Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS One ; 10(4): e0122410, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25830552

RESUMO

Ischaemic stroke is caused by occlusive thrombi in the cerebral vasculature. Although tissue-plasminogen activator (tPA) can be administered as thrombolytic therapy, it has major limitations, which include disruption of the blood-brain barrier and an increased risk of bleeding. Treatments that prevent or limit such deleterious effects could be of major clinical importance. Activated protein C (APC) is a natural anticoagulant that regulates thrombin generation, but also confers endothelial cytoprotective effects and improved endothelial barrier function mediated through its cell signalling properties. In murine models of stroke, although APC can limit the deleterious effects of tPA due to its cell signalling function, its anticoagulant actions can further elevate the risk of bleeding. Thus, APC variants such as APC(5A), APC(Ca-ins) and APC(36-39) with reduced anticoagulant, but normal signalling function may have therapeutic benefit. Human and murine protein C (5A), (Ca-ins) and (36-39) variants were expressed and characterised. All protein C variants were secreted normally, but 5-20% of the protein C (Ca-ins) variants were secreted as disulphide-linked dimers. Thrombin generation assays suggested reductions in anticoagulant function of 50- to 57-fold for APC(36-39), 22- to 27-fold for APC(Ca-ins) and 14- to 17-fold for APC(5A). Interestingly, whereas human wt APC, APC(36-39) and APC(Ca-ins) were inhibited similarly by protein C inhibitor (t½ - 33 to 39 mins), APC(5A) was inactivated ~9-fold faster (t½ - 4 mins). Using the murine middle cerebral artery occlusion ischaemia/repurfusion injury model, in combination with tPA, APC(36-39), which cannot be enhanced by its cofactor protein S, significantly improved neurological scores, reduced cerebral infarct area by ~50% and reduced oedema ratio. APC(36-39) also significantly reduced bleeding in the brain induced by administration of tPA, whereas wt APC did not. If our data can be extrapolated to clinical settings, then APC(36-39) could represent a feasible adjunctive therapy for ischaemic stroke.


Assuntos
Anticoagulantes/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Proteína C/uso terapêutico , Animais , Anticoagulantes/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Infarto da Artéria Cerebral Média/sangue , Cinética , Masculino , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Fármacos Neuroprotetores/farmacologia , Proteína C/química , Proteína C/farmacologia , Inibidor da Proteína C/química , Inibidor da Proteína C/farmacologia , Proteólise , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/prevenção & controle , Trombina/metabolismo , Tempo de Trombina
2.
Thromb Haemost ; 112(5): 883-92, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25230930

RESUMO

Despite years of research and efforts to translate stroke research to clinical therapy, ischaemic stroke remains a major cause of death, disability, and diminished quality of life. Primary and secondary preventive measures combined with improved quality of care have made significant progress. However, no novel drug for ischaemic stroke therapy has been approved in the past decade. Numerous studies have shown beneficial effects of activated protein C (APC) in rodent stroke models. In addition to its natural anticoagulant functions, APC conveys multiple direct cytoprotective effects on many different cell types that involve multiple receptors including protease activated receptor (PAR) 1, PAR3, and the endothelial protein C receptor (EPCR). Application of molecular engineered APC variants with altered selectivity profiles to rodent stroke models demonstrated that the beneficial effects of APC primarily require its cytoprotective activities but not its anticoagulant activities. Extensive basic, preclinical, and clinical research provided a compelling rationale based on strong evidence for translation of APC therapy that has led to the clinical development of the cytoprotective-selective APC variant, 3K3A-APC, for ischaemic stroke. Recent identification of non-canonical PAR1 and PAR3 activation by APC that give rise to novel tethered-ligands capable of inducing biased cytoprotective signalling as opposed to the canonical signalling provides a mechanistic explanation for how APC-mediated PAR activation can selectively induce cytoprotective signalling pathways. Collectively, these paradigm-shifting discoveries provide detailed insights into the receptor targets and the molecular mechanisms for neuroprotection by cytoprotective-selective 3K3A-APC, which is currently a biologic drug in clinical trials for ischaemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Proteína C/uso terapêutico , Animais , Antígenos CD/fisiologia , Coagulação Sanguínea , Isquemia Encefálica/epidemiologia , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Receptor de Proteína C Endotelial , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Hemorragia/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Moleculares , Proteína C/metabolismo , Proteína C/farmacologia , Conformação Proteica , Receptores de Superfície Celular/fisiologia , Receptores Ativados por Proteinase/fisiologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Trombina/farmacologia , Trombina/fisiologia , Ativador de Plasminogênio Tecidual/efeitos adversos , Ativador de Plasminogênio Tecidual/uso terapêutico
3.
Scand J Clin Lab Invest ; 73(5): 407-13, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23713554

RESUMO

AIM: In view of the central role of granulocytic neutrophils in the context of inflammatory reactions, the present study focuses on anti-inflammatory effects of drugs on activated neutrophils in neonates and adults. METHODS: Sixteen blood samples of neonates and adults were investigated in a prospective study. Loss of deformability, morphological changes, and increases in neutrophil elastase were determined as measures of neutrophil activation due to incubation with the pro-inflammatory cytokine interleukin-8. For inhibition experiments, the blood samples were also incubated with the phosphodiesterase inhibitors milrinone and piclamilast, the protease inhibitor urinastatin, ketamine, protein C concentrate, and the nitric oxide donor FK 409. Changes in deformability were investigated with a cell transit analyzer, morphological changes by microscopic observation, and the extent of neutrophil elastase release with an enzyme immunoassay. RESULTS: The drugs milrinone, piclamilast, urinastatin, ketamine, protein C concentrate and FK 409 showed deactivating effects on activated neutrophils in recommended clinical doses. They improved deformability as well as reduced pseudopod formation and the release of neutrophil elastase. The effects on neutrophils did not differ between neonates and adults despite their functional differences. CONCLUSION: We conclude that these drugs may reduce the inflammatory response and improve microcirculation in neonates and adults during inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Neutrófilos/efeitos dos fármacos , Adulto , Benzamidas/farmacologia , Forma Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Glicoproteínas/farmacologia , Humanos , Recém-Nascido , Ketamina/farmacologia , Elastase de Leucócito/metabolismo , Masculino , Milrinona/farmacologia , Ativação de Neutrófilo , Neutrófilos/enzimologia , Neutrófilos/imunologia , Nitrocompostos/farmacologia , Estudos Prospectivos , Proteína C/farmacologia , Piridinas/farmacologia
4.
Med Sci (Paris) ; 27(5): 501-7, 2011 May.
Artigo em Francês | MEDLINE | ID: mdl-21609671

RESUMO

Sepsis is defined as a systemic response to infection, characterized by an intense inflammatory response linked to coagulation activation and fibrinolysis inhibition, two processes which are intimately associated. In a field where mortality remains very high, administration of activated protein C, a physiological coagulation inhibitor with cytoprotective properties, has demonstrated its effectiveness and was able to reduce mortality. Protein C belongs to a system that involves plasma proteins and endothelial cell receptors. In addition to well documented effects on coagulation and fibrinolysis, activated protein C exhibits anti-inflammatory, anti-apoptotic but also anti-histone activities. Indeed, a recent study focusing on the cytoprotective effects of activated protein C showed that extracellular histones are released during severe sepsis and may participate in the pathophysiology of severe sepsis. These histones appear to be new targets of activated protein C.


Assuntos
Coagulação Sanguínea/fisiologia , Inflamação/sangue , Proteína C/fisiologia , Sepse/fisiopatologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Anticoagulantes/uso terapêutico , Antígenos CD/fisiologia , Fatores de Coagulação Sanguínea/fisiologia , Ensaios Clínicos como Assunto , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Receptor de Proteína C Endotelial , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Fibrinólise , Transtornos Hemorrágicos/induzido quimicamente , Histonas/antagonistas & inibidores , Histonas/metabolismo , Humanos , Inflamação/tratamento farmacológico , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Modelos Biológicos , Estudos Multicêntricos como Assunto , Proteína C/efeitos adversos , Proteína C/farmacologia , Proteína C/uso terapêutico , Receptor PAR-1/efeitos dos fármacos , Receptor PAR-1/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/uso terapêutico , Sepse/sangue , Sepse/tratamento farmacológico
5.
J Infect Dis ; 202(10): 1600-7, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20925531

RESUMO

BACKGROUND: Recombinant human activated protein C (APC) improves survival of patients with severe sepsis; this beneficial effect is especially apparent in patients with pneumococcal pneumonia. The aim of this study was to determine the effect of APC treatment initiated after induction of pneumococcal pneumonia on pulmonary coagulation, inflammation, and survival, with or without concurrent antibiotic therapy. METHODS: Mice were infected intranasally with viable Streptococcus pneumoniae and were treated intraperitoneally after 24 h of infection with vehicle, recombinant mouse (rm) APC (125 µg), ceftriaxone (500 µg), or rm-APC plus ceftriaxone. Treatment with rm-APC or vehicle was repeated every 8 h for a maximum of 96 h. Animals were either killed 48 h after infection or were monitored in a survival study (with an extra dose of ceftriaxone given after 72 h). RESULTS: Rm-APC treatment inhibited pulmonary activation of coagulation, as reflected by lower levels of thrombin-antithrombin complexes and D-dimer. Rm-APC did not affect the pulmonary levels of 55 inflammatory mediators in the context of antibiotic therapy. Rm-APC added to ceftriaxone markedly improved survival, compared with ceftriaxone treatment alone. CONCLUSIONS: Rm-APC inhibits pulmonary activation of coagulation and, when added to antibiotic therapy, improves survival in murine pneumococcal pneumonia.


Assuntos
Transtornos de Proteínas de Coagulação/tratamento farmacológico , Transtornos de Proteínas de Coagulação/microbiologia , Pneumonia Pneumocócica/tratamento farmacológico , Proteína C/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Fatores de Coagulação Sanguínea/antagonistas & inibidores , Ceftriaxona/administração & dosagem , Ceftriaxona/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/complicações , Proteína C/administração & dosagem , Proteína C/farmacologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
6.
Eur J Pharmacol ; 635(1-3): 103-8, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20123096

RESUMO

The present study was attempted to evaluate the therapeutic effects of activated protein C and/or hyperbaric oxygen in an animal model of heatstroke. Sixty-eight minutes heat stress (43 degrees C) initiated, the anesthetized rats were randomized to several groups and administered: 1) no resuscitation (vehicle solution plus normabaric air, 2) intravenous activated protein C (1mg in 1ml of normal saline per kg of body weight), 3) hyperbaric oxygen (100% oxygen at 202kpa for 17min), and 4) intravenous activated protein C plus hyperbaric oxygen. Another group of rats exposed to room temperature (26 degrees C) was used as normothermic controls. Blood sampling was 0min, 70min, and 85min after heat stress initiated. When the vehicle-treated rats underwent heat exposure, their survival time values found were to be 19-25min. Resuscitation with activated protein C or hyperbaric oxygen significantly and equally improved survival during heatstroke (134-159min). As compared with those of activated protein C or hyperbaric oxygen alone, combined activated protein C and hyperbaric oxygen significantly had higher survival time values (277-347min). All vehicle-treated heatstroke animals displayed systemic response, hypercoagulable state, and hepatic and renal dysfunction. Combined activated protein C and hyperbaric oxygen therapy reduced these heatstroke reactions better than activated protein C or hyperbaric oxygen alone. The results indicate consequently, combined activated protein C and hyperbaric oxygen therapy heightens benefit in combating heatstroke reactions.


Assuntos
Golpe de Calor/terapia , Oxigenoterapia Hiperbárica , Proteína C/metabolismo , Proteína C/uso terapêutico , Animais , Ativação Enzimática , Golpe de Calor/complicações , Golpe de Calor/tratamento farmacológico , Hipotensão/complicações , Hipotensão/tratamento farmacológico , Hipotensão/terapia , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/terapia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Fígado/efeitos dos fármacos , Fígado/fisiopatologia , Insuficiência de Múltiplos Órgãos/complicações , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Insuficiência de Múltiplos Órgãos/terapia , Proteína C/farmacologia , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Trombofilia/complicações , Trombofilia/tratamento farmacológico , Trombofilia/terapia
7.
Blood ; 109(9): 3733-40, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17227834

RESUMO

The anticoagulant and anti-inflammatory enzyme, activated protein C (APC), naturally controls thrombosis without affecting hemostasis. We therefore evaluated whether the integrity of primary hemostasis was preserved during limited pharmacological antithrombotic protein C activator (PCA) treatment in baboons. The double-mutant thrombin (Trp215Ala/Glu217Ala) with less than 1% procoagulant activity was used as a relatively selective PCA and compared with systemic anticoagulation by APC and low-molecular-weight heparin (LMWH) at doses that inhibited fibrin deposition on thrombogenic segments of arteriovenous shunts. As expected, both systemic anticoagulants, APC (0.028 or 0.222 mg/kg for 70 minutes) and LMWH (0.325 to 2.6 mg/kg for 70 minutes), were antithrombotic and prolonged the template bleeding time. In contrast, PCA at doses (0.0021 to 0.0083 mg/kg for 70 minutes) that had antithrombotic effects comparable with LMWH did not demonstrably impair primary hemostasis. PCA bound to platelets and leukocytes, and accumulated in thrombi. APC infusion at higher circulating APC levels was less antithrombotic than PCA infusion at lower circulating APC levels. The observed dissociation of antithrombotic and antihemostatic effects during PCA infusion thus appeared to emulate the physiological regulation of intravascular blood coagulation (thrombosis) by the endogenous protein C system. Our data suggest that limited pharmacological protein C activation might exhibit considerable thrombosis specificity.


Assuntos
Anticoagulantes/farmacologia , Hemostasia/efeitos dos fármacos , Hemostáticos/farmacologia , Heparina de Baixo Peso Molecular/farmacologia , Proteína C/farmacologia , Trombina/farmacologia , Trombose/tratamento farmacológico , Substituição de Aminoácidos , Animais , Anticoagulantes/farmacocinética , Tempo de Sangramento , Avaliação Pré-Clínica de Medicamentos , Hemostáticos/farmacocinética , Heparina de Baixo Peso Molecular/farmacocinética , Humanos , Mutação de Sentido Incorreto , Papio , Proteína C/agonistas , Proteína C/análise , Proteína C/farmacocinética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Trombina/genética , Trombina/farmacocinética , Trombose/sangue
8.
Thromb Res ; 120(4): 531-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17169412

RESUMO

The antithrombotic and anticoagulant effects of recombinant wild type (WT) and mutated human activated protein C (hAPC) were investigated using a rat model of arterial thrombosis. Recent in vitro studies using human plasma have shown enhanced anticoagulant effects of hAPC by mutagenesis of either loop 148 in the serine protease domain or of the Gla domain. The Gla-domain mutant QGNSEDY-hAPC (= H10Q/S11G/S12N/D23S/Q32E/N33D/H44Y) was found to be particularly active as an anticoagulant. We now combined the two mutations to create the variant QGNSEDY-hAPC:B148 and investigated the in vivo effects of this variant as well as of QGNSEDY-hAPC and WT hAPC using a rat model of arterial thrombosis. In vitro clotting experiments using rat plasma demonstrated WT hAPC to be inefficient, whereas both mutant hAPC variants yielded distinct dose dependent anticoagulant effects. In the arterial injury model, a segment of the left common carotid artery was opened longitudinally. An endarterectomy was performed and the arteriotomy was closed, whereafter the vessel was reperfused and the patency rate determined after 31 min. Three treatment groups each containing 10 rats and a control group of 20 animals were in a blind random fashion given intravenous bolus injections of 0.8 mg/kg WT or mutant hAPC or vehicle only. The ex vivo clotting times of plasma drawn 3 min after the injections, as compared to baseline clotting times, were approximately doubled by QGNSEDY-hAPC and tripled by QGNSEDY-hAPC:B148 infusions, while WT APC had little effect. Compared to the control group, none of the hAPC preparations had significant antithrombotic effect or increased arteriotomy bleeding.


Assuntos
Anticoagulantes/farmacologia , Fibrinolíticos/farmacologia , Mutação , Proteína C/genética , Proteína C/farmacologia , Animais , Anticoagulantes/administração & dosagem , Trombose das Artérias Carótidas , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Fibrinolíticos/administração & dosagem , Humanos , Proteína C/administração & dosagem , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes
9.
Curr Drug Saf ; 2(3): 227-31, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18690972

RESUMO

Severe sepsis and septic shock are common in the critically ill patient and account for considerable morbidity and mortality not to mention the high associated costs. Advances in our understanding of sepsis pathophysiology and in the important link between the inflammatory response to sepsis and activation of coagulation led to the development and licensing of the first ever, specific, immunomodulatory anti-sepsis drug. Drotrecogin alfa (activated), a recombinant version of activated protein C, was shown in a large randomized controlled clinical trial to reduce mortality rates from 30.8% in the placebo group to 24.7% in the treatment group, which equated to one additional life saved for every 16 patients treated. Vasopressor requirements and duration of mechanical ventilation were also reduced. Apart from an expected increased risk of severe bleeding, mostly associated with interventions, drotrecogin alfa (activated) was not associated with any other adverse reactions. In this article, I will briefly summarize the events leading to the development of drotrecogin alfa (activated) including aspects of sepsis epidemiology and pathophysiology and the results of early animal and clinical studies. The results of the large multicenter phase III PROWESS study will then be reviewed, along with results from subsequent open-label studies. Finally, I will focus on the key side effect issue with drotrecogin alfa (activated), that of increased bleeding, drawing data from the available clinical studies, and highlighting the contraindications and precautions when prescribing this drug.


Assuntos
Anti-Infecciosos/uso terapêutico , Proteína C/uso terapêutico , Sepse/tratamento farmacológico , Animais , Anti-Infecciosos/efeitos adversos , Anti-Infecciosos/farmacologia , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Proteína C/efeitos adversos , Proteína C/farmacologia , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Sepse/epidemiologia , Sepse/fisiopatologia , Índice de Gravidade de Doença , Choque Séptico/tratamento farmacológico
10.
Biophys Chem ; 95(2): 109-24, 2002 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11897150

RESUMO

Protein C is a vitamin-K dependent zymogen of the anti-coagulant serine protease activated protein C (APC). In this paper, we report four lines of evidence that APC can activate protein C in pooled normal plasma, and purified protein C. First, the addition of APC to protein C-deficient plasma supplemented with protein C produces a prolongation of the clotting time of plasma that is proportional to the amount of protein C. This behavior was observed with APC from the Chromogenix APC resistance kit (Dia Pharm, Franklin, OH, USA) and from APC derived from the thrombin activation of human protein C (Enzyme Research Laboratories, South Bend, IN, USA). Secondly, using immunoblotting after gel electrophoresis, the disappearance of epitopes for monoclonal antibodies that recognize protein C but not APC indicates a time course for the activation by APC of protein C in pooled normal plasma and protein C purified from plasma. Thirdly, the same time course for the disappearance of protein C specific epitope can be followed using ELISA. Finally, protein C can be activated by APC as indicated by the increase in APC specific synthetic substrate Tryp-Arg-Arg-p nitroaniline hydrolysis. Kinetic data indicate a value of 4.7+/-0.4 mM(-1) s(-1) for the activation of protein C by APC under physiological conditions and in the presence of calcium. These observations document that APC must function not only in the inactivation of activated factors V and VIII, but also in the activation of protein C. This additional action of APC may be important to consider more broadly because of APC in the treatment of sepsis.


Assuntos
Anticoagulantes/química , Proteína C/química , Serina Endopeptidases/química , Anticorpos Monoclonais/química , Anticoagulantes/antagonistas & inibidores , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Técnicas In Vitro , Indicadores e Reagentes , Cinética , Proteína C/antagonistas & inibidores , Proteína C/farmacologia , Inibidores de Serina Proteinase/farmacologia
12.
J Clin Invest ; 101(3): 667-76, 1998 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9449701

RESUMO

Activated protein C (APC) is a potent physiologic anticoagulant with profibrinolytic properties, and has been shown to prevent thrombosis in different experimental models. We investigated the effect of human APC on thrombin-induced thromboembolism in mice, a model of acute intravascular fibrin deposition leading to death within minutes. APC given intravenously (i.v.) as a bolus 2 min before thrombin challenge (1,250 U/kg) reduced mortality in a dose-dependent manner despite the lack of thrombin inhibitor activity. Significant inhibition of thrombin-induced death was observed at the dose of 0.05 mg/kg, and maximal protection was obtained with 2 mg/kg (> 85% reduction in mortality rate). Histology of lung tissue revealed that APC treatment (2 mg/kg) reduced significantly vascular occlusion rate (from 89.2 to 46.6%, P < 0.01). The protective effect of APC was due to the inhibition of endogenous thrombin formation as indicated by the fact that (a) the injection of human thrombin caused a marked decrease in the coagulation factors of the intrinsic and common pathways (but not of Factor VII), suggesting the activation of blood clotting via the contact system; (b) APC pretreatment reduced markedly prothrombin consumption; (c) the lethal effect of thrombin was almost abolished when the animals were made deficient in vitamin K-dependent factors by warfarin treatment, and could be restored only by doubling the dose of thrombin, indicating that the generation of endogenous thrombin contributes significantly to death; and (d) APC failed to protect warfarin-treated animals, in which mortality is entirely due to injected thrombin, even after protein S supplementation. Other results suggest that APC protects from thrombin-induced thromboembolism by rendering the formed fibrin more susceptible to plasmin degradation rather than by reducing fibrin formation: in thrombin-treated mice, fibrinogen consumption was not inhibited by APC; and inhibition of endogenous fibrinolysis by epsilon-aminocaproic or tranexamic acid resulted in a significant reduction of the protective effect of APC. Since APC did not enhance plasma fibrinolytic activity, as assessed by the measurement of plasminogen activator (PA) or PA inhibitor (PAI) activities, PAI-1 antigen, or 125I-fibrin degrading activity, we speculate that the inhibition of additional (endogenous) thrombin formation by APC interrupts thrombin-dependent mechanisms that make fibrin clots more resistant to lysis, so that the intravascular deposited fibrin can be removed more rapidly by the endogenous fibrinolytic system.


Assuntos
Anticoagulantes/farmacologia , Coagulantes/farmacologia , Fibrinolíticos/farmacologia , Proteína C/farmacologia , Embolia Pulmonar/prevenção & controle , Trombina/biossíntese , Animais , Anticoagulantes/administração & dosagem , Coagulantes/administração & dosagem , Modelos Animais de Doenças , Ativação Enzimática , Fibrina/metabolismo , Fibrinolíticos/administração & dosagem , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Proteína C/administração & dosagem , Embolia Pulmonar/mortalidade , Trombina/administração & dosagem , Trombina/efeitos dos fármacos , Trombina/farmacologia
13.
Haemostasis ; 26(4): 203-9, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8872131

RESUMO

The importance of bleeding as a complication of anticoagulant therapy is clearly recognized. We previously reported that amelioration of hemorrhage associated with disseminated intravascular coagulation by the human activated protein C (APC) was greater than that by heparin. In this study, we compared the bleeding complication of intravenously administered APC and heparin in rabbits, and also estimated primary hemostasis. When both anticoagulants were intravenously infused, the bleeding time from a punctured ear vein was prolonged dose-dependently. However, at doses which prolonged the activated partial thromboplastin time nearly equally, the prolongation of bleeding was greater in heparin-administered rabbits. Blood withdrawn from heparin-administered animals showed increases in in vitro bleeding parameters which correlated with the in vivo bleeding time. However, only small changes were observed in the blood withdrawn from APC-administered animals. Both drugs induced either no change or only a slight decrease in the platelet count, hematocrit and fibrinogen content. These observations suggest that APC may be a more useful anticoagulant than heparin since it causes less bleeding tendency.


Assuntos
Anticoagulantes/toxicidade , Testes de Coagulação Sanguínea/instrumentação , Hemorragia/induzido quimicamente , Heparina/toxicidade , Proteína C/toxicidade , Animais , Anticoagulantes/farmacologia , Tempo de Sangramento , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Ativação Enzimática , Fibrinogênio/análise , Hematócrito , Heparina/farmacologia , Humanos , Infusões Intravenosas , Masculino , Tempo de Tromboplastina Parcial , Contagem de Plaquetas/efeitos dos fármacos , Proteína C/metabolismo , Proteína C/farmacologia , Coelhos
14.
Thromb Res ; 82(2): 147-57, 1996 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9163068

RESUMO

Activated protein C (APC) possesses species specificity in its anticoagulant activity. Human APC exerts only weak activity in rat plasma compared with that in human plasma. The present study was undertaken to estimate the difference in interaction of human and rat factors with human APC and to assess the cause of the species specificity. Human or rat protein S (PS), factor V, or factor VIII was used to supplement human plasma depleted of each respective factor, and the anticoagulant activity of human APC was measured in term of the elongation of activated partial thromboplastin time (APTT). The activity of human APC in rat PS- or factor V-supplemented plasma was weaker than that in the human PS- or factor V-supplemented plasma. Furthermore, using purified human and rat factor V, human APC showed weaker inactivation of rat factor V than human factor V. Equal anticoagulant activity was observed in human or rat factor VIII-supplemented plasma. And there was a little difference in the interaction of APC with its inhibitors in human or rat plasma during a few minutes of incubation as judged by measurement of residual activity by an enzyme capture assay. From these results factor V as well as PS seems to play a major role in the species specificity of APC.


Assuntos
Anticoagulantes/farmacologia , Fator V/fisiologia , Proteína C/farmacologia , Proteína S/fisiologia , Animais , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática , Estudos de Avaliação como Assunto , Humanos , Tempo de Tromboplastina Parcial , Ratos , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA