Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
J Cell Biol ; 104(6): 1505-13, 1987 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3584239

RESUMEN

The 20-kD regulatory light chain (RLC) plays a central role in the regulation of smooth muscle contraction. Little is known about the structure or expression of smooth muscle myosin light chain (MLC) genes. A cDNA library was constructed in the expression vector, lambda gt-11, with mRNA derived from cultured rat aortic smooth muscle cells. Using antibody generated against tracheal smooth muscle myosin, three cDNA clones encoding a RLC were isolated, one of which, SmRLC-2, represents a full-length transcript of the RLC mRNA. The derived amino acid sequence shows 94.2% homology with the chicken gizzard RLC, and 70 and 52% homology with the rat skeletal and cardiac muscle MLC-2 proteins, respectively. Thus, the gene encoding the putative smooth muscle RLC appears to have originated by duplication of the same ancestor that gave rise to the sarcomeric MLC-2 genes. Contrary to the stringent tissue-specific expression of sarcomeric MLC-2 genes, RNA blot hybridization and S1 nuclease mapping demonstrates that the putative smooth muscle RLC gene is expressed in smooth, sarcomeric, and nonmuscle tissues at significant levels. Primer extension analysis suggests that the same promoter region is used in these different tissues. Thus the putative smooth muscle RLC gene appears to be a gene that is constitutively expressed in a large variety of cells and has a differentiated function in smooth muscle.


Asunto(s)
Fibroblastos/análisis , Músculo Liso/análisis , Miofibrillas/análisis , Miosinas/genética , Fragmentos de Péptidos/genética , ARN Mensajero/genética , Sarcómeros/análisis , Animales , Línea Celular , Clonación Molecular , ADN , Genes , Miocardio/análisis , Subfragmentos de Miosina , Miosinas/biosíntesis , Fragmentos de Péptidos/biosíntesis , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Ratas , Transcripción Genética
2.
J Cell Biol ; 114(2): 303-12, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1649198

RESUMEN

The ryanodine receptor/junctional channel complex (JCC) forms the calcium release channel and foot structures of the sarcoplasmic reticulum. The JCC and the dihydropyridine (DHP) receptor in the transverse tubule are two of the major components involved in excitation-contraction (E-C) coupling in skeletal muscle. The DHP receptor is believed to serve as the voltage sensor in E-C coupling. Both the JCC and DHP receptor, as well as many skeletal muscle-specific contractile protein genes, are expressed in the BC3H1 muscle cell line. In the present study, we find that during differentiation of BC3H1 cells, induced by mitogen withdrawal, induction of the JCC and DHP receptor mRNAs is temporally similar to that of the skeletal muscle contractile protein genes alpha-tropomyosin and alpha-actin. Our data suggest that there is coordinate regulation of both the contractile protein genes (which have been studied in detail previously) and the genes encoding the calcium channels involved in E-C coupling. Induction of both calcium channels is accompanied by profound changes in BC3H1 cell morphology including the development of many components of mature skeletal muscle cells, despite lack of myoblast fusion. Visualized by electron microscopy, the JCC appears as "foot structures" located in the dyad junction between the plasmalemma and the sarcoplasmic reticulum of the BC3H1 cells. Development of foot structures is concomitant with JCC mRNA expression. Expression of the JCC and DHP receptor mRNAs and formation of the foot structures are inhibited specifically by fibroblast growth factor.


Asunto(s)
Canales de Calcio/fisiología , Sustancias de Crecimiento/farmacología , Uniones Intercelulares/fisiología , Músculos/citología , Receptores Colinérgicos/fisiología , Actinas/genética , Actinas/metabolismo , Animales , Bloqueadores de los Canales de Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Línea Celular , Factores de Crecimiento de Fibroblastos/farmacología , Uniones Intercelulares/efectos de los fármacos , Ratones , Microscopía Electrónica , Contracción Muscular/efectos de los fármacos , Músculos/metabolismo , Músculos/fisiología , Músculos/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Colinérgicos/efectos de los fármacos , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/farmacología , Canal Liberador de Calcio Receptor de Rianodina , Tropomiosina/genética , Tropomiosina/metabolismo
3.
J Cell Biol ; 111(3): 1127-35, 1990 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2391362

RESUMEN

The myosin 20,000-D regulatory light chain (RLC) has a central role in smooth muscle contraction. Previous work has suggested either the presence of two RLC isoforms, one specific for nonmuscle and one specific for smooth muscle, or the absence of a true smooth muscle-specific isoform, in which instance smooth muscle cells would use nonmuscle isoforms. To address this issue directly, we have isolated rat RLC cDNAs and corresponding genomic sequences of two smooth muscle RLC based on homology to the amino acid sequence of the chicken gizzard RLC. These cDNAs are highly homologous in their amino acid coding regions and contain unique 3'-untranslated regions. RNA analyses of rat tissue using these unique 3'-untranslated regions revealed that their expression is differentially regulated. However, one cDNA (RLC-B), predominantly a nonmuscle isoform, based on abundant expression in nonmuscle tissues including brain, spleen, and lung, is easily detected in smooth muscle tissues. The other cDNA (RLC-A; see Taubman, M., J. W. Grant, and B. Nadal-Ginard. 1987. J. Cell Biol. 104:1505-1513) was detected in a variety of nonmuscle, smooth muscle, and sarcomeric tissues. RNA analyses comparing expression of both RLC genes with the actin gene family and smooth muscle specific alpha-tropomyosin demonstrated that neither RLC gene was strictly smooth muscle specific. RNA analyses of cell lines demonstrated that both of the RLC genes are expressed in a variety of cell types. The complete genomic structure of RLC-A and close linkage to RLC-B is described.


Asunto(s)
Miosinas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/metabolismo , Línea Celular , Clonación Molecular , Regulación de la Expresión Génica , Ligamiento Genético , Biblioteca Genómica , Datos de Secuencia Molecular , Músculo Liso/metabolismo , Especificidad de Órganos , ARN Mensajero/metabolismo , Ratas , Homología de Secuencia de Ácido Nucleico , Bazo/metabolismo
4.
J Cell Biol ; 108(5): 1799-806, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2715180

RESUMEN

The BC3H1 cell line has been used widely as a model for studying regulation of muscle-related proteins, such as the acetylcholine receptor, myokinase, creatine kinase, and actin. These cells, derived from a nitrosourea-induced mouse brain neoplasm, have some of the morphological characteristics of smooth muscle and have been shown to express the vascular smooth muscle isoform of alpha-actin. To provide further information about the contractile protein phenotype of BC3H1 and to gain additional insights into the possible tissue of origin of these cells, we have examined the expression of a battery of contractile protein genes. During rapid growth, subconfluent BC3H1 cells express the nonmuscle isoform of alpha-tropomyosin (alpha-Tm) and the nonsarcomeric isoforms of myosin heavy and light chains (MHCs and MLCs, respectively), but do not express troponin T(TnT). However, when BC3H1 cells differentiate in response to incubation in serum-deprived medium or upon approaching confluence, they express TnT as well as sarcomeric muscle isoforms of MHC, MLC 2 and 3, alpha-Tm, and alpha-actin. These results suggest that BC3H1 is a skeletal muscle cell line of ectodermal origin that is defective for commitment to terminal differentiation.


Asunto(s)
Genes , Proteínas Musculares/genética , Músculos/metabolismo , Miofibrillas/metabolismo , Sarcómeros/metabolismo , Transcripción Genética , Actinas/genética , Animales , Diferenciación Celular , Línea Celular , ADN/genética , Sondas de ADN , Ratones , Músculos/citología , Miosinas/genética , Hibridación de Ácido Nucleico , Fenotipo , ARN Mensajero/genética
5.
Science ; 186(4169): 1115-7, 1974 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-4478921

RESUMEN

Rabbit antiserums were produced against the procollagen molecule secreted into the medium of cultured human skin fibroblasts. The isolated antigenic, amino terminal portion of the procollagen molecule was purified, labeled with iodine-125, and used in a radioimmunoassay which detected nanogram quantities of the same antigen. With the assay, immunologically identical molecules were detected in the culture mediumn of different strains of human fibroblasts and in normal human serums. Serumns from human cord blood contained a 12-fold higher concentration of the antigen than serums from adults, while serums other vertebrates gave reactions to incomplete cross-reactivity or non-reactivity.


Asunto(s)
Colágeno/análisis , Precursores de Proteínas/análisis , Animales , Bovinos , Línea Celular , Pollos , Colágeno/sangre , Reacciones Cruzadas , Humanos , Ratones , Precursores de Proteínas/sangre , Radioinmunoensayo , Ovinos , Especificidad de la Especie
6.
J Clin Invest ; 91(5): 2253-9, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-7683701

RESUMEN

Tissue factor (TF) is a major activator of the coagulation cascade and may play a role in initiating thrombosis after intravascular injury. To investigate whether medial vascular smooth muscle provides a source of TF following arterial injury, the induction of TF mRNA and protein was studied in balloon-injured rat aorta. After full length aortic injury, aortas were harvested at various times and the media and adventitia separated using collagenase digestion and microscopic dissection. In uninjured aortic media, TF mRNA was undetectable by RNA blot hybridization. 2 h after balloon injury TF mRNA levels increased markedly. Return to near baseline levels occurred at 24 h. In situ hybridization with a 35S-labeled antisense rat TF cRNA probe detected TF mRNA in the adventitia but not in the media or endothelium of uninjured aorta. 2 h after balloon dilatation, a marked induction of TF mRNA was observed in the adventitia and media. Using a functional clotting assay, TF procoagulant activity was detected at low levels in uninjured rat aortic media and rose by approximately 10-fold 2 h after balloon dilatation. Return to baseline occurred within 4 d. These data demonstrate that vascular injury rapidly induces active TF in arterial smooth muscle, providing a procoagulant that may result in thrombus initiation or propagation.


Asunto(s)
Aorta Torácica/metabolismo , Aorta Torácica/patología , Cateterismo/efectos adversos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , ARN Mensajero/biosíntesis , Tromboplastina/biosíntesis , Animales , Aorta Torácica/lesiones , Células Cultivadas , Sondas de ADN , Embolia/metabolismo , Embolia/patología , Biblioteca de Genes , Hibridación in Situ , Cinética , Masculino , Músculo Liso Vascular/lesiones , ARN/genética , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Tromboplastina/genética , Factores de Tiempo
7.
J Clin Invest ; 98(10): 2277-83, 1996 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8941644

RESUMEN

Abnormal vascular smooth muscle cell (SMC) proliferation and migration contribute to the development of restenosis after percutaneous transluminal coronary angioplasty and accelerated arteriopathy after cardiac transplantation. Previously, we reported that the macrolide antibiotic rapamycin, but not the related compound FK506, inhibits both human and rat aortic SMC proliferation in vitro by inhibiting cell cycle-dependent kinases and delaying phosphorylation of retinoblastoma protein (Marx, S.O., T. Jayaraman, L.O. Go, and A.R. Marks. 1995. Circ. Res. 362:801). In the present study the effects of rapamycin on SMC migration were assayed in vitro using a modified Boyden chamber and in vivo using a porcine aortic SMC explant model. Pretreatment with rapamycin (2 ng/ml) for 48 h inhibited PDGF-induced migration (PDGF BB homodimer; 20 ng/ml) in cultured rat and human SMC (n = 10; P < 0.0001), whereas FK506 had no significant effect on migration. Rapamycin administered orally (1 mg/kg per d for 7 d) significantly inhibited porcine aortic SMC migration compared with control (n = 15; P < 0.0001). Thus, in addition to being a potent immunosuppressant and antiproliferative, rapamycin also inhibits SMC migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Inmunosupresores/farmacología , Músculo Liso/fisiología , Polienos/farmacología , Administración Oral , Secuencia de Aminoácidos , Animales , Aorta/citología , Secuencia de Bases , Northern Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proteínas Portadoras/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/fisiología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/inmunología , Proteínas de Choque Térmico/fisiología , Humanos , Immunoblotting , Inmunosupresores/administración & dosificación , Datos de Secuencia Molecular , Músculo Liso/citología , Sistemas de Lectura Abierta , Factor de Crecimiento Derivado de Plaquetas/fisiología , Polienos/administración & dosificación , ARN/análisis , Ratas , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Sirolimus , Porcinos , Tacrolimus/farmacología , Proteínas de Unión a Tacrolimus
8.
J Clin Invest ; 91(2): 547-52, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8432863

RESUMEN

Tissue factor (TF) is a low molecular weight glycoprotein that initiates the clotting cascade and is considered to be a major regulator of coagulation, hemostasis, and thrombosis. TF is not expressed in the intima or media of normal adult blood vessels. Accordingly, it has been hypothesized that the initiation of intravascular coagulation may require the "induced" expression of TF in the vessel wall. We report that TF mRNA and protein are rapidly and markedly induced in early and late passaged vascular smooth muscle cells (VSMC) by growth factors (serum, platelet-derived growth factor, epidermal growth factor), vasoactive agonists (angiotensin II), and a clotting factor (alpha-thrombin). The induction of TF mRNA by these agents is dependent upon mobilization of intracellular Ca2+ and is blocked by Ca2+ chelation. In contrast to other growth factor-responsive genes, such as KC and c-fos, downregulation of protein kinase C activity by prolonged treatment with phorbol esters fails to block agonist-mediated TF induction. This raises the possibility that protein kinase C activation may not be necessary for TF mRNA induction in VSMC. VSMC may play a role in the generation or propagation of thrombus through the induction of TF, particularly in settings, such as those associated with acute vessel injury, where the endothelium is denuded and the VSMC are exposed to circulating blood.


Asunto(s)
Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Proteína Quinasa C/metabolismo , Tromboplastina/biosíntesis , Angiotensina II/farmacología , Animales , Células Cultivadas , Activación Enzimática , Masculino , Factor de Crecimiento Derivado de Plaquetas/farmacología , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Trombina/farmacología , Tromboplastina/genética
9.
J Clin Invest ; 100(9): 2276-85, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9410905

RESUMEN

Tissue factor (TF) is a transmembrane glycoprotein that initiates the coagulation cascade. Because of the potential role of TF in mediating arterial thrombosis, we have examined its expression in human aortic and coronary artery smooth muscle cells (SMC). TF mRNA and protein were induced in SMC by a variety of growth agonists. Exposure to PDGF AA or BB for 30 min provided all of the necessary signals for induction of TF mRNA and protein. This result was consistent with nuclear runoff analyses, demonstrating that PDGF-induced TF transcription occurred within 30 min. A newly developed assay involving binding of digoxigenin-labeled FVIIa (DigVIIa) and digoxigenin-labeled Factor X (DigX) was used to localize cellular TF. By light and confocal microscopy, prominent TF staining was seen in the perinuclear cytoplasm beginning 2 h after agonist treatment and persisting for 10-12 h. Surface TF activity, measured on SMC monolayers under flow conditions, increased transiently, peaking 4-6 h after agonist stimulation and returning to baseline within 16 h. Peak surface TF activity was only approximately 20% of total TF activity measured in cell lysates. Surface TF-blocking experiments demonstrated that the remaining TF was found as encrypted surface TF, and also in an intracellular pool. The relatively short-lived surface expression of TF may be critical for limiting the thrombotic potential of intact SMC exposed to growth factor stimulation. In contrast, the encrypted surface and intracellular pools may provide a rich source of TF under conditions associated with SMC damage, such as during atherosclerotic plaque rupture or balloon arterial injury.


Asunto(s)
Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Tromboplastina/metabolismo , Aorta , Compartimento Celular , Células Cultivadas , Factor VIIa/metabolismo , Factor X/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reología , Tromboplastina/genética , Transcripción Genética/efectos de los fármacos
10.
J Clin Invest ; 75(3): 1083-6, 1985 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3872314

RESUMEN

Atherosclerotic arteries have enhanced reactivity to vasoconstrictors, which suggests that features of the atherosclerotic process itself may result in this abnormal responsiveness. Since vascular smooth muscle proliferation is a prominent feature of atherosclerosis, we postulated that vasoactive agonists and smooth muscle mitogens may share certain common cellular mechanisms of action which potentially contribute to this hyperreactivity. To test this hypothesis, we studied the effects of epidermal growth factor (EGF), a well-characterized mitogen, on rat aortic vascular smooth muscle, both in intact aortic strips and in culture. EGF caused contraction (EC50 = 19 nM) of rat aortic strips which maximally was equivalent to 40% of that induced by angiotensin II, a potent vasoconstrictor. EGF increased 45Ca efflux (EC50 = 3 nM) from cultured rat aortic smooth muscle cells, which was an effect shared by angiotensin II and thought to reflect increased cytosolic-free calcium concentration. EGF (7.5 nM) also stimulated growth of these cultured cells to the same extent as 10% calf serum. These results demonstrate that EGF is both a vasoconstrictor and mitogen for rat aortic smooth muscle cells. The similarities in the effects of EGF and angiotensin II suggest that certain common intracellular mechanisms of action may exist for vasoactive agonists and growth factors which may contribute to the altered vasoreactivity of atherosclerotic vessels.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Mitógenos/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Animales , Aorta , Calcio/farmacología , Masculino , Mitosis/efectos de los fármacos , Músculo Liso Vascular/citología , Ratas , Ratas Endogámicas
11.
J Thromb Haemost ; 5(5): 942-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17461928

RESUMEN

OBJECTIVES: To evaluate the effects of pioglitazone on insulin sensitivity and levels of biomarkers associated with thrombotic risk in overweight and obese, non-diabetic subjects with coronary artery disease. BACKGROUND: Little information is available regarding the effects of thiazolidinediones in the absence of diabetes. Further, although postprandial hyperlipemia is a risk factor for cardiovascular diseases, there is limited information about the postprandial effects. METHODS: Twenty overweight and obese, non-diabetic patients with coronary artery disease were enrolled in a randomized, placebo-controlled, double-blind study. Subjects were on atorvastatin for the duration of the study and received either placebo or pioglitazone (45 mg day(-1)) for 12 weeks and then crossed over to the alternative therapy for an additional 12 weeks. Insulin sensitivity, fasting and postprandial levels of lipid, hemostatic, and inflammatory variables were measured, and endothelial function was assessed. RESULTS: Insulin sensitivity improved from 0.03 micromol kg(-1) x min pM(-1) on placebo to 0.04 on pioglitazone (P = 0.0002), and there were decreases in fasting levels of factor (F) VII:C (102 +/- 17% to 92 +/- 18%, P = 0.001), FVII:Ag (68 +/- 12% to 60 +/- 14%, P = 0.01) and in von Willebrand factor (VWF) (174 +/- 94% to 142 +/- 69%, P = 0.01). Pioglitazone lowered postprandial levels of FVII:Ag, FVII:C, plasminogen activator inhibitor-1, VWF, and triglycerides, and increased high-density lipoproteins (+9%, P = 0.02). CONCLUSIONS: Pioglitazone improves insulin sensitivity and favorably modifies fasting and postprandial lipid, hemostatic and inflammatory markers of the metabolic syndrome in overweight and obese non-diabetic patients with coronary artery disease.


Asunto(s)
Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Ayuno , Hemostasis/efectos de los fármacos , Hiperlipidemias/tratamiento farmacológico , Periodo Posprandial , Tiazolidinedionas/uso terapéutico , Adulto , Anciano , Enfermedad de la Arteria Coronaria/complicaciones , Estudios Cruzados , Método Doble Ciego , Femenino , Humanos , Hiperlipidemias/complicaciones , Insulina/sangre , Masculino , Persona de Mediana Edad , Sobrepeso , Pioglitazona , Tiazolidinedionas/farmacología
12.
Mol Cell Biol ; 19(10): 6471-8, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10490587

RESUMEN

Glucocorticoids are potent anti-inflammatory agents widely used in the treatment of human disease. We have previously shown that the inflammatory cytokine monocyte chemoattractant protein 1 (MCP-1) is regulated posttranscriptionally by glucocorticoids in arterial smooth muscle cells (SMC). To elucidate the mechanism mediating this effect, in vitro-transcribed radiolabeled MCP-1 mRNA was incubated with cytoplasmic extracts from SMC and analyzed by gel electrophoresis. Extracts from SMC treated with platelet-derived growth factor (PDGF) did not degrade the transcripts for up to 3 h. In contrast, extracts from cells treated with 1 microM dexamethasone (Dex) alone or in combination with PDGF degraded the probe with a half-life of approximately 15 min. Dex had maximal effect at concentrations above 0.01 microM and was effective on both rat and human MCP-1 transcripts. By deletion analysis, the Dex-sensitive region of the MCP-1 mRNA was localized to the initial 224 nucleotides (nt) at the 5' end and did not involve an AU-rich sequence in the 3' untranslated end. The 224-nt region conferred Dex sensitivity to heterologous mRNA. These studies provide new insights into the molecular mechanisms underlying the effect of glucocorticoids on gene expression.


Asunto(s)
Quimiocina CCL2/genética , Dexametasona/farmacología , Glucocorticoides/farmacología , Músculo Liso Vascular/metabolismo , ARN Mensajero/metabolismo , Animales , Interacciones Farmacológicas , Semivida , Humanos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Estabilidad del ARN/efectos de los fármacos , Ratas , Eliminación de Secuencia , Fracciones Subcelulares/metabolismo
13.
J Thromb Haemost ; 4(1): 158-67, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16409465

RESUMEN

Tissue factor (TF) is a transmembrane glycoprotein that initiates coagulation and plays a critical role in regulating hemostasis and thrombosis. We have recently reported a naturally occurring, soluble form of human tissue factor (asTF) generated by alternative splicing. This splice variant has a novel C-terminus with no homology to that of the full-length TF (flTF), lacks a transmembrane domain, and is active in the presence of phospholipids. Mouse models offer unique opportunities to examine the relative importance of flTF and asTF in mediating thrombosis, the response to arterial injury, and ischemic damage. To that end, we have identified and characterized murine asTF (masTF). Like the human splice variant, masTF lacks a transmembrane domain and has a unique C-terminus. We have generated antibodies specific to masTF and murine flTF (mflTF) to examine the expression of both forms of TF. masTF antigen is widely and abundantly expressed, with a pattern similar to that of mflTF, in adult tissues, in experimentally induced thrombi, and during development. These studies demonstrate that masTF contributes to the pool of total TF and may thus play an important role in mediating TF-dependent processes.


Asunto(s)
Empalme Alternativo , Tromboplastina/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso , ARN Mensajero/análisis , Solubilidad , Tromboplastina/análisis , Tromboplastina/química , Trombosis , Distribución Tisular
14.
Circulation ; 100(11): 1215-22, 1999 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-10484543

RESUMEN

BACKGROUND: The mechanisms by which lipid lowering reduces the incidence of acute thrombotic complications of coronary atheroma in clinical trials remains unknown. Tissue factor (TF) overexpressed in atheroma may accelerate thrombus formation at the sites of plaque disruption. A cell surface cytokine CD40 ligand (CD40L) enhances TF expression in vitro. METHODS AND RESULTS: To test the hypothesis that lipid lowering reduces TF expression and activity, we produced atheroma in rabbit aortas by balloon injury and cholesterol feeding for 4 months (Baseline group, n=15), followed by either a chow diet (Low group, n=10) or a continued high-cholesterol diet for 16 months (High group, n=5). Immunolocalization of TF, CD40L, and its receptor CD40 was quantified by computer-assisted color image analysis. Macrophages in atheroma of the Baseline and High groups strongly expressed TF. Intimal smooth muscle cells and endothelial cells also contained immunoreactive TF. Regions of expression of CD40L and CD40 colocalized with TF. Protein expression of TF diminished substantially in the Low group in association with reduced expression of CD40L and CD40. In situ binding of TF to factors VIIa and X, detected by digoxigenin-labeled factors VIIa and X, colocalized with TF protein in atheroma and decreased after lipid lowering. We also determined reduced TF biological activity in the Low group by use of a chromogenic assay. The level of TF mRNA detected by reverse transcription-polymerase chain reaction also decreased after lipid lowering. CONCLUSIONS: These results suggest decreased expression and activity of TF as a novel mechanism of reduced incidence of thrombotic complications of atherosclerosis by lipid lowering.


Asunto(s)
Arteriosclerosis/metabolismo , Colesterol en la Dieta/administración & dosificación , Tromboplastina/biosíntesis , Animales , Antígenos CD40/análisis , Factor VIIa/análisis , Factor X/análisis , Lípidos/sangre , Masculino , ARN Mensajero/análisis , Conejos , Tromboplastina/genética
15.
Circulation ; 102(7): 786-92, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10942748

RESUMEN

BACKGROUND: Lipoprotein(a) [Lp(a)] is a risk factor for atherosclerosis; however, the mechanisms are unclear. We previously reported that Lp(a) stimulated human vascular endothelial cells to produce monocyte chemotactic activity. The apolipoprotein(a) [apo(a)] portion of Lp(a) was the active moiety. METHODS AND RESULTS: We now describe the identification of the chemotactic activity as being due to the CC chemokine I-309. The carboxy-terminal domain of apo(a) containing 6 type-4 kringles (types 5 to 10), kringle V, and the protease domain was demonstrated to contain the I-309-inducing portion. Polyclonal and monoclonal anti-I-309 antibodies as well as an antibody against a portion of the extracellular domain of CCR8, the I-309 receptor, inhibited the increase in monocyte chemotactic activity induced by apo(a). I-309 antisense oligonucleotides also inhibited the induction of endothelial monocyte chemotactic activity by apo(a). I-309 mRNA was identified in human umbilical vein endothelial cells. Apo(a) induced an increase in I-309 protein in the endothelial cytoplasm and in the conditioned medium. Immunohistochemical studies have identified I-309 in endothelium, macrophages, and extracellular areas of human atherosclerotic plaques and have found that I-309 colocalized with apo(a). CONCLUSIONS: These data establish that I-309 is responsible for the monocyte chemotactic activity induced in human umbilical vein endothelial cells by Lp(a). The identification of the endothelial cell as a source for I-309 suggests that this chemokine may participate in vessel wall biology. Our data also suggest that I-309 may play a role in mediating the effects of Lp(a) in atherosclerosis.


Asunto(s)
Apolipoproteínas A/fisiología , Quimiocinas CC/metabolismo , Factores Quimiotácticos/metabolismo , Endotelio Vascular/metabolismo , Monocitos/metabolismo , Anticuerpos/farmacología , Apolipoproteínas A/farmacología , Western Blotting , Células Cultivadas , Quimiocina CCL1 , Quimiocinas CC/genética , Quimiocinas CC/inmunología , Factores Quimiotácticos/antagonistas & inhibidores , Factores Quimiotácticos/genética , Factores Quimiotácticos/inmunología , Medios de Cultivo/metabolismo , Citoplasma/metabolismo , Endotelio Vascular/citología , Humanos , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/metabolismo , Receptores CCR8 , Receptores de Quimiocina/inmunología , Proteínas Recombinantes/metabolismo
16.
J Am Coll Cardiol ; 15(7): 1667-87, 1990 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2188991

RESUMEN

Vascular injury represents a critical initiating event in the pathogenesis of various vascular diseases, including atherosclerosis. This review discusses 1) the current understanding and a new pathologic classification of vascular injury; 2) the resultant cellular pathophysiologic responses, specifically, lipid accumulation, platelet aggregation, thrombus formation and smooth muscle cell proliferation; 3) the role of vascular injury in the pathogenesis of spontaneous and accelerated atherosclerosis; and 4) emerging therapeutic approaches in preventing these vascular diseases. The process of type I vascular injury (nondenuding functional injury) followed by lipid accumulation, monocyte and platelet adhesion, smooth muscle cell proliferation and resultant plaque formation represents the prevalent view of the early stages of spontaneous atherogenesis. The syndromes of accelerated atherosclerosis (namely, heart transplant atherosclerosis, coronary vein graft disease and restenosis after percutaneous transluminal coronary angioplasty) appear to share etiologic mechanisms with spontaneous atherosclerosis by means of the "response to injury" hypothesis. However, type II and type III vascular injury (denuding endothelial and intimal injury with or without medial damage) followed by thrombus and its organization by smooth muscle cell proliferation and subsequent fibrosis appear to be responsible for the vascular process. This accelerated and premature occlusive process accounts for significant morbidity and mortality in patients with these conditions. Better understanding of the nature of vascular injury and its pathophysiologic responses in these clinical situations may aid in developing therapeutic strategies for preventing these vascular diseases.


Asunto(s)
Arteriosclerosis/patología , Vasos Sanguíneos/patología , Músculo Liso Vascular/patología , Animales , Arteriosclerosis/fisiopatología , Vasos Sanguíneos/fisiopatología , División Celular , Predicción , Humanos , Síndrome , Factores de Tiempo
17.
J Am Coll Cardiol ; 36(7): 2303-10, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11127477

RESUMEN

OBJECTIVES: We investigated the in vivo effects of tissue factor (TF) inhibition with recombinant tissue factor pathway inhibitor (rTFPI) on acute thrombus formation and intimal hyperplasia and the in vitro effects on smooth muscle cell migration and proliferation. BACKGROUND: Inhibition of TF with TFPI has been shown to reduce intimal hyperplasia in experimental models. However, its effects after coronary angioplasty and the cellular mechanisms involved have not been investigated. METHODS: Twenty-three swine underwent multivessel coronary angioplasty. Fifteen (n = 25 arteries) were euthanized at 72 h to assess thrombus formation and eight (n = 24 arteries) at 28 days to assess intimal hyperplasia. Animals in the 72-h time point received: 1) human rTFPI (0.5 mg bolus plus 25 microg/kg/min continuous infusion for 3 days) plus heparin (150 IU/kg intravenous bolus) plus acetyl salicylic acid (ASA) (325 mg/day); 2) rTFPI regimen plus ASA and 3) heparin (150 IU/kg intravenous bolus) plus ASA. RESULTS: On histology the control group had evidence of mural thrombus (area 0.8+/-0.4 mm2). Treatment with TFPI plus heparin abolished thrombus formation (mean area: 0.0+/-0.0 mm2, p < 0.05) but was associated with prolonged activated partial thromboplastin time and extravascular hemorrhage. Recombinant TFPI alone inhibited thrombosis without bleeding complications (mean area: 0.03+/-0.02 mm2, p < 0.05 vs. control). Animals in the 28-day time point received continuous intravenous infusion of rTFPI or control solution for 14 days. Tissue factor pathway inhibitor reduced neointimal formation with mean intimal area of 1.2+/-0.3 mm2 versus 3.2+/-0.4 mm2 in the control group; p < 0.01. Recombinant TFPI had no effect on human aortic smooth muscle cell growth but inhibited platelet-derived growth factor BB-induced migration. CONCLUSIONS: Inhibition of TF with rTFPI can prevent acute thrombosis and intimal hyperplasia after injury. Tissue factor plasma inhibitor may prove useful as an adjunct to intracoronary interventions.


Asunto(s)
Angioplastia Coronaria con Balón , Trombosis Coronaria/prevención & control , Fibrinolíticos/farmacología , Lipoproteínas/farmacología , Fragmentos de Péptidos/farmacología , Tromboplastina/antagonistas & inhibidores , Túnica Íntima/patología , Angioplastia Coronaria con Balón/efectos adversos , Animales , Células Cultivadas , Trombosis Coronaria/etiología , Vasos Coronarios/patología , Sinergismo Farmacológico , Heparina/farmacología , Hiperplasia , Modelos Animales , Porcinos
18.
Atherosclerosis ; 155(2): 371-80, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11254907

RESUMEN

Macrophages play a critical role in the development and progression of atherosclerosis. This study was designed to examine the effect of the glucocorticoid, dexamethasone, (Dex), on macrophage accumulation after acute arterial injury. Twenty New Zealand white rabbits were fed a 2% cholesterol, 6% peanut oil, rabbit chow diet for one month prior to bilateral balloon dilatation of the femoral arteries. Ten rabbits received Dex (1 mg/kg, im.) the day before and then daily for 7 days after arterial injury; control rabbits received vehicle only. Seven days after injury, Dex treatment resulted in a 96% and 77% reduction (P < 0.002) in the mean number of macrophages accumulating in the intima and media, respectively. This effect was apparently not due to a reduction in the number of circulating monocytes or to the ability of monocytes from Dex treated animals to adhere to endothelium or migrate in response to a chemotactic signal, determined in vitro under static conditions. It was associated with a 61% reduction in monocyte chemoattractant protein-1 (MCP-1) antigen (P < 0.004) in the injured arterial wall (media+intima). Glucocorticoids may be useful in attenuating the inflammatory response and subsequent foam-cell accumulation after arterial injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Arteriosclerosis/cirugía , Cateterismo/efectos adversos , Quimiotaxis/efectos de los fármacos , Colesterol en la Dieta/toxicidad , Dexametasona/uso terapéutico , Dieta Aterogénica , Arteria Femoral/lesiones , Oclusión de Injerto Vascular/prevención & control , Macrófagos/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Arteriosclerosis/inducido químicamente , Arteriosclerosis/patología , Adhesión Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Dexametasona/farmacología , Evaluación Preclínica de Medicamentos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Arteria Femoral/metabolismo , Arteria Femoral/patología , Hiperplasia , Macrófagos/patología , Masculino , Monocitos/efectos de los fármacos , Monocitos/patología , Conejos , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Heridas y Lesiones/tratamiento farmacológico
19.
Thromb Haemost ; 70(1): 180-3, 1993 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-8236099

RESUMEN

VSMC have been long thought to play a critical role in restenosis by proliferating and migrating from the vessel media to the intima, resulting in intimal hyperplasia. Recently, it has been suggested that the VSMC may play an earlier role in the events leading to restenosis, for example through the production of PDGF. The work described above suggests that the VSMC may mediate both the early inflammatory and thrombotic responses associated with vessel injury. Thus the VSMC may be involved in all phases of vascular injury, including thrombosis, inflammation, and intimal hyperplasia. Additional work will be necessary to fully elucidate the programs activated in VSMC in response to growth and migratory factors. The recent advances in recombinant DNA technology provide the hope that this will lead to novel approaches to attenuate the response of the VSMC to injury.


Asunto(s)
Angioplastia Coronaria con Balón , Arteriosclerosis/genética , Expresión Génica , Túnica Íntima/lesiones , Animales , Células Cultivadas , Enfermedad Crónica , Constricción Patológica/genética , Constricción Patológica/terapia , Hiperplasia/genética , Ratas , Túnica Íntima/metabolismo , Túnica Íntima/patología
20.
Thromb Haemost ; 82(2): 801-5, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10605785

RESUMEN

The last few years have provided increasing evidence to support a major role for TF in the initiation and propagation of thrombosis after acute arterial injury. Although thrombotic occlusion occurs in a small minority of patients undergoing acute coronary interventions or bypass surgery, mural thrombi are likely to be present in almost all cases. These thrombi may stimulate SMC and promote the development of intimal hyperplasia and luminal narrowing. The use of inhibitors of TF and factor VIIa, therefore, may not only be valuable for inhibiting thrombus formation associated with acute arterial interventions, but may also have benefit in attenuating intimal hyperplasia. Although this paper focuses on the role of TF in establishing a procoagulant state after arterial injury, the fibrinolytic system undoubtedly plays a role in balancing the effects of increased TF production in the arterial wall. This is underscored by the success of activators of fibrinolysis (tissue plasminogen activator, streptokinase, urokinase) in revascularization in the setting of acute myocardial infarction and is reviewed elsewhere. Likewise, local regulation of TFPI in the atherosclerotic plaque and injured vessel wall may be important in attenuating the effects of increased TF synthesis and accumulation. It has been assumed that the primary source of active TF after arterial injury is either SMC or invading macrophages and that active TF is anchored to the surface of these cells. Recent data have suggested that the majority of cell-associated TF is either encrypted on the cell surface or present in an intracellular pool. Arterial injury may, therefore, involve the de-encryption of surface TF or the release of intracellular TF. In addition, active vascular TF may be present in microparticles that are not anchored to the arterial wall and may be washed into the circulation. The procoagulant state may be further accentuated by the accumulation of bloodborne TF at sites of arterial injury and in developing thrombi. This TF is likely to arise from circulating leukocytes, including neutrophils and monocytes. These studies suggest that the cellular processing of TF may be an important target for inhibiting thrombotic complications associated with arterial injury and acute coronary events.


Asunto(s)
Arterias/fisiología , Arteriosclerosis/fisiopatología , Tromboplastina/fisiología , Trombosis/etiología , Animales , Arterias/lesiones , Arterias/patología , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Fibrinólisis , Humanos , Hiperplasia , Músculo Liso Vascular/fisiología , ARN Mensajero , Tromboplastina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA