Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Thromb Haemost ; 15(11): 2208-2217, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28834179

RESUMEN

Essentials Tumor-bearing mice have larger venous clots than controls. Human tissue factor is present in clots in tumor-bearing mice. Inhibition of human tissue factor reduces clot size in tumor-bearing mice. This new mouse model may be useful to study mechanisms of cancer-associated thrombosis. SUMMARY: Background Pancreatic cancer patients have a high rate of venous thromboembolism. Human pancreatic tumors and cell lines express high levels of tissue factor (TF), and release TF-positive microvesicles (TF+ MVs). In pancreatic cancer patients, tumor-derived TF+ MVs are present in the blood, and increased levels are associated with venous thromboembolism and decreased survival. Previous studies have shown that mice with orthotopic human or murine pancreatic tumors have circulating tumor-derived TF+ MVs, an activated clotting system, and increased incidence and mean clot weight in an inferior vena cava stenosis model. These results suggest that TF+ MVs contribute to thrombosis. However, the specific role of tumor-derived TF+ MVs in venous thrombosis in mice has not been determined. Objectives To test the hypothesis that tumor-derived TF+ MVs enhance thrombosis in mice. Methods We determined the contribution of TF+ MVs derived from human pancreatic tumors grown orthotopically in nude mice to venous clot formation by using an anti-human TF mAb. We used an inferior vena cava stasis model of venous thrombosis. Results Tumor-bearing mice had significantly larger clots than control mice. Clots from tumor-bearing mice contained human TF, suggesting the incorporation of tumor-derived MVs. Importantly, administration of an anti-human TF mAb reduced clot size in tumor-bearing mice but did not affect clot size in control mice. Conclusions Our results indicate that TF+ MVs released from orthotopic pancreatic tumors increase venous thrombosis in mice. This new model may be useful for evaluating the roles of different factors in cancer-associated thrombosis.


Asunto(s)
Coagulación Sanguínea , Micropartículas Derivadas de Células/metabolismo , Neoplasias Pancreáticas/complicaciones , Tromboplastina/metabolismo , Trombosis de la Vena/etiología , Animales , Anticuerpos Monoclonales/farmacología , Plaquetas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Fibrinolíticos/farmacología , Xenoinjertos , Humanos , Masculino , Ratones Desnudos , Trasplante de Neoplasias , Neutrófilos/metabolismo , Neoplasias Pancreáticas/sangre , Tromboplastina/antagonistas & inhibidores , Trombosis de la Vena/sangre , Trombosis de la Vena/prevención & control
8.
J Thromb Haemost ; 15(9): 1829-1833, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28677246

RESUMEN

Essentials Factor XI (FXI) deficient mice have normal hemostasis in a tail transection model. The hemostatic capacity of FXI-/- mice was assessed in three different bleeding models. FXI-/- mice have increased saphenous vein bleeding. FXI-/- mice may be a useful experimental model to study bleeding associated with FXI deficiency. SUMMARY: Background Factor XI (FXI) is a key component of the intrinsic pathway of coagulation. It can be activated by either FXIIa or thrombin and amplifies thrombin generation during clot formation. Congenital FXI deficiency in humans (known as hemophilia C) is associated with bleeding after hemostatic challenge. However, to date there are no reports of excess bleeding in FXI-deficient mice. Objectives To determine if the absence of FXI in mice prolongs bleeding in different models. Methods We assessed the hemostatic capacity of FXI-/- mice in three different bleeding models: tail bleeding, surgical bleeding and saphenous vein bleeding. Results We found that tail bleeding and surgical bleeding of FXI-/- mice were similar to wild-type mice. However, FXI-/- mice had an impaired hemostatic capacity in the saphenous vein bleeding model compared with wild-type controls. Conclusions Our results indicate that FXI-/- mice have a mild hemostatic defect after injury to the saphenous vein but not after transection of the tail or vessels in the abdominal wall.


Asunto(s)
Pared Abdominal/irrigación sanguínea , Deficiencia del Factor XI/sangre , Factor XI/metabolismo , Hemorragia/sangre , Vena Safena/lesiones , Cola (estructura animal)/irrigación sanguínea , Lesiones del Sistema Vascular/sangre , Animales , Tiempo de Sangría , Modelos Animales de Enfermedad , Factor XI/genética , Deficiencia del Factor XI/complicaciones , Deficiencia del Factor XI/genética , Predisposición Genética a la Enfermedad , Hemorragia/etiología , Hemorragia/genética , Hemostasis/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Lesiones del Sistema Vascular/complicaciones , Lesiones del Sistema Vascular/genética
9.
J Thromb Haemost ; 15(8): 1625-1639, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28509332

RESUMEN

Essentials Tissue factor (TF) represents a central link between hemostasis and inflammation. We studied the roles of myeloid and airway epithelial TF in acid-caused acute lung injury (ALI). TF on myeloid cells displays a non-coagulatory role regulating the inflammatory response in ALI. Airway epithelial TF contributes to hemostatic functions, but is dispensable in ALI pathogenesis. SUMMARY: Introduction Acute lung injury (ALI) is a life-threatening condition characterized by damaged alveolar-capillary structures and activation of inflammatory and hemostatic processes. Tissue factor (TF) represents a crucial link between inflammation and coagulation, as inflammatory mediators induce myeloid TF expression, and TF initiates extrinsic coagulation. Objective As pulmonary inflammation stimulates TF expression and TF modulates immune responses, we aimed to elucidate its impact on ALI. In particular, we wanted to distinguish the contributions of TF expressed on airway epithelial cells and TF expressed on myeloid cells. Methods Mice with different cell type-specific TF deficiency and wild-type littermates were intratracheally treated with hydrochloric acid, and leukocyte recruitment, cytokine levels, thrombin-antithrombin (TAT) complexes and pulmonary protein-rich infiltrates were analyzed. Results Our data demonstrate that a lack of epithelial TF did not influence acute responses, as bronchoalveolar neutrophil accumulation 8 h after ALI induction was unaltered. However, it led to mild, prolonged inflammation, as pulmonary leukocyte and erythrocyte numbers were still increased after 24 h, whereas those in wild-type mice had returned to basal levels. In contrast, myeloid TF was primarily involved in regulating the acute phase of ALI without affecting local coagulation, as indicated by increased bronchoalveolar neutrophil infiltration, pulmonary interleukin-6 levels, and edema formation, but equal TAT complex formation, 8 h after ALI induction. This augmented inflammatory response associated with myeloid TF deficiency was confirmed in vitro, as lipopolysaccharide-stimulated TF-deficient alveolar macrophages released increased levels of chemokine (C-X-C motif) ligand 1 and tumor necrosis factor-α as compared with wild-type macrophages. Conclusion We conclude that myeloid TF dampens inflammation in acid-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Células Epiteliales/metabolismo , Ácido Clorhídrico , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Neumonía/prevención & control , Tromboplastina/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Antitrombina III/metabolismo , Coagulación Sanguínea , Células Cultivadas , Quimiotaxis de Leucocito , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Péptido Hidrolasas/metabolismo , Fenotipo , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/metabolismo , Edema Pulmonar/inducido químicamente , Edema Pulmonar/metabolismo , Edema Pulmonar/prevención & control , Tromboplastina/deficiencia , Tromboplastina/genética , Factores de Tiempo
10.
Thromb Res ; 140 Suppl 1: S169-70, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27161676

RESUMEN

INTRODUCTION: Cancer patients have a 4- to 7- fold increased risk of venous thromboembolism (VTE) compared with general population. Most tumor cells express tissue factor (TF) and constitutively release small membrane microvesicles called tumor microvesicles (TMVs). Clinical studies have shown that circulating MP-TF activity is associated with VTE in pancreatic cancer but not in other types of cancer. Thrombin is a potent platelet agonist and activates platelets via protease activated receptors (PARs). AIM: To determine the contribution of the TF+ TMV-thrombin-platelet pathway to cancer-associated thrombosis. MATERIALS AND METHODS: A human pancreatic adenocarcinoma cell line expressing high levels of TF (BxPc-3) was selected to study the effect of TF+ TMVs on platelet activation and thrombosis. RESULTS: TF+ TMVs induced platelet activation in vitro in a thrombin-dependent manner. The presence of orthotopically grown BxPc-3 tumors in mice was associated with increased levels of thrombin-antithrombin III complexes (TATc) and larger thrombi in an inferior vena cava stenosis model compared with control mice. Furthermore, injection of BxPc-3 TF+ TMVs into mice triggered platelet activation and enhanced venous thrombosis in a TF-dependent manner. Importantly, BxPc-3 TF+ TMV-enhanced thrombosis was reduced in Par4-deficient mice and wild-type mice treated with the platelet inhibitor clopidogrel, suggesting that platelet activation was required for the enhanced thrombosis. CONCLUSIONS: These studies suggest that platelet inhibitors may reduce thrombosis in cancer patients with elevated levels of TF+ TMVs.

11.
J Thromb Haemost ; 14(6): 1238-48, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26947929

RESUMEN

UNLABELLED: Essentials H1N1 Influenza A virus (IAV) infection is a hemostatic challenge for the lung. Tissue factor (TF) on lung epithelial cells maintains lung hemostasis after IAV infection. Reduced TF-dependent activation of coagulation leads to alveolar hemorrhage. Anticoagulation might increase the risk for hemorrhages into the lung during severe IAV infection. SUMMARY: Background Influenza A virus (IAV) infection is a common respiratory tract infection that causes considerable morbidity and mortality worldwide. Objective To investigate the effect of genetic deficiency of tissue factor (TF) in a mouse model of IAV infection. Methods Wild-type mice, low-TF (LTF) mice and mice with the TF gene deleted in different cell types were infected with a mouse-adapted A/Puerto Rico/8/34 H1N1 strain of IAV. TF expression was measured in the lungs, and bronchoalveolar lavage fluid (BALF) was collected to measure extracellular vesicle TF, activation of coagulation, alveolar hemorrhage, and inflammation. Results IAV infection of wild-type mice increased lung TF expression, activation of coagulation and inflammation in BALF, but also led to alveolar hemorrhage. LTF mice and mice with selective deficiency of TF in lung epithelial cells had low basal levels of TF and failed to increase TF expression after infection; these two strains of mice had more alveolar hemorrhage and death than controls. In contrast, deletion of TF in either myeloid cells or endothelial cells and hematopoietic cells did not increase alveolar hemorrhage or death after IAV infection. These results indicate that TF expression in the lung, particularly in epithelial cells, is required to maintain alveolar hemostasis after IAV infection. Conclusion Our study indicates that TF-dependent activation of coagulation is required to limit alveolar hemorrhage and death after IAV infection.


Asunto(s)
Células Epiteliales/virología , Hemorragia/virología , Infecciones por Orthomyxoviridae/patología , Alveolos Pulmonares/metabolismo , Tromboplastina/deficiencia , Animales , Anticoagulantes/uso terapéutico , Coagulación Sanguínea , Líquido del Lavado Bronquioalveolar , Eliminación de Gen , Hemostasis , Inflamación , Subtipo H1N1 del Virus de la Influenza A , Integrasas/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Tromboplastina/metabolismo
13.
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
14.
J Thromb Haemost ; 14(1): 153-66, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26516108

RESUMEN

UNLABELLED: ESSENTIALS: Cancer patients have a high rate of venous thrombosis (VT) but the underlying mechanisms are unknown. Tumor-derived, tissue factor-positive microvesicles in platelet activation in vitro and in vivo were studied. Tumor-derived, tissue factor-positive microvesicles enhanced VT in mice. Platelets may contribute to VT in some cancer patients, and this could be prevented with antiplatelet drugs. BACKGROUND: Cancer patients have an approximately 4-fold increased risk of venous thromboembolism (VTE) compared with the general population, and cancer patients with VTE have reduced survival. Tumor cells constitutively release small membrane vesicles called microvesicles (MVs) that may contribute to thrombosis in cancer patients. Clinical studies have shown that levels of circulating tumor-derived, tissue factor-positive (TF(+) ) MVs in pancreatic cancer patients are associated with VTE. Objectives We tested the hypothesis that TF(+) tumor-derived MVs (TMVs) activate platelets in vitro and in mice. MATERIALS AND METHODS: We selected two human pancreatic adenocarcinoma cell lines expressing high (BxPc-3) and low (L3.6pl) levels of TF as models to study the effect of TF(+) TMVs on platelets and thrombosis. RESULTS AND CONCLUSIONS: We found that both types of TF(+) TMVs activated human platelets and induced aggregation in vitro in a TF and thrombin-dependent manner. Further, injection of BxPc-3 TF(+) TMVs triggered platelet activation in vivo and enhanced thrombosis in two mouse models of venous thrombosis in a TF-dependent manner. Importantly, BxPc-3 TF(+) TMV-enhanced thrombosis was reduced in Par4-deficient mice and in wild-type mice treated with clopidogrel, suggesting that platelet activation was required for enhanced thrombosis. These studies suggest that TF(+) TMV-induced platelet activation contributes to thrombosis in cancer patients.


Asunto(s)
Micropartículas Derivadas de Células , Tromboplastina/fisiología , Trombosis/tratamiento farmacológico , Adenocarcinoma/fisiopatología , Animales , Plaquetas/citología , Línea Celular Tumoral , Clopidogrel , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Activación Plaquetaria , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Embolia Pulmonar/tratamiento farmacológico , Trombina/metabolismo , Ticlopidina/análogos & derivados , Ticlopidina/farmacología
15.
J Thromb Haemost ; 13(8): 1372-82, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25988873

RESUMEN

Cancer patients have a ~4 fold increased risk of venous thromboembolism (VTE) compared with the general population and this is associated with significant morbidity and mortality. This review summarizes our current knowledge of VTE and cancer, from mouse models to clinical studies. Notably, the risk of VTE varies depending on the type and stage of cancer. For instance, pancreatic and brain cancer patients have a higher risk of VTE than breast and prostate cancer patients. Moreover, patients with metastatic disease have a higher risk than those with localized tumors. Tumor-derived procoagulant factors and growth factors may directly and indirectly enhance VTE. For example, increased levels of circulating tumor-derived, tissue factor-positive microvesicles may trigger VTE. In a mouse model of ovarian cancer, tumor-derived IL-6 and hepatic thrombopoietin have been linked to increased platelet production and thrombosis. In addition, mouse models of mammary and lung cancer showed that tumor-derived granulocyte colony-stimulating factor causes neutrophilia and activation of neutrophils. Activated neutrophils can release neutrophil extracellular traps (NETs) that enhance thrombosis. Cell-free DNA in the blood derived from cancer cells, NETs and treatment with cytotoxic drugs can activate the clotting cascade. These studies suggest that there are multiple mechanisms for VTE in patients with different types of cancer. Preventing and treating VTE in cancer patients is challenging; the current recommendations are to use low-molecular-weight heparin. Understanding the underlying mechanisms may allow the development of new therapies to safely prevent VTE in cancer patients.


Asunto(s)
Coagulación Sanguínea , Neoplasias/complicaciones , Trombosis de la Vena/etiología , Animales , Anticoagulantes/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrinolíticos/uso terapéutico , Humanos , Ratones , Neoplasias/sangre , Neoplasias/patología , Pronóstico , Medición de Riesgo , Factores de Riesgo , Trombosis de la Vena/sangre , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/prevención & control
16.
J Vet Intern Med ; 29(3): 908-16, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25871966

RESUMEN

BACKGROUND: Studies of some human prothrombotic diseases suggest that phosphatidylserine-positive (PS+) and tissue factor-positive (TF+) microparticles (MPs) might play a role in the pathogenesis of thrombosis or serve as biomarkers of thrombotic risk. HYPOTHESIS/OBJECTIVES: To determine if circulating levels of PS+MP and procoagulant activity (PCA) associated with PS+MPs and TF+ MPs are increased in dogs with IMHA. ANIMALS: Fifteen dogs with primary or secondary IMHA and 17 clinically healthy dogs. METHODS: Prospective case-controlled observational study. Circulating PS+MPs were measured by flow cytometry. PCA associated with PS+MPs and TF+MPs was measured by thrombin and Factor Xa generating assays, respectively. RESULTS: Circulating numbers of PS+MPs were not significantly higher in dogs with IMHA [control median 251,000/µL (36,992-1,141,250/µL); IMHA median 361,990/µL (21,766-47,650,600/µL) P = .30]. However, PS+MP PCA [control median 2.2 (0.0-16.8) nM PS eq; IMHA median 8.596, (0-49.33 nM PS eq) P = .01] and TF+MP PCA [control median 0.0, (0.0-0.0 pg/mL); IMHA median 0.0; (0-22.34 pg/mL], P = .04) were increased. Intravascular hemolysis, which we showed might increase PS+ and TF+MP PCA, was evident in 3 of 5 dogs with PS+MP PCA and 2 of 4 dogs with TF+MP PCA higher than controls. Underlying disease in addition to IMHA was detected in 1 of 5 dogs with PS+PCA and 3 of 4 dogs with TF+MP PCA higher than controls. CONCLUSIONS AND CLINICAL IMPORTANCE: TF+ and PS+MP PCA is increased in some dogs with IMHA. Further studies that determine if measuring TF+ and PS+ MP PCA can help identify dogs at risk for thrombosis are warranted.


Asunto(s)
Anemia Hemolítica Autoinmune/veterinaria , Coagulantes/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Anemia Hemolítica Autoinmune/tratamiento farmacológico , Animales , Pruebas de Coagulación Sanguínea/veterinaria , Estudios de Casos y Controles , Coagulantes/administración & dosificación , Perros , Femenino , Citometría de Flujo , Hematócrito/veterinaria , Masculino , Memoria Episódica , Microesferas
17.
Hamostaseologie ; 35(1): 37-46, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25434707

RESUMEN

UNLABELLED: Tissue factor (TF) is expressed in the heart where it is required for haemostasis. High levels of TF are also expressed in atherosclerotic plaques and likely contribute to atherothrombosis after plaque rupture. Indeed, risk factors for atherothrombosis, such as diabetes, hypercholesterolaemia, smoking and hypertension, are associated with increased TF expression in circulating monocytes, microparticles and plasma. Several therapies that reduce atherothrombosis, such as statins, ACE inhibitors, beta-blockers and anti-platelet drugs, are associated with reduced TF expression. In addition to its haemostatic and pro-thrombotic functions, the TF : FVIIa complex and downstream coagulation proteases activate cells by cleavage of protease-activated receptors (PARs). In mice, deficiencies in either PAR-1 or PAR-2 reduce cardiac remodelling and heart failure after ischaemia-reperfusion injury. This suggests that inhibition of coagulation proteases and PARs may be protective in heart attack patients. In contrast, the TF/thrombin/PAR-1 pathway is beneficial in a mouse model of Coxsackievirus B3-induced viral myocarditis. We found that stimulation of PAR-1 increases the innate immune response by enhancing TLR3-dependent IFN-ß expression. Therefore, inhibition of the TF/thrombin/PAR-1 pathway in patients with viral myocarditis could have detrimental effects. CONCLUSION: The TF : FVIIa complex has both protective and pathological roles in the heart.


Asunto(s)
Coagulación Sanguínea/inmunología , Citocinas/inmunología , Factor VIII/inmunología , Hemostasis/inmunología , Tromboplastina/inmunología , Trombosis/inmunología , Animales , Humanos , Modelos Cardiovasculares , Modelos Inmunológicos
18.
J Thromb Haemost ; 13(3): 417-25, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25442192

RESUMEN

INTRODUCTION: Hemostasis is a rapid response by the body to stop bleeding at sites of vessel injury. Both platelets and fibrin are important for the formation of a hemostatic plug. Mice have been used to uncover the molecular mechanisms that regulate the activation of platelets and coagulation under physiologic conditions. However, measurements of hemostasis in mice are quite variable, and current methods do not quantify platelet adhesion or fibrin formation at the site of injury. METHODS: We describe a novel hemostasis model that uses intravital fluorescence microscopy to quantify platelet adhesion, fibrin formation and time to hemostatic plug formation in real time. Repeated vessel injuries of ~ 50-100 µm in diameter were induced with laser ablation technology in the saphenous vein of mice. RESULTS: Hemostasis in this model was strongly impaired in mice deficient in glycoprotein Ibα or talin-1, which are important regulators of platelet adhesiveness. In contrast, the time to hemostatic plug formation was only minimally affected in mice deficient in the extrinsic tissue factor (TF(low)) or the intrinsic factor IX coagulation pathways, even though platelet adhesion was significantly reduced. A partial reduction in platelet adhesiveness obtained with clopidogrel led to instability within the hemostatic plug, especially when combined with impaired coagulation in TF(low) mice. CONCLUSIONS: In summary, we present a novel, highly sensitive method to quantify hemostatic plug formation in mice. On the basis of its sensitivity to platelet adhesion defects and its real-time imaging capability, we propose this model as an ideal tool with which to study the efficacy and safety of antiplatelet agents.


Asunto(s)
Tiempo de Sangría , Plaquetas/metabolismo , Hemostasis , Vena Safena/metabolismo , Lesiones del Sistema Vascular/sangre , Animales , Coagulación Sanguínea , Plaquetas/efectos de los fármacos , Clopidogrel , Modelos Animales de Enfermedad , Factor IX/genética , Factor IX/metabolismo , Fibrina/metabolismo , Hemostasis/genética , Microscopía Intravital , Terapia por Láser , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Microscopía por Video , Adhesividad Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Vena Safena/cirugía , Talina/deficiencia , Talina/genética , Tromboplastina/deficiencia , Tromboplastina/genética , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Factores de Tiempo , Lesiones del Sistema Vascular/etiología , Lesiones del Sistema Vascular/genética
19.
J Thromb Haemost ; 12(12): 2065-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25267332

RESUMEN

BACKGROUND: Immune responses to therapeutic factor VIII remain a major problem, affecting 30% of patients with severe hemophilia A. The primary factors that drive immune responses in these patients remain elusive. There have been conflicting reports on a role of coagulation (or thrombin) in anti-FVIII immune responses. OBJECTIVE: To assess the importance of coagulation-associated processes for the onset of the anti-FVIII immune response. METHODS: Using FVIII-deficient mice, we compared the immunogenicity of recombinant FVIII or the inactive FVIII(V) (634M) mutant. In parallel, the involvement of tissue factor (TF) activity in the anti-FVIII immune response was investigated upon injection of a neutralizing anti-TF antibody or by the use of chimeric mice that lack TF expression in myeloid cells. The development of the anti-FVIII immune response was also monitored after treatment with warfarin. RESULTS: The kinetics of the development of antibody responses to FVIII(V) (634M) were indistinguishable from those of wild-type FVIII. Inhibition of TF activity did not modulate immune responses to exogenous FVIII. Additionally, global inhibition of coagulation with warfarin failed to reduce the anti-FVIII immune response. CONCLUSIONS: Thrombin generation or coagulation-associated processes do not modulate the anti-FVIII antibody response in mouse model of severe hemophilia A.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/sangre , Inmunidad Humoral , Animales , Anticuerpos Neutralizantes/inmunología , Coagulación Sanguínea , Modelos Animales de Enfermedad , Hemofilia A/genética , Inflamación , Ratones , Mutación , Plásmidos , Estructura Terciaria de Proteína , Proteínas Recombinantes/inmunología , Trombina/química , Tromboplastina/química , Warfarina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA