Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Haematologica ; 105(10): 2484-2495, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33054088

RESUMO

Tissue factor is highly expressed in sub-endothelial tissue. The extracellular allosteric disulfide bond Cys186-Cys209 of human tissue factor shows high evolutionary conservation and in vitro evidence suggests that it significantly contributes to tissue factor procoagulant activity. To investigate the role of this allosteric disulfide bond in vivo, we generated a C213G mutant tissue factor mouse by replacing Cys213 of the corresponding disulfide Cys190-Cys213 in murine tissue factor. A bleeding phenotype was prominent in homozygous C213G tissue factor mice. Pre-natal lethality of 1/3rd of homozygous offspring was observed between E9.5 and E14.5 associated with placental hemorrhages. After birth, homozygous mice suffered from bleedings in different organs and reduced survival. Homozygous C213G tissue factor male mice showed higher incidence of lung bleedings and lower survival rates than females. In both sexes, C213G mutation evoked a reduced protein expression (about 10-fold) and severely reduced pro-coagulant activity (about 1000-fold). Protein glycosylation was impaired and cell membrane exposure decreased in macrophages in vivo. Single housing of homozygous C213G tissue factor males reduced the occurrence of severe bleeding and significantly improved survival, suggesting that inter-male aggressiveness might significantly account for the sex differences. These experiments show that the tissue factor allosteric disulfide bond is of crucial importance for normal in vivo expression, post-translational processing and activity of murine tissue factor. Although C213G tissue factor mice do not display the severe embryonic lethality of tissue factor knock-out mice, their postnatal bleeding phenotype emphasizes the importance of fully functional tissue factor for hemostasis.


Assuntos
Dissulfetos , Tromboplastina , Animais , Feminino , Hemorragia/genética , Masculino , Camundongos , Mutação , Fenótipo , Gravidez , Tromboplastina/genética
2.
J Thromb Haemost ; 17(4): 627-634, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30740873

RESUMO

Essentials Prothrombotic extracellular vesicles (EV) carry agonist pathway-specific proteomes Agonists for protease activated receptor (PAR) 2 signaling have distinct effects on EV composition PAR2 signaling rapidly generates prothrombotic EV and slowly EV with inactive tissue factor (TF) FVIIa integrin ligation restricts TF incorporation into EV from endothelial cells SUMMARY: Background Cell injury signal-induced activation and release of tissue factor (TF) on extracellular vesicles (EVs) from immune and vessel wall cells propagate local and systemic coagulation initiation. TF trafficking and release on EVs occurs in concert with the release of cell adhesion receptors, including integrin ß1 heterodimers, which control trafficking of the TF-activated factor VII (FVIIa) complex. Activation of the TF signaling partner, protease-activated receptor (PAR) 2, also triggers TF release on integrin ß1+ EVs from endothelial cells, but the physiological signals for PAR2-dependent EV generation at the vascular interface remain unknown. Objective To define relevant protease ligands of TF contributing to PAR2-dependent release on EVs from endothelial cells. Methods In endothelial cells with balanced expression of TF and PAR2, we evaluated TF release on EVs by using a combination of activity and antigen assays, immunocapture, and confocal imaging. Results and Conclusions PAR2 stimulation generated time-dependent release of distinct TF+ EVs with high coagulant activity (early) and high antigen levels (late). Whereas PAR2 agonist peptide and a stabilized TF-FVIIa-activated FX complex triggered TF+ EV release, stimulation with FVIIa alone promoted cellular retention of TF, despite comparable PAR2 activation. On endothelial cells, FVIIa uniquely induced formation of a complex of TF with integrin α5 ß1 . Internalization of TF by FVIIa or anti-TF and activating antibodies against integrin ß1 prevented PAR2 agonist-induced release of TF on EVs. These data demonstrate that intracellular trafficking controlled by FVIIa forcing interaction with integrin ß1 regulates TF availability for release on procoagulant EVs.


Assuntos
Células Endoteliais/efeitos dos fármacos , Vesículas Extracelulares/efeitos dos fármacos , Fator VIIa/farmacologia , Integrina beta1/metabolismo , Oligopeptídeos/farmacologia , Receptor PAR-2/agonistas , Tromboplastina/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Transporte Proteico , Receptor PAR-2/metabolismo , Transdução de Sinais , Fatores de Tempo
3.
Blood ; 131(6): 674-685, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29246902

RESUMO

The tissue factor (TF) pathway serves both hemostasis and cell signaling, but how cells control these divergent functions of TF remains incompletely understood. TF is the receptor and scaffold of coagulation proteases cleaving protease-activated receptor 2 (PAR2) that plays pivotal roles in angiogenesis and tumor development. Here we demonstrate that coagulation factor VIIa (FVIIa) elicits TF cytoplasmic domain-dependent proangiogenic cell signaling independent of the alternative PAR2 activator matriptase. We identify a Lys-Gly-Glu (KGE) integrin-binding motif in the FVIIa protease domain that is required for association of the TF-FVIIa complex with the active conformer of integrin ß1. A point mutation in this motif markedly reduces TF-FVIIa association with integrins, attenuates integrin translocation into early endosomes, and reduces delayed mitogen-activated protein kinase phosphorylation required for the induction of proangiogenic cytokines. Pharmacologic or genetic blockade of the small GTPase ADP-ribosylation factor 6 (arf6) that regulates integrin trafficking increases availability of TF-FVIIa with procoagulant activity on the cell surface, while inhibiting TF-FVIIa signaling that leads to proangiogenic cytokine expression and tumor cell migration. These experiments delineate the structural basis for the crosstalk of the TF-FVIIa complex with integrin trafficking and suggest a crucial role for endosomal PAR2 signaling in pathways of tissue repair and tumor biology.


Assuntos
Fator VIIa/química , Fator VIIa/metabolismo , Integrina beta1/metabolismo , Domínios e Motivos de Interação entre Proteínas , Receptor PAR-2/metabolismo , Fator 6 de Ribosilação do ADP , Animais , Sítios de Ligação/genética , Células Cultivadas , Fator VIIa/genética , Humanos , Integrina beta1/química , Camundongos , Células NIH 3T3 , Neovascularização Fisiológica/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Mapas de Interação de Proteínas , Receptor PAR-2/genética , Transdução de Sinais/genética , Tromboplastina/química , Tromboplastina/metabolismo
4.
Blood ; 130(14): 1661-1670, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28729433

RESUMO

Safe and effective antithrombotic therapy requires understanding of mechanisms that contribute to pathological thrombosis but have a lesser impact on hemostasis. We found that the extrinsic tissue factor (TF) coagulation initiation complex can selectively activate the antihemophilic cofactor, FVIII, triggering the hemostatic intrinsic coagulation pathway independently of thrombin feedback loops. In a mouse model with a relatively mild thrombogenic lesion, TF-dependent FVIII activation sets the threshold for thrombus formation through contact phase-generated FIXa. In vitro, FXa stably associated with TF-FVIIa activates FVIII, but not FV. Moreover, nascent FXa product of TF-FVIIa can transiently escape the slow kinetics of Kunitz-type inhibition by TF pathway inhibitor and preferentially activates FVIII over FV. Thus, TF synergistically primes FIXa-dependent thrombin generation independently of cofactor activation by thrombin. Accordingly, FVIIa mutants deficient in direct TF-dependent thrombin generation, but preserving FVIIIa generation by nascent FXa, can support intrinsic pathway coagulation. In ex vivo flowing blood, a TF-FVIIa mutant complex with impaired free FXa generation but activating both FVIII and FIX supports efficient FVIII-dependent thrombus formation. Thus, a previously unrecognized TF-initiated pathway directly yielding FVIIIa-FIXa intrinsic tenase complex may be prohemostatic before further coagulation amplification by thrombin-dependent feedback loops enhances the risk of thrombosis.


Assuntos
Coagulação Sanguínea , Fator VIII/metabolismo , Fator VIIa/metabolismo , Fator Xa/metabolismo , Tromboplastina/metabolismo , Fator VIIIa/metabolismo , Humanos , Trombina/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 37(7): 1323-1331, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28495929

RESUMO

OBJECTIVE: Coagulation initiation by tissue factor (TF) is regulated by cellular inhibitors, cell surface availability of procoagulant phosphatidylserine, and thiol-disulfide exchange. How these mechanisms contribute to keeping TF in a noncoagulant state and to generating prothrombotic TF remain incompletely understood. APPROACH AND RESULTS: Here, we study the activation of TF in primary macrophages by a combination of pharmacological, genetic, and biochemical approaches. We demonstrate that primed macrophages effectively control TF cell surface activity by receptor internalization. After cell injury, ATP signals through the purinergic receptor P2rx7 induce release of TF+ microvesicles. TF cell surface availability for release onto microvesicles is regulated by the GTPase arf6 associated with integrin α4ß1. Furthermore, microvesicles proteome analysis identifies activation of Gαi2 as a participating factor in the release of microvesicles with prothrombotic activity in flowing blood. ATP not only prevents TF and phosphatidylserine internalization but also induces TF conversion to a conformation with high affinity for its ligand, coagulation factor VII. Although inhibition of dynamin-dependent internalization also exposes outer membrane procoagulant phosphatidylserine, the resulting TF+ microvesicles distinctly lack protein disulfide isomerase and high affinity TF and fail to produce fibrin strands typical for microvesicles generated by thrombo-inflammatory P2rx7 activation. CONCLUSIONS: These data show that procoagulant phospholipid exposure is not sufficient and that TF affinity maturation is required to generate prothrombotic microvesicles from a variety of cell types. These findings are significant for understanding TF-initiated thrombosis and should be considered in designing functional microvesicles-based diagnostic approaches.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Coagulação Sanguínea , Integrina alfa4/metabolismo , Integrina alfa4beta1/metabolismo , Macrófagos/metabolismo , Tromboplastina/metabolismo , Trombose/metabolismo , Fator 6 de Ribosilação do ADP , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Fator VIIa/metabolismo , Técnicas de Introdução de Genes , Genótipo , Humanos , Integrina alfa4/genética , Integrina alfa4beta1/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Fosfolipídeos/metabolismo , Transporte Proteico , Receptores Purinérgicos P2X7/metabolismo , Transdução de Sinais , Trombose/sangue , Trombose/genética , Transfecção
6.
Thromb Res ; 140 Suppl 1: S1-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27067961

RESUMO

Cancer-associated thrombosis remains a significant complication in the clinical management of cancer and interactions of the hemostatic system with cancer biology continue to be elucidated. Here, we review recent progress in our understanding of tissue factor (TF) regulation and procoagulant activation, TF signaling in cancer and immune cells, and the expanding roles of the coagulation system in stem cell niches and the tumor microenvironment. The extravascular functions of coagulant and anti-coagulant pathways have significant implications not only for tumor progression, but also for the selection of appropriate target specific anticoagulants in the therapy of cancer patients.


Assuntos
Anticoagulantes/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Neoplasias/complicações , Tromboplastina/metabolismo , Trombose/complicações , Trombose/tratamento farmacológico , Animais , Anticoagulantes/farmacologia , Humanos , Terapia de Alvo Molecular , Neoplasias/sangue , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombose/sangue , Trombose/metabolismo , Microambiente Tumoral/efeitos dos fármacos
7.
J Clin Invest ; 125(4): 1471-84, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25705884

RESUMO

Extracellular ATP is a signal of tissue damage and induces macrophage responses that amplify inflammation and coagulation. Here we demonstrate that ATP signaling through macrophage P2X7 receptors uncouples the thioredoxin (TRX)/TRX reductase (TRXR) system and activates the inflammasome through endosome-generated ROS. TRXR and inflammasome activity promoted filopodia formation, cellular release of reduced TRX, and generation of extracellular thiol pathway-dependent, procoagulant microparticles (MPs). Additionally, inflammasome-induced activation of an intracellular caspase-1/calpain cysteine protease cascade degraded filamin, thereby severing bonds between the cytoskeleton and tissue factor (TF), the cell surface receptor responsible for coagulation activation. This cascade enabled TF trafficking from rafts to filopodia and ultimately onto phosphatidylserine-positive, highly procoagulant MPs. Furthermore, caspase-1 specifically facilitated cell surface actin exposure, which was required for the final release of highly procoagulant MPs from filopodia. Together, the results of this study delineate a thromboinflammatory pathway and suggest that components of this pathway have potential as pharmacological targets to simultaneously attenuate inflammation and innate immune cell-induced thrombosis.


Assuntos
Trifosfato de Adenosina/fisiologia , Caspase 1/fisiologia , Micropartículas Derivadas de Células/fisiologia , Macrófagos/fisiologia , Transdução de Sinais/fisiologia , Actinas/fisiologia , Animais , Líquido Extracelular , Imunidade Inata , Inflamassomos/fisiologia , Inflamação/fisiopatologia , Microdomínios da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Pseudópodes/fisiologia , Receptores Purinérgicos P2X7/fisiologia , Tiorredoxina Dissulfeto Redutase/fisiologia , Tiorredoxinas/fisiologia , Trombose/fisiopatologia
8.
Semin Immunopathol ; 34(1): 133-49, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21971685

RESUMO

Protease-activated receptors (PARs) are G protein-coupled receptors that are activated by proteolytical cleavage of the amino-terminus and thereby act as sensors for extracellular proteases. While coagulation proteases activate PARs to regulate hemostasis, thrombosis, and cardiovascular function, PAR2 is also activated in extravascular locations by a broad array of serine proteases, including trypsin, tissue kallikreins, coagulation factors VIIa and Xa, mast cell tryptase, and transmembrane serine proteases. Administration of PAR2-specific agonistic and antagonistic peptides, as well as studies in PAR2 knockout mice, identified critical functions of PAR2 in development, inflammation, immunity, and angiogenesis. Here, we review these roles of PAR2 with an emphasis on the role of coagulation and other extracellular protease pathways that cleave PAR2 in epithelial, immune, and neuronal cells to regulate physiological and pathophysiological processes.


Assuntos
Inflamação/imunologia , Receptor PAR-2/imunologia , Transdução de Sinais/imunologia , Animais , Coagulação Sanguínea/genética , Coagulação Sanguínea/imunologia , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/imunologia , Proteínas Sanguíneas/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/imunologia , Receptor PAR-2/genética , Transdução de Sinais/genética , Trombose/sangue , Trombose/genética , Trombose/imunologia , Trombose/patologia
9.
Int J Cancer ; 127(5): 1096-105, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20143389

RESUMO

Treatment of pancreatic cancer remains a major challenge and new anticancer drugs are urgently required. Our study presents the marine natural compound spongistatin 1 as a promising experimental drug. Spongistatin 1 was applied in an orthotopic in vivo model of human pancreatic cancer. Spongistatin 1 significantly reduced tumor growth, which correlates with a strong apoptosis induction (DNA-fragmentation) and long-term effects on clonogenic survival of pancreatic tumor cells (L3.6pl) in vitro. In addition, the formation of metastasis was reduced in spongistatin 1-treated mice, which is in line with a diminished MMP-9 activity in tumor tissue determined by zymography. Based on the pronounced efficacy of spongistatin 1, the underlying mechanisms were studied in more detail. In vitro adhesion, as well as migration, and invasion assays showed spongistatin 1 to influence these critical steps in the metastatic cascade. Furthermore, spongistatin 1 induced anoikis in L3.6pl cells. Exposure to spongistatin 1 leads to phosphorylation, and thus inactivation of the antiapoptotic protein Bcl-2 in pancreatic tumor cells. siRNA experiments silencing Bcl-2 suggest a role of Bcl-2 in anoikis and cell migration. Taken together, spongistatin 1 not only proved to be a potent experimental drug but also served as a chemical tool to examine the role of the antiapoptotic protein Bcl-2 in pancreas carcinoma, thereby supporting the hypothesis of a link between apoptosis signaling and metastasis.


Assuntos
Neoplasias Hepáticas Experimentais/tratamento farmacológico , Macrolídeos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Moduladores de Tubulina/farmacologia , Animais , Anoikis/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Western Blotting , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Feminino , Humanos , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/secundário , Metástase Linfática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
10.
FASEB J ; 23(4): 1127-37, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19056838

RESUMO

The aims of the study were to meet the demand of new tubulin antagonists with fewer side effects by characterizing the antiangiogenic properties of the experimental compound spongistatin 1, and to elucidate nonmitotic mechanisms by which tubulin antagonists inhibit angiogenesis. Although tubulin-inhibiting drugs and their antiangiogenic properties have been investigated for a long time, surprisingly little is known about their underlying mechanisms of action. Antiangiogenic effects of spongistatin 1 were investigated in endothelial cells in vitro, including functional cell-based assays, live-cell imaging, and a kinome array, and in the mouse cornea pocket assay in vivo. Spongistatin 1 inhibited angiogenesis at nanomolar concentrations (IC(50): cytotoxicity>50 nM, proliferation 100 pM, migration 1.0 nM, tube formation 1.0 nM, chemotaxis 1.0 nM, aortic ring sprouting 500 pM, neovascularization in vivo 10 microg/kg). Further, a kinome array and validating data showed that spongistatin 1 inhibits the phosphorylation activity of protein kinase Calpha (PKCalpha), an essential kinase in angiogenesis, and its translocation to the membrane. Thus, we conclude that PKCalpha might be an important target for the antiangiogenic effects of tubulin antagonism. In addition, the data from the kinase array suggest that different tubulin antagonists might have individual intracellular actions.


Assuntos
Inibidores da Angiogênese/farmacologia , Macrolídeos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Proteína Quinase C-alfa/antagonistas & inibidores , Tubulina (Proteína)/metabolismo , Inibidores da Angiogênese/metabolismo , Animais , Aorta Abdominal/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio/citologia , Humanos , Imuno-Histoquímica , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos , Microtúbulos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Poríferos/química , Transfecção , Veias Umbilicais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...