Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Toxicon ; 58(1): 123-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21641921

ABSTRACT

Disintegrins and disintegrins-like proteins are able to inhibit platelet aggregation and integrin-mediated cell adhesion. The aim of this study was to produce one disintegrin-like cloned from Bothrops leucurus venom gland and to characterize it regarding biological activity. The recombinant protein was purified by one step procedure involving anion-exchange chromatography (DEAE-cellulose) and presented a molecular mass of 10.4 kDa. The purified protein was able to inhibit platelet aggregation induced by collagen (IC50 = 0.65 µM) and to inhibit growth of Ehrlich tumor implanted in mice by more than 50% after 7 days administration of 10 µg/day. No effects were observed upon adenosine 5'-diphosphate (ADP)-and arachidonic acid (AA)-induced platelet aggregation. The recombinant protein was recognized by an antibody specific for jararhagin one metalloproteinase isolated from Bothrops jararaca venom, and therefore it was named leucurogin. Anti-angiogenesis effect of leucurogin was evaluated by the sponge implant model. After 7 days administration leucurogin inhibited, in a dose dependent way, the vascularization process in the sponge. Leucurogin represents a new biotechnological tool to understand biological processes where disintegrins-like are involved and may help to characterize integrins that can be involved in development and progression of malignant cells.


Subject(s)
Bothrops/metabolism , Carcinoma, Ehrlich Tumor/drug therapy , Disintegrins/pharmacology , Recombinant Proteins/pharmacology , Amino Acid Sequence , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/isolation & purification , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Bothrops/genetics , Cloning, Molecular , Crotalid Venoms , Disintegrins/chemistry , Disintegrins/genetics , Disintegrins/isolation & purification , Male , Metalloendopeptidases , Mice , Molecular Sequence Data , Neovascularization, Physiologic/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/isolation & purification , Platelet Aggregation Inhibitors/pharmacology , Recombinant Proteins/metabolism , Sequence Alignment , Bothrops jararaca Venom
2.
Genes Immun ; 11(8): 637-48, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861859

ABSTRACT

The natural killer gene complex (NKC) on chromosome 6 contains clusters of genes that encode both activation and inhibitory receptors expressed on mouse natural killer (NK) cells. NKC genes, particularly belonging to the Nkrp1 and Ly49 gene families, display haplotype differences between different mouse strains and allelic polymorphisms of individual genes, as previously revealed by conventional analysis in a small number of inbred mouse strains. Herein we used array-based comparative genomic hybridization (aCGH) to efficiently compare the NKC in 21 mouse strains to the reference C57BL/6 strain. By using unsupervised clustering methods, we could sort these variations into the same groups as determined by previous RFLP analyses of Nkrp1 and Ly49 genes. Prospective analyses of aCGH and RFLP data validated these relationships. Moreover, aCGH data predicted monoclonal antibody reactivity with an allospecific determinant on molecules expressed by NK cells. Taken together, these data demonstrate the structural variation in the NKC between mouse strains as well as the usefulness of aCGH in analysis of complex, polymorphic gene clusters.


Subject(s)
Comparative Genomic Hybridization , Killer Cells, Natural/immunology , Multigene Family/genetics , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily B/genetics , Alleles , Animals , Antigens, CD/genetics , Chromosome Mapping , Haplotypes , Immunity, Cellular/genetics , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Polymorphism, Restriction Fragment Length , Species Specificity
3.
Biochimie ; 89(3): 319-28, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17110015

ABSTRACT

Two proteins with phospholipase A(2) (PLA(2)) activity were purified to homogeneity from Bothrops leucurus (white-tailed-jararaca) snake venom through three chromatographic steps: Conventional gel filtration on Sephacryl S-200, ion-exchange on Q-Sepharose and reverse phase on Vydac C4 HPLC column. The molecular mass for both enzymes was estimated to be approximately 14 kDa by SDS-PAGE. The N-terminal sequences (48 residues) show that one enzyme presents lysine at position 48 and the other an aspartic acid in this position, and therefore they were designated blK-PLA(2) and blD-PLA(2) respectively. blK-PLA(2) presented negligible levels of PLA(2) activity as compared to that of blD-PLA(2). The PLA(2) activity of both enzymes is Ca(2+)-dependent. blD-PLA(2) did not have any effect upon platelet aggregation induced by arachidonic acid, ADP or collagen, but strongly inhibits coagulation and is able to stimulate Ehrlich tumor growth but not angiogenesis.


Subject(s)
Bothrops/metabolism , Phospholipases A/metabolism , Snake Venoms/enzymology , Amino Acid Sequence , Animals , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Calcium/metabolism , Carcinoma, Ehrlich Tumor/chemically induced , Carcinoma, Ehrlich Tumor/enzymology , Carcinoma, Ehrlich Tumor/pathology , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Enzyme-Linked Immunosorbent Assay , Hemoglobins/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Isoenzymes/metabolism , K562 Cells , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Molecular Weight , Phospholipases A/chemistry , Phospholipases A/isolation & purification , Platelet Aggregation/drug effects , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Snake Venoms/pharmacology
4.
Surg Endosc ; 15(9): 1004-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11605112

ABSTRACT

BACKGROUND: Sphincter of Oddi dysfunction (SOD) is one of the causes of postcholecytectomy syndrome and biliary pain. Endoscopic sphincterotomy (EST) is recommended in some cases for patients refractory to conservative treatment. By the Milwaukee classification, patients with biliary pain can be divided into three groups. Group I patients show all the objective signs suggestive of a disturbed bile outflow-i.e., elevated liver function tests, dilated common bile duct (CBD), and delayed contrast drainage during endoscopic retrograde cholangio pancreatography (ERCP). Group II patients have biliary-type pain along with one or two of the criteria from group I. Group III patients have only biliary pain, with no other abnormalities. This study confirms the effectiveness of EST for the relief of symptoms in group I patients (papillary stenosis). METHODS: Between 1989 and 1999, we treated eight patients clinically diagnosed as having group I papillary stenosis by EST. Their ages ranged from 52 to 73 years. In addition to biliary pain, all patients were found to have dilated CBD, elevated enzyme levels, and delayed contrast drainage at ERCP. None of the patients had CBD stones or other causes of obstruction. Sphincter of Oddi manometry was not performed. RESULTS: EST was successfully performed in eight patients. Each patient had a very large papilla. A false orifice was found in one patient. In five patients, endoscopic cannulation of the bile duct was very difficult. The use of a long, tapered catheter and guidewire papillotomy was necessary in four patients. A precut papillotomy was performed in one patient. All patients achieved resolution of their symptoms after EST. There were no complications. The average length of the follow-up period was 26 months. CONCLUSIONS: SOD is a real entity that continues to pose a diagnostic dilemma. EST is an effective and safe modality for the treatment of papillary stenosis (group I patients). SOD manometry is not necessary before EST in group I patients.


Subject(s)
Common Bile Duct Diseases/surgery , Sphincter of Oddi/surgery , Sphincterotomy, Endoscopic/methods , Abdominal Pain/diagnosis , Abdominal Pain/surgery , Aged , Cholangiopancreatography, Endoscopic Retrograde , Common Bile Duct Diseases/diagnosis , Constriction, Pathologic/surgery , Female , Humans , Male , Middle Aged , Treatment Outcome
5.
Kekkaku ; 76(5): 419-26, 2001 May.
Article in Japanese | MEDLINE | ID: mdl-11449697

ABSTRACT

Forty isolates of rapidly growing Mycobacteria, Mycobacterium fortuitum group including M. fortuitum and M. peregrinum and M. chelonae group including M. chelonae subsp. chelonae and M. chelonae subsp. abscessus at Showa University Fujigaoka Hospital collected between February 1981 and December 1997 were investigated in this study. These isolates were from the patients who were not infected with HIV. The average age of fourteen patients, from whom M. fortuitum group was isolated, was 58 years, ranging from 17 to 80 years old. One patient (71-year-old) with chronic myelogenous leukemia and another (64-year-old) with chronic diabetes mellitus were diagnosed with skin abscesses of M. fortuitum group, which were located on the right site of the neck and in the scar after injecting insulin (injection abscess), respectively. The average age of twenty-six patients, from whom M. chelonae group was isolated, was 57 years, ranging from 32 to 84 years old. One patient (75-year-old) with articular rheumatism was diagnosed with a lung infection of mixed M. chelonae group and Pseudomonas aeruginosa, and another (74-year-old) with diabetes mellitus and kidney failure was strongly suspected of a lung infection. The isolates of the two mycobacteria from the remaining patients were due to colonization, while these patients had the following underlying diseases contributing to infections: pulmonary emphysema; diabetes mellitus; leukemia; collagen diseases; lung cancer; chronic kidney diseases; systemic lupus erythematosus; carcinomatous pleurisy; bronchiectasis; post-tuberculosis. Most isolates of the two mycobacteria were separated from the specimens of patients' respiratory tracts, but since M. chelonae group was a contaminant in the tap-water for diluting concentrated chlorhexidine, the organism happened to be isolated with the mucous membranes of the 6 patients' colons that were picked up while using the washed fiber-scope. These findings suggest that M. fortuitum and M. chelonae groups, in spite of the fact that they rarely cause infection, have a significant risk of infecting aged patients in general hospitals with various underlying diseases attributable to infections. As only a few colonies were isolated from patients' specimens in the majority of cases, it took time to carry out these clinical examinations, and to improve this "laboratory's delay", it is needed to make faster report to clinicians.


Subject(s)
Hospitals, General , Laboratories, Hospital , Mycobacterium/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Inpatients , Male , Middle Aged , Mycobacterium Infections/epidemiology , Mycobacterium Infections/microbiology , Outpatients
7.
Nihon Shokakibyo Gakkai Zasshi ; 95(9): 1001-7, 1998 Sep.
Article in Japanese | MEDLINE | ID: mdl-9785889

ABSTRACT

We have treated 19 HBV carriers who developed acute severe exacerbation using interferon and immunosuppressive agents. Of these 14 patients developed fulminant hepatic failure. Of 10 patients with positive result for serum HBV DNA polymerase before the start of te treatment, five patients in whom HBV DNA polymerase turned negative and one patient whose HBV DNA polymerase level fluctuated in a low abnormal range after the start of the treatment survived. While, four patients whose HBV DNA polymerase level remained high after the start of interferon treatment died. Thus, it is suggested that suppression of HBV virus replication is closely related to prognosis in HB carriers developing acute severe exacervation of hepatitis.


Subject(s)
Carrier State/therapy , Hepatitis B/therapy , Interferons/therapeutic use , Acute Disease , Adolescent , Adult , Aged , Anti-Inflammatory Agents/administration & dosage , Carrier State/virology , Cyclosporine/administration & dosage , Drug Therapy, Combination , Female , Hepatic Encephalopathy/etiology , Hepatitis B/virology , Humans , Male , Middle Aged , Prednisolone/administration & dosage , Prognosis
8.
Biochem J ; 327 ( Pt 2): 577-83, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9359432

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a primary regulator of the initiation of blood coagulation. TFPI is internalized and degraded by HepG2 cells through the low-density-lipoprotein receptor-related protein (LRP) but also binds another molecule present on the cell surface at approx. 10-fold the abundance of LRP [Warshawsky, Broze and Schwartz (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 6664-6668]. When HepG2 cells are washed with heparin or dextran sulphate, a substance that binds TFPI is removed from the cell surface and can be detected in a slot-blot assay. Preincubation with trypsin destroys the reactivity of the TFPI-binding component in the slot-blot assay, suggesting that it is a protein. In addition, when the sulphation of glycosaminoglycans (GAGs) is prevented by growing the HepG2 cells in the presence of 30 mM sodium chlorate, TFPI binding is unaffected, whereas the binding of bovine lipoprotein lipase, a protein known to associate with cell-surface GAGs, falls to 50% of control levels. Dextran sulphate washes of HepG2 cells grown in sodium chlorate have an equal reactivity in slot-blot experiments to that of non-treated cells, suggesting that GAGs are not totally responsible for the binding activity observed. By using the slot blot to follow binding activity and conventional protein purification techniques, a protein species that migrates at 40 kDa after reduction was identified in the HepG2 cell wash. The binding of this protein to TFPI was confirmed with immobilized TFPI. Amino acid sequence analysis identified this protein species as a proteolytic fragment of glypican-3 (also called OCI-5), a member of the glypican family of glycosylphosphatidylinositol-anchored proteoglycans.


Subject(s)
Glycosaminoglycans/metabolism , Heparan Sulfate Proteoglycans , Heparitin Sulfate/metabolism , Lipoproteins/metabolism , Proteoglycans/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/metabolism , Cattle , Cell Membrane/metabolism , Chlorates/pharmacology , Chromatography, Affinity , Cloning, Molecular , Dextran Sulfate/pharmacology , Fibrinolytic Agents/metabolism , Glycosylphosphatidylinositols/metabolism , Glypicans , Heparin/pharmacology , Heparitin Sulfate/chemistry , Heparitin Sulfate/isolation & purification , Humans , Kinetics , Lipoprotein Lipase/metabolism , Molecular Sequence Data , Peptide Fragments/chemistry , Proteoglycans/chemistry , Proteoglycans/isolation & purification , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Trypsin/pharmacology , Tumor Cells, Cultured
9.
Blood ; 90(3): 944-51, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9242522

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a multivalent Kunitz-type proteinase inhibitor that directly inhibits factor Xa and, in a factor Xa-dependent fashion, produces feedback inhibition of the factor VIIa/TF catalytic complex responsible for the initiation of coagulation. To further define the physiologic role of TFPI, gene-targeting techniques were used to disrupt exon 4 of the TFPI gene in mice. This exon encodes Kunitz domain-1 of TFPI, which is required for factor VIIa/TF inhibition. In mice heterozygous for TFPI gene-disruption, TFPI(K1)(+/-), an altered form of TFPI lacking Kunitz domain-1, circulates in plasma at a concentration approximately 40% that of wild-type TFPI. TFPI(K1)(+/-) animals have plasma TFPI activity approximately 50% that of wild-type mice, based on a functional assay that measures factor VIIa/TF inhibition, and have a normal phenotype. Sixty percent of TFPI(K1)(-/-) mice die between embryonic days E9.5 and E11.5 with signs of yolk sac hemorrhage. The extent of structural abnormalities within the yolk sac vascular system appears to mirror the condition of the embryo, suggesting that the embryonic and extra-embryonic tissues are both responding to same insult, presumably circulatory insufficiency. Organogenesis is normal in TFPI(K1) null animals that progress beyond E11.5, but hemorrhage, particularly in the central nervous system and tail, is evident during later gestation and none of the TFPI(K1)(-/-) mice survive to the neonatal period. The presence of immunoreactive fibrin(ogen) in the liver and intravascular thrombi is consistent with the notion that unregulated factor VIIa/TF action and a consequent consumptive coagulopathy underlies the bleeding diathesis in these older embryos. Human TFPI-deficient embryos may suffer a similar fate because an individual with TFPI deficiency has not been identified.


Subject(s)
Abnormalities, Multiple/genetics , Fetal Death/genetics , Hemorrhagic Disorders/embryology , Lipoproteins/physiology , Abnormalities, Multiple/embryology , Animals , Blood Vessels/abnormalities , Blood Vessels/embryology , Central Nervous System/blood supply , Central Nervous System/embryology , Disseminated Intravascular Coagulation/embryology , Disseminated Intravascular Coagulation/genetics , Embryonic and Fetal Development , Exons/genetics , Factor VIIa/antagonists & inhibitors , Factor VIIa/physiology , Factor Xa/physiology , Female , Fetal Death/physiopathology , Fetal Diseases/embryology , Fetal Diseases/genetics , Genes, Lethal , Genotype , Gestational Age , Hemorrhagic Disorders/genetics , Humans , Lipoproteins/genetics , Male , Mice , Mice, Knockout , Morphogenesis , Tail/abnormalities , Tail/blood supply , Yolk Sac/abnormalities , Yolk Sac/blood supply
10.
Blood ; 88(10): 3815-23, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8916945

ABSTRACT

Coagulation is initiated by the binding of factor VIIa to tissue factor, with resultant limited factor IX and X activation and thrombin production. Owing to the feedback inhibition of the factor VIIa/tissue factor complex by tissue factor pathway inhibitor (TFPI), additional factor X activation and thrombin generation must proceed through a pathway involving factors VIII, IX, and XI. Experiments designed to elucidate the requirement for amplified factor Xa and thrombin generation in normal hemostasis show that the resistance of plasma clots to tissue plasminogen activator (tPA)- and urokinase-induced fibrinolysis is related to the extent of thrombin generation. Inhibition of fibrinolysis is mediated in part by plasma carboxypeptidase-U ([CPU] carboxypeptidase-R, procarboxypeptidase-B, thrombin-activatable fibrinolysis inhibitor), a proenzyme that is proteolytically activated by thrombin in a process enhanced dramatically by the cofactor thrombomodulin. A clot induced in factor IX-deficient plasma with limited amounts of tissue factor in the presence of urokinase (100 U/mL) lyses prematurely, and this defect is corrected by supplementation of the deficient plasma with factor IX (5 micrograms/mL) or thrombomodulin (20 ng/mL). These additions enhance the rate and extent of CPU activation: in the case of factor IX, presumably by permitting amplified generation of factor Xa and thrombin, and in the case of thrombomodulin, presumably by increasing the degree of CPU activation produced by the low levels of thrombin generated in the absence of factor IX. Pretreatment of the factor IX-deficient plasma with specific anti-CPU antibodies prevents the increased resistance to fibrinolysis produced by addition of factor IX and thrombomodulin. Likewise, when coagulation is induced by thrombin (2 U/mL) in the presence of tPA (60 U/mL), clots formed from plasmas deficient in factors VIII, IX, X, or XI lyse prematurely unless the missing factor is replaced or thrombomodulin (20 ng/mL) is added.


Subject(s)
Blood Coagulation Factors/physiology , Carboxypeptidases/physiology , Fibrinolysis/physiology , Hemophilia A/blood , Animals , Carboxypeptidase B2 , Carboxypeptidases/isolation & purification , Cations, Divalent/blood , Edetic Acid/pharmacology , Factor IX/pharmacology , Factor X Deficiency/blood , Hemophilia B/blood , Humans , Rabbits , Thrombin/metabolism , Thrombomodulin/physiology , Urokinase-Type Plasminogen Activator/pharmacology
11.
J Biochem ; 119(1): 29-36, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8907172

ABSTRACT

Bleomycin (BLM) hydrolase, which hydrolyzes the carboxyamide bond in the beta-amino-alanine moiety, was purified from newborn rat skin. The enzyme was purified 2,500-fold over the crude extract to apparent homogeneity in five steps in the presence of 2-mercaptoethanol: 45-55% ammonium sulfate fractionation, followed by chromatographies on Sephacryl S-200, DEAE-cellulofine, Phe-Superose, and Mono Q ion-exchange. The native enzyme had a molecular mass of 280 kDa according to gel filtration. The subunit molecular mass was estimated as 48 kDa by SDS-PAGE, indicating that the enzyme was comprised of six identical subunits. The amino acid sequence of its NH2-terminus was determined to be acetyl-Met-Asn-Asn-Ala-Gly-Leu-Asn-Ser-Glu-Lys-, which was not found in the amino acid sequence database. The optimum pH of the enzyme was 7.5 with pepleomycin (PLM). The Km and Vmax values were 2.1 mM and 6.8 mu mol center dot mg-1 center dot h-1 for PLM, and 1.8 mM and 7.2 mu mol center dot mg-1 center dot h-1 for BLM-A2, respectively. The enzyme activity was inhibited by iodoacetic acid, N-ethylmaleinimide (NEM), and p-chloromercuribenzoic acid (pCMB) as well as divalent cations such as Cu2+, Cd2+, Hg2+, and Zn2+. It was effectively inhibited by a cysteine protease inhibitor E-64. However, cystatins A and C did not inhibit the activity. BLM hydrolase exhibited broad aminopeptidase substrate specificity towards aminoacyl-beta-naphthylamides such as basic, neutral, and hydrophobic amino acid residues, as well as acidic residues. These results indicated that BLM hydrolase represents a new family of cysteine proteases. Western blotting and immunohistochemical analyses showed that BLM hydrolase is ubiquitous in various rat tissues but at low levels in lung and adult skin tissues, suggesting that this enzyme plays an important role in the metabolism of antibiotics.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Skin/enzymology , Amino Acid Sequence , Amino Acids/analysis , Amino Acids/metabolism , Animals , Animals, Newborn , Cysteine Endopeptidases/isolation & purification , Metals/pharmacology , Models, Chemical , Molecular Sequence Data , Rabbits , Rats , Rats, Sprague-Dawley , Skin/chemistry , Substrate Specificity , Tissue Distribution
12.
J Biol Chem ; 270(42): 24800-4, 1995 Oct 20.
Article in English | MEDLINE | ID: mdl-7559599

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a potent inhibitor of the blood coagulation factor VIIa-tissue factor complex, as well as a direct inhibitor of factor Xa. Intravenously administered TFPI is rapidly cleared from circulation predominantly via liver. We previously reported that the low density lipoprotein receptor-related protein (LRP), a multifunctional endocytic receptor, mediates the uptake and degradation of TFPI in hepatoma cells. This process is inhibited by a 39-kDa receptor-associated protein which binds to LRP and regulates its ligand binding activity. However, a distinct, low affinity binding site (perhaps heparin sulfate proteoglycans, HSPGs) on the endothelium and liver is thought to be responsible for the majority of TFPI cell surface binding. In the current study, we investigated the role of LRP and this second binding site in the clearance of 125I-TFPI in vivo using competitors and inhibitors of the receptors. Mice overexpressing the 39-kDa protein via adenoviral-mediated gene transfer displayed diminished plasma clearance of 125I-TFPI. Blockade of cell surface HSPGs sites by incubation with the positively charged molecule, protamine, inhibited 125I-TFPI binding to the hepatoma cells in vitro. In addition, preadministration of protamine in vivo prolonged the plasma clearance of 125I-TFPI in a dose-dependent manner. However, a dramatic increase of the plasma half-life of 125I-TFPI and virtual elimination of 125I-TFPI clearance was observed in mice overexpressing the 39-kDa protein and administered protamine. Taken together, our results suggest that two receptor mechanisms are involved in the clearance of TFPI in vivo.


Subject(s)
Anticoagulants/pharmacokinetics , Heparin/analogs & derivatives , Lipoproteins/pharmacokinetics , Proteoglycans/physiology , Receptors, Immunologic/physiology , Serine Proteinase Inhibitors/pharmacokinetics , Animals , Heparin/pharmacology , Heparin/physiology , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Protamines/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacokinetics , Tumor Cells, Cultured
15.
Blood ; 79(7): 1712-9, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1558967

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a multivalent Kunitz-type inhibitor that directly inhibits factor Xa and, in a factor Xa-dependent fashion, also inhibits the factor VIIa/tissue factor (TF) catalytic complex. The Kunitz-2 domain in TFPI is needed for the binding and inhibition of factor Xa, while the Kunitz-1 domain appears to be responsible for binding factor VIIa in a quaternary factor Xa-TFPI-factor VIIa/TF inhibitory complex. Human leukocyte elastase (HLE) proteolytically cleaves TFPI between threonine-87 and threonine-88 within the polypeptide that links the Kunitz-1 and Kunitz-2 domains in the TFPI molecule. HLE treatment not only affects the ability of TFPI to inhibit factor VIIa/TF, but also dramatically reduces its inhibition of factor Xa. Both purified HLE and stimulated neutrophils regenerate TF activity from a preformed factor Xa-TFPI-factor VIIa/TF inhibitory complex. Kinetic analysis suggests that HLE cleavage does not effect the affinity of the initial encounter interaction between factor Xa and TFPI, whereas it markedly reduces the affinity of the final factor Xa:TFPI complex with Ki (final) values for untreated and HLE-treated TFPI of 58 pmol/L and 4.4 nmol/L, respectively. Thus, an epitope in the amino-terminal region of TFPI or a conformation of the TFPI molecule that requires the presence of this region is needed in concert with the Kunitz-2 domain to produce optimal inhibition of factor Xa by TFPI.


Subject(s)
Factor VII/antagonists & inhibitors , Lipoproteins/metabolism , Pancreatic Elastase/metabolism , Thromboplastin/antagonists & inhibitors , Amino Acid Sequence , Factor VII/chemistry , Factor VII/metabolism , Factor VII/pharmacology , Factor VIIa/antagonists & inhibitors , Factor VIIa/metabolism , Factor Xa/metabolism , Factor Xa Inhibitors , Humans , Kinetics , Leukocyte Elastase , Lipoproteins/chemistry , Lipoproteins/pharmacology , Molecular Sequence Data , Molecular Weight , Neutrophils/metabolism , Protein Conformation , Thromboplastin/chemistry , Thromboplastin/metabolism , Thromboplastin/pharmacology
17.
J Dermatol ; 18(9): 545-8, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1787226

ABSTRACT

Keratotic erythematous lesions developed on the face, ears, neck, and upper back of a 38-year-old man. The patient had a history of photosensitivity to sunlight manifested by the development of new lesions. One week after an MED (2.7 mJ/cm2) was determined using UV-B light, persistent erythema with fine scales were observed at test sites irradiated over 72 mJ/cm2. These lasted for at least 2 months. A biopsy specimen obtained from the test site irradiated at 72 mJ/cm2 showed liquefaction degeneration of the basal cell layer. Within the upper part of the dermis, perivascular and disseminated lymphocytic infiltrates were seen. However, when irradiated again under the same conditions 7 and 12 months after the initial phototest, no persistent erythema was observed.


Subject(s)
Lupus Erythematosus, Discoid/pathology , Photosensitivity Disorders/pathology , Ultraviolet Rays/adverse effects , Adult , Animals , Biopsy , Erythema/etiology , Humans , Keratosis/etiology , Keratosis/pathology , Male , Photosensitivity Disorders/etiology
18.
Science ; 248(4959): 1126-8, 1990 Jun 01.
Article in English | MEDLINE | ID: mdl-2111585

ABSTRACT

An inhibitor of coagulation factor XIa was purified from serum-free conditioned medium of HepG2 liver cells. Platelets stimulated with thrombin or calcium ionophore (A23187) secrete a protein functionally and immunologically identical to the inhibitor, implying a role for this inhibitor in hemostasis. Analysis of the amino-terminal amino acid sequence and immunologic reactivity showed the inhibitor to be a truncated form of the Alzheimer's amyloid precursor protein that contains a Kunitz-type serine protease inhibitor domain and at least a portion of the amyloid beta protein. It inhibits factor XIa and trypsin with a Ki of 450 +/- 50 pM and 20 +/- 10 pM, respectively. Heparin (1 unit/ml) did not significantly effect inhibition of trypsin, but inhibition of XIa was 15 times greater (Ki = 25 +/- 15 pM) in the presence of heparin.


Subject(s)
Blood Platelets/analysis , Blood Proteins/isolation & purification , Factor XIa/antagonists & inhibitors , Serine Proteinase Inhibitors/isolation & purification , Amino Acid Sequence , Amyloid/immunology , Amyloid beta-Protein Precursor , Blood Proteins/immunology , Blood Proteins/physiology , Cross Reactions , Humans , Molecular Sequence Data , Protein Precursors/immunology , Serine Proteinase Inhibitors/blood
19.
Blood ; 71(2): 335-43, 1988 Feb.
Article in English | MEDLINE | ID: mdl-3422166

ABSTRACT

Blood coagulation is initiated when plasma factor VII(a) binds to its essential cofactor tissue factor (TF) and proteolytically activates factors X and IX. Progressive inhibition of TF activity occurs upon its addition to plasma. This process is reversible and requires the presence of VII(a), catalytically active Xa, Ca2+, and another component that appears to be associated with the lipoproteins in plasma, a lipoprotein-associated coagulation inhibitor (LACI). A protein, LACI(HG2), possessing the same inhibitory properties as LACI, has recently been isolated from the conditioned media of cultured human liver cells (HepG2). Rabbit antisera raised against a synthetic peptide based on the N-terminal sequence of LACI(HG2) and purified IgG from a rabbit immunized with intact LACI(HG2) inhibit the LACI activity in human serum. In a reaction mixture containing VIIa, Xa, Ca2+, and purified LACI(HG2), the apparent half-life (t1/2) for TF activity was 20 seconds. The presence of heparin accelerated the initial rate of inhibition threefold. Antithrombin III alpha alone had no effect, but antithrombin III alpha with heparin abrogated the TF inhibition. LACI(HG2) also inhibited Xa with an apparent t1/2 of 50 seconds. Heparin enhanced the rate of Xa inhibition 2.5-fold, whereas phospholipids and Ca2+ slowed the reaction 2.5-fold. Xa inhibition was demonstrable with both chromogenic substrate (S-2222) and bioassays, but no complex between Xa and LACI(HG2) could be visualized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Nondenaturing PAGE, however, showed that LACI(HG2) bound to Xa but not to X or Xa inactivated by diisopropyl fluorophosphate. Thus, LACI(HG2) appears to bind to Xa at or near its active site. Bovine factor Xa lacking its gamma-carboxyglutamic acid-containing domain, BXa(-GD), through treatment with alpha-chymotrypsin, was used to further investigate the Xa requirement for VIIa/TF inhibition by LACI(HG2). LACI(HG2) bound to BXa(-GD) and inhibited its catalytic activity against a small molecular substrate (Spectrozyme Xa), though at a rate approximately sevenfold slower than native BXa. Preincubation of LACI(HG2) with saturating concentrations of BXa(-GD) markedly retarded the subsequent inhibition of BXa. The VII(a)/TF complex was not inhibited by LACI(HG2) in the presence of BXa(-GD), and further, preincubation of LACI(HG2) with BXa(-GD) slowed the inhibition of VIIa/TF after the addition of native Xa. The results are consistent with the hypothesis that inhibition of VII(a)/TF involves the formation of a VIIa-TF-XA-LACI complex that requires the GD of XA.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Anticoagulants , Blood Coagulation , Factor VII/antagonists & inhibitors , Lipoproteins , Neoplasm Proteins/pharmacology , Serine Proteinase Inhibitors , Calcium/physiology , Factor Xa , In Vitro Techniques , Peptide Fragments/antagonists & inhibitors , Protein Binding , Structure-Activity Relationship , Thrombin/antagonists & inhibitors , Thromboplastin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...