Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
BMC Pharmacol ; 1: 7, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11686853

RESUMEN

BACKGROUND: The excitatory amino acid domoic acid, a glutamate and kainic acid analog, is the causative agent of amnesic shellfish poisoning in humans. No studies to our knowledge have investigated the potential contribution to short-term neurotoxicity of the brain microglia, a cell type that constitutes circa 10% of the total glial population in the brain. We tested the hypothesis that a short-term in vitro exposure to domoic acid, might lead to the activation of rat neonatal microglia and the concomitant release of the putative neurotoxic mediators tumor necrosis factor-alpha (TNF-alpha), matrix metalloproteinases-2 and-9 (MMP-2 and -9) and superoxide anion (O2-). RESULTS: In vitro, domoic acid [10 microM-1 mM] was significantly neurotoxic to primary cerebellar granule neurons. Although neonatal rat microglia expressed ionotropic glutamate GluR4 receptors, exposure during 6 hours to domoic acid [10 microM-1 mM] had no significant effect on viability. By four hours, LPS (10 ng/mL) stimulated an increase in TNF-alpha mRNA and a 2,233 % increase in TNF-alpha protein In contrast, domoic acid (1 mM) induced a slight rise in TNF-alpha expression and a 53 % increase (p < 0.01) of immunoreactive TNF-alpha protein. Furthermore, though less potent than LPS, a 4-hour treatment with domoic acid (1 mM) yielded a 757% (p < 0.01) increase in MMP-9 release, but had no effect on MMP-2. Finally, while PMA (phorbol 12-myristate 13-acetate) stimulated O2- generation was elevated in 6 hour LPS-primed microglia, a similar pretreatment with domoic acid (1 mM) did not prime O2- release. CONCLUSIONS: To our knowledge this is the first experimental evidence that domoic acid, at in vitro concentrations that are toxic to neuronal cells, can trigger a release of statistically significant amounts of TNF-alpha and MMP-9 by brain microglia. These observations are of considerable pathophysiological significance because domoic acid activates rat microglia several days after in vivo administration.


Asunto(s)
Ácido Kaínico/análogos & derivados , Ácido Kaínico/toxicidad , Metaloproteinasa 9 de la Matriz/metabolismo , Microglía/efectos de los fármacos , Superóxidos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antígeno CD11b/análisis , Supervivencia Celular/efectos de los fármacos , Cerebelo/efectos de los fármacos , Interacciones Farmacológicas , Técnica del Anticuerpo Fluorescente , Humanos , Lipopolisacáridos/farmacología , Toxinas Marinas/toxicidad , Espectrometría de Masas , Microglía/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/análisis , Receptores de Glutamato/análisis
2.
Am J Physiol Renal Physiol ; 281(2): F309-17, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11457723

RESUMEN

Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes that degrade the extracellular matrix (ECM). The membrane-type matrix metalloproteinases (MT-MMPs) are a new family of MMPs that differ from other MMPs in that they have a transmembrane domain that anchors them to the cell surface. MT-MMPs have been shown to function as receptors and activators for other MMPs and to localize extracellular matrix proteolysis at the pericellular region. Here we report on mRNA and protein expression of the fifth human MT-MMP (MT5-MMP), a 64-kDa protein that is capable of converting pro-MMP-2 to its active form, in human kidney as well as its upregulation in diabetes. We also demonstrate upregulation of the active form of MMP-2 in kidney samples from patients with diabetes. Through immunohistochemistry, MT5-MMP expression was localized to the epithelial cells of the proximal and distal tubules, the collecting duct, and the loop of Henle. Furthermore, the tubular epithelial cells that expressed MT5-MMP were associated with tubular atrophy. Because renal tubular atrophy is a significant factor in the pathogenesis of diabetic nephropathy and renal failure and the molecular mechanisms regulating this process remain unknown, it is hypothesized that the elevated expression of MT5-MMP contributes to the activation of pro-MMP-2, which participates in the remodeling of the proximal and distal tubules as well as in the collecting duct. These results provide the first evidence of the expression of a MT-MMP in diabetes and suggest a novel role for MT5-MMP in the pathogenesis of renal tubular atrophy and end-stage renal disease.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Riñón/enzimología , Metaloendopeptidasas/metabolismo , Adolescente , Adulto , Anciano , Diabetes Mellitus Tipo 1/patología , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Riñón/citología , Riñón/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Metaloendopeptidasas/genética , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Extractos de Tejidos/metabolismo , Regulación hacia Arriba
3.
Life Sci ; 68(7): 799-814, 2001 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-11205871

RESUMEN

Myocardial infarction (MI), leads to cardiac remodeling, thinning of the ventricle wall, ventricular dilation, and heart failure, and is a leading cause of death. Interactions between the contractile elements of the cardiac myocytes and the extracellular matrix (ECM) help maintain myocyte alignment required for the structural and functional integrity of the heart. Following MI, reorganization of the ECM and the myocytes occurs, contributing to loss of heart function. In certain pathological circumstances, the ECM is modulated such that the structure of the tissue becomes damaged. The matrix metalloproteinases (MMPs) are a family of enzymes that degrade molecules of the ECM. The present experiments were performed to define the time-course, isozyme subtypes, and cellular source of increased MMP expression that occurs following MI in an experimental rabbit model. Heart tissue samples from infarcted and sham animals were analyzed over a time-course of 1-14 days. By zymography, it was demonstrated that, unlike the sham controls, MMP-9 expression was induced within 24 hours following MI. MMP-3 expression, also absent in sham controls, was induced 2 days after MI. MMP-2 expression was detected in both the sham and infarcted samples and was modestly up-regulated following MI. Tissue inhibitor of metalloproteinase-1 (TIMP-1) expression was evaluated and shown to be down-regulated following MI, inverse of MMP-9 and MMP-3 expression. Further, MMP-9 and MMP-3 expression was detected by immunohistochemistry in myocytes within the infarct. Additional studies were conducted in which cultured rat cardiac myocytes were exposed to a hypoxic environment (2% O2) for 24 hours and the media analyzed for MMP expression. MMP-9 and MMP-3 were induced following exposure to hypoxia. It is speculated that the net increase in proteolytic activity by myocytes is a contributing factor leading to myocyte misalignment and slippage. Additional studies with a MMP inhibitor would elucidate this hypothesis.


Asunto(s)
Metaloproteinasas de la Matriz/biosíntesis , Infarto del Miocardio/enzimología , Miocardio/metabolismo , Animales , Western Blotting , Hipoxia de la Célula , Electroforesis en Gel de Poliacrilamida , Femenino , Regulación Enzimológica de la Expresión Génica , Inmunohistoquímica , Metaloproteinasa 3 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasas de la Matriz/genética , Infarto del Miocardio/patología , Isquemia Miocárdica/metabolismo , Miocardio/citología , Proteínas/química , Conejos , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
4.
Neuroreport ; 11(16): 3551-4, 2000 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-11095516

RESUMEN

Up-regulation of matrix metalloproteinases MMP-9 and MMP-2 after injury to the spinal cord (SCI) is demonstrated. MMP-9 activity maximized at 12-24 h, and MMP-2 rose at 5 days post-injury. MMP-3 was not detectable by zymographic analysis, so its level of expression was, at most, very low. The level of tissue inhibitor of metalloproteinases in the spinal cord was not altered by injury, perhaps permitting increased MMP-9 and MMP-2 activities in situ. Ablating them with an antibody demonstrated that infiltrating neutrophils were the principal source of MMP-9 activity after spinal cord injury, suggesting that neutrophils utilize that proteinase in responding to spinal cord injury. MMP-9 and MMP-2 probably contribute to breakdown of the extracellular matrix following SCI.


Asunto(s)
Metaloendopeptidasas/metabolismo , Traumatismos de la Médula Espinal/enzimología , Médula Espinal/enzimología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neutrófilos/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
5.
Am J Physiol Lung Cell Mol Physiol ; 279(5): L895-902, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11053025

RESUMEN

The effects of a second generation p38 mitogen-activated protein kinase (MAPK) inhibitor, SB 239063 [trans-1-(4-hydroxycyclohexyl)-4-(4-fluorophenyl)-5-(2-methoxypyridim idi n-4-yl)imidazole; IC(50) = 44 nM vs. p38 alpha], were assessed in models that represent different pathological aspects of chronic obstructive pulmonary disease (COPD) [airway neutrophilia, enhanced cytokine formation and increased matrix metalloproteinase (MMP)-9 activity] and in a model of lung fibrosis. Airway neutrophil infiltration and interleukin (IL)-6 levels, assessed by bronchoalveolar lavage 48 h after lipopolysaccharide (LPS) inhalation, were inhibited dose dependently by 3-30 mg/kg of SB 239063 given orally twice a day. In addition, SB 239063 (30 mg/kg orally) attenuated IL-6 bronchoalveolar lavage fluid concentrations (>90% inhibition) and MMP-9 activity (64% inhibition) assessed 6 h after LPS exposure. In guinea pig cultured alveolar macrophages, SB 239063 inhibited LPS-induced IL-6 production (IC(50) of 362 nM). In a bleomycin-induced pulmonary fibrosis model in rats, treatment with SB 239063 (2.4 or 4.8 mg/day via osmotic pump) significantly inhibited bleomycin-induced right ventricular hypertrophy (indicative of secondary pulmonary hypertension) and increases in lung hydroxyproline synthesis (indicative of collagen synthesis and fibrosis). Therefore, SB 239063 demonstrates activity against a range of sequelae commonly associated with COPD and fibrosis, supporting the therapeutic potential of p38 MAPK inhibitors such as SB 239063 in chronic airway disease.


Asunto(s)
Citocinas/biosíntesis , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Lipopolisacáridos/toxicidad , Enfermedades Pulmonares Obstructivas/fisiopatología , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neutrófilos/fisiología , Fibrosis Pulmonar/prevención & control , Pirimidinas/farmacología , Animales , Bleomicina/toxicidad , Células Cultivadas , Citocinas/sangre , Modelos Animales de Enfermedad , Cobayas , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/prevención & control , Inflamación/fisiopatología , Inflamación/prevención & control , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1/sangre , Interleucina-6/sangre , Interleucina-8/sangre , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Masculino , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neutrófilos/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/inmunología , Fibrosis Pulmonar/inducido químicamente , Ratas , Ratas Endogámicas Lew , Sialoglicoproteínas/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
6.
Cardiovasc Res ; 47(4): 759-68, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10974224

RESUMEN

OBJECTIVE: Recent studies suggest that tranilast inhibits a variety of agents implicated in neointimal growth and restenosis in experimental animal models and humans. We report here a study evaluating the efficacy of tranilast in the rat carotid artery balloon angioplasty model, a model that mimics many aspects of the percutaneous transluminal angioplasty procedure in humans. Efficacy was determined based on in vivo and ex vivo magnetic resonance imaging (MRI) as well as by histomorphometry. The utility of this study, using a reverse paradigm, is to investigate if agents successful in the clinic can demonstrate efficacy in this animal model primary screen as measured by MRI and histomorphometry. METHODS: Tranilast (300 mg/kg/day, p.o.) was administered to Sprague-Dawley rats 3 days prior to balloon injury and continued for 14 days after injury. Three methods of measuring the vascular injury that occurs in this model were employed: (1) in vivo MRI, used to measure in vivo lumen volumes for the carotid artery once at baseline (pre-surgery) and again at 14 days post angioplasty; (2) ex vivo MRI (and histomorphometry), used to evaluate the total arterial wall thickness and the intima-to-media ratio; and (3) analysis of collagen density, used to evaluate the efficacy of tranilast to abrogate collagen synthesis and deposition following vascular injury. RESULTS: Tranilast provided 33% protection (P<0.05) from angioplasty-induced lumen narrowing as measured by MRI in vivo. The results of the ex vivo MR analysis of total wall thickness showed a 14% protection of angioplasty-induced narrowing (P<0.05), and the mean intima-to-media ratio showed a 39% (P<0.006) protection for the tranilast-treated rats. Histological analysis of the mean intima-to-media ratio demonstrated that tranilast provided 36% (P<0. 01) protection in the intima-to-media ratio. Further, treatment with tranilast showed a 52% reduction in collagen density of the intimal layer and a 70% reduction in collagen density of the medial layer of the injured arteries. CONCLUSION: The data obtained by in vivo MRI, ex vivo MRI, histology and collagen analysis demonstrate that tranilast provided significant beneficial effects in inhibiting neointimal formation in the rat carotid artery model. Also this study, to the best of our knowledge, is the first to harness complimentary information from various technologies, including lumen patency by in vivo MRI, neointimal formation by ex vivo MRI and conventional histomorphometry, and histological analysis for collagen density, to provide a comprehensive understanding of the pathology in this disease model.


Asunto(s)
Angioplastia Coronaria con Balón , Antialérgicos/uso terapéutico , Enfermedad Coronaria/terapia , Imagen por Resonancia Magnética , Túnica Íntima/anatomía & histología , ortoaminobenzoatos/uso terapéutico , Análisis de Varianza , Animales , Arterias Carótidas , Cateterismo , Colágeno/análisis , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Recurrencia , Túnica Íntima/efectos de los fármacos , Túnica Íntima/metabolismo
7.
Biochem Biophys Res Commun ; 271(1): 138-43, 2000 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-10777693

RESUMEN

Interleukin-1beta (IL-1beta) is a pleiotropic cytokine capable of inducing smooth muscle activation and leukocyte recruitment in restenosis and atherosclerosis. Our present study investigated the expression of IL-1beta, IL-1 receptor antagonist (IL-1ra), and IL-1 receptor (IL-1RI and IL-1RII) mRNA in carotid artery after balloon angioplasty using semiquantitative reverse transcription/polymerase chain reaction (RT/PCR) and Northern analysis. Time course studies revealed that IL-1beta mRNA was rapidly induced at 6 h (30-fold increase over control, P < 0.001) post balloon injury and diminished to basal levels at 24 h. The increased expression of IL-1ra mRNA was delayed, reaching a peak at 24 h (400-fold increase, P < 0.001) and sustained up to 14 days. The expression of IL-1RII mRNA was remarkably similar to that of IL-1beta, whereas the IL-1RI (the signaling receptor) mRNA expression was delayed (significantly induced at day 1; 14.2-fold increase, P < 0.01) post balloon injury. Immunohistochemical studies revealed a strong induction of IL-1beta in the area with actively proliferating and migrating smooth muscle cells (i.e., in the inner medial layer at day 1 and in neointima at day 14 after balloon injury). The differential but concomitant expression of IL-1beta, IL-1ra, IL-1RI, and IL-1RII mRNAs after balloon angioplasty suggests that each of these IL-1 system components may play a distinct role in neointima formation.


Asunto(s)
Angioplastia de Balón , Arterias Carótidas/metabolismo , Interleucina-1/biosíntesis , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/biosíntesis , Animales , Northern Blotting , Inmunohistoquímica , Interleucina-1/genética , Cinética , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Regulación hacia Arriba
8.
Nature ; 401(6750): 282-6, 1999 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-10499587

RESUMEN

Urotensin-II (U-II) is a vasoactive 'somatostatin-like' cyclic peptide which was originally isolated from fish spinal cords, and which has recently been cloned from man. Here we describe the identification of an orphan human G-protein-coupled receptor homologous to rat GPR14 and expressed predominantly in cardiovascular tissue, which functions as a U-II receptor. Goby and human U-II bind to recombinant human GPR14 with high affinity, and the binding is functionally coupled to calcium mobilization. Human U-II is found within both vascular and cardiac tissue (including coronary atheroma) and effectively constricts isolated arteries from non-human primates. The potency of vasoconstriction of U-II is an order of magnitude greater than that of endothelin-1, making human U-II the most potent mammalian vasoconstrictor identified so far. In vivo, human U-II markedly increases total peripheral resistance in anaesthetized non-human primates, a response associated with profound cardiac contractile dysfunction. Furthermore, as U-II immunoreactivity is also found within central nervous system and endocrine tissues, it may have additional activities.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/metabolismo , Receptores de Superficie Celular/agonistas , Receptores Acoplados a Proteínas G , Urotensinas/farmacología , Vasoconstrictores/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcio/metabolismo , Línea Celular , Clonación Molecular , ADN Complementario , Proteínas de Unión al GTP/genética , Humanos , Macaca fascicularis , Masculino , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Homología de Secuencia de Aminoácido , Distribución Tisular , Urotensinas/metabolismo , Vasoconstrictores/metabolismo
9.
Shock ; 11(3): 180-6, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10188770

RESUMEN

The effects of lipopolysaccharide (LPS) on the central nervous system, one of the first organs to be affected by sepsis, are still incompletely understood. Rat microglia (BMphi) constitute the main leukocyte-dependent source of reactive oxygen species in the central nervous system. The in vitro effect of LPS on agonist-stimulated superoxide (O2-) generation from BMphi appears controversial. Our purpose was to determine the time- and concentration-dependent effect of Escherichia coil LPS on phorbol-12 myristate 13-acetate-stimulated O2- generation from BMphi. Our results demonstrate that BMphi O2- generation in vitro peaked 17 h after stimulation of with .3 ng/mL LPS. Furthermore, stimulation of BMphi with LPS for 17 h resulted in the following concentration-dependent responses: .1-1 ng/mL LPS induced no prior mediator generation but potently enhanced subsequent phorbol-12 myristate 13-acetate-stimulated O2- generation; 3-10 ng/mL LPS caused nitric oxide, tumor necrosis factor-alpha (TNF-alpha), thromboxane B2 and matrix metalloproteinase-9 release although partially inhibiting ensuing phorbol-12 myristate 13-acetate-stimulated O2- generation; 30-100 ng/mL LPS, maximized nitric oxide, TNF-alpha, thromboxane B2, matrix metalloproteinase-9 generation with concomitant lactic dehydrogenase release although strongly deactivating successive phorbol-12 myristate 13-acetate-stimulated O2 production. Our in vitro studies suggest that enhanced release of these four mediators (nitric oxide, TNF-alpha, thromboxane B2, and matrix metalloproteinase-9) during stimulation of BMphi with LPS might play a critical role in the subsequent ability of BMphi to generate O2- in vivo. Potential clinical implications of our findings are suggested by the fact that LPS levels similar to the ones used in this study have been observed in cerebrospinal fluid both in Gram-negative meningitis and sepsis.


Asunto(s)
Escherichia coli/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lipopolisacáridos/farmacología , Microglía/metabolismo , Superóxidos/metabolismo , Animales , Animales Recién Nacidos , Aniones/metabolismo , Colagenasas/efectos de los fármacos , Colagenasas/metabolismo , Relación Dosis-Respuesta a Droga , L-Lactato Deshidrogenasa/efectos de los fármacos , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/efectos de los fármacos , Metaloendopeptidasas/metabolismo , Microglía/efectos de los fármacos , Microglía/microbiología , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacología , Tromboxano B2/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Biol Chem ; 274(3): 1479-86, 1999 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9880523

RESUMEN

TL1 is a recently discovered novel member of the tumor necrosis factor (TNF) cytokine family. TL1 is abundantly expressed in endothelial cells, but its function is not known. The present study was undertaken to explore whether TL1 induces apoptosis in endothelial cells and, if so, to explore its mechanism of action. Cultured bovine pulmonary artery endothelial cells (BPAEC) exposed to TL1 showed morphological (including ultrastructural) and biochemical features characteristic of apoptosis. TL1-induced apoptosis in BPAEC was a time- and concentration-dependent process (EC50 = 72 ng/ml). The effect of TL1 was not inhibited by soluble TNF receptors 1 or 2. TL1 up-regulated Fas expression in BPAEC at 8 and 24 h after treatment, and significantly activated stress-activated protein kinase (SAPK) and p38 mitogen-activated protein kinase (p38 MAPK). The peak activities of SAPK and p38 MAPK in TL1-treated BPAEC were increased by 9- and 4-fold, respectively. TL1-induced apoptosis in the BPAEC was reduced by expression of a dominant-interfering mutant of c-Jun (62.8%, p < 0.05) or by a specific p38 inhibitor, SB203580 (1-10 microM) dose-dependently. TL1 also activated caspases in BPAEC, and TL1-induced apoptosis in BPAEC was significantly attenuated by the caspase inhibitor, ZVAD-fluromethyl-ketone. The major component activated by TL1 in BPAEC was caspase-3, which was based on substrate specificity and immunocytochemical analysis. These findings suggest that TL1 may act as an autocrine factor to induce apoptosis in endothelial cells via activation of multiple signaling pathways, including stress protein kinases as well as certain caspases.


Asunto(s)
Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Caspasas/metabolismo , Endotelio Vascular/fisiología , Proteínas Quinasas Activadas por Mitógenos , Receptores del Factor de Necrosis Tumoral/fisiología , Animales , Antígenos CD/metabolismo , Caspasa 3 , Bovinos , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Arteria Pulmonar , Piridinas/farmacología , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral , Regulación hacia Arriba , Receptor fas/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos
11.
Circ Res ; 83(3): 264-9, 1998 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-9710118

RESUMEN

During the development of atherosclerotic lesions, lipoprotein(a) [Lp(a)], a highly atherogenic lipoprotein, accumulates within fibrin clots attached to blood vessel walls. As Lp(a) accumulates within the fibrin clot with time, fatty streaks are formed that develop into occlusive atherosclerotic plaques. It is not understood, however, which mechanisms are involved in the binding of Lp(a) to fibrin and, hence, the stable incorporation of Lp(a) into the fibrin clot. The results of the present study demonstrate that factor XIIIa, a transglutaminase that catalyzes the formation of amide bonds between endo-gamma-glutaminyl and endo-epsilon-lysyl residues of proteins, is capable of cross-linking Lp(a) to fibrinogen, the soluble precursor of fibrin. Biochemical assays were conducted to demonstrate that factor XIIIa cross-links Lp(a) with fibrinogen in a time- and concentration-dependent manner. Additionally, immunohistochemical studies revealed that factor XIII protein expression colocalizes with Lp(a) expression in human atherosclerotic plaques. It is proposed that factor XIIIa-mediated cross-linking of Lp(a) to fibrin effectively increases the local concentration of Lp(a) within a fibrin clot. The accumulation of Lp(a) within the blood vessel promotes an antifibrinolytic environment, foam cell formation, the generation of a fatty streak, and an increase in smooth muscle cell content, all of which may contribute to the pathogenesis of atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Fibrinógeno/metabolismo , Lipoproteína(a)/metabolismo , Transglutaminasas/metabolismo , Western Blotting , Reactivos de Enlaces Cruzados/metabolismo , Ensayo de Inmunoadsorción Enzimática , Fibrina/metabolismo , Humanos , Técnicas In Vitro
12.
J Mol Cell Cardiol ; 30(3): 495-507, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9515027

RESUMEN

Cardiomyocyte apoptosis has been demonstrated in animal models of cardiac injury as well as in patients with congestive heart failure or acute myocardial infarction. Therefore, apoptosis has been proposed as an important process in cardiac remodeling and progression of myocardial dysfunction. However, the mechanisms underlying cardiac apoptosis are poorly understood. The present study was designed to determine whether the family of caspase proteases and stress-activated protein kinase (SAPK/JNK) are involved in cardiac apoptosis. Cultured rat neonatal cardiac myocytes were treated with staurosporine to induce apoptosis as evidenced by the morphological (including ultrastructural) characteristics of cell shrinkage, cytoplasmic and nuclear condensation, and fragmentation. Nucleosomal DNA fragmentation in myocytes was further identified by agarose gel electrophoresis (DNA ladder) as well as in situ nick end-labeling (TUNEL). Staurosporine-induced apoptosis in myocytes was a time- and concentration-(0.25-1 micro M)-dependent process. Staurosporine-induced apoptosis in myocytes was reduced by a cell-permeable, irreversible tripeptide inhibitor of caspases, ZVAD-fmk, but not by the ICE-specific inhibitor, Ac-YVAD-CHO. At 10, 50 and 100 muM of ZVAD-fmk, staurosporine-induced myocyte apoptosis was reduced by 5.8, 39.1 (P<0.01) and 53.8% (P<0.01), respectively. Staurosporine, at 0.25-1 micro M, increased caspase activity in cardiomyocytes by five- to eight-fold, peaking at 4-8 h after stimulation. Based on substrate specificity analysis, the major component of caspases activated in myocytes was consistent with caspase-3 (CPP32). Moreover, the appearance of the 17-kD subunit of active caspase-3 in staurosporine-treated myocytes was demonstrated by immunocytochemical analysis. In contrast, staurosporine induced a rapid and transient inhibition of SAPK/JNK in myocytes. The SAPK activity in myocytes was reduced by 68.3 and 58.3% (P<0.01 v basal) at 10 and 30 min after treatment with 1 micro M of staurosporine, respectively. Our results suggest that staurosporine-induced cardiac myocyte apoptosis involves activation of caspases, mainly caspase-3, but not activation of the SAPK signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas , Cisteína Endopeptidasas/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Miocardio/citología , Miocardio/enzimología , Estaurosporina/farmacología , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Caspasa 3 , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Corazón/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos , Oligopéptidos/química , Ratas , Transducción de Señal , Especificidad por Sustrato
13.
Stroke ; 29(5): 1020-30, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9596253

RESUMEN

BACKGROUND AND PURPOSE: Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes that degrade the extracellular matrix and are implicated in numerous pathological conditions including atherosclerosis, inflammation, and tumor growth and metastasis. In the brain, the endothelial cell wall, strengthened by tight junctions, defines the blood-brain barrier (BBB). The extracellular matrix molecules constitute the basement membrane underlying the vasculature and play a critical role in maintaining the integrity of the BBB. After focal stroke, there is a breakdown of the BBB with an associated increase in vascular permeability, inflammatory cell influx, and neuronal cell death. The present study was designed to investigate the effects of MMP expression after stroke. METHODS: Focal stroke was produced by permanent middle cerebral artery occlusion (MCAO) in the rat, and MMP protein expression was measured by Western blot and zymogram analysis over a time course ranging from 6 hours to 30 days (n=32). Immunohistochemistry at 1 and 5 days (n=8 and 6, respectively) was also utilized to characterize the expression of several MMPs and related proteins after stroke, including their cellular source. To test the hypothesis that early increased MMP-9 expression is involved in ischemic brain injury, a neutralizing monoclonal antibody directed against MMP-9 was administered intravenously (n=7 per group) 1 hour before MCAO, and infarct size was measured 24 hours later. RESULTS: MMP expression increased progressively over time after stroke. After 12 hours, significant (P<0.05) MMP-9 activity was observed that reached maximum levels by 24 hours (P<0.001), then persisted for 5 days at this level and returned to basal (zero) levels by 15 days. On the basis of morphological criteria, MMP-9 appeared to stain with endothelial cells and neutrophils identified both within and at the periphery of the infarct within 24 hours of focal ischemia. After 5 days, MMP-9 appeared to stain with macrophages present within the infarcted brain. MMP-2 activity was significantly (P<0.001) increased by 24 hours and was maximum after 5 days following MCAO. MMP-2 appeared to stain with macrophages present within the infarcted region. Unlike MMP-9 and MMP-2, tissue inhibitor of metalloproteinase-1 was identified at comparable levels in both control and ischemic tissue after MCAO. MMP-1 and MMP-3 could not be detected in the brain after focal stroke. When an MMP-9-neutralizing monoclonal antibody was administered systemically, animals exhibited significantly reduced infarct size (ie, a 30% reduction compared with non-immune antibody controls; P<0.05). CONCLUSIONS: These results demonstrate that early increased MMP-9 expression in endothelial cells and infiltrating neutrophils is a significant response to cerebral focal ischemia and that selective inhibition of MMP-9 activity can significantly reduce brain injury after stroke.


Asunto(s)
Isquemia Encefálica/enzimología , Colagenasas/biosíntesis , Colagenasas/metabolismo , Metaloproteinasa 3 de la Matriz/biosíntesis , Metaloendopeptidasas/biosíntesis , Animales , Anticuerpos Monoclonales/inmunología , Arteriopatías Oclusivas/enzimología , Arteriopatías Oclusivas/fisiopatología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/enzimología , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Infarto Cerebral/enzimología , Infarto Cerebral/patología , Colagenasas/análisis , Electroforesis en Gel de Poliacrilamida , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Inducción Enzimática , Gelatinasas/análisis , Gelatinasas/metabolismo , Inmunohistoquímica , Macrófagos/citología , Macrófagos/enzimología , Masculino , Metaloproteinasa 1 de la Matriz , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Inhibidores de la Metaloproteinasa de la Matriz , Metaloendopeptidasas/análisis , Metaloendopeptidasas/metabolismo , Neutrófilos/citología , Neutrófilos/enzimología , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/inmunología , Ratas , Ratas Endogámicas SHR , Dodecil Sulfato de Sodio , Factores de Tiempo , Extractos de Tejidos/química , Inhibidor Tisular de Metaloproteinasa-1/análisis , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidores Tisulares de Metaloproteinasas/administración & dosificación , Inhibidores Tisulares de Metaloproteinasas/inmunología
14.
J Mol Cell Cardiol ; 30(1): 167-73, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9500873

RESUMEN

Carvedilol, a multiple action neurohumoral antagonist, has been reported recently to significantly reduce mortality in congestive heart failure (CHF) patients. In addition to being a beta-adrenoceptor antagonist, one of the unique aspects of the biological effects of carvedilol is that it is also a potent antioxidant with antimitogenic properties. As there is a significant correlation between plasma immunoreactive endothelin-1 (ET-1) levels and the severity of CHF, the present study was designed to determine the effects of carvedilol on ET-1 biosynthesis in cultured human coronary artery endothelial cells (HCAECs). HCAECs were treated with carvedilol 15 min prior to the addition of serum and ET-1 levels were measured in the cell culture conditioned medium 24 h later. Carvedilol (10 microM) significantly inhibited basal ET-1 production in HCAECs by 62 +/- 8%. Carvedilol produced a concentration-dependent inhibition of serum-mediated stimulation of ET-1 biosynthesis with an IC50 = 1.2 microM. PreproET-1 mRNA expression was also inhibited by carvedilol. Other beta-adrenoceptor antagonists, such as propranolol (10 microM) or celiprolol (10 microM), did not effect ET-1 biosynthesis. Furthermore, the antioxidant probucol (10 microM) did not effect ET-1 production. Immunohistochemical analysis of HCAECs demonstrated that resting HCAECs have expression of ET-1 and can be inhibited by carvedilol. The results of the present study demonstrate that serum stimulation of HCAECs produced an increase in ET-1 expression, and carvedilol treatment caused a marked decrease in stimulated ET-1 expression as compared to serum-treated HCAECs. These data indicate that carvedilol directly inhibits the biosynthesis of ET-1 in HCAECs, and this effect may contribute to its vasodilating and antiproliferative actions. Furthermore, these effects may contribute to the ability of carvedilol to improve clinical outcome in CHF patients.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Antioxidantes/farmacología , Carbazoles/farmacología , Endotelina-1/biosíntesis , Endotelio Vascular/metabolismo , Propanolaminas/farmacología , Carvedilol , Células Cultivadas , Vasos Coronarios , Endotelina-1/genética , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , ARN Mensajero/biosíntesis
15.
Circ Res ; 82(2): 166-74, 1998 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-9468187

RESUMEN

Carvedilol, a new vasodilating beta-adrenoceptor antagonist and a potent antioxidant, produces a high degree of cardioprotection in a variety of experimental models of ischemic cardiac injury. Recent clinical studies in patients with heart failure have demonstrated that carvedilol reduces morbidity and mortality and inhibits cardiac remodeling. The present study was designed to explore whether the protective effects of carvedilol on the ischemic myocardium include inhibition of apoptosis of cardiomyocytes and, if so, to determine its mechanism of action. Anesthetized rabbits were subjected to 30 minutes of coronary artery occlusion followed by 4 hours of reperfusion. Detection of apoptosis of cardiomyocytes was based on the presence of nucleosomal DNA fragments on agarose gels (DNA ladder) and in situ nick end labeling. Carvedilol (1 mg/kg IV), administered 5 minutes before reperfusion, reduced the number of apoptotic myocytes in the ischemic area from 14.7 +/- 0.4% to 3.4 +/- 1.8% (77% reduction, P<.001). Propranolol, administered at equipotent beta-blocking dosage, reduced the number of apoptotic myocytes to 8.9 +/- 2.1% (39% reduction, P<.05). DNA ladders were observed in the hearts of all six vehicle-treated rabbits but only one of six carvedilol-treated rabbits (P<.01). Immunocytochemical analysis of rabbit hearts demonstrated an upregulation of Fas protein in ischemic cardiomyocytes, and treatment with carvedilol reduced both the intensity of staining as well as the area stained. Myocardial ischemia/reperfusion led to a rapid activation of stress-activated protein kinase (SAPK) in the ischemic area but not in nonischemic regions. SAPK activity was increased from 2.1 +/- 0.3 mU/mg (basal) to 8.9 +/- 0.8 mU/mg after 30 minutes of ischemia followed by 20 minutes of reperfusion. Carvedilol inhibited the activation of SAPK by 53.4 +/- 6.5% (P<.05). Under the same conditions, propranolol (1 mg/kg) had no effect on SAPK activation. Taken together, these results suggest that carvedilol prevents myocardial ischemia/reperfusion-induced apoptosis in cardiomyocytes possibly by downregulation of the SAPK signaling pathway, by inhibition of Fas receptor expression, and by beta-adrenergic blockade. The former two actions represent novel and important mechanisms that may contribute to the cardioprotective effects of carvedilol.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Apoptosis/efectos de los fármacos , Carbazoles/uso terapéutico , Glicoproteínas de Membrana/metabolismo , Isquemia Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Propanolaminas/uso terapéutico , Proteínas Quinasas/metabolismo , Animales , Carvedilol , Fragmentación del ADN/fisiología , Regulación hacia Abajo/fisiología , Activación Enzimática/fisiología , Proteína Ligando Fas , Hemodinámica/efectos de los fármacos , Masculino , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Conejos , Transducción de Señal/fisiología , Estrés Fisiológico/metabolismo
16.
Lab Invest ; 76(1): 11-23, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9010446

RESUMEN

During inflammation, T cells transmigrate from the bloodstream into perivascular tissues. As T cells transmigrate, they undergo a series of attachments to and detachments from the endothelium and then extravasate into the extracellular matrix (ECM). T cell migration into the ECM involves a number of mechanisms that influence cell-ECM interactions. The modulation of integrin expression and affinity are two such mechanisms in which cells can alter their ability to interact with other cells and ECM. We show in vitro that transmigrated T cells exhibit down-regulation of very late activation antigen-4 and leukocyte function-associated antigen-1 integrin surface expression and a decrease in binding to recombinant vascular cell adhesion molecule-1 and recombinant intercellular adhesion molecule-1. Also, transmigrated T cells displayed an increase in binding to collagens I and IV and fibronectin. Further, brain sections of experimental autoimmune encephalomyelitis mice demonstrated that as T cells migrated farther into the tissue, very late activation antigen-4 expression was lost while CD4 expression remained unchanged. The significance of these findings in the modulation of the inflammatory response is discussed.


Asunto(s)
Adhesión Celular , Endotelio Vascular/fisiología , Matriz Extracelular/fisiología , Linfocitos T/fisiología , Animales , Encéfalo/inmunología , Encéfalo/patología , Movimiento Celular , Células Cultivadas , Colágeno/fisiología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Endotelio Vascular/inmunología , Fibronectinas/fisiología , Citometría de Flujo , Integrina alfa4beta1 , Integrinas/biosíntesis , Molécula 1 de Adhesión Intercelular/farmacología , Interleucina-2/farmacología , Antígeno-1 Asociado a Función de Linfocito/biosíntesis , Ratones , Ratones Endogámicos , Proteína Básica de Mielina/farmacología , Fragmentos de Péptidos/farmacología , Receptores Mensajeros de Linfocitos/biosíntesis , Receptores de Antígeno muy Tardío/biosíntesis , Proteínas Recombinantes/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/farmacología , Molécula 1 de Adhesión Celular Vascular/fisiología
17.
Cell Growth Differ ; 7(11): 1453-60, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8930394

RESUMEN

One of the most critical cellular events in disease states such as vascular restenosis is the cytokine-induced activation of vascular smooth muscle cells (VSMCs) resulting in intimal thickening. Identification of the molecular mechanisms of VSMC activation is crucial in understanding the initiation of vascular restenosis. In this report, we show that one 14-3-3 protein family isoform, gamma, is transcriptionally up-regulated in rat carotid arteries after balloon angioplasty. 14-3-3 gamma protein induction localizes to both the media and neointima in such injured vessels. Because it has been shown that some members of the 14-3-3 family may play an important role in cellular proliferation by binding to and activating the protein kinase Raf-1 and VSMCs constitute the major cellular component of the restenotic lesion, we investigated the expression of this message in serum- and cytokine-stimulated human VSMCs. Both serum and selected cytokines induce 14-3-3 gamma mRNA and protein, the magnitude of which correlates with the degree of cellular stimulation. 14-3-3 gamma mRNA, however, does not increase when other cell types are stimulated with specific growth factors. Human tissue distribution of 14-3-3 gamma mRNA indicates that in contrast to other 14-3-3 proteins, the gamma isoform is highly expressed in VSMCs and skeletal and heart muscle, suggesting an important role for the gamma isoform in muscle tissue as well. These results indicate that 14-3-3 gamma expression increases in response to vessel damage and proliferative signals and may implicate a role for the gamma isoform of 14-3-3 in VSMC activation and metabolism.


Asunto(s)
Arterias Carótidas/metabolismo , Músculo Liso Vascular/metabolismo , Biosíntesis de Proteínas , Tirosina 3-Monooxigenasa , Proteínas 14-3-3 , Angioplastia de Balón , Animales , Arterias Carótidas/patología , Células Cultivadas , Cicloheximida/farmacología , Regulación de la Expresión Génica , Sustancias de Crecimiento/farmacología , Humanos , Masculino , Especificidad de Órganos , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas/genética , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Linfocitos T , Túnica Íntima/metabolismo
18.
Anal Biochem ; 223(2): 173-80, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7887459

RESUMEN

The enzymes procollagen C- and N-proteinases specifically cleave carboxyl- and amino-terminal propeptides of procollagens. After cleavage of the propeptides, the resulting collagens self-assemble into fibrils. In most previous experiments with the enzymes, the substrate was monomeric type I procollagen. Here we have prepared aggregates of type I procollagen from chick embryo tendons by using 1 to 100 micrograms/ml of 500-kDa dextran sulfate or 3 to 5% (w/v) polyethylene glycol (M(r) 3350). Aggregation of the substrate with dextran sulfate increased its rate of cleavage by purified or crude C-proteinase from chick embryo tendons 10- to 15-fold. Aggregation of the substrate with 25 to 100 microgram/ml of dextran sulfate increased the rate of cleavage by purified N-proteinase about 4-fold. The rate of cleavage by crude N-proteinase was enhanced only about 2-fold, apparently because of partial precipitation of the enzyme by dextran sulfate. Using polyethylene glycol to aggregate the substrate increased the rate of cleavage by procollagen C-proteinases 5- to 20-fold. Aggregation with polyethylene glycol also increased the rate of cleavage by purified procollagen N-proteinases 2- to 5-fold. With crude N-proteinase, the rate of cleavage was increased only 1.5-fold. The results suggest that the rate of cleavage of the substrate by both enzymes is increased by the aggregation of the substrate itself by dextran sulfate or polyethylene glycol. The increased rates of cleavage seen after aggregation of substrate can be used to develop more sensitive assays for the enzymic activities.


Asunto(s)
Proteínas Morfogenéticas Óseas , Endopeptidasas/metabolismo , Metaloendopeptidasas , Procolágeno N-Endopeptidasa/metabolismo , Procolágeno/metabolismo , Animales , Proteína Morfogenética Ósea 1 , Precipitación Química , Embrión de Pollo , Sulfato de Dextran , Técnicas In Vitro , Indicadores y Reactivos , Polietilenglicoles , Procolágeno/química , Procolágeno/aislamiento & purificación , Especificidad por Sustrato , Tendones/metabolismo
19.
J Cell Biol ; 125(5): 1165-78, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7515069

RESUMEN

T cell extravasation from the bloodstream into the perivascular tissue during inflammation involves transmigration through the endothelial cell layer and basement membrane into the interstitial matrix. The specific mechanisms by which T cells transmigrate, however, are poorly understood. Matrix degradation by enzymes such as 72-kD gelatinase has been implicated as an important component in tissue invasion by various types of cells. In this study, we have demonstrated that 72-kD gelatinase is induced in T cells upon adhesion to endothelial cells. We also provide evidence that the induction of 72-kD gelatinase is mediated by binding to vascular cell adhesion molecule-1 (VCAM-1). The T cells used in this study were cloned murine Th1 cells antigenic to myelin basic protein. These cells express very late antigen-4 on their cell surface and have been shown to infiltrate the brain parenchyma and cause experimental autoimmune encephalomyelitis when infused into normal mice (Baron, J. L., J. A. Madri, N. H. Ruddle, G. Hashim, and C. A. Janeway. 1993. J. Exp. Med. 177:57-68). In the experiments presented here, T cells were cocultured with VCAM-1-positive and -negative endothelial cells grown in a monolayer in order to study the expression of 72-kD gelatinase upon T cell adhesion. Additional experiments were conducted in which T cells were cocultured with VCAM-1 positive cells grown on microporous membranes suspended in transwells to study 72-kD gelatinase following T cell transmigration. T cells were also incubated with recombinant VCAM-1 in order to study the role of VCAM-1 in inducing 72-kD gelatinase. The results demonstrated that T cells adhered to both VCAM-1-positive and -negative endothelial cells. T cells that adhered to the VCAM-1-positive endothelial cells exhibited an induction in 72-kD gelatinase protein, activity, and mRNA whereas 72-kD gelatinase was not induced in the T cells that adhered to the VCAM-1-negative endothelial cells. Incubating T cells with recombinant VCAM-1 coated onto tissue culture plastic showed that T cells adhered to the molecule and that adhesion to recombinant VCAM-1 was sufficient to induce 72-kD gelatinase. Further, T cells that had transmigrated through a VCAM-1-positive endothelial cell monolayer exhibited 72-kD gelatinase activity but not mRNA expression. In addition, 72-kD gelatinase was detected on the cell surface of the transmigrated T cells by FACS analysis. In other experiments, TIMP-2 was added to the transmigration studies and was shown to reduce T cell transmigration.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Linfocitos T CD4-Positivos/enzimología , Moléculas de Adhesión Celular/fisiología , Gelatinasas/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Adhesión Celular , Línea Celular , Movimiento Celular , Endotelio Vascular/citología , Inducción Enzimática , Ratones , Proteínas/farmacología , Inhibidor Tisular de Metaloproteinasa-2 , Molécula 1 de Adhesión Celular Vascular
20.
J Biol Chem ; 269(15): 11614-9, 1994 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8157695

RESUMEN

Procollagen I was isolated from cultured skin fibroblasts from a proband who was homozygous for a mutation in the COL1A2 gene that substituted a serine codon for a glycine codon at position 661 of the alpha 2(I) chain. The procollagen I was cleaved to pCcollagen I by procollagen N-proteinase and the pCcollagen I was used as a substrate for assay of self-assembly of collagen I into fibrils. The mutated pCcollagen I was cleaved to collagen I by procollagen C-proteinase at the same rate as control pCcollagen I. However, self-assembly of the mutated collagen I had a lag period that was 15-fold greater than the lag period observed with normal collagen I under the same conditions. Also, self-assembly of the mutated collagen I had a propagation rate of about one-fourth of the propagation rate of normal collagen I. In addition, the critical concentration for fibril assembly was slightly increased. Rotary shadowing electron microscopy of the mutated procollagen I did not reveal any increased flexibility of the triple helix as was seen previously with two mutated procollagens I in which there were substitutions of cysteine for glycine residues in the alpha 1(I) chain (Vogel, B. E., Doelz, R., Kadler, K. E., Hojima, Y., Engel, J., and Prockop, D. J. (1988) J. Biol. Chem. 263, 19249-19255; Lightfoot, S. J., Holmes, D. F., Brass, A., Grant, M. E., Byers, P. H., and Kadler, K. E. (1992) J. Biol. Chem. 267, 25521-25528). However, morphometric analysis by dark-field light microscopy and electron microscopy showed that the fibrils formed from the mutated collagen I appeared thicker in diameter than the fibrils formed from the normal collagen I. Comparison of the results with similar data on four mutated procollagens previously studied raised the possibility that mutations which markedly increase the critical concentration of fibril assembly produce more severe phenotypes than mutations which change other parameters of fibril assembly.


Asunto(s)
Colágeno/biosíntesis , Colágeno/genética , Glicina , Mutación Puntual , Procolágeno/genética , Serina , Piel/metabolismo , Secuencia de Aminoácidos , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Humanos , Cinética , Sustancias Macromoleculares , Microscopía Electrónica , Fenotipo , Procolágeno/ultraestructura , Procolágeno N-Endopeptidasa/metabolismo , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA