Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Clin Exp Immunol ; 185(3): 372-81, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27218304

RESUMEN

Recently, there has been considerable interest in using 4-methylumbelliferone (4-MU) to inhibit hyaluronan (HA) synthesis in mouse models of cancer, autoimmunity and a variety of other inflammatory disorders where HA has been implicated in disease pathogenesis. In order to facilitate future studies in this area, we have examined the dosing, treatment route, treatment duration and metabolism of 4-MU in both C57BL/6 and BALB/c mice. Mice fed chow containing 5% 4-MU, a dose calculated to deliver 250 mg/mouse/day, initially lose substantial weight but typically resume normal weight gain after 1 week. It also takes up to a week to see a reduction in serum HA in these animals, indicating that at least a 1-week loading period on the drug is required for most protocols. At steady state, more than 90% of the drug is present in plasma as the glucuronidated metabolite 4-methylumbelliferyl glucuronide (4-MUG), with the sulphated metabolite, 4-methylumbelliferyl sulphate (4-MUS) comprising most of the remainder. Chow containing 5% but not 0·65% 4-MU was effective at preventing disease in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis, as well as in the DORmO mouse model of autoimmune diabetes. While oral 4-MU was effective at preventing EAE, daily intraperitoneal injections of 4-MU were not. Factors potentially affecting 4-MU uptake and plasma concentrations in mice include its taste, short half-life and low bioavailability. These studies provide a practical resource for implementing oral 4-MU treatment protocols in mice.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Ácido Hialurónico/antagonistas & inhibidores , Ácido Hialurónico/biosíntesis , Himecromona/administración & dosificación , Himecromona/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Semivida , Ácido Hialurónico/sangre , Himecromona/sangre , Himecromona/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
2.
Immunol Res ; 58(2-3): 186-92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24614953

RESUMEN

The extracellular matrix polysaccharide hyaluronan (HA) exerts size-dependent effects on leukocyte behavior. Low-molecular weight HA is abundant at sites of active tissue catabolism and promotes inflammation via effects on Toll-like receptor signaling. Conversely, high-molecular weight HA is prevalent in uninjured tissues and is anti-inflammatory. We propose that the ability of high-molecular weight but not low-molecular weight HA to cross-link CD44 functions as a novel form of pattern recognition that recognizes intact tissues and communicates "tissue integrity signals" that promote resolution of local immune responses.


Asunto(s)
Ácido Hialurónico/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Transducción de Señal , Animales , Matriz Extracelular/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/química , Peso Molecular , Unión Proteica , Receptores de Reconocimiento de Patrones/metabolismo
3.
Acta Biomater ; 5(4): 1019-26, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19109082

RESUMEN

The addition of exogenous hyaluronan to biomaterial scaffolds has been an important area of investigation for many decades. The ability to manipulate endogenous production of hyaluronan via the hyaluronan syntheses has offered another mechanism to study the effect of hyaluronan. While the literature suggests that exogenously added hyaluronan and endogenously produced hyaluronan will have varying impacts on extracellular matrix organization and function, no studies have directly shown this phenomenon. In this investigation, we demonstrate that the addition of exogenous high molecular weight (approximately 1 MDa) hyaluronan and hyaluronan oligosaccharides have a distinct impact on both contraction and strength of smooth muscle cell-seeded collagen gels when compared to the effects of hyaluronan that is endogenously produced by the hyaluronan synthases. More specifically, the addition of exogenous high molecular weight hyaluronan resulted in more compact collagen gels with a higher ultimate tensile strength, whereas the endogenous overproduction of hyaluronan resulted in the opposite effect. We suggest that the addition of exogenous HA to collagen gels represents a model for the therapeutic administration of HA, whereas the addition of excess HA to a tissue via the endogenous overexpression of has represents a model for the pathological accumulation of HA.


Asunto(s)
Colágeno/química , Ácido Hialurónico/química , Animales , Células Cultivadas , Colágeno/metabolismo , ADN/genética , Proteínas de la Matriz Extracelular/metabolismo , Geles/química , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Inmunohistoquímica , Masculino , Ratas
4.
Diabetologia ; 47(12): 2105-13, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15592811

RESUMEN

AIMS/HYPOTHESIS: Vascular disease in type 2 diabetes is associated with an up-regulation of atherogenic growth factors, which stimulate matrix synthesis including proteoglycans. We have examined the direct actions of fenofibrate on human vascular smooth muscle cells (VSMCs) and have specifically investigated proteoglycan synthesis and binding to LDL. METHODS: Proteoglycans synthesised by human VSMCs treated with fenofibrate (30 micromol/l) were assessed for binding to human LDL using a gel mobility shift assay, metabolically labelled with [(35)S]-sulphate and quantitated by cetylpyridinium chloride. They were then assessed for electrophoretic mobility by SDS-PAGE, for size by gel filtration, for sulphation pattern by fluorophore-assisted carbohydrate electrophoresis, and for glycosaminoglycan (GAG) composition by enzyme digestion. RESULTS: Proteoglycans synthesised in the presence of fenofibrate showed an increase in the half-maximum saturation concentration of LDL from 36.8+/-12.4 microg/ml to 77.7+/-17 microg/ml under basal conditions, from 24.9+/-4.6 microg/ml to 39.1+/-6.1 microg/ml in the presence of TGF-beta1, and from 9.5+/-4.4 microg/ml to 31.1+/-3.4 microg/ml in the presence of platelet-derived growth factor/insulin. Fenofibrate treatment in the presence of TGF-beta1 inhibited the incorporation of [(35)S]-sulphate into secreted and cell-associated proteoglycans synthesised by human VSMCs by 59.2% (p<0.01) and 39.8% (p<0.01) respectively. The changes in sulphate incorporation following treatment with fenofibrate were associated with a concentration-related increase in the electrophoretic mobility due to a reduction in GAG length. There was no change in the sulphation pattern; however, there was an alteration in the disaccharide composition of the GAGs. CONCLUSIONS/INTERPRETATION: Fenofibrate modifies the structure of vascular proteoglycans by reducing the length of the GAG chains and GAG composition, resulting in reduced binding to human LDL, a mechanism which may lead to a reduction of atherosclerosis and cardiovascular disease in people with diabetes treated with fenofibrate.


Asunto(s)
Fenofibrato/farmacología , Hipolipemiantes/farmacología , Lipoproteínas/metabolismo , Músculo Liso Vascular/fisiología , Proteoglicanos/metabolismo , Células Cultivadas , Glicosaminoglicanos/metabolismo , Humanos , Insulina/farmacología , Lipoproteínas/efectos de los fármacos , Arterias Mamarias , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteoglicanos/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
5.
Diabetologia ; 47(5): 837-43, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15071727

RESUMEN

AIMS/HYPOTHESIS: Retention of atherogenic lipoproteins in the artery wall by proteoglycans is a key step in the development of atherosclerosis. Thiazolidinediones have been shown to reduce atherosclerosis in mouse models. The aim of this study was to determine whether thiazolidinediones modify vascular proteoglycan synthesis in a way that decreases LDL binding. METHODS: Primate aortic smooth muscle cells were exposed to troglitazone or rosiglitazone, or no stimulus at all for a 24-hour steady-state labelling period. Sulphate incorporation, size and LDL binding affinity of proteoglycans were determined. Proteoglycans secreted by cells in the presence or absence of troglitazone were separated into large and small classes by size exclusion chromatography, and LDL binding affinity was determined. RESULTS: Proteoglycans synthesised by cells exposed to troglitazone or rosiglitazone were smaller, with decreased sulphate incorporation and decreased LDL binding affinity. However, troglitazone had a greater effect than rosiglitazone. Troglitazone reduced the LDL binding affinities of both the large and small proteoglycans compared with control. The binding differences persisted when glycosaminoglycan chains released from proteoglycans were incubated with LDL, indicating that troglitazone affects the glycosaminoglycan synthetic machinery of these cells. CONCLUSIONS/INTERPRETATION: Thiazolidinediones decrease the LDL binding affinity of the proteoglycans synthesised by primate aortic smooth muscle cells. This could, in part, account for the reduced atherosclerosis observed in animal models.


Asunto(s)
Lipoproteínas LDL/sangre , Proteoglicanos/metabolismo , Tiazolidinedionas/farmacología , Animales , Sitios de Unión , Células Cultivadas , Cromanos/farmacología , Hipoglucemiantes/farmacología , Macaca nemestrina , Músculo Liso Vascular/metabolismo , Troglitazona
6.
Arch Biochem Biophys ; 394(1): 29-38, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11566024

RESUMEN

Hyaluronan and versican-rich pericellular matrices form around arterial smooth muscle cells (ASMC) preferentially during the detachment phase of proliferation and migration. PDGF is a potent mitogen and chemotactic agent for ASMC and also stimulates the production of extracellular matrix molecules which may regulate the proliferative and migratory capacity of the cells. We have examined the effect of PDGF on the formation of hyaluronan-dependent pericellular matrices, and on the synthesis and interaction of several major pericellular coat constituents. As demonstrated using a particle exclusion assay, PDGF stimulated the formation of pericellular matrices and was seen both in an increased proportion of cells with a coat and a greater coat size. This increase was accompanied by a transient increase in hyaluronan synthase 2 (HAS2) expression and an increase in hyaluronan synthesis and polymer length. PDGF also increased the synthesis of versican and link protein as measured at the mRNA and protein levels. The amount of native versican-hyaluronan aggregates and link-stabilized aggregate was also increased following PDGF treatment. Time lapse imaging showed that pericellular matrix formation occurred around trailing cell processes prior to their detachment. These data suggest that PDGF modulates the synthesis and organization of ASMC pericellular coat-forming molecules such as versican, hyaluronan, and link protein, which leads to extracellular matrix expansion and alterations in ASMC phenotype.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Matriz Extracelular/efectos de los fármacos , Glicosiltransferasas , Ácido Hialurónico/metabolismo , Proteínas de la Membrana , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Transferasas , Proteínas de Xenopus , Aorta , Adhesión Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Proteoglicanos Tipo Condroitín Sulfato/biosíntesis , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Humanos , Hialuronano Sintasas , Ácido Hialurónico/biosíntesis , Recién Nacido , Lectinas Tipo C , Músculo Liso Vascular/metabolismo , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Versicanos
9.
J Cell Physiol ; 188(1): 67-74, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11382923

RESUMEN

Smooth muscle cell migration, proliferation, and deposition of extracellular matrix are key events in atherogenesis and restenosis development. To explore the mechanisms that regulate smooth muscle cell function, we have investigated whether perlecan, a basement membrane heparan sulfate proteoglycan, modulates interaction between smooth muscle cells and other matrix components. A combined substrate of fibronectin and perlecan showed a reduced adhesion of rat aortic smooth muscle cells by 70-90% in comparison to fibronectin alone. In contrast, perlecan did not interfere with cell adhesion to laminin. Heparinase treated perlecan lost 60% of its anti-adhesive effect. Furthermore, heparan sulfate as well as heparin reduced smooth muscle cell adhesion when combined with fibronectin whereas neither hyaluronan nor chondroitin sulfate had any anti-adhesive effects. Addition of heparin as a second coating to a preformed fibronectin matrix did not affect cell adhesion. Cell adhesion to the 105- and 120 kDa cell-binding fragments of fibronectin, lacking the main heparin-binding domains, was also inhibited by heparin. In addition, co-coating of fibronectin and (3)H-heparin showed that heparin was not even incorporated in the substrate. Morphologically, smooth muscle cells adhering to a substrate prepared by co-coating of fibronectin and perlecan or heparin were small, rounded, lacked focal contacts, and showed poorly developed stress fibers of actin. The results show that the heparan sulfate chains of perlecan lead to altered interactions between smooth muscle cells and fibronectin, possibly due to conformational changes in the fibronectin molecule. Such interactions may influence smooth muscle cell function in atherogenesis and vascular repair processes.


Asunto(s)
Adhesión Celular/fisiología , Fibronectinas/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Animales , Aorta/anatomía & histología , Tamaño de la Célula , Sulfatos de Condroitina/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Laminina/metabolismo , Masculino , Unión Proteica , Ratas , Ratas Endogámicas F344 , Fibras de Estrés/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 21(5): 777-84, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11348874

RESUMEN

Decorin is a member of the family of small leucine-rich proteoglycans that are present in blood vessels and synthesized by arterial smooth muscle cells (ASMCs). This proteoglycan accumulates in topographically defined regions of atherosclerotic lesions and may play a role in the development of this disease. However, little is known about whether decorin has specific effects on the cellular events that contribute to atherosclerotic lesion formation. In the present study, rat ASMCs were transduced with a retroviral vector (LDSN) that carries the bovine decorin gene. Compared with vector control cells (LXSN), these cells constitutively overexpress decorin, as verified by Northern and Western analysis and by metabolic labeling. Experiments were performed to examine the responsiveness of decorin-overexpressing rat ASMCs to platelet-derived growth factor (PDGF) and transforming growth factor-beta1 (TGF-beta1), 2 growth factors that affect cell proliferation and extracellular matrix production in atherosclerosis. Decorin-overexpressing cells had decreased [(3)H]thymidine incorporation into DNA and increased the levels of the cyclin-dependent kinase inhibitors p21 and p27 in the first 24 hours of response to serum and PDGF-BB. However, these effects of decorin were not apparent at 48 or 72 hours after plating and did not result in reduced growth of decorin-overexpressing cells in response to serum and PDGF-BB. In contrast, the growth response of decorin-overexpressing ASMCs to TGF-beta1, as well as the expression of TGF-beta1-responsive genes, such as plasminogen activator inhibitor-1 and versican (an extracellular matrix proteoglycan), was diminished. These results indicate that decorin selectively inhibits the responsiveness of rat ASMCs to TGF-beta1 and suggests that the induction of constitutive decorin overexpression by ASMCs in vivo may have therapeutic value in the inhibition of TGF-beta1-mediated effects on the development of atherosclerotic lesions.


Asunto(s)
Proteínas de Ciclo Celular , Músculo Liso Vascular/metabolismo , Proteoglicanos/fisiología , Factor de Crecimiento Transformador beta/farmacología , Proteínas Supresoras de Tumor , Animales , Arterias/citología , Arterias/efectos de los fármacos , Arterias/metabolismo , División Celular/efectos de los fármacos , Proteoglicanos Tipo Condroitín Sulfato/biosíntesis , Proteoglicanos Tipo Condroitín Sulfato/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , ADN/biosíntesis , Decorina , Proteínas de la Matriz Extracelular , Lectinas Tipo C , Proteínas Asociadas a Microtúbulos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Inhibidor 1 de Activador Plasminogénico/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteoglicanos/biosíntesis , Proteoglicanos/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas F344 , Retroviridae/genética , Transducción Genética , Factor de Crecimiento Transformador beta1 , Versicanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA