Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Br J Haematol ; 192(5): 909-921, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33528045

RESUMEN

Lowe syndrome (LS) is an oculocerebrorenal syndrome of Lowe (OCRL1) genetic disorder resulting in a defect of the OCRL protein, a phosphatidylinositol-4,5-bisphosphate 5-phosphatase containing various domains including a Rho GTPase-activating protein (RhoGAP) homology domain catalytically inactive. We previously reported surgery-associated bleeding in patients with LS, suggestive of platelet dysfunction, accompanied with a mild thrombocytopenia in several patients. To decipher the role of OCRL in platelet functions and in megakaryocyte (MK) maturation, we conducted a case-control study on 15 patients with LS (NCT01314560). While all had a drastically reduced expression of OCRL, this deficiency did not affect platelet aggregability, but resulted in delayed thrombus formation on collagen under flow conditions, defective platelet spreading on fibrinogen and impaired clot retraction. We evidenced alterations of the myosin light chain phosphorylation (P-MLC), with defective Rac1 activity and, inversely, elevated active RhoA. Altered cytoskeleton dynamics was also observed in cultured patient MKs showing deficient proplatelet extension with increased P-MLC that was confirmed using control MKs transfected with OCRL-specific small interfering(si)RNA (siOCRL). Patients with LS also had an increased proportion of circulating barbell-shaped proplatelets. Our present study establishes that a deficiency of the OCRL protein results in a defective actomyosin cytoskeleton reorganisation in both MKs and platelets, altering both thrombopoiesis and some platelet responses to activation necessary to ensure haemostasis.


Asunto(s)
Plaquetas/citología , Megacariocitos/citología , Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolasas/fisiología , Trombopoyesis/fisiología , Actomiosina/análisis , Adolescente , Adulto , Anemia/etiología , Coagulación Sanguínea , Plaquetas/ultraestructura , Estudios de Casos y Controles , Forma de la Célula , Niño , Colágeno , Citoesqueleto/ultraestructura , Femenino , Silenciador del Gen , Humanos , Masculino , Megacariocitos/ultraestructura , Persona de Mediana Edad , Mutación , Cadenas Ligeras de Miosina/metabolismo , Síndrome Oculocerebrorrenal/sangre , Síndrome Oculocerebrorrenal/patología , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , Dominios Proteicos , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/genética , Transducción de Señal , Trombocitopenia/etiología , Adulto Joven
2.
Eur J Anaesthesiol ; 36(6): 449-456, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30308522

RESUMEN

BACKGROUND: Rapid detection of the anticoagulant effect of oral factor Xa (FXa) inhibitors may be essential in several emergency clinical situations. Specific assays quantifying the drugs are performed in plasma and require a turnaround time that is too long to be useful in emergency situations. Rotational thromboelastometry (ROTEM) is a whole blood coagulation assay of blood viscoelasticity and could be of interest for FXa inhibitor detection in emergency. However, conventional ROTEM reagents only detect high amounts of inhibitors. OBJECTIVE: The aim of this study was first to assess the effect of whole blood components on the viscoelastic measurement of the effects of FXa inhibitors, and second to evaluate whether a modified ROTEM, triggered with a low amount of tissue factor and a saturating amount of phospholipid vesicles, can reliably detect low levels of FXa inhibitor activity in whole blood. DESIGN: Diagnostic test study. SETTINGS: A university research laboratory. From November 2014 to April 2016. PATIENTS: Sixty-six patients: 30 treated with rivaroxaban, 17 with apixaban and 19 without treatment. INTERVENTION: ROTEM was triggered with 2.5 pmol l of tissue factor and 10 µmol l of phospholipid vesicles. MAIN OUTCOME MEASURES: Modified ROTEM parameters were measured in different experimental conditions: platelet-poor plasma (PPP), platelet-rich plasma, PPP supplemented with fibrinogen and reconstituted whole blood with various haematocrit levels adjusted between 30 and 60%. Modified ROTEM was further validated using whole blood from patients who were either treated or not treated with FXa inhibitors. RESULTS: Modified ROTEM allowed detection of as little as 25 ng ml FXa inhibitors in PPP, with at least a 1.4-fold increase of the clotting time (P ≤ 0.02). Neither changes of fibrinogen concentration nor variations of platelet count or haematocrit precluded FXa inhibitor detection. A lengthened modified ROTEM clotting time of more than 197 s allowed detection of FXa inhibitor concentrations above 30 ng ml in whole blood with 90% sensitivity and 85% specificity. CONCLUSION: Modified ROTEM may be applicable in emergency situations for the detection of FXa inhibitors in whole blood.


Asunto(s)
Inhibidores del Factor Xa/sangre , Tromboelastografía/métodos , Administración Oral , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Coagulación Sanguínea/efectos de los fármacos , Cuidados Críticos/métodos , Inhibidores del Factor Xa/administración & dosificación , Inhibidores del Factor Xa/farmacocinética , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pirazoles/administración & dosificación , Pirazoles/sangre , Pirazoles/farmacocinética , Piridonas/administración & dosificación , Piridonas/sangre , Piridonas/farmacocinética , Rivaroxabán/administración & dosificación , Rivaroxabán/sangre , Rivaroxabán/farmacocinética , Sensibilidad y Especificidad , Factores de Tiempo , Adulto Joven
3.
Mar Drugs ; 13(12): 7446-62, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26694425

RESUMEN

Low-molecular-weight fucoidan (LMWF) is a sulfated polysaccharide extracted from brown seaweed that presents antithrombotic and pro-angiogenic properties. However, its mechanism of action is not well-characterized. Here, we studied the effects of LMWF on cell signaling and whole genome expression in human umbilical vein endothelial cells and endothelial colony forming cells. We observed that LMWF and vascular endothelial growth factor had synergistic effects on cell signaling, and more interestingly that LMWF by itself, in the absence of other growth factors, was able to trigger the activation of the PI3K/AKT pathway, which plays a crucial role in angiogenesis and vasculogenesis. We also observed that the effects of LMWF on cell migration were PI3K/AKT-dependent and that LMWF modulated the expression of genes involved at different levels of the neovessel formation process, such as cell migration and cytoskeleton organization, cell mobilization and homing. This provides a better understanding of LMWF's mechanism of action and confirms that it could be an interesting therapeutic approach for vascular repair.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Polisacáridos/farmacología , Citoesqueleto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peso Molecular , Neovascularización Fisiológica/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Polisacáridos/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Am J Respir Crit Care Med ; 188(6): 684-92, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23924269

RESUMEN

RATIONALE: Purpura fulminans in adults is a rare but devastating disease. Its pathophysiology is not well known. OBJECTIVES: To understand the pathophysiology of skin lesions in purpura fulminans, the interplay between circulating blood and vascular alterations was assessed. METHODS: Prospective multicenter study in four intensive care units. Patients with severe sepsis without skin lesions were recruited as control subjects. MEASUREMENTS AND MAIN RESULTS: Twenty patients with severe sepsis and purpura fulminans were recruited for blood sampling, and skin biopsy was performed in deceased patients. High severity of disease and mortality rates (80%) was observed. Skin biopsies in purpura fulminans lesions revealed thrombosis and extensive vascular damage: vascular congestion and dilation, endothelial necrosis, alteration of markers of endothelial integrity (CD31) and of the protein C pathway receptors (endothelial protein C receptor, thrombomodulin). Elevated plasminogen activating inhibitor-1 mRNA was also observed. Comparison with control patients showed that these lesions were specific to purpura fulminans. By contrast, no difference was observed for blood hemostasis parameters, including soluble thrombomodulin, activated protein C, and disseminated intravascular coagulation markers. Bacterial presence at the vascular wall was observed specifically in areas of vascular damage in eight of nine patients tested (including patients with Streptococcus pneumoniae, Neisseria meningitidis, Escherichia coli, and Pseudomonas aeruginosa infection). CONCLUSIONS: Thrombi and extensive vascular damage with multifaceted prothrombotic local imbalance are characteristics of purpura fulminans. A "vascular wall infection" hypothesis, responsible for endothelial damage and subsequent skin lesions, can be put forward.


Asunto(s)
Endotelio Vascular/patología , Púrpura Fulminante/patología , Trombosis/complicaciones , Malformaciones Vasculares/complicaciones , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Biopsia , Endotelio Vascular/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidor 1 de Activador Plasminogénico/metabolismo , Estudios Prospectivos , Púrpura Fulminante/complicaciones , Púrpura Fulminante/metabolismo , Sepsis/metabolismo , Piel/irrigación sanguínea , Trombomodulina/metabolismo , Trombosis/patología , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología
5.
J Thromb Haemost ; 20(7): 1653-1664, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35445541

RESUMEN

BACKGROUND: Protein S (PS) is a natural anticoagulant acting as a cofactor for activated protein C (APC) in the proteolytic inactivation of activated factors V (FVa) and VIII (FVIIIa), but also for tissue factor pathway inhibitor α (TFPIα) in the inhibition of activated factor X (FXa). OBJECTIVE: For therapeutic purposes, we aimed at generating single-domain antibodies (sdAbs) that could specifically modulate the APC-cofactor activity of PS in vivo. METHODS: A llama-derived immune library of sdAbs was generated and screened on recombinant human PS by phage display. PS binders were tested in a global activated partial thromboplastin time (APTT)-based APC-cofactor activity assay. RESULTS: A PS-specific sdAb (PS003) was found to enhance the APC-cofactor activity of PS in our APTT-based assay, and this enhancing effect was greater for a bivalent form of PS003 (PS003biv). Further characterization of PS003biv demonstrated that PS003biv also enhanced the APC-cofactor activity of PS in a tissue factor (TF)-induced thrombin generation assay and stimulated APC in the inactivation of FVa, but not FVIIIa, in plasma-based assays. Furthermore, PS003biv was directed against the sex hormone-binding globulin (SHBG)-like domain but did not inhibit the binding of PS to C4b-binding protein (C4BP) and did not interfere with the TFPIα-cofactor activity of PS. In mice, PS003biv exerted an antithrombotic effect in a FeCl3 -induced thrombosis model, while not affecting physiological hemostasis in a tail-clip bleeding model. DISCUSSION: Altogether, these results showed that pharmacological enhancement of the APC-cofactor activity of PS through an original anti-PS sdAb might constitute a promising and safe antithrombotic strategy.


Asunto(s)
Proteína S , Anticuerpos de Dominio Único , Animales , Factor VIIIa/química , Fibrinolíticos/farmacología , Humanos , Ratones , Proteína C/metabolismo , Proteína S/metabolismo
6.
Int J Biol Macromol ; 184: 209-217, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34126147

RESUMEN

Alpha2-macroglobulin (α2M) is a physiological macromolecule that facilitates the clearance of many proteinases, cytokines and growth factors in human. Here, we explored the effect of induced forms of α2M on anticoagulant drugs. Gla-domainless factor Xa (GDFXa) and methylamine (MA)-induced α2M were prepared and characterized by electrophoresis, immunonephelometry, chromogenic, clot waveform and rotational thromboelastometry assays. Samples from healthy volunteers and anticoagulated patients were included. In vivo neutralization of anticoagulants was evaluated in C57Bl/6JRj mouse bleeding-model. Anticoagulant binding sites on induced α2M were depicted by computer-aided energy minimization modeling. GDFXa-induced α2M neutralized dabigatran and heparins in plasma and whole blood. In mice, a single IV dose of GDFXa-induced α2M following anticoagulant administration significantly reduced blood loss and bleeding time. Being far easier to prepare, we investigated the efficacy of MA-induced α2M. It neutralized rivaroxaban, apixaban, dabigatran and heparins in spiked samples in a concentration-dependent manner and in samples from treated patients. Molecular docking analysis evidenced the ability of MA-induced α2M to bind non-covalently these compounds via some deeply buried binding sites. Induced forms of α2M have the potential to neutralize direct oral anticoagulants and heparins, and might be developed as a universal antidote in case of major bleeding or urgent surgery.


Asunto(s)
Inhibidores del Factor Xa/efectos adversos , Factor Xa/química , Hemorragia/tratamiento farmacológico , Heparina/efectos adversos , alfa 2-Macroglobulinas Asociadas al Embarazo/administración & dosificación , Administración Oral , Animales , Modelos Animales de Enfermedad , Femenino , Voluntarios Sanos , Hemorragia/inducido químicamente , Humanos , Metilaminas/farmacología , Ratones , Simulación del Acoplamiento Molecular , Embarazo , alfa 2-Macroglobulinas Asociadas al Embarazo/química , alfa 2-Macroglobulinas Asociadas al Embarazo/farmacología , Dominios Proteicos
7.
Pediatr Cardiol ; 31(5): 657-62, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20195855

RESUMEN

The reversibility of pulmonary arterial hypertension (PAH) in children with congenital heart disease (CHD) is strongly associated with the degree of intimal proliferation, vessel narrowing, and number of circulating endothelial cells (CECs). Circulating endothelial cells may arise from either endothelial damage or accelerated turnover during vessel remodeling, but nothing is known about endothelial microparticles (EMPs) and other biomarkers reflecting endothelial alterations. This study aimed to document endothelial markers further according to the irreversibility of PAH secondary to CHD. The study investigated soluble markers of endothelial damage or activation (thrombomodulin, soluble endothelial protein C receptor, and soluble E-selectin), inflammation (interleukin-6), and angiogenic cytokine levels [vascular endothelial growth factor (VEGF) and placental growth factor (PlGF)] in 26 patients with CHD, 16 with reversible PAH (median age, 2 years) and 10 with irreversible PAH (median age, 9 years). Endothelial activation/apoptosis was evaluated by measuring EMP levels. Plasma procoagulant activity also was measured. The results show that the levels of soluble markers indicating endothelial activation were not predictors of PAH irreversibility. Lower levels of PlGF were observed in reversible compared with irreversible PAH but were not associated with the CEC level, the mean pulmonary artery pressure (mPAP), or age. No significant difference in procoagulant activity or EMP level was found between irreversible and reversible PAH. Among a large panel of biomarkers reflecting endothelial activation, regeneration, and injury, the high CEC levels previously described proved to be the only marker allowing discrimination between reversible and irreversible PAH secondary to CHD.


Asunto(s)
Biomarcadores/sangre , Endotelio Vascular/metabolismo , Cardiopatías Congénitas/complicaciones , Hipertensión Pulmonar/etiología , Factores de Edad , Niño , Preescolar , Femenino , Cardiopatías Congénitas/sangre , Cardiopatías Congénitas/fisiopatología , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/fisiopatología , Lactante , Masculino , Estudios Retrospectivos , Estadísticas no Paramétricas , Resistencia Vascular/fisiología
8.
Thromb Haemost ; 120(9): 1270-1281, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32717757

RESUMEN

Inherited protein C (PC) deficiency caused by mutations in the PROC gene is a well-known risk factor for venous thromboembolism. Few studies have investigated the relationship between PROC genotype and plasma or clinical phenotypes. We addressed this issue in a large retrospective cohort of 1,115 heterozygous carriers of 226 PROC pathogenic or likely pathogenic mutations. Mutations were classified in three categories according to their observed or presumed association with type I, type IIa, or type IIb PC deficiency. The study population comprised 876 carriers of type I category mutations, 55 carriers of type IIa category mutations, and 184 carriers of type IIb category mutations. PC anticoagulant activity significantly influenced risk of first venous thrombosis (p trend < 10-4). No influence of mutation category on risk of whole or unprovoked thrombotic events was observed. Both PC anticoagulant activity and genotype significantly influenced risk of venous thrombosis. Effect of detrimental mutations on plasma phenotype was ambiguous in several carriers, whatever the mutation category. Altogether, our findings confirm that diagnosing PC inherited deficiency based on plasma measurement may be difficult but show that diagnosis can be improved by PROC genotyping.


Asunto(s)
Estudios de Asociación Genética , Deficiencia de Proteína C/congénito , Deficiencia de Proteína C/genética , Proteína C/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Preescolar , Femenino , Francia/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Deficiencia de Proteína C/epidemiología , Estudios Retrospectivos , Adulto Joven
10.
Am J Respir Crit Care Med ; 176(9): 878-85, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17673691

RESUMEN

RATIONALE: Severe sepsis is associated with an exacerbated procoagulant state with protein C (PC) system impairment. In contrast, the inflammatory and coagulation status of nonseptic patients with organ failure (OF) is less documented. OBJECTIVES: To compare coagulation activation, focusing on the PC system, and inflammatory status in septic and nonseptic patients with OF. METHODS: Thirty patients with severe sepsis and 30 nonseptic patients were recruited at the onset of OF and compared with 30 matched healthy subjects. We performed an extensive analysis of the PC pathway, including plasma protein measurements and quantification of leukocyte expression of PC system receptors. In addition, we analyzed the inflammatory status, based on inflammation-related gene leukocyte expression. MEASUREMENTS AND MAIN RESULTS: We observed coagulation activation, reflected by a similar increase in tissue factor mRNA expression, in the two patient groups when compared with the healthy subjects. Soluble thrombomodulin levels were higher in septic patients than in healthy control subjects, whereas PC, protein S, and soluble endothelial cell PC receptor levels were lower. Similar results were obtained in nonseptic patients with OF. Monocyte thrombomodulin overexpression, together with increased circulating levels of activated PC, suggests that the capacity for PC activation is at least partly preserved in both settings. No difference in the inflammatory profile was found between septic and nonseptic patients. CONCLUSIONS: The pathogenesis of OF in critical care patients is characterized by an overwhelming systemic inflammatory response and by exacerbated coagulation activation, independently of whether or not infection is the triggering event. Clinical trial registered with www.clinicaltrials.gov (NCT 00361725).


Asunto(s)
Coagulación Sanguínea/fisiología , Insuficiencia Multiorgánica/sangre , Proteína C/metabolismo , Sepsis/sangre , Adulto , Anciano , Antígenos CD/sangre , Antígenos CD/genética , Estudios de Casos y Controles , Receptor de Proteína C Endotelial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Multiorgánica/complicaciones , Proteína S/metabolismo , ARN Mensajero/metabolismo , Receptores de Superficie Celular/sangre , Receptores de Superficie Celular/genética , Sepsis/complicaciones , Trombomodulina/sangre , Trombomodulina/genética , Tromboplastina/genética , Tromboplastina/metabolismo
11.
Thromb Haemost ; 118(9): 1535-1544, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30071567

RESUMEN

Increasing number of patients are treated with direct oral anticoagulants (DOAC). An antidote for dabigatran inhibiting thrombin (idarucizumab) is available but no antidote is yet approved for the factor Xa (FXa) inhibitors (xabans). We hypothesized that a complex between Gla-domainless FXa and α2-macroglobulin (GDFXa-α2M) may neutralize the xabans without interfering with normal blood coagulation.Purified α2M was incubated with GDFXa to form GDFXa-α2M. Affinity of apixaban and rivaroxaban for GDFXa-α2M was only slightly decreased compared to FXa. Efficacy and harmlessness of GDFXa-α2M were tested in vitro and in vivo. Stoichiometric excess of GDFXa-α2M neutralized rivaroxaban and apixaban as attested by clot waveform assay and rotational thromboelastometry, whereas GDFXa-α2M alone had no effect on these assays. Efficacy and pro-thrombotic potential of GDFXa-α2M were also assessed in vivo. Half-life of GDFXa-α2M in C57BL6 mice was 4.9 ± 1.1 minutes, but a 0.5 mg/mouse dose resulted in uptake saturation such that 50% persistence was still observed after 170 minutes. Single administration of GDFXa-α2M significantly decreased the rivaroxaban-induced bleeding time (p < 0.001) and blood loss (p < 0.01). GDFXa-α2M did not increase D-dimer or thrombin-antithrombin complex formation, suggesting a lack of pro-thrombotic potential.GDFXa-α2M is therefore an attractive candidate for xaban neutralization neither pro- nor anticoagulant in vitro as well as in vivo.


Asunto(s)
Anticoagulantes/química , Inhibidores del Factor Xa/química , alfa 2-Macroglobulinas Asociadas al Embarazo/metabolismo , Pirazoles/química , Piridonas/química , Rivaroxabán/química , Administración Oral , Animales , Anticoagulantes/uso terapéutico , Inhibidores del Factor Xa/uso terapéutico , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Pirazoles/uso terapéutico , Piridonas/uso terapéutico , Rivaroxabán/uso terapéutico , Tromboelastografía , Trombina/metabolismo
12.
Thromb Haemost ; 96(3): 356-60, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16953279

RESUMEN

Thromboxane A2 receptor (TP) is an important actor in vascular physiology and plays a crucial role in the platelet activation process. Genetic polymorphisms of the gene coding for the TP have been described, but their impacts on platelet function tests are unknown. The aim of this study was to investigate the relationship between genetic polymorphisms of the coding sequence of the TP gene and platelet function tests. We investigated 100 healthy volunteers twice, one week apart by performing platelet aggregation and secretion tests. We sequenced the coding region of the TP gene and confronted the genetic variants with the phenotypic results. We identified five single nucleotide polymorphisms (SNP); one of them, T1712C, replaces Leu by Pro at position 133 of the isoform beta of the TP. Homozygosity for the minor allele of the C795T, C924T or the G1686A SNP was associated with a decreased expression of CD62P when platelets were stimulated with the TP agonist U46619. As C795T and C924T have been linked to clinical disorders in which TxA2 plays a key role, the possible role of the G1686A and T1712C SNP should also be examined in selected diseases.


Asunto(s)
Polimorfismo Genético , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Adolescente , Adulto , Citometría de Flujo , Variación Genética , Genotipo , Homocigoto , Humanos , Masculino , Modelos Genéticos , Selectina-P/biosíntesis , Fenotipo , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Polimorfismo de Nucleótido Simple
13.
Med Sci (Paris) ; 22(11): 985-9, 2006 Nov.
Artículo en Francés | MEDLINE | ID: mdl-17101102

RESUMEN

The coagulation system is governed by a subtle balance between clotting activators and inhibitors. Many genes can contribute to the overall phenotype, and polymorphisms may act to up regulate or down regulate the generation of thrombin, the coagulation-key enzyme. An increase in coagulation factor (gain function) or/and a decrease in coagulation inhibitors (loss of function) may favor venous thromboembolism (VTE). It has been observed since a long time that VTE may be a familial disease, but it was only in 1965 that Egeberg published the first case of inherited antithrombin (AT) deficiency. This was followed by similar reports of protein C (PC) and protein S (PS) deficiencies. Hereditary thrombophilia was thus initially considered as a rare monogenic disorder with incomplete penetrance. AT, PC and PS deficiencies are due to multiple and mostly private mutations of the corresponding genes. Most patients are heterozygous and experience VTE at adult age. Homozygosity associated with severe thrombosis at birth has been observed in newborns with undetectable PC or PS concentrations. The discovery of factor (F) V Leiden and F2 g.20210 G>A, two gain of function mutations, challenged the view of thrombophilia as a rare monogenic disorder. FV Leiden and F2 g.20210 G>A are due to a founder effect and affect populations of European descent with frequencies at 5% and 3% respectively. These two mutations are moderate of risk factor for thrombosis and paved the way for gene-gene and gene-environment interactions. Patients carrying more than one genetic risk factor are at higher risk to develop VTE. The exposition to acquired risk factors such as estrogen based oral contraception may also have a synergistic effect favoring thrombosis in patients with FV Leiden or other genetic risk factors.


Asunto(s)
Trastornos de la Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/genética , Mutación , Trombosis/genética , Variación Genética , Hemostasis , Humanos , Trombofilia/genética
14.
Circulation ; 108(8): 989-95, 2003 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-12912815

RESUMEN

BACKGROUND: The adenosine diphosphate (ADP) receptor P2Y12 plays a pivotal role in platelet aggregation, as demonstrated by the benefit conferred by its blockade in patients with cardiovascular disease. Some studies have shown interindividual differences in ADP-induced platelet aggregation responses ex vivo, but the mechanisms underlying this variability are unknown. METHODS AND RESULTS: We examined ADP-induced platelet aggregation responses in 98 healthy volunteers, and we identified 2 phenotypic groups of subjects with high and low responsiveness to 2 micromol/L ADP. This prompted us to screen the recently identified Gi-coupled ADP receptor gene P2Y12 for sequence variations. Among the 5 frequent polymorphisms thus identified, 4 were in total linkage disequilibrium, determining haplotypes H1 and H2, with respective allelic frequencies of 0.86 and 0.14. The number of H2 alleles was associated with the maximal aggregation response to ADP in the overall study population (P=0.007). Downregulation of the platelet cAMP concentration by ADP was more marked in 10 selected H2 carriers than in 10 noncarriers. CONCLUSIONS: In healthy subjects, ADP-induced platelet aggregation is associated with a haplotype of the P2Y12 receptor gene. Given the crucial role of the P2Y12 receptor in platelet functions, carriers of the H2 haplotype may have an increased risk of atherothrombosis and/or a lesser clinical response to drugs inhibiting platelet function.


Asunto(s)
Adenosina Difosfato/farmacología , Variación Genética , Proteínas de la Membrana , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/genética , Receptores Purinérgicos P2/genética , Adolescente , Adulto , Alelos , Secuencia de Bases , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos/genética , Pruebas Genéticas , Haplotipos , Heterocigoto , Humanos , Integrina beta3/genética , Masculino , Datos de Secuencia Molecular , Polimorfismo Genético , Receptores Purinérgicos P2Y12 , Valores de Referencia
15.
PLoS One ; 10(8): e0136652, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26313154

RESUMEN

Hematopoietic stem cells (HSC) differentiate into megakaryocytes (MK), whose function is to release platelets. Attempts to improve in vitro platelet production have been hampered by the low amplification of MK. Providing HSC with an optimal three-dimensional (3D) architecture may favor MK differentiation by mimicking some crucial functions of the bone marrow structure. To this aim, porous hydrogel scaffolds were used to study MK differentiation from HSC as well as platelet production. Flow cytometry, qPCR and perfusion studies showed that 3D was suitable for longer kinetics of CD34+ cell proliferation and for delayed megakaryocytic differentiation far beyond the limited shelf-life observed in liquid culture but also increased production of functional platelets. We provide evidence that these 3D effects were related to 1) persistence of MK progenitors and precursors and 2) prolongation of expression of EKLF and c-myb transcription factors involved in early MK differentiation. In addition, presence of abundant mature MK with increased ploidy and impressive cytoskeleton elongations was in line with expression of NF-E2 transcription factor involved in late MK differentiation. Platelets produced in flow conditions were functional as shown by integrin αIIbß3 activation following addition of exogenous agonists. This study demonstrates that spatial organization and biological cues synergize to improve MK differentiation and platelet production. Thus, 3D environment constitutes a powerful tool for unraveling the physiological mechanisms of megakaryopoiesis and thrombopoiesis in the bone marrow environment, potentially leading to an improved amplification of MK and platelet production.


Asunto(s)
Plaquetas/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , Trombopoyesis , Andamios del Tejido/química , Antígenos CD34/metabolismo , Plaquetas/citología , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Hidrogeles/química , Integrina alfa2/biosíntesis , Integrina beta3/biosíntesis , Factores de Transcripción de Tipo Kruppel/biosíntesis , Masculino , Megacariocitos/citología , Subunidad p45 del Factor de Transcripción NF-E2/biosíntesis , Proteínas Proto-Oncogénicas c-myb/biosíntesis
19.
Thromb Res ; 129(2): 152-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21824644

RESUMEN

The endothelial protein C receptor (EPCR) plays a crucial role in the anticoagulant and anti-inflammatory effects of the protein C pathway, whereas its soluble form (sEPCR) exhibits opposite properties. High plasma levels of sEPCR have been observed in subjects carrying the A3 haplotype of PROCR, the EPCR gene. Elevated plasma levels of sEPCR were also recently reported in women with preeclampsia (PE), a multisystemic syndrome involving inflammation, endothelial dysfunction and thrombosis. To determine whether this increase is genetically mediated or acquired, we analyzed sEPCR levels and the A3 haplotype distribution in 145 preeclamptic women and 145 age- and term-matched women with normal pregnancies enrolled in a case-control study. Plasma sEPCR levels were higher in the women with PE than in the controls, and this difference was not due to A3 haplotype over-representation. We observed a positive correlation between sEPCR levels and two markers of endothelium activation/damage (von Willebrand factor and soluble thrombomodulin), and a trend towards a third (sVCAM1). We also found an association between sEPCR levels in the highest quartile and the PE risk. The modest increase of sEPCR levels, together with the correlation with other endothelium activation/damage markers, suggest that it is more an innocent bystander of the endothelium activation/damage than an actor in PE.


Asunto(s)
Antígenos CD/sangre , Preeclampsia/sangre , Preeclampsia/epidemiología , Receptores de Superficie Celular/sangre , Disfunción Ventricular Izquierda/sangre , Disfunción Ventricular Izquierda/epidemiología , Adulto , Biomarcadores/sangre , Comorbilidad , Receptor de Proteína C Endotelial , Femenino , Francia/epidemiología , Humanos , Preeclampsia/diagnóstico , Embarazo , Prevalencia , Medición de Riesgo , Factores de Riesgo , Sensibilidad y Especificidad , Disfunción Ventricular Izquierda/diagnóstico
20.
Thromb Res ; 125(2): e33-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19878975

RESUMEN

INTRODUCTION: Protein S (PS) is a vitamin K-dependent plasma glycoprotein with a key role in the control of coagulation pathway on phospholipid membranes. We compared anticoagulant and membrane binding properties of PS altered by natural mutations (N217S, DelI203D204) affecting the epidermal growth factor like-domain 4 (EGF4) and causing PS deficiency. MATERIALS AND METHODS: Binding of recombinant, immunopurified PS (rPS) to several conformation-specific antibodies, to C4BP and to phospholipid liposomes was investigated by ELISA. PS binding to cells was analysed by flow cytometry. PS inhibitory activities were studied in plasma and purified systems. RESULTS AND CONCLUSIONS: Conformational changes produced by mutations were revealed by mapping with calcium-dependent antibodies. The immunopurified recombinant mutants (rPS) showed at 200-800 nM concentration reduced inhibition of coagulation (rPS217S, 10.2-17.3%; rPSDelI203D204, 5.8-8.9% of rPSwt) in FXa 1-stage clotting assay with APC. In thrombin generation assays the inhibition of ETP was reduced to 51.6% (rPS217S) and 24.1% (rPSDelI203D204) of rPSwt. A slightly shortened lag time (minutes) was also observed (rPS217S, 2.58; rPSDelI203D204, 2.33; rPSwt, 3.17; PS deficient plasma, 2.17). In flow cytometry analysis both mutants efficiently bound apoptotic cells in adhesion or in suspension. The affinity for phosphatidylserine-rich vesicles (apparent Kd: rPSwt 27.7+/-1.6 nM, rPS217S 146.0+/-16.1 nM and rPSDelI203D204 234.1+/-28.1 nM) was substantially increased by membrane oxidation (10.9+/-0.6, 38.2+/-3.5 and 81.4+/-6.0 nM), which resulted in a virtually normal binding capacity of mutants at physiological PS concentration. These properties help to define the molecular bases of PS deficiency, and provide further elements for PS-mediated bridging of coagulation and inflammation.


Asunto(s)
Anticoagulantes/metabolismo , Membrana Celular/metabolismo , Liposomas/metabolismo , Proteína S/genética , Proteína S/metabolismo , Animales , Sitios de Unión/genética , Coagulación Sanguínea/genética , Línea Celular , Línea Celular Transformada , Transformación Celular Viral , Proteína de Unión al Complemento C4b , Cricetinae , Fibroblastos/citología , Herpesvirus Humano 4/fisiología , Antígenos de Histocompatibilidad/metabolismo , Humanos , Riñón/citología , Membranas/metabolismo , Fosfolípidos/metabolismo , Unión Proteica/genética , Proteína C/metabolismo , Conformación Proteica , Deficiencia de Proteína S/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Virus 40 de los Simios/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA