Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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