Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Vet Clin Pathol ; 52(4): 569-575, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37438861

RESUMEN

Bernard-Soulier syndrome (BSS), also known as hemorrhagiparous thrombocytic dystrophy (OMIA 002207-9615), is a rare defect in platelet function recognized in both dogs and humans. It is caused by a deficiency in glycoprotein 1b-IX-V, the platelet surface protein which acts as a receptor for the von Willebrand factor. The characteristic features of BSS in humans and dogs include macrothrombocytes and mild-to-moderate thrombocytopenia with a bleeding tendency. This condition has previously been reported in European Cocker Spaniel dogs; however, the results of platelet function tests in these animals have not been reported. This case report describes a European Cocker Spaniel dog with spontaneously occurring Bernard-Soulier syndrome and the results of point-of-care platelet function tests, including a prolonged buccal mucosal bleeding time (>8 min), prolongation (>300 s) of PFA-200 COL/ADP, COL/EPI, and P2Y closure times, and reduced aggregation (15%-48%) with Plateletworks ADP, but with normal aggregation (92%) with Plateletworks AA. This is the first description of the results of platelet function tests in canine Bernard-Soulier syndrome.


Asunto(s)
Síndrome de Bernard-Soulier , Enfermedades de los Perros , Trastornos Hemorrágicos , Humanos , Perros , Animales , Síndrome de Bernard-Soulier/diagnóstico , Síndrome de Bernard-Soulier/veterinaria , Síndrome de Bernard-Soulier/metabolismo , Sistemas de Atención de Punto , Plaquetas/metabolismo , Hemostasis , Complejo GPIb-IX de Glicoproteína Plaquetaria , Trastornos Hemorrágicos/veterinaria , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/metabolismo
3.
Platelets ; 29(8): 827-833, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30332551

RESUMEN

Mutations in the GP1BA gene have been associated with platelet-type von Willebrand disease and Bernard-Soulier syndrome. Here, we report a novel GP1BA mutation in a family with autosomal dominant macrothrombocytopenia and mild bleeding. We performed analyses of seven family members. Using whole-exome sequencing of germline DNA samples, we identified a heterozygous single-nucleotide change in GP1BA (exone2:c.176T>G), encoding a p.Leu59Arg substitution in the N-terminal domain, segregating with macrothrombocytopenia. This variant has not been previously reported. We also analysed the structure of the detected sequence variant in silico. In particular, we used the crystal structure of the human platelet receptor GP Ibα N-terminal domain. Replacement of aliphatic amino-acid Leu 59 with charged, polar and larger arginine probably disrupts the protein structure. An autosomal dominant mode of inheritance, a family history of mild bleeding episodes, aggregation pattern in affected individuals together with evidence of mutation occurring in part of the GP1BA gene encoding the leucine-rich repeat region suggest a novel variant causing monoallelic Bernard-Soulier syndrome.


Asunto(s)
Síndrome de Bernard-Soulier/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Mutación Puntual , Síndrome de Bernard-Soulier/metabolismo , Cristalografía por Rayos X , Femenino , Humanos , Masculino , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Dominios Proteicos
5.
Stem Cell Res ; 20: 10-13, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395735

RESUMEN

Bernard Soulier Syndrome (BSS) is a rare autosomal platelet disorder characterized by mutations in the von Willebrand factor platelet receptor complex GPIb-V-IX. In this work we have generated an induced pluripotent stem cell (BSS3-PBMC-iPS4F8) from peripheral blood mononuclear cells of a BSS patient with a p.Phe55Ser mutation in the GPIX gene. Characterization of BSS3-PBMC-iPS4F8 showed that these cells maintained the original mutation present in the BSS patient, expressed pluripotent stem cell markers and were able to differentiate into the three germline layers. This new iPSC line will contribute to better understand the biology of BSS disease.


Asunto(s)
Síndrome de Bernard-Soulier/patología , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Secuencia de Bases , Síndrome de Bernard-Soulier/genética , Síndrome de Bernard-Soulier/metabolismo , Diferenciación Celular , Línea Celular , Reprogramación Celular , Análisis Mutacional de ADN , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Femenino , Homocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Polimorfismo de Nucleótido Simple , Secuencias Repetidas en Tándem/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Haematologica ; 101(8): 896-908, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27478199

RESUMEN

Constitutional thrombocytopenias result from platelet production abnormalities of hereditary origin. Long misdiagnosed and poorly studied, knowledge about these rare diseases has increased considerably over the last twenty years due to improved technology for the identification of mutations, as well as an improvement in obtaining megakaryocyte culture from patient hematopoietic stem cells. Simultaneously, the manipulation of mouse genes (transgenesis, total or conditional inactivation, introduction of point mutations, random chemical mutagenesis) have helped to generate disease models that have contributed greatly to deciphering patient clinical and laboratory features. Most of the thrombocytopenias for which the mutated genes have been identified now have a murine model counterpart. This review focuses on the contribution that these mouse models have brought to the understanding of hereditary thrombocytopenias with respect to what was known in humans. Animal models have either i) provided novel information on the molecular and cellular pathways that were missing from the patient studies; ii) improved our understanding of the mechanisms of thrombocytopoiesis; iii) been instrumental in structure-function studies of the mutated gene products; and iv) been an invaluable tool as preclinical models to test new drugs or develop gene therapies. At present, the genetic determinants of thrombocytopenia remain unknown in almost half of all cases. Currently available high-speed sequencing techniques will identify new candidate genes, which will in turn allow the generation of murine models to confirm and further study the abnormal phenotype. In a complementary manner, programs of random mutagenesis in mice should also identify new candidate genes involved in thrombocytopenia.


Asunto(s)
Trombocitopenia/etiología , Trombocitopenia/metabolismo , Animales , Autoantígenos/metabolismo , Síndrome de Bernard-Soulier/etiología , Síndrome de Bernard-Soulier/metabolismo , Plaquetas/metabolismo , Diferenciación Celular/genética , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Yoduro Peroxidasa/metabolismo , Proteínas de Unión a Hierro/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Trombocitopenia/diagnóstico , Trombopoyesis , Factores de Transcripción/metabolismo , Síndrome de Wiskott-Aldrich/etiología , Síndrome de Wiskott-Aldrich/metabolismo
7.
Stem Cell Res ; 16(3): 692-5, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27346198

RESUMEN

We generated an induced pluripotent stem cell (iPSC) line from a Bernard-Soulier Syndrome (BSS) patient carrying the mutation p.Trp71Arg in the GPIX locus (BSS1-PBMC-iPS4F4). Peripheral blood mononuclear cells (PBMCs) were reprogrammed using heat sensitive non-integrative Sendai viruses containing the reprogramming factors Oct3/4, SOX2, KLF4 and c-MYC. Successful silencing of the exogenous reprogramming factors was checked by RT-PCR. Characterization of BSS1-PBMC-iPS4F4 included mutation analysis of GPIX locus, Short Tandem Repeats (STR) profiling, alkaline phosphatase enzymatic activity, analysis of conventional pluripotency-associated factors at mRNA and protein level and in vivo differentiation studies. BSS1-PBMC-iPS4F4 will provide a powerful tool to study BSS.


Asunto(s)
Síndrome de Bernard-Soulier/patología , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Animales , Síndrome de Bernard-Soulier/metabolismo , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Cariotipo , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Teratoma/metabolismo , Teratoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Thromb Haemost ; 112(6): 1252-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25104056

RESUMEN

The pathogenesis of atherosclerosis involves the interplay of haematopoietic, stromal and endothelial cells. Platelet interactions with endothelium and leukocytes are pivotal for atherosclerosis promotion. Glycoprotein (GP) Ibα is the ligand-binding subunit of the platelet GPIb-IX-V receptor complex; its deficiency causes the Bernard-Soulier syndrome (BSS), characterised by absent platelet GPIb-IX-V, macrothrombocytopenia and bleeding. We designed this study to determine the role of platelet GPIbα in the pathogenesis of atherosclerosis using two unique knockout models. Ldlr-/- mice were reconstituted with wild-type (wt), GPIbα-/- (lacks GPIbα) or chimeric IL-4R/GPIbα-Tg (lacks GPIbα extracellular domain) bone marrow and assayed for atherosclerosis development after feeding with pro-atherogenic "western diet". Here, we report that Ldlr-/-mice reconstituted with GPIbα-/- bone marrow developed less atherosclerosis compared to wt controls; accompanied by augmented accumulation of pro-inflammatory CD11b+ and CD11c+ myeloid cells, reduced oxLDL uptake and decreased TNF and IL 12p35 gene expression in the aortas. Flow cytometry and live cell imaging in whole blood-perfused microfluidic chambers revealed reduced platelet-monocyte aggregates in GPIbα-/- mice, which resulted in decreased monocyte activation. Interestingly, Ldlr-/-mice reconstituted with IL-4R/GPIbα-Tg bone marrow, producing less abnormal platelets, showed atherosclerotic lesions similar to wt mice. Platelet interaction with blood monocytes and accumulation of myeloid cells in the aortas were also essentially unaltered. Moreover, only complete GPIbα ablation altered platelet microparticles and CCL5 chemokine production. Thus, atherosclerosis reduction in mice lacking GPIbα may not result from the defective GPIbα-ligand binding, but more likely is a consequence of functional defects of GPIbα-/- platelets and reduced blood platelet counts.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Síndrome de Bernard-Soulier/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Síndrome de Bernard-Soulier/genética , Plaquetas/metabolismo , Trasplante de Médula Ósea , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Quimiocina CCL5/metabolismo , Dieta Occidental , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Subunidad p35 de la Interleucina-12/metabolismo , Lipoproteínas LDL/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Adhesividad Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Estructura Terciaria de Proteína , Receptores de Interleucina-4/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Pediatr Int ; 55(4): 434-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23566026

RESUMEN

BACKGROUND: Bernard-Soulier syndrome (BSS) is a rare autosomal recessive bleeding disorder characterized by giant platelets, thrombocytopenia, and a prolonged bleeding time, which is caused by homozygous mutations in the GPIbα, GPIbß, or GPIX genes. The 22q11.2 deletion syndrome (22q11.2DS) is caused by a microdeletion on chromosome 22, which includes the GPIbß gene, and is characterized by abnormal development of the pharyngeal apparatus and heart. Thus, patients with 22q11.2DS are obligate carriers for BSS. METHODS: We evaluated two infants with BSS and performed the genetic analysis of the GPIbα, GPIbß, or GPIX genes, and investigated the segregation of the mutation within the families. The status of the 22q11.2 deletion was examined by fluorescence in situ hybridization and single-nucleotide polymorphism array copy number analysis. RESULTS: DNA sequencing analysis revealed that the infants were compound heterozygous for a hemizygous mutation in the GPIbß gene (p.Trp148X and p.Leu97Phe, respectively) and 22q11.2 deletion in the other chromosome. Both infants had the common 3Mb 22q11.2 deletion but did not show major phenotypic features of 22q11.2DS, such as developmental delay, cardiac defects, dysmorphic facial features, palatal anomalies, hypocalcemia, and immune deficiency. The 22q11.2DS would not have become clear if detailed molecular genetic analyses of BSS had not been performed. CONCLUSIONS: Our cases illustrate that a suspicion of 22q11.2 deletion is warranted in pediatric BSS patients with a mutation in the GPIbß gene, even without remarkable symptoms.


Asunto(s)
Síndrome de Bernard-Soulier/genética , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Mutación , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Síndrome de Bernard-Soulier/metabolismo , Preescolar , Femenino , Hemicigoto , Humanos , Hibridación Fluorescente in Situ , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Análisis de Secuencia de ADN
10.
Haematologica ; 96(12): 1878-82, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21993687

RESUMEN

Platelet glycoprotein GPIbα mutations are the basic defect behind Bernard-Soulier syndrome, a rare inherited macrothrombocytopenia characterized by anomalies of the GPIbα, GPIbß and GPIX subunits of von Willebrand factor receptor. A 32-year old man was investigated for suspected Bernard-Soulier syndrome. Ristocetin induced agglutination was absent. Flow cytometry and Western blot analysis showed a severe reduction in GPIbα, but sequencing revealed only a biallelic c.386A>G substitution, theoretically leading to a p.Asn110Glu variation. To further clarify the data, megakaryocyte cultures were set. Though the maturation of megakaryocytes was normal, proplatelet formation was defective and GPIbα mRNA was not detectable. GPIX protein was slightly reduced and GPIbß polypeptide almost absent. Computational analysis showed that the c.386A>G mutation disrupted an exon splicing enhancer motif involved in the proper maturation of the GPIbα transcript. The c.386A>G mutation suggests a unique mutational mechanism causing the virtual absence of GPIbα without creating a stop codon.


Asunto(s)
Alelos , Sustitución de Aminoácidos , Síndrome de Bernard-Soulier/genética , Glicoproteínas de Membrana/genética , Trastornos Mentales/genética , Mutación Missense , Adulto , Síndrome de Bernard-Soulier/metabolismo , Síndrome de Bernard-Soulier/fisiopatología , Elementos de Facilitación Genéticos/genética , Exones/genética , Humanos , Masculino , Trastornos Mentales/metabolismo , Trastornos Mentales/fisiopatología , Complejo GPIb-IX de Glicoproteína Plaquetaria , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA