrier organic anion transporter family member 2A1). Five downregulated genes were confirmed but only in one or the other model. Of the seven upregulated genes, ADAMTS4, PLAT, hyaluronidase 2, solute carrier organic anion transporter family member 2A1, leucine-rich repeat family containing 8, and chromosome 8 open reading frame 4 were also upregulated in vitro in cultured smooth muscle cells or cultured iliac artery by treatment with FasL, which causes cell death. However, blockade of caspase activity with Z-VAD inhibited FasL-mediated cell death, but not gene induction. CONCLUSION: Seven gene products were upregulated in two distinctly different in vivo nonhuman primate vascular atrophy models. Induction of cell death by FasL in vitro induced six of these genes, including the ECM-degrading factors ADAMTS4, hyaluronidase 2, and PLAT, suggesting a mechanism by which the program of tissue atrophy coordinately removes extracellular matrix as cells die. These genes may be key regulators of vascular atrophy.


Assuntos
Apoptose , Derivação Arteriovenosa Cirúrgica/efeitos adversos , Implante de Prótese Vascular/efeitos adversos , Matriz Extracelular/metabolismo , Músculo Liso Vascular/cirurgia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Complicações Pós-Operatórias/etiologia , Animais , Atrofia , Células Cultivadas , Modelos Animais de Doenças , Proteína Ligante Fas/metabolismo , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Artéria Femoral/cirurgia , Veia Femoral/metabolismo , Veia Femoral/patologia , Veia Femoral/cirurgia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Artéria Ilíaca/metabolismo , Artéria Ilíaca/patologia , Artéria Ilíaca/cirurgia , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Papio , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
10.
Arterioscler Thromb Vasc Biol ; 29(9): 1356-62, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19592464

RESUMO

OBJECTIVE: Arterial injury induces smooth muscle cell (SMC) proliferation, migration, and intimal accumulation of cells and extracellular matrix. These processes are regulated by the administration of the glycosaminoglycans heparin and heparan sulfate, but little is known about the role of endogenous heparan sulfate proteoglycans in the vessel wall. We investigated the response to carotid injury of syndecan-1-null mice to assess the function of one of a conserved family of transmembrane heparan and chondroitin sulfate proteoglycans. METHODS AND RESULTS: Syndecan-1-null mice developed a large neointimal lesion after injury, whereas wild-type mice made little or none. This was accompanied by a significant increase in both medial and intimal SMC replication. Cultured syndecan-1-null SMCs showed a significant increase in proliferation in response to PDGF-BB, thrombin, FGF2, EGF, and serum. In response to thrombin, PDGF-BB, and serum syndecan-1-null SMCs expressed more PDGF-B chain message than did wild-type SMCs. Downregulation of PDGF-BB or PDGFRbeta inhibited thrombin-, PDGF-BB-, and serum-induced DNA synthesis in syndecan-1-null SMCs. CONCLUSIONS: These results suggest the possibility that syndecan-1 may limit intimal thickening in injured arteries by suppressing SMC activation through inhibition of SMC PDGF-B chain expression and PDGFRbeta activation.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Proliferação de Células , Músculo Liso Vascular/metabolismo , Sindecana-1/metabolismo , Túnica Íntima/metabolismo , Animais , Becaplermina , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Movimento Celular , Células Cultivadas , Replicação do DNA , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hiperplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Sindecana-1/deficiência , Sindecana-1/genética , Trombina/metabolismo , Fatores de Tempo , Túnica Íntima/patologia
11.
J Histochem Cytochem ; 57(9): 889-97, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19506088

RESUMO

High blood flow through baboon polytetrafluorethylene aorto-iliac grafts increases neointimal vascular smooth muscle cell (SMC) death, neointimal atrophy, and cleavage of versican to generate the DPEAAE neoepitope, a marker of ADAMTS-mediated proteolysis. In this study, we have determined the effect of high blood flow on transcript abundance in the neointima for ADAMTS1, -4, -5, -8, -9, -15, and -20. We found that after 24 hr of flow, the mRNA for ADAMTS4 was significantly increased, whereas that for the other family members was unchanged. Because vascular SMC death is markedly increased in the graft after 24 hr of high flow, we next examined the possibility that the ADAMTS4 induction and the cell death are causally related. The addition of Fas ligand to SMC cultures increased both ADAMTS4 mRNA and cell death approximately 5-fold, consistent with the idea that ADAMTS4-dependent cleavage of versican may be partly responsible for cell death and tissue atrophy under these conditions.


Assuntos
Proteínas ADAM/biossíntese , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Versicanas/metabolismo , Proteínas ADAM/genética , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/fisiopatologia , Aorta Torácica/metabolismo , Atrofia , Prótese Vascular , Morte Celular , Células Cultivadas , Modelos Animais de Doenças , Proteína Ligante Fas/farmacologia , Humanos , Artéria Ilíaca/fisiopatologia , Masculino , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/patologia , Papio cynocephalus , Politetrafluoretileno , RNA Mensageiro/biossíntese , Fluxo Sanguíneo Regional , Túnica Íntima/metabolismo , Túnica Íntima/patologia
12.
J Vasc Surg ; 49(5): 1282-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19307078

RESUMO

OBJECTIVE: About a quarter of peripheral vein grafts fail due in part to intimal hyperplasia. The proliferative capacity and response to growth inhibitors of medial smooth muscle cells and adventitial fibroblasts in vitro were studied to test the hypothesis that intrinsic differences in cells of vein grafts are associated with graft failure. METHODS: Cells were grown from explants of the medial and adventitial layers of samples of vein grafts obtained at the time of implantation. Vein graft patency and function were monitored over the first 12 months using ankle pressures and Duplex ultrasound to determine vein graft status. Cells were obtained from veins from 11 patients whose grafts remained patent (non-stenotic) and from seven patients whose grafts developed stenosis. Smooth muscle cells (SMCs) derived from media and fibroblasts derived from adventitia were growth arrested in serum-free medium and then stimulated with 1 muM sphingosine-1-phosphate (S1P), 10 nM thrombin, 10 ng/ml epidermal growth factor (EGF), 10 ng/ml platelet-derived growth factor-BB (PDGF-BB), PDGF-BB plus S1P, or PDGF-BB plus thrombin for determination of incorporation of [(3)H]-thymidine into DNA. Cells receiving PDGF-BB or thrombin were also treated with or without 100 microg/ml heparin, which is a growth inhibitor. Cells receiving thrombin were also treated with or without 150 nM AG1478, an EGF receptor kinase inhibitor. RESULTS: SMCs and fibroblasts from veins of patients that developed stenosis responded more to the growth factors, such as PDGF-BB alone or in combination with thrombin or S1P, than cells from veins of patients that remained patent (P = .012). In addition, while PDGF-BB-mediated proliferation of fibroblasts from grafts that remained patent was inhibited by heparin (P < .03), PDGF-BB-mediated proliferation of fibroblasts from veins that developed stenosis was not (P > .5). CONCLUSION: Inherent differences in the proliferative response of vein graft cells to PDGF-BB and heparin may explain, in part, the variability among patients regarding long term patency of vein grafts.


Assuntos
Tornozelo/irrigação sanguínea , Proliferação de Células , Fibroblastos/patologia , Oclusão de Enxerto Vascular/etiologia , Extremidade Inferior/irrigação sanguínea , Miócitos de Músculo Liso/patologia , Doenças Vasculares Periféricas/cirurgia , Veia Safena/patologia , Veia Safena/transplante , Idoso , Becaplermina , Pressão Sanguínea , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Constrição Patológica , Replicação do DNA , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Oclusão de Enxerto Vascular/patologia , Oclusão de Enxerto Vascular/fisiopatologia , Heparina/farmacologia , Humanos , Hiperplasia , Lisofosfolipídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Doenças Vasculares Periféricas/patologia , Doenças Vasculares Periféricas/fisiopatologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-sis , Quinazolinas , Veia Safena/efeitos dos fármacos , Veia Safena/fisiopatologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Trombina/metabolismo , Fatores de Tempo , Tirfostinas/farmacologia , Ultrassonografia Doppler Dupla , Grau de Desobstrução Vascular
13.
J Vasc Surg ; 47(5): 1039-47, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18358668

RESUMO

OBJECTIVE: Increased blood flow causes neointimal atrophy, whereas relief of wall tension with an external wrap causes arterial medial atrophy. To study the effects of blood flow and wall tension separately and together, we applied tight or loose wraps on high-flow or normal-flow iliac arteries in baboons. METHOD: Baboon external iliac arteries were wrapped with loose-fitting and tight-fitting expanded polytetrafluoroethylene (ePTFE), leaving part unwrapped. A downstream arteriovenous fistula was constructed on one side to increase blood flow approximately twofold. The arteries were perfusion-fixed with 10% formalin after 4 (n = 5) and 28 days (n = 5). RESULTS: At 4 days, compared with the unwrapped artery, the loosely and tightly wrapped normal-flow artery showed significant medial atrophy (23% and 30%, respectively; P < .05). The tightly wrapped artery showed a loss of cells (27%; P = .02) but no change in cell density. At 28 days, the medial cross-sectional area was decreased by the tight wrap and loose wrap under normal (45% and 28%, respectively; P < .05) and high (43% and 29%, respectively; P < .05) flow. High flow did not alter the effect of wrapping nor did it affect the unwrapped medial area. At 28 days, the normal and high flow tightly wrapped media showed an insignificant loss of cells but had increased cell density (47% and 30%, respectively; P < .05), suggesting preferential loss of extracellular matrix. Decorin was expressed at the late time only in the tightly wrapped normal and high-flow media and was associated with tight packing of the collagen, as detected by picrosirius red staining. CONCLUSION: Loose-fitting and tight-fitting ePTFE wraps induced an inflammatory foreign body response that caused medial atrophy with loss of cells and extracellular matrix; the tight wrap was more effective. High blood flow did not prevent or augment medial atrophy. CLINICAL RELEVANCE: Research in arterial restenosis has focused on the biologic mechanisms and pharmacologic approaches to the prevention of intimal hyperplasia. An alternative therapeutic approach might be to induce atrophy of established intimal hyperplasia. We have previously reported that high blood flow induces neointimal regression in expanded polytetrafluoroethylene grafts in baboons. Here we provide another model of vascular atrophy induced by external wrapping. The similarity between baboons and humans in their vascular systems and individual genetic heterogeneity makes these experiments of great relevance. Up- or down-regulated genes common to both models might be key regulators of vascular atrophy and therefore suitable therapeutic targets for pharmacologic treatment of established lesions.


Assuntos
Derivação Arteriovenosa Cirúrgica , Bandagens/efeitos adversos , Migração de Corpo Estranho/etiologia , Artéria Ilíaca , Politetrafluoretileno , Animais , Atrofia , Morte Celular , Proliferação de Células , Desenho de Equipamento , Matriz Extracelular/metabolismo , Artéria Femoral/cirurgia , Veia Femoral/cirurgia , Migração de Corpo Estranho/metabolismo , Migração de Corpo Estranho/patologia , Migração de Corpo Estranho/fisiopatologia , Artéria Ilíaca/metabolismo , Artéria Ilíaca/patologia , Artéria Ilíaca/fisiopatologia , Masculino , Modelos Animais , Papio , Pressão , Fluxo Sanguíneo Regional , Estresse Mecânico , Fatores de Tempo
14.
J Vasc Surg ; 47(3): 662-70, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17950562

RESUMO

Regardless of the type of arterial reconstruction, luminal narrowing (stenosis or restenosis) develops in approximately one third of the vessels. In the past, the focus of research has been on the mechanisms of stenosis (intimal hyperplasia, pathologic remodeling) and pharmacologic approaches to prevention. An alternative approach is to induce intimal atrophy after luminal narrowing has developed, thus limiting treatment to only those patients that develop a problem. This approach to treat established disease by reducing wall mass through induction of cell death and extracellular matrix removal would be particularly useful for treating stenosis in synthetic bypass grafts or stented vessels, in which intimal hyperplasia is the primary mechanism of stenosis. This approach may be applicable as well to other vascular proliferative disorders, such as pulmonary hypertension and chronic transplant arteriopathy. Proof of principle has been shown in experiments with antibodies to platelet-derived growth factor (PDGF) receptors that cause neointimal regression in baboon polytetrafluoroethylene (PTFE) grafts and with angiotensin-converting enzyme inhibitors that induce medial atrophy in hypertensive arteries. Possible molecular targets could include PDGF receptors, A20, and BMP4. Further studies are needed to determine the utility of such a therapeutic approach to vascular disease.


Assuntos
Arteriopatias Oclusivas/tratamento farmacológico , Vasos Sanguíneos/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Proliferação de Células/efeitos dos fármacos , Doença das Coronárias/tratamento farmacológico , Animais , Arteriopatias Oclusivas/etiologia , Arteriopatias Oclusivas/metabolismo , Arteriopatias Oclusivas/patologia , Atrofia , Prótese Vascular , Implante de Prótese Vascular/efeitos adversos , Implante de Prótese Vascular/instrumentação , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Fármacos Cardiovasculares/uso terapêutico , Morte Celular/efeitos dos fármacos , Constrição Patológica , Doença das Coronárias/etiologia , Doença das Coronárias/metabolismo , Doença das Coronárias/patologia , Modelos Animais de Doenças , Transplante de Coração/efeitos adversos , Humanos , Hiperplasia , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Hipertensão/patologia , Hipertrofia , Recidiva , Indução de Remissão , Transdução de Sinais/efeitos dos fármacos , Stents
15.
Trends Cardiovasc Med ; 16(1): 25-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16387627

RESUMO

Platelet-derived growth factor (PDGF) is a major stimulant for smooth muscle cell migration and proliferation, and blockade of PDGF receptors in vivo reduces intimal growth after arterial injury. Signalling by PDGF receptors has been extensively studied and involves multiple signal transduction pathways. These include ras/Extracellular Signal Regulated Kinase (ERK), a pathway critical for controlling cell cycle progression. We have recently discovered that release of fibroblast growth factor 2 and the subsequent activation of fibroblast growth factor receptor 1 are required for maximal induction by PDGF of ERK and of human smooth muscle cell proliferation. This review summarizes our latest findings and discusses the potential implications of this novel signaling mechanism for restenosis.


Assuntos
Músculo Liso Vascular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Becaplermina , Ativação Enzimática/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/fisiologia , Proteoglicanas de Heparan Sulfato/fisiologia , Humanos , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Proto-Oncogênicas c-sis , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais , Ativação Transcricional/fisiologia
16.
J Vasc Surg ; 43(1): 150-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16414402

RESUMO

OBJECTIVE: Placement in baboons of a distal femoral arteriovenous fistula increases shear stress through aortoiliac polytetrafluoroethylene (PTFE) grafts and induces regression of a preformed neointima. Atrophy of the neointima might be controlled by shear stress-induced genes, including the bone morphogenetic proteins (BMPs). We have investigated the expression and function of BMPs 2, 4, and 5 in the graft neointima and in cultured baboon smooth muscle cells (SMCs). METHODS: Baboons received bilateral aortoiliac PTFE grafts and 8 weeks later, a unilateral femoral arteriovenous fistula. RESULTS: Quantitative polymerase chain reaction showed that high shear stress increased BMP2, 4, and 5 messenger RNA (mRNA) in graft intima between 1 and 7 days, while noggin (a BMP inhibitor) mRNA was decreased. BMP4 most potently (60% inhibition) inhibited platelet-derived growth factor-stimulated SMC proliferation compared with BMP2 and BMP5 (31% and 26%, respectively). BMP4 also increased SMC death by 190% +/- 10%. Noggin reversed the antiproliferative and proapoptotic effects of BMP4. Finally, Western blotting confirmed BMP4 protein upregulation by high shear stress at 4 days. BMP4 expression demonstrated by in situ hybridization was confined to endothelial cells. CONCLUSIONS: Increased BMPs (particularly BMP4) coupled with decreased noggin may promote high shear stress-mediated graft neointimal atrophy by inhibiting SMC proliferation and increasing SMC death.


Assuntos
Prótese Vascular , Proteínas Morfogenéticas Ósseas/fisiologia , Túnica Íntima/patologia , Animais , Atrofia/etiologia , Proteína Morfogenética Óssea 4 , Masculino , Papio , Resistência ao Cisalhamento , Estresse Mecânico
17.
Circ Res ; 96(2): 172-9, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15625285

RESUMO

We have shown that the G protein-coupled receptor (GPCR) agonists, thrombin and Factor Xa, stimulate smooth muscle cell (SMC) proliferation through transactivation of the EGF receptor (EGFR) or the FGF receptor (FGFR), both of which are tyrosine kinase receptors. In the present study, we investigated whether platelet-derived growth factor (PDGF), a tyrosine kinase receptor agonist, might transactivate another tyrosine kinase receptor to induce SMC proliferation. Because heparin inhibits PDGF-mediated proliferation in human SMCs, we investigated whether the heparin-binding growth factor basic fibroblast growth factor (bFGF) and one of its receptors, FGFR-1, play a role in the response of human arterial SMCs to PDGF-BB. PDGF-BB induced the release of bFGF and sustained phosphorylation of FGFR-1 (30 minutes to 6 hours). A bFGF-neutralizing antibody inhibited PDGF-BB-mediated phosphorylation of FGFR-1, DNA synthesis, and cell proliferation. In the presence of bFGF antibody, PDGF-BB-induced early activation of ERK (0 to 60 minutes) was not affected, whereas late ERK activation (2 to 4 hours) was reduced. When FGFR-1 expression was suppressed using small interfering RNA (siRNA), ERK activation was reduced at late, but not early, time points after PDGF-BB stimulation. Addition of bFGF antibody to cells treated with siRNA to FGFR-1 had no further effect on ERK activation. Our results provide support for a novel mechanism by which PDGF-BB induces the release of bFGF and activation of FGFR-1 followed by the sustained activation of ERK and proliferation of human SMCs.


Assuntos
Fator 2 de Crescimento de Fibroblastos/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Aorta Abdominal , Becaplermina , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Cromonas/farmacologia , Replicação do DNA/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Flavonoides/farmacologia , Heparina/farmacologia , Humanos , Indóis/farmacologia , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Maleimidas/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Morfolinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-sis , RNA Interferente Pequeno/farmacologia , Receptores Proteína Tirosina Quinases/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Proteínas Recombinantes/farmacologia , Tirfostinas/farmacologia
18.
Arterioscler Thromb Vasc Biol ; 24(5): 839-44, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15044209

RESUMO

BACKGROUND: Membrane-type matrix metalloproteinases (MT-MMPs) were initially identified as cell surface activators of MMP-2 (gelatinase A). We have reported that MT1-MMPs and MT3-MMPs are expressed by activated vascular smooth muscle cells (SMCs) and play a role in the regulation of cell function. Overexpression of MT-MMPs results in cell rounding, decreased adherence, and increased migration. Because integrin-mediated cell adhesion regulates these events, we have investigated the functional relationship between MT-MMPs and focal adhesion assembly. METHODS AND RESULTS: Using adenoviral vectors we show that overexpression of MT-MMPs reduces the number of focal contacts, whereas the cell surface expression of integrin subunits remains unchanged. The 125-kDa focal adhesion kinase (FAK) is cleaved resulting in a 90-kDa fragment under these conditions, and paxillin is partially dissociated from FAK after its cleavage. Pretreatment of cells with BB94, a synthetic MMP inhibitor, rescues cell adhesion and prevents changes in focal adhesions, supporting a potential role for MT-MMP enzymatic activities. CONCLUSIONS: This study provides the first evidence that MT-MMPs are not only important in matrix degradation but also may affect the function of focal adhesions through FAK cleavage.


Assuntos
Metaloendopeptidases/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/enzimologia , Proteínas Tirosina Quinases/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Adesão Celular , Células Cultivadas/enzimologia , Células Cultivadas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Metaloproteinases da Matriz Associadas à Membrana , Metaloendopeptidases/genética , Microscopia de Fluorescência , Peso Molecular , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Paxilina , Fosfoproteínas/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia
19.
J Vasc Surg ; 39(2): 440-6, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14743150

RESUMO

OBJECTIVE: Although current treatments for restenosis attempt to prevent the development of intimal hyperplasia, an alternative strategy is to induce intimal atrophy after restenosis has developed. Because platelet-derived growth factor (PDGF) is a smooth muscle cell growth and survival factor, we tested the hypothesis that complete blockade of PDGF by using antibodies against PDGF receptors alpha and beta would cause intimal atrophy in a baboon vascular graft model. METHODS: We administered chimeric antibodies against PDGF receptor alpha or PDGF receptor beta, either separately or together, to baboons with bilateral prosthetic aortoiliac grafts, the intimas of which had reached maximal size before treatment was begun. High blood flow, which we have previously shown to cause intimal atrophy, was induced through one graft to serve as a positive control. After 2 weeks, the intima lining the grafts was assessed for cross-sectional area, cell proliferation, and apoptosis by standard morphologic and immunohistochemical techniques. RESULTS: Blocking both PDGF receptors simultaneously reduced the cross-sectional area of the normal-flow graft intima by 44% (P <.05 vs control), whereas treatment with the individual antibodies did not significantly alter intimal area. Blockade of both receptors also inhibited smooth muscle cell proliferation by 66% (P <.05 vs control), whereas neither antibody alone altered proliferation. In contrast, all treatments increased smooth muscle cell apoptosis threefold to fivefold. CONCLUSIONS: These data suggest that simultaneous inhibition of cell proliferation and stimulation of cell death by the administration of antibodies to both PDGF receptor alpha and receptor beta is required for intimal atrophy in this baboon graft model. In addition, these data provide an in vivo model for the pharmacologic induction of intimal atrophy and introduce a novel clinical approach to treat intimal hyperplasia. Clinical relevance This study introduces the concept of pharmacologic induction of intimal atrophy. Intimal hyperplasia plagues all forms of arterial reconstruction. Currently, the only effective treatment of these restenotic lesions is balloon angioplasty or operative revision. An alternative approach to patients with clinically significant intimal hyperplasia might be to stimulate intimal regression by modulating growth and survival factors required for intimal maintenance. Although PDGF is known to be critical in intimal formation, the results of this study suggest that PDGF is also critical for intimal maintenance. Inhibition of the PDGF system may prove to be a clinically applicable approach for inducing intimal atrophy.


Assuntos
Prótese Vascular , Oclusão de Enxerto Vascular/prevenção & controle , Politetrafluoretileno , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Túnica Íntima/patologia , Animais , Atrofia , Masculino , Papio , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/imunologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/imunologia
20.
J Vasc Surg ; 36(6): 1248-55, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12469058

RESUMO

OBJECTIVE: We have previously shown that baboon grafts subjected to elevated shear stress exhibit an increase in luminal area through atrophy of the neointimal layer. This study was designed to investigate the smooth muscle cell (SMC) growth kinetics during early regression and evaluate the influence of nitric oxide (NO) in the regulation of this process. METHODS: Sixteen baboons underwent bilateral polytetrafluorethylene aortoiliac graft placement. After development of a neointima over an 8-week period, blood flow through one graft was increased with placement of a downstream arteriovenous fistula. Grafts were harvested at 4 (n = 6), 7 (n = 5), and 14 (n = 5) days and assessed for neointimal cross-sectional area, SMC proliferation and apoptosis, and macrophage infiltration. High-flow grafts were compared with contralateral normal-flow controls. Eleven baboons underwent an identical experimental preparation to evaluate the effect of NO inhibition. Eight weeks after graft implantation, the animals were treated with an initial bolus (100 mg/kg) followed by continuous infusion (60 mg/kg/d) of either N(G)-nitro-L-arginine methyl ester (L-NAME; n = 6) or the inactive stereoisomer N(G)-nitro-D-arginine methyl ester (n = 5). Grafts were harvested at 7 days and evaluated with the experimental endpoints detailed previously. RESULTS: Distal fistula placement resulted in a 3.8-fold increase in mean centerline velocity and wall shear stress. Grafts harvested during the initial 14 days after flow manipulation showed a progressive reduction in neointimal cross-sectional area. This change was associated with a decrease in subendothelial SMC proliferation and an increase in neointimal SMC apoptosis, the latter being in the region adjacent to the graft. Animals treated with L-NAME showed a 20% reduction in platelet cyclic guanosine monophosphate and a 17% reduction in serum nitrate/nitrite concentrations. Despite this inhibition of NO production, no effect on the flow-dependent changes in neointimal area, cell proliferation, or apoptosis was observed in the L-NAME-treated baboons. CONCLUSION: The local hemodynamic environment within healing prosthetic grafts modulates neointimal SMC proliferation and apoptosis. An increase in graft flow leads to atrophy of the neointima.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Implante de Prótese Vascular , Prótese Vascular/efeitos adversos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Politetrafluoretileno/efeitos adversos , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/fisiopatologia , Doenças Vasculares/fisiopatologia , Doenças Vasculares/cirurgia , Animais , Modelos Animais de Doenças , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Óxido Nítrico/análise , Óxido Nítrico/farmacologia , Papio , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA