Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Surg Res ; 253: 53-62, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32320897

RESUMEN

BACKGROUND: Proteoglycan 4 (PRG4; lubricin) is a member of two gene co-expression network modules associated with human vein graft failure. However, little is known about PRG4 and the vascular system. Therefore, we have investigated the effects of recombinant human PRG4 (rhPRG4) on cell migration and proliferation in human veins. METHODS: Effects of rhPRG4 on cell migration, proliferation, and neointima formation were determined in human venous tissue and cultured venous smooth muscle cells (SMCs), adventitial cells, and endothelial cells. Expression of PRG4 by cultured human saphenous veins, failed vein grafts, and varicose veins was determined by immunostaining or Western blotting. RESULTS: Limited expression of PRG4 in fresh saphenous veins was dramatically increased around medial SMCs after culture ex vivo. rhPRG4 inhibited the migration of cultured SMCs, adventitial cells, and endothelial cells, as well as the proliferation of endothelial cells. rhPRG4 also inhibited the migration of SMCs and adventitial cells from tissue explants, but there was no effect on cell proliferation or neointima formation in ex vivo whole veins. Finally, PRG4 was largely absent in two examples of venous pathology, that is, failed human vein grafts and varicose veins. CONCLUSIONS: Although rhPRG4 can inhibit the migration of venous SMCs, endothelial cells, and adventitial cells, and the proliferation of endothelial cells, PRG4 was only increased around medial SMCs in veins after ex vivo culture. PRG4 was not observed around medial SMCs in failed human vein grafts and varicose veins, suggesting the possibility that a failure of PRG4 upregulation may promote these pathologies.


Asunto(s)
Rechazo de Injerto/patología , Neointima/patología , Proteoglicanos/metabolismo , Vena Safena/trasplante , Várices/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Endoteliales/patología , Rechazo de Injerto/etiología , Humanos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/patología , Neointima/etiología , Técnicas de Cultivo de Órganos , Enfermedad Arterial Periférica/cirugía , Cultivo Primario de Células , Proteoglicanos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vena Safena/citología , Vena Safena/patología , Técnicas de Cultivo de Tejidos , Injerto Vascular/efectos adversos
2.
Eur J Vasc Endovasc Surg ; 57(2): 259-266, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30343000

RESUMEN

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.


Asunto(s)
Oclusión de Injerto Vascular/cirugía , Rechazo de Injerto/diagnóstico , Inmunoglobulina M/metabolismo , Enfermedad Arterial Periférica/cirugía , Fosforilcolina/inmunología , Injerto Vascular/métodos , Anciano , Anciano de 80 o más Años , Autoinjertos , Femenino , Oclusión de Injerto Vascular/inmunología , Rechazo de Injerto/inmunología , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Enfermedad Arterial Periférica/inmunología , Estudios Prospectivos , Vena Safena/cirugía , Resultado del Tratamiento , Grado de Desobstrucción Vascular
3.
J Vasc Surg ; 68(6S): 165S-176S.e6, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29914830

RESUMEN

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.


Asunto(s)
Isquemia/cirugía , Extremidad Inferior/irrigación sanguínea , Enfermedad Arterial Periférica/cirugía , Vena Safena/trasplante , Recolección de Tejidos y Órganos/métodos , Injerto Vascular/métodos , Anciano , Autoinjertos , Proliferación Celular , Células Cultivadas , Microambiente Celular , Técnicas de Cocultivo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/patología , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Enfermedad Arterial Periférica/genética , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/patología , Polimorfismo Genético , Estudios Prospectivos , Factores de Riesgo , Vena Safena/metabolismo , Vena Safena/patología , Fumar/efectos adversos , Fumar/metabolismo , Fumar/patología , Técnicas de Cultivo de Tejidos , Recolección de Tejidos y Órganos/efectos adversos , Injerto Vascular/efectos adversos , Grado de Desobstrucción Vascular , Remodelación Vascular , Infección de Heridas/metabolismo , Infección de Heridas/patología
4.
J Vasc Surg ; 67(5): 1556-1570.e9, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28647196

RESUMEN

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.


Asunto(s)
Movimiento Celular , Proliferación Celular , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Lesiones del Sistema Vascular/patología , Válvulas Venosas/patología , Becaplermina , Muerte Celular , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Humanos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima , Proteínas Proto-Oncogénicas c-sis/farmacología , Vena Safena/lesiones , Vena Safena/metabolismo , Vena Safena/patología , Semaforina-3A/genética , Semaforina-3A/metabolismo , Factores de Tiempo , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/metabolismo , Válvulas Venosas/efectos de los fármacos , Válvulas Venosas/lesiones , Válvulas Venosas/metabolismo
5.
J Vasc Surg ; 67(1): 309-317.e7, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526559

RESUMEN

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.


Asunto(s)
Adventicia/fisiología , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Rechazo de Injerto/genética , Miocitos del Músculo Liso/fisiología , Vena Safena/trasplante , Injerto Vascular/efectos adversos , Adventicia/citología , Anciano , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/metabolismo , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Vena Safena/citología , Túnica Íntima/citología , Túnica Íntima/fisiología
6.
J Vasc Surg ; 64(1): 202-209.e6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-25935274

RESUMEN

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.


Asunto(s)
Antígenos de Diferenciación/genética , Oclusión de Injerto Vascular/genética , Proteínas de Neoplasias/genética , Receptores Depuradores de Clase A/genética , Injerto Vascular/efectos adversos , Grado de Desobstrucción Vascular/genética , Venas/trasplante , Becaplermina , Línea Celular , Movimiento Celular , Proliferación Celular , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Oclusión de Injerto Vascular/diagnóstico , Oclusión de Injerto Vascular/metabolismo , Oclusión de Injerto Vascular/fisiopatología , Humanos , Hiperplasia , Neointima , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Proteínas Proto-Oncogénicas c-sis/farmacología , Interferencia de ARN , Factores de Riesgo , Biología de Sistemas , Transfección , Resultado del Tratamiento , Venas/efectos de los fármacos , Venas/metabolismo , Venas/fisiopatología , Cicatrización de Heridas
7.
J Vasc Surg ; 63(4): 1044-50, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25935273

RESUMEN

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.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colorantes/toxicidad , Violeta de Genciana/toxicidad , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Equipo Quirúrgico , Cicatrización de Heridas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Equipo , Humanos , Antígeno Ki-67/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/cirugía , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Técnicas de Cultivo de Órganos , Vena Safena/efectos de los fármacos , Vena Safena/metabolismo , Vena Safena/patología , Factores de Tiempo
8.
J Vasc Surg ; 57(5): 1179-85.e1-2, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23312942

RESUMEN

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.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Oclusión de Injerto Vascular/genética , Claudicación Intermitente/cirugía , Isquemia/cirugía , Extremidad Inferior/irrigación sanguínea , Enfermedad Arterial Periférica/cirugía , Polimorfismo de Nucleótido Simple , Injerto Vascular/efectos adversos , Grado de Desobstrucción Vascular/genética , Venas/trasplante , Anciano , Enfermedad Crítica , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Oclusión de Injerto Vascular/diagnóstico por imagen , Oclusión de Injerto Vascular/fisiopatología , Humanos , Claudicación Intermitente/genética , Claudicación Intermitente/fisiopatología , Isquemia/genética , Isquemia/fisiopatología , Masculino , Persona de Mediana Edad , Análisis Multivariante , Enfermedad Arterial Periférica/genética , Enfermedad Arterial Periférica/fisiopatología , Fenotipo , Regiones Promotoras Genéticas , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Ultrasonografía Doppler Dúplex , Estados Unidos , Venas/diagnóstico por imagen , Venas/fisiopatología
9.
J Vasc Surg ; 54(1): 182-191.e24, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21493032

RESUMEN

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.


Asunto(s)
Apoptosis , Derivación Arteriovenosa Quirúrgica/efectos adversos , Implantación de Prótesis Vascular/efectos adversos , Matriz Extracelular/metabolismo , Músculo Liso Vascular/cirugía , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Complicaciones Posoperatorias/etiología , Animales , Atrofia , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Ligando Fas/metabolismo , Arteria Femoral/metabolismo , Arteria Femoral/patología , Arteria Femoral/cirugía , Vena Femoral/metabolismo , Vena Femoral/patología , Vena Femoral/cirugía , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Arteria Ilíaca/metabolismo , Arteria Ilíaca/patología , Arteria Ilíaca/cirugía , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Papio , Complicaciones Posoperatorias/genética , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
10.
J Cell Physiol ; 222(1): 230-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19798690

RESUMEN

Nitric oxide triggers cGMP-dependent kinase-mediated phosphorylation of the actin regulator vasodilator-stimulated phosphoprotein (VASP) at residue serine239. The function of this phosphorylation for smooth muscle cell (SMC) adhesion, spreading, matrix contraction, and invasion is not well understood. We reconstituted VASP deficient SMC with wild-type VASP (wt-VASP) or VASP mutants that mimic "locked" serine239 phosphorylation (S239D-VASP) or "blocked" serine239 phosphorylation (S239A-VASP). Collagen gel contraction was reduced in S239D-VASP compared to S239A-VASP and wt-VASP expressing cells and nitric oxide (NO) stimulation decreased gel contraction of wt-VASP reconstituted SMC. Invasion of collagen was enhanced in S239D-VASP and NO-stimulated wild-type SMCs compared to S239A-VASP expressing cells. Expression of S239D-VASP impaired SMC attachment to collagen, reduced the number of membrane protrusions, and caused cell rounding compared to expression of S239A-VASP. Treatment of wt-VASP reconstituted SMCs with NO exerted similar effects as expression of S239D-VASP. As unstimulated cells were spreading on collagen S239A-VASP and wt-VASP localized to actin fibers whereas S239D-VASP was enriched in the cytosol. NO interferes with SMC invasion and contraction of collagen matrices. This requires phosphorylation of VASP on serine239, which reduces VASP binding to actin fibers. These findings support the conclusion that VASP phosphorylation at serine239 regulates cytoskeleton remodeling.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Colágeno/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/farmacología , Fosfoproteínas/metabolismo , Fosfoserina/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/deficiencia , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Geles , Humanos , Ratones , Proteínas de Microfilamentos/deficiencia , Proteínas Mutantes/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Fosfoproteínas/deficiencia , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
11.
Arterioscler Thromb Vasc Biol ; 29(9): 1356-62, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19592464

RESUMEN

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.


Asunto(s)
Traumatismos de las Arterias Carótidas/metabolismo , Proliferación Celular , Músculo Liso Vascular/metabolismo , Sindecano-1/metabolismo , Túnica Íntima/metabolismo , Animales , Becaplermina , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Movimiento Celular , Células Cultivadas , Replicación del ADN , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Hiperplasia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Sindecano-1/deficiencia , Sindecano-1/genética , Trombina/metabolismo , Factores de Tiempo , Túnica Íntima/patología
12.
J Histochem Cytochem ; 57(9): 889-97, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19506088

RESUMEN

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.


Asunto(s)
Proteínas ADAM/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Versicanos/metabolismo , Proteínas ADAM/genética , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/fisiopatología , Aorta Torácica/metabolismo , Atrofia , Prótesis Vascular , Muerte Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Ligando Fas/farmacología , Humanos , Arteria Ilíaca/fisiopatología , Masculino , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , Papio cynocephalus , Politetrafluoroetileno , ARN Mensajero/biosíntesis , Flujo Sanguíneo Regional , Túnica Íntima/metabolismo , Túnica Íntima/patología
13.
J Vasc Surg ; 49(5): 1282-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19307078

RESUMEN

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.


Asunto(s)
Tobillo/irrigación sanguínea , Proliferación Celular , Fibroblastos/patología , Oclusión de Injerto Vascular/etiología , Extremidad Inferior/irrigación sanguínea , Miocitos del Músculo Liso/patología , Enfermedades Vasculares Periféricas/cirugía , Vena Safena/patología , Vena Safena/trasplante , Anciano , Becaplermina , Presión Sanguínea , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Constricción Patológica , Replicación del ADN , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Oclusión de Injerto Vascular/patología , Oclusión de Injerto Vascular/fisiopatología , Heparina/farmacología , Humanos , Hiperplasia , Lisofosfolípidos/metabolismo , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Enfermedades Vasculares Periféricas/patología , Enfermedades Vasculares Periféricas/fisiopatología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-sis , Quinazolinas , Vena Safena/efectos de los fármacos , Vena Safena/fisiopatología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Trombina/metabolismo , Factores de Tiempo , Tirfostinos/farmacología , Ultrasonografía Doppler Dúplex , Grado de Desobstrucción Vascular
14.
PLoS One ; 13(9): e0204045, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30265729

RESUMEN

Changes in extracellular matrix proteins may contribute significantly to the adaptation of vein grafts to the arterial circulation. We examined the production and distribution of versican and hyaluronan in intact human vein rings cultured ex vivo, veins perfused ex vivo, and cultured venous adventitial and smooth muscle cells. Immunohistochemistry revealed higher levels of versican in the intima/media compared to the adventitia, and no differences in hyaluronan. In the vasa vasorum, versican and hyaluronan associated with CD34+ progenitor cells. Culturing the vein rings for 14 days revealed increased versican immunostaining of 30-40% in all layers, with no changes in hyaluronan. Changes in versican accumulation appear to result from increased synthesis in the intima/media and decreased degradation in the adventitia as versican transcripts were increased in the intima/media, but unchanged in the adventitia, and versikine (the ADAMTS-mediated cleavage product of versican) was increased in the intima/media, but decreased in the adventitia. In perfused human veins, versican was specifically increased in the intima/media in the presence of venous pressure, but not with arterial pressure. Unexpectedly, cultured adventitial cells express and accumulate more versican and hyaluronan than smooth muscle cells. These data demonstrate a differential regulation of versican and hyaluronan in human venous adventitia vs. intima/media and suggest distinct functions for these extracellular matrix macromolecules in these venous wall compartments during the adaptive response of vein grafts to the arterial circulation.


Asunto(s)
Venas/metabolismo , Venas/trasplante , Versicanos/metabolismo , Adventicia/metabolismo , Antígenos CD34/metabolismo , Presión Arterial/fisiología , Células Cultivadas , Humanos , Ácido Hialurónico/metabolismo , Inmunohistoquímica , Miocitos del Músculo Liso/metabolismo , Vena Safena/citología , Vena Safena/metabolismo , Células Madre/metabolismo , Técnicas de Cultivo de Tejidos , Túnica Íntima/citología , Túnica Íntima/metabolismo , Túnica Media/citología , Túnica Media/metabolismo , Vasa Vasorum/citología , Vasa Vasorum/metabolismo , Venas/citología , Versicanos/genética
15.
Circ Res ; 96(2): 172-9, 2005 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15625285

RESUMEN

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.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Aorta Abdominal , Becaplermina , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Cromonas/farmacología , Replicación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Flavonoides/farmacología , Heparina/farmacología , Humanos , Indoles/farmacología , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Maleimidas/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfolinas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-sis , ARN Interferente Pequeño/farmacología , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes/farmacología , Tirfostinos/farmacología
16.
Trends Cardiovasc Med ; 16(6): 209-15, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16839865

RESUMEN

Versican is an abundant proteoglycan in the blood vessel wall that is increased after vascular injury and accumulates in advanced atherosclerotic plaques. Versican is a large molecule with domains that mediate binding to cytokines, enzymes, lipoproteins, other extracellular matrix molecules, and signaling receptors. There is evidence that versican exists in the normal, as well as the diseased, vessel wall as discrete fragments, which represent these functional domains. We review the literature on versican degradation in vascular tissue and the function of versican domains, all of which suggest that proteolytic modification of versican may have physiologic as well as pathologic implications for the vascular system.


Asunto(s)
Enfermedades Vasculares/metabolismo , Versicanos/metabolismo , Animales , Humanos , Hiperplasia/metabolismo , Hiperplasia/patología , Túnica Íntima/metabolismo , Túnica Íntima/patología , Enfermedades Vasculares/patología
17.
Trends Cardiovasc Med ; 16(1): 25-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16387627

RESUMEN

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.


Asunto(s)
Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/efectos de los fármacos , Becaplermina , Activación Enzimática/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteoglicanos de Heparán Sulfato/fisiología , Humanos , Músculo Liso Vascular/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteínas Proto-Oncogénicas c-sis , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal , Activación Transcripcional/fisiología
18.
Circ Res ; 94(3): 340-5, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14670838

RESUMEN

Thrombin and factor Xa (FXa) are agonists for G protein-coupled receptors (GPRCs) and may contribute to vascular lesion formation by stimulating proliferation of vascular smooth muscle cells (SMCs). Mitogenic signaling of GPCRs requires transactivation of receptor tyrosine kinases (RTKs). In rat SMCs, thrombin transactivates the epidermal growth factor receptor (EGFR) via a pathway that involves heparin-binding EGF-like growth factor (HB-EGF) as ligand for EGFR. The purpose of this study was to investigate in human SMCs the role of receptor transactivation in the mitogenic response to thrombin and FXa. Thrombin (10 nmol/L) and FXa (100 nmol/L) cause a 3.3- and 2.6-fold increase in DNA synthesis, respectively. In human SMCs, neither thrombin nor FXa causes EGFR phosphorylation, and blockade of EGFR kinase does not inhibit DNA synthesis. However, DNA synthesis and phosphorylation of fibroblast growth factor receptor-1 (FGFR-1) induced by thrombin or FXa are inhibited by antibodies neutralizing basic fibroblast growth factor (bFGF) or by heparin. Hirudin inhibits thrombin-, but not FXa-induced mitogenesis, indicating that FXa acts independently of thrombin. We further demonstrate by ELISA that upon thrombin and FXa stimulation, bFGF is released and binds to the extracellular matrix. Our data suggest that in human vascular SMCs, both thrombin and FXa rapidly release bFGF into the pericellular matrix. This is followed by transactivation of the FGFR-1 and increased proliferation. Heparin may inhibit the mitogenic effects of thrombin and FXa in human SMCs by preventing bFGF binding to FGFR-1.


Asunto(s)
ADN/efectos de los fármacos , Factor Xa/farmacología , Músculo Liso Vascular/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Trombina/farmacología , Western Blotting , Células Cultivadas , ADN/biosíntesis , Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptor PAR-1/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/genética , Activación Transcripcional
19.
Cardiovasc Res ; 66(2): 402-9, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15820209

RESUMEN

OBJECTIVE: Constrictive remodeling accounts for lumen loss in postangioplasty restenosis. Matrix metalloproteinase-9 (MMP-9) has been shown to prevent constrictive remodeling in vivo. To investigate potential mechanisms for this observation, we investigated the role of MMP-9 in smooth muscle cell (SMC)-mediated collagen gel contraction, an in vitro model of constrictive remodeling. METHODS: Fischer rat SMCs were stably transfected with a construct-expressing rat-MMP-9 under the control of a tetracycline (Tet)-off promoter. SMCs were seeded in type I collagen gels (2.4 mg/ml) in the presence or not of tetracycline (1 microg/ml), and gel contraction was defined as the percentage of retraction of the collagen gel. The depletion of MMP-9 was obtained by using siRNA targeting MMP-9 mRNA or a blocking antibody. RESULTS: Gel contraction was significantly reduced at all times when MMP-9 was overexpressed (Tet-) as compared with the control condition (Tet+). However, MMP-9 depletion of control (Tet+) SMCs (using siRNA or antibody) also inhibited gel contraction. To resolve the apparent discrepancy and determine if MMP-9 exerts a dose-dependent biphasic effect on gel contraction, conditioned medium and purified rat-MMP-9 were prepared. Gel contraction was significantly increased by addition of 0.8 ng/ml of MMP-9, while high concentrations of MMP-9 (> or =100 ng/ml) inhibited contraction. The addition of BB94 and TIMP-1 did not alter the inhibitory or stimulatory effect of MMP-9. CONCLUSIONS: Our data suggest that MMP-9, independent of its proteolytic function, has a biphasic effect on SMC-mediated collagen gel contraction. Understanding the different roles of MMP-9 should allow the development of better therapeutic strategies for restenotic vascular disease.


Asunto(s)
Metaloproteinasa 9 de la Matriz/fisiología , Músculo Liso Vascular/efectos de los fármacos , Animales , Aorta , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Colágeno , Relación Dosis-Respuesta a Droga , Geles , Inhibidores de la Metaloproteinasa de la Matriz , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Ratas , Ratas Endogámicas F344 , Tiofenos/farmacología , Inhibidor Tisular de Metaloproteinasa-1/farmacología , Vasoconstricción/efectos de los fármacos
20.
J Histochem Cytochem ; 53(1): 131-40, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15637346

RESUMEN

The composition of extracellular matrix during growth and regression of the neointima was analyzed during healing in a baboon aorto-iliac polytetrafluoroethylene graft. Graft neointimal thickening can be modulated by altering blood flow by construction of downstream arteriovenous fistulas. Normal flow with normal shear stress induces neointimal thickening, whereas high flow with high shear stress upstream of a fistula induces regression of established neointima. The neointima formed under normal shear stress is enriched in hyaluronan and proteoglycans, particularly versican. On the other hand, the neointima near the graft material is enriched in collagen and biglycan. Neointimal regression in response to high shear stress is associated with a loss of proteoglycans as detected by histochemical staining. Immunostaining with an antibody against an ADAMTS cleavage neoepitope of versican increases after switching to high flow, although immunostaining for versican core protein is not appreciably changed by high flow. The present data demonstrate that the graft neointima is enriched with proteoglycans, particularly versican and hyaluronan, as well as collagen, and there is a differential distribution of each. Neointimal atrophy occurs with an apparent loss of proteoglycans and evidence of versican degradation.


Asunto(s)
Prótesis Vascular/efectos adversos , Matriz Extracelular/patología , Túnica Íntima/patología , Túnica Íntima/ultraestructura , Animales , Aorta/patología , Hiperplasia , Arteria Ilíaca/patología , Masculino , Papio , Politetrafluoroetileno , Flujo Sanguíneo Regional , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA