Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Carbohydr Polym ; 256: 117513, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33483034

RESUMEN

Marine sulfated polysaccharides have aroused widespread concern for their various structures and bioactivities. Peroxide depolymerization is a common strategy in analysis of structures and structure-activity relationships of polysaccharides. However, confirming the depolymerization process and exact structures of the degradation products is still a considerable challenge. This study reported the structures of a fucan sulfate (FS) from sea cucumber Stichopus herrmanni and its depolymerized products (dFS) prepared by peroxide degradation. The FS was elucidated with a highly regular structure, {-3)-L-Fuc2S-(α1-}n. Structure analysis of oligosaccharides purified from dFS suggested that peroxide degradation involved in cleavage of glycosidic bonds and oxidative modification of reducing end of sugar residue, while no break in sugar ring was observed. Both FS and series of dFSs exhibited significant anticoagulant activities due to their anti-thrombin effects in presence of heparin cofactor II and their potencies were related to their molecular sizes, dFS with ∼ 20 kDa showed the strongest activity.


Asunto(s)
Anticoagulantes/química , Coagulación Sanguínea/efectos de los fármacos , Peróxidos/química , Polisacáridos/química , Stichopus/química , Animales , Anticoagulantes/aislamiento & purificación , Anticoagulantes/farmacología , Organismos Acuáticos , Pruebas de Coagulación Sanguínea , Secuencia de Carbohidratos , Cofactor II de Heparina/farmacología , Humanos , Hidrólisis , Peso Molecular , Polisacáridos/aislamiento & purificación , Polisacáridos/farmacología , Stichopus/fisiología , Relación Estructura-Actividad , Trombina/antagonistas & inhibidores , Trombina/metabolismo
2.
Int J Biol Macromol ; 164: 87-94, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32663563

RESUMEN

A sulfated fucan was extracted, purified, and characterized from Acaudina leucoprocta (a low value sea cucumber) to better understand and utilize this species. The structure of the sulfated fucan was elucidated using chemical and modern spectroscopic analyses including HPGPC, IR, AFM, GC-MS, and NMR, and its bioactivity was investigated. Our results showed that the sulfated fucan was mainly composed of → 3)-α-L-Fucp-(1→ linkage, and that the sulfate groups were substituted at the O-2 and/or O-4 positions of the fucose ring. In detail, the sulfated fucan consisted of Fuc0S (40%), Fuc2S4S (24%), Fuc2S (24%), and Fuc4S (12%). On average, there were seven sulfate groups on every eight fucose residues. Assay for anticoagulant activity indicated that the sulfated fucan displayed intrinsic anticoagulant activity and specific anti-thrombin activity through heparin cofactor II. Our results showed that this bioactive sulfated fucan could enable the high-value utilization of this low-value sea cucumber.


Asunto(s)
Anticoagulantes/aislamiento & purificación , Polisacáridos/aislamiento & purificación , Animales , Anticoagulantes/química , Anticoagulantes/farmacología , Pruebas de Coagulación Sanguínea , Conformación de Carbohidratos , Cromatografía por Intercambio Iónico , Cromatografía de Gases y Espectrometría de Masas , Cofactor II de Heparina/farmacología , Humanos , Microscopía de Fuerza Atómica , Estructura Molecular , Peso Molecular , Polisacáridos/química , Polisacáridos/farmacología , Pepinos de Mar/química , Análisis Espectral , Sulfatos/análisis
4.
Mar Drugs ; 15(1)2016 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-28042854

RESUMEN

Waste generated from the processing of marine organisms for food represents an underutilized resource that has the potential to provide bioactive molecules with pharmaceutical applications. Some of these molecules have known anti-thrombotic and anti-coagulant activities and are being investigated as alternatives to common anti-thrombotic drugs, like heparin and warfarin that have serious side effects. In the current study, extracts prepared from blacklip abalone (Haliotis rubra) processing waste, using food grade enzymes papain and bromelain, were found to contain sulphated polysaccharide with anti-thrombotic activity. Extracts were found to be enriched with sulphated polysaccharides and assessed for anti-thrombotic activity in vitro through heparin cofactor-II (HCII)-mediated inhibition of thrombin. More than 60% thrombin inhibition was observed in response to 100 µg/mL sulphated polysaccharides. Anti-thrombotic potential was further assessed as anti-coagulant activity in plasma and blood, using prothrombin time (PT), activated partial thromboplastin time (aPTT), and thromboelastography (TEG). All abalone extracts had significant activity compared with saline control. Anion exchange chromatography was used to separate extracts into fractions with enhanced anti-thrombotic activity, improving HCII-mediated thrombin inhibition, PT and aPTT almost 2-fold. Overall this study identifies an alternative source of anti-thrombotic molecules that can be easily processed offering alternatives to current anti-thrombotic agents like heparin.


Asunto(s)
Organismos Acuáticos/química , Fibrinolíticos/química , Fibrinolíticos/farmacología , Gastrópodos/química , Animales , Anticoagulantes/química , Anticoagulantes/farmacología , Pruebas de Coagulación Sanguínea/métodos , Cofactor II de Heparina/farmacología , Tiempo de Tromboplastina Parcial/métodos , Polisacáridos/química , Polisacáridos/farmacología , Tiempo de Protrombina/métodos , Trombina/metabolismo , Trombosis/tratamiento farmacológico
5.
Biochem Biophys Res Commun ; 457(4): 585-8, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600805

RESUMEN

Heparan sulfate normally binds to heparin cofactor II and modulates the coagulation pathway by inhibiting thrombin. However, when human heparin cofactor II was incubated with heparan sulfate, heparin cofactor II became degraded. Other glycosaminoglycans were tested, including hyaluronic acid, chondroitin sulfates, dermatan sulfate, and heparin, but only dextran sulfate also degraded heparin cofactor II. Pretreatment of heparan sulfate with heparinase reduced its heparin cofactor II-degrading activity. Heparan sulfate and dextran sulfate diminished the thrombin inhibitory activity of heparin cofactor II. Other serpins, including antithrombin III and pigment epithelium-derived factor, were also degraded by heparan sulfate. This is the first evidence of acidic polysaccharides exhibiting protein-degrading activity without the aid of other proteins.


Asunto(s)
Antitrombinas/metabolismo , Sulfato de Dextran/metabolismo , Cofactor II de Heparina/metabolismo , Heparitina Sulfato/metabolismo , Proteolisis , Animales , Antitrombina III/metabolismo , Antitrombinas/farmacología , Bovinos , Flavobacterium/enzimología , Cofactor II de Heparina/farmacología , Liasa de Heparina/metabolismo , Humanos , Indicadores y Reactivos/metabolismo
6.
J Biol Chem ; 289(43): 29790-800, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25202017

RESUMEN

Antithrombin III (ATIII) is a key antiproteinase involved in blood coagulation. Previous investigations have shown that ATIII is degraded by Staphylococcus aureus V8 protease, leading to release of heparin binding fragments derived from its D helix. As heparin binding and antimicrobial activity of peptides frequently overlap, we here set out to explore possible antibacterial effects of intact and degraded ATIII. In contrast to intact ATIII, the results showed that extensive degradation of the molecule yielded fragments with antimicrobial activity. Correspondingly, the heparin-binding, helix D-derived, peptide FFFAKLNCRLYRKANKSSKLV (FFF21) of human ATIII, was found to be antimicrobial against particularly the Gram-negative bacteria Escherichia coli and Pseudomonas aeruginosa. Fluorescence microscopy and electron microscopy studies demonstrated that FFF21 binds to and permeabilizes bacterial membranes. Analogously, FFF21 was found to induce membrane leakage of model anionic liposomes. In vivo, FFF21 significantly reduced P. aeruginosa infection in mice. Additionally, FFF21 displayed anti-endotoxic effects in vitro. Taken together, our results suggest novel roles for ATIII-derived peptide fragments in host defense.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Antitrombina III/química , Antitrombina III/farmacología , Secuencia de Aminoácidos , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Dicroismo Circular , Modelos Animales de Enfermedad , Cofactor II de Heparina/química , Cofactor II de Heparina/farmacología , Humanos , Lipopolisacáridos/metabolismo , Liposomas/metabolismo , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Elastasa Pancreática/metabolismo , Péptidos/química , Péptidos/farmacología , Estructura Secundaria de Proteína , Proteolisis/efectos de los fármacos , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/metabolismo , Staphylococcus aureus/ultraestructura
7.
Biochim Biophys Acta ; 1838(5): 1225-34, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24522010

RESUMEN

Host defense peptides are key components of the innate immune system, providing multi-facetted responses to invading pathogens. Here, we describe that the peptide GKS26 (GKSRIQRLNILNAKFAFNLYRVLKDQ), corresponding to the A domain of heparin cofactor II (HCII), ameliorates experimental septic shock. The peptide displays antimicrobial effects through direct membrane disruption, also at physiological salt concentration and in the presence of plasma and serum. Biophysical investigations of model lipid membranes showed the antimicrobial action of GKS26 to be mirrored by peptide incorporation into, and disordering of, bacterial lipid membranes. GKS26 furthermore binds extensively to bacterial lipopolysaccharide (LPS), as well as its endotoxic lipid A moiety, and displays potent anti-inflammatory effects, both in vitro and in vivo. Thus, for mice challenged with ip injection of LPS, GKS26 suppresses pro-inflammatory cytokines, reduces vascular leakage and infiltration in lung tissue, and normalizes coagulation. Together, these findings suggest that GKS26 may be of interest for further investigations as therapeutic against severe infections and septic shock.


Asunto(s)
Antiinflamatorios/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Endotoxinas/antagonistas & inhibidores , Cofactor II de Heparina/farmacología , Inflamación/tratamiento farmacológico , Animales , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Lípidos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Choque Séptico/tratamiento farmacológico , Choque Séptico/metabolismo
8.
Biochim Biophys Acta ; 1828(11): 2709-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23806651

RESUMEN

Lipid membrane and lipopolysaccharide (LPS) interactions were investigated for a series of amphiphilic and cationic peptides derived from human heparin cofactor II (HCII), using dual polarization interferometry, ellipsometry, circular dichroism (CD), cryoTEM, and z-potential measurements. Antimicrobial effects of these peptides were compared to their ability to disorder bacterial lipid membranes, while their capacity to block endotoxic effects of LPS was correlated to the binding of these peptides to LPS and its lipid A moiety, and to charge, secondary structure, and morphology of peptide/LPS complexes. While the peptide KYE28 (KYEITTIHNLFRKLTHRLFRRNFGYTLR) displayed potent antimicrobial and anti-endotoxic effects, its truncated variants KYE21 (KYEITTIHNLFRKLTHRLFRR) and NLF20 (NLFRKLTHRLFRRNFGYTLR) provide some clues on structure-activity relations, since KYE21 retains both the antimicrobial and anti-endotoxic effects of KYE28 (although both attenuated), while NLF20 retains the antimicrobial but only a fraction of the anti-endotoxic effect, hence locating the anti-endotoxic effects of KYE28 to its N-terminus. The antimicrobial effect, on the other hand, is primarily located at the C-terminus of KYE28. While displaying quite different endotoxic effects, these peptides bind to a similar extent to both LPS and lipid A, and also induce comparable LPS scavenging on model eukaryotic membranes. In contrast, fragmentation and densification of LPS aggregates, in turn dependent on the secondary structure in the peptide/LPS aggregates, correlate to the anti-endotoxic effect of these peptides, thus identifying peptide-induced packing transitions in LPS aggregates as key for anti-endotoxic functionality. This aspect therefore needs to be taken into account in the development of novel anti-endotoxic peptide therapeutics.


Asunto(s)
Endotoxinas/antagonistas & inhibidores , Cofactor II de Heparina/farmacología , Lipopolisacáridos/metabolismo , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular , Dicroismo Circular , Cofactor II de Heparina/química , Humanos , Ratones , Datos de Secuencia Molecular , Péptidos/química , Espectrometría de Fluorescencia , Relación Estructura-Actividad
9.
J Biol Chem ; 287(41): 34256-63, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22904320

RESUMEN

We previously clarified that heparin cofactor II (HCII), a serine proteinase inhibitor, exerts various protective actions on cardiovascular diseases in both experimental and clinical studies. In the present study, we aimed to clarify whether HCII participates in the regulation of angiogenesis. Male heterozygous HCII-deficient (HCII(+/-)) mice and male littermate wild-type (HCII(+/+)) mice at the age of 12-16 weeks were subjected to unilateral hindlimb ligation surgery. Laser speckle blood flow analysis showed that blood flow recovery in response to hindlimb ischemia was delayed in HCII(+/-) mice compared with that in HCII(+/+) mice. Capillary number, arteriole number, and endothelial nitric-oxide synthase (eNOS), AMP-activated protein kinase (AMPK), and liver kinase B1 (LKB1) phosphorylation in ischemic muscles were decreased in HCII(+/-) mice. Human purified HCII (h-HCII) administration almost restored blood flow recovery, capillary density, and arteriole number as well as phosphorylation levels of eNOS, AMPK, and LKB1 in ischemic muscles of HCII(+/-) mice. Although treatment with h-HCII increased phosphorylation levels of eNOS, AMPK, and LKB1 in human aortic endothelial cells (HAECs), the h-HCII-induced eNOS phosphorylation was abolished by compound C, an AMPK inhibitor, and by AMPK siRNA. In a similar fashion, tube formation, proliferation, and migration of HAECs were also promoted by h-HCII treatment and were abrogated by pretreatment with compound C. HCII potentiates the activation of vascular endothelial cells and the promotion of angiogenesis in response to hindlimb ischemia via an AMPK-eNOS signaling pathway. These findings suggest that HCII is a novel therapeutic target for treatment of patients with peripheral circulation insufficiency.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cofactor II de Heparina/metabolismo , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transducción de Señal/fisiología , Animales , Aorta/citología , Aorta/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Cofactor II de Heparina/farmacología , Humanos , Masculino , Ratones , Ratones Mutantes , Neovascularización Fisiológica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Comp Biochem Physiol B Biochem Mol Biol ; 156(3): 206-15, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20363356

RESUMEN

The dermatan sulfate (DS) isolated from the ray skin Raja montagui was identified and characterized. Its average molecular weight (Mw) and sulfate content were 39 kDa and 25% w/w, respectively. This DS prolonged thrombin time and activated partial thromboplastin time and inhibited the thrombin generation in a concentration-dependent manner whereas it had no effect on the anti-Xa assay and on platelet function. Data from the anti-IIa assay allowed the assessment of the specific anticoagulant activity which was 40 units/mg. The kinetics of the thrombin inhibition by heparin cofactor II (HCII) has been studied as a function of DS concentration according to a kinetic model in which the polysaccharide binds quickly to the inhibitor and forms a complex more reactive than the free inhibitor towards thrombin. This DS accelerated thrombin inhibition exclusively by HCII. The dissociation constant of the DS-HCII complex, K(DSHCII), and the rate constant of the thrombin inhibition by this complex, k, were (2.93+/-0.25)x10(-6)M and (2.2+/-0.35)x10(9)M(-1)min(-1), respectively. Our findings indicated that the major polysaccharide in the skin of the ray Raja montagui was a DS endowed with a high anticoagulant effect mediated by HCII and which may constitute an anticoagulant drug of interest in anticoagulant therapy.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Dermatán Sulfato/química , Dermatán Sulfato/farmacología , Rajidae , Animales , Anticoagulantes/aislamiento & purificación , Dermatán Sulfato/aislamiento & purificación , Inhibidores del Factor Xa , Cofactor II de Heparina/farmacología , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Protrombina/antagonistas & inhibidores , Piel/química , Sulfatos/análisis , Trombina/metabolismo
11.
Thromb Res ; 123(6): 902-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19046760

RESUMEN

INTRODUCTION: The kinetics of the thrombin inhibition by heparin cofactor II (HCII) and antithrombin (AT) have been studied as a function of the concentration of a dermatan sulfate (DS) from the skin of the ray Raja radula. MATERIALS AND METHODS: The initial concentrations of inhibitor (I), HCII or AT, and thrombin (E) were set at equimolecular levels (3.10(-9) M). Analysis of the experimental data obtained for DS concentrations ranging from 10(-8) to 10(-4) M was performed according to a previously described model in which DS binds quickly to the inhibitor and forms a complex more reactive than the free inhibitor towards thrombin. RESULTS: The apparent rate constant of the thrombin inhibition, k(app), by either HCII or AT, increased in a concentration-dependent manner for DS concentrations up to 10(-5) M or 10(-6) M, respectively. At higher DS concentrations, k(app) remained unchanged for thrombin inhibition by HCII whereas a decrease in k(app) was observed for the thrombin-AT reaction. The dissociation constant of the polysaccharide-inhibitor complex, K(DSI), and the rate constant of the thrombin inhibition by this complex, k, were (7.81+/-0.75).10(-7) M and (2.84+/-0.42).10(9) M(-1).min(-1), whereas they were (4.93+/-0.31).10(-7) M and (2.47+/-0.28).10(8) M(-1).min(-1), when the inhibitor was either HCII or AT, respectively. CONCLUSION: DS from ray skin catalyzes the thrombin inhibition by HCII or AT primarily by forming a DS-inhibitor complex more reactive than the free inhibitor towards the protease. The affinity of DS for HCII was approximately 2-fold higher whereas the catalyzed reaction rate constant was approximately 20-fold higher when compared to AT.


Asunto(s)
Antitrombinas/farmacología , Dermatán Sulfato/farmacología , Fibrinolíticos/química , Fibrinolíticos/farmacología , Cofactor II de Heparina/farmacología , Rajidae/metabolismo , Trombina/antagonistas & inhibidores , Animales , Coagulación Sanguínea/efectos de los fármacos , Dermatán Sulfato/aislamiento & purificación , Fibrinolíticos/aislamiento & purificación , Humanos , Técnicas In Vitro , Cinética , Piel/química
12.
Blood ; 111(8): 4118-25, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18281504

RESUMEN

Heparin cofactor II (HCII)-deficient mice form occlusive thrombi more rapidly than do wild-type mice following injury to the carotid arterial endothelium. Dermatan sulfate (DS) and heparan sulfate (HS) increase the rate of inhibition of thrombin by HCII in vitro, but it is unknown whether vascular glycosaminoglycans play a role in the antithrombotic effect of HCII in vivo. In this study, we found that intravenous injection of either wild-type recombinant HCII or a variant with low affinity for HS (K173H) corrected the abnormally short thrombosis time of HCII-deficient mice, while a variant with low affinity for DS (R189H) had no effect. When HCII was incubated with frozen sections of the mouse carotid artery, it bound specifically to DS in the adventitia. HCII was undetectable in the wall of the uninjured carotid artery, but it became concentrated in the adventitia following endothelial injury. These results support the hypothesis that HCII interacts with DS in the vessel wall after disruption of the endothelium and that this interaction regulates thrombus formation in vivo.


Asunto(s)
Arterias Carótidas/metabolismo , Dermatán Sulfato/metabolismo , Fibrinolíticos/metabolismo , Cofactor II de Heparina/metabolismo , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Arterias Carótidas/efectos de la radiación , Condroitín Liasas/metabolismo , Femenino , Cofactor II de Heparina/deficiencia , Cofactor II de Heparina/farmacocinética , Cofactor II de Heparina/farmacología , Heparitina Sulfato/metabolismo , Humanos , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mutantes/farmacología , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/efectos de la radiación , Porcinos , Trombina/antagonistas & inhibidores , Trombosis/patología , Factores de Tiempo
13.
J Thromb Haemost ; 5(11): 2219-26, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17958740

RESUMEN

BACKGROUND: In a previous study, we found that thrombin induced proliferation of TM-1 and T98G human glioma cells and that the mitogenic effect was abolished by hirudin. OBJECTIVES: We investigated thrombin's effects on the proliferation of A172 human glioblastoma cells and the induction of growth factors. Furthermore, we examined whether or not the expression of heparin cofactor II (HCII) in A172 cells using adenovirus vector could suppress thrombin's effects. METHODS: The effect of thrombin on cell proliferation was assessed using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay. The amount of growth factors in the conditioned medium was measured by enzyme-linked immunosorbent assay. The level of platelet-derived growth factor (PDGF)-B mRNA was assessed by reverse transcriptase-polymerase chain reaction analysis. RESULTS: Thrombin-induced proliferation of A172 cells primarily depended on the enhanced secretion of PDGF-AB by thrombin. The action of thrombin depended on its proteolytic activity. However, thrombin-induced PDGF-AB secretion was not abolished by anti-protease-activated receptor (PAR) antibody. The PAR-1 agonist peptide had no effect on cell growth and PDGF-AB levels. Thrombin did not increase PDGF-B gene expression. Expression of HCII effectively suppressed thrombin-induced PDGF-AB release. CONCLUSIONS: These results indicate that thrombin may play an important role in the proliferation of A172 cells by inducing PDGF-AB secretion and that thrombin's action is mediated by its proteolytic activity. Inhibition of thrombin's proteolytic activity may be a new therapeutic method for gliomas.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glioblastoma/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Trombina/farmacología , Línea Celular Tumoral , Medios de Cultivo Condicionados/química , Glioblastoma/metabolismo , Cofactor II de Heparina/administración & dosificación , Cofactor II de Heparina/farmacología , Humanos , Proteínas Proto-Oncogénicas c-sis/análisis , Proteínas Proto-Oncogénicas c-sis/genética , ARN Mensajero/análisis
14.
Blood Coagul Fibrinolysis ; 18(3): 227-36, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17413758

RESUMEN

Endogenous heparinoids impair coagulation, evidenced by thrombelastography in cirrhotic patients with bacterial infection, but it is not clear which glycosaminoglycans can be detected by native and heparinase-modified thrombelastography. To assess the effects of different glycosaminoglycans on thrombelastography parameters and the reversibility of these effects by heparinase-I-modified thrombelastography. Twenty volunteers were enrolled. Solutions of heparan sulphate, dermatan sulphate, and chondroitin-4-sulphate were prepared at 'equivalent' concentrations, based on the composition and anticoagulant activity of danaparoid. Serial dilutions of each glycosaminoglycan were prepared to achieve 1.0, 0.5, 0.1, and 0.05 U/ml. Native and heparinase-modified thrombelastography, anti-activated factor X activity and heparin cofactor II activity were evaluated at each concentration. A statistically significant heparin-like effect was seen with 1 and 0.5 U/ml heparan sulphate, and 1 and 0.5 U/ml dermatan sulphate, which was completely reversed by heparinase-modified thrombelastography. Anti-activated factor X activity was significantly increased in samples containing heparan and dermatan sulphates. The heparin cofactor II activity decreased with 1.0 and 0.5 U/ml dermatan sulphate and chondroitin-4-sulphate, but not with heparan sulphate. Heparan and dermatan sulphates affect haemostasis when added to whole blood in vitro, detectable by native thrombelastography and completely reversed by heparinase-I-modified thrombelastography. They may therefore be responsible for the heparin-like effect seen by thrombelastography in patients with cirrhosis and bacterial infection.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Glicosaminoglicanos/farmacología , Tromboelastografía/métodos , Infecciones Bacterianas/sangre , Sulfatos de Condroitina/farmacología , Dermatán Sulfato/farmacología , Fibrosis/sangre , Cofactor II de Heparina/farmacología , Liasa de Heparina/farmacología , Humanos
15.
Carbohydr Res ; 340(12): 2015-23, 2005 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16023626

RESUMEN

Marine red algae are an abundant source of sulfated galactans with potent anticoagulant activity. However, the specific structural motifs that confer biological activity remain to be elucidated. We have now isolated and purified a sulfated galactan from the marine red alga, Gellidium crinale. The structure of this polysaccharide was determined using NMR spectroscopy. It is composed of the repeating structure -4-alpha-Galp-(1-->3)-beta-Galp1--> but with a variable sulfation pattern. Clearly 15% of the total alpha-units are 2,3-di-sulfated and another 55% are 2-sulfated. No evidence for the occurrence of 3,6-anhydro alpha-galactose units was observed in the NMR spectra. We also compared the anticoagulant activity of this sulfated galactan with a polysaccharide from the species, Botryocladia occidentalis, with a similar saccharide chain but with higher amounts of 2,3-di-sulfated alpha-units. The sulfated galactan from G. crinale has a lower anticoagulant activity on a clotting assay when compared with the polysaccharide from B. occidentalis. When tested in assays using specific proteases and coagulation inhibitors, these two galactans showed significant differences in their activity. They do not differ in thrombin inhibition mediated by antithrombin, but in assays where heparin cofactor II replaces antithrombin, the sulfated galactan from G. crinale requires a significantly higher concentration to achieve the same inhibitory effect as the polysaccharide from B. occidentalis. In contrast, when factor Xa instead of thrombin is used as the target protease, the sulfated galactan from G. crinale is a more potent anticoagulant. These observations suggest that the proportion and/or the distribution of 2,3-di-sulfated alpha-units along the galactan chain may be a critical structural motif to promote the interaction of the protease with specific protease and coagulation inhibitors.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Galactanos/química , Galactanos/farmacología , Rhodophyta/química , Secuencia de Carbohidratos , Inhibidores del Factor Xa , Cofactor II de Heparina/farmacología , Humanos , Resonancia Magnética Nuclear Biomolecular , Tiempo de Tromboplastina Parcial , Trombina/antagonistas & inhibidores
16.
Thromb Res ; 116(4): 357-63, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16038721

RESUMEN

Heparin cofactor II functions as a physiological inhibitor of thrombin activity. The rate of inactivation of thrombin by heparin cofactor II is increased in the presence of dermatan sulfate, which is produced by fibroblasts or smooth muscle cells. To elucidate the role of heparin cofactor II in the extravascular cells, we induced expression of heparin cofactor II in cultured human fibroblasts or vascular smooth muscle cells using adenovirus-mediated gene transfer. After infection of adenovirus vector, these cells secreted heparin cofactor II protein into culture medium. The expressed heparin cofactor II formed the complex with exogenous thrombin and inhibited the proteolytic activity of thrombin. Expression of heparin cofactor II by infection of adenovirus vector inhibited thrombin-induced tissue-type plasminogen activator and interleukin-6 releases from fibroblasts and thrombin-induced interleukin-6 release from vascular smooth muscle cells. These findings show that fibroblasts and vascular smooth muscle cells expressing heparin cofactor II are resistant to thrombin-induced cellular responses.


Asunto(s)
Adenoviridae/genética , Fibroblastos/efectos de los fármacos , Cofactor II de Heparina/genética , Cofactor II de Heparina/farmacología , Músculo Liso Vascular/efectos de los fármacos , Trombina/antagonistas & inhibidores , Células Cultivadas , Cofactor II de Heparina/administración & dosificación , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Trombina/farmacología , Activador de Tejido Plasminógeno/antagonistas & inhibidores , Activador de Tejido Plasminógeno/metabolismo , Transducción Genética
17.
Biochem J ; 372(Pt 3): 747-55, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12656676

RESUMEN

Negatively charged organochemical inactivators of the anti-proteolytic activity of plasminogen activator inhibitor-1 (PAI-1) convert it to inactive polymers. As investigated by native gel electrophoresis, the size of the PAI-1 polymers ranged from dimers to multimers of more than 20 units. As compared with native PAI-1, the polymers exhibited an increased resistance to temperature-induced unfolding. Polymerization was associated with specific changes in patterns of digestion with non-target proteases. During incubation with urokinase-type plasminogen activator, the polymers were slowly converted to reactive centre-cleaved monomers, indicating substrate behaviour of the terminal PAI-1 molecules in the polymers. A quadruple mutant of PAI-1 with a retarded rate of latency transition also had a retarded rate of polymerization. Studying a number of serpins by native gel electrophoresis, ligand-induced polymerization was observed only with PAI-1 and heparin cofactor II, which were also able to copolymerize. On the basis of these results, we suggest that the binding of ligands in a specific region of PAI-1 leads to so-called loop-sheet polymerization, in which the reactive centre loop of one molecule binds to beta-sheet A in another molecule. Induction of serpin polymerization by small organochemical ligands is a novel finding and is of protein chemical interest in relation to pathological protein polymerization in general.


Asunto(s)
Biopolímeros/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Biopolímeros/química , Electroforesis en Gel de Poliacrilamida , Cofactor II de Heparina/farmacología , Calor , Humanos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Papaína/metabolismo , Inhibidor 1 de Activador Plasminogénico/química , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serpinas/metabolismo , Espectrometría de Fluorescencia/métodos , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
18.
Carbohydr Res ; 337(21-23): 2231-8, 2002 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-12433487

RESUMEN

Marine alga is an abundant source of sulfated polysaccharides with potent anticoagulant activity. However, several attempts to identify the specific structural features in these compounds, which confer the biological activity, failed due to their complex, heterogeneous structure. We isolated and characterized several sulfated alpha-L-galactans and sulfated alpha-L-fucans from marine invertebrates. In contrast to the algal fucans and galactans, these invertebrate polysaccharides have a simple structure, composed of well-defined units of oligosaccharides. We employed two of these compounds to elucidate their structure-anticoagulant action relationship. Our results indicate that a 2-sulfated, 3-linked alpha-L-galactan, but not an alpha-L-fucan, is a potent thrombin inhibitor mediated by antithrombin or heparin cofactor II. The difference between the activities of these two polysaccharides is not very pronounced when factor Xa replaces thrombin. Thus, the anticoagulant activity of sulfated galactan and sulfated fucan is not merely a consequence of their charge density. The interaction of these polysaccharides with coagulation cofactors and their target proteases are specific. Identification of specific structural requirements in sulfated galactans and sulfated fucans necessary for interaction with coagulation cofactors is an essential step for a more rational approach to develop new anticoagulant and antithrombotic drugs.


Asunto(s)
Anticoagulantes/aislamiento & purificación , Galactanos/aislamiento & purificación , Galactanos/farmacología , Animales , Anticoagulantes/química , Anticoagulantes/farmacología , Antitrombina III/farmacología , Pruebas de Coagulación Sanguínea , Interacciones Farmacológicas , Inhibidores del Factor Xa , Galactanos/química , Cofactor II de Heparina/farmacología , Humanos , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Polisacáridos/farmacología , Erizos de Mar/química , Relación Estructura-Actividad , Ésteres del Ácido Sulfúrico/química , Ésteres del Ácido Sulfúrico/aislamiento & purificación , Ésteres del Ácido Sulfúrico/farmacología , Trombina/antagonistas & inhibidores
19.
Thromb Res ; 107(1-2): 67-73, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12413592

RESUMEN

Antithrombin (ATIII), heparin cofactor II (HCII) and protein C inhibitor (PCI; also named plasminogen activator inhibitor-3) are serine protease inhibitors (serpins) whose thrombin inhibition activity is accelerated in the presence of glycosaminoglycans. We compared the inhibition properties of PCI and HCII to ATIII using R93A/R97A/R101A thrombin, an anion-binding exosite-2 (exosite-2) mutant that has greatly reduced heparin-binding properties. Heparin-enhanced PCI inhibition of R93A/R97A/R101A thrombin was only approximately 2-fold compared to 40-fold enhancement with wild-type recombinant thrombin. Thrombomodulin (TM) (with or without the chondroitin sulfate moiety) accelerated PCI inhibition of both wild-type and R93A/R97A/R101A thrombins. HCII achieved the same maximum activity in the presence of heparin with both wild-type and R93A/R97A/R101A thrombins; however, the optimum heparin concentration was 20 times greater than the reaction with wild-type thrombin, indicative of a decrease in heparin affinity. Dermatan sulfate (DSO4)-catalyzed HCII thrombin inhibition was unchanged in R93A/R97A/R101A thrombin compared to wild-type recombinant thrombin. These results suggest that PCI is similar to ATIII and depends upon ternary complex formation with heparin and these specific thrombin exosite-2 residues to accelerate thrombin inhibition. In contrast, HCII does not require Arg(93), Arg(97) and Arg(101) of thrombin exosite-2 and further supports the hypothesis that HCII uses an allosteric process following glycosaminoglycan binding to inhibit thrombin.


Asunto(s)
Cofactor II de Heparina/farmacología , Mutación , Inhibidor de Proteína C/farmacología , Trombina/antagonistas & inhibidores , Regulación Alostérica , Secuencia de Aminoácidos , Sitios de Unión , Glicosaminoglicanos/metabolismo , Cofactor II de Heparina/química , Humanos , Inhibidor de Proteína C/química , Trombina/química , Trombina/genética
20.
Blood ; 100(4): 1347-53, 2002 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12149217

RESUMEN

In a patient who presented with a severe coagulation deficiency in plasma contrasting with a very mild hemorrhagic diathesis a homozygous Arg67His mutation was identified in the prothrombin gene. Wild-type (factor IIa [FIIa]-WT) and mutant Arg67His thrombin (FIIa-MT67) had similar amidolytic activity. By contrast, the k(cat)/K(m) value of fibrinopeptide A hydrolysis by FIIa-WT and FIIa-MT67 was equal to 2.1 x 10(7) M(-1)s(-1) and 9 x 10(5) M(-1)s(-1). Decreased activation of protein C (PC) correlated with the 33-fold decreased binding affinity for thrombomodulin (TM; K(d) = 65.3 nM vs 2.1 nM, in FIIa-MT67 and in FIIa-WT, respectively). In contrast, hydrolysis of PC in the absence of TM was normal. The Arg67His mutation had a dramatic effect on the cleavage of protease-activated G protein-coupled receptor 1 (PAR-1) 38-60 peptide (k(cat/)K(m) = 4 x 10(7) M(-1)s(-1) to 1.2 x 10(6) M(-1)s(-1)). FIIa-MT67 showed a weaker platelet activating capacity, attributed to a defective PAR-1 interaction, whereas the interaction with glycoprotein Ib was normal. A drastic decrease (up to 500-fold) of the second-order rate constant pertaining to heparin cofactor II (HCII) interaction, especially in the presence of dermatan sulfate, was found for the FIIa-MT67 compared with FIIa-WT, suggesting a severe impairment of thrombin inhibition by HCII in vivo. Finally, the Arg67His mutation was associated with a 5-fold decrease of prothrombin activation by the factor Xa-factor Va complex, perhaps through impairment of the prothrombin-factor Va interaction. These experiments show that the Arg67His substitution affects drastically both the procoagulant and the anticoagulant functions of thrombin as well as its inhibition by HCII. The mild hemorrhagic phenotype might be explained by abnormalities that ultimately counterbalance each other.


Asunto(s)
Arginina , Trastornos de la Coagulación Sanguínea/genética , Trastornos Hemorrágicos/genética , Histidina , Mutación , Protrombina/genética , Línea Celular , Consanguinidad , Factor Va/metabolismo , Factor Xa/metabolismo , Femenino , Fibrinopéptido A/metabolismo , Cofactor II de Heparina/farmacología , Homocigoto , Humanos , Hidrólisis , Lactante , Mutagénesis Sitio-Dirigida , Proteína C/metabolismo , Protrombina/metabolismo , Receptor PAR-1 , Receptores de Trombina/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Tromboplastina/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...