Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Thromb Haemost ; 4(8): 1813-24, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16879225

RESUMO

BACKGROUND: Thrombomodulin (TM) is predominantly a vascular endothelial cell plasma membrane glycoprotein that, via distinct structural domains, interacts with multiple ligands, thereby modulating coagulation, fibrinolysis, complement activation, inflammation and cell proliferation. We previously reported that by mediating signals that interfere with mitogen-activated protein kinase and nuclear factor kappaB pathways, the amino-terminal C-type lectin-like domain of TM has direct anti-inflammatory properties. METHODS: In the current study, we use murine models of acute inflammatory arthritis and biochemical approaches to assess the mechanism by which the lectin-like domain of TM modifies disease progression. RESULTS: Mice lacking the lectin-like domain of TM (TM(LeD/LeD)mice) develop inflammatory arthritis that is more rapid in onset and more severe than that developed in their wildtype counterparts. In two models of arthritis, treatment of mice with recombinant soluble lectin-like domain of TM significantly suppresses clinical evidence of disease and diminishes monocyte/macrophage infiltration into the synovium, with weaker expression of the pro-inflammatory high mobility group box chromosomal protein 1. While thrombin-TM mediated activation of thrombin activatable fibrinolysis inhibitor inactivates complement factors C3a and C5a, we show that TM has a second independent mechanism to regulate complement: the lectin-like domain of TM directly interferes with complement activation via the classical and lectin pathways. CONCLUSIONS: These data extend previous insights into the mechanisms by which TM modulates innate immunity, and highlight its potential as a therapeutic target for inflammatory diseases.


Assuntos
Artrite/metabolismo , Carboxipeptidase B2/metabolismo , Lectinas/química , Trombomodulina/química , Trombomodulina/genética , Animais , Artrite/prevenção & controle , Artrite Experimental/metabolismo , Coagulação Sanguínea , Ativação do Complemento , Técnicas de Transferência de Genes , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Estrutura Terciária de Proteína , Membrana Sinovial/patologia , Trombina/metabolismo , Trombomodulina/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 21(10): 1604-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11597933

RESUMO

The clinical outcome of vascular stenting is limited by in-stent stenosis. Increased nitric oxide (NO)/cGMP signaling by L-arginine (L-Arg) supplementation, the substrate for NO synthase (NOS), or NOS gene transfer may reduce in-stent neointima formation. After stenting, vascular cell proliferation in rat carotid arteries, as measured by 5'-bromodeoxyuridine (5'-BrdU) incorporation, indicated 15+/-8%, 28+/-5%, and 33+/-7% 5'-BrdU-positive vascular cells at 4, 7, and 14 days, respectively. Reporter beta-galactosidase gene transfer efficacy was evidenced by 30% beta-galactosidase-expressing medial smooth muscle cells at 14 days. The intima-to-media ratio (I/M) progressively increased to 2.32+/-0.24 at 14 days. To target in-stent neointima formation, animals were infected with adenoviral vectors (4x10(10) plaque-forming units per mL) expressing NOS2 (AdNOS2) or no transgene (AdRR5), or they received daily doses of L-Arg (500 mg. kg(-1). (d-1) IP). The neointima at 14 days was smaller in L-Arg-treated than in untreated rats (I/M 1.25+/-0.35 vs 2.32+/-0.24, P<0.05, n=7 each) or in AdRR5- and AdNOS2-infected rats (I/M 2.57+/-0.43, n=7 and 1.82+/-0.75, n=8, respectively; P<0.05 for both). The effect of L-Arg was abolished by simultaneous administration of N(G)-nitro L-arginine methyl ester, an NOS inhibitor (2.03+/-0.39, P<0.05, vs L-Arg). Inflammation was markedly less in L-Arg- and AdNOS2-treated than in AdRR5-infected rats. Supplemental L-Arg reduces neointima formation after stenting by way of an NOS-dependent mechanism and may be a valuable strategy to target in-stent stenosis.


Assuntos
Arginina/farmacologia , Estenose das Carótidas/terapia , Óxido Nítrico/fisiologia , Stents/efeitos adversos , Adenoviridae/genética , Angioplastia com Balão/efeitos adversos , Animais , Arginina/administração & dosagem , Estenose das Carótidas/etiologia , Estenose das Carótidas/patologia , Terapia Genética , Vetores Genéticos , Masculino , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Ratos , Ratos Wistar , Transdução Genética
3.
J Biol Chem ; 275(21): 16329-36, 2000 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-10748062

RESUMO

According to current views, peroxisomal beta-oxidation is organized as two parallel pathways: the classical pathway that is responsible for the degradation of straight chain fatty acids and a more recently identified pathway that degrades branched chain fatty acids and bile acid intermediates. Multifunctional protein-2 (MFP-2), also called d-bifunctional protein, catalyzes the second (hydration) and third (dehydrogenation) reactions of the latter pathway. In order to further clarify the physiological role of this enzyme in the degradation of fatty carboxylates, MFP-2 knockout mice were generated. MFP-2 deficiency caused a severe growth retardation during the first weeks of life, resulting in the premature death of one-third of the MFP-2(-/-) mice. Furthermore, MFP-2-deficient mice accumulated VLCFA in brain and liver phospholipids, immature C(27) bile acids in bile, and, after supplementation with phytol, pristanic and phytanic acid in liver triacylglycerols. These changes correlated with a severe impairment of peroxisomal beta-oxidation of very long straight chain fatty acids (C(24)), 2-methyl-branched chain fatty acids, and the bile acid intermediate trihydroxycoprostanic acid in fibroblast cultures or liver homogenates derived from the MFP-2 knockout mice. In contrast, peroxisomal beta-oxidation of long straight chain fatty acids (C(16)) was enhanced in liver tissue from MFP-2(-/-) mice, due to the up-regulation of the enzymes of the classical peroxisomal beta-oxidation pathway. The present data indicate that MFP-2 is not only essential for the degradation of 2-methyl-branched fatty acids and the bile acid intermediates di- and trihydroxycoprostanic acid but also for the breakdown of very long chain fatty acids.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/genética , Enoil-CoA Hidratase/genética , Ácidos Graxos/metabolismo , Complexos Multienzimáticos/genética , 3-Hidroxiacil-CoA Desidrogenases/deficiência , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Animais , Ácidos e Sais Biliares/química , Ácidos e Sais Biliares/metabolismo , Dieta , Enoil-CoA Hidratase/deficiência , Enoil-CoA Hidratase/metabolismo , Fibroblastos , Crescimento/genética , Fígado/enzimologia , Fígado/metabolismo , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/deficiência , Complexos Multienzimáticos/metabolismo , Peroxissomos/enzimologia , Peroxissomos/metabolismo , Fitol/metabolismo
4.
Blood ; 95(3): 936-42, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10648406

RESUMO

Recombinant staphylokinase (SakSTAR) variants obtained by site-directed substitution with cysteine, in the core (lysine 96 [Lys96], Lys102, Lys109, and/or Lys135) or the NH(2)-terminal region that is released during activation of SakSTAR (serine 2 [Ser2] and/or Ser3), were derivatized with thiol-specific (ortho-pyridyl-disulfide or maleimide) polyethylene glycol (PEG) molecules with molecular weights of 5,000 (P5), 10,000 (P10), or 20,000 (P20). The specific activities and thrombolytic potencies in human plasma were unaltered for most variants derivatized with PEG (PEGylates), but maleimide PEG derivatives had a better temperature stability profile. In hamsters, SakSTAR was cleared at 2.2 mL/min; variants with 1 P5 molecule were cleared 2-to 5-fold; variants with 2 P5 or 1 P10 molecules were cleared 10-to 30-fold; and variants with 1 P20 molecule were cleared 35-fold slower. A bolus injection induced dose-related lysis of a plasma clot, fibrin labeled with 125 iodine ((125)I-fibrin plasma clot), and injected into the jugular vein. A 50% clot lysis at 90 minutes required 110 microg/kg SakSTAR; 50 to 110 microg/kg of core-substitution derivatives with 1 P5; 25 microg/kg for NH(2)-terminal derivatives with 1 P5; 5 to 25 microg/kg with derivatives with 2 P5 or 1 P10; and 7 microg/kg with P20 derivatives. Core substitution with 1 or 2 P5 molecules did not significantly reduce the immunogenicity of SakSTAR in rabbits. Derivatization of staphylokinase with a single PEG molecule allows controllable reduction of the clearance while maintaining thrombolytic potency at a reduced dose. This indicates that mono-PEGylated staphylokinase variants may be used for single intravenous bolus injection.


Assuntos
Substituição de Aminoácidos , Cisteína/química , Fibrinolíticos/farmacologia , Metaloendopeptidases/farmacologia , Polietilenoglicóis/química , Animais , Cricetinae , Reagentes de Ligações Cruzadas/farmacologia , Cistina/química , Portadores de Fármacos , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/química , Fibrinolíticos/farmacocinética , Fibrinolíticos/uso terapêutico , Meia-Vida , Humanos , Maleatos/química , Metaloendopeptidases/química , Metaloendopeptidases/farmacocinética , Metaloendopeptidases/uso terapêutico , Mutagênese Sítio-Dirigida , Plasminogênio/metabolismo , Processamento de Proteína Pós-Traducional , Embolia Pulmonar/tratamento farmacológico , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Relação Estrutura-Atividade
5.
Circulation ; 100(25): 2541-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10604893

RESUMO

BACKGROUND: The effects of alteplase (rtPA), streptokinase, and staphylokinase (rSak) on focal cerebral ischemia (FCI) and on pulmonary clot lysis (PCL) were studied in hamsters. METHODS AND RESULTS: ++FCI was produced by ligation of the left middle cerebral artery (MCA) and common carotid artery (CCA) and a 10-minute occlusion of the right CCA. FCI was measured after 24 hours by 2,3, 5-triphenyltetrazolium chloride staining. (125)I-fibrin-labeled plasma clots were injected via the jugular vein, and clot lysis was determined from residual radioactivity at 90 minutes. Study drugs were given intravenously over 60 minutes. FCI increased from 1.2 (0. 27 to 2.3) mm(3) (median and 17th to 83rd percentile range, n=24) in controls to 19 to 27 mm(3) with thrombolytic agent, with maximal rates at 0.13+/-0.05 mg/kg rtPA, 0.23+/-0.09 mg/kg streptokinase, and 0.037+/-0.025 mg/kg rSak. PCL increased from 18+/-2% (mean+/-SEM, n=27) in controls to approximately 85% with thrombolytics, with maximal rates at 0.12+/-0.03 mg/kg rtPA, 0.17+/-0.05 mg/kg streptokinase, and 0.018+/-0.002 mg/kg rSak. All agents caused maximal FCI and PCL rates at similar doses without alpha(2)-antiplasmin and fibrinogen depletion. Injection of 6 mg/kg human plasminogen combined with streptokinase caused a "systemic fibrinolytic state" with fibrinogen depletion. Maximal rates of FCI were obtained with 0.097+/-0.077 mg/kg streptokinase (P=0.26 versus streptokinase alone) and of PCL with 0.010+/-0.002 mg/kg (P=0.006 versus streptokinase alone). CONCLUSIONS: Thrombolytic agents cause similar dose-related extension of FCI after MCA ligation and PCL, irrespective of the agent or systemic plasmin generation.


Assuntos
Infarto Cerebral/tratamento farmacológico , Fibrinolíticos/uso terapêutico , Metaloendopeptidases/farmacologia , Embolia Pulmonar/tratamento farmacológico , Estreptoquinase/uso terapêutico , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Artéria Carótida Primitiva , Cricetinae , Avaliação Pré-Clínica de Medicamentos , Fibrinogênio/metabolismo , Fibrinolisina/análise , Fibrinolíticos/toxicidade , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Mesocricetus , Metaloendopeptidases/toxicidade , Proteínas Recombinantes/uso terapêutico , Estreptoquinase/toxicidade , Terapia Trombolítica/efeitos adversos , Ativador de Plasminogênio Tecidual/toxicidade , alfa 2-Antiplasmina/metabolismo
6.
Circulation ; 99(18): 2440-4, 1999 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-10318667

RESUMO

BACKGROUND: The role of plasminogen system components in focal cerebral ischemic infarction (FCI) was studied in mice deficient in plasminogen (Plg-/-), in tissue or urokinase plasminogen activator (tPA-/- or uPA-/-), or in plasminogen activator inhibitor-1 or alpha2-antiplasmin (PAI-1(-/-) or alpha2-AP-/-). METHODS AND RESULTS: FCI was produced by ligation of the left middle cerebral artery and measured after 24 hours by planimetry of stained brain slices. In control (wild-type) mice, infarct size was 7.6+/-1.1 mm3 (mean+/-SEM), uPA-/- mice had similar infarcts (7.8+/-1.0 mm3, P=NS), tPA-/- mice smaller (2.6+/-0.80 mm3, P<0.0001), PAI-1(-/-) mice larger (16+/-0.52 mm3, P<0.0001), and Plg-/- mice larger (12+/-1.2 mm3, P=0.037) infarcts. alpha2-AP-/- mice had smaller infarcts (2. 2+/-1.1 mm3, P<0.0001 versus wild-type), which increased to 13+/-2.5 mm3 (P<0.005 versus alpha2-AP-/-) after intravenous injection of human alpha2-AP. Injection into alpha2-AP-/- mice of Fab fragments of affinospecific rabbit IgG against human alpha2-AP, after injection of 200 microg human alpha2-AP, reduced FCI from 11+/-1.5 to 5.1+/-1.1 mm3 (P=0.004). CONCLUSIONS: Plg system components affect FCI at 2 different levels: (1) reduction of tPA activity (tPA gene inactivation) reduces whereas its augmentation (PAI-1 gene inactivation) increases infarct size, and (2) reduction of Plg activity (Plg gene inactivation or alpha2-AP injection) increases whereas its augmentation (alpha2-AP gene inactivation or alpha2-AP neutralization) reduces infarct size. Inhibition of alpha2-AP may constitute a potential avenue to treatment of FCI.


Assuntos
Isquemia Encefálica/metabolismo , Infarto Cerebral/metabolismo , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Plasminogênio/fisiologia , Ativador de Plasminogênio Tecidual/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , alfa 2-Antiplasmina/fisiologia , Adenoviridae/genética , Animais , Encéfalo/patologia , Isquemia Encefálica/patologia , Infarto Cerebral/patologia , Avaliação Pré-Clínica de Medicamentos , Feminino , Fibrinólise , Marcação de Genes , Terapia Genética , Vetores Genéticos/genética , Humanos , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Plasminogênio/deficiência , Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/deficiência , Inibidor 1 de Ativador de Plasminogênio/genética , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Proteínas Recombinantes de Fusão/uso terapêutico , Ativador de Plasminogênio Tecidual/deficiência , Ativador de Plasminogênio Tecidual/genética , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/deficiência , Ativador de Plasminogênio Tipo Uroquinase/genética , alfa 2-Antiplasmina/deficiência , alfa 2-Antiplasmina/genética , alfa 2-Antiplasmina/imunologia , alfa 2-Antiplasmina/uso terapêutico
7.
Atherosclerosis ; 137 Suppl: S33-8, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9694539

RESUMO

Malondialdehyde (MDA)-modified and oxidized low density lipoproteins (LDL) have been demonstrated in atherosclerotic lesions. Elevated titers of autoimmune antibodies specific for MDA-modified LDL predicted the progression of carotid atherosclerosis and of myocardial infarction. Recently, elevated levels of MDA-modified LDL were detected in the plasma of patients with ischemic heart disease, whereas, elevated levels of oxidized LDL were detected in the plasma of patients with ischemic heart disease and of heart transplant patients with post-transplant cardiovascular disease. Although increased levels of autoimmune antibodies against oxidatively modified LDL and increased levels of oxidized LDL antigen appear to be associated with atherosclerotic cardiovascular disease, there is to date no direct proof of the causal role of oxidized LDL in atherothrombosis. However, the decreased risk of cardiovascular disease associated with the administration of antioxidants (e.g. vitamin E), estrogen supplementation and increased levels of high density lipoproteins (HDL) may, at least partially, be due to the inhibition of oxidation of LDL or to the reversal of the atherothrombotic effects of oxidized LDL.


Assuntos
Arteriosclerose/etiologia , Lipoproteínas LDL/metabolismo , Animais , Antioxidantes/farmacologia , Arteriosclerose/prevenção & controle , Estrogênios/farmacologia , Humanos , Lipoproteínas HDL/fisiologia , Lipoproteínas LDL/sangue , Oxirredução
9.
DNA Cell Biol ; 15(6): 429-42, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8672239

RESUMO

A 5.4-kb cDNA encoding the protein that binds to the B Box of the plasminogen activator inhibitor-1 (PAI-1) gene was isolated and sequenced. The protein, named helicase-like transcription factor (HLTF), contains a DNA-binding domain, a RING finger domain, and seven helicase domains and is homologous to SWI/SNF proteins. Two HLTF mRNAs of 5.5 and 4.5 kb were detected in most human tissues, a single gene was located on chromosome 3q24-25, and the protein was located in the nucleoplasm. Two HLTF proteins differing in translation start site (Met-1 or Met-123) were obtained by in vitro translation in reticulocyte lysate or by immunoprecipitation from HeLa cell nuclear extracts. In vitro transcription from the PAI-1 promoter in HeLa cell extracts was inhibited by HLTF antibodies and by the HLTF DNA binding domain. Over-expression of HLTF or HLTFMet123 produced a three-fold induction of PAI-1-LUC transient expression in HeLa cells. Mutation of the PAI-1 B Box led to an eight-fold reduction of basal PAI-1-LUC expression in these cell lines, but did not affect the four- to six-fold induction by phorbol esters.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/fisiologia , Inibidor 1 de Ativador de Plasminogênio/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Sequência de Bases , Núcleo Celular/química , Cromossomos Humanos Par 3 , Clonagem Molecular , DNA/metabolismo , DNA Helicases/genética , DNA Complementar/genética , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Peso Molecular , Músculo Esquelético , Especificidade de Órgãos , Ligação Proteica , RNA Mensageiro/análise , Sequências Reguladoras de Ácido Nucleico/genética , Análise de Sequência de DNA , Fatores de Transcrição/análise , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Dedos de Zinco/genética
10.
FASEB J ; 8(15): 1279-84, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8001740

RESUMO

Oxidized low density lipoproteins (LDL) and autoimmune antibodies against oxidized LDL have been demonstrated in human atherosclerotic lesions; they may enhance the progression of these lesions by 1) enhancing monocyte adhesion and macrophage foam cell generation; 2) inducing smooth muscle cell migration, proliferation, and foam cell generation; 3) enhancing platelet adhesion and aggregation, which may stimulate macrophage foam cell generation and smooth muscle cell proliferation; 4) triggering thrombosis; and 5) impairing vasodilation, which results in increased shear stress. The oxidation of LDL probably occurs in the arterial wall, where it is sequestered from circulating antioxidants. Atherosclerotic arterial walls contain increased levels of redox-active metal ions, and the LDL of patients with atherosclerotic cardiovascular disease are more susceptible to oxidation, possibly as a result of reduced endogenous antioxidants such as vitamin E. Dietary supplementation with vitamin E (up to 1,000 IU/day) or administration of probucol reduce the oxidation rate of LDL and may significantly decrease the risk of coronary heart disease.


Assuntos
Arteriosclerose/etiologia , Lipoproteínas LDL/fisiologia , Trombose/etiologia , Animais , Antioxidantes/farmacologia , Humanos , Oxirredução
11.
J Am Coll Cardiol ; 19(6): 1350-9, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1342779

RESUMO

The chimeric molecule K1K2Pu, comprising the two kringle domains (K1 and K2) of tissue-type plasminogen activator (t-PA) and the COOH-terminal region with the serine protease domain (Pu) of urokinase-type plasminogen activator (u-PA), was previously shown to have a 5- to 10-fold reduced clearance rate with maintained specific thrombolytic activity, resulting in an increased thrombolytic potency in animal models of venous and arterial thrombosis. To document the thrombolytic potential of K1K2Pu, the thrombolytic potency and fibrin specificity were studied in a combined platelet-rich arterial eversion graft thrombosis and venous whole blood clot model in heparinized dogs (100 U/kg bolus and 50 U/kg per h infusion). Dose-response effects of bolus injections of K1K2Pu (0.032 to 0.25 mg/kg) were compared with those of recombinant t-PA (rt-PA) and of recombinant single chain u-PA (rscu-PA) (0.25 to 1.0 mg/kg each) in groups of five or six dogs, each given heparin with or without the thromboxane synthase inhibitor/prostaglandin endoperoxide receptor antagonist ridogrel. Heparin and ridogrel in the absence of a thrombolytic agent did not produce arterial reflow or venous clot lysis in five dogs. Addition of K1K2Pu, rt-PA or rscu-PA resulted in a dose-dependent induction of arterial reflow and of venous clot lysis in the absence of systemic fibrinolytic activation and fibrinogen breakdown. Consistent arterial reflow required 0.063 mg/kg of K1K2Pu and 0.5 mg/kg of rt-PA or of rscu-PA. The thrombolytic potency for venous clot lysis, expressed as percent lysis per mg compound administered per kg body weight, was (mean +/- SEM) 750 +/- 160 for K1K2Pu, 68 +/- 17 for rscu-PA (p less than 0.001 vs. K1K2Pu) and 110 +/- 29 for rt-PA (p less than 0.001 vs. K1K2Pu). The plasma clearance rates were significantly lower for K1K2Pu than for rscu-PA and rt-PA. In the absence of ridogrel, arterial reflow was significantly slower and was followed by cyclic reocclusion and reflow; however, venous clot lysis was unaffected. Template bleeding times were not significantly altered in the absence but were markedly prolonged in the presence of ridogrel. These results confirm and establish that, when given as a bolus injection, K1K2Pu has an approximately 10-fold higher thrombolytic potency for arterial and venous thrombolysis than does rt-PA or rscu-PA. Thrombolysis with K1K2Pu is obtained in the absence of systemic fibrinolytic activation and fibrinogen breakdown. These properties suggest that K1K2Pu offers potential for thrombolytic therapy by bolus administration in patients with thromboembolic disease.


Assuntos
Modelos Animais de Doenças , Artéria Femoral , Veia Femoral , Terapia Trombolítica/métodos , Trombose/tratamento farmacológico , Ativador de Plasminogênio Tecidual/uso terapêutico , Ativador de Plasminogênio Tipo Uroquinase/uso terapêutico , Animais , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Artéria Femoral/fisiopatologia , Veia Femoral/fisiopatologia , Membro Posterior/irrigação sanguínea , Infusões Intravenosas , Ativadores de Plasminogênio/administração & dosagem , Ativadores de Plasminogênio/uso terapêutico , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Fluxo Sanguíneo Regional/efeitos dos fármacos , Trombose/sangue , Trombose/fisiopatologia , Ativador de Plasminogênio Tecidual/administração & dosagem , Ativador de Plasminogênio Tipo Uroquinase/administração & dosagem
12.
Circulation ; 83(6 Suppl): IV65-72, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2040073

RESUMO

BACKGROUND: Two thrombosis models for the evaluation of thrombolytic agents in small animals (less than 100 g) were evaluated: an iodine-125 fibrin-labeled rat plasma clot in the inferior caval vein of 3-4-week-old rats and a pulmonary embolus in adult hamsters that had been obtained by injection of a 125I fibrin-labeled human plasma clot. The extent of thrombolysis was determined by continuous external monitoring of radioisotope over the thrombus region and by ex vivo recovery of residual clot. METHODS AND RESULTS: In the rat model, infusion of solvent for 60 minutes was associated with mean +/- SEM lysis within 90 minutes of 13 +/- 3% (n = 8) by external counting and 26 +/- 4% (n = 8) by radioisotope recovery. Intravenous infusion of recombinant tissue-type plasminogen activator (rt-PA) over 60 minutes caused dose-dependent progressive clot lysis; with 0.5 mg/kg, producing a plasma level of 0.14 +/- 0.04 microgram/ml, lysis was 64 +/- 9% (n = 4) by external gamma counting and 78 +/- 4% (n = 4) by residual isotope in the vein segment and was not associated with significant fibrinogen or alpha 2-antiplasmin breakdown. In the hamster model, infusion of solvent for 60 minutes was associated with lysis within 90 minutes of 19 +/- 4% (n = 11) by external gamma counting and 31 +/- 3% (n = 14) by residual radioisotope. Intravenous rt-PA during 60 minutes resulted in dose-dependent progressive thrombolysis; with 0.5 mg/kg, producing a plasma level of 0.14 +/- 0.01 micrograms/ml, lysis was 50 +/- 4% (n = 4) by external gamma counting and 78 +/- 5% (n = 4) by residual radioactivity, without an extensive decrease in fibrinogen or alpha 2-antiplasmin. Parallel experiments in the rabbit jugular vein thrombosis model with a rabbit blood clot with intravenous infusion over 4 hours produced 7 +/- 2% (n = 9) lysis with solvent and dose-dependent progressive lysis with rt-PA; with 1 mg/kg, producing a plasma level of 0.20 +/- 0.03 microgram/ml, lysis was 56 +/- 7% (n = 7) by external gamma counting and 61 +/- 7% (n = 7) by residual radioactivity, without extensive consumption of fibrinogen or alpha 2-antiplasmin. CONCLUSIONS: These two thrombosis models in small animals are as reproducible and quantitative as the extensively used rabbit jugular vein thrombosis model. The hamster pulmonary embolism model is superior because it is simpler and more straightforward and allows the performance of as many as 10 experiments by one investigator in 1 day.


Assuntos
Fibrinolíticos/farmacologia , Trombose/sangue , Animais , Coagulação Sanguínea/efeitos dos fármacos , Cricetinae , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Veias Jugulares , Masculino , Embolia Pulmonar/sangue , Coelhos , Ratos , Ratos Endogâmicos , Veia Cava Inferior
13.
Circulation ; 75(4): 830-6, 1987 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3103951

RESUMO

The effect of beta-adrenergic blockade on the salvage and functional recovery of reperfused myocardium was investigated in anesthetized dogs. Immediately after thrombotic occlusion of the left anterior descending coronary artery, the cardioselective beta-blocking agent metoprolol was given intravenously at a dose of 0.5 mg/kg infused over 10 min. One hour after the onset of occlusion, recanalization was initiated by intravenous infusion of recombinant human tissue-type plasminogen activator (rt-PA, 10 micrograms/kg/min for 30 min). Anatomic infarct size expressed as percent of the left ventricular mass (I/LV), global ejection fraction, and mean systolic shortening of the segmental radii (SS) of the infarcted area were measured either after 24 hr or 1 week in six groups of six dogs each: group I (rt-PA + metoprolol, evaluated at 24 hr), group II (rt-PA + metoprolol, evaluated at 1 week, group III (rt-PA alone, evaluated at 24 hr), group IV (rt-PA alone, evaluated at 1 week), group V (persistent occlusion, evaluated at 24 hr), and group VI (persistent occlusion, evaluated at 1 week). The smallest infarcts were found in reperfused dogs given metoprolol, but the differences from dogs receiving rt-PA alone were not statistically significant (I/LV, expressed as mean +/- SEM: 5.5 +/- 0.9% in group I, 6.7 +/- 1.9% in group II, 15.4 +/- 5.0% in group III, 11.4 +/- 3.5% in group IV, 23.6 +/- 2.5% in group V, and 26.9 +/- 2.3% in group VI).(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Doença das Coronárias/tratamento farmacológico , Trombose Coronária/tratamento farmacológico , Metoprolol/uso terapêutico , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Sístole/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Trombose Coronária/mortalidade , Trombose Coronária/fisiopatologia , Cães , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Hemodinâmica/efeitos dos fármacos , Infarto do Miocárdio/mortalidade , Infarto do Miocárdio/fisiopatologia , Proteínas Recombinantes/uso terapêutico , Fatores de Tempo
14.
Arteriosclerosis ; 4(6): 579-85, 1984.
Artigo em Inglês | MEDLINE | ID: mdl-6439177

RESUMO

Tissue-type plasminogen activator (t-PA), purified from the culture fluid of a stable human melanoma cell line, is a serine protease, different from urokinase, with a molecular weight of about 70,000. It is composed of one polypeptide chain, which is converted to a two-chain molecule by limited plasmic action. Activation of plasminogen to plasmin occurs by cleavage of the Arg 560-Val 561 peptide bond. Kinetic analysis has shown that the activation obeys Michaelis-Menten kinetics and that the presence of fibrin strikingly enhances the activation rate by increasing the affinity of plasminogen for fibrin-bound t-PA. The directed action of plasmin toward fibrin in vivo, might be explained by the low Michaelis constant in the presence of fibrin (0.16 microM), which allows efficient plasminogen activation on a fibrin clot, while its high value in the absence of fibrin (65 microM) prevents efficient activation in plasma. Plasmin formed on the fibrin surface would then be protected from rapid inactivation by alpha 2-antiplasmin. An important consequence of this molecular model for physiological fibrinolysis is that specific thrombolysis is only expected with the use of a specific plasminogen activator, which confines activation to the fibrin surface. Studies on the thrombolytic properties of purified t-PA in various animal species and in humans have revealed a higher specific thrombolytic activity than urokinase. Thrombolysis could be achieved without causing significant plasminogen activation, alpha 2-antiplasmin consumption, or fibrinogen breakdown. Alternatively, pro-urokinase, the zymogen precursor of urokinase, also displays a certain degree of fibrin specificity. Its mechanism of action and potential therapeutic value remain to be established.


Assuntos
Fibrinolíticos/uso terapêutico , Animais , Fenômenos Químicos , Físico-Química , Doença das Coronárias/tratamento farmacológico , Cães , Avaliação Pré-Clínica de Medicamentos , Precursores Enzimáticos/farmacologia , Precursores Enzimáticos/uso terapêutico , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Humanos , Técnicas In Vitro , Ativadores de Plasminogênio/antagonistas & inibidores , Ativadores de Plasminogênio/farmacologia , Ativadores de Plasminogênio/uso terapêutico , Inativadores de Plasminogênio , Embolia Pulmonar/tratamento farmacológico , Coelhos , Relação Estrutura-Atividade , Tromboflebite/tratamento farmacológico , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/uso terapêutico
15.
Biochim Biophys Acta ; 579(1): 142-54, 1979 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-157166

RESUMO

Plasminogen and plasminogen derivatives which contain lysine-binding sites were found to decrease the reaction rate between plasmin and alpha2-antiplasmin by competing with plasmin for the complementary site(s) in alpha2-antiplasmin. The dissocwation constant Kd for the interaction between intact plasminogen (Glu-plasminogen) and alpha2-antiplasmin is 4.0 microM but those for Lys-plasminogen or TLCK-plasmin are about 10-fold lower indicating a stronger interaction. The lysine-binding site(s) which is situated in triple-loops 1--3 in the plasmin A-chain is mainly responsible for the interaction with alpha2-antiplasmin. The interaction between Glu-plasminogen and alpha2-antiplasmin furthermore enhances the activation of Glu-plasminogen by urokinase to a comparable extent as 6-aminohexanoic acid, suggesting that similar conformational changes occur in the proenzyme after complex formation. Fibrinogen, fibrinogen digested with plasmin, purified fragment E and purified fragment D interfere with the reaction between plasmin and alpha2-antiplasmin by competing with alpha2-antiplasmin for the lysine-binding site(s) in the plasmin A-chain. The Kd obtained for these interactions varied between 0.2 microM and 1.4 microM; fragment E being the most effective. Thus the fibrinogen molecule contains several complementary sites to the lysine-binding sites located both in its NH2-terminal and COOH-terminal regions; these sites are to a large extent.


Assuntos
Proteínas Sanguíneas , Fibrinogênio , Fibrinolisina/antagonistas & inibidores , Lisina , Sítios de Ligação , Humanos , Cinética , Ligantes , Ligação Proteica , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA