Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Platelets ; 29(1): 84-86, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28726538

RESUMEN

Congenital platelet function disorders are often the result of defects in critical signal transduction pathways required for platelet adhesion and clot formation. Mutations affecting RASGRP2, the gene encoding the Rap GTPase activator, CalDAG-GEFI, give rise to a novel, and rare, group of platelet signal transduction abnormalities. We here report platelet function studies for two brothers (P1 and P2) expressing a novel variant of RASGRP2, CalDAG-GEFI(p.Gly305Asp). P1 and P2 have a lifelong history of bleeding with severe epistaxis successfully treated with platelet transfusions or rFVIIa. Other bleedings include extended hemorrhage from minor wounds. Platelet counts and plasma coagulation were normal, as was αIIbß3 and GPIb expression on the platelet surface. Aggregation of patients' platelets was significantly impaired in response to select agonists including ADP, epinephrine, collagen, and calcium ionophore A23187. Integrin αIIbß3 activation and granule release were also impaired. CalDAG-GEFI protein expression was markedly reduced but not absent. Homology modeling places the Gly305Asp substitution at the GEF-Rap1 interface, suggesting that the mutant protein has very limited catalytic activity. In summary, we here describe a novel mutation in RASGRP2 that affects both expression and function of CalDAG-GEFI and that causes impaired platelet adhesive function and significant bleeding in humans.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/genética , Plaquetas/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Hemorragia/etiología , Biomarcadores , Trastornos de las Plaquetas Sanguíneas/complicaciones , Niño , Índices de Eritrocitos , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hemorragia/diagnóstico , Humanos , Masculino , Modelos Moleculares , Linaje , Fenotipo , Agregación Plaquetaria/genética , Recuento de Plaquetas , Conformación Proteica , Transducción de Señal
2.
Blood ; 130(8): 1026-1030, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28637664

RESUMEN

Heritable platelet function disorders (PFDs) are genetically heterogeneous and poorly characterized. Pathogenic variants in RASGRP2, which encodes calcium and diacylglycerol-regulated guanine exchange factor I (CalDAG-GEFI), have been reported previously in 3 pedigrees with bleeding and reduced platelet aggregation responses. To better define the phenotype associated with pathogenic RASGRP2 variants, we compared high-throughput sequencing and phenotype data from 2042 cases in pedigrees with unexplained bleeding or platelet disorders to data from 5422 controls. Eleven cases harbored 11 different, previously unreported RASGRP2 variants that were biallelic and likely pathogenic. The variants included 5 high-impact variants predicted to prevent CalDAG-GEFI expression and 6 missense variants affecting the CalDAG-GEFI CDC25 domain, which mediates Rap1 activation during platelet inside-out αIIbß3 signaling. Cases with biallelic RASGRP2 variants had abnormal mucocutaneous, surgical, and dental bleeding from childhood, requiring ≥1 blood or platelet transfusion in 78% of cases. Platelets displayed reduced aggregation in response to adenosine 5'-diphosphate and epinephrine, but variable aggregation defects with other agonists. There were no other consistent clinical or laboratory features. These data enable definition of human CalDAG-GEFI deficiency as a nonsyndromic, recessive PFD associated with a moderate or severe bleeding phenotype and complex defects in platelet aggregation.


Asunto(s)
Plaquetas/patología , Factores de Intercambio de Guanina Nucleótido/genética , Hemorragia/genética , Mutación/genética , Alelos , Secuencia de Bases , Femenino , Humanos , Masculino , Linaje
3.
Platelets ; 28(5): 484-490, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27819553

RESUMEN

Despite the increased worldwide awareness, over the last decade, of the platelet-type von Willebrand Disease (PT-VWD), many uncertainties remain around this rare platelet bleeding disorder. This report aims to correctly identify and study the phenotype of new patients and highlights the diagnostic and therapeutic challenges this disease remains to pose. We describe four PT-VWD cases confirmed by genetic analysis in which either the diagnosis and/or the treatment posed challenge. We provide the details of the clinical presentation, laboratory analysis, and the treatment and the responses in each case. We show that in addition to type 2B VWD, PT-VWD can be misdiagnosed as idiopathic thrombocytopenic purpura, neonatal alloimmune thrombocytopenia, and unexplained gestational thrombocytopenia. The disease can be diagnosed as early as 1 year of age and with phenotypically normal parents. Bleeding in some patients can be managed successfully using Humate P and DDAVP combined with tranexamic acid with no significant thrombocytopenia. We provide for the first time an evidence of an efficient response to rFVIIa in PT-VWD. Anaphylactic reaction to VWF preparations may be related to PT-VWD and the development of HLA antibodies is not uncommon. Progressive thrombocytopenia with normal VWF levels can be seen with PT-VWD and the platelet count was normalized at 2.5 weeks postpartum in one case. We conclude that these studies represent a record of clinical observations/interventions that help improve diagnoses/management of PT-VWD, highlight the variations in age and clinical presentations, laboratory diagnostic approaches, the importance of genetic testing for accurate diagnosis and consideration of therapeutic alternatives.


Asunto(s)
Desamino Arginina Vasopresina/administración & dosificación , Factor VIII/administración & dosificación , Factor VIIa/administración & dosificación , Hemorragia , Ácido Tranexámico/administración & dosificación , Enfermedades de von Willebrand , Adolescente , Preescolar , Quimioterapia Combinada/métodos , Femenino , Hemorragia/sangre , Hemorragia/diagnóstico , Hemorragia/tratamiento farmacológico , Humanos , Recién Nacido , Masculino , Proteínas Recombinantes/administración & dosificación , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/diagnóstico , Enfermedades de von Willebrand/tratamiento farmacológico
4.
Semin Thromb Hemost ; 43(1): 92-100, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27978591

RESUMEN

von Willebrand disease type 2B (VWD2B) expresses gain-of-function mutations that enhance binding of an individual's von Willebrand factor (VWF) to its platelet ligand, glycoprotein Ib (GPIb), and which are usually identified by increased ristocetin-induced platelet aggregation (RIPA). We describe here the phenotypic profile of 38 genotypically selected VWD2B-affected family members (AFMs) belonging to 19 unrelated families. Major bleeding was observed in 68.4% of AFMs (previous to their diagnosis and registered by lifetime interviews), with a total of 46 episodes (1.21/patient), and was found to be highly related to the individual bleeding score and presence of thrombocytopenia, but otherwise unrelated to other laboratory parameters. Excessive muco-cutaneous bleeding symptoms were often reported, the most frequent of which comprised menorrhagia, epistaxis, easy bruising, and bleeding after teeth extraction/in oral cavity. Eight unaffected family members were also studied. The prevalence of VWD2B within families was 0.826, and the penetrance of mutations was complete, making it mandatory to study entire family sets to complete diagnostic profiles. Seven heterozygous missense mutations were found, the most common being p.V1316M. In the p.R1308C group, 75% of the AFMs showed absence of RIPA at 0.5 mg/mL, 66.6% of whom had VWF:RCo < 10 IU/dL, and 50% of whom had VWF:CB < 10 IU/dL. In the p.S1310F group, none of the AFMs had VWF:RCo/VWF:Ag < 0.6 (RCo/Ag), but 100% had VWF:CB/VWF:Ag < 0.6/(CB/Ag). Patients with p.P1266L and p.R1304V were characterized as atypical VWD2B. Two de novo mutations were found in four AFMs belonging to two families. We also describe a novel mutation: p.Y1258C. Of our patients, 70.5% had O blood group. In conclusion, a normal RCo/Ag and a negative RIPA at 0.5 mg/mL do not necessarily rule out a diagnosis of VWD2B.


Asunto(s)
Enfermedad de von Willebrand Tipo 2/genética , Enfermedades de von Willebrand/genética , Factor de von Willebrand/genética , Femenino , Genotipo , Humanos , Masculino , Mutación
5.
Acta bioquím. clín. latinoam ; 50(2): 273-289, jun. 2016. graf, tab
Artículo en Español | LILACS | ID: biblio-837606

RESUMEN

El factor von Willebrand (VWF) es una glicoproteína que se sintetiza en células endoteliales y en megacariocitos. Su vida media es de ~12 horas. Está formado por multímeros de diferentes pesos moleculares, pequeños, intermedios, grandes y extragrandes. La actividad funcional reside en los multímeros grandes, y los extragrandes son trombogénicos. Promueve la adhesión plaquetaria al subendotelio, la agregación plaquetaria y transporta al FVIII en plasma, protegiéndolo de su degradación por proteasas. La enfermedad de von Willebrand es el trastorno hemorrágico más frecuente; se describen deficiencias cuantitativas (parcial: VWD1; total: VWD3) o defectos cualitativos (VWD2A, VWD2M, VWD2B y VWD2N). La expresión clínica es variable (sangrado muco-cutáneo) y su herencia autosómica, dominante o recesiva, según las variantes. Los niveles del VWF dependen de factores genéticos y no genéticos que afectan el diagnóstico y la expresión clínica. Para llegar al diagnóstico se precisan varias pruebas, algunas inespecíficas. El laboratorio comienza con pruebas orientadoras, se continúa con pruebas confirmatorias, y posteriormente pruebas para definir la variante de VWD. El diagnóstico genotípico es fundamental para lograr el diagnóstico diferencial entre VWD2B vs. PT-VWD y VWD2N vs. Hemofilia A (leve-moderada), diferenciar VWD de AVWS y discriminar variantes VWD2.


Von Willebrand factor (VWF) is a glycoprotein with essential roles in both primary and secondary hemostasis, synthesized by endothelial cells and megakaryocytes. Its half-life is ~12 hours. VWF consists in multimers of different molecular weight: small, intermediate, large and ultra large. The functional activity resides in the large multimers; the ultra large are thrombogenic. VWF promotes platelet adhesion to subendothelium, platelet aggregation and binds FVIII, protecting it from proteolysis and preserving its hemostatic function. Von Willebrand disease is the most common bleeding disorder; qualitative defects (VWD2A, VWD2M, VWD2B and VWD2N) and quantitative deficiencies (VWD1 and VWD3) are described. The clinical expression is variable (mucocutaneous bleeding); VWF levels depend on genetic and non-genetic factors affecting diagnosis and clinical expression. The inheritance can be autosomal, dominant or recessive according to the variants. To reach diagnosis, several tests are required, being some of them unspecific. The laboratory testing begins with global tests, followed by confirmatory tests and further tests to define the variant of VWD. Genotypic studies are essential to achieve the differential diagnosis between VWD2B vs. PT-VWD, VWD2N vs. Hemophilia A (mild to moderate) and differentiate VWD from AVWS and discriminate VWD2 variants.


O fator de von Willebrand (vWF) é uma glicoproteína sintetizada em células endoteliais e em megacariócitos. Sua vida média é de ~12 horas. É constituído por multímeros de pesos moleculares diferentes, pequenos, intermediários, grandes e extragrandes. A atividade funcional reside nos multímeros grandes, sendo os extragrandes, trombogênicos. Promove adesão das plaquetas ao subendotélio, a agregação plaquetária e transporta o FVIII em plasma, protegendo-o de sua degradação. A doença de von Willebrand é o distúrbio hemorrágico mais frequente; são descritas deficiências quantitativas (parcial: VWD1; total: VWD3) ou defeitos qualitativos (VWD2A, VWD2M, VWD2B e VWD2N). A expressão clínica é variável, (sangramento mucocutâneo), e sua herança autossômica dominante ou recessiva de acordo com as variantes. Os níveis de vWF dependem de fatores genéticos e não-genéticos que afetam o diagnóstico e a expressão clínica. Para fazer o diagnóstico, vários testes são necessários, alguns inespecíficos. O laboratório começa com testes orientadores, continua com testes de confirmação e, mais tarde, com testes para definir a variante de VWD. O diagnóstico genotípico é essencial para alcançar o diagnóstico diferencial entre VWD2B vs. PT-VWD e VWD2N vs. Hemofilia A (leve a moderada), diferenciar VWD de AVWS, discriminar variantes VWD2.


Asunto(s)
Humanos , Masculino , Femenino , Enfermedades de von Willebrand , Factor de von Willebrand , Hemostasis , Fenotipo , Genotipo
6.
Blood ; 127(23): 2791-803, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27084890

RESUMEN

Inherited bleeding, thrombotic, and platelet disorders (BPDs) are diseases that affect ∼300 individuals per million births. With the exception of hemophilia and von Willebrand disease patients, a molecular analysis for patients with a BPD is often unavailable. Many specialized tests are usually required to reach a putative diagnosis and they are typically performed in a step-wise manner to control costs. This approach causes delays and a conclusive molecular diagnosis is often never reached, which can compromise treatment and impede rapid identification of affected relatives. To address this unmet diagnostic need, we designed a high-throughput sequencing platform targeting 63 genes relevant for BPDs. The platform can call single nucleotide variants, short insertions/deletions, and large copy number variants (though not inversions) which are subjected to automated filtering for diagnostic prioritization, resulting in an average of 5.34 candidate variants per individual. We sequenced 159 and 137 samples, respectively, from cases with and without previously known causal variants. Among the latter group, 61 cases had clinical and laboratory phenotypes indicative of a particular molecular etiology, whereas the remainder had an a priori highly uncertain etiology. All previously detected variants were recapitulated and, when the etiology was suspected but unknown or uncertain, a molecular diagnosis was reached in 56 of 61 and only 8 of 76 cases, respectively. The latter category highlights the need for further research into novel causes of BPDs. The ThromboGenomics platform thus provides an affordable DNA-based test to diagnose patients suspected of having a known inherited BPD.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Predisposición Genética a la Enfermedad , Hemorragia/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Trombosis/genética , Estudios de Casos y Controles , Variaciones en el Número de Copia de ADN , Femenino , Estudios de Asociación Genética/métodos , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/métodos
7.
Thromb Res ; 137: 92-96, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26577258

RESUMEN

BACKGROUND: The aim of our study was to evaluate the effect of programmed physical activity and a single exercise test on the number of CD309+ circulating endothelial progenitor cell (EPC) and their relation to the variation in plasma levels of VEGF in chronic coronary patients. METHODS: 21 patients <75 years with chronic stable coronary artery disease were included. All patients underwent exercise myocardial perfusion SPECT. Then, participants were divided into two groups: one group (11 patients) underwent cardiac rehabilitation program and the other (10 patients) continued with the standard treatment. Blood samples were obtained at baseline, 30 min after exercise ended and at one and three months during follow-up. RESULTS: VEGF values decreased significantly after exercise SPECT test. After one month, there was a significant increase in VEGF levels compared to those measured immediately after exercise. All patients showed a decrease in the values of EPC at 1 and 3-month follow-up. There was an inverse and statistically significant relation between change of EPC and VEGF between the baseline and 1 month. CONCLUSIONS: The increase of VEGF at 1-month, with respect to baseline values correlated with decreased levels of EPC. This association was independent of the onset of ischemia in the perfusion study.


Asunto(s)
Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/terapia , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Terapia por Ejercicio/métodos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adaptación Fisiológica , Anciano , Línea Celular , Proliferación Celular , Supervivencia Celular , Enfermedad de la Arteria Coronaria/fisiopatología , Prueba de Esfuerzo , Femenino , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/fisiología
8.
Bol. Acad. Nac. Med. B.Aires ; 93(2): 269-276, jul.-dic. 2015. graf
Artículo en Español | LILACS | ID: biblio-997434

RESUMEN

El inhibidor lúpico (IL) es uno de los criterios de laboratorio para Síndrome Antifosfolipídico (SAF); sin embargo, puede detectarse en individuos asintomáticos o estar asociado a otras situaciones clínicas. Presentamos un análisis retrospectivo de 1000 exámenes consecutivos para IL (TTPA, DRVVT) de los cuales 249 casos no presentaban criterios clínicos de SAF. Aplicando los criterios SSC-ISTH, hallamos IL+ en 27,30% (205/751) y 43,37% (108/249) de los casos con y sin criterios clínicos de SAF respectivamente; analizándose en estos últimos casos las características clínicas y de laboratorio. Contexto clínico de casos IL+ sin SAF: 18,52% asintomáticos, 34,26% síntomas de sangrado y 47,22% otras manifestaciones. Otras alteraciones de laboratorio en casos IL+ sin SAF, con síntomas de sangrado: detectamos alteraciones plaquetarias, descenso de VWF:RCo y/o VWF:Ag, disminución de FVIII, FV, FVII, FXI o fibrinógeno e hiperfibrinolisis en el 54,05% de los casos. El análisis mostró detección de IL+ en un número importante de estudios (108/1000) sin criterios SAF. Los casos con IL+ y sangrado representan un desafío particular, al requerir evaluar otros posibles defectos subyacentes, que pudiesen justificar el comportamiento clínico. La detección e identificación de defectos combinados requiriere de un análisis minucioso, a fin de alcanzar un diagnóstico correcto, esencial para tomar decisiones terapéuticas adecuadas. (AU)


Despite lupus anticoagulant (LA) is one of the laboratory criteria for antiphospholipid syndrome (APS), it can be present in asymptomatic subjects or it can be associated with other clinical settings. We present a retrospective analysis of 1000 consecutive LA assays (APTT, DRVVT), 249 of them were performed in patients without clinical criteria for APS. According to ISTH criteria, positive LA was found in 27.30% (205/751) and 43.37% (108/249) of cases with or without APS criteria respectively; in the last group, the analysis of clinical background and laboratory characteristics was done. Clinical background of LA+ cases without APS: 18.52% asymptomatic, 34.26% bleeding symptoms and 47.22% other clinical settings. Other abnormal laboratory tests in LA+ cases without APS and bleeding symptoms: platelet dysfunction; low VWF:RCo and/or VWF:Ag; decrease of FVIII, FV, FVII, FXI or fibrinogen and hyperfibrinolysis were found in the 54.05% of the cases. The analysis showed positive LA in an important number of cases (108/1000) without criteria of APS. Those LA+ cases with bleeding symptoms represent a particular challenge because other possible underlying defects have to be analysed in order to explain the clinical behaviour. The detection and identifications of combined defects required a careful analysis in order to achieve accurate diagnosis, essential for therapeutic decisions. (AU)


Asunto(s)
Humanos , Inhibidor de Coagulación del Lupus/análisis , Inhibidor de Coagulación del Lupus/sangre , Síndrome Antifosfolípido , Trastornos de las Plaquetas Sanguíneas , Diagnóstico Diferencial
9.
Bol. Acad. Nac. Med. B.Aires ; 92(2): 309-314, jul.-dic. 2014. tab, ilus
Artículo en Español | LILACS | ID: biblio-998762

RESUMEN

La enfermedad de von Willebrand tipo plaquetario (PT-VWD) y tipo 2B (2B-VWD) son trastornos hemorrágicos raros, caracterizados por agregación plaquetaria a bajas concentraciones de ristocetina (RIPA). El diagnóstico diferencial no es fácil y representa un desafío. Hasta el presente, sólo se habían reportado cinco mutaciones en el gen GP1BA relacionadas con este desorden. Describimos aquí la sexta mutación relacionada con PT-VWD, en un paciente con sintomatología hemorrágica severa, macro-trombocitopenia, leve agregación plaquetaria espontánea, RIPA positivo a 0,3 y 0,4 mg/mL, VWF:RCo/VWF: Ag<0,2 y estudios discriminatorios positivos para PT-VWD. VWFpp/VWF: Ag resultó normal a diferencia del 2B-VWD que en algunas oportunidades resulta afectado. El exón 28 del gen VWF del paciente y su madre no reveló mutaciones. Identificamos una sustitución G>T en el nucleótido 3805 en el gen GP1BA del paciente, resultando en un cambio de Trp a Leu en el residuo 246 (p.W246L), en la región de la GPIBa que une al VWF. Esta mutación no se identificó en su madre ni en 100 controles sanos. Es considerada como dañina por análisis in sílico. Consideramos que esta sustitución es responsable del fenotipo PT-VWD del paciente. Dada la ausencia de la misma en los 100 normales estudiados, no se considera un polimorfismo


Platelet-type von Willebrand disease (PT-VWD) and type 2B von Willebrand disease (2B-VWD) are rare bleeding disorders characterized by increased ristocetin-induced platelet aggregation (RIPA) at low concentrations. Diagnosis of either condition is not easy and the differential diagnosis is especially challenging. Five mutations in the GP1BA gene related to PT-VWD and near 50 patients are currently reported worldwide. We herein describe a patient with severe bleeding symptoms, macro thrombocytopenia, mild spontaneous platelet aggregation, positive RIPA at 0.3 and 0.4 mg/mL, VWF: RCo/VWF: Ag <0.2, normal VWFpp/VWF: Ag ratio, and RIPA mixing tests and cryoprecipitate challenge positive for PT-VWD. GP1BA gene was studied in the patient, his mother, and 100 healthy control subjects. We identified a substitution G>T at nucleotide 3805 in the patient's GP1BA gene, resulting in a Trp to Leu amino acid change at residue 246 (p.W246L), within the VWF binding region. This mutation was absent in his unaffected mother and also in the 100 controls, and was predicted as damaging by in silico analysis. The residue is located in a strongly conserved position in the phylogenetic tree. These findings argue in favor of considering this substitution does not represent a polymorphism, and is therefore responsible for the PT-VWD phenotype of the patient


Asunto(s)
Humanos , Masculino , Enfermedades de von Willebrand/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Predisposición Genética a la Enfermedad/genética , Mutación Missense , Enfermedades de von Willebrand/sangre , Análisis Mutacional de ADN , Salud de la Familia , Secuencia de Aminoácidos
10.
Semin Thromb Hemost ; 40(2): 151-60, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24474090

RESUMEN

Platelet-type von Willebrand disease (PT-VWD) and type 2B von Willebrand disease (2B-VWD) are rare bleeding disorders characterized by increased ristocetin-induced platelet aggregation (RIPA) at low concentrations of ristocetin. Diagnosis of either condition is not easy and the differential diagnosis between the two entities is especially challenging as evidenced by high levels of misdiagnosis of both conditions, but particularly PT-VWD. Five mutations in the GP1BA gene related to PT-VWD and less than 50 patients are currently reported worldwide. We herein describe a patient with severe bleeding symptoms, macrothrombocytopenia, mild spontaneous platelet aggregation, positive RIPA at 0.3 and 0.4 mg/mL, von Willebrand factor ristocetin cofactor (VWF:RCo) to antigen (VWF:Ag) < 0.2, normal VWF propeptide/VWF:Ag ratio, and RIPA mixing tests and cryoprecipitate challenge positive for PT-VWD. GP1BA gene was studied in the patient, in his mother, and in 100 healthy control subjects. We identified a heterozygous substitution G > T located at nucleotide 3805 in the g.DNA of the patient's GP1BA gene, resulting in a Trp to Leu amino acid change at residue 246 (p.W246L). This mutation was absent in his unaffected mother and also in the 100 controls, and was predicted as damaging by in silico analysis. The residue W246 is located within the VWF-binding region and exists in a strongly conserved position in the phylogenetic tree, which is expected to be unable to tolerate substitutions without changing its functional characteristics. These findings argue strongly in favor of the view that this substitution does not represent a polymorphism and is therefore responsible for the PT-VWD phenotype of the patient.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Mutación Missense , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Enfermedades de von Willebrand/genética , Secuencia de Aminoácidos , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Madres , Adulto Joven , Enfermedades de von Willebrand/sangre
11.
Thromb Haemost ; 100(1): 45-51, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18612537

RESUMEN

The Gray platelet syndrome (GPS) is a rare inherited disorder linked to undefined molecular abnormalities that prevent the formation and maturation of alpha-granules. Here, we report studies on two patients from unrelated families that confirm phenotypic heterogeneity in the disease. First we used immunoelectron microscopy (I-EM) to confirm that TREM-like transcript-1 (TLT-1) is mostly localized to alpha-granule membranes of normal platelets. Then we performed Western blotting (WB) and flow cytometry with permeabilized platelets to show that TLT-1 is selectively reduced in the platelets of patient 1, previously noted to be deficient in glycoprotein (GP)VI (Nurden et al., Blood 2004; 104: 107-114). Yet both TLT-1 and GPVI were normally expressed in platelets of patient 2. Usual levels of JAM-C and claudin-5, also members of the immunoglobulin receptor family, were detected in platelets of both patients. In contrast, P-selectin was markedly decreased for patient 1 but not patient 2. Two metalloproteases, MMP-2 and MMP-9 were normally present. As predicted, platelets of patient 1 showed little labelling for TLT-1 in I-EM, whereas residual Fg was seen in small vesicular structures and P-selectin lining vacuoles or channels of what may be elements of the surface-connected canalicular system. Our results identify TLT-1 as a glycoprotein potentially targeted in platelets of GPS patients, while decreases in at least three membrane glycoproteins suggest that an unidentified proteolytic activity may contribute to the phenotype in some patients with this rare disease.


Asunto(s)
Plaquetas/metabolismo , Deficiencia de Almacenamiento del Pool Plaquetario/metabolismo , Receptores Inmunológicos/sangre , Plaquetas/ultraestructura , Western Blotting , Antígenos CD36/sangre , Moléculas de Adhesión Celular/sangre , Femenino , Citometría de Flujo , Humanos , Metaloproteinasa 2 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/sangre , Microscopía Inmunoelectrónica , Selectina-P/sangre , Fenotipo , Deficiencia de Almacenamiento del Pool Plaquetario/patología , Síndrome
12.
Rev. argent. cardiol ; 75(6): 429-435, nov.-dic. 2007. ilus, graf, tab
Artículo en Español | LILACS | ID: lil-633957

RESUMEN

Introducción El ejercicio físico es útil para los pacientes con enfermedad coronaria y es un estímulo para el desarrollo de circulación colateral. Ésta podría estar determinada por un aumento en la producción y circulación de células progenitoras. Objetivo Evaluar el efecto del ejercicio físico programado sobre la producción y el número circulante de células progenitoras en pacientes coronarios crónicos estables. Material y métodos Estudio prospectivo, controlado, aleatorizado y abierto con la inclusión de 18 pacientes (8 en grupo ejercicio y 10 en grupo control) con enfermedad coronaria estable, < 75 años, que no hubieran participado en grupos de ejercicio programado en los últimos 3 meses. La determinación de las células progenitoras se realizó por citometría de flujo utilizando marcaciones con anticuerpos monoclonales CD45-FITC, CD34-FITC y CD133/1-PE. Resultados En el grupo control, el nivel de CD45 no tuvo variación significativa (0,724 ± 0,256 versus 0,765 ± 0,216 [media ± EE cada 100.000 eventos]), mientras que en el grupo ejercicio el nivel de CD45(+)/CD133(+) se incrementó de 0,497 ± 0,161 a 2,265 ± 1,003 luego de un mes de actividad física programada sin alcanzar significación estadística. Al analizar los niveles de CD34 se observó que en el grupo control se incrementaron de 0,196 ± 0,086 a 0,235 ± 0,063 (p = NS). En el grupo ejercicio, la variación fue mucho mayor: 0,220 ± 0,078 a 0,844 ± 0,172 (p = 0,0046; p = 0,0092 versus el grupo control). Conclusión El ejercicio físico programado en pacientes coronarios promueve un incremento de las células progenitoras circulantes. Su estímulo persistente podría ser la base para un mayor desarrollo de circulación colateral.


Background Exercise is useful for patients with coronary artery disease, and it works as a stimulus for the development of collateral circulation, which could be the result of an increase in the production and circulation of endothelial progenitor cells. Objective The objective of this study was to assess the effect of programmed exercise on the production and number of circulating endothelial progenitor cells in patients with chronic and stable coronary disease. Material and Methods We conducted a prospective, randomized, controlled and open study that included 18 patients (8 in the exercise group and 10 in the control group) with demonstrated chronic stable angina, < 75 years. Patients eligible should not have participated in programmed exercise groups within the last 3 months. Progenitor cells were determined by flow cytometry using marked monoclonal antibodies CD45-FITC, CD34-FITC y CD133/1-PE. Results After 1 month of programmed physical activity, CD45 level did not show any significant change in the control group (0.724±0.256 vs 0.765±0.216 [mean ± SE each 100,000 events]), whereas it increased from basal levels of 0.497±0.161 to 2.265±1.003 in the exercise group, though this change was not statistically significant. Analyzing CD34 levels, an increase from 0.196±0.086 to 0.235±0.063 was seen in the control group (p = NS). This increase was greater in the exercise group: 0.220±0.078 to 0.844±0.172 (p=0.0046; p=0.0092 vs control group). Conclusions In patients with coronary artery disease, programmed exercise promotes an increase in the level of circulating progenitor cells. Its persistent stimulus could be the basis of a greater development of collateral circulation.

13.
Thromb Haemost ; 94(3): 578-84, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16268475

RESUMEN

There is a growing body of evidence on the role of nitric oxide (NO) in human platelet physiology regulation. Recently, interest has developed in the functional role of an alternative redox form of NO, namely nitroxyl (HNO/NO-), because it is formed by a number of diverse biochemical reactions. The aim of the present study was to comparatively analyze the effect of HNO and NO on several functional parameters of human platelets. For this purpose, sodium trioxodinitrate (Angeli's salt,AS) and sodium nitroprusside (SNP) were used as HNO and NO releasers, respectively. BothAS and SNP significantly inhibited platelet aggregation and ATP release induced by different agonists and adrenaline. AS or SNP did not modify the expression of platelet glycoproteins (Ib, IIb-IIIa, la-IIa, IV), whereas they substantially decreased the levels of CD62P, CD63 and of PAC-1 (a platelet activated glycoprotein IIb/IIIa epitope) after the stimulation with ADP. AS and SNP significantly increased cGMP accumulation in a 1H-[1,2,4]oxadiazolo [4,3-a] quinoxalin-1-one (ODQ)-sensitive manner. However, while L-cysteine reduced the effect of AS, it increased the effect of SNP on this parameter. Accordingly, a differential effect of L-cysteine was observed on the antiaggregatory effect of both compounds. In summary, these results indicate that HNO is an effective inhibitor of human platelet aggregation.


Asunto(s)
Plaquetas/efectos de los fármacos , Óxidos de Nitrógeno/farmacología , Agregación Plaquetaria/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Antígenos CD/metabolismo , Plaquetas/metabolismo , GMP Cíclico/metabolismo , Cisteína/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Técnicas In Vitro , Óxido Nítrico/farmacología , Donantes de Óxido Nítrico , Nitritos , Nitroprusiato , Selectina-P/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Tetraspanina 30 , Factores de Tiempo
14.
Thromb Haemost ; 92(4): 820-8, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467914

RESUMEN

Abciximab is an anti-GPIIb-IIIa drug widely used to prevent thrombotic complications during percutaneous coronary intervention. We now report on the immunologic origin of thrombocytopenia developing between 7 and 12 days after the onset of abciximab infusion. Antibodies directed against abciximabcoated platelets were located in 5 patients with delayed thrombocytopenia, just as they were present in a patient whose platelet count fell within a few hours after receiving the drug. Abciximab-dependent IgG antibody was revealed in serum using control platelets in the monoclonal antibody immobilization of platelet antigens assay (MAIPA) performed with SZ22, a MoAb to GPIIb. The presence of IgG antibodies specific for platelets sensitized with abciximab was confirmed by flow cytometry. They were not located in 13 patients receiving abciximab but whose platelet counts remained stable. For three patients, antibodies were transient and their presence related to the extent of the thrombocytopenia. Surprisingly, antibodycontaining plasma from three patients induced abciximabdependent activation and aggregation of normal platelets, a finding confirmed by electron microscopy. Immunogold labeling revealed that abciximab was associated with platelets in the aggregate, suggesting that its inhibitory effect was overcome by the platelet stimulation. In summary, these results show that abciximab-dependent thrombocytopenia can be delayed and potentially prothrombotic.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Fragmentos Fab de Inmunoglobulinas/efectos adversos , Trombocitopenia/inmunología , Abciximab , Anciano , Autoanticuerpos/sangre , Autoanticuerpos/farmacología , Plaquetas/metabolismo , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/inmunología , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/inmunología , Recuento de Plaquetas , Trombocitopenia/inducido químicamente , Trombofilia/inducido químicamente , Factores de Tiempo
15.
Blood Coagul Fibrinolysis ; 15(3): 235-40, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15060419

RESUMEN

Bleeding and thrombosis in myeloproliferative disorders (MPD) are common events, sometimes both are present in the same patient during the course of the disease. Platelet activation in patients with MPD is often suggested. The present study analyses the presence of circulating activated platelets, using simultaneously flow cytometry and aggregometric studies in MPD. We studied 28 patients: 13 with polycythaemia vera, seven with essential thrombocythaemia, and eight chronic myeloid leukaemia. We performed functional tests, aggregation and adenosine triphosphate (ATP) release and flow cytometric assays (mepacrine staining and platelet activation markers CD62, CD63 and fibrinogen binding (B-FG)). Twenty-one MPD samples (75%) had reduced aggregation and ATP release. Acquired delta-SPD was detected in 11 of 28 MPD patients (39%), and we found no association between reduced mepacrine labelling and abnormal ATP release. High levels of activation markers were obtained: CD62 in 19 of 28 patients (68%), CD63 in 13 of 28 patients (46%) and B-FG in 19 of 28 patients (68%). The most prevalent abnormality was a reduced aggregation and ATP release. The lack of association between ATP release and mepacrine labelling suggests that other mechanisms, besides the deficit of intraplatelet ATP/adenosine diphosphate, might occur. High levels of activation markers were also observed. We conclude that both tests are complementary and necessary to understand the functional status of platelets in MPD.


Asunto(s)
Trastornos Mieloproliferativos/sangre , Activación Plaquetaria , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Biomarcadores/sangre , Femenino , Citometría de Flujo/métodos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Masculino , Persona de Mediana Edad , Nefelometría y Turbidimetría/métodos , Pruebas de Función Plaquetaria/métodos , Policitemia Vera/sangre , Trombocitemia Esencial/sangre
16.
Kidney Int ; 62(4): 1338-48, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12234304

RESUMEN

BACKGROUND: Nitric oxide (NO) is an endogenous vasodilator and platelet inhibitor. An enhanced NO production has been detected in patients with hemolytic uremic syndrome (HUS), although its implication in HUS pathogenesis has not been clarified. METHODS: A mouse model of Shiga toxin 2 (Stx2)-induced HUS was used to study the role of NO in the development of the disease. Modulation of l-arginine-NO pathway was achieved by oral administration of NO synthase (NOS) substrate or inhibitors, and renal damage, mortality and platelet activity were evaluated. The involvement of platelets was studied by means of a specific anti-platelet antibody. RESULTS: Inhibition of NO generation by the NOS inhibitor L-NAME enhanced Stx2-mediated renal damage and lethality; this effect was prevented by the addition of l-arginine. The worsening effect of L-NAME involved enhanced Stx2-mediated platelet activation, and it was completely prevented by platelet depletion. CONCLUSIONS: NO exerts a protective role in the early pathogenesis of HUS, and its inhibition potentiates renal damage and mortality through a mechanism involving enhanced platelet activation.


Asunto(s)
Síndrome Hemolítico-Urémico/metabolismo , Óxido Nítrico/metabolismo , Toxina Shiga II/toxicidad , Animales , Degranulación de la Célula/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fibrinógeno/análisis , Síndrome Hemolítico-Urémico/inducido químicamente , Síndrome Hemolítico-Urémico/mortalidad , Síndrome Hemolítico-Urémico/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Ratones , Ratones Endogámicos BALB C , NG-Nitroarginina Metil Éster/farmacología , Activación Plaquetaria/efectos de los fármacos , Trombosis/inducido químicamente , Trombosis/metabolismo , Trombosis/mortalidad , Trombosis/patología , Urea/sangre
17.
Pathophysiol Haemost Thromb ; 32(4): 155-7, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12759515

RESUMEN

An increased risk of bleeding is associated with a more intense oral anticoagulation, a greater international normalized ratio (INR) variability and the use of aspirin. We studied the INR variability of patients (n = 121) with modern heart valves who had been prospectively randomized to receive acenocoumarol at a targeted INR of 2.4-3.6 plus aspirin 100 mg/day or acenocoumarol alone at the same dosage, to evaluate whether aspirin influences variability and thus the risk of bleeding. Variability was similar in patients with no events regardless of the use of aspirin. A statistically significantly higher variability was observed in patients with bleeding events independently of the use of aspirin. Nevertheless, the concomitant use of aspirin in patients with a high variability should be monitored closely and thoroughly.


Asunto(s)
Aspirina/farmacología , Prótesis Valvulares Cardíacas/efectos adversos , Relación Normalizada Internacional/normas , Acenocumarol/administración & dosificación , Acenocumarol/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticoagulantes/administración & dosificación , Anticoagulantes/farmacología , Aspirina/administración & dosificación , Femenino , Hemorragia/inducido químicamente , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Riesgo , Trombosis/etiología , Trombosis/prevención & control
18.
Bol. Acad. Nac. Med. B.Aires ; 77(2): 240-6, ene.-jun. 1999. tab
Artículo en Español | LILACS | ID: lil-262108

RESUMEN

Se estudiaron 91 embarazadas normales. Hubo 2 casos de plaquetopenia leve en el 2§ trimestre y 6 en el 3§; se observó aumento del factor von Willebrand. El Fibrinógeno aumentó hacia el tercer trimestre. La proteína S (PS) biológica e inmunológica libre disminuyeron. La Resistencia a la Proteína C Activada observó un acortamiento entre el primer y segundo trimestre. Aumentó el tiempo de lisis de euglobulinas basal y disminuyó la diferencia entre euglobulinas pre y post isquemia. Dentro de la marcación de receptores plaquetarios sólo la unión de fibrinógeno resultó incrementado (p<0.05). Se analizaron las causas que inducen la aparición de 2 o más abortos/pérdidas fetales (a/PF) y la frecuencia de alteraciones de la hemostasia. La población se dividió en 3 grupos. A: A/PF primarias (media 32 años, 204 gestas, 190 A/PF espontáneas, 73 por ciento 1er. trimestre (T), 18 por ciento 2do. T, 9 por ciento 3er. T). B: A/PF secundarias (32 años, 164 gestas, 96 pérdidas espontáneas, 57 por ciento 1er. T, 35 por ciento 2do. T, 8 por ciento 3er. T). C: misceláneas (30 años, 35 gestas, 21 pérdidas espontáneas, 45 por ciento 1er. T, 15 por ciento 2do. T, 40 por ciento 3er. T). Hallamos anormalidades de la hemostasia en los 3 grupos estudiados (A: 94 por ciento, B: 92 por ciento y C: 100 por ciento), como defectos aislados (36 por ciento) o combinados (58 por ciento). Se detallan los porcentajes de anomalías encontradas en las pacientes que han concluído la evaluación.


Asunto(s)
Humanos , Femenino , Embarazo , Adulto , Antitrombina III , Inhibidores de Factor de Coagulación Sanguínea , Fibrinógeno , Hemostasis , Tamización de Portadores Genéticos , Complicaciones Hematológicas del Embarazo , Proteína C , Proteína S , Protrombina/genética , Pruebas de Función Plaquetaria , Aborto Espontáneo/etiología , Factor VIII , Factor de von Willebrand
20.
Bol. Acad. Nac. Med. B.Aires ; 77(2): 240-6, ene.-jun. 1999. tab
Artículo en Español | BINACIS | ID: bin-12527

RESUMEN

Se estudiaron 91 embarazadas normales. Hubo 2 casos de plaquetopenia leve en el 2º trimestre y 6 en el 3º; se observó aumento del factor von Willebrand. El Fibrinógeno aumentó hacia el tercer trimestre. La proteína S (PS) biológica e inmunológica libre disminuyeron. La Resistencia a la Proteína C Activada observó un acortamiento entre


Asunto(s)
Humanos , Femenino , Embarazo , Adulto , Hemostasis , Complicaciones Hematológicas del Embarazo , Antitrombina III , Proteína C , Proteína S , Inhibidores de Factor de Coagulación Sanguínea , Fibrinógeno , Pruebas de Función Plaquetaria , Tamización de Portadores Genéticos , Protrombina/genética , Aborto Espontáneo/etiología , Factor VIII , Factor de von Willebrand
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...