Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Lupus ; 25(2): 162-76, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26391610

RESUMEN

Patients with antiphospholipid syndrome (APS) produce antiphospholipid antibodies (aPL) and develop vascular thrombosis that may occur in large or small vessels in the arterial or venous beds. On the other hand, many individuals produce aPL and yet never develop thrombotic events. Toll-like receptor 4 (TLR4) appears to be necessary for aPL-mediated prothrombotic effects in venous and microvascular models of thrombosis, but its role in arterial thrombosis has not been studied. Here, we propose that aPL alone are insufficient to cause thrombotic events in an arterial model of APS, and that a concomitant trigger of innate immunity (e.g. TLR4 activation) is required. We show specifically that anti-ß2-glycoprotein I (anti-ß2GPI) antibodies, a subset of aPL, accelerated thrombus formation in C57BL/6 wild-type, but not TLR4-deficient, mice in a ferric chloride-induced carotid artery injury model. These aPL bound to arterial and venous endothelial cells, particularly in the presence of ß2GPI, and to human TLR4 by enzyme-linked immunoassay. Arterial endothelium from aPL-treated mice had enhanced leukocyte adhesion, compared to control IgG-treated mice. In addition, aPL treatment of mice enhanced expression of tissue factor (TF) in leukocytes induced by the TLR4 ligand lipopolysaccharide (LPS). aPL also enhanced LPS-induced TF expression in human leukocytes in vitro. Our findings support a mechanism in which aPL enhance TF expression by leukocytes, as well as augment adhesion of leukocytes to the arterial endothelium. The activation of TLR4 in aPL-positive individuals may be required to trigger thrombotic events.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Trombosis/inmunología , Receptor Toll-Like 4/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Síndrome Antifosfolípido/inmunología , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Femenino , Humanos , Inmunidad Innata , Leucocitos/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Tromboplastina/inmunología , beta 2 Glicoproteína I/antagonistas & inhibidores , beta 2 Glicoproteína I/inmunología
2.
J Mol Cell Cardiol ; 52(5): 1056-65, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22326437

RESUMEN

Tissue Factor (TF) is expressed in various cell types of the heart, such as cardiomyocytes. In addition to its role in the initiation of blood coagulation, the TF:FVIIa complex protects cells from apoptosis. There are two isoforms of Tissue Factor (TF): "full length" (fl)TF--an integral membrane protein, and alternatively spliced (as)TF--a protein that lacks a transmembrane domain and can thus be secreted in a soluble form. Whether asTF or flTF affects apoptosis of cardiomyocytes is unknown. In this study, we examined whether asTF or flTF protects murine cardiomyocytes from TNF-α-induced apoptosis. We used murine cardiomyocytic HL-1 cells and primary murine embryonic cardiomyocytes that overexpressed either murine asTF or murine flTF, and stimulated them with TNF-α to initiate cell death. Apoptosis was assessed by annexin-V assay, propidium iodide assay, as well as activation of caspase-3 and -9. In addition, signaling via integrins, Akt, NFκB and Erk1/2, and gene-expression of Bcl-2 family members were analyzed. We here report that overexpression of asTF reduced phosphatidylserine exposure upon TNF-α-stimulation. asTF overexpression led to an increased expression and phosphorylation of Akt, as well as up-regulation of the anti-apoptotic protein Bcl-x(L). The anti-apoptotic effects of asTF overexpression were mediated via α(V)ß(3)/Akt/NFκB signaling and were dependent on Bcl-x(L) expression in HL-1 cells. The anti-apoptotic activity of asTF was also observed using primary cardiomyocytes. Analogous yet less pronounced anti-apoptotic sequelae were observed due to overexpression of flTF. Importantly, cardiomyocytes deficient in TF exhibited increased apoptosis compared to wild type cells. We propose that asTF and flTF protect cardiomyocytes against TNF-α-induced apoptosis via activation of specific signaling pathways, and up-regulation of anti-apoptotic members of the Bcl-2 protein family.


Asunto(s)
Apoptosis , Miocitos Cardíacos/fisiología , Tromboplastina/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Línea Celular , Expresión Génica , Sistema de Señalización de MAP Quinasas , Ratones , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Regulación hacia Arriba , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
3.
Nat Med ; 6(12): 1355-61, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11100120

RESUMEN

Activation of the zinc-finger transcription factor early growth response (Egr)-1, initially linked to developmental processes, is shown here to function as a master switch activated by ischemia to trigger expression of pivotal regulators of inflammation, coagulation and vascular hyperpermeability. Chemokine, adhesion receptor, procoagulant and permeability-related genes are coordinately upregulated by rapid ischemia-mediated activation of Egr-1. Deletion of the gene encoding Egr-1 strikingly diminished expression of these mediators of vascular injury in a murine model of lung ischemia/reperfusion, and enhanced animal survival and organ function. Rapid activation of Egr-1 in response to oxygen deprivation primes the vasculature for dysfunction manifest during reperfusion. These studies define a central and unifying role for Egr-1 activation in the pathogenesis of ischemic tissue damage.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Pulmón/patología , Daño por Reperfusión/etiología , Factores de Transcripción/metabolismo , Animales , Factores de Coagulación Sanguínea/biosíntesis , Quimiocinas/biosíntesis , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Factores de Crecimiento Endotelial/biosíntesis , Genes de Cambio , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Lipopolisacáridos/toxicidad , Pulmón/irrigación sanguínea , Linfocinas/biosíntesis , Ratones , Ratones Mutantes , Factores de Transcripción/genética , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Dedos de Zinc/genética
4.
Thromb Res ; 199: 21-31, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33385797

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is associated with a hypercoagulable state and high mortality. Increases in the plasma levels of tumor marker carbohydrate antigen (CA) 19-9 are used in diagnosis and follow-up but have also been reported to precede venous thromboembolism (VTE). AIMS: We examined the association between CA 19-9 and thrombin generation (TG) in plasma from PDAC patients, as well as their association with coagulation biomarkers prior to pancreatic surgery. In addition, we determined the effect of commercial sources of CA 19-9 on TG. METHODS: We collected plasma from 58 treatment-naïve PDAC patients without any signs of VTE. We measured levels of CA 19-9, FVIII, fibrinogen, D-dimer, antithrombin and extracellular vesicle (EV) tissue factor (TF) activity and TG using a Calibrated Automated Thrombogram (CAT). The effect of different commercial sources of CA 19-9 on TG in Standard Human Plasma (SHP) was also studied. RESULTS: Patient plasma samples were divided into 4 preoperative groups based on the level of CA 19-9: none < 2, low = 3-200, high = 201-1000, and very high > 1000 U/mL. CA 19-9 levels were associated with several of the TG parameters, including endogenous thrombin potential, peak, and time to peak. CA 19-9 did not associate with any of the coagulation biomarkers. Spiking of SHP with CA 19-9 increased TG but this was decreased by an anti-TF antibody. CONCLUSIONS: CA 19-9 was associated with TG in patients prior to any pancreatic cancer treatments or signs of VTE. Some commercial sources of CA 19-9 enhanced TG in SHP seemingly due to contaminating TF.


Asunto(s)
Neoplasias Pancreáticas , Trombina , Biomarcadores de Tumor , Pruebas de Coagulación Sanguínea , Antígeno CA-19-9 , Carcinoma Ductal Pancreático , Humanos
5.
J Exp Med ; 174(6): 1517-26, 1991 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-1744583

RESUMEN

Lipopolysaccharide (LPS) activation of cells of monocytic lineage leads to rapid and transient expression of a set of inflammatory gene products, including tissue factor (TF). This transmembrane receptor is the major cellular initiator of the blood coagulation cascades, and induced expression of TF is postulated to play a role in inflammation. Functional studies using transfected THP-1 monocytic cells revealed the presence of a 56-bp LPS response element (LRE) within the TF promoter that conferred LPS responsiveness to a heterologous promoter. LPS stimulation of these cells activated proteins that bound to nucleotide sequences within the LRE resembling consensus binding sites for activator protein 1 (AP-1) and nuclear factor kappa B (NF-kappa B). Induction of the TF gene may represent a prototypic example of gene activation in monocytic cells by assembly of transcription factor complexes, and may clarify the role of AP-1 and NF-kappa B in the regulation of other LPS-responsive genes.


Asunto(s)
Lipopolisacáridos/metabolismo , FN-kappa B/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Tromboplastina/genética , Transcripción Genética/efectos de los fármacos , Secuencia de Bases , Sitios de Unión , Humanos , Datos de Secuencia Molecular , Mutagénesis , Regiones Promotoras Genéticas , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
6.
Lupus ; 19(4): 370-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20353972

RESUMEN

Antiphospholipid syndrome (APS) is an acquired autoimmune disorder defined by the presence of an antiphospholipid antibody (aPL) and the occurrence of at least one associated clinical condition that includes venous thrombosis, arterial thrombosis or pregnancy morbidity. The aPL detected in APS have long been thought to have a direct prothrombotic effect in vivo. However, the pathophysiology underlying their coagulopathic effect has not been defined. Emerging data suggest a role for the procoagulant protein tissue factor (TF). In this review we provide an overview of TF, describe mouse models used in the evaluation of the role of TF in thrombosis, as well as summarize recent work on TF and APS.


Asunto(s)
Síndrome Antifosfolípido/fisiopatología , Tromboplastina/metabolismo , Trombosis/etiología , Animales , Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Embarazo , Complicaciones del Embarazo/inmunología , Trombosis/inmunología
7.
Thromb Res ; 178: 145-154, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31030034

RESUMEN

The mammalian hemostatic system involves complex interactions between protein components of the coagulation cascade and platelets. The fibrinolytic system removes the hemostatic plug. Dysregulation of coagulation or fibrinolytic systems can induce bleeding or thrombosis. Animals, such as snakes, worms and insects, have evolved to express proteins that modulate the mammalian coagulation and fibrinolytic systems. Many of these proteins have been isolated and characterized. Understanding the mechanisms by which these exogenous factors from venoms and animal saliva modulate the mammalian coagulation and fibrinolytic systems has led to a better understanding of these systems. Furthermore, some of these exogenous proteins are used in diagnostic assays and as therapeutic drugs. This review summarizes our current knowledge of exogenous proteins from venom and saliva that either activate or inhibit the mammalian coagulation and fibrinolytic systems.


Asunto(s)
Artrópodos/química , Coagulación Sanguínea/efectos de los fármacos , Fibrinólisis/efectos de los fármacos , Hemostasis/efectos de los fármacos , Insectos/química , Nematodos/química , Serpientes/sangre , Ponzoñas/sangre , Animales , Humanos
8.
J Thromb Haemost ; 17(1): 169-182, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30472780

RESUMEN

Essentials Tissue factor (TF) isoforms are expressed in pancreatic neuroendocrine tumors (pNET). TF knockdown inhibits proliferation of human pNET cells in vitro. mTOR kinase inhibitor sapanisertib/MLN0128 suppresses TF expression in human pNET cells. Sapanisertib suppresses TF expression and activity and reduces the growth of pNET tumors in vivo. SUMMARY: Background Full-length tissue factor (flTF) and alternatively spliced TF (asTF) contribute to growth and spread of pancreatic ductal adenocarcinoma. It is unknown, however, if flTF and/or asTF contribute to the pathobiology of pancreatic neuroendocrine tumors (pNETs). Objective To assess TF expression in pNETs and the effects of mTOR complex 1/2 (mTORC1/2) inhibition on pNET growth. Methods Human pNET specimens were immunostained for TF. Human pNET cell lines QGP1 and BON were evaluated for TF expression and responsiveness to mTOR inhibition. shRNA were used to knock down TF in BON. TF cofactor activity was assessed using a two-step FXa generation assay. TF promoter activity was assessed using transient transfection of human TF promoter-driven reporter constructs into cells. Mice bearing orthotopic BON tumors were treated with the mTORC1/2 ATP site competitive inhibitor sapanisertib/MLN0128 (3 mg kg-1 , oral gavage) for 34 days. Results Immunostaining of pNET tissue revealed flTF and asTF expression. BON and QGP1 expressed both TF isoforms, with BON exhibiting higher levels. shRNA directed against TF suppressed BON proliferation in vitro. Treatment of BON with sapanisertib inhibited mTOR signaling and suppressed TF levels. BON tumors grown in mice treated with sapanisertib had significantly less TF protein and cofactor activity, and were smaller compared with tumors grown in control mice. Conclusions TF isoforms are expressed in pNETs. Sapanisertib suppresses TF mRNA and protein expression as well as TF cofactor activity in vitro and in vivo. Thus, further studies are warranted to evaluate the clinical utility of TF-suppressing mTORC1/2 inhibitor sapanisertib in pNET management.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tromboplastina/metabolismo , Animales , Línea Celular Tumoral , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones Desnudos , Tumores Neuroendocrinos/enzimología , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Tromboplastina/genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Thromb Haemost ; 16(4): 749-758, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29427323

RESUMEN

Essentials Androgen deprivation increases the rate of venous thromboembolism in prostate cancer patients. We characterized androgen receptor-mediated tissue factor regulation in prostate epithelial cells. Androgen receptor is dampening tissue factor expression in prostate epithelial cells. Androgen deprivation could enhance tissue factor expression and raise venous thromboembolism rates. SUMMARY: Background Prostate cancer is one of the leading causes of cancer death in men. Advanced prostate cancer is usually treated by androgen deprivation therapy (ADT), which is aimed at reducing circulating testosterone levels to reduce cancer growth. There is growing evidence that ADT can increase the rate of venous thromboembolism (VTE) in prostate cancer patients. The tissue factor (TF) gene is one of the most important mediators of coagulation and VTE, but, so far, there are limited data on androgen receptor (AR)-mediated TF gene expression. Objectives To characterize AR-mediated TF regulation in vitro and in vivo. Methods We used the androgen-dependent prostate cancer cell lines LNCaP and MyC-CaP to test whether TF expression is regulated by AR. Furthermore, we cloned the TF gene promoter into a luciferase reporter vector to identify the transcription factor-binding sites that mediate TF regulation downstream of AR. Finally, we used castration experiments in mice to characterize AR-mediated TF regulation in vivo. Results TF is directly regulated by AR. In LNCaP cells, nuclear factor-κB signaling and EGR1 mediate TF expression. By using castration experiments in mice, we could detect upregulation of TF and early growth response protein 1 mRNA and protein expression in prostate epithelial cells. Conclusion AR is crucial for dampening TF expression, which could be important for increased TF expression and TF-positive microvesicle release in androgen-deprived prostate cancer patients.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Células Epiteliales/metabolismo , FN-kappa B/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Tromboplastina/metabolismo , Antagonistas de Andrógenos/efectos adversos , Andrógenos/farmacología , Animales , Sitios de Unión , Línea Celular Tumoral , Dihidrotestosterona/farmacología , Regulación hacia Abajo , Humanos , Masculino , Ratones Endogámicos C57BL , Orquiectomía , Regiones Promotoras Genéticas , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Unión Proteica , Receptores Androgénicos/efectos de los fármacos , Transducción de Señal , Tromboplastina/genética , Tromboembolia Venosa/inducido químicamente , Tromboembolia Venosa/genética , Tromboembolia Venosa/metabolismo
10.
PLoS One ; 13(11): e0207387, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30412630

RESUMEN

Coagulation activation and venous thromboembolism (VTE) are hallmarks of cancer; however, there is an unmet need of improved biomarkers for individualized anticoagulant treatment. The present sub-study of the RASTEN trial was designed to explore the role of coagulation biomarkers in predicting VTE risk and outcome in a homogenous cancer patient population. RASTEN is a multicenter, randomized phase-3 trial investigating the survival effect of low molecular weight heparin enoxaparin when added to standard treatment in newly diagnosed small cell lung cancer (SCLC) patients. Plasma collected at baseline, during treatment, and at follow-up was used in this ad hoc sub-study (N = 242). Systemic coagulation was assessed using four assays reflecting various facets of the coagulation system: Total tissue factor (TF); extracellular vesicle associated TF (EV-TF); procoagulant phospholipids (PPL); and thrombin generation (TG). We found small variations of biomarker levels between baseline, during treatment and at follow-up, and appeared independent on low molecular weight heparin treatment. Overall, none of the measured biomarkers at any time-point did significantly associate with VTE incidence, although increased total TF at baseline showed significant association in control patients not receiving low molecular weight heparin (P = 0.03). Increased TG-Peak was significantly associated with decreased overall survival (OS; P = 0.03), especially in patients with extensive disease. Low baseline EV-TF predicted a worse survival in the low molecular weight heparin as compared with the control group (HR 1.42; 95% CI 1.04-1.95; P = 0.03; P for interaction = 0.12). We conclude that the value of the analyzed coagulation biomarkers for the prediction of VTE risk was very limited in SCLC patients. The associations between TG-Peak and EV-TF with patient survival and response to low molecular weight heparin therapy, respectively, warrant further studies on the role of coagulation activation in SCLC aggressiveness.


Asunto(s)
Biomarcadores de Tumor/sangre , Heparina de Bajo-Peso-Molecular/administración & dosificación , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Tromboembolia Venosa , Anciano , Supervivencia sin Enfermedad , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Incidencia , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Fosfolípidos/sangre , Carcinoma Pulmonar de Células Pequeñas/sangre , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Tasa de Supervivencia , Tromboplastina/metabolismo , Tromboembolia Venosa/sangre , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/mortalidad
11.
Thromb Haemost ; 118(2): 229-250, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29378352

RESUMEN

Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.


Asunto(s)
Tromboembolia/terapia , Trombosis/sangre , Trombosis/terapia , Anticoagulantes/uso terapéutico , Biomarcadores/sangre , Coagulación Sanguínea , Eritrocitos/metabolismo , Factor VIII/metabolismo , Factor XII/metabolismo , Factor XIII/metabolismo , Humanos , Macrófagos/metabolismo , Países Bajos , Fenotipo , Placa Aterosclerótica/sangre , Placa Aterosclerótica/diagnóstico , Placa Aterosclerótica/terapia , Polifosfatos/metabolismo , Factores de Riesgo , Transducción de Señal , Tromboembolia/sangre , Tromboembolia/diagnóstico , Trombosis/diagnóstico
12.
J Clin Invest ; 105(11): 1547-54, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10841513

RESUMEN

Recent studies indicate that tissue factor (TF) acts in embryogenesis, metastasis, and angiogenesis. Three independent groups showed that targeted disruption of the murine TF (mTF) gene results in 90% lethality of mTF null embryos at embryonic days 9. 5-10.5. We have demonstrated that expression of wild-type human TF (hTF) from a minigene rescues the embryonic lethality of mTF null embryos. To investigate the role of TF in embryogenesis, we made mutant hTF minigenes whose products either bound FVII/VIIa at a reduced level or lacked the cytoplasmic domain. Two independent transgenic lines expressing the hTF extracellular domain mutant failed to rescue the embryonic lethality of mTF null embryos, suggesting that FVII/VIIa binding by TF, proteolytic activity by the TF/FVIIa complex, or both were required for embryogenesis. In contrast, two transgenic lines expressing the hTF cytoplasmic domain mutant rescued the embryonic lethality of mTF null embryos, indicating that the cytoplasmic domain of TF was not required for embryogenesis. We propose that TF/FVIIa-dependent extracellular protease activity is required for embryogenesis.


Asunto(s)
Desarrollo Embrionario y Fetal , Tromboplastina/fisiología , Animales , Células CHO , Cricetinae , Citoplasma/química , Femenino , Humanos , Ratones , Ratones Transgénicos , Embarazo , ARN Mensajero/análisis , Tromboplastina/genética , Transgenes
13.
J Clin Invest ; 91(5): 2031-9, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8486772

RESUMEN

Macrophage-derived apolipoprotein (apo) E and multimers of a synthetic apo E-peptide display monokine-like functions by inhibiting mitogen- or antigen-driven lymphocyte proliferation. This study demonstrated how the target lymphocyte itself can modulate macrophage apo E production. The lymphokine interferon-gamma (IFN) dramatically inhibited the accumulation of apo E in the supernatant of human monocytic THP-1 cells when present during phorbol myristate acetate-induced differentiation. A similar effect was observed when IFN was added to differentiated THP-1 cells. Treatment with IFN did not change the steady-state levels of apo E mRNA. Furthermore, in the presence of IFN no increased degradation or increased uptake of extracellular apo E was detected. Pulse-chase experiments indicated that IFN reduced the accumulation of extracellular apo E and increased the degradation of intracellular apo E. The inhibitory effect of IFN on apo E production also was observed in human monocyte-derived macrophages. Thus, our data demonstrated that IFN inhibited macrophage apo E production by posttranslational mechanisms. This represents a previously uncharacterized immunoregulatory interaction and lends further support to a relationship between lipid metabolism and the immune system.


Asunto(s)
Apolipoproteínas E/biosíntesis , Interferón gamma/farmacología , Macrófagos/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/aislamiento & purificación , Diferenciación Celular/efectos de los fármacos , Línea Celular , Sondas de ADN , Humanos , Cinética , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Monocitos/citología , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Proteínas Recombinantes , Acetato de Tetradecanoilforbol/farmacología
14.
J Clin Invest ; 101(3): 560-9, 1998 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9449688

RESUMEN

Tissue factor (TF) expression is associated with life-threatening thrombosis in a variety of human diseases, including sepsis, cancer, and atherosclerosis. Recently, it was shown that inactivation of the murine TF (mTF) gene results in embryonic lethality. To date, despite extensive studies on the regulation of the TF promoter in vitro, no studies have examined the cis-acting regulatory elements that control TF gene expression in vivo. Here we report that a human TF (hTF) minigene containing the human TF promoter and human TF cDNA directed a low level (approximately 1% relative to mouse TF) of both constitutive and LPS-inducible human TF expression in transgenic mice. Importantly, the human TF minigene rescued the embryonic lethality of murine TF null embryos, suggesting that human TF substituted for murine TF during embryogenesis. Rescued mice (mTF-/-, hTF+), which expressed low levels (approximately 1%) of TF activity, developed normally with no signs of a bleeding diathesis, suggesting that low TF expression can maintain hemostasis compatible with normal survival. These studies establish a novel mouse model system that can be used to examine the regulation of the human TF gene in vivo and the impact of low TF levels on the hemostatic balance in various thrombotic diseases.


Asunto(s)
Hemostáticos/metabolismo , Tromboplastina/metabolismo , Animales , Femenino , Expresión Génica , Hemostasis , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Tromboplastina/biosíntesis , Tromboplastina/genética , Distribución Tisular
15.
J Clin Invest ; 99(4): 737-44, 1997 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9045878

RESUMEN

Using flow channel, we report that the application of a laminar shear stress induced a transient increase of tissue factor (TF) procoagulant activity in human umbilical vein endothelial cells (HUVEC), which was accompanied by a rapid and transient induction of the TF mRNA in the HUVEC. Functional analysis of the 2.2 kb TF 5' promoter indicated that a GC-rich region containing three copies each of the EGR-1 and Sp1 sites was required for induction. Mutation of the Sp1 sites, but not the EGR-1 sites, attenuated the response of TF promoter to shear stress. Thus, Sp1 is a newly defined shear stress responsive element. Electrophoretic mobility shift assays showed there was no increase in binding of nuclear extracts from sheared cells to an Sp1 consensus site. In contrast, immunoblotting of these nuclear extracts with antibody against transcription factor Sp1 demonstrated that shear stress increased the phosphorylation of Sp1. We also showed that shear stress, like the phosphatase inhibitor okadaic acid, increased the transcriptional activity of Sp1. These findings suggest that the shear stress induction of TF gene expression is mediated through an increased Sp1 transcriptional activity with a concomitant hyperphosphorylation of Sp1.


Asunto(s)
Regulación de la Expresión Génica , Tromboplastina/genética , Endotelio Vascular/metabolismo , Hemorreología , Humanos , Fosforilación , Regiones Promotoras Genéticas/fisiología , Unión Proteica/genética , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/metabolismo , Tromboplastina/metabolismo , Transcripción Genética , Venas Umbilicales
16.
J Clin Invest ; 102(9): 1645-52, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9802878

RESUMEN

Recognition of bacterial endotoxin (LPS) elicits multiple host responses, including activation of cells of the innate immune system. LPS exposure occurs repeatedly during septicemia, making strict regulation of gene expression necessary. Such regulation might prevent, for example, the continuous production of proinflammatory cytokines such as tumor necrosis factor (TNF), which could lead to severe vascular collapse. Tolerance to LPS is characterized by a diminished production of TNF during prolonged exposure to LPS, and is therefore likely to represent an essential control mechanism during sepsis. In the present study, which uses mice with genetic deletions of the proteins of NF-kappaB complex, we provide data demonstrating that increased expression of the p50 subunit of NF-kappaB directly results in the downregulation of LPS-induced TNF production. This contention is supported by the following observations: (1) tolerance to LPS is not induced in macrophages from p50-/- mice; (2) long-term pretreatment with LPS does not block synthesis of TNF mRNA in p50-/- macrophages (in contrast to wild-type macrophages); (3) ectopic overexpression of p50 reduces transcriptional activation of the murine TNF promoter; and (4) analysis of the four kappaB sites from the murine TNF promoter demonstrates that binding of p50 homodimers to the positively acting kappaB3 element is associated with development of the LPS-tolerant phenotype. Thus, p50 expression plays a key role in the development of LPS tolerance.


Asunto(s)
Inmunidad Innata/fisiología , Macrófagos Peritoneales/inmunología , FN-kappa B/inmunología , Animales , Células Cultivadas , Regulación hacia Abajo , Tolerancia Inmunológica , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Noqueados , Subunidad p50 de NF-kappa B , Regiones Promotoras Genéticas , ARN Mensajero , Factor de Necrosis Tumoral alfa/genética
17.
J Clin Invest ; 96(3): 1621-30, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7544811

RESUMEN

Bleomycin-induced lung injury is an established murine model of human pulmonary fibrosis. Although procoagulant molecules (e.g., tissue factor [TF]) and fibrinolytic components (e.g., urokinase [u-PA] and type 1 plasminogen activator inhibitor [PAI-1]) have been detected in alveolar fluid from injured lungs, the origin of these molecules remains unknown. We therefore examined the expression of procoagulant and fibrinolytic components in relation to the distribution of parenchymal fibrin in bleomycin-injured lungs. Extravascular fibrin localized to the alveolar and extracellular matrix in injured lung tissue. Injured lung tissue extracts contained elevated levels of PAI-1 activity and decreased levels of u-PA activity. Whole lung PAI-1 and TF mRNAs were dramatically induced by lung injury. In situ hybridization of injured lungs revealed that PAI-1, u-PA, and TF mRNAs were induced within the fibrin-rich fibroproliferative lesions, primarily in fibroblast-like and macrophagelike cells, respectively, while TF mRNA was also induced in perilesional alveolar cells. Taken together, these observations suggest that the induction of PAI-1 and TF gene expression plays and important role in the formation and persistence of extracellular fibrin in bleomycin injured murine lungs.


Asunto(s)
Bleomicina/toxicidad , Expresión Génica , Pulmón/patología , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Tromboplastina/biosíntesis , Activador de Tejido Plasminógeno/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Animales , Modelos Animales de Enfermedad , Femenino , Fibrina/análisis , Fibrina/biosíntesis , Fibrinólisis , Expresión Génica/efectos de los fármacos , Hibridación in Situ , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Factores de Tiempo
18.
J Clin Invest ; 92(1): 349-58, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8326003

RESUMEN

Hemostasis in the brain is of paramount importance because bleeding into the neural parenchyma can result in paralysis, coma, and death. Consistent with this sensitivity to hemorrhage, the brain contains large amounts of tissue factor (TF), the major cellular initiator of the coagulation protease cascades. However, to date, the cellular source for TF in the central nervous system has not been identified. In this study, analysis of murine brain sections by in situ hybridization demonstrated high levels of TF mRNA in cells that expressed glial fibrillary acidic protein, a specific marker for astrocytes. Furthermore, primary mouse astrocyte cultures and astrocyte cell lines from mouse, rat, and human constitutively expressed TF mRNA and functional protein. These data indicated that astrocytes are the primary source of TF in the central nervous system. We propose that astrocytes forming the glia limitans around the neural vasculature and deep to the meninges are intimately involved in controlling hemorrhage in the brain. Finally, we observed an increase in TF mRNA expression in the brains of scrapie-infected mice. This modulation of TF expression in the absence of hemorrhage suggested that TF may function in processes other than hemostasis by altering protease generation in normal and diseased brain.


Asunto(s)
Astrocitos/metabolismo , Tromboplastina/metabolismo , Animales , Línea Celular , Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemostasis , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , ARN Mensajero/genética , Scrapie/metabolismo
19.
J Thromb Haemost ; 5(8): 1693-700, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17663739

RESUMEN

BACKGROUND: The tissue-specific pattern of tissue factor (TF) expression suggests that it plays a major role in the hemostatic protection of specific organs, such as the heart and lung. In support of this notion, we found that mice expressing very low levels of TF exhibit hemostatic defects in the heart and lung. Hemosiderosis and fibrosis are observed in the hearts of all low TF mice as early as 3 months of age. In contrast, TF(+/-) mice expressing approximately 50% of wild-type levels of TF had no detectable hemostatic defects. OBJECTIVE AND METHODS: The objective of this study was to determine the threshold of TF that is required to maintain hemostasis under normal and pathologic conditions, and to investigate the specific role of cardiac myocyte TF in heart hemostasis using mice with altered levels of TF expression in cardiac myocytes. RESULTS: First, we found that mice with 20% of wild-type levels of TF activity in their hearts had hemosiderosis and fibrosis by 6 months of age. Secondly, mice with a selective deletion of the TF gene in cardiac myocytes had a mild hemostatic defect under normal conditions but exhibited a significant increase in hemosiderosis and fibrosis after challenge with isoproterenol. Finally, we showed that cardiac myocyte-specific overexpression of TF abolished hemosiderin deposition and fibrosis in the hearts of low TF mice. CONCLUSIONS: Taken together, our results indicate that TF expression by cardiac myocytes is important to maintain heart hemostasis under normal and pathologic conditions.


Asunto(s)
Corazón/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Animales , Genotipo , Hemostasis , Humanos , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Modelos Genéticos , Tromboplastina/genética , Tromboplastina/fisiología , Distribución Tisular
20.
Mol Cell Biol ; 11(9): 4732-8, 1991 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1875949

RESUMEN

Tissue factor (TF) is transiently expressed in human monocytes exposed to the inflammatory agonist bacterial lipopolysaccharide (LPS). Since TF is the major cellular initiator of the coagulation protease cascades, it is inferred that its expression within the vasculature is strictly regulated. In this study, we investigated mechanisms which control TF mRNA expression in the human monocytic cell line THP-1. LPS induced a rapid and transient accumulation of the mature 2.2-kb TF mRNA, which was maximal at 2 h. After stimulation, the rate of transcription of the TF gene was increased (3.3 +/- 1.3)fold. In addition, we observed a significant change in TF mRNA stability: at 1 h after LPS stimulation, TF mRNA was stable during a 60-min period and had a half-life of greater than 120 min, whereas at 2 h, the half-life had declined to 25 +/- 5 min. Furthermore, a larger (3.4-kb) TF RNA species was induced in these cells; the size of this species and data from selective hybridizations with intron-specific probes are consistent with the presence of an unspliced copy of intron 1. These results demonstrate that the LPS-induced accumulation of TF mRNA levels in these monocytic cells is accomplished by both transcriptional and posttranscriptional control mechanisms.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Procesamiento Postranscripcional del ARN , Tromboplastina/genética , Transcripción Genética , Cicloheximida/farmacología , Humanos , Cinética , Leucemia Mieloide , ARN Mensajero/biosíntesis , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA