Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
J Thromb Haemost ; 21(2): 237-254, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36700498

RESUMEN

BACKGROUND: Severe high-molecular-weight kininogen (HK) deficiency is a poorly studied autosomal recessive contact system defect caused by pathogenic, biallelic KNG1 variants. AIM: We performed the first comprehensive analysis of diagnostic, clinical, genetic, and epidemiological aspects of HK deficiency. METHODS: We collected clinical information and blood samples from a newly detected HK-deficient individual and from published cases identified by a systematic literature review. Activity and antigen levels of coagulation factors were determined. Genetic analyses of KNG1 and KLKB1 were performed by Sanger sequencing. The frequency of HK deficiency was estimated considering truncating KNG1 variants from GnomAD. RESULTS: We identified 48 cases of severe HK deficiency (41 families), of these 47 have been previously published (n = 19 from gray literature). We genotyped 3 cases and critically appraised 10 studies with genetic data. Ten HK deficiency-causing variants (one new) were identified. All of them were truncating mutations, whereas the only known HK amino acid substitution with a relevant phenotype instead causes hereditary angioedema. Conservative estimates suggest an overall prevalence of severe HK deficiency of approximately one case per 8 million population, slightly higher in Africans. Individuals with HK deficiency appeared asymptomatic and had decreased levels of prekallikrein and factor XI, which could lead to misdiagnosis. CONCLUSION: HK deficiency is a rare condition with only few known pathogenic variants. It has an apparently good prognosis but is prone to misdiagnosis. Our understanding of its clinical implications is still limited, and an international prekallikrein and HK deficiency registry is being established to fill this knowledge gap.


Asunto(s)
Quininógeno de Alto Peso Molecular , Precalicreína , Quininógeno de Alto Peso Molecular/genética , Quininógeno de Alto Peso Molecular/metabolismo , Precalicreína/genética , Precalicreína/metabolismo , Prevalencia , Factores de Coagulación Sanguínea
3.
Haemophilia ; 24(6): e395-e401, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30144214

RESUMEN

INTRODUCTION: The study is the first application of the Principles of Haemophilia Care for Europe (PHCE) in other regions of the world, specifically in Latin America. OBJECTIVE: To identify strengths in the care of haemophilia, and the aspects that should be improved. METHODS: The information was obtained through a questionnaire designed according to the PHCE and answered by specialists in mid-2016. The countries included were as follows: Argentina, Brazil, Chile, Colombia, Costa Rica, El Salvador, Mexico, Panama, Dominican Republic and Venezuela. RESULTS: In most countries, there is a central organization for haemophilia care supported by local groups. The existence of a national registry of people with haemophilia (PWH) was verified in eight countries. Centres of integrated care are located in large cities. In the majority of countries, there was no evidence of the participation of multiple actors in the decision-making. The supply of factor concentrates presents constraints, although it is reported as adequate in half of the countries. In most countries, home treatment is available under special conditions. In most countries, there are restrictions on the use of prophylaxis. The coordination of specialized and emergency services depends on each centre. Unrestricted treatment of inhibitors is performed in most countries. In all countries, there are human resources training programmes; however, clinical and health services researches are not widely developed. CONCLUSION: The study identifies the initial situation of principles of care, as well as the alternatives that must be implemented to achieve improvements in the quality of life of PWH in the region.


Asunto(s)
Hemofilia A , Atención al Paciente/estadística & datos numéricos , Relaciones Comunidad-Institución , Servicios Médicos de Urgencia , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Humanos , América Latina , Educación del Paciente como Asunto , Calidad de Vida , Sistema de Registros , Encuestas y Cuestionarios
4.
Semin Thromb Hemost ; 39(6): 684-92, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23929306

RESUMEN

Paradoxically, there are reports of thrombotic events for some rare bleeding disorders associated with significant bleeding tendency. Afibrinogenemia, factor (F) VII, or FXI deficiencies are those most commonly associated with venous or arterial thrombosis. Pathogenesis is multifactorial and the main conditions associated with this complication relate to the coexistence of inherited or acquired thrombotic risk factors linked to certain specific characteristics of the underlying defect. Patients with afibrinogenemia can develop severe, spontaneous, or recurrent thromboembolic disease. Up to 20% of congenital dysfibrinogenemia patients show predisposition to thrombosis. Thrombotic episodes, particularly deep vein thrombosis, have been reported in 3 to 4% FVII deficient patients, even those who were severely affected. These events have been reported either after infusion of plasma derived FXI concentrate or recombinant activated FVII in FXI deficient patients. So, in addition to factor level, replacement therapy must be individualized and should take into account past personal or family history of bleeding and thrombosis, and other prothrombotic risk factors. Treatment of thrombosis represents a challenge. For mild factor deficiencies, antithrombotic prophylaxis must be considered with or without concomitant use of replacement therapy. For all patients, it is also recommended to control known cardiovascular disease risk factors.


Asunto(s)
Trastornos de la Coagulación Sanguínea/complicaciones , Guías de Práctica Clínica como Asunto , Trombosis/complicaciones , Trombosis/terapia , Afibrinogenemia/complicaciones , Deficiencia del Factor VII/complicaciones , Deficiencia del Factor XI/complicaciones , Humanos , Factores de Riesgo , Trombosis/diagnóstico
5.
Thromb Haemost ; 109(2): 238-47, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23238632

RESUMEN

Patients with inherited factor VII (FVII) deficiency display different clinical phenotypes requiring ad hoc management. This study evaluated treatments for spontaneous and traumatic bleeding using data from the Seven Treatment Evaluation Registry (STER). One-hundred one bleeds were analysed in 75 patients (41 females; FVII coagulant activity <1-20%). Bleeds were grouped as haemarthroses (n=30), muscle/subcutaneous haematomas (n=16), epistaxis (n=12), gum bleeding (n=13), menorrhagia (n=16), central nervous system (CNS; n=9), gastrointestinal (GI; n=2) and other (n=3). Of 93 evaluable episodes, 76 were treated with recombinant, activated FVII (rFVIIa), eight with fresh frozen plasma (FFP), seven with plasma-derived FVII (pdFVII) and two with prothrombin-complex concentrates. One-day replacement therapy resulted in very favourable outcomes in haemarthroses, and was successful in muscle/subcutaneous haematomas, epistaxis and gum bleeding. For menorrhagia, single- or multiple-dose schedules led to favourable outcomes. No thrombosis occurred; two inhibitors were detected in two repeatedly treated patients (one post-rFVIIa, one post-pdFVII). In FVII deficiency, most bleeds were successfully treated with single 'intermediate' doses (median 60 µg/kg) of rFVIIa. For the most severe bleeds (CNS, GI) short- or long-term prophylaxis may be optimal.


Asunto(s)
Factores de Coagulación Sanguínea/administración & dosificación , Transfusión de Componentes Sanguíneos , Coagulantes/administración & dosificación , Deficiencia del Factor VII/terapia , Factor VIIa/administración & dosificación , Hemorragia/tratamiento farmacológico , Adolescente , Adulto , Anciano , Factores de Coagulación Sanguínea/efectos adversos , Transfusión de Componentes Sanguíneos/efectos adversos , Niño , Preescolar , Coagulantes/efectos adversos , Esquema de Medicación , Deficiencia del Factor VII/complicaciones , Deficiencia del Factor VII/diagnóstico , Deficiencia del Factor VII/genética , Factor VIIa/efectos adversos , Femenino , Hemorragia/genética , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Proteínas Recombinantes/administración & dosificación , Sistema de Registros , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
6.
Blood Cells Mol Dis ; 50(3): 177-81, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23266225

RESUMEN

Routine coagulation tests on a 14year-old male with frequent epistaxis showed a prolonged thrombin time together with diminished functional (162mg/dl) and gravimetric (122mg/dl) fibrinogen concentrations. His father showed similar aberrant results and sequencing of the three fibrinogen genes revealed a novel heterozygous nonsense mutation in the FGB gene c.1105C>T, which converts the codon for residue Bß 339Q to stop, causing deletion of Bß chain residues 339-461. Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and RP-HPLC (reverse-phase high-pressure liquid chromatography) of purified fibrinogen showed only normal Aα, Bß, and γ chains, indicating that molecules with the truncated 37,990Da ß chain were not secreted into plasma. Functional analysis showed impaired fibrin polymerization, fibrin porosity, and elasticity compared to controls. By laser scanning confocal microscopy the patient's fibers were slightly thinner than normal. Electrospray ionization mass spectrometry (ESI MS) presented normal sialylation of the oligosaccharide chains, and liver function tests showed no evidence of liver dysfunction that might explain the functional abnormalities.


Asunto(s)
Afibrinogenemia/genética , Codón sin Sentido , Fibrinógeno/genética , Mutación , Adolescente , Afibrinogenemia/sangre , Afibrinogenemia/diagnóstico , Coagulación Sanguínea , Fibrina/metabolismo , Fibrinógeno/metabolismo , Humanos , Masculino , Multimerización de Proteína
7.
Hematology ; 17 Suppl 1: S141-3, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22507803

RESUMEN

Hemophilia is a chronic and inherited X-linked bleeding disorder that requires life-long medical care. Hemophilia treatment is costly and complex partly because of the cost of the factor concentrates used in replacement therapy. However, the management of hemophilia is not based solely on achieving access to better treatment with safe factor concentrates; it also includes accurately diagnosing the disorder and providing specialized comprehensive care by a multidisciplinary team of specialists trained in hemophilia management. Comprehensive care for the person with hemophilia is defined as the continuous supervision of all medical and psychological aspects affecting the patient and his family and it demands the establishment of specialized centers, called Hemophilia Treatment Centers. The services that should be offered by a comprehensive hemophilia healthcare center are diverse and the multidisciplinary team should be coordinated preferably by a hematologist with the participation of other health professionals. It has been demonstrated that the benefits of establishing hemophilia centers are observed even in developing countries and that changes can be achieved when resources are re-organized, especially when education and training are provided at all levels. To reach these objectives, it is essential to have the participation of the patient and family members, and to strive to obtain the financial and legislative support from the State or Government in order to achieve a national comprehensive care program contemplating all the aspects needed for improving the quality of life for the community of patients with hemophilia and other bleeding disorders.


Asunto(s)
Atención Integral de Salud/métodos , Hemofilia A/terapia , Hospitales Especializados , Humanos , Cuerpo Médico
8.
Hematology ; 17 Suppl 1: S156-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22507808

RESUMEN

Inherited deficiencies of blood coagulation factors are usually associated with lifelong bleeding tendency. In addition to Haemophilias A and B and von Willebrand disease, congenital deficiencies of such factors as fibrinogen, prothrombin (FII)), FV, FVII, FX, FXI, FXIII, and combined deficiencies occur and can lead to a diversity of clinical conditions. Paradoxically, for some of these disorders associated with significant bleeding tendency there are reports of thrombotic events, both arterial and venous. Thrombosis in hemophilia patients has a multifactorial pathogenesis and the main conditions associated with this complication are the use of long-term central venous catheters, intensive replacement therapy usually in the setting of surgical procedures, the use of bypassing agents or the coexistence of acquired or inherited prothrombotic risk factors. Regarding other rare bleeding disorders, thrombotic phenomena has been described particularly in patients with afibrinogenemia, FXI and FVII deficiency and the events can occur even in young patients, in the presence of concomitant risk factors or spontaneously. Replacement therapy must be individualized and should take into account past history of haemostatic challenges, family history of bleeding and thrombosis, just like the level of factor. For mild deficiencies when patients are asymptomatic the use of antithrombotic prophylaxis must be considered with or without concomitant use of replacement therapy. In patients with history of thrombosis it may be helpful to perform a thrombophilia screening to exclude coexisting prothrombotic defects and for all patients it is recommended to control known cardiovascular disease risk factors.


Asunto(s)
Hemorragia/complicaciones , Trastornos Hemorrágicos/complicaciones , Enfermedades Raras/complicaciones , Trombosis/complicaciones , Trastornos de la Coagulación Sanguínea Heredados/complicaciones , Trastornos de la Coagulación Sanguínea Heredados/metabolismo , Deficiencia del Factor VII/complicaciones , Deficiencia del Factor VII/metabolismo , Deficiencia del Factor XI/complicaciones , Deficiencia del Factor XI/metabolismo , Fibrinógeno/metabolismo , Hemorragia/metabolismo , Trastornos Hemorrágicos/metabolismo , Humanos , Enfermedades Raras/metabolismo , Trombosis/metabolismo
9.
Srp Arh Celok Lek ; 138 Suppl 1: 39-42, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20229681

RESUMEN

One of the best procedures to prevent haemarthrosis in haemophilia has been radioactive synovectomy (radiosynoviorthesis). Since 1976 we have performed 119 radiosynoviortheses in 110 patients, aged from 3 to 40 years (mean 10), and of whom 71 were under 12 years of age. The knees were injected in 71, elbow in 29, ankles in 16, and shoulders in 3 cases. Clinical results of the procedure gave excellent results 80% of patients with no further bleeding. In the case of failure a reinjection can be given in the same joint at a 6 month interval. One of the criticisms against this method is possible chromosomal damage. In our centre, 4 studies have been made in order to see whether these changes are permanent, but all have demonstrated that chromosomal changes are reversible. Radioactive material used in 2 studies was Au-189. In 1978, 354 metaphases were studied with 61 ruptures, with 17.23% non-premalignant and 6 structural changes considered premalignant (1.69%). Further study was done in 1982, in the same group of patients with the result of 21 ruptures (3.34%) and no structural changes. The third study was performed in 13 patients that sustained radiosynoviorthesis with Re-186 in 1991. We compared the chromosomal study before and 6 months after the radioactive material injection and the results confirmed that changes appeared equally in non-irradiated and radiated patients and disappeared with time, never reaching the dangerous zone of 2%. In the group treated with Re-186 we studied an additional number of 130 metaphases with identical results and no structural changes. A study performed before and after radiosynoviorthesis with Y-90 revealed no premalignant changes. It seems than radiosynoviorthesis is safe and highly beneficial to haemophilic patients.


Asunto(s)
Hemartrosis/radioterapia , Hemofilia A/complicaciones , Radioisótopos/administración & dosificación , Membrana Sinovial/efectos de la radiación , Adolescente , Adulto , Niño , Preescolar , Aberraciones Cromosómicas/efectos de la radiación , Hemartrosis/etiología , Humanos , Inyecciones Intraarticulares , Radioisótopos/efectos adversos , Adulto Joven
10.
Medicina (B Aires) ; 68(3): 227-42, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18689157

RESUMEN

The Committee of Latin America on the Therapeutics of Inhibitor Groups (CLOTTING) is composed of a number of hemophilia specialists from Latin America. The group aims to encourage the adoption of a good standard of care for Latin American patients with hemophilia. The occurrence of inhibitors in patients with hemophilia poses clinical challenges, and it is estimated that between 1000 and 3000 patients in Latin America are affected by hemophilia with inhibitors. There is an urgent need to establish a regional consensus and clinical guidelines for the diagnosis and treatment of these patients. We present an extensive review based on best current clinical practice and published literature, as seen from a Latin American perspective, taking into account the variable nature of hemophilia care available in the various countries in this Region.


Asunto(s)
Coagulantes/administración & dosificación , Factor IX/administración & dosificación , Factor VIII/administración & dosificación , Hemofilia A/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Adulto , Factores de Coagulación Sanguínea/administración & dosificación , Niño , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Quimioterapia Combinada , Hemofilia A/clasificación , Hemofilia A/diagnóstico , Hemofilia B/clasificación , Hemofilia B/diagnóstico , Humanos , América Latina , Guías de Práctica Clínica como Asunto , Índice de Severidad de la Enfermedad
11.
Medicina (B.Aires) ; 68(3): 227-242, mayo-jun. 2008. tab
Artículo en Inglés | LILACS | ID: lil-633546

RESUMEN

The Committee of Latin America on the Therapeutics of Inhibitor Groups (CLOTTING) is composed of a number of hemophilia specialists from Latin America. The group aims to encourage the adoption of a good standard of care for Latin American patients with hemophilia. The occurrence of inhibitors in patients with hemophilia poses clinical challenges, and it is estimated that between 1000 and 3 000 patients in Latin America are affected by hemophilia with inhibitors. There is an urgent need to establish a regional consensus and clinical guidelines for the diagnosis and treatment of these patients. We present an extensive review based on best current clinical practice and published literature, as seen from a Latin American perspective, taking into account the variable nature of hemophilia care available in the various countries in this Region.


El Comité Latinoamericano sobre la Terapéutica de Personas con Inhibidores (CLOTTING) está compuesto por un grupo de especialistas en hemofilia de Latinoamérica. El objetivo del grupo es promover la adopción de un estándar de tratamiento óptimo para los pacientes con hemofilia en Latinoamérica. La prevalencia de inhibidores en pacientes con hemofilia en Latinoamérica determina desafíos clínicos y se estima que de 1000 a 3000 pacientes en esta región están afectados con hemofilia e inhibidores. Existe una necesidad urgente de establecer un consenso regional y guías clínicas para el diagnóstico y tratamiento de estos pacientes. Nosotros presentamos una revisión exhaustiva basada en las mejores prácticas clínicas vigentes y en los datos publicados en la literatura, con una perspectiva latinoamericana, tomando en cuenta la variabilidad existente de los tratamientos de la hemofilia disponibles en los diferentes países de esta Región.


Asunto(s)
Adulto , Niño , Humanos , Coagulantes/administración & dosificación , Factor IX/administración & dosificación , Factor VIII/administración & dosificación , Hemofilia A/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Factores de Coagulación Sanguínea/administración & dosificación , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Quimioterapia Combinada , Hemofilia A/clasificación , Hemofilia A/diagnóstico , Hemofilia B/clasificación , Hemofilia B/diagnóstico , América Latina , Guías de Práctica Clínica como Asunto , Índice de Severidad de la Enfermedad
12.
Arch. venez. farmacol. ter ; 27(1): 86-89, 2008. tab
Artículo en Español | LILACS | ID: lil-517081

RESUMEN

Comparar la eficacia de dos presentaciones de clopidogrel en la Inhibición de la Agregación Plaquetaria (IAP). Estudio prospectivo, secuencial, en 22 voluntarios sanos, 8 mujeres y 14 hombres, con edades entre 21 y 55 años, mediana 30 años. Se les realizó agregación plaquetaria (AP), con ADP 4µM y 10µM, para excluir patología plaquetaria. Todos recibieron clopidogrel (Cirgrel®), 75 mg diarios por 15 días y se les realizó AP. Posteriormente a los 30 días, recibieron clopidogrel (Plavix®) por 15 días y se les realizó AP para comparar los efectos. Los promedios de AP basales fueron: 66,45±7.00% y 66,45±7.88%, respectivamente para cada concentración utilizada. Los promedios de IAP fueron: con clopidogrel (Cirgrel®), 30,21±19.51% y 28,96±21.75%, y con clopidogrel (Plavix®) 35,64±24.84% y 30,91±27.4% (p=0.45 y 0.81), respectivamente para cada concentración de ADP. No hubo diferencias significativas, por lo que podemos concluir que estas dos presentaciones de clopidogrel son equivalentes.


Asunto(s)
Humanos , Masculino , Adulto , Femenino , Persona de Mediana Edad , Enfermedades Cardiovasculares/prevención & control , Agregación Plaquetaria , Química Farmacéutica/métodos
13.
Thromb Res ; 118(5): 637-50, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16406498

RESUMEN

Fibrinogen Guarenas is a dysfibrinogenemia with a nonsense mutation at G4731T that causes an Aalpha-chain truncation at Ser 466. This abnormal fibrinogen is associated with a bleeding diathesis, severe in the proposita and mild in one brother, even though the fibrinogen levels in plasma are normal. All other family members are asymptomatic. Fibrinogens from the proposita and one family member, the mother of the proposita, both heterozygous for the mutation, were studied. Turbidity curves of fibrin polymerization showed that the lateral association of protofibrils was impaired and the maximum rate of polymerization was slightly diminished. The binding of albumin to fibrinogen was increased compared to control due to the presence of a free sulfhydryl group because of the missing disulphide bridge between Aalpha-Cys 442-472 in the mutated molecules. The abnormal fibrinogen formed much less alpha-polymer, and gamma-dimer formation was delayed compared to the control. Plasminogen activation by t-PA in the presence of fibrin was decreased. When Guarenas clots were perfused with fibrinolytic enzymes, clot degradation was retarded. Clot structure studied by confocal 3D microscopy showed that the fibrin network was dense, made up of thin and highly branched fibers, which accounted for the decreased flow rates by buffer permeation and increased rigidity of the fibrin clots, measured using a torsion pendulum. It seems that the increased clot rigidity, decreased porosity, hypofibrinolysis and t-PA induced fibrinolysis, by itself are not necessarily associated with thrombotic disorders in dysfibrinogenemia.


Asunto(s)
Afibrinogenemia/genética , Fibrinógenos Anormales/química , Fibrinógenos Anormales/inmunología , Codón sin Sentido/genética , Fibrinógenos Anormales/genética , Humanos , Relación Estructura-Actividad
14.
Blood ; 107(5): 1903-7, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16304051

RESUMEN

Mutations in LMAN1 (ERGIC-53) or MCFD2 cause combined deficiency of factor V and factor VIII (F5F8D). LMAN1 and MCFD2 form a protein complex that functions as a cargo receptor ferrying FV and FVIII from the endoplasmic reticulum to the Golgi. In this study, we analyzed 10 previously reported and 10 new F5F8D families. Mutations in the LMAN1 or MCFD2 genes accounted for 15 of these families, including 3 alleles resulting in no LMAN1 mRNA accumulation. Combined with our previous reports, we have identified LMAN1 or MCFD2 mutations as the causes of F5F8D in 71 of 76 families. Among the 5 families in which no mutations were identified, 3 were due to misdiagnosis, with the remaining 2 likely carrying LMAN1 or MCFD2 mutations that were missed by direct sequencing. Our results suggest that mutations in LMAN1 and MCFD2 may account for all cases of F5F8D. Immunoprecipitation and Western blot analysis detected a low level of LMAN1-MCFD2 complex in lymphoblasts derived from patients with missense mutations in LMAN1 (C475R) or MCFD2 (I136T), suggesting that complete loss of the complex may not be required for clinically significant reduction in FV and FVIII.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Portadoras/genética , Deficiencia del Factor V/genética , Hemofilia A/genética , Lectinas de Unión a Manosa/genética , Proteínas de la Membrana/genética , Mutación Missense , Mutación Puntual , Alelos , Western Blotting/métodos , Proteínas Portadoras/metabolismo , Análisis Mutacional de ADN/métodos , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Factor V/metabolismo , Deficiencia del Factor V/metabolismo , Factor VIII/metabolismo , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Hemofilia A/metabolismo , Humanos , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Transporte de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Transporte Vesicular
15.
Br J Haematol ; 130(4): 620-7, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16098078

RESUMEN

Mutation of the non-muscle myosin heavy chain type II-A results in MYH9-related hereditary macrothrombocytopenia (HMTC), including four autosomal dominant platelet disorders: May-Hegglin anomaly (MHA), Sebastian (SBS), Fechtner (FS) and Epstein (EPS) syndrome. Denaturing high-performance liquid chromatography (DHPLC) was optimised for rapid screening of the seven exons harbouring all but one of the previously reported mutations of MYH9. Individuals from 13 families with phenotypes suggestive of MYH9-related HMTC were screened for mutations by DHPLC followed by direct sequencing of samples with aberrant column retention time. Mutations were identified in all 13 families. Six distinct missense heterozygous mutations were found in 10 families, including six families with MHA or SBS (E1841K, D1424N), three families with FS (R702H, R1165C, and D1424Y), and one family with EPS (S96L). A truncating mutation (R1933X) was found in three MHA families. A review of all published mutations suggests that mutation in the C-terminal coiled coil region or truncation of the tailpiece is associated with haematological-only phenotype, while mutation of the head ATPase domain frequently is associated with nephropathy and/or hearing loss. Mutations of other regions have intermediate expression of non-haematological characteristics. Further study is required to confirm these associations and understand the molecular basis for this genotype-phenotype relationship.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Trombocitopenia/genética , Cromatografía Líquida de Alta Presión/métodos , Análisis Mutacional de ADN , Genes Dominantes , Genotipo , Pérdida Auditiva/genética , Heterocigoto , Humanos , Enfermedades Renales/genética , Mutación Missense , Fenotipo
16.
Am J Trop Med Hyg ; 73(2): 381-5, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16103609

RESUMEN

Platelet dysfunction was detected in six children with purpura and eosinophilia. We conducted clinical evaluations, hematologic and platelet function tests, clotting studies (bleeding time, prothrombin time, partial thromboplastin time, thrombin time, factor XIII, factor VIII, and von Willebrand factor), assays for IgG and IgM antibodies to platelets, and a search for stool parasites. Mild bleeding phenomena (ecchymoses, petechiae, epistaxis, and gingival) were transient. All children showed intestinal parasites and marked eosinophilia (mean count = 2,615.2 cells/muL, 95% confidence interval = 1,259.6-5,429.8). Main abnormalities included prolonged bleeding times (50%) and defective aggregation with collagen (100%) adrenaline (66%), or ADP (66%). Antibodies to platelets were not detected. Anti-parasite therapy reversed the hemorrhagic manifestations and normalized eosinophil counts and platelet alterations. No relationship could be established between excess eosinophils, intensity of bleeding, or type and degree of platelet abnormalities. Thrombocytopathic features mimicked the intrinsic defect of storage pool disease. The possible pathogenic roles of eosinophilia and parasitism are reviewed. This is the first report of this pathologic combination in Latin American children.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/complicaciones , Eosinofilia/complicaciones , Parasitosis Intestinales/complicaciones , Pruebas de Coagulación Sanguínea , Trastornos de las Plaquetas Sanguíneas/patología , Niño , Preescolar , Eosinofilia/patología , Femenino , Humanos , Parasitosis Intestinales/parasitología , Masculino , Pruebas de Función Plaquetaria , Púrpura Trombocitopénica/complicaciones , Púrpura Trombocitopénica/patología , Síndrome , Venezuela
17.
Nat Genet ; 34(2): 220-5, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12717434

RESUMEN

Mutations in LMAN1 (also called ERGIC-53) result in combined deficiency of factor V and factor VIII (F5F8D), an autosomal recessive bleeding disorder characterized by coordinate reduction of both clotting proteins. LMAN1 is a mannose-binding type 1 transmembrane protein localized to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC; refs. 2,3), suggesting that F5F8D could result from a defect in secretion of factor V and factor VIII (ref. 4). Correctly folded proteins destined for secretion are packaged in the ER into COPII-coated vesicles, which subsequently fuse to form the ERGIC. Secretion of certain abundant proteins suggests a default pathway requiring no export signals (bulk flow; refs. 6,7). An alternative mechanism involves selective packaging of secreted proteins with the help of specific cargo receptors. The latter model would be consistent with mutations in LMAN1 causing a selective block to export of factor V and factor VIII. But approximately 30% of individuals with F5F8D have normal levels of LMAN1, suggesting that mutations in another gene may also be associated with F5F8D. Here we show that inactivating mutations in MCFD2 cause F5F8D with a phenotype indistinguishable from that caused by mutations in LMAN1. MCFD2 is localized to the ERGIC through a direct, calcium-dependent interaction with LMAN1. These findings suggest that the MCFD2-LMAN1 complex forms a specific cargo receptor for the ER-to-Golgi transport of selected proteins.


Asunto(s)
Proteínas Portadoras/genética , Deficiencia del Factor V/genética , Hemofilia A/genética , Hemorragia/genética , Lectinas de Unión a Manosa/genética , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Transporte Biológico Activo/genética , Retículo Endoplásmico/metabolismo , Deficiencia del Factor V/metabolismo , Femenino , Aparato de Golgi/metabolismo , Células HeLa , Hemofilia A/metabolismo , Hemorragia/etiología , Hemorragia/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Homología de Secuencia de Aminoácido , Transfección , Proteínas de Transporte Vesicular
18.
Centro méd ; 47(1): 13-16, mayo 2002. tab
Artículo en Español | LILACS | ID: lil-393058

RESUMEN

Se hace una evaluación de la influencia que ha tenido la infeción por el virus VIH en la torpidez de la evolución de hemartrosis agudas y crónica. También la proporción de tratamiento


Asunto(s)
Humanos , Hemartrosis , Hemofilia A , VIH , Venezuela
19.
Thromb Haemost ; 87(2): 282-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11858488

RESUMEN

Prothrombin Perijá is a dysprothrombin derived from a homozygous patient that manifests low thrombin activity upon activation in a one-stage assay. Purified prothrombin Perijá showed normal appearance on SDS-PAGE. and was cleaved normally to form alpha-thrombin by the prothrombinase complex. The activated form, thrombin Perijá, however, did not show any proteolytic activity towards native substrates, fibrinogen, protein C or various synthetic substrates for alpha-thrombin, but it was able to bind to antithrombin III, although the binding capacity was markedly reduced even in the presence of heparin. Thrombin Perijá showed full reactivity toward a small inhibitor, DFP, indicating that the molecular defect is in the substrate binding site in the thrombin molecule but not in the active site itself. By DNA sequence analysis of the patient prothrombin gene, we identified a G to C mutation at nucleotide 20016 in exon 14, which predicts a Gly-548 to Ala substitution in the prothrombin Perijá molecule. The structural modeling of thrombin Perijá suggests that Ala-548 is located close to the limb of the cavity wall of the substrate binding pocket, and that the methyl group blocks protrusion of the guanidino group of Arg into the cavity. This steric hindrance may well inhibit the access of Arg-containing substrates to the catalytic Ser-525 leading to the loss of proteolytic activity.


Asunto(s)
Sustitución de Aminoácidos , Protrombina/química , Trombina/química , Alanina/química , Antitrombina III/metabolismo , Arginina/química , Sitios de Unión , Dominio Catalítico , Codón/genética , Análisis Mutacional de ADN , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Exones/genética , Factor V/metabolismo , Factor Xa/metabolismo , Glicina/química , Humanos , Modelos Moleculares , Ácidos Pipecólicos/farmacología , Polimorfismo Conformacional Retorcido-Simple , Conformación Proteica , Protrombina/genética , Serina/química , Relación Estructura-Actividad , Sulfonamidas , Trombina/antagonistas & inhibidores , Trombina/metabolismo
20.
Arch. Hosp. Vargas ; 31(1/2): 75-82, ene.-jun. 1989. tab, ilus
Artículo en Español | LILACS | ID: lil-86860

RESUMEN

Se reporta el caso clínico de un paciente de sexo masculino de 36 años, con historia de diátesis hemorrágica de un año de evolución, sin antecedentes personales o familiares de hemorragia y disminución de los valores de factor VIII:C, factor vWAg y de la actividad Cofactor de Ristocetina. Los estudios realizados con el plasma del paciente y con las fracciones purificadas del mismo, permiten concluir que se trata de una enfermedad de von Willebrand adquirida por acción inhibidora de la paraproteína IgA-K, caracteristica de un mieloma múltiple diagnosticado durante la evaluación de su problema hemorrágico. La acción inhibidora fue dirigida principalmente a la actividad de Cofactor de Ristocetina. El cuadro hemorrágico se correlacionó con los valores elevados de proteínas séricas caracteristicos de su enfermedad base, mejorando en forma temporal con plasmaféresis


Asunto(s)
Adulto , Humanos , Masculino , Coagulación Sanguínea/métodos , Trastornos Hemorrágicos/diagnóstico , Hemostasis/métodos , Mieloma Múltiple/complicaciones , Enfermedades de von Willebrand/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...