Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nat Commun ; 11(1): 2992, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32532974

RESUMO

Activated protein C (APC) is a plasma serine protease with antithrombotic and cytoprotective functions. Based on the hypothesis that specific inhibition of APC's anticoagulant but not its cytoprotective activity can be beneficial for hemophilia therapy, 2 types of inhibitory monoclonal antibodies (mAbs) are tested: A type I active-site binding mAb and a type II mAb binding to an exosite on APC (required for anticoagulant activity) as shown by X-ray crystallography. Both mAbs increase thrombin generation and promote plasma clotting. Type I blocks all APC activities, whereas type II preserves APC's cytoprotective function. In normal monkeys, type I causes many adverse effects including animal death. In contrast, type II is well-tolerated in normal monkeys and shows both acute and prophylactic dose-dependent efficacy in hemophilic monkeys. Our data show that the type II mAb can specifically inhibit APC's anticoagulant function without compromising its cytoprotective function and offers superior therapeutic opportunities for hemophilia.


Assuntos
Anticorpos Monoclonais/farmacologia , Hemofilia A/prevenção & controle , Fragmentos Fab das Imunoglobulinas/imunologia , Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Animais , Anticorpos Monoclonais/classificação , Anticorpos Monoclonais/imunologia , Tempo de Sangramento , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Cristalografia por Raios X , Hemofilia A/sangue , Hemorragia/prevenção & controle , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Macaca fascicularis , Masculino , Proteína C/química , Proteína C/imunologia , Proteína C/metabolismo , Inibidor da Proteína C/sangue , Inibidor da Proteína C/farmacocinética
2.
Exp Cell Res ; 391(1): 111987, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32240661

RESUMO

BACKGROUND: The protein plasminogen activator inhibitor-1 (PAI-1), an inhibitor specific for urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA), has been shown to have a key role in cancer metastases. Currently, it is unknown as to whether the exocellular inhibition of PAI-1 can inhibit the migration of cancer cells. METHODS: By fusing the mutated serine protease domain (SPD) of uPA and human serum albumin (HSA), PAItrap3, a protein that traps PAI-1, was synthesized and experiments were conducted to determine if exocellular PAItrap3 attenuates PAI-1-induced cancer cell migration in vitro. RESULTS: PAItrap3 (0.8 µM) significantly inhibited the motility of MCF-7, MDA-MB-231, HeLa and 4T1 cancer cells, by 90%, 50%, 30% and 20%, respectively, without significantly altering their proliferation. The PAI-1-induced rearrangement of F-actin was significantly inhibited by PAItrap3, which produced a decrease in the number of cell protrusions by at least 20%. CONCLUSIONS: In vitro, PAItrap3 inhibited PAI-1-induced cancer cell migration, mainly through inhibiting the rearrangement of F-actin. Overall, these results, provided they can be extrapolated to humans, suggest that the PAItrap3 protein could be used as an exocellular inhibitor to attenuate cancer metastases.


Assuntos
Actinas/genética , Movimento Celular/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Inibidor da Proteína C/farmacologia , Actinas/antagonistas & inibidores , Actinas/metabolismo , Sítios de Ligação , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Células HeLa , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Células MCF-7 , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Pichia/genética , Pichia/metabolismo , Inibidor 1 de Ativador de Plasminogênio/química , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Inibidor da Proteína C/química , Inibidor da Proteína C/genética , Inibidor da Proteína C/metabolismo , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
3.
Blood ; 129(1): 105-113, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27789479

RESUMO

Hemophilia is a bleeding disorder caused by deficiency in factors VIII or IX, the two components of the intrinsic Xase complex. Treatment with replacement factor can lead to the development of inhibitory antibodies, requiring the use of bypassing agents such as factor VIIa and factor concentrates. An alternative approach to bypass the Xase complex is to inhibit endogenous anticoagulant activities. Activated protein C (APC) breaks down the complex that produces thrombin by proteolytically inactivating factor Va. Defects in this mechanism (eg, factor V Leiden) are associated with thrombosis but result in less severe bleeding when co-inherited with hemophilia. Selective inhibition of APC might therefore be effective for the treatment of hemophilia. The endogenous inhibitors of APC are members of the serpin family: protein C inhibitor (PCI) and α1-antitrypsin (α1AT); however, both exhibit poor reactivity and selectivity for APC. We mutated residues in and around the scissile P1-P1' bond in PCI and α1AT, resulting in serpins with the desired specificity profile. The lead candidate was shown to promote thrombin generation in vitro and to restore fibrin and platelet deposition in an intravital laser injury model in hemophilia B mice. The power of targeting APC was further demonstrated by the complete normalization of bleeding after a severe tail clip injury in these mice. These results demonstrate that the protein C anticoagulant system can be successfully targeted by engineered serpins and that administration of such agents is effective at restoring hemostasis in vivo.


Assuntos
Hemofilia B/tratamento farmacológico , Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Serpinas/farmacologia , Animais , Modelos Animais de Doenças , Desenho de Fármacos , Eletroforese em Gel de Poliacrilamida , Humanos , Camundongos
4.
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
5.
Thromb Res ; 133(6): 1105-14, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24513148

RESUMO

INTRODUCTION: Activated protein C (APC) is the central enzyme of the anticoagulant protein C pathway. Low concentrations of APC circulate in plasma and are believed to contribute to the maintenance of a normal haemostatic balance. MATERIALS AND METHODS: We have used a structure-based virtual screening approach to discover small drug-like molecules that inhibit the interaction between APC and its substrate FVa through inhibition of a predominant APC exosite, known to be involved in FVa substrate binding. We have combined in silico selection with functional screening and direct binding analysis to identify novel molecules and to ascertain and characterize the inhibition of the interaction between APC and FVa. RESULTS: We have identified a number of novel molecules that bind to APC and protein C with Kd values in the range of 10(-3)- 10(-5)M. Inhibition by these molecules is incomplete, which most likely reflects the extended surface that is involved in the interaction between APC and its substrates. Direct binding of hit molecules to variant APC molecules that were mutated in the targeted binding site revealed that several of the molecules presented a 100-500 fold lower affinity for the variant molecule, suggesting that these molecules indeed bind the exosite of APC. CONCLUSIONS: The protein-protein interaction inhibitors discovered here, could function as starting molecules for further development of small molecules with anti-APC properties. Such molecules may be of clinical interest, in particular in individuals where thrombin formation is compromised and the haemostatic balance is tipped towards bleeding tendencies, such as in haemophilia A.


Assuntos
Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Sítios de Ligação , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Modelos Moleculares , Proteína C/química , Proteína C/metabolismo , Inibidor da Proteína C/química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
6.
Bioorg Med Chem Lett ; 24(3): 821-7, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24418773

RESUMO

A design strategy was used to identify inhibitors of activated protein C with selectivity over thrombin featured by a basic and/or aromatic functionality for binding to the S2 pocket. Our strongest inhibitor showed an IC50-material value and selectivity for APC vs thrombin similar to a compound previously reported in the literature. However, in contrast to the reference compound, our compound showed a retained coagulant effect of thrombin with increasing substrate concentration in a modified Calibrated Automated Thrombogram (CAT) method. This was likely related to our compound being inactive against FVIIa, while the reference compound showed an IC50 of 8.9 µM. Thus, the higher selectivity of our compound against all relevant coagulation factors likely explained its higher therapeutic potential in comparison to the reference compound. The data indicate that at least a 100-fold selectivity over other serine proteases in the coagulation cascade will be required for an effective APC inhibitor.


Assuntos
Desenho de Fármacos , Inibidor da Proteína C/síntese química , Inibidor da Proteína C/farmacologia , Trombina/antagonistas & inibidores , Sítios de Ligação , Coagulantes/síntese química , Coagulantes/química , Coagulantes/farmacologia , Hemofilia A/tratamento farmacológico , Concentração Inibidora 50 , Ligação Proteica/efeitos dos fármacos , Inibidor da Proteína C/química , Relação Estrutura-Atividade , Especificidade por Substrato
7.
PLoS One ; 7(6): e39262, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22723979

RESUMO

The serine protease inhibitor protein C inhibitor (PCI) is expressed in many human tissues and exhibits broad protease reactivity. PCI binds glycosaminoglycans and certain phospholipids, which modulate its inhibitory activity. Enteropeptidase (EP) is a type II transmembrane serine protease mainly found on the brush border membrane of epithelial cells in the duodenum, where it activates trypsinogen to initiate the digestion of food proteins. Some active EP is also present in duodenal fluid and has been made responsible for causing pancreatitis in case of duodeno-pancreatic reflux. Together with its substrate trypsinogen, EP is furthermore present in the epidermis and in some cancer cells. In this report, we show that PCI inhibited EP with an apparent 2nd order rate constant of 4.48 × 10(4) M(-1) s(-1). Low molecular weight (LMWH) and unfractionated heparin (UFH) slightly reduced the inhibitory effect of PCI. The SI (stoichiometry of inhibition) value for the inhibition of EP by PCI was 10.8 in the absence and 17.9 in the presence of UFH (10 U/ml). By inhibiting trypsin, chymotrypsin, and additionally EP, PCI might play a role in the protection of the pancreas from autodigestion. Furthermore the interaction of PCI with EP may influence the regulation of epithelial differentiation.


Assuntos
Enteropeptidase/metabolismo , Inibidor da Proteína C/metabolismo , Inibidores de Serina Proteinase/metabolismo , Animais , Antitrombinas/metabolismo , Antitrombinas/farmacologia , Bovinos , Relação Dose-Resposta a Droga , Enteropeptidase/antagonistas & inibidores , Heparina/farmacologia , Humanos , Camundongos , Ligação Proteica , Inibidor da Proteína C/farmacologia , Inibidores de Serina Proteinase/farmacologia , Serpinas/metabolismo , Serpinas/farmacologia
8.
J Biol Chem ; 287(19): 15739-48, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22416129

RESUMO

The activation of coagulation factors V and X by Russell's viper venom (RVV) has been implicated in the development of consumptive coagulopathies in severely envenomed patients. However, factor Va is prone to inactivation by activated protein C (APC), an important serine protease that negatively regulates blood coagulation. It is therefore hypothesized that APC may be down-regulated by some of the venom components. In this study, we managed to isolate a potent Kunitz-type APC inhibitor, named DrKIn-I. Using chromogenic substrate, DrKIn-I dose-dependently inhibited the activity of APC. Heparin potentiated the inhibition and reduced the IC(50) of DrKIn-I by 25-fold. DrKIn-I, together with heparin, also protected factor Va from APC-mediated inactivation. Using surface plasmon resonance, DrKIn-I exhibited fast binding kinetics with APC (association rate constant = 1.7 × 10(7) M(-1) s(-1)). Direct binding assays and kinetic studies revealed that this inhibition (K(i) = 53 pM) is due to the tight binding interactions of DrKIn-I with both heparin and APC. DrKIn-I also effectively reversed the anticoagulant activity of APC and completely restored the thrombin generation in APC-containing plasma. Furthermore, although the injection of either DrKIn-I or RVV-X (the venom factor X-activator) into ICR mice did not significantly deplete the plasma fibrinogen concentration, co-administration of DrKIn-I with RVV-X resulted in complete fibrinogen consumption and the deposition of fibrin thrombi in the glomerular capillaries. Our results provide new insights into the pathogenesis of RVV-induced coagulopathies and indicate that DrKIn-I is a novel APC inhibitor that is associated with potentially fatal thrombotic complications in Russell's viper envenomation.


Assuntos
Daboia/metabolismo , Coagulação Intravascular Disseminada/metabolismo , Heparina/farmacologia , Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Coagulação Sanguínea/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Coagulação Intravascular Disseminada/induzido quimicamente , Sinergismo Farmacológico , Humanos , Cinética , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Ligação Proteica , Proteína C/metabolismo , Inibidor da Proteína C/genética , Inibidor da Proteína C/metabolismo , Daboia/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Venenos de Víboras/genética , Venenos de Víboras/metabolismo , Venenos de Víboras/farmacologia , Venenos de Víboras/intoxicação
9.
PLoS One ; 6(12): e29011, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22205989

RESUMO

Protein C inhibitor (PCI) is a serpin type of serine protease inhibitor that is found in many tissues and fluids in human, including blood plasma, seminal plasma and urine. This inhibitor displays an unusually broad protease specificity compared with other serpins. Previous studies have shown that the N-glycan(s) and the NH2-terminus affect some blood-related functions of PCI. In this study, we have for the first time determined the N-glycan profile of seminal plasma PCI, by mass spectrometry. The N-glycan structures differed markedly compared with those of both blood-derived and urinary PCI, providing evidence that the N-glycans of PCI are expressed in a tissue-specific manner. The most abundant structure (m/z 2592.9) had a composition of Fuc3Hex5HexNAc4, consistent with a core fucosylated bi-antennary glycan with terminal Lewis(x). A major serine protease in semen, prostate specific antigen (PSA), was used to evaluate the effects of N-glycans and the NH2-terminus on a PCI function related to the reproductive tract. Second-order rate constants for PSA inhibition by PCI were 4.3±0.2 and 4.1±0.5 M⁻¹ s⁻¹ for the natural full-length PCI and a form lacking six amino acids at the NH2-terminus, respectively, whereas these constants were 4.8±0.1 and 29±7 M⁻¹ s⁻¹ for the corresponding PNGase F-treated forms. The 7-8-fold higher rate constants obtained when both the N-glycans and the NH2-terminus had been removed suggest that these structures jointly affect the rate of PSA inhibition, presumably by together hindering conformational changes of PCI required to bind to the catalytic pocket of PSA.


Assuntos
Regulação da Expressão Gênica , Polissacarídeos , Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Humanos , Masculino , Modelos Moleculares , Especificidade de Órgãos , Antígeno Prostático Específico/antagonistas & inibidores , Antígeno Prostático Específico/química , Antígeno Prostático Específico/metabolismo , Inibidor da Proteína C/sangue , Inibidor da Proteína C/farmacologia , Conformação Proteica , Sêmen/metabolismo
10.
J Thromb Haemost ; 9(11): 2262-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21920012

RESUMO

BACKGROUND: Replacement therapy for hemophilic patient treatment is costly, because of the high price of pharmacologic products, and is not affordable for the majority of patients in developing countries. OBJECTIVE: To generate and evaluate low molecular weight agents that could be useful for hemophilia treatment. METHODS: Potential agents were generated by synthesizing specific inhibitors [6-(Lys-Lys-Thr-[homo]Arg)amino-2-(Lys[carbobenzoxy]-Lys[carbobenzoxy]-O-benzyl)naphthalenesulfonamide] (PNASN-1)] for activated protein C (APC) and tested in plasma and fresh blood from hemophilia A patients. RESULTS: In the activated partial thromboplastin time-based APC resistance assay, PNASN-1 partially neutralized the effect of APC. In calibrated automated thrombography, PNASN-1 neutralized the effect of APC on thrombin generation in normal and congenital factor VIII-deficient plasma (FVIII:C < 1%). The addition of PNASN-1 to tissue factor-triggered (5 pm) contact pathway-inhibited fresh blood from 15 hemophilia A patients with various degrees of FVIII deficiency (FVIII:C < 1-51%) increased the maximum level of thrombin generated from 78 to 162 nm, which approached that observed in blood from a healthy individual (201 nm). PNASN-1 also caused a 47% increase in clot weight in hemophilia A blood. CONCLUSIONS: Specific APC inhibitors compensate to a significant extent for FVIII deficiency, and could be used for hemophilia treatment.


Assuntos
Hemofilia A/tratamento farmacológico , Inibidor da Proteína C/farmacologia , Trombina/biossíntese , Sangue/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Testes de Coagulação Sanguínea , Estudos de Casos e Controles , Avaliação de Medicamentos , Humanos , Tempo de Tromboplastina Parcial , Inibidor da Proteína C/síntese química , Trombina/efeitos dos fármacos
11.
Blood ; 118(10): 2882-8, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21772054

RESUMO

Heparin-induced thrombocytopenia (HIT) is caused by antibodies that recognize complexes between platelet factor 4 (PF4) and heparin or glycosaminoglycan side chains. These antibodies can lead to a limb- and life-threatening prothrombotic state. We now show that HIT antibodies are able to inhibit generation of activated protein C (aPC) by thrombin/thrombomodulin (IIa/TM) in the presence of PF4. Tetrameric PF4 potentiates aPC generation by formation of complexes with chondroitin sulfate (CS) on TM. Formation of these complexes occurs at a specific molar ratio of PF4 to glycosaminoglycan. This observation and the finding that the effect of heparin on aPC generation depends on the concentration of PF4 suggest similarity between PF4/CS complexes and those that bind HIT antibodies. HIT antibodies reduced the ability of PF4 to augment aPC formation. Cationic protamine sulfate, which forms similar complexes with heparin, also enhanced aPC generation, but its activity was not blocked by HIT antibodies. Our studies provide evidence that complexes formed between PF4 and TM's CS may play a physiologic role in potentiating aPC generation. Recognition of these complexes by HIT antibodies reverses the PF4-dependent enhancement in aPC generation and may contribute to the prothrombotic nature of HIT.


Assuntos
Anticorpos Monoclonais/farmacologia , Heparina/efeitos adversos , Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Proteína C/metabolismo , Protrombina/metabolismo , Trombocitopenia/induzido quimicamente , Trombocitopenia/metabolismo , Adulto , Animais , Anticoagulantes/efeitos adversos , Células Cultivadas , Glicosaminoglicanos/metabolismo , Humanos , Integrases/metabolismo , Rim/citologia , Rim/imunologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator Plaquetário 4/fisiologia , Multimerização Proteica , Proteínas Recombinantes/metabolismo , Trombina/metabolismo , Trombocitopenia/imunologia , Trombomodulina/metabolismo
12.
Crit Care ; 14(3): 156, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20497609

RESUMO

An animal model of H1N1 influenza demonstrates that this infection is associated with pulmonary and systemic activation of coagulation and impairment of fibrinolysis in addition to systemic inflammation and intense neutrophil influx into the lung. Activated protein C attenuates coagulation activation and restores fibrinolytic capacity but has little effect on inflammation or survival from this infection. This animal model points to a profound inflammatory state developing in H1N1 infection that impacts mortality. Additional modifications to the model and the type and amount of activated protein C dosing will provide the data to determine the possible use of activated protein C as a therapy in human H1N1 infection.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/fisiopatologia , Inibidor da Proteína C/uso terapêutico , Inibidores de Serina Proteinase/uso terapêutico , Animais , Fibrinólise/efeitos dos fármacos , Humanos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Camundongos , Modelos Animais , Infecções por Orthomyxoviridae , Inibidor da Proteína C/administração & dosagem , Inibidor da Proteína C/farmacologia , Inibidores de Serina Proteinase/administração & dosagem , Inibidores de Serina Proteinase/farmacologia
13.
Biochim Biophys Acta ; 1800(6): 580-90, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20230872

RESUMO

BACKGROUND: Protein C inhibitor (PCI) is a plasma serine protease inhibitor (serpin) that regulates several serine proteases in coagulation including thrombin and activated protein C. However, the physiological role of PCI remains under investigation. The cysteine protease, cathepsin L, has a role in many physiological processes including cardiovascular diseases, blood vessel remodeling, and cancer. METHODS AND RESULTS: We found that PCI inhibits cathepsin L with an inhibition rate (k(2)) of 3.0x10(5)M(-)(1)s(-)(1). Whereas, the PCI P1 mutant (R354A) inhibits cathepsin L at rates similar to wild-type PCI, mutating the P2 residue results in a slight decrease in the rate of inhibition. We then assessed the effect of PCI and cathepsin L on the migration of human breast cancer (MDA-MB-231) cells. Cathepsin L was expressed in both the cell lysates and conditioned media of MDA-MB-231 cells. Wound-induced and transwell migration of MDA-MB-231 cells was inhibited by exogenously administered wtPCI and PCI P1 but not PCI P14 mutant. In addition, migration of MDA-MB-231 cells expressing wtPCI was significantly decreased compared to non-expressing MDA-MB-231 cells or MDA-MB-231 cells expressing the PCI P14 mutant. Downregulation of cathepsin L by either a specific cathepsin L inhibitor or siRNA technology also resulted in a decrease in the migration of MDA-MB-231 cells. CONCLUSIONS: Overall, our data show that PCI regulates tumor cell migration partly by inhibiting cathepsin L. GENERAL SIGNIFICANCE: Consequently, inhibiting cathepsin L by serpins like PCI may be a new pathway of regulating hemostasis, cardiovascular and metastatic diseases.


Assuntos
Neoplasias da Mama/patologia , Catepsina L/metabolismo , Metástase Neoplásica , Inibidor da Proteína C/farmacologia , Sequência de Bases , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Primers do DNA , Imunofluorescência , Humanos , Mutagênese Sítio-Dirigida
14.
PLoS Pathog ; 5(12): e1000698, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20019810

RESUMO

Protein C inhibitor (PCI) is a heparin-binding serine proteinase inhibitor belonging to the family of serpin proteins. Here we describe that PCI exerts broad antimicrobial activity against bacterial pathogens. This ability is mediated by the interaction of PCI with lipid membranes, which subsequently leads to their permeabilization. As shown by negative staining electron microscopy, treatment of Escherichia coli or Streptococcus pyogenes bacteria with PCI triggers membrane disruption followed by the efflux of bacterial cytosolic contents and bacterial killing. The antimicrobial activity of PCI is located to the heparin-binding site of the protein and a peptide spanning this region was found to mimic the antimicrobial activity of PCI, without causing lysis or membrane destruction of eukaryotic cells. Finally, we show that platelets can assemble PCI on their surface upon activation. As platelets are recruited to the site of a bacterial infection, these results may explain our finding that PCI levels are increased in tissue biopsies from patients suffering from necrotizing fasciitis caused by S. pyogenes. Taken together, our data describe a new function for PCI in innate immunity.


Assuntos
Imunidade Inata , Lipossomos/metabolismo , Inibidor da Proteína C/imunologia , Anti-Infecciosos , Sítios de Ligação , Plaquetas/fisiologia , Membrana Celular/metabolismo , Escherichia coli/imunologia , Humanos , Ativação Plaquetária , Inibidor da Proteína C/farmacologia , Streptococcus pyogenes/imunologia
15.
Thromb Res ; 122(6): 838-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18055002

RESUMO

In the past decade development of aptamer technology is poised into several fields with various clinical applications. With this progress, aptamer based anti-coagulant agents are evolved and continuing their applications with clinical trials. At present several anti-coagulant aptamers are available against different proteins from blood coagulation cascade. In this review, the mechanism and functions of anti-coagulant aptamers, as an alternate candidate to other available anti-coagulant agents are discussed.


Assuntos
Anticoagulantes/farmacologia , Aptâmeros de Nucleotídeos/farmacologia , Técnica de Seleção de Aptâmeros , Animais , Antitrombinas/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Fator IX/antagonistas & inibidores , Fator VIIa/antagonistas & inibidores , Humanos , Inibidor da Proteína C/farmacologia
16.
Semin Thromb Hemost ; 33(7): 667-72, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18000793

RESUMO

Activated protein C (APC) and protein C inhibitor (PCI) are the major components of the anticoagulant protein C pathway. Recently, APC and PCI have been demonstrated to play many roles not only in the regulation of hemostasis but also in cell inflammation, proliferation, apoptosis, tumor cell migration, invasion, and metastasis. Here we summarize the role of APC and PCI in malignancy. APC increases migration of ovarian cancer cells and choriocarcinoma cells in a Transwell invasion assay in the presence of plasminogen activator inhibitor (PAI)-1; this finding suggests that APC stimulates urokinase-type plasminogen activator (uPA) by forming a complex with PAI-1 leading to activation of extracellular matrix proteases and increased invasion. It was recently reported that APC, independent of PAI-1, may increase invasion and chemotaxis of breast cancer cells by activating specific signaling pathways through endothelial protein C receptor (EPCR) and protease-activated receptor (PAR)-1. APC also increased proliferation of vascular endothelial cells and angiogenesis by EPCR-mediated activation of mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K), and endothelial nitric oxide synthase (eNOS) pathways. On the other hand, we have previously reported that both uPA and PCI are synthesized in renal proximal tubular epithelial cells (RPTECs) and that PCI expression in RPTEC-derived tumor cells is significantly decreased compared with normal RPTECs. The RPTEC-derived renal carcinoma cell line Caki-1 also showed decreased expression of PCI. PCI inhibited in vitro invasive activity of Caki-1 and breast cancer cells by its protease inhibitory activity. However, PCI was found to inhibit the growth and metastatic potential of breast cancer cells independent of its protease inhibitory activity in severe combined immunodeficient mice. PCI can also inhibit angiogenesis in vivo and in vitro assays independent of its protease inhibitory activity. Overall, these data show that APC promotes tumor cell invasion by EPCR-mediated and PAR-1-mediated protease activity and that PCI inhibits tumor cell invasion in vitro by its protease inhibitory activity and suppresses tumor cell growth, metastasis, and angiogenesis independent of its protease inhibitory activity.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Inibidor da Proteína C/metabolismo , Proteína C/metabolismo , Indutores da Angiogênese/sangue , Indutores da Angiogênese/metabolismo , Inibidores da Angiogênese/sangue , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Anticoagulantes/sangue , Anticoagulantes/metabolismo , Anticoagulantes/farmacologia , Antígenos CD/metabolismo , Receptor de Proteína C Endotelial , Fibrinolíticos/sangue , Fibrinolíticos/metabolismo , Fibrinolíticos/farmacologia , Humanos , Metástase Neoplásica , Neovascularização Patológica , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor da Proteína C/sangue , Inibidor da Proteína C/farmacologia , Receptor PAR-1/metabolismo , Receptores de Superfície Celular/metabolismo , Inibidores de Serina Proteinase/sangue , Inibidores de Serina Proteinase/metabolismo , Inibidores de Serina Proteinase/farmacologia
17.
J Thromb Haemost ; 5(7): 1486-92, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17635698

RESUMO

BACKGROUND: Protein C inhibitor (PCI) and antithrombin (AT) are serine protease inhibitors (serpins) that inhibit a wide array of blood coagulation serine proteases including thrombin. OBJECTIVE: Fifty-five Ala-scanned recombinant thrombin mutants were used to determine thrombin residues important for inhibition by PCI with and without the cofactors heparin and thrombomodulin (TM) and compared with the prototypical serpin, AT. RESULTS: Residues around the active site (Tyr50 and Glu202) and the sodium-binding site (Glu229 and Arg233) were required for thrombin inhibition by PCI with and without cofactors. Exosite-2 residues (Arg89, Arg93, Glu94, Arg98, Arg245, Arg248, and Gln251) were critical for heparin-accelerated inhibition of thrombin by PCI. Exosite-1 residues (especially Lys65 and Tyr71) were required for enhanced PCI inhibition of thrombin-TM. Interestingly, we also found that the TM chondroitin sulfate moiety is not required for the approximately 150-fold enhanced rate of thrombin inhibition by PCI. Using the aforementioned thrombin exosite-2 mutants that were essential for heparin-catalyzed PCI-thrombin inhibition reactions we found no change in PCI inhibition rates for thrombin-TM. CONCLUSIONS: Collectively, these results show that (i) similar thrombin exosite-2 residues are critical for the heparin-catalyzed inhibition by PCI and AT, (ii) PCI and AT are different in their thrombin-TM inhibition properties, and (iii) PCI has a distinct advantage over AT in the regulation of the activity of thrombin-TM.


Assuntos
Heparina/metabolismo , Inibidor da Proteína C/farmacologia , Trombina/antagonistas & inibidores , Trombina/genética , Trombomodulina/metabolismo , Substituição de Aminoácidos , Sítios de Ligação/genética , Heparina/farmacologia , Humanos , Técnicas In Vitro , Modelos Moleculares , Complexos Multiproteicos , Mutagênese Sítio-Dirigida , Inibidor da Proteína C/genética , Conformação Proteica , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Trombina/química , Trombina/metabolismo
18.
J Thromb Haemost ; 5(7): 1477-85, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17456194

RESUMO

BACKGROUND: Hepatocyte growth factor (HGF) plays an important role in tissue repair and regeneration. HGF activator (HGFA), a factor XIIa-like serine protease, activates HGF precursor to HGF. The precursor of HGFA, proHGFA, is activated by thrombin generated at sites of tissue injury. It is known that protein C inhibitor (PCI), an inhibitor of activated protein C (APC), also inhibits thrombin-thrombomodulin (TM) complex. OBJECTIVES: In the present study we evaluated the effect of PCI on thrombin-catalyzed proHGFA activation in the presence of TM, and on HGFA activity. RESULTS: PCI did not inhibit thrombin-TM-mediated proHGFA activation, but it directly inhibited activated HGFA by forming an enzyme inhibitor complex. The second-order rate constants (m(-1) min(-1)) of the reaction between HGFA and PCI in the presence or absence of heparin (10 U mL(-1)) were 4.3 x 10(6) and 4.0 x 10(6), respectively. The inhibition of HGFA by PCI resulted in a significant decrease of HGFA-catalyzed activation of HGF precursor. Exogenous HGFA added to normal human plasma formed a complex with plasma PCI, and this complex formation was competitively inhibited by APC in the presence of heparin, but very weakly in the absence of heparin. We also demonstrated using recombinant R362A-PCI that Arg362 residue of PCI is important for HGFA inhibition by PCI as judged from the three-dimensional structures constructed using docking models of PCI and HGFA or APC. CONCLUSION: These observations indicate that PCI is a potent inhibitor of activated HGFA, suggesting a novel function for PCI in the regulation of tissue repair and regeneration.


Assuntos
Inibidor da Proteína C/farmacologia , Serina Endopeptidases/efeitos dos fármacos , Adulto , Sequência de Bases , DNA Complementar/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Técnicas In Vitro , Substâncias Macromoleculares , Pessoa de Meia-Idade , Modelos Moleculares , Proteína C/metabolismo , Proteína C/farmacologia , Inibidor da Proteína C/química , Inibidor da Proteína C/genética , Precursores de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Serina Endopeptidases/química , Trombina/metabolismo , Trombina/farmacologia , Trombomodulina/metabolismo
19.
Semin Thromb Hemost ; 33(1): 41-5, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17253188

RESUMO

Protein C inhibitor (PCI) is a heparin-dependent serine protease inhibitor found in human plasma, urine, and other body fluids. In blood plasma, PCI is present at approximately 0.08 microM and inactivates activated protein C and other coagulation and fibrinolytic enzymes. In seminal plasma, PCI is present at 2.2 to 3.7 microM. The main sources of seminal PCI are the seminal vesicles, where it remains fully active. Following ejaculation, PCI completely looses its activity in approximately 2 hours, when it partially complexes with prostate-specific antigen, two plasminogen activators (urokinase-type and tissue-type plasminogen activators), and tissue kallikrein. PCI is also present in an active form in follicular fluid at approximately 0.1 microM. Purified functionally active human blood plasma-derived as well as inactive semen-derived PCI inhibited both binding and penetration of zona-free hamster oocytes by human sperm. The binding inhibition by PCI was dose dependent. A concentration of 0.04 microM PCI (approximately 100-fold lower than that present in seminal plasma) inhibited 50% of the binding and penetration ability. Given that capacitated sperm used for in vitro fertilization usually contains more than 0.05 microM of PCI, fertilization rates might be significantly reduced. All of these data suggest that PCI is involved in human reproduction at several steps, including the fertilization process.


Assuntos
Antígeno Prostático Específico/metabolismo , Inibidor da Proteína C/metabolismo , Sêmen/metabolismo , Capacitação Espermática/fisiologia , Interações Espermatozoide-Óvulo/fisiologia , Animais , Cricetinae , Feminino , Fertilização in vitro , Humanos , Masculino , Ligação Proteica , Proteína C/metabolismo , Inibidor da Proteína C/química , Inibidor da Proteína C/farmacologia , Capacitação Espermática/efeitos dos fármacos , Interações Espermatozoide-Óvulo/efeitos dos fármacos
20.
J Zhejiang Univ Sci B ; 7(11): 899-905, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17048305

RESUMO

OBJECTIVE: To investigate the effect of activated protein C (APC) on inflammatory responses in human umbilical vein endothelial cells (HUVEC) stimulated with lipopolysaccharide (LPS). METHODS: The second passage of collagenase digested HUVEC was divided into the following groups: serum free medium control group (SFM control), phosphate buffer solution control group (PBS control), LPS group with final concentration of 1 microg/ml (LPS group), APC group with final concentration of 7 microg/ml, Pre-APC group (APC pretreatment for 30 min prior to LPS challenge), and Post-APC group (APC administration 30 min after LPS challenge). Supernatant was harvested at 0, 4, 8, 12 and 24 h after LPS challenge. Interleukin-6 (IL-6) and Interleukin-8 (IL-8) levels were analyzed with ELISA. Cells were harvested at 24 h after LPS challenge, and total RNA was extracted. Messenger RNA levels for IL-6 and IL-8 were semi-quantitatively determined by RT-PCR. RESULTS: Compared with control group, IL-6 and IL-8 levels steadily increased 4 to 24 h after LPS stimulation. APC treatment could increase LPS-induced IL-6 and IL-8 production. The mRNA levels of IL-6 and IL-8 exhibited a similar change. CONCLUSION: APC can further increase the level of IL-6 and IL-8 induced by LPS. The effect of these elevated cytokines is still under investigation.


Assuntos
Células Endoteliais/efeitos dos fármacos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lipopolissacarídeos/farmacologia , Inibidor da Proteína C/farmacologia , Proteína C/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Humanos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA