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










Base de dados
Intervalo de ano de publicação
1.
J Thromb Haemost ; 14(11): 2274-2286, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27589692

RESUMO

Essentials Dysregulated DNA and histone release can promote pathological immunothrombosis. Weibel-Palade bodies (WPBs) are sentinel-like organelles that respond to proinflammatory stimuli. Histones induce WPB exocytosis in a caspase, calcium and charge-dependent mechanism. A targetable axis may exist between DNA/histones and WPBs in inflammation and immunothrombosis. SUMMARY: Background Damage-associated molecular patterns (DAMPs), including molecules such as DNA and histones, are released into the blood following cell death. DAMPs promote a procoagulant phenotype through enhancement of thrombin generation and platelet activation, thereby contributing to immunothrombosis. Weibel-Palade bodies (WPBs) are dynamic endothelial cell organelles that contain procoagulant and proinflammatory mediators, such as von Willebrand factor (VWF), and are released in response to cell stresses. VWF mediates platelet adhesion and aggregation, and has been implicated as a procoagulant component of the innate immune response. Objective To determine the influence of histones and DNA on WPB release, and characterize their association in models of inflammation. Methods We treated C57BL/6J mice and cultured endothelial cells with histones (unfractionated, lysine-rich or arginine-rich) and DNA, and measured WPB exocytosis. We used inhibitors to determine a mechanism of histone-induced WPB release in vitro. We characterized the release of DAMPs and WPBs in response to acute and chronic inflammation in human and murine models. Results and conclusions Histones, but not DNA, induced the release of VWF (1.46-fold) from WBPs and caused thrombocytopenia (0.74-fold), which impaired arterial thrombus formation in mice. Histones induced WPB release from endothelial cells in a caspase-dependent, calcium-dependent and charge-dependent manner, and promoted platelet capture in a flow chamber model of VWF-platelet string formation. The levels of DAMPs and WPB-released proteins were elevated during inflammation, and were positively correlated in chronic inflammation. These studies showed that DAMPs can regulate the function and level of VWF by inducing its release from endothelial WPBs. This DAMP-WPB axis may propagate immunothrombosis associated with inflammation.


Assuntos
Exocitose , Histonas/metabolismo , Trombose/metabolismo , Corpos de Weibel-Palade/metabolismo , Animais , Arginina/química , Caspases/metabolismo , DNA/química , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Lisina/química , Camundongos , Camundongos Endogâmicos C57BL , Adesividade Plaquetária , Trombose/patologia
2.
Thromb Res ; 146: 119-125, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27354153

RESUMO

BACKGROUND: The rate of recurrent venous thromboembolism (VTE) in patients with a first unprovoked VTE who had a negative qualitative D-dimer test one month after stopping anticoagulant therapy was higher than expected in the D-dimer Optimal Duration Study (DODS). OBJECTIVES: To determine whether quantitative D-dimer levels using a low threshold, age- and sex-specific thresholds, or repeated measurements, would improve identification of patients at low risk of recurrent VTE. MATERIALS AND METHODS: D-dimer levels were quantified in banked samples from 307 patients in DODS who had a negative qualitative D-dimer test while on, and 1month after stopping, anticoagulant therapy and the rates of recurrent VTE were determined in patients with D-dimer levels below various predefined thresholds. RESULTS: The rate (per patient year) of recurrent VTE was: 5.9% with D-dimer levels<250µg/l at one month; 5.2% with D-dimer levels between 250 and 499µg/l at one month; 5.0% with D-dimer levels less than predefined age- and sex-specific thresholds at one month; and 6.3% when D-dimer levels were <500µg/l at both one and 7months after stopping anticoagulant therapy. These rates are similar to the overall event rate of 6.3% in patients who stopped treatment. CONCLUSIONS: Among unprovoked VTE patients who had a negative qualitative D-dimer test during and after anticoagulant therapy, low D-dimer thresholds, age and sex-adjusted thresholds or repeated measurements, did not identify subgroups with a very low rate of recurrence.


Assuntos
Anticoagulantes/uso terapêutico , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Tromboembolia Venosa/tratamento farmacológico , Estudos de Coortes , Feminino , Humanos , Masculino , Prognóstico , Recidiva , Medição de Risco , Fatores de Risco
3.
J Thromb Haemost ; 14(2): 366-74, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26663133

RESUMO

UNLABELLED: ESSENTIALS: It is unknown if thrombin activatable fibrinolysis inhibitor (TAFI) and protein C compete on cells. TAFI and protein C activation on endothelial cells was simultaneously quantified. TAFI and protein C do not compete for activation on endothelial cells. TAFI and protein C are independently recognized by the thrombin-thrombomodulin complex. BACKGROUND: When bound to thrombomodulin (TM), thrombin is a potent activator of protein C (PC) and thrombin activable fibrinolysis inhibitor (TAFI). By binding PC and presenting it to the thrombin-TM complex, endothelial cell PC receptor (EPCR) enhances PC activation. It is unknown whether PC and TAFI compete for the thrombin-TM complex on endothelial cells. OBJECTIVE: To compare PC and TAFI activation on the surface of cultured human endothelial cells in the absence or presence of JRK1535 and/or CTM1009, inhibitory antibodies directed against EPCR and TM, respectively, and to determine whether PC and TAFI compete with each other for activation. METHODS: PC and TAFI activation on endothelial cells were compared, and the effect of PC on TAFI activation and TAFI on PC activation was determined in the absence or presence of JRK1535 and/or CTM1009. RESULTS: In the absence of antibodies, activation of PC was four-fold faster than that of TAFI. Blocking EPCR with JRK1535 resulted in a 53-fold decrease in PC activation and no effect on TAFI activation. Blocking TM with CTM1009 inhibited both TAFI and PC activation. Neither TAFI nor PC competed with each other in the absence or presence of JRK1535. CONCLUSIONS: PC and TAFI are concurrently activated in a TM-dependent manner and do not compete for the thrombin-TM complex, raising the possibility that they interact with distinct activation complexes. EPCR selectively enhances PC activation so that PC and TAFI activation kinetics become comparable on endothelial cells.


Assuntos
Antígenos CD/metabolismo , Carboxipeptidase B2/metabolismo , Células Endoteliais/enzimologia , Proteína C/metabolismo , Receptores de Superfície Celular/metabolismo , Ligação Competitiva , Células Cultivadas , Receptor de Proteína C Endotelial , Ativação Enzimática , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Cinética , Ligação Proteica , Trombina/metabolismo , Trombomodulina/metabolismo
4.
J Thromb Haemost ; 13 Suppl 1: S82-91, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26149054

RESUMO

The existence of extracellular DNA in human plasma, also known as cell-free DNA (cfDNA), was first described in the 1940s. In recent years, there has been a resurgence of interest in the functional significance of cfDNA, particularly in the context of neutrophil extracellular traps (NETs). cfDNA and histones are key components of NETs that aid in the host response to infection and inflammation. However, cfDNA and histones may also exert harmful effects by triggering coagulation, inflammation, and cell death and by impairing fibrinolysis. In this article, we will review the pathologic nature of cfDNA and histones in macrovascular and microvascular thrombosis, including venous thromboembolism, cancer, sepsis, and trauma. We will also discuss the prognostic value of cfDNA and histones in these disease states. Understanding the molecular and cellular pathways regulated by cfDNA and histones may provide novel insights to prevent pathological thrombus formation and vascular occlusion.


Assuntos
Coagulação Sanguínea , DNA/sangue , Armadilhas Extracelulares/metabolismo , Histonas/sangue , Inflamação/sangue , Embolia Pulmonar/sangue , Trombose/sangue , Animais , DNA/imunologia , Armadilhas Extracelulares/genética , Armadilhas Extracelulares/imunologia , Histonas/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Neoplasias/sangue , Neoplasias/genética , Neoplasias/imunologia , Embolia Pulmonar/genética , Embolia Pulmonar/imunologia , Sepse/sangue , Sepse/genética , Sepse/imunologia , Transdução de Sinais , Trombose/genética , Trombose/imunologia , Tromboembolia Venosa/sangue , Tromboembolia Venosa/genética , Tromboembolia Venosa/imunologia , Ferimentos e Lesões/sangue , Ferimentos e Lesões/genética , Ferimentos e Lesões/imunologia
5.
J Thromb Haemost ; 9(12): 2424-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21972922

RESUMO

BACKGROUND: Serpins form a widely distributed protein superfamily, but no integral membrane serpins have been described. OBJECTIVES: To anchor three serpins -α(1) -proteinase inhibitor (α(1) PI) (M358R), antithrombin (AT), and heparin cofactor II (HCII) - in the plasma membranes of transfected mammalian cells and assess their ability to inhibit thrombin. METHODS: Serpin cDNAs were altered to include N-terminal, non-cleavable plasma membrane-targeting sequences from the human transferrin receptor (TR) (TR-serpin) or the human asialoglycoprotein receptor (AR) (AR-serpin), and used to transfect COS-1 or HEK 293 cells. Cells were analyzed for serpin expression by immunoblotting of subcellular fractions, by immunofluorescence microscopy, or by flow cytometry, with or without exposure to exogenous thrombin; AR-serpins and TR-serpins were also compared with their soluble recombinant counterparts. RESULTS: Both TR-α(1) PI (M358R) and AR-α(1) PI (M358R) were enriched in the integral membrane fraction of transfected COS-1 or HEK 293 cells, and formed inhibitory complexes with thrombin, although less rapidly than soluble α(1) PI (M358R). Thrombin inhibition was abrogated by an additional T345R mutation in AR-α(1) PI (M358R). Surface-displayed AR-AT also formed serpin-enzyme complexes with thrombin, but to a lesser extent than AR-α(1) PI (M358R); AR-HCII inhibitory function was not detected. Immunofluorescence detection and flow cytometric quantification of bound thrombin also supported the status of AR-α(1) PI (M358R) and AR-AT as thrombin inhibitors. CONCLUSIONS: Two of three thrombin-inhibitory serpins retained functionality when expressed as integral membrane proteins. Our findings could be applied to create and screen hypervariable serpin libraries expressed in mammalian cells, or to confer protease resistance on engineered cells in vivo.


Assuntos
Antitrombinas/farmacologia , Proteínas de Membrana/genética , Serpinas/genética , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Humanos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Proteínas Recombinantes/genética
6.
J Thromb Haemost ; 9(11): 2313-21, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21838758

RESUMO

BACKGROUND: Thrombosis is a common complication for breast cancer patients receiving chemotherapy. However, the mechanisms by which breast cancer chemotherapeutic agents increase this risk are largely uncharacterized. Nucleic acids released by injured cells may enhance coagulation via the activation of the contact pathway. OBJECTIVES: In this study, we examined the effects of breast cancer chemotherapy agents on the release of cell-free DNA (CFDNA) and its relationship to thrombin generation using in vitro and in vivo methods. METHODS: CFDNA release and thrombin-antithrombin (TAT) levels were measured in plasma of breast cancer patients and healthy mice receiving chemotherapy. Venous whole blood and cultured cells were exposed to chemotherapy and CFDNA release and levels of DNA-histone complexes were measured. The procoagulant activity of isolated CFDNA was measured with calibrated, automated thrombin generation. RESULTS: Breast cancer patients receiving chemotherapy had elevated levels of CFDNA 24 h post-chemotherapy, a time-point at which elevated thrombin-antithrombin levels have been previously reported. Treatment of healthy mice with doxorubicin, epirubicin and 5-fluorouracil increased CFDNA release, with a corresponding elevation in TAT complex formation. Venous whole blood and neutrophils incubated with chemotherapeutic agents had elevated CFDNA in plasma or cell supernatants. In addition, incubation of venous whole blood with chemotherapy decreased histone-DNA complex levels. CFDNA released from epirubicin-treated whole blood significantly elevated thrombin generation in a dose-dependent manner, and involved activation of the contact pathway. CONCLUSIONS: Release of CFDNA from chemotherapy-injured cells may represent a novel mechanism by which thrombosis is triggered in cancer patients.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , DNA/sangue , Trombina/biossíntese , Animais , Antineoplásicos/farmacologia , Antitrombina III , Neoplasias da Mama/sangue , DNA/efeitos dos fármacos , DNA/metabolismo , Doxorrubicina , Epirubicina , Feminino , Fluoruracila , Humanos , Camundongos , Peptídeo Hidrolases/sangue , Trombose/induzido quimicamente , Fatores de Tempo
7.
J Thromb Haemost ; 9(4): 767-75, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21320281

RESUMO

BACKGROUND: Thrombosis is a common complication in cancer patients receiving chemotherapy regimens that include cyclophosphamide. However, the mechanisms by which these agents increase this risk are largely uncharacterized. OBJECTIVES: To examine the effects of cyclophosphamide and its metabolite acrolein on procoagulant and anticoagulant pathways in both cell-based and animal-based models. METHODS: Thrombin and activated protein C (APC) generation were measured in defibrinated plasma exposed to acrolein-treated endothelial and smooth muscle cells. Tissue factor (TF) activity was measured on acrolein-treated cells. Cell surface levels of phosphatidylserine, TF, endothelial protein C receptor and thrombomodulin were measured. Healthy BALB/c mice received injections of saline (control), acrolein, or cyclophosphamide; blood was collected, and plasma thrombin-antithrombin (TAT) complex, protein C and APC levels were analyzed. RESULTS: Exposure of acrolein-treated endothelial and smooth muscle cells to defibrinated plasma increased thrombin generation in the plasma. This was associated with enhanced phosphatidylserine exposure and/or increased TF activity on acrolein-treated cells. Despite elevated levels of thrombin generation, plasma APC levels were not elevated. In vivo, treatment of mice with cyclophosphamide and acrolein resulted in elevations of plasma TAT complex levels, whereas APC levels remained low. CONCLUSIONS: This is the first study to examine thrombin generation and the APC pathway in chemotherapy-treated mice. Cyclophosphamide and acrolein appear to upregulate procoagulant pathways, while impairing endogenous anticoagulant pathways. This may explain, in part, the increased risk of thrombosis observed in cancer patients receiving cyclophosphamide-containing chemotherapy.


Assuntos
Acroleína/farmacologia , Antineoplásicos/farmacologia , Modelos Teóricos , Proteína C/antagonistas & inibidores , Trombina/biossíntese , Regulação para Cima/efeitos dos fármacos , Animais , Células Cultivadas , Ciclofosfamida/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C
8.
J Thromb Haemost ; 9(3): 582-92, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21166994

RESUMO

BACKGROUND: The endoplasmic reticulum (ER) is responsible for the synthesis and folding of secretory, transmembrane and ER-resident proteins. Conditions that impair protein folding or overwhelm its protein folding capacity disrupt ER homeostasis, thereby causing ER stress. ER stress-induced apoptosis and inflammation are involved in the pathogenesis of inflammatory diseases. Activated protein C (APC) inhibits inflammation and apoptosis in monocytes, and this may partly explain the protective effects of APC treatment in severe sepsis. However, the precise molecular pathways by which APC modulates these effects remain unknown. OBJECTIVES: To investigate whether APC modulates the ER stress response in human monocytes. METHODS: We treated monocytes with ER stress-inducing agents in the presence or absence of APC to determine the effect on this response. Protein and mRNA levels were determined by immunoblotting and real-time PCR, respectively. Enzyme assays and flow cytometry were used to determine the role of APC in this model. RESULTS: In thapsigargin (Tg)-treated cells, APC dampened unfolded protein response activation, as indicated by reduced levels of the 78-kDa glucose-regulated protein (GRP78), in an endothelial protein C receptor-independent and protease-activated receptor-1-independent manner. Consistent with this, APC decreased phosphorylated eukaryotic translational initiation factor 2α and C/EBP homologous protein levels induced by Tg. APC inhibited Tg-induced ER Ca(2+) flux and reactive oxygen species generation. Functionally, APC diminished Tg-induced caspase-3 activity and degradation of the nuclear factor kappaB inhibitor IκBα. Furthermore, APC dampened the induction of tissue factor procoagulant activity facilitated by Tg. CONCLUSIONS: These studies suggest that APC modulates the adverse effects of ER Ca(2+) depletion in human monocytes.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/sangue , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Proteína C/farmacologia , Tromboplastina/metabolismo , Anti-Inflamatórios/farmacologia , Sequência de Bases , Sinalização do Cálcio/efeitos dos fármacos , Caspase 3/sangue , Chaperona BiP do Retículo Endoplasmático , Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico/sangue , Proteínas de Choque Térmico/genética , Humanos , Técnicas In Vitro , Monócitos/citologia , NF-kappa B/sangue , RNA Mensageiro/sangue , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/sangue , Proteínas Recombinantes/farmacologia , Sepse/tratamento farmacológico , Sepse/metabolismo , Sepse/patologia , Estresse Fisiológico/efeitos dos fármacos , Tapsigargina/farmacologia
9.
J Thromb Haemost ; 7(4): 619-26, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19187077

RESUMO

BACKGROUND: Although chemotherapy is associated with an increased risk of thrombosis, the pathogenic mechanisms by which chemotherapeutic agents exert prothrombotic effects are unclear. OBJECTIVES: In this study we explored the possibility that chemotherapeutic agents doxorubicin, epirubicin, 5-fluorouracil and methotrexate induce a procoagulant phenotype on vascular endothelial cells and/or on blood monocytes. METHODS: Thrombin generation was measured in defibrinated plasma exposed to chemotherapy-treated human umbilical vein endothelial cells (HUVECs). Tissue factor activity assays were performed on chemotherapy-treated HUVECs and blood monocytes. The effects of chemotherapy drugs on phosphatidylserine exposure and the protein C pathway were also measured. RESULTS: Exposure of defibrinated plasma to either doxorubicin- or epirubicin-treated HUVECs resulted in an increase in plasma thrombin generation. The procoagulant activity of doxorubicin- and epirubicin-treated HUVECs reflects an increase in tissue factor activity and phosphatidylserine exposure. Doxorubicin-mediated increase in tissue factor activity is related to increased levels of phosphatidylserine rather than to protein disulfide isomerase activity, and is likely to involve reactive oxygen species generation. Unlike doxorubicin, epirubicin does not have an impact on the protein C anticoagulant pathway. Interestingly, neither methotrextate nor 5-fluorouracil altered endothelial or monocyte hemostatic properties. CONCLUSIONS: These studies suggest that doxorubicin and epirubicin have the greatest 'prothrombotic potential' by virtue of their ability to alter endothelial and monocyte hemostatic properties.


Assuntos
Doxorrubicina/farmacologia , Células Endoteliais/efeitos dos fármacos , Epirubicina/farmacologia , Monócitos/efeitos dos fármacos , Trombofilia/induzido quimicamente , Antibióticos Antineoplásicos/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Testes de Coagulação Sanguínea , Células Cultivadas , Endotélio Vascular , Humanos , Fenótipo , Plasma/efeitos dos fármacos
10.
J Thromb Haemost ; 1(4): 662-70, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12871399

RESUMO

Activated protein C (APC) serves as an 'on demand' anticoagulant. Defects in the APC anticoagulant pathway are underlying risk factors for the development of venous and arterial thrombosis. APC has recently been shown to significantly reduce mortality in patients with severe sepsis, presumably by virtue of its ability to down-regulate coagulation as well as inflammation. Our objective was to develop an assay that, for the first time, permits rapid detection of plasma APC. This assay will expedite studies of APC in a variety of vascular disease states including sepsis, severe atherosclerosis, diabetes, and vasculitis. By generating a highly APC-specific monoclonal antibody (HAPC 1555), we have developed an assay that, for the first time, allows rapid detection of plasma APC. The Kd measured for the interaction between APC and HAPC 1555 based on BIAcore studies and binding to immobilized HAPC on microtiter plates is 6.2 +/- 0.9 and 8.8 +/- 1.0 nmol L(-1), respectively. The interaction between HAPC 1555 and APC is Ca2+-dependent, with a Ca2+ concentration of 313 +/- 48 micro mol L(-1) required for half maximal binding. HAPC 1555 interferes with APC-mediated inactivation of factor (F)Va in the presence and absence of phospholipids, suggesting that HAPC 1555 binds to the FVa binding domain of APC. When HAPC 1555 was used in an APC enzyme capture assay, therapeutic APC levels could be measured in 1.5 h, and physiologic levels of APC could be detected between 3 and 19 h. APC levels were also shown to vary markedly in patients with severe sepsis. The rapidity of our APC assay makes APC detection in patients practical clinically. This assay will expedite studies of APC in a variety of vascular disease states including sepsis, severe atherosclerosis, diabetes, and vasculitis.


Assuntos
Anticorpos Monoclonais , Epitopos , Fator Va/metabolismo , Proteína C/análise , Afinidade de Anticorpos/efeitos dos fármacos , Especificidade de Anticorpos , Sítios de Ligação , Cálcio/farmacologia , Estudos de Casos e Controles , Humanos , Técnicas Imunoenzimáticas/normas , Proteína C/imunologia , Proteína C/metabolismo , Sepse/sangue , Fatores de Tempo
11.
Thromb Haemost ; 86(4): 945-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11686350

RESUMO

EPCR is a type I transmembrane protein, highly expressed on the endothelium of large vessels, that binds protein C and augments its activation. In this study, a 23bp insertion in the EPCR gene was found in 4/198 survivors of myocardial infarction and 3/194 patients with deep vein thrombosis. The EPCR gene with the insertion predicts a protein that lacks part of the extracellular domain, the transmembrane domain and the cytoplasmic tail. Expression studies showed that the truncated protein is not localized on the cell surface, cannot be secreted in the culture medium, and does not bind activated protein C. Since protein C activation depends on the concentration of EPCR, patients with the EPCR insertion could have a diminished protein C activation capacity. Further clinical studies of adequate samples size are necessary to establish whether or not the EPCR insertion predisposes to the development of thrombotic events.


Assuntos
Fatores de Coagulação Sanguínea , Endotélio Vascular/metabolismo , Infarto do Miocárdio/genética , Receptores de Superfície Celular/genética , Trombofilia/genética , Trombose Venosa/genética , Adulto , Idade de Início , Animais , Membrana Celular/metabolismo , Células Cultivadas , Análise Mutacional de DNA , Ativação Enzimática , Éxons/genética , Feminino , Frequência do Gene , Predisposição Genética para Doença , Glicosilação , Humanos , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Peso Molecular , Mutagênese Insercional , Infarto do Miocárdio/epidemiologia , Projetos Piloto , Ligação Proteica/genética , Proteína C/metabolismo , Conformação Proteica , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transporte Proteico/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/fisiologia , Fatores de Risco , Relação Estrutura-Atividade , Trombofilia/epidemiologia , Trombose Venosa/epidemiologia
12.
J Biol Chem ; 276(24): 20959-65, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11294849

RESUMO

Although fibrin-bound thrombin is resistant to inactivation by heparin.antithrombin and heparin.heparin cofactor II complexes, indirect studies in plasma systems suggest that the dermatan sulfate.heparin cofactor II complex can inhibit fibrin-bound thrombin. Herein we demonstrate that fibrin monomer produces a 240-fold decrease in the heparin-catalyzed rate of thrombin inhibition by heparin cofactor II but reduces the dermatan sulfate-catalyzed rate only 3-fold. The protection of fibrin-bound thrombin from inhibition by heparin.heparin cofactor II reflects heparin-mediated bridging of thrombin to fibrin that results in the formation of a ternary heparin.thrombin.fibrin complex. This complex, formed as a result of three binary interactions (thrombin.fibrin, thrombin.heparin, and heparin.fibrin), limits accessibility of heparin-catalyzed inhibitors to thrombin and induces conformational changes at the active site of the enzyme. In contrast, dermatan sulfate binds to thrombin but does not bind to fibrin. Although a ternary dermatan sulfate. thrombin.fibrin complex forms, without dermatan sulfate-mediated bridging of thrombin to fibrin, only two binary interactions exist (thrombin.fibrin and thrombin. dermatan sulfate). Consequently, thrombin remains susceptible to inactivation by heparin cofactor II. This study explains why fibrin-bound thrombin is susceptible to inactivation by heparin cofactor II in the presence of dermatan sulfate but not heparin.


Assuntos
Dermatan Sulfato/farmacologia , Fibrina/metabolismo , Cofator II da Heparina/metabolismo , Heparina/farmacologia , Trombina/química , Trombina/metabolismo , Sítios de Ligação , Dermatan Sulfato/isolamento & purificação , Fibrina/isolamento & purificação , Heparina/isolamento & purificação , Cofator II da Heparina/isolamento & purificação , Humanos , Cinética , Ligação Proteica , Espectrofotometria , Trombina/isolamento & purificação
13.
J Biol Chem ; 276(11): 8364-70, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11099506

RESUMO

The endothelial cell protein C receptor (EPCR) is an endothelial cell-specific transmembrane protein that binds both protein C and activated protein C (APC). EPCR regulates the protein C anticoagulant pathway by binding protein C and augmenting protein C activation by the thrombin-thrombomodulin complex. EPCR is homologous to the MHC class 1/CD1 family, members of which contain two alpha-helices that sit upon an 8-stranded beta-sheet platform. In this study, we identified 10 residues that, when mutated to alanine, result in the loss of protein C/APC binding (Arg-81, Leu-82, Val-83, Glu-86, Arg-87, Phe-146, Tyr-154, Thr-157, Arg-158, and Glu-160). Glutamine substitutions at the four N-linked carbohydrate attachment sites of EPCR have little affect on APC binding, suggesting that the carbohydrate moieties of EPCR are not critical for ligand recognition. We then mapped the epitopes for four anti-human EPCR monoclonal antibodies (mAbs), two of which block EPCR/Fl-APC (APC labeled at the active site with fluorescein) interactions, whereas two do not. These epitopes were localized by generating human-mouse EPCR chimeric proteins, since the mAbs under investigation do not recognize mouse EPCR. We found that 5 of the 10 candidate residues for protein C/APC binding (Arg-81, Leu-82, Val-83, Glu-86, Arg-87) colocalize with the epitope for one of the blocking mAbs. Three-dimensional molecular modeling of EPCR indicates that the 10 protein C/APC binding candidate residues are clustered at the distal end of the two alpha-helical segments. Protein C activation studies on 293 cells that coexpress EPCR variants and thrombomodulin demonstrate that protein C binding to EPCR is necessary for the EPCR-dependent enhancement in protein activation by the thrombin-thrombomodulin complex. These studies indicate that EPCR has exploited the MHC class 1 fold for an alternative and possibly novel mode of ligand recognition. These studies are also the first to identify the protein C/APC binding region of EPCR and may provide useful information about molecular defects in EPCR that could contribute to cardiovascular disease susceptibility.


Assuntos
Fatores de Coagulação Sanguínea , Antígenos de Histocompatibilidade Classe I/metabolismo , Proteína C/metabolismo , Receptores de Superfície Celular/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Sítios de Ligação , Bovinos , Glicosilação , Humanos , Camundongos , Dados de Sequência Molecular , Inibidor da Proteína C , Receptores de Superfície Celular/metabolismo
14.
J Biol Chem ; 275(8): 5447-52, 2000 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-10681521

RESUMO

The endothelial cell protein C receptor (EPCR) functions as an important regulator of the protein C anticoagulant pathway by binding protein C and enhancing activation by the thrombin-thrombomodulin complex. EPCR binds to both protein C and activated protein C (APC) with high affinity. A soluble form of EPCR (sEPCR) circulates in plasma and inhibits APC anticoagulant activity. In this study, we investigate the mechanisms by which sEPCR modulates APC function. Soluble EPCR inhibited the inactivation of factor Va by APC only in the presence of phospholipid vesicles. By using flow cytometric analysis in the presence of 3 mM CaCl(2) and 0. 6 mM MgCl(2), sEPCR inhibited the binding of protein C and APC to phospholipid vesicles (K(i) = 40 +/- 7 and 33 +/- 4 nM, respectively). Without MgCl(2), the K(i) values increased approximately 4-fold. Double label flow cytometric analysis using fluorescein-APC and Texas Red-sEPCR indicated that the APC.sEPCR complex does not interact with phospholipid vesicles. By using surface plasmon resonance, we found that sEPCR also inhibited binding of protein C to phospholipid in a dose-dependent fashion (K(i) = 32 nM). To explore the possibility that sEPCR evokes structural changes in APC, fluorescence spectroscopy studies were performed to monitor sEPCR/Fl-APC interactions. sEPCR binds saturably to Fl-APC (K(d) = 27 +/- 13 nM) with a maximum decrease in Fl-APC fluorescence of 10.8 +/- 0.6%. sEPCR also stimulated the amidolytic activity of APC toward synthetic substrates. We conclude that sEPCR binding to APC blocks phospholipid interaction and alters the active site of APC.


Assuntos
Fatores de Coagulação Sanguínea , Proteína C/metabolismo , Proteína S/metabolismo , Receptores de Superfície Celular/metabolismo , Sítios de Ligação , Cloreto de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Fator Va/metabolismo , Citometria de Fluxo , Humanos , Cinética , Lipossomos/metabolismo , Cloreto de Magnésio/farmacologia , Ligação Proteica , Conformação Proteica , Ressonância de Plasmônio de Superfície , Fatores de Tempo
15.
J Biol Chem ; 274(39): 27597-604, 1999 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-10488098

RESUMO

Heparin and dermatan sulfate activate heparin cofactor II (HCII) comparably, presumably by liberating the amino terminus of HCII to bind to exosite I of thrombin. To explore this model of activation, we systematically substituted basic residues in the glycosaminoglycan-binding domain of HCII with neutral amino acids and measured the rates of thrombin inactivation by the mutants. Mutant D, with changes at Arg(184), Lys(185), Arg(189), Arg(192), Arg(193), demonstrated a approximately 130-fold increased rate of thrombin inactivation that was unaffected by the presence of glycosaminoglycans. The increased rate reflects displacement of the amino terminus of mutant D because (a) mutant D inactivates gamma-thrombin at a 65-fold slower rate than alpha-thrombin, (b) hirudin-(54-65) decreases the rate of thrombin inactivation, and (c) deletion of the amino terminus of mutant D reduces the rate of thrombin inactivation approximately 100-fold. We also examined the contribution of glycosaminoglycan-mediated bridging of thrombin to HCII to the inhibitory process. Whereas activation of HCII by heparin was chain-length dependent, stimulation by dermatan sulfate was not, suggesting that dermatan sulfate does not utilize a template mechanism to accelerate the inhibitory process. Fluorescence spectroscopy revealed that dermatan sulfate evokes greater conformational changes in HCII than heparin, suggesting that dermatan sulfate stimulates HCII by producing more effective displacement of the amino terminus.


Assuntos
Dermatan Sulfato/farmacologia , Cofator II da Heparina/metabolismo , Heparitina Sulfato/farmacologia , Trombina/metabolismo , Sequência de Aminoácidos , Animais , Catálise , Linhagem Celular , Cricetinae , Variação Genética , Glicosaminoglicanos/farmacologia , Cofator II da Heparina/química , Cofator II da Heparina/genética , Humanos , Cinética , Dados de Sequência Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Trombina/antagonistas & inibidores , Transfecção , Células Tumorais Cultivadas
16.
Biochem J ; 332 ( Pt 1): 213-21, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9576870

RESUMO

The mechanism by which homocysteine causes endothelial cell (EC) injury and/or dysfunction is not fully understood. To examine the stress-inducing effects of homocysteine on ECs, mRNA differential display and cDNA microarrays were used to evaluate changes in gene expression in cultured human umbilical-vein endothelial cells (HUVEC) exposed to homocysteine. Here we show that homocysteine increases the expression of GRP78 and GADD153, stress-response genes induced by agents or conditions that adversely affect the function of the endoplasmic reticulum (ER). Induction of GRP78 was specific for homocysteine because other thiol-containing amino acids, heat shock or H2O2 did not appreciably increase GRP78 mRNA levels. Homocysteine failed to elicit an oxidative stress response in HUVEC because it had no effect on the expression of heat shock proteins (HSPs) including HSP70, nor did it activate heat shock transcription factor 1. Furthermore homocysteine blocked the H2O2-induced expression of HSP70. In support of our findings in vitro, steady-state mRNA levels of GRP78, but not HSP70, were elevated in the livers of cystathionine beta-synthase-deficient mice with hyperhomocysteinaemia. These studies indicate that the activation of stress response genes by homocysteine involves reductive stress leading to altered ER function and is in contrast with that of most other EC perturbants. The observation that homocysteine also decreases the expression of the antioxidant enzymes glutathione peroxidase and natural killer-enhancing factor B suggests that homocysteine could potentially enhance the cytotoxic effect of agents or conditions known to cause oxidative stress.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT , Endotélio Vascular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico , Homocisteína/farmacologia , Estresse Fisiológico/metabolismo , Animais , Proteínas de Transporte/genética , Células Cultivadas , Cistationina beta-Sintase/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico HSP70/metabolismo , Homocisteína/sangue , Humanos , Peróxido de Hidrogênio/farmacologia , Imuno-Histoquímica , Camundongos , Chaperonas Moleculares/genética , Músculo Liso/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Fator de Transcrição CHOP , Fatores de Transcrição/genética
17.
J Biol Chem ; 273(15): 8932-9, 1998 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-9535876

RESUMO

Co-crystallographic studies have shown that the interaction of human prothrombin fragment 2 (F2) with thrombin involves the formation of salt bridges between the kringle inner loop of F2 and anion-binding exosite II of thrombin. When F2 binds to thrombin, it has been shown to evoke conformational changes at the active site and at exosite I of the enzyme. Using plasma, recombinant, and synthetic F2 peptides (F2, rF2, and sF2, respectively) we have further localized the thrombin-binding domain on F2. F2, rF2-(1-116), rF2-(55-116), and sF2-(63-116), all of which contain the kringle inner loop (residues 64-93) and the acidic COOH-terminal connecting peptide (residues 94-116), bind to thrombin-agarose. In contrast, analogues of the kringle inner loop, sF2-(63-90), or the COOH-terminal connecting peptide, sF2-(92-116), do not bind. Thus, contrary to predictions from the crystal structure, the COOH-terminal connecting peptide as well as the kringle inner loop are involved in the interaction of F2 with thrombin. F2 and sF2-(63-116) bind saturably to fluorescently labeled active site-blocked thrombin with Kd values of 4.1 and 51.3 microM, respectively. The affinity of sF2-(63-116) for thrombin increases about 5-fold (Kd = 10 microM) when Val at position 78 is substituted with Glu. F2 and sF2-(63-116) bind to exosite II on thrombin because both reduce the heparin-catalyzed rate of thrombin inhibition by antithrombin approximately 4-fold. In contrast, only F2 slows the uncatalyzed rate of thrombin inactivation by antithrombin. Like F2, sF2-(63-116) induces allosteric changes in the active site and exosite I of thrombin because it alters the rates of thrombin-mediated hydrolysis of chromogenic substrates and displaces fluorescently labeled hirudin54-65 from active site-blocked thrombin, respectively. Both peptides also prolong the thrombin clotting time of fibrinogen in a concentration-dependent fashion, reflecting their effects on the active site and/or exosite I. These studies provide further insight into the regions of F2 that evoke functional changes in thrombin.


Assuntos
Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Protrombina/química , Protrombina/metabolismo , Trombina/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Cromatografia de Afinidade , Primers do DNA , Dissulfetos/análise , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/isolamento & purificação , Reação em Cadeia da Polimerase , Protrombina/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
19.
Yeast ; 10(6): 771-87, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7975895

RESUMO

We have used a random selection protocol to define the consensus and range of binding sites for the Saccharomyces cerevisiae REB1 protein. Thirty-five elements were sequenced which bound specifically to a GST-REB1p fusion protein coupled to glutathione-Sepharose under conditions in which more than 99.9% of the random sequences were not retained. Twenty-two of the elements contained the core sequence CGGGTRR, with all but one of the remaining elements containing only one deviation from the core. Of the core sequence, the only residues that were absolutely conserved were the three consecutive G residues. Statistical analysis of a nucleotide-use matrix suggested that the REB1p binding site also extends into flanking sequences with the optimal sequence for REB1p binding being GNGCCGGGGTAACNC. There was a positive correlation between the ability of the sites to bind in vitro and activate transcription in vivo; however, the presence of non-conformants suggests that the binding site may contribute more to transcriptional activation than simply allowing protein binding. Interestingly, one of the REB1p binding elements had a DNAse 1 footprint appreciably longer than other elements with similar affinity. Analysis of its sequence indicated the potential for a second REB1p binding site on the opposite strand. This suggests that two closely positioned low-affinity sites can function together as a highly active site. In addition, database searches with some of the randomly defined REB1p binding sites suggest that related elements are commonly found within 'TATA-less' promoters.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Fúngicas/metabolismo , Saccharomyces cerevisiae/genética , Sequência de Bases , Sítios de Ligação , DNA/metabolismo , Desoxirribonuclease I/farmacologia , Metilação , Dados de Sequência Molecular , Oligonucleotídeos/metabolismo , Regiões Promotoras Genéticas , Proteínas de Saccharomyces cerevisiae , Fatores de Transcrição , Ativação Transcricional
20.
J Biol Chem ; 267(29): 20943-52, 1992 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-1400410

RESUMO

In the gal-his3 hybrid promoter, his3-GG1, GCN4 stimulates transcription at the position normally occupied by a TATA element. This expression requires two elements within gal1-10 sequences, a REB1-binding site and a second element, Z, which resides 20 base pairs upstream of the GCN4-binding site. No obvious TATA element is present in this promoter. To characterize the function of Z, we replaced it with short random oligonucleotides and selected for expression in vivo. Fourteen elements were identified and classified into groups based upon sequence and phenotypic similarities. Group 1 elements contained functional TATA sequences that were essential for activity. TATA elements can thus function when positioned upstream of a GCN4-binding site. The Group 2 elements activated transcription poorly when used as conventional TATA elements; however, mutational analyses demonstrated that their activity required TATA-like sequences. These TATA-like sequences bound the yeast TATA-binding protein (TBP) poorly in vitro but function in vivo as TBP interaction sites based upon two criteria. First mutations that improved their TATA character correspondingly improved function and second their activity could be enhanced in the presence of an altered binding specificity mutant of TBP. Furthermore, the Group 2 elements enabled the identification of mutations outside of the TATA-like core that contribute to transcriptional activation without adversely affecting TBP binding. The finding that low affinity TBP-binding sites can be used at unconventional positions suggests that many "TATA-less" promoters contain a cryptic interaction site for TBP.


Assuntos
DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Regiões Promotoras Genéticas , Proteínas Quinases , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , TATA Box , Fatores de Transcrição/metabolismo , Transcrição Gênica , Alelos , Sequência de Bases , Sítios de Ligação , Clonagem Molecular , Sondas de DNA , DNA Fúngico/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Sondas de Oligonucleotídeos , Fenótipo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteína de Ligação a TATA-Box
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...