Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
J Clin Invest ; 75(2): 496-501, 1985 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3838317

RESUMO

Heparin cofactor II is a plasma protein that inhibits thrombin rapidly in the presence of either heparin or dermatan sulfate. We have determined the effects of two glycosaminoglycan-binding proteins, i.e., histidine-rich glycoprotein and platelet factor 4, on these reactions. Inhibition of thrombin by heparin cofactor II and heparin was completely prevented by purified histidine-rich glycoprotein at the ratio of 13 micrograms histidine-rich glycoprotein/microgram heparin. In contrast, histidine-rich glycoprotein had no effect on inhibition of thrombin by heparin cofactor II and dermatan sulfate at ratios of less than or equal to 128 micrograms histidine-rich glycoprotein/microgram dermatan sulfate. Removal of 85-90% of the histidine-rich glycoprotein from plasma resulted in a fourfold reduction in the amount of heparin required to prolong the thrombin clotting time from 14 s to greater than 180 s but had no effect on the amount of dermatan sulfate required for similar anti-coagulant activity. In contrast to histidine-rich glycoprotein, purified platelet factor 4 prevented inhibition of thrombin by heparin cofactor II in the presence of either heparin or dermatan sulfate at the ratio of 2 micrograms platelet factor 4/micrograms glycosaminoglycan. Furthermore, the supernatant medium from platelets treated with arachidonic acid to cause secretion of platelet factor 4 prevented inhibition of thrombin by heparin cofactor II in the presence of heparin or dermatan sulfate. We conclude that histidine-rich glycoprotein and platelet factor 4 can regulate the antithrombin activity of heparin cofactor II.


Assuntos
Glicoproteínas/metabolismo , Fator Plaquetário 4/fisiologia , Proteínas/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Dermatan Sulfato/farmacologia , Heparina/metabolismo , Heparina/farmacologia , Cofator II da Heparina , Humanos , Técnicas In Vitro , Proteínas/metabolismo
2.
J Clin Invest ; 55(6): 1259-68, 1975 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1169259

RESUMO

Monovalent goat antibody fragments (Fab) that were monospecific for human fibrinogen were isolated by affinity chromatography on fibrinogen-Sepharose and used as a direct probe for the involvement of fibrinogen in platelet aggregation and the release reaction. The antifibrinogen Fab inhibited aggregation of washed human platelets induced by thrombin (0.1-10 U/ml) by 50-95%, but had no effect on (14-C)-serotinin release and only a slight inhibitory effect on 125-I-thrombin binding to platelets. Inhibition of aggregation was not observed with nonimmune goat Fab or rabbit antihuman albumie bound tightly at saturation to surface fibrinogen molecules. After washing the platelets once to remove unbound Fab, aggregation by subsequently added thrombin was no longer inhibited. The antifibrinogen Fab inhibited the clotting of fibrinogen by thrombin but did not effect the rate of fibrinopeptide A release, indicating that the Fab inhibits clotting by interfering with the polymerization of fibrin monomers. Our experiments suggest that fibrinogen released from platelets is directly involved in thrombin-induced aggregation of washed platelets, perhaps through polymerization of fibrin monomers generated by proteolytic cleavage of released fibrinogen.


Assuntos
Fibrinogênio/fisiologia , Fragmentos Fab das Imunoglobulinas , Adesividade Plaquetária , Agregação Plaquetária , Trombina/fisiologia , Animais , Radioisótopos de Carbono , Cabras/imunologia , Humanos , Radioisótopos do Iodo , Coelhos/imunologia , Serotonina/fisiologia , Trombina/farmacologia
3.
J Clin Invest ; 68(3): 589-96, 1981 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6168653

RESUMO

We have demonstrated that human plasma contains a heparin-dependent inhibitor of thrombin that is distinguishable from antithrombin III (AT III). When a 1:50 dilution of plasma was incubated with greater than or equal to 0.01 U/ml heparin and 1 U/ml 125I-thrombin, the labeled thrombin B-chains became incorporated into two complexes of Mr-96,000 and Mr-85,000 that were separated by polyacrylamide gel electrophoresis in the presence of sodium dodecyl sulfate and beta-mercaptoethanol. Neither complex was detectable at heparin concentrations less than 0.01 U/ml. When a limiting amount of 125I-thrombin was present, the proportion of radioactivity incorporated into each of the two complexes varied with the heparin concentration. Thus, the Mr-85,000 complex predominated at 0.01-5 U/ml heparin, whereas the Mr-96,000 complex predominated at 5-100 U/ml heparin. The Mr-85,000 complex reacted with antibodies to human AT III and comigrated with the purified thrombin-AT III complex. The Mr-96,000 complex did not react with antibodies to AT III or to alpha 1-antitrypsin, and it was detected in normal quantities after incubating 125I-thrombin with plasma immunodepleted of AT III, alpha 2-antiplasmin, alpha 2-macroglobulin, C1 inactivator, alpha 1-antichymotrypsin, or inter-alpha-trypsin inhibitor. The protein that combines with thrombin to form the Mr-96,000 complex was estimated to be present at a minimum concentration of 90 +/- 26 micrograms/ml (mean +/- SD) in identical to any of the known plasma protease inhibitors and that at relatively high heparin concentrations in vitro it reacts with thrombin more rapidly than does AT III.


Assuntos
Antitrombinas , Heparina/farmacologia , Antitrombina III/análise , Antitrombinas/imunologia , Epitopos , Humanos , Peso Molecular
4.
J Clin Invest ; 81(1): 69-74, 1988 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3335644

RESUMO

The effect of administration of N-desulfated/acetylated heparin, almost completely devoid of anticoagulant activity, on the progression of renal disease was examined in rats with 13/4 nephrectomy. Three groups of rats with 13/4 nephrectomy were studied. Group I (control, n = 11) received 0.15 ml of 0.15 M NaCl subcutaneously twice daily for 5 wk; group 2 (n = 11) received 0.15 ml twice daily of N-desulfated/acetylated heparin (5.4 mg/ml; less than 0.5 U/ml); group 3 (n = 9) received 0.15 ml twice daily of standard beef lung heparin (5.4 mg/ml; 977 U/ml). Clearances and renal histological studies were done at the end of 5 wk of heparin or saline administration. Rats given the heparin preparations had significantly higher inulin clearances (2.55 +/- 0.38 ml/min per body weight (BW) for group 2, or 2.60 +/- 0.24 ml/min per kg BW for group 3) than control rats (1.59 +/- 0.20 ml/min per kg BW). Histological analysis revealed a greater number of glomeruli with segmental or global sclerosis, hyalinosis, or fibrosis (36.6%) in control rats than in rats receiving N-desulfated/acetylated heparin (6.2%) or standard heparin (3.0%). Blood pressure averaged 169.4 +/- 6.2 mmHg in controls, 119.1 +/- 6.1 in rats of group 2, and 124.3 +/- 2.5 in rats of group 3. The values for blood pressure were significantly lower in the two groups receiving heparin than in controls. These studies indicate that a heparin preparation, almost completely devoid of anticoagulant properties, affords the same degree of protection against progression of renal disease as does standard heparin in rats with subtotal renal ablation. It is suggested that other biological properties of heparin may be responsible for the effects observed.


Assuntos
Heparina/análogos & derivados , Nefropatias/fisiopatologia , Nefrectomia , Acetilação , Animais , Feminino , Heparina/farmacologia , Heparina/uso terapêutico , Nefropatias/sangue , Nefropatias/patologia , Testes de Função Renal , Glomérulos Renais/patologia , Tempo de Tromboplastina Parcial , Ratos , Ratos Endogâmicos
5.
J Clin Invest ; 53(1): 211-8, 1974 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-4357613

RESUMO

We have previously shown that the erythroagglutinating phytohemagglutinin (E-PHA) from Phaseolus vulgaris binds to the surface of intact human platelets and that adenylate cyclase activity in the particulate fraction of E-PHA-treated platelets is lower than in comparable controls. We now find that E-PHA induces release of [(14)C]serotonin from platelets. Release follows binding of E-PHA, and a haptenic inhibitor of E-PHA binding prevents induction of release. E-PHA does not produce platelet lysis and has little effect on [(14)C]serotonin uptake. Platelets possess approximately 300,000 receptor sites of E-PHA per cell, and we estimate that about 15% of these sites must be occupied by E-PHA to initiate the release reaction. Prior incubation of platelets with prostaglandin E(1), theophylline, or dibutyryl cyclic AMP prevents E-PHA-induced release, although these agents have little effect on E-PHA binding to platelets. Thrombin and E-PHA produce different rates and extents of serotonin release. Thrombin (1 U/ml) causes release of 75-85% of platelet [(14)C]-serotonin, with half-maximal release occurring less than 0.5 min after thrombin addition. E-PHA, however, induces release of only 30-60% of platelet serotonin at a 10-fold slower rate. In addition, utilizing electron microscopy, we have observed striking differences in the morphological changes that occur in platelets exposed to E-PHA as compared with thrombin. Thus, the platelet release reaction may be triggered in part by binding of E-PHA to the cell surface, but this reaction only partially resembles that produced by thrombin.


Assuntos
Plaquetas/efeitos dos fármacos , Lectinas/farmacologia , Plaquetas/metabolismo , Bucladesina/farmacologia , Radioisótopos de Carbono , Humanos , Técnicas In Vitro , Radioisótopos do Iodo , Microscopia Eletrônica , Prostaglandinas/farmacologia , Receptores de Droga , Serotonina/metabolismo , Teofilina/farmacologia , Trombina/farmacologia
6.
J Clin Invest ; 56(1): 241-5, 1975 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1141438

RESUMO

Binding of prothrombin and activation intermediates 1 and 2 to human platelets was tested with 125-I-labeled protein preparations. None of these precursors of thrombin bound to platelets under conditions in which high affinity binding of thrombin was observed,nor did they cause platelet aggregation or serotonin release. The molecular conformation required for binding to platelets as well as for induction of platelet aggregation and release is present, therefore, only after the final step in prothrombin activation.


Assuntos
Plaquetas/metabolismo , Protrombina/metabolismo , Sítios de Ligação , Plaquetas/efeitos dos fármacos , Cálcio/farmacologia , Radioisótopos de Carbono , Eletroforese em Gel de Poliacrilamida , Humanos , Técnicas In Vitro , Radioisótopos do Iodo , Conformação Molecular , Agregação Plaquetária , Ligação Proteica , Serotonina/metabolismo , Trombina/metabolismo
7.
Biochim Biophys Acta ; 1431(1): 148-56, 1999 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-10209287

RESUMO

A variety of sulphated polyanions in addition to heparin and dermatan sulphate stimulate the inhibition of thrombin by heparin cofactor II (HCII). Previous investigations indicated that the binding sites on HCII for heparin and dermatan sulphate overlap but are not identical. In this study we determined the concentrations (IC50) of various polyanions required to stimulate thrombin inhibition by native recombinant HCII in comparison with three recombinant HCII variants having decreased affinity for heparin (Lys-173-->Gln), dermatan sulphate (Arg-189-->His), or both heparin and dermatan sulphate (Lys-185-->Asn). Pentosan polysulphate, sulphated bis-lactobionic acid amide, and sulphated bis-maltobionic acid amide resembled dermatan sulphate, since their IC50 values were increased to a much greater degree (>/=8-fold) by the mutations Arg-189-->His and Lys-185-->Asn than by Lys-173-->Gln (Gln and Lys-185-->Asn (>/=6-fold) than by Arg-189-->His (

Assuntos
Aminoácidos/análise , Cofator II da Heparina/química , Polímeros/farmacologia , Trombina/antagonistas & inibidores , Sítios de Ligação , Cofator II da Heparina/biossíntese , Cofator II da Heparina/genética , Mutação , Polieletrólitos , Proteínas Recombinantes/química
8.
Thromb Haemost ; 80(5): 784-90, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9843172

RESUMO

Heparin cofactor II (HCII) is a serpin that inhibits thrombin rapidly in the presence of heparin or dermatan sulfate. HCII activity has been detected in human, rabbit, and mouse plasma, and cDNA clones for HCII have been isolated previously from human, rabbit, rat, and mouse liver libraries, suggesting a conserved physiologic role for HCII among mammals. In this report, we show that both frog and chicken plasma contain a dermatan sulfate-dependent inhibitor that forms a 118-kDa complex with human 125I-thrombin. Screening of frog and chicken liver cDNA libraries in bacteriophage lambda with a human HCII cDNA probe yielded nearly full-length clones with inserts of 1.8 and 1.7 kb, respectively. The amino acid sequences deduced from the frog and chicken HCII cDNAs are approximately 60% identical to one another and to each of the mammalian sequences. In particular, the N-terminal acidic domain, the glycosaminoglycan-binding site, and the reactive site sequences are highly conserved. Our results indicate that HCII is widely distributed among vertebrates and may have a common function in birds, amphibians, and mammals.


Assuntos
Galinhas/genética , DNA Complementar/genética , Cofator II da Heparina/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , DNA Complementar/isolamento & purificação , Dermatan Sulfato/farmacologia , Biblioteca Gênica , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/química , Fígado/química , Mamíferos/genética , Modelos Moleculares , Dados de Sequência Molecular , RNA Mensageiro/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Trombina/antagonistas & inibidores , Trombina/metabolismo
9.
Am J Clin Pathol ; 89(4): 515-7, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3354504

RESUMO

Hereditary deficiency of antithrombin-III (AT-III), the major heparin cofactor in human plasma, is a well-established cause of recurrent venous thrombosis. Cross-reactivity of heparin cofactor II (HC II) in assays of AT-III may, in some cases, interfere with the ability to diagnose hereditary deficiency of AT-III. For that reason, we have evaluated the interference by HC II in the new DuPont aca antithrombin assay. Response of the assay to purified AT-III and HC II was compared. Inhibition of the bovine thrombin in the assay was sixfold less per molecule of HC II than of AT-III. This level of selectivity should be adequate to prevent misdiagnosis of patients. Analysis of patient samples showed close correlation (r = 0.91) of values from the automated aca assay with those of a manual assay (Coatest antithrombin, Helena Laboratories).


Assuntos
Antitrombina III/análise , Antitrombinas/análise , Glicoproteínas/fisiologia , Kit de Reagentes para Diagnóstico , Deficiência de Antitrombina III , Reações Cruzadas , Erros de Diagnóstico , Cofator II da Heparina , Humanos
10.
Ann N Y Acad Sci ; 714: 21-31, 1994 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-8017769

RESUMO

The binding sites for dermatan sulfate and heparin in HCII overlap but are not identical. This may explain the observation that HCII binds nonspecifically to heparin oligosaccharides, but preferentially binds to a minor hexasaccharide isolated from dermatan sulfate. The tissue distribution of dermatan sulfate molecules containing the high-affinity HCII binding site may regulate HCII activity in vivo. Finally, in the presence of dermatan sulfate or heparin, the N-terminal acidic region of HCII may interact with the hirudin-binding site of thrombin to produce maximal stimulation of the thrombin-HCII reaction.


Assuntos
Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/química , Cofator II da Heparina/metabolismo , Conformação Proteica , Trombina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Configuração de Carboidratos , Sequência de Carboidratos , Dermatan Sulfato/química , Humanos , Dados de Sequência Molecular , Oligossacarídeos/química , Oligossacarídeos/isolamento & purificação , Oligossacarídeos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Trombina/química , Trombina/isolamento & purificação , Difração de Raios X
11.
Thromb Res ; 100(5): 443-51, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11150588

RESUMO

The rate of thrombin inhibition by heparin cofactor II (HCII) is facilitated by heparin or dermatan sulfate in vitro. The distributions of these glycosaminoglycans (GAGs) in vivo are not the same; heparin-like substance is rich on the surface of endothelial cells and dermatan sulfate is relatively dominant in the extravascular region. When inflammation takes place, at least two other possible existent forms of HCII, the complexed form with thrombin and the cleaved form by leukocyte elastase, are assumed to be present at relatively high concentrations in a local circumstance. We examined the interactions of HCII with the two forms of HCII on thrombin inhibition in the presence of the GAGs. By HCII in complex with thrombin or cleaved by leukocyte elastase, the affinity of HCII moiety for heparin increases and that for dermatan sulfate decreases. The two forms possibly occur at relatively high concentrations in a local pathological situation, although the heparin cofactor activity for thrombin inhibition by HCII decreases and dermatan sulfate determines the cofactor activity. These results indicate efficient thrombin inhibitory activity of HCII in the extravascular region.


Assuntos
Cofator II da Heparina/metabolismo , Cofator II da Heparina/farmacologia , Inibidores de Serina Proteinase/metabolismo , Inibidores de Serina Proteinase/farmacologia , Trombina/antagonistas & inibidores , Trombina/metabolismo , Humanos , Elastase Pancreática/metabolismo
12.
Adv Exp Med Biol ; 313: 167-76, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1442260

RESUMO

The binding sites for dermatan sulfate and heparin in HCII overlap but are not identical. This may explain the observation that HCII binds non-specifically to heparin oligosaccharides but preferentially binds to a minor hexasaccharide isolated from dermatan sulfate having the structure shown in Fig. 4B. The tissue distribution of dermatan sulfate molecules containing the high-affinity HCII binding site may regulate HCII activity in vivo. Finally, in the presence of dermatan sulfate or heparin, the N-terminal acidic domain of HCII may interact with the hirudin-binding site of thrombin to produce maximal stimulation of the thrombin-HCII reaction.


Assuntos
Dermatan Sulfato/metabolismo , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Sequência de Carboidratos , Dermatan Sulfato/química , Cofator II da Heparina/química , Cofator II da Heparina/genética , Humanos , Modelos Químicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Trombina/antagonistas & inibidores
13.
J Thromb Haemost ; 9(2): 350-60, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21138526

RESUMO

BACKGROUND: We found a high incidence of thrombotic deaths in COX-1(+/-)COX-2(-/-) mice and sought to define the mechanism of these events. The cyclooxygenase products thromboxane A(2) and prostacyclin are important in the regulation of coagulation but their role in fibrinolysis is largely unexplored. PAI-1 blocks fibrinolysis by inhibiting plasminogen activator. AIM: Our objective was to explain the mechanism of increased thrombosis associated with the COX-1(+/-)COX-2(-/-) genotype. METHODS: Carotid artery occlusion times were measured after photochemical injury. PAI-1 levels were measured in the plasma by ELISA. PAI-1 levels in the aorta were measured by RT-PCR and Western blotting. Urinary metabolites of Thromboxane A(2) and prostacyclin were measured by ELISA. RESULTS: The COX-1(+/-)COX-2(-/-) genotype is associated with a decreased time to occlusion in the carotid artery thrombosis model (30 ± 5 minutes vs 60 ± minutes in wild type, p<.001). The COX-1(-/-)COX-2(+/+), COX-1(+/-)COX-2(+/-) and COX-1(+/-)COX-2(+/+) all had occlusion times similar to wild type. COX-1(+/+)COX-2(-/-) had a prolonged occlusion time. COX-1(+/-)COX-2(-/-) had increased PAI-1 levels in the plasma and aorta and with a prolonged euglobulin lysis time (37.4 ± 10.2 hours vs 15.6 ± 9.8 hours in wild type, p<.004). The decreased time to occlusion in the COX-1(+/-)COX2(-/-) mice was normalized by an inhibitory antibody to PAI-1 whereas the antibody had no effect on the time to occlusion in wild type mice. CONCLUSION: The COX-1(+/-)COX-2(-/-) genotype is associated with a shortened time to occlusion in the carotid thrombosis model and the shortened time to occlusion is mediated through increased PAI-1 levels resulting in decreased fibrinolysis.


Assuntos
Estenose das Carótidas/genética , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Animais , Estenose das Carótidas/metabolismo , Primers do DNA , Modelos Animais de Doenças , Feminino , Fibrinólise , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Semin Thromb Hemost ; 16(2): 162-8, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2191430

RESUMO

Antithrombin activity should probably be determined in persons with early onset of recurrent thrombosis, especially if a family history is present. The initial screening test should be a heparin cofactor assay optimized to reduce the contribution of heparin cofactor II. If the heparin cofactor assay is low, an antigenic determination should be performed to rule out the possibility of an antithrombin variant. At the present time, there is insufficient evidence to recommend the routine determination of heparin cofactor II levels in patients with thrombosis.


Assuntos
Antitrombinas/deficiência , Cofator II da Heparina/deficiência , Antitrombinas/análise , Antitrombinas/genética , Deleção Cromossômica , Cofator II da Heparina/análise , Cofator II da Heparina/genética , Humanos
18.
Nouv Rev Fr Hematol (1978) ; 26(4): 233-7, 1984.
Artigo em Inglês | MEDLINE | ID: mdl-6548014

RESUMO

Heparin cofactor II is a glycoprotein present in human plasma at a concentration of approximately 1.2 microM. It inhibits thrombin by forming a stable, 1:1 complex with the protease. The rate of complex formation is increased approximately 1,000-fold by heparin or dermatan sulfate. Heparin cofactor II appears to be the only thrombin inhibitor in plasma that can be activated by dermatan sulfate. Platelet factor 4 abolishes the activation of heparin cofactor II by dermatan sulfate, but plasma histidine-rich glycoprotein does not. Heparin cofactor II is activated by dermatan sulfate oligosaccharides that are at least 12-14 sugar residues in length and contain a high-affinity binding site for the inhibitor protein.


Assuntos
Condroitina/análogos & derivados , Dermatan Sulfato/farmacologia , Glicoproteínas/metabolismo , Heparina/farmacologia , Dermatan Sulfato/metabolismo , Glicoproteínas/farmacologia , Antagonistas de Heparina/farmacologia , Cofator II da Heparina , Histidina/farmacologia , Humanos , Cinética , Peso Molecular , Fator Plaquetário 4/farmacologia
19.
J Biol Chem ; 265(30): 18263-71, 1990 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-2211700

RESUMO

Dermatan sulfate increases the rate of inhibition of thrombin by heparin cofactor II (HCII) approximately 1000-fold by providing a catalytic template to which both the inhibitor and the protease bind. Dermatan sulfate is a linear polymer of D-glucuronic acid (GlcA) or L-iduronic acid (IdoA) alternating with N-acetyl-D-galactosamine (GalNAc) residues. Heterogeneity in dermatan sulfate results from varying degrees of O-sulfation and from the presence of the two types of uronic acid residues. To characterize the HCII-binding site in dermatan sulfate, we isolated the smallest fragment of dermatan sulfate that bound to HCII with high affinity. Dermatan sulfate was partially N-deacetylated by hydrazinolysis, cleaved with nitrous acid at pH 4, and reduced with [3H]NaBH4. The resulting fragments, containing an even number of monosaccharide units with the reducing terminal GalNAc converted to [3H]2,5-anhydro-D-talitol (ATalR), were size-fractionated and then chromatographed on an HCII-Sepharose column. The smallest HCII-binding fragments were hexasaccharides, of which approximately 6% bound. Based on ion-exchange chromatography, the bound material appeared to comprise a heterogeneous mixture of molecules possessing four, five, or six sulfate groups per hexasaccharide. Subsequently, hexasaccharides with the highest affinity for HCII were isolated by overloading the HCII-Sepharose column. The high-affinity hexasaccharides were fractionated by strong anion-exchange chromatography, and one major peak representing approximately 2% of the starting hexasaccharides was isolated. The high-affinity hexasaccharide was cleaved to disaccharides that were analyzed by anion-exchange chromatography, paper electrophoresis, and paper chromatography. A single disulfated disaccharide, IdoA(2-SO4)----ATalR(4-SO4) was observed, indicating that the hexasaccharide has the following structure: IdoA(2-SO4)----GalNAc(4-SO4)----IdoA(2-SO4)---- GalNAc(4-SO4)----IdoA(2-SO4)----ATalR(4-SO4). Since IdoA(2-SO4)----GalNAc(4-SO4) comprises only approximately 5% of the disaccharides present in intact dermatan sulfate, clustering of these disaccharides must occur during biosynthesis to form the high-affinity binding site for HCII.


Assuntos
Dermatan Sulfato/química , Cofator II da Heparina/metabolismo , Oligossacarídeos/química , Animais , Sequência de Carboidratos , Cromatografia em Gel , Cromatografia por Troca Iônica , Eletroforese , Dados de Sequência Molecular , Suínos , Trombina/antagonistas & inibidores
20.
J Biol Chem ; 262(1): 169-75, 1987 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-3793724

RESUMO

Inhibition of thrombin by heparin cofactor II (HCII) is accelerated by dermatan sulfate, heparan sulfate, and heparin. Purified HCII or defibrinated plasma was incubated with washed confluent cell monolayers, 125I-thrombin was added, and the rate of formation of covalent 125I-thrombin-inhibitor complexes was determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and autoradiography. Fibroblasts and porcine aortic smooth muscle cells accelerated inhibition of thrombin by HCII 2.3-7.5-fold but had no effect on other thrombin inhibitors in plasma. Human umbilical vein endothelial cells and mouse macrophage-derived cells did not accelerate the thrombin-HCII reaction. IMR-90 normal human fetal lung fibroblasts treated with heparinase or heparitinase accelerated the thrombin-HCII reaction to the same degree as untreated cells. In contrast, treatment with chondroitinase ABC almost totally abolished the ability of these cells to activate HCII while chondroitinase AC had little or no effect, suggesting that dermatan sulfate was responsible for the activity observed. [35S]Sulfate-labeled proteoglycans were isolated from IMR-90 fibroblast monolayers and conditioned medium and fractionated into two peaks on Sepharose CL-2B. The lower Mr proteoglycans contained 74-76% dermatan sulfate and were 11-25 times more active with HCII than the higher Mr proteoglycans which contained 68-97% heparan sulfate. The activity of the lower Mr proteoglycans decreased 70-90% by degradation of the dermatan sulfate component with chondroitinase ABC. These results confirm that dermatan sulfate proteoglycans are primarily responsible for activation of HCII by IMR-90 fibroblasts. We suggest that HCII may inhibit thrombin when plasma is exposed to vascular smooth muscle cells or fibroblasts.


Assuntos
Fibroblastos/fisiologia , Glicoproteínas/metabolismo , Músculo Liso Vascular/fisiologia , Animais , Antitrombina III/metabolismo , Linhagem Celular , Condroitina Liases/farmacologia , Cricetinae , Meios de Cultura , Dermatan Sulfato/farmacologia , Cofator II da Heparina , Humanos , Camundongos , Proteoglicanas/farmacologia , Suínos , Trombina/antagonistas & inibidores , Trombina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA