Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Thromb Haemost ; 16(9): 1817-1829, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29879294

RESUMO

Essentials Factor VIIa binds activated platelets to promote hemostasis in hemophilia patients with inhibitors. The interactions and sites responsible for platelet-FVIIa binding are not fully understood. Endothelial cell protein C receptor (EPCR) is expressed on activated human platelets. EPCR binding enhances the efficacy of a FVIIa variant and could impact design of new therapeutics. SUMMARY: Background High-dose factor VIIa (FVIIa) is routinely used as an effective bypassing agent to treat hemophilia patients with inhibitory antibodies that compromise factor replacement. However, the mechanism by which FVIIa binds activated platelets to promote hemostasis is not fully understood. FVIIa-DVQ is an analog of FVIIa with enhanced tissue factor (TF)-independent activity and hemostatic efficacy relative to FVIIa. Our previous studies have shown that FVIIa-DVQ exhibits greater platelet binding, thereby suggesting that features in addition to lipid composition contribute to platelet-FVIIa interactions. Objectives Endothelial cell protein C receptor (EPCR) also functions as a receptor for FVIIa on endothelial cells. We therefore hypothesized that an interaction with EPCR might play a role in platelet-FVIIa binding. Methods/results In the present study, we used flow cytometric analyses to show that platelet binding of both FVIIa and FVIIa-DVQ is partially inhibited in the presence of excess protein C or an anti-EPCR antibody. This decreased binding results in a corresponding decrease in the activity of both molecules in FXa and thrombin generation assays. Enhanced binding to EPCR was sufficient to account for the increased platelet binding of FVIIa-DVQ compared with wild-type FVIIa. As EPCR protein expression has not previously been shown in platelets, we confirmed the presence of EPCR in platelets using immunofluorescence, flow cytometry, immunoprecipitation, and mass spectrometry. Conclusions This work represents the first demonstration that human platelets express EPCR and suggests that modulation of EPCR binding could be utilized to enhance the hemostatic efficacy of rationally designed FVIIa analogs.


Assuntos
Plaquetas/metabolismo , Receptor de Proteína C Endotelial/sangue , Fator VIIa/metabolismo , Ativação Plaquetária , Adulto , Ligação Competitiva , Proteínas de Transporte/farmacologia , Venenos de Crotalídeos/farmacologia , Células Endoteliais/metabolismo , Receptor de Proteína C Endotelial/biossíntese , Fator VIIa/genética , Fator Xa/biossíntese , Hemostasia , Humanos , Lectinas Tipo C , Peptídeos/farmacologia , Ligação Proteica , Proteína C/metabolismo
2.
Blood Adv ; 2(11): 1300-1314, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29891592

RESUMO

Venous thromboembolism (VTE) is a leading cause of morbidity and mortality in elderly people. Increased expression of tumor suppressor protein 53 (p53) has been implicated in vascular senescence. Here, we examined the importance of endothelial p53 for venous thrombosis and whether endothelial senescence and p53 overexpression are involved in the exponential increase of VTE with age. Mice with conditional, endothelial-specific deletion of p53 (End.p53-KO) and their wild-type littermates (End.p53-WT) underwent subtotal inferior vena cava (IVC) ligation to induce venous thrombosis. IVC ligation in aged (12-month-old) End.p53-WT mice resulted in higher rates of thrombus formation and greater mean thrombus size vs adult (12-week-old) End.p53-WT mice, whereas aged End.p53-KO mice were protected from vein thrombosis. Analysis of primary endothelial cells from aged mice or human vein endothelial cells after induction of replicative senescence revealed significantly increased early growth response gene-1 (Egr1) and heparanase expression, and plasma factor Xa levels were elevated in aged End.p53-WT, but not in End.p53-KO mice. Increased endothelial Egr1 and heparanase expression also was observed after doxorubicin-induced p53 overexpression, whereas p53 inhibition using pifithrin-α reduced tissue factor (TF) expression. Importantly, inhibition of heparanase activity using TF pathway inhibitor-2 (TFPI2) peptides prevented the enhanced venous thrombus formation in aged mice and restored it to the thrombotic phenotype of adult mice. Our findings suggest that p53 accumulation and heparanase overexpression in senescent endothelial cells are critically involved in mediating the increased risk of venous thrombosis with age and that heparanase antagonization may be explored as strategy to ameliorate the prothrombotic endothelial phenotype with age.


Assuntos
Envelhecimento/metabolismo , Células Endoteliais/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Trombose Venosa/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/genética , Células Endoteliais/patologia , Fator Xa/biossíntese , Fator Xa/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Heparina Liase/genética , Camundongos , Camundongos Knockout , Proteína Supressora de Tumor p53/genética , Trombose Venosa/genética
3.
Arch Pharm Res ; 37(11): 1454-63, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24817443

RESUMO

Sulforaphane (SFN), a natural isothiocyanate that is present in cruciferous vegetables such as broccoli and cabbage, is effective in preventing carcinogenesis, diabetes and inflammatory responses. Here, the anticoagulant activities of SFN were examined by monitoring activated partial thromboplastin time (aPTT), prothrombin time, and the activities of thrombin (Factor IIa, FIIa) and activated factor X (FXa). And, the effects of SFN on expression of plasminogen activator inhibitor type 1 (PAI-1) and tissue-type plasminogen activator (t-PA) were evaluated in tumor necrosis factor-α activated human umbilical vein endothelial cells (HUVECs). Treatment with SFN resulted in prolonged aPTT and PT and inhibition of the activities of thrombin and FXa, as well as inhibited production of thrombin and FXa in HUVECs. In addition, SFN inhibited thrombin-catalyzed fibrin polymerization and platelet aggregation. SFN also elicited anticoagulant effects in mice. In addition, treatment with SFN resulted in significant reduction of the PAI-1 to t-PA ratio. Collectively, SFN possesses antithrombotic activities and offers a basis for development of a novel anticoagulant.


Assuntos
Inibidores do Fator Xa/farmacologia , Fator Xa/biossíntese , Fibrinolíticos/farmacologia , Isotiocianatos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Protrombina/antagonistas & inibidores , Adulto , Animais , Sobrevivência Celular/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Estrutura Molecular , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Sulfóxidos , Ativador de Plasminogênio Tecidual/biossíntese , Adulto Jovem
4.
Fitoterapia ; 91: 1-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23973654

RESUMO

Pellitorine (PLT), an active amide compound, is well known to possess insecticidal, antibacterial and anticancer properties. However, the anti-coagulant functions of PLT are not studied yet. Here, the anticoagulant activities of PLT were examined by monitoring activated partial thromboplastin time (aPTT), prothrombin time (PT), and the activities of cell-based thrombin and activated factor X (FXa). Furthermore, the effects of PLT on the expressions of plasminogen activator inhibitor type 1 (PAI-1) and tissue-type plasminogen activator (t-PA) were tested in tumor necrosis factor (TNF)-α activated human umbilical vein endothelial cells (HUVECs). Treatment with PLT resulted in prolonged aPTT and PT and inhibition of the activities of thrombin and FXa, and PLT inhibited production of thrombin and FXa in HUVECs. And PLT inhibited thrombin-catalyzed fibrin polymerization and platelet aggregation. In accordance with these anticoagulant activities, PLT elicited anticoagulant effects in mouse. In addition, treatment with PLT resulted in the inhibition of TNF-α-induced production of PAI-1 and in the significant reduction of the PAI-1 to t-PA ratio. Collectively, PLT possesses antithrombotic activities and offers bases for development of a novel anticoagulant.


Assuntos
Anticoagulantes/farmacologia , Asarum/química , Ácidos Graxos Insaturados/farmacologia , Extratos Vegetais/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Trombose/metabolismo , Animais , Fator Xa/biossíntese , Fibrina/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos ICR , Tempo de Tromboplastina Parcial , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Tempo de Protrombina , Trombina/biossíntese , Ativador de Plasminogênio Tecidual/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
5.
Haematologica ; 92(8): 1091-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17640852

RESUMO

BACKGROUND AND OBJECTIVES: Ionizing radiation (IR) is associated with thrombotic vascular occlusion predicting a poor clinical outcome. Our study examined whether IR induced tissue factor (TF) expression and procoagulability. We further investigated coordinated gene alterations associated with TF upregulation in the myelomonocytic leukemia THP-1 cells. DESIGN AND METHODS: TF expression was determined by quantitative Reverse Transcriptase (TaqMan) PCR, TF ELISA and TF activity by a two stage chromogenic assay in the time course of days 1, 3, 7, 10, and 17 post IR. To detect IR-induced alterations in gene expression, Affymetrix HG U133 Plus 2.0 microarrays were used. RESULTS IR induced a significant increase in TF/GAPDH mRNA ratios and cellular TF protein on days 3 and 7 post IR (20 Gy [p>or=0.01] and 40 Gy [p or=0.001] vs. control respectively), suggesting IR immediately alters the cellular thrombogenicity. TF upregulation post IR was confirmed in PBMNCs. Gene expression profiling showed IR increased the expression of inflammatory and apoptosis-related pathways known to be involved in the regulation of TF expression. INTERPRETATION AND CONCLUSIONS: TF upregulation together with inflammation and apoptosis may increase the thrombogenicity of tissues. The demonstrated upregulation of TF might play a pivotal role in radiation associated thrombosis.


Assuntos
Fatores de Coagulação Sanguínea/biossíntese , Regulação Leucêmica da Expressão Gênica/efeitos da radiação , Leucemia Mielomonocítica Aguda/patologia , Proteínas de Neoplasias/biossíntese , Trombofilia/etiologia , Tromboplastina/biossíntese , Apoptose/genética , Apoptose/efeitos da radiação , Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/efeitos da radiação , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/efeitos da radiação , Ensaio de Imunoadsorção Enzimática , Fator Xa/biossíntese , Perfilação da Expressão Gênica , Humanos , Inflamação , Leucemia Mielomonocítica Aguda/sangue , Leucemia Mielomonocítica Aguda/complicações , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Nitrilas/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Aceleradores de Partículas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Radiação Ionizante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonas/farmacologia , Tromboplastina/genética
6.
J Am Soc Nephrol ; 16(8): 2477-86, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15976001

RESUMO

Priming of the coagulation cascade during hemodialysis (HD) leads to the release of activated factor X (FXa). The binding of FXa to its specific receptors, effector protease receptor-1 (EPR-1) and protease-activated receptor-2 (PAR-2), may induce the activation of peripheral blood mononuclear cells (PBMC) and promote a chronic inflammatory state that is responsible for several HD-related morbidities. In the attempt to elucidate the mechanisms underlying the coagulation-associated inflammation in HD, 10 HD patients were randomized to be treated subsequently with a cellulose acetate membrane (CA) and Ethylen-vinyl-alcohol (EVAL), a synthetic membrane that has been shown to reduce FXa generation. At the end of each experimental period, surface FXa and thrombin receptors (EPR-1 and PAR-1, -2, and -4) and CCR2 (monocyte chemoattractant protein-1 receptor) gene expression in isolated PBMC were examined. the ability of dialytic membranes to activate protein-tyrosine kinases and the stress-activated kinase JNK and to modulate the generation of terminal complement complex (TCC) was also investigated. EPR-1 and PAR-2 and -4 mRNA expression, barely detectable in normal PBMC, were significantly upregulated in HD patients, particularly in those who were treated with CA. A striking increase of tyrosine-phosphorylated proteins and JNK activation was observed at the end of HD only in CA-treated patients. Simultaneously, an increased gene expression for both splicing isoforms of CCR2, A and B, only in PBMC from CA-treated patients was demonstrated. The increased CCR-2 mRNA abundance was followed by a significant increase in its protein synthesis. The high expression of CCR2 was associated with an increased generation of plasma TCC and a significant drop in leukocyte and monocyte count. By contrast, EVAL treatment slightly lowered TCC generation and normalized leukocyte count. In vitro FXa induced CCR2 A and B expression and JNK activation in freshly isolated PBMC. FXa-induced CCR2 mRNA expression was completely abolished by JNK and tyrosine kinase inhibition. In conclusion, these data suggest that subclinical clotting activation may cause an increased CCR2 gene and protein expression on uremic PBMC, contributing to HD-related chronic microinflammation. The use of the less coagulation-activating membrane, EVAL, may reduce PBMC activation through the modulation of the stress-activated kinase JNK.


Assuntos
Regulação da Expressão Gênica , Leucócitos Mononucleares/citologia , Receptores de Quimiocinas/biossíntese , Adulto , Idoso , Proteínas Reguladoras de Apoptose/biossíntese , Western Blotting , Membrana Celular/metabolismo , Celulose/análogos & derivados , Celulose/química , Complexo de Ataque à Membrana do Sistema Complemento/biossíntese , Proteínas de Ligação a DNA/biossíntese , Inibidores Enzimáticos/farmacologia , Fator Xa/biossíntese , Fator Xa/metabolismo , Feminino , Humanos , Inflamação , Leucócitos/citologia , Leucócitos Mononucleares/metabolismo , MAP Quinase Quinase 4/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/citologia , Fosforilação , Polivinil/química , Ligação Proteica , Isoformas de Proteínas , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-ets , RNA/metabolismo , RNA Mensageiro/metabolismo , Receptor PAR-1/biossíntese , Receptor PAR-2/biossíntese , Receptores CCR2 , Diálise Renal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fatores de Transcrição/biossíntese
7.
Thromb Res ; 115(1-2): 123-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15567463

RESUMO

A number of studies indicate that coagulation proteases play significant roles in cancer biology. Melanoma is a highly metastatic cancer, and there is evidence that thrombin contributes to this aggressive pattern. However, few studies correlate this type of cancer with formation of the prothrombinase complex, which is responsible for conversion of prothrombin into thrombin in the coagulation system. The aim of this study was to investigate the assembly and regulation of prothrombinase complex on the murine melanoma cell line, B16F10. B16F10 cells were unable to activate prothrombin except when previously incubated with factor Xa. This effect was dependent on factor Xa binding to cell membranes, since no activation was detected with Gla-domainless factor Xa. The thrombin formation by B16F10-bound factor Xa was enhanced approximately 10 fold in the presence of factor Va, indicating the assembly of prothrombinase complex. Differently from platelets, B16F10-assembled prothrombinase complex was inhibited by prothrombin fragment 1 but not by fragment 2. In addition, bothrojaracin, a specific ligand of proexosite I on prothrombin, caused a significant decrease in the zymogen activation. Our data demonstrate that B16F10 melanoma cells generate thrombin by promoting assembly of the prothrombinase complex. This ability might be correlated with the increased metastatic potential of this cell line. Moreover, B16F10-assembled prothrombinase complex seems to be modulated in a different way from that found for the physiological complex assembled on platelets.


Assuntos
Fator V/biossíntese , Fator Xa/biossíntese , Melanoma/enzimologia , Animais , Coagulação Sanguínea , Linhagem Celular Tumoral , Venenos de Crotalídeos/farmacologia , Fator V/metabolismo , Fator Xa/metabolismo , Humanos , Melanoma/patologia , Camundongos , Metástase Neoplásica , Fragmentos de Peptídeos/farmacologia , Protrombina/farmacologia , Trombina/biossíntese
8.
Thromb Haemost ; 92(5): 947-55, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15543320

RESUMO

Tissue factor (TF) is the blood coagulation initiator, whose cofactor function is required for physiological factor VIIa (FVIIa)-mediated activation of factor X (FX) to FXa. A previous study reported TF on herpes simplex virus type 1 (HSV1), but this explained only part of FVIIa-dependent FXa generation observed on the virus surface (Sutherland et al. (1997) Proc. Natl. Acad. Sci. USA. 94:13510-14). In the current study, we investigated the role of HSV1-encoded glycoprotein C (gC) in this process. Purified gC-deficient HSV1 facilitated several fold less FX activation by FVIIa than either wild type or gC-rescued strains. To confirm the implication of gC in FVIIa-dependent FX activation, purified soluble gC (sgC) enhanced FXa production in the absence of TF. sgC required FVIIa, calcium and anionic phospholipid to participate in FX activation, suggesting similarity to TF. When purified virus was combined with sgC, the sgC-dependent FXa generation was enhanced three orders of magnitude, suggesting synergy with an additional HSV1 component and explaining the relatively low activity of purified sgC compared to the viral counterpart. FX activation on gC-competent HSV1 was inhibited 20% by a gC-specific antibody, inhibited 40% by a TF-specific antibody, inhibited 65% by combining the gC- and TF-specific antibodies, and nearly completely inhibited by the TF antibody alone on gC-deficient HSV-1. Cumulatively, these observations show that two pathways initiating FX activation function in parallel on the virus surface. In addition to the previously described TF-dependent pathway, HSV-1-encoded gC also enhances FXa generation, and like TF, requires FVIIa.


Assuntos
Fator VIIa/efeitos dos fármacos , Herpesvirus Humano 1 , Proteínas do Envelope Viral/farmacologia , Fator VIIa/metabolismo , Fator X/metabolismo , Fator Xa/biossíntese , Fator Xa/efeitos dos fármacos , Herpesvirus Humano 1/isolamento & purificação , Herpesvirus Humano 1/fisiologia , Humanos , Trombina/biossíntese , Trombina/efeitos dos fármacos , Tromboplastina/fisiologia , Doenças Vasculares/virologia , Proteínas do Envelope Viral/isolamento & purificação
9.
Blood Coagul Fibrinolysis ; 15(6): 483-90, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15311157

RESUMO

Abnormal tissue factor (TF) expression on vascular endothelial cells may account for thrombotic events associated with cardiovascular disease. The transcription factor nuclear factor-kappa B (NF-kappa B) activation plays a key role in endothelial cell injury and TF expression. Disruption of NF-kappa B activation in endothelial cells may inhibit TF expression and be protective in thrombosis. The purpose of the study was to determine whether NF-kappa B transcription factor decoy (TFD) could block TF expression. NF-kappa B TFD was transferred into cultured human umbilical cord vein endothelial cells (HUVEC) by liposomes, and the transfection efficiency was detected by flow cytometry. The effect of NF-kappa B TFD on TF mRNA levels was determined by reverse transcription-polymerase chain reaction. The expression of surface TF antigen was analyzed by flow cytometry. TF activity was studied by measuring enzymatic activation of factor X by the TF-activated factor VII complex. The results suggested that NF-kappa B decoy could be successfully transferred into HUVEC by liposome. The NF-kappa B TFD competed with the endogenous kappa B site sequence in the TF promoter for binding to transcription factor NF-kappa B in tumor necrosis factor-alpha-stimulated HUVEC, which could block the tumor necrosis factor-alpha-induced increase in TF mRNA levels, the upregulation of surface TF antigen and TF activity. This study demonstrated that NF-kappa B decoy could block HUVEC TF gene expression. Targeted genetic disruption of endothelial TF expression by NF-kappa B decoy may provide a possible therapeutic method for cardiovascular and thrombosis disease.


Assuntos
Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Oligodesoxirribonucleotídeos/farmacologia , Tromboplastina/biossíntese , Sítios de Ligação , Ligação Competitiva , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fator Xa/biossíntese , Humanos , Lipossomos , Oligodesoxirribonucleotídeos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Sequências Reguladoras de Ácido Nucleico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tromboplastina/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Veias Umbilicais
10.
Circulation ; 110(4): 452-9, 2004 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15238452

RESUMO

BACKGROUND: Tissue factor (TF) is the main initiator of the arterial blood coagulation system, and aggregated LDL (agLDL) are found in the arterial intima. Our hypothesis is that agLDL internalization by vascular smooth muscle cells (VSMCs) may trigger TF-procoagulant activity. METHODS AND RESULTS: Cultured human VSMCs were obtained from human coronary arteries of explanted hearts during transplant operations. VSMCs were incubated with native LDL (nLDL) or agLDL. TF mRNA was analyzed by real-time polymerase chain reaction, and cellular and released TF protein antigen were analyzed by Western blot. TF microparticle (MP) content was analyzed by flow cytometry and TF activity by a factor Xa generation test. Both nLDL and agLDL strongly and equally increased TF mRNA and cell membrane protein expression, by approximately 5- and 9-fold, respectively. A sustained TF procoagulant activity was induced by agLDL but not by nLDL (agLDL 2.46+/-0.22 versus nLDL 0.72+/-0.12 mU/mg protein at 12 hours). AgLDL increased TF antigen release (agLDL 5.64+/-0.4 versus nLDL 3.28+/-0.22 AU) and TF MP release (agLDL 89.85+/-8.51 versus nLDL 19.69+/-4.59 TF MP/10(3) cells). TF activation and release induced by agLDL is not related to apoptosis. Blockade of LDL receptor-related protein, a receptor for agLDL, prevented the agLDL-induced release of TF protein and TF MP. CONCLUSIONS: VSMC-TF expression is upregulated by both nLDL and agLDL. However, only agLDL engagement to LDL receptor-related protein induced cellular TF procoagulant activity and TF release by human VSMCs.


Assuntos
Lipoproteínas LDL/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Tromboplastina/metabolismo , Anexina A5/análise , Células Cultivadas/citologia , Células Cultivadas/metabolismo , Vasos Coronários/citologia , Fator Xa/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Genes bcl-2 , Humanos , Lipoproteínas LDL/farmacologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Lipídeos de Membrana/metabolismo , Músculo Liso Vascular/citologia , Fosfolipídeos/metabolismo , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteína X Associada a bcl-2
11.
J Thromb Haemost ; 2(1): 93-101, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14717972

RESUMO

Tissue factor (TF) is a transmembrane glycoprotein that initiates blood coagulation when complexed with factor (F)VIIa. Recently, TF has been shown to promote cellular signaling, tumor growth, angiogenesis, and metastasis. In the present study, we examined the pathway by which TF-FVIIa complex induces cellular signaling in human breast cancer cells using the Adr-MCF-7 cell line. This cell line has high endogenous TF expression as measured by flow cytometry and expression of protease-activated receptors 1 and 2 (PAR1 and PAR2) as determined by reverse transcriptase-polymerase chain reaction analysis. Both PAR1 and PAR2 are functionally active as determined by induction of p44/42 mitogen-activated protein kinase (MAPK) phosphorylation using specific agonist peptides. We found that MAPK phosphorylation in this cell line was strongly induced by the combination of FVIIa and factor (F)X, but not by FVIIa alone at a concentration of FVIIa that approaches physiological levels. Induction of MAPK phosphorylation involved the formation of TF-FVIIa-FXa complex and occurred by a pathway that did not require thrombin formation, indicating a critical role for FXa generation. In addition, induction of MAPK phosphorylation was found to be independent of PAR1 activation. We then examined whether TF-FVIIa complex formation could promote tumor cell migration using a modified Boyden chamber chemotaxis assay. The combination of FVIIa and FX, but not FVIIa alone, strongly induced migration of tumor cells by a pathway that probably involves PAR2, but not PAR1 activation. MAPK phosphorylation was found to be required for the induction of cell migration by the combination of FVIIa and FX. These data suggest that TF-FVIIa-mediated signaling in human breast cancer cells occurs most efficiently by formation of the TF-FVIIa-FXa complex. One of the physiological consequences of this signaling pathway is enhanced cell migration that is probably mediated by PAR2, but not PAR1 activation.


Assuntos
Neoplasias da Mama/fisiopatologia , Fator VIIa/fisiologia , Fator Xa/fisiologia , Tromboplastina/fisiologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Fator VIIa/biossíntese , Fator Xa/biossíntese , Feminino , Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Substâncias Macromoleculares , Receptor PAR-1/genética , Receptor PAR-1/fisiologia , Receptor PAR-2/genética , Receptor PAR-2/fisiologia , Transdução de Sinais , Tromboplastina/biossíntese , Tromboplastina/genética
12.
Am J Hematol ; 75(1): 34-9, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14695630

RESUMO

We investigated the rare case of a patient with IgGlambda multiple myeloma for whom both prothrombin time and APTT were significantly prolonged. The IgG inhibited coagulation reactions upstream from prothrombin when coagulation was initiated by mRVVT, but not by FXa, as indicated by a chromogenic substrate for FXa. The mPT and the mAPTT showed inhibition of FXa generation in both the intrinsic and extrinsic pathways. The IgG inhibited both protein C (indicated by APTT) and FX (indicated by RVV) but not amidolysis for either activated protein C or FXa. The addition of excess phospholipid significantly shortened the prolonged RVVT of the patient. It inhibited the coagulation reactions of normal plasma and was dependent on decreasing the PS concentration in the APTT reagent. It was suggested that the IgG showed lupus anticoagulant (LA)-like activity that inhibited phospholipid-dependent coagulation reactions in the intrinsic, extrinsic, and common pathways. However, the IgG did not bind cardiolipin-beta2GPI complex, beta2GPI, or prothrombin in ELISA assays. The IgG did not bind to either PS or phospholipid complexes in the presence or absence of prothrombin, FX, or FXa. Interestingly, the IgG lost its LA like-activity when it was degraded to F(ab')2 and Fc fragments by pepsin. We suspected that the IgG might inhibit the interaction between coagulation factors and acid phospholipid non-immunologically and that this process requires an intact IgG conformation, although the reaction mode is still not clear.


Assuntos
Imunoglobulina G/metabolismo , Inibidor de Coagulação do Lúpus/metabolismo , Mieloma Múltiplo/metabolismo , Fosfolipídeos/metabolismo , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Fator Xa/biossíntese , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/biossíntese , Fragmentos Fc das Imunoglobulinas/biossíntese , Imunoglobulina G/fisiologia , Pessoa de Meia-Idade , Pepsina A/farmacologia , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Receptores Imunológicos/biossíntese
13.
Blood Coagul Fibrinolysis ; 14(6): 539-44, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12960606

RESUMO

The cellular initiation of coagulation by the tissue factor (TF)-activated factor VII complex is transiently inhibited by endogenous tissue factor pathway inhibitor-1 (TFPI-1), whereas exogenously added TFPI-1 is targeted to a degradation pathway. This study investigates the relevance of glycosyl phosphatidylinositol (GPI) anchoring for the anticoagulant properties of TFPI-1. Experiments were performed with the human cell line ECV304 using liposomal gene transfer. For GPI anchoring of TFPI-1 we used a fusion protein of TFPI-1 and the GPI attachment sequence of decay-accelerating factor (GPI-TFPI-1), and compared it with wild-type TFPI-1. We measured TF and TFPI-1 surface expression by flow cytometry and TF proteolytic activity by a chromogenic assay for activated factor X generation. After transfection of GPI-TFPI-1, surface expression of TFPI-1 increased to 134 +/- 9% of mock transfected cells (mean +/- SEM, P = 0.004), and transfection with wild-type TFPI-1 did not significantly alter TFPI-1 surface expression. After transfection with GPI-TFPI-1, TF activity was reduced by 18 +/- 9% compared with mock transfections (P = 0.003), whereas after transfection with TFPI-1 wild type no significant inhibition was observed. This effect was not due to altered TF expression. GPI anchoring is an essential prerequisite for surface expression of TFPI-1 and inhibition of TF activity. Gene transfer of GPI-anchored TFPI, therefore, may be an efficient tool to inhibit local TF-induced coagulation.


Assuntos
Glicosilfosfatidilinositóis/farmacologia , Lipoproteínas/farmacologia , Tromboplastina/antagonistas & inibidores , Anticoagulantes/administração & dosagem , Linhagem Celular Tumoral , Compostos Cromogênicos , Fator Xa/biossíntese , Terapia Genética/métodos , Glicosilfosfatidilinositóis/genética , Humanos , Lipoproteínas/genética , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Tromboplastina/metabolismo , Transfecção/métodos
14.
Arterioscler Thromb Vasc Biol ; 23(5): 905-10, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12649087

RESUMO

OBJECTIVE: This study was performed to evaluate the mechanisms leading to tumor vessel occlusion by tissue factor-based drugs, which are used in vascular targeting approaches for the treatment of malignant tumors. METHODS AND RESULTS: The effects of nontargeted soluble tissue factor were evaluated in vitro and in vivo. Tumor-bearing mice were treated with (1) the extracellular portion of tissue factor (soluble tissue factor), (2) low nontoxic doses of lipopolysaccharides, or (3) a combination thereof. The combination treatment showed the best effects and resulted in selective thrombosis of tumor vessels. On the basis of our data from subsequent in vitro analyses, including surface plasmon resonance measurements and endothelial cell based coagulation assays, we propose a model on how soluble tissue factor, although lacking its membrane anchor, can promote selective tumor vessel occlusion. CONCLUSIONS: To our knowledge, this is the first report to describe the molecular mechanisms of coagulation induction by untargeted soluble tissue factor in vivo. Combination treatments including soluble tissue factor might represent an alternative vascular targeting approach for the treatment of malignant tumors.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Embolização Terapêutica/métodos , Lipopolissacarídeos/uso terapêutico , Linfoma não Hodgkin/terapia , Teratocarcinoma/terapia , Tromboplastina/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Ativação Enzimática/efeitos dos fármacos , Fator Xa/biossíntese , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Linfoma não Hodgkin/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Solubilidade , Ressonância de Plasmônio de Superfície , Teratocarcinoma/irrigação sanguínea , Tromboplastina/administração & dosagem , Tromboplastina/genética , Tromboplastina/farmacologia , Fator de Necrose Tumoral alfa/análise , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Thromb Haemost ; 88(6): 1012-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12529753

RESUMO

Aim of our study was to investigate effects of eptifibatide and anticoagulants on platelet aggregation and thrombin generation under low and high coagulant challenge in tissue factor-activated platelet rich plasma using a model allowing simultaneous determination of the time course of platelet aggregation and thrombin generation. Eptifibatide exerted a dose-dependent anti-aggregating effect under both high and significantly stronger under low coagulant challenge. Combination of eptifibatide and anticoagulants resulted in significant additive prolongation of the lag phase until the onset of platelet aggregation, more pronounced under low coagulant challenge. Under high, but not under low coagulant challenge combination of eptifibatide and anticoagulants had a significant synergistic inhibitory effect on platelet aggregation. Under low coagulant challenge combination of eptifibatide with LMWH, but not with UH, or rH, resulted in significantly reduced thrombin potential, F 1+2 generation, and FXa formation compared to measurements in the absence of eptifibatide. We demonstrate a synergistic effect of eptifibatide and anticoagulants on platelet aggregation inhibition and an additional inhibitory effect of LMWH and eptifibatide on thrombin generation. Our results support the notion that combination of eptifibatide and anticoagulants might be beneficial in atherosclerotic disease to palliate the thrombogenic potency of ruptured atherosclerotic plaques.


Assuntos
Anticoagulantes/administração & dosagem , Peptídeos/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Trombina/biossíntese , Arteriosclerose/sangue , Arteriosclerose/tratamento farmacológico , Sinergismo Farmacológico , Eptifibatida , Fator Xa/biossíntese , Heparina/administração & dosagem , Heparina de Baixo Peso Molecular/administração & dosagem , Hirudinas/administração & dosagem , Humanos , Técnicas In Vitro , Inibidores da Agregação Plaquetária/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Trombose/prevenção & controle
16.
Thromb Haemost ; 85(1): 93-100, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11204595

RESUMO

Recombinant factor VIII SQ (r-VIII SQ), ReFacto, is a recombinant factor VIII product similar to the smallest active factor VIII protein found in plasma-derived factor VIII (p-VIII) concentrates. The protein comprises two polypeptide chains of 80 and 90 kDa and lacks the major part of the heavily glycosylated B-domain i.e. amino acids Gln744 to Ser1637. r-VIII SQ retains six potential glycosylation sites for N-linked oligosaccharides at asparagine residues 41, 239, 582, 1685, 1810 and 2118. We describe a thorough comparison of the characteristics of r-VIII SQ with those of p-VIII. The primary and secondary structures of r-VIII SQ were in good agreement with that of B-domain-deleted p-VIII (p-VIII-LMW) as shown by SDS-PAGE, Western blotting with antifactor VIII antibodies, tryptic mapping, amino acid sequence analysis and circular dichroism spectroscopy. A few divergences also existed. Thus r-VIII SQ was shown to contain a small amount of the single chain primary translation product of 170 kDa and also the product specific sequence of 14 amino acids, the SQ-link, in the C-terminal end of the 90 kDa chain. It was shown that r-VIII SQ had a high specific activity of about 14,000 IU VIII:C/mg as determined by use of a chromogenic substrate assay. The r-VIII SQ protein was comparable to p-VIII forms with a retained B-domain, in terms of potency measured by a chromogenic substrate or a two-stage clotting assay, in interactions with thrombin, and with activated protein C (APC) in combination with Protein S. The ability of r-VIII SQ to participate as a cofactor in factor Xa generation in a mixture of factors IXa and X, phospholipid and calcium was in conformity with that of p-VIII. Furthermore r-VIII SQ had a good binding capacity for phospholipid vesicles and von Willebrand factor (vWF) as shown in gel filtration studies. The same kinetics in binding to von Willebrand factor was found for r-VIII SQ and p-VIII as determined by real-time biospecific interaction analysis (BIA) with use of the BIAcore instrument. The apparent association rate constant was 4 x 10(6) M(-1)s(-1). Two dissociation rate constants were found, 1 X 10(-2)s(-1) and 4 x 10(-4)s(-1). The results extend the present knowledge that the factor VIII B-domain is dispensable for the factor VIII cofactor function in hemostasis.


Assuntos
Fator VIII/química , Proteínas Recombinantes/química , Sítios de Ligação , Testes de Coagulação Sanguínea , Western Blotting , Dicroísmo Circular , Fator VIII/metabolismo , Fator VIII/farmacologia , Fator Xa/biossíntese , Fator Xa/efeitos dos fármacos , Humanos , Cinética , Fragmentos de Peptídeos/química , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Análise de Sequência de Proteína , Ressonância de Plasmônio de Superfície , Fator de von Willebrand/metabolismo
17.
Thromb Haemost ; 84(2): 319-24, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10959707

RESUMO

Influenza virus epidemics are associated with excess mortality due to cardiovascular diseases. There are several case reports of excessive coagulation during generalised influenza virus infection. In this study, we demonstrate the ability of respiratory viruses (influenza A, influenza B, parainfluenza-1, respiratory syncytial virus, adenovirus, cytomegalovirus) to infect lung fibroblasts and human umbilical vein endothelial cells in culture. All viral pathogens induced procoagulant activity in infected endothelial cells, as determined in a one-stage clotting assay, by causing an average 55% reduction in the clotting time. When factor VII deficient plasma was used clotting time was not reduced. The induction of procoagulant activity was associated with a 4- to 5-fold increase in the expression of tissue factor, as measured by the generation of factor Xa. Both experiments indicate that the procoagulant activity of endothelial cells in response to infection with respiratory viruses is caused by upregulation of the extrinsic pathway. Although both enveloped viruses and a non-enveloped virus (adenovirus) induced procoagulant activity in endothelial cells by stimulating tissue factor expression, the role of the viral envelope in the assembly of the prothrombinase complex remains uncertain. We conclude that both enveloped and non-enveloped respiratory viruses are capable of infecting cultured human endothelial cells and causing a shift from anticoagulant to procoagulant activity associated with the induction of tissue factor expression.


Assuntos
Endotélio Vascular/virologia , Infecções Respiratórias/sangue , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Fator Xa/biossíntese , Fibroblastos/metabolismo , Fibroblastos/virologia , Hemaglutininas Virais/metabolismo , Humanos , Pulmão/citologia , Pulmão/patologia , Pulmão/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Tromboplastina/biossíntese , Fatores de Tempo , Células Tumorais Cultivadas/virologia , Veias Umbilicais/patologia , Veias Umbilicais/fisiopatologia , Veias Umbilicais/virologia
18.
Thromb Res ; 96(1): 37-49, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10554083

RESUMO

The effects of a fucoidan (C-II), which was purified from the brown seaweed Ecklonia kurome, on the generation of thrombin and factor Xa have been investigated by measuring the amidolytic activities by using the respective specific chromogenic substrates in both plasma and purified systems. C-II inhibited significantly the generation of thrombin in both the intrinsic and the extrinsic pathways, although the intrinsic inhibitory effect by C-II was more remarkable than the extrinsic one. On the other hand, C-II was a good inhibitor of the factor Xa generation in the intrinsic pathway, while it was a poor one in the extrinsic pathway. In the purified systems C-II also inhibited the formation of prothrombin-activating complex (i.e., prothrombinase), but not its activity. The concentration of C-II required for 50% inhibition of thrombin generation was about one-tenth to one-seventh of that of the activity of the generated thrombin in plasma. These results indicate that C-II has an inhibitory effect on the generation of thrombin by blocking the formation of prothrombinase and by preventing the generation of intrinsic factor Xa in addition to its antithrombin activity, and also that the generation-inhibitory effect is more remarkable than C-II's enhancement effect on the antithrombin activity by heparin cofactor II in plasma.


Assuntos
Inibidores do Fator Xa , Fator Xa/biossíntese , Polissacarídeos/farmacologia , Trombina/antagonistas & inibidores , Trombina/biossíntese , Animais , Anticoagulantes/farmacologia , Dermatan Sulfato/farmacologia , Endopeptidases/farmacologia , Fator IXa/farmacologia , Fator X/metabolismo , Fator Xa/efeitos dos fármacos , Fucose/farmacologia , Cofator II da Heparina/farmacologia , Humanos , Poliéster Sulfúrico de Pentosana/farmacologia , Phaeophyceae/química , Protrombina/metabolismo , Daboia , Alga Marinha , Ésteres do Ácido Sulfúrico/farmacologia , Trombina/efeitos dos fármacos , Tromboplastina/antagonistas & inibidores , Tromboplastina/biossíntese , Venenos de Víboras/farmacologia
19.
Arterioscler Thromb Vasc Biol ; 19(3): 531-7, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10073954

RESUMO

This study was undertaken to characterize tissue factor (TF) induction, localization, and functional activity in cultured human umbilical vein endothelial cells (HUVECs) exposed to recombinant vascular endothelial growth factor (rVEGF) and recombinant tumor necrosis factor-alpha (rTNF-alpha). rVEGF (1 nmol/L) and rTNF-alpha (500 U/mL) synergistically increased TF mRNA, protein, and total activity, as measured in cell lysates. To examine surface TF expression, living cells were treated with antibody to TF and examined microscopically. Almost no staining was seen in control cells or cells treated with a single agent. In contrast, cells treated with both agonists showed intense membrane staining with surface patches, appearing as buds by confocal microscopy. To determine surface TF activity, studies were performed using a parallel-plate flow chamber, which allows detection of factor Xa generation on living cells. rVEGF and rTNF-alpha induced little surface TF activity (0.032+/-0.008 and 0.014+/-0.008 fmol/cm2, respectively). In combination, they significantly increased TF expression on the cell surface (0.429+/-0.094 fmol/cm2, P<0.05). These data indicate that the synergistic effect of rVEGF and rTNF-alpha is necessary to generate functional TF on the surface of endothelial cells. The requirement for multiple agonists to expose active TF may serve to protect endothelial cells from acting as a procoagulant surface, even under conditions of cell perturbation.


Assuntos
Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/metabolismo , Linfocinas/farmacologia , Tromboplastina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Antígenos de Superfície/análise , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/química , Endotélio Vascular/citologia , Fator Xa/biossíntese , Expressão Gênica/efeitos dos fármacos , Humanos , Linfocinas/metabolismo , RNA Mensageiro/análise , Proteínas Recombinantes/farmacologia , Tromboplastina/análise , Tromboplastina/genética , Fator de Necrose Tumoral alfa/metabolismo , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Rocz Akad Med Bialymst ; 42 Suppl 1: 110-7, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9337529

RESUMO

A review of literature concerning cancer procoagulant (CP) has been carried out. This procoagulant directly activates coagulation factor X to factor Xa. Possibilities of utilising determinations of this activator in diagnostics and prognostics of the cancerous disease are discussed.


Assuntos
Cisteína Endopeptidases/fisiologia , Proteínas de Neoplasias , Neoplasias/enzimologia , Animais , Biomarcadores Tumorais/sangue , Transtornos da Coagulação Sanguínea/etiologia , Cricetinae , Cisteína Endopeptidases/análise , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Fator X/metabolismo , Fator Xa/biossíntese , Humanos , Camundongos , Neoplasias/sangue , Neoplasias/complicações , Neoplasias Experimentais/sangue , Neoplasias Experimentais/complicações , Neoplasias Experimentais/enzimologia , Coelhos , Ratos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA