Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
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
2.
Thromb Res ; 140 Suppl 1: S171, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27161680

RESUMEN

INTRODUCTION: Severe thrombocytopenia (≤50×10(9) platelets/L) is often the consequence of hematological malignancies and intensive chemotherapy. The risk of clinically significant bleeding is increased in these patients, despite the use of prophylactic platelet transfusions. The fact that there is no clear correlation between the platelet count and the risk of hemorrhage, suggests that there are other contributing factors. The contribution of impairments in platelet and coagulant function remains poorly understood. AIM: In patients with chemotherapy-induced thrombocytopenia due to hematological malignancies, we evaluate platelet and coagulant functions and determine the effects of platelet transfusion. Ultimately, we can identify specific hemostatic factors that aid in the prediction of bleeding. MATERIALS AND METHODS: In total 58 patients were included and blood was collected before and, if indicated (≤10×10(9) platelets/L), 1 hour after transfusion with platelet concentrate. Platelet function was assessed using flow cytometry by determining: 1) integrin αIIbß3 activation (PAC-1 antibody), 2) P-selectin expression (anti-P-selectin antibody), 3) phosphatidylserine exposure (Annexin-V) and 4) intracellular calcium (Fluo-4 AM). Factor levels were determined in plasma. Thrombus and fibrin formation was assessed by perfusion of whole blood over a collagen-tissue factor surface at a shear rate of 1,000 s-1. RESULTS: Platelets from the thrombocytopenic patients before transfusion showed markedly reduced integrin αIIbß3 activation and P-selectin expression in response to thrombin, collagen-related peptide and ADP, compared to healthy donor platelets. Also, agonist-induced intracellular calcium fluxes were greatly reduced. However, calcium fluxes with thapsigargin, a SERCA pump inhibitor, were similar in patient and control platelets, suggesting a normal calcium store content in the patient platelets. Furthermore, phosphatidylserine exposure was increased in unstimulated patient platelets compared to control platelets (8.2 vs. 1.8%, p<0.0001). Coagulation factor levels were within the normal range, with the exception of von Willebrand factor and fibrinogen levels, which were elevated. Platelet transfusion partly recovered the platelet integrin αIIbß3 activation and P-selectin expression induced by agonists. Platelet deposition (6.7 vs. 1.7%, p<0.0001) and fibrin formation (7.6 vs. 0.9%, p=0.0005) under flow conditions were substantially improved after platelet transfusion. CONCLUSIONS: Platelets from cancer patients undergoing chemotherapy appear to display impaired functional responses to activating stimuli. Platelet transfusion partly restores these functional defects, resulting in improved thrombus and fibrin formation.

3.
Thromb Res ; 140 Suppl 1: S183, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27161708

RESUMEN

INTRODUCTION: The myeloproliferative neoplasms ET and PV are characterized by a high incidence of both arterial and venous thrombosis, and/or microcirculatory disturbances. Three somatic mutations, i.e. JAK2-V617F, Calreticulin (CalR) and MPL, commonly found in these diseases, correlate with different thrombotic risk levels. AIM: To analyze the influence of JAK2-V617F, CalR and MPL mutations on PLT adhesion, evaluated by a dynamic method under flow conditions in a group of patients with ET and PV. MATERIALS AND METHODS: 86 patients, i.e. 51 ET (19 M/32 F; age range 32-86 years) and 35PV (22 M/13 F; 41-83 yrs.), and 24 healthy controls (13 M/11 F; 28-61 yrs.) were enrolled upon informed consent. For the adhesion assay, peripheral venous whole blood was perfused over collagen for 4' at a 1,000 s-1 shear rate. PLTs were then stained with an anti-P-selectin-FITC antibody to evaluate PLT activation, and annexin V-AlexaFluor647 to detect procoagulant phosphatidylserine expression. Then, images of adherent PLTs in random fields were taken using phase contrast and fluorescence imaging by EVOS® fluorescence microscope. Results are mean±SEM of the % area covered by PLTs, or as the % of adherent PLTs positive for P-selectin or phosphatidylserine. Main hematological parameters and mutational status were recorded. RESULTS: PLT adhesion was significantly (p<0.01) greater in ET (44.6±1.6%) and PV patients (49.0±1.9%) compared to controls (37.9±1.7%). In ET, PLT adhesion was highest in JAK2-V617F mutation carriers (n=23), followed by CalR-positive (n=16) and triple negative subjects (n=9), and lowest in the MPL-positive patients (n=3). In PV, no difference in PLT adhesion was observed between JAK2-V617F heterozygous and homozygous subjects. P-selectin expression by adherent PLTs was not statistically different between patients and controls. Differently, phosphatidylserine expression on adherent PLTs was significantly reduced (p<0.01) in both ET and PV compared to healthy subjects. In ET patients, a significant (p<0.05) correlation was found between PLT adhesion and PLT count in JAK2-V617F and CalR-positive mutation carriers. Multivariate regression analysis adjusted for age and sex, confirmed PLT count as a significant determinant of PLT adhesion in JAK2-V617F positive patients only. CONCLUSIONS: ET and PV platelets show an increased adhesion to collagen in vitro, particularly in those carrying the JAK2-V617F mutation. A prospective study is ongoing to evaluate the predictive value of our PLT thrombus formation dynamic model for the thrombotic risk in ET and PV patients. ACKNOWLEDGEMENT: Project funded by "AIRC-IG2013" grant Nr. 14505 from the "Italian Association for Cancer Research" (A.I.R.C.).

4.
J Thromb Haemost ; 13(8): 1503-13, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25988848

RESUMEN

BACKGROUND: Patients undergoing major cardiothoracic surgery are subjected to dilution, owing to massive fluid infusion and blood component transfusion. These patients may experience bleeding perioperatively, and are frequently treated with the endothelium-activating agent desmopressin. OBJECTIVES: To investigate the effect of desmopressin administration on von Willebrand factor (VWF)-dependent coagulant and platelet functions under flow conditions. PATIENTS/METHODS: Blood from 16 patients with postoperative bleeding was obtained before and after desmopressin treatment (0.3 µg kg(-1) body weight), and assessed for coagulant properties and platelet function. Furthermore, VWF antigen levels and multimer composition were determined in both samples. RESULTS: Desmopressin treatment did not change thrombin generation in plasma or whole blood thromboelasticity. Also coagulation factor levels (other than factor VIII) and coagulation times were unchanged, suggesting that desmopressin treatment did not have a major effect on the coagulant activity. On the other hand, desmopressin treatment raised the already high plasma levels of VWF from a median of 116 IU mL(-1) (interquartile range [IQR] 102-154 IU mL(-1) ) to a median of 160 IU mL(-1) (IQR 126-187 IU mL(-1) ) (P = 0.007), owing to accumulation of the high molecular weight VWF multimers. Furthermore, desmopressin treatment caused an increase in collagen-dependent thrombus formation and platelet phosphatidylserine exposure. Markers of thrombus formation correlated with the plasma levels of VWF. In vitro control experiments confirmed a major contribution of VWF to thrombus formation and procoagulant activity under conditions of blood dilution. CONCLUSIONS: Desmopressin treatment of patients with bleeding complications after cardiothoracic surgery induces the release of high molecular weight VWF multimers, which enhance platelet activation and thrombus formation under flow conditions.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Desamino Arginina Vasopresina/uso terapéutico , Hemostáticos/uso terapéutico , Hemorragia Posoperatoria/tratamiento farmacológico , Anciano , Pruebas de Coagulación Sanguínea , Plaquetas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosfatidilserinas/sangre , Activación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Hemorragia Posoperatoria/sangre , Hemorragia Posoperatoria/etiología , Resultado del Tratamiento , Factor de von Willebrand/metabolismo
5.
Thromb Haemost ; 111(3): 447-57, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24258426

RESUMEN

Blood dilution after transfusion fluids leads to diminished coagulant activity monitored by rotational thromboelastometry, assessing elastic fibrin clot formation, or by thrombin generation testing. We aimed to determine the contributions of blood cells (platelets, red blood cells) and plasma factors (fibrinogen, prothrombin complex concentrate) to fibrin clot formation under conditions of haemodilution in vitro or in vivo.Whole blood or plasma diluted in vitro was supplemented with platelets, red cells, fibrinogen or prothrombin complex concentrate (PCC). Thromboelastometry was measured in whole blood as well as plasma; thrombin generation was determined in parallel. Similar tests were performed with blood from 48 patients, obtained before and after massive fluid infusion during cardiothoracic surgery.Addition of platelets or fibrinogen, in additive and independent ways, reversed the impaired fibrin clot formation (thromboelastometry) in diluted whole blood. In contrast, supplementation of red blood cells or prothrombin complex concentrate was ineffective. Platelets and fibrinogen independently restored clot formation in diluted plasma, resulting in thromboelastometry curves approaching those in whole blood. In whole blood from patients undergoing dilution during surgery, elastic clot formation was determined by both the platelet count and the fibrinogen level. Thrombin generation in diluted (patient) plasma was not changed by fibrinogen, but improved markedly by prothrombin complex concentrate. In conclusion, in dilutional coagulopathy, platelets and fibrinogen, but not red blood cells or vitamin K-dependent coagulation factors, independently determine thromboelastometry parameters measured in whole blood and plasma. Clinical decisions for transfusion based on thromboelastometry should take into account the platelet concentration.


Asunto(s)
Plaquetas/patología , Puente Cardiopulmonar , Fibrina/metabolismo , Fibrinógeno/metabolismo , Hemorragia/prevención & control , Anciano , Coagulación Sanguínea , Plaquetas/metabolismo , Eritrocitos/patología , Femenino , Hemodilución/efectos adversos , Hemorragia/etiología , Humanos , Masculino , Persona de Mediana Edad , Recuento de Plaquetas , Protrombina/metabolismo , Tromboelastografía , Trombina/metabolismo , Reacción a la Transfusión
6.
J Urol ; 148(5): 1577-82, 1992 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1433571

RESUMEN

At present, isoniazid (INH) is being used prophylactically to reduce the side effects of intravesical BCG therapy for superficial bladder cancer, although it is not clear whether or not this reduces the antitumor efficacy of BCG. In this study the impact of INH treatment on the immune response after repeated intravesical BCG administration was investigated in guinea pigs. INH was given on the 3 days around each BCG instillation. We found that the administration of INH severely impaired the immunological effects of BCG. The induction of mononuclear cell infiltration in the bladder wall was reduced. Enlargement of the regional lymph nodes (weight and number of cells), and increase of MHC Class II expression on the lymph node cells, normally observed after intravesical BCG administration, were inhibited by INH. Systemic immunity, measured by the DTH reaction in the skin to PPD, was also diminished due to the combined treatment of BCG with INH. When INH was administered during the last 4 of 6 BCG instillations, the immune response to BCG was still impaired. A five-fold increase of the dose of BCG did not overcome the effect of INH. INH probably did not exert a direct suppression of the immune system of the guinea pig as the DNCB skin reactivity was not influenced. Although INH concentrations in the urine were high at the onset of the instillation, in vitro experiments indicated that the effect of INH may not be caused by killing of the BCG organisms shortly after application in the bladder. In conclusion, our data in guinea pigs suggest that the use of INH may impair the immune response to intravesical BCG. As this response may be important for the antitumor effect of BCG, urologists should be cautious with the prophylactic use of INH. The influence on the antitumor efficacy is now investigated in man.


Asunto(s)
Vacuna BCG/inmunología , Isoniazida/farmacología , Administración Intravesical , Animales , Vacuna BCG/administración & dosificación , Recuento de Colonia Microbiana , Femenino , Cobayas , Antígenos de Histocompatibilidad Clase II/análisis , Hipersensibilidad Tardía , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Linfocitos/patología , Mycobacterium bovis/efectos de los fármacos , Pruebas Cutáneas , Vejiga Urinaria/patología
7.
J Urol ; 147(3): 596-600, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1538436

RESUMEN

Intravesical therapy with bacillus Calmette-Guerin (BCG) has proved to be more effective in the prophylaxis and treatment of superficial bladder tumors and carcinoma in situ than most chemotherapeutic agents. Compared to intravesical chemotherapy, instillations with BCG provoke more local and systemic reactions. In addition to the commonly induced granulomatous inflammatory changes in the bladder, which produce irritative symptoms, this therapy may cause systemic side effects varying from mild malaise and fever to, in rare instances, life-threatening or fatal sepsis. We report the incidence and varieties of toxicities in 2,602 patients treated with intravesical BCG. Side effects are classified according to local and systemic toxicity. Treatment options vary according to the severity of toxicity from delaying or withholding instillations to treatment with antituberculous drugs for up to 6 months. In general, 95% of the patients have no serious side effects. Recognition of risk factors, particularly traumatic catheterization or concurrent cystitis, that result in systemic BCG absorption, as well as the prompt and appropriate treatment of early side effects should significantly decrease the incidence of severe toxicity.


Asunto(s)
Vacuna BCG/efectos adversos , Neoplasias de la Vejiga Urinaria/terapia , Administración Intravesical , Vacuna BCG/administración & dosificación , Vacuna BCG/uso terapéutico , Contractura , Cistitis/epidemiología , Cistitis/etiología , Humanos , Incidencia , Estadificación de Neoplasias , Tuberculosis/tratamiento farmacológico , Tuberculosis/epidemiología , Tuberculosis/etiología , Obstrucción Ureteral/epidemiología , Obstrucción Ureteral/etiología , Enfermedades de la Vejiga Urinaria/epidemiología , Enfermedades de la Vejiga Urinaria/etiología , Neoplasias de la Vejiga Urinaria/patología
9.
J Urol ; 146(2): 444-6, 1991 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1856950

RESUMEN

Intravesical treatment with Bacillus Calmette-Guérin is an established treatment of carcinoma in situ and an effective prophylaxis for the prevention of recurrence of transitional cell carcinoma. During instillation therapy oral antibiotics may be used to prevent or to treat urinary tract infections. Tuberculostatic agents are employed to prevent or to treat local irritative symptoms and systemic side effects caused by Bacillus Calmette-Guérin. We investigated the susceptibility of four different Bacillus Calmette-Guérin preparations to 18 antibiotics and to 11 tuberculostatic agents in vitro. All preparations were equally susceptible to most of the commonly used antibiotics and to all of the tuberculostatic agents with the exception of pyrazinamide. Our results suggest that in the absence of cystitis, instillations should not be accompanied by antibiotics because of the possible inhibition of antitumor efficacy by eradication of living Bacillus Calmette-Guérin organisms. The same inhibition may occur when tuberculostatic agents are used to prevent local and systemic complications of Bacillus Calmette-Guérin instillation therapy.


Asunto(s)
Antibacterianos/farmacología , Vacuna BCG/farmacología , Carcinoma in Situ/terapia , Neoplasias de la Vejiga Urinaria/terapia , Administración Intravesical , Antituberculosos/farmacología , Vacuna BCG/efectos adversos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Técnicas In Vitro , Pruebas de Sensibilidad Microbiana , Mycobacterium bovis/efectos de los fármacos
11.
Biochem Biophys Res Commun ; 118(3): 760-6, 1984 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-6367742

RESUMEN

Methanol: 5-hydroxybenzimidazolylcobamide methyltransferase (MT1) from Methanosarcina barkeri, which is one of the enzymes responsible for the transmethylation from methanol to coenzyme M, was found to be activated in the presence of hydrogenase and ferredoxin. This activation was shown to involve a reduction of the bound corrinoid to the Co (I) level, and was demonstrated by spectrophotometry and chemical conversion of reduced MT1 to its methylated form. The reducing system of hydrogenase and ferredoxin was able to reduce dithiols, like dithiodiethanesulfonate and cystine to their monomers, in the presence of a corrinoid, which acts as an electron carrier. The ferredoxin was purified 133-fold and was tentatively identified on the basis of spectral properties and iron content of 3.8-4.0 atoms iron per molecule ferredoxin (12,000 daltons).


Asunto(s)
Euryarchaeota/enzimología , Metiltransferasas/metabolismo , Corrinoides , Activación Enzimática/efectos de los fármacos , Ferredoxinas/farmacología , Hidrogenasas , Metilación , Oxidación-Reducción , Oxidorreductasas/farmacología , Espectrofotometría , Vitamina B 12/farmacología
12.
J Bacteriol ; 153(1): 6-11, 1983 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-6294063

RESUMEN

Methanol is converted to methane by crude extracts of Methanosarcina barkeri. The first reaction involved in this process, is catalyzed by methanol:2-mercaptoethanesulfonic acid methyltransferase (EC 2.1.1.-). The methyltransferase has an optimum at pH 6.5 and is not inhibited by 2-bromoethanesulfonic acid. Pyridoxal-5'-phosphate acts as an inhibitor (Ki = 0.30 mM). The methyltransferase was tested in the presence of 2-bromoethanesulfonic acid, which inhibits the conversion of 2-(methylthio)ethanesulfonic acid to methane. The reaction is subject to activation and inactivation. Inactivation is brought about by the presence of oxygen, flavin mononucleotide, flavin adenine dinucleotide, and 2-(methylthio)ethanesulfonic acid, the product of the reaction. Activation of the system requires the presence of ATP and Mg2+ and of hydrogen. Hydrogen can be replaced by enzymatic systems, such as pyruvate dehydrogenase, which deliver free hydrogen.


Asunto(s)
Ácidos Alcanesulfónicos , Euryarchaeota/enzimología , Metiltransferasas/metabolismo , Adenosina Trifosfato/farmacología , Alcanosulfonatos/farmacología , Activación Enzimática , Hidrógeno/farmacología , Concentración de Iones de Hidrógeno , Magnesio/farmacología , Metanol/metabolismo , Oxígeno/farmacología , Fosfato de Piridoxal/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA