Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Haematol ; 111(1): 91-5, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11091186

ABSTRACT

Factor XI (FXI) is a plasma glycoprotein produced by the liver that plays an essential role in blood coagulation. Individuals deficient in this protein are prone to bleeding following trauma or surgery. Well-washed platelets possess FXI-like activity and FXI antigen that differs in molecular weight from plasma FXI and is associated with FXI mRNA from platelets in which exon V is absent. Some individuals deficient in plasma FXI produce the platelet form of the protein. The molecular basis for the presence of platelet FXI in plasma FXI-deficient patients is unknown. In the current study, to help determine the mechanism for this differential expression, the FXI genotype was determined for three plasma FXI-deficient individuals who express platelet FXI. All three individuals had a nonsense mutation encoding a stop codon in exon V of the FXI gene that would result in a severely truncated polypeptide. An exon V nonsense mutation in the FXI gene combined with the absence of exon V in platelet FXI mRNA provides a plausible mechanism for the differential expression of platelet FXI in plasma FXI-deficient patients.


Subject(s)
Blood Platelets/metabolism , Factor XI Deficiency/genetics , Factor XI/genetics , Gene Expression Regulation , Mutation, Missense , Exons , Factor XI/metabolism , Factor XI Deficiency/blood , Female , Genotype , Humans , Sequence Analysis, DNA
2.
J Biol Chem ; 275(24): 18085-92, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10749870

ABSTRACT

Biglycan and decorin are small dermatan sulfate-containing proteoglycans in the extracellular matrix of the artery wall. The dermatan sulfate chains are known to stimulate thrombin inhibition by heparin cofactor II (HCII), a plasma proteinase inhibitor that has been detected within the artery wall. The purpose of this study was to analyze the HCII-stimulatory activity of biglycan and decorin isolated from normal human aorta and atherosclerotic lesions type II through VI and to correlate activity with dermatan sulfate chain composition and structure. Biglycan and decorin from plaque exhibited a 24-75% and 38-79% loss of activity, respectively, in thrombin-HCII inhibition assays relative to proteoglycan from normal aorta. A significant negative linear relationship was observed between lesion severity and HCII stimulatory activity (r = 0.79, biglycan; r = 0.63, decorin; p < 0.05). Biglycan, but not decorin, from atherosclerotic plaque contained significantly reduced amounts of iduronic acid and disulfated disaccharides DeltaDi-2,4S and DeltaDi-4,6S relative to proteoglycan from normal artery. Affinity coelectrophoresis analysis of a subset of samples demonstrated that increased interaction of proteoglycan with HCII in agarose gels paralleled increased activity in thrombin-HCII inhibition assays. In conclusion, both biglycan and decorin from atherosclerotic plaque possessed reduced activity with HCII, but only biglycan demonstrated a correlation between activity and specific glycosaminoglycan structural features. Loss of the ability of biglycan and decorin in atherosclerotic lesions to regulate thrombin activity through HCII may be critical in the progression of the disease.


Subject(s)
Arteriosclerosis/pathology , Dermatan Sulfate/chemistry , Heparin Cofactor II/metabolism , Proteoglycans/metabolism , Aorta/chemistry , Biglycan , Cells, Cultured , Chromatography, Gel , Decorin , Extracellular Matrix Proteins , Glycosaminoglycans/chemistry , Humans , Protein Conformation , Structure-Activity Relationship
3.
Arterioscler Thromb Vasc Biol ; 16(9): 1138-46, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8792767

ABSTRACT

Heparin cofactor II (HCII) is a potent thrombin inhibitor in the presence of heparin and dermatan sulfate, glycosaminoglycans that accelerate the inhibition reaction. HCII is postulated to be an extravascular thrombin inhibitor that is stimulated physiologically by dermatan sulfate proteoglycans. To understand how thrombin activity may be downregulated within the artery wall, cultured monkey aorta smooth muscle cell (SMC) proteoglycans were tested for their ability to accelerate thrombin inhibition by HCII. Early confluent SMC monolayers increased thrombin-HCII inhibition rates 2-fold to 4-fold compared with reactions in cell-free control wells (7.3 +/- 0.5 versus 2.7 +/- 0.2 x 10(4) mol.L-1.min-1, with and without SMCs, respectively; n = 7 experiments). Extracellular matrix obtained by cell monolayer removal also accelerated the thrombin-HCII inhibition reaction 3-fold to 5-fold. Rate increases were abolished by Polybrene or protamine sulfate. Pretreatment of monolayers with heparitinase I (and of extracellular matrix with HNO2) to degrade heparan sulfate blocked the thrombin-HCII inhibition rate increase. In contrast, pretreatment with chondroitinase ABC in the presence of proteinase inhibitors had no effect. "Pericellular" (cell surface- and extracellular matrix-derived) SMC heparan sulfate proteoglycans (HSPGs) were purified and fractionated by charge on DEAE-Sephacel. At a concentration of 1 microgram/mL hexuronic acid, high-charge HSPG stimulated a 7-fold thrombin-HCII inhibition rate increase relative to reactions without proteoglycan, whereas low-charge HSPG induced a 2-fold rate increase. In comparison, an 18-fold rate increase was observed with 1 microgram/mL dermatan sulfate proteoglycan purified from SMC culture media. These results indicate that SMC HSPG could contribute significantly to thrombin inhibition by HCII in the artery wall.


Subject(s)
Heparin Cofactor II/metabolism , Heparitin Sulfate/metabolism , Muscle, Smooth, Vascular/metabolism , Thrombin/metabolism , Animals , Aorta, Thoracic/metabolism , Cells, Cultured , Extracellular Matrix/metabolism , Heparin Cofactor II/pharmacology , Heparitin Sulfate/pharmacology , Macaca fascicularis , Thrombin/antagonists & inhibitors
4.
J Biol Chem ; 269(46): 28690-5, 1994 Nov 18.
Article in English | MEDLINE | ID: mdl-7961820

ABSTRACT

Protein C inhibitor (PCI) is a plasma serine proteinase inhibitor (serpin) that is a major physiological regulator of activated protein C. Inhibition of its target proteinase is accelerated by heparin in a reaction that involves the binding of both inhibitor and proteinase to heparin to form a ternary complex. This study was undertaken to understand the role of the H helix region (residues 264-278) of PCI in heparin binding and used (i) a recombinant truncated PCI fusion protein of the first 294 residues, (ii) H helix synthetic peptides containing single Arg/Lys-->Glu substitutions, and (iii) site-directed Ala mutagenesis of 4 basic residues (Arg-269, Lys-270, Lys-276, and Lys-277) in the H helix region of full-length recombinant PCI (rPCI) expressed in Baculovirus. The PCI fusion protein interfered in heparin-accelerated PCI-proteinase inhibition reactions, and it bound to heparin-Sepharose. Compared to the wild-type PCI fusion protein, deletion of the H helix from the fusion protein resulted in a reduction of both heparin-Sepharose binding and the ability to compete for heparin during PCI-proteinase inhibition reactions. Competition assays with H helix synthetic peptides revealed that the R269E altered peptide was the least effective at blocking heparin-catalyzed PCI-proteinase inhibition reactions. Compared with full-length active wild-type rPCI, R269A: K270A and K276A:K277A rPCI both had reduced heparin-Sepharose binding, but only R269A:K270A rPCI showed a loss of heparin-accelerated proteinase inhibition for both activated protein C and thrombin. We conclude that a major heparin-binding site of PCI is the H helix, unlike its heparin-binding serpin homologues antithrombin and heparin cofactor II, which bind heparin primarily through the D helix.


Subject(s)
Heparin/metabolism , Protein C Inhibitor/metabolism , Amino Acid Sequence , Baculoviridae/genetics , Base Sequence , Binding Sites , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Protein Binding , Protein C Inhibitor/chemistry , Protein C Inhibitor/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
5.
J Biol Chem ; 268(5): 3321-7, 1993 Feb 15.
Article in English | MEDLINE | ID: mdl-8429008

ABSTRACT

Heparin cofactor II and antithrombin are plasma serine proteinase inhibitors whose ability to inhibit alpha-thrombin is accelerated by glycosaminoglycans. Dysfunctional thrombin mutants Quick I (Arg67-->Cys) and Quick II (Gly226-->Val) were used to further compare heparin cofactor II and antithrombin interactions. Quick I, Quick II, and alpha-thrombin were eluted at the same salt concentration from heparin-Sepharose suggesting that the putative heparin-binding site (also termed anion binding exosite-II) is functional. Antithrombin yielded similar inhibition rates for Quick I and alpha-thrombin in the absence or presence of various amounts of heparin. Also, Quick I was inhibited similarly to alpha-thrombin by heparin cofactor II in the absence of glycosaminoglycan. In contrast, glycosaminoglycan-accelerated Quick I inhibition by heparin cofactor II was greatly reduced indicating that anion binding exosite-I (where the mutation occurs in Quick I) is critical for increased inhibition by heparin cofactor II. We also found that heparin cofactor II formed a SDS-resistant bimolecular complex with Quick II and alpha-thrombin at similar rates and the rate of complex formation was accelerated in the presence of glycosaminoglycans. A three-dimensional molecular model of the Quick II active site compared to alpha-thrombin suggested that the heparin cofactor II Leu-Ser-reactive site sequence (P1-P1') is a compatible "pseudosubstrate" in contrast to the Arg-Ser sequence found in antithrombin. The importance of heparin cofactor II as a thrombin regulator will depend upon its ability to interact with glycosaminoglycans and the functional availability of thrombin exosites.


Subject(s)
Antithrombins/pharmacology , Heparin Cofactor II/pharmacology , Mutation , Thrombin/antagonists & inhibitors , Amino Acid Sequence , Chromatography, Affinity , Heparin/metabolism , Humans , Kinetics , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Thrombin/chemistry , Thrombin/isolation & purification , Thrombin/metabolism
6.
Appl Opt ; 5(5): 863-7, 1966 May 01.
Article in English | MEDLINE | ID: mdl-20048961

ABSTRACT

The preparation and properties of a highly cross-linked dibutyl phthalate-modified methyl methacrylate polymer are described. The material is virtually optically isotropic with a refractive index near 1.5 and exhibits low absorption in the wavelength region 3000 A to 2 micro. It exhibits good dimensional stability when properly annealed and can be easily polished to close optical tolerances. It is unaffected by many organic and inorganic solvents. Careful preparation and annealing have removed gross density gradients and adventitious particle scattering in the medium.

SELECTION OF CITATIONS
SEARCH DETAIL
...