Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Br J Haematol ; 205(1): 306-315, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38815995

RESUMEN

Thrombocytopenia 4 (THC4) is an autosomal-dominant thrombocytopenia caused by mutations in CYCS, the gene encoding cytochrome c (CYCS), a small haeme protein essential for electron transport in mitochondria and cell apoptosis. THC4 is considered an extremely rare condition since only a few patients have been reported so far. These subjects presented mild thrombocytopenia and no or mild bleeding tendency. In this study, we describe six Italian families with five different heterozygous missense CYCS variants: p.Gly42Ser and p.Tyr49His previously associated with THC4, and three novel variants (p.Ala52Thr, p.Arg92Gly, and p.Leu99Val), which have been classified as pathogenic by bioinformatics and segregation analyses. Moreover, we supported functional effects of p.Ala52Thr and p.Arg92Gly on oxidative growth and respiratory activity in a yeast model. The clinical characterization of the 22 affected individuals, the largest series of THC4 patients ever reported, showed that this disorder is characterized by mild-to-moderate thrombocytopenia, normal platelet size, and function, low risk of bleeding, and no additional clinical phenotypes associated with reduced platelet count. Finally, we describe a significant correlation between the region of CYCS affected by mutations and the extent of thrombocytopenia, which could reflect different degrees of impairment of CYCS functions caused by different pathogenetic variants.


Asunto(s)
Citocromos c , Trombocitopenia , Humanos , Trombocitopenia/genética , Femenino , Masculino , Citocromos c/genética , Adulto , Persona de Mediana Edad , Linaje , Mutación Missense , Anciano , Adolescente , Mutación , Adulto Joven , Niño
2.
Haematologica ; 108(7): 1909-1919, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-36519321

RESUMEN

Inherited thrombocytopenias (IT) are genetic diseases characterized by low platelet count, sometimes associated with congenital defects or a predisposition to develop additional conditions. Next-generation sequencing has substantially improved our knowledge of IT, with more than 40 genes identified so far, but obtaining a molecular diagnosis remains a challenge especially for patients with non-syndromic forms, having no clinical or functional phenotypes that raise suspicion about specific genes. We performed exome sequencing (ES) in a cohort of 116 IT patients (89 families), still undiagnosed after a previously validated phenotype-driven diagnostic algorithm including a targeted analysis of suspected genes. ES achieved a diagnostic yield of 36%, with a gain of 16% over the diagnostic algorithm. This can be explained by genetic heterogeneity and unspecific genotype-phenotype relationships that make the simultaneous analysis of all the genes, enabled by ES, the most reasonable strategy. Furthermore, ES disentangled situations that had been puzzling because of atypical inheritance, sex-related effects or false negative laboratory results. Finally, ES-based copy number variant analysis disclosed an unexpectedly high prevalence of RUNX1 deletions, predisposing to hematologic malignancies. Our findings demonstrate that ES, including copy number variant analysis, can substantially contribute to the diagnosis of IT and can solve diagnostic problems that would otherwise remain a challenge.


Asunto(s)
Pruebas Genéticas , Trombocitopenia , Humanos , Secuenciación del Exoma , Fenotipo , Pruebas Genéticas/métodos , Genotipo , Trombocitopenia/diagnóstico , Trombocitopenia/genética
3.
Haematologica ; 107(1): 260-267, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33472357

RESUMEN

GFI1B is a transcription factor essential for the regulation of erythropoiesis and megakaryopoiesis, and pathogenic variants have been associated with thrombocytopenia and bleeding. Analysing thrombocytopenic families by whole exome sequencing, we identified a novel GFI1B variant (c.648+5G>A), which causes exon 9 skipping and overexpression of a shorter p32 isoform. We report the clinical data of our patients and critically review the phenotype observed in individuals with different GFI1B variants leading to the same effect on the p32 expression. Since p32 is increased in acute and chronic leukemia cells, we tested the expression level of genes playing a role in various type of cancers, including hematological tumors and found that they are significantly dysregulated, suggesting a potential role for GFI1B in carcinogenesis regulation. Increasing the detection of individuals with GFI1B variants will allow us to better characterize this rare disease and determine whether it is associated with an increased risk of developing malignancies.


Asunto(s)
Mutación de Línea Germinal , Trombocitopenia , Carcinogénesis , Humanos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Trombocitopenia/genética
4.
Blood ; 133(12): 1346-1357, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30591527

RESUMEN

Inherited thrombocytopenias (ITs) are a heterogeneous group of disorders characterized by low platelet count that may result in bleeding tendency. Despite progress being made in defining the genetic causes of ITs, nearly 50% of patients with familial thrombocytopenia are affected with forms of unknown origin. Here, through exome sequencing of 2 siblings with autosomal-recessive thrombocytopenia, we identified biallelic loss-of-function variants in PTPRJ . This gene encodes for a receptor-like PTP, PTPRJ (or CD148), which is expressed abundantly in platelets and megakaryocytes. Consistent with the predicted effects of the variants, both probands have an almost complete loss of PTPRJ at the messenger RNA and protein levels. To investigate the pathogenic role of PTPRJ deficiency in hematopoiesis in vivo, we carried out CRISPR/Cas9-mediated ablation of ptprja (the ortholog of human PTPRJ) in zebrafish, which induced a significantly decreased number of CD41+ thrombocytes in vivo. Moreover, megakaryocytes of our patients showed impaired maturation and profound defects in SDF1-driven migration and formation of proplatelets in vitro. Silencing of PTPRJ in a human megakaryocytic cell line reproduced the functional defects observed in patients' megakaryocytes. The disorder caused by PTPRJ mutations presented as a nonsyndromic thrombocytopenia characterized by spontaneous bleeding, small-sized platelets, and impaired platelet responses to the GPVI agonists collagen and convulxin. These platelet functional defects could be attributed to reduced activation of Src family kinases. Taken together, our data identify a new form of IT and highlight a hitherto unknown fundamental role for PTPRJ in platelet biogenesis.


Asunto(s)
Plaquetas/patología , Predisposición Genética a la Enfermedad , Megacariocitos/patología , Mutación , Trombocitopenia/patología , Adolescente , Adulto , Animales , Plaquetas/metabolismo , Sistemas CRISPR-Cas , Niño , Femenino , Estudios de Seguimiento , Hematopoyesis , Humanos , Masculino , Megacariocitos/metabolismo , Persona de Mediana Edad , Linaje , Pronóstico , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Trombocitopenia/etiología , Trombocitopenia/genética , Pez Cebra
5.
Int J Mol Sci ; 22(19)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34638529

RESUMEN

Bernard-Soulier syndrome (BSS) is an autosomal-recessive bleeding disorder caused by biallelic variants in the GP1BA, GP1BB, and GP9 genes encoding the subunits GPIbα, GPIbß, and GPIX of the GPIb-IX complex. Pathogenic variants usually affect the extracellular or transmembrane domains of the receptor subunits. We investigated a family with BSS caused by the homozygous c.528_550del (p.Arg177Serfs*124) variant in GP1BB, which is the first mutation ever identified that affects the cytoplasmic domain of GPIbß. The loss of the intracytoplasmic tail of GPIbß results in a mild form of BSS, characterized by only a moderate reduction of the GPIb-IX complex expression and mild or absent bleeding tendency. The variant induces a decrease of the total platelet expression of GPIbß; however, all of the mutant subunit expressed in platelets is correctly assembled into the GPIb-IX complex in the plasma membrane, indicating that the cytoplasmic domain of GPIbß is not involved in assembly and trafficking of the GPIb-IX receptor. Finally, the c.528_550del mutation exerts a dominant effect and causes mild macrothrombocytopenia in heterozygous individuals, as also demonstrated by the investigation of a second unrelated pedigree. The study of this novel GP1BB variant provides new information on pathophysiology of BSS and the assembly mechanisms of the GPIb-IX receptor.


Asunto(s)
Síndrome de Bernard-Soulier/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Trombocitopenia/genética , Adulto , Síndrome de Bernard-Soulier/sangre , Síndrome de Bernard-Soulier/patología , Plaquetas/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Dominios Proteicos/genética , Trombocitopenia/patología , Factor de von Willebrand/metabolismo
7.
Blood ; 124(6): e4-e10, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24990887

RESUMEN

Abnormalities of platelet size are one of the distinguishing features of inherited thrombocytopenias (ITs), and evaluation of blood films is recommended as an essential step for differential diagnosis of these disorders. Nevertheless, what we presently know about this subject is derived mainly from anecdotal evidence. To improve knowledge in this field, we evaluated platelet size on blood films obtained from 376 patients with all 19 forms of IT identified so far and found that these conditions differ not only in mean platelet diameter, but also in platelet diameter distribution width and the percentage of platelets with increased or reduced diameters. On the basis of these findings, we propose a new classification of ITs according to platelet size. It distinguishes forms with giant platelets, with large platelets, with normal or slightly increased platelet size, and with normal or slightly decreased platelet size. We also measured platelet diameters in 87 patients with immune thrombocytopenia and identified cutoff values for mean platelet diameter and the percentage of platelets with increased or reduced size that have good diagnostic accuracy in differentiating ITs with giant platelets and with normal or slightly decreased platelet size from immune thrombocytopenia and all other forms of IT.


Asunto(s)
Plaquetas/patología , Trombocitopenia/sangre , Trombocitopenia/genética , Adolescente , Adulto , Estudios de Casos y Controles , Tamaño de la Célula , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Pérdida Auditiva Sensorineural/sangre , Pérdida Auditiva Sensorineural/clasificación , Pérdida Auditiva Sensorineural/genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/genética , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/diagnóstico , Trombocitopenia/clasificación , Trombocitopenia/congénito , Adulto Joven
9.
Haematologica ; 101(11): 1333-1342, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27365488

RESUMEN

ETV6-related thrombocytopenia is an autosomal dominant thrombocytopenia that has been recently identified in a few families and has been suspected to predispose to hematologic malignancies. To gain further information on this disorder, we searched for ETV6 mutations in the 130 families with inherited thrombocytopenia of unknown origin from our cohort of 274 consecutive pedigrees with familial thrombocytopenia. We identified 20 patients with ETV6-related thrombocytopenia from seven pedigrees. They have five different ETV6 variants, including three novel mutations affecting the highly conserved E26 transformation-specific domain. The relative frequency of ETV6-related thrombocytopenia was 2.6% in the whole case series and 4.6% among the families with known forms of inherited thrombocytopenia. The degree of thrombocytopenia and bleeding tendency of the patients with ETV6-related thrombocytopenia were mild, but four subjects developed B-cell acute lymphoblastic leukemia during childhood, resulting in a significantly higher incidence of this condition compared to that in the general population. Clinical and laboratory findings did not identify any particular defects that could lead to the suspicion of this disorder from the routine diagnostic workup. However, at variance with most inherited thrombocytopenias, platelets were not enlarged. In vitro studies revealed that the maturation of the patients' megakaryocytes was defective and that the patients have impaired proplatelet formation. Moreover, platelets from patients with ETV6-related thrombocytopenia have reduced ability to spread on fibrinogen. Since the dominant thrombocytopenias due to mutations in RUNX1 and ANKRD26 are also characterized by normal platelet size and predispose to hematologic malignancies, we suggest that screening for ETV6, RUNX1 and ANKRD26 mutations should be performed in all subjects with autosomal dominant thrombocytopenia and normal platelet size.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/genética , Trombocitopenia/genética , Adolescente , Adulto , Transformación Celular Neoplásica/genética , Niño , Preescolar , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Familia , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular , Persona de Mediana Edad , Mutación , Proteínas Nucleares/genética , Linaje , Trombocitopenia/patología , Adulto Joven , Proteína ETS de Variante de Translocación 6
10.
Hum Mutat ; 35(2): 236-47, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24186861

RESUMEN

MYH9-related disease (MYH9-RD) is a rare autosomal-dominant disorder caused by mutations in the gene for nonmuscle myosin heavy chain IIA (NMMHC-IIA). MYH9-RD is characterized by a considerable variability in clinical evolution: patients present at birth with only thrombocytopenia, but some of them subsequently develop sensorineural deafness, cataract, and/or nephropathy often leading to end-stage renal disease (ESRD). We searched for genotype-phenotype correlations in the largest series of consecutive MYH9-RD patients collected so far (255 cases from 121 families). Association of genotypes with noncongenital features was assessed by a generalized linear regression model. The analysis defined disease evolution associated to seven different MYH9 genotypes that are responsible for 85% of MYH9-RD cases. Mutations hitting residue R702 demonstrated a complete penetrance for early-onset ESRD and deafness. The p.D1424H substitution associated with high risk of developing all the noncongenital manifestations of disease. Mutations hitting a distinct hydrophobic seam in the NMMHC-IIA head domain or substitutions at R1165 associated with high risk of deafness but low risk of nephropathy or cataract. Patients with p.E1841K, p.D1424N, and C-terminal deletions had low risk of noncongenital defects. These findings are essential to patients' clinical management and genetic counseling and are discussed in view of molecular pathogenesis of MYH9-RD.


Asunto(s)
Catarata/genética , Estudios de Asociación Genética , Pérdida Auditiva Sensorineural/genética , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Trombocitopenia/congénito , Adulto , Edad de Inicio , Sustitución de Aminoácidos , Femenino , Genotipo , Pérdida Auditiva Sensorineural/complicaciones , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Italia , Modelos Lineales , Masculino , Mutación , Fenotipo , Factores de Riesgo , Trombocitopenia/complicaciones , Trombocitopenia/diagnóstico , Trombocitopenia/genética
11.
Am J Hum Genet ; 88(1): 115-20, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21211618

RESUMEN

THC2, an autosomal-dominant thrombocytopenia described so far in only two families, has been ascribed to mutations in MASTL or ACBD5. Here, we show that ANKRD26, another gene within the THC2 locus, and neither MASTL nor ACBD5, is mutated in eight unrelated families. ANKRD26 was also found to be mutated in the family previously reported to have an ACBD5 mutation. We identified six different ANKRD26 mutations, which were clustered in a highly conserved 19 bp sequence located in the 5' untranslated region. Mutations were not detected in 500 controls and are absent from the 1000 Genomes database. Available data from an animal model and Dr. Watson's genome give evidence against haploinsufficiency as the pathogenetic mechanism for ANKRD26-mediated thrombocytopenia. The luciferase reporter assay suggests that these 5' UTR mutations might enhance ANKRD26 expression. ANKRD26 is the ancestor of a family of primate-specific genes termed POTE, which have been recently identified as a family of proapoptotic proteins. Dysregulation of apoptosis might therefore be the pathogenetic mechanism, as demonstrated for another thrombocytopenia, THC4. Further investigation is needed to provide evidence supporting this hypothesis.


Asunto(s)
Repetición de Anquirina/genética , Genes Dominantes , Mutación , Secuencia de Bases , Rotura Cromosómica , Trastornos de los Cromosomas/genética , Secuencia Conservada/genética , Femenino , Sitios Genéticos , Haploinsuficiencia , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Trombocitopenia/congénito , Trombocitopenia/genética
13.
Autophagy ; 19(3): 984-999, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35857791

RESUMEN

Endoplasmic reticulum stress is an emerging significant player in the molecular pathology of connective tissue disorders. In response to endoplasmic reticulum stress, cells can upregulate macroautophagy/autophagy, a fundamental cellular homeostatic process used by cells to degrade and recycle proteins or remove damaged organelles. In these scenarios, autophagy activation can support cell survival. Here we demonstrated by in vitro and in vivo approaches that megakaryocytes derived from col6a1-/- (collagen, type VI, alpha 1) null mice display increased intracellular retention of COL6 polypeptides, endoplasmic reticulum stress and apoptosis. The unfolded protein response is activated in col6a1-/- megakaryocytes, as evidenced by the upregulation of molecular chaperones, by the increased splicing of Xbp1 mRNA and by the higher level of the pro-apoptotic regulator DDIT3/CHOP. Despite the endoplasmic reticulum stress, basal autophagy is impaired in col6a1-/- megakaryocytes, which show lower BECN1 levels and reduced autophagosome maturation. Starvation and rapamycin treatment rescue the autophagic flux in col6a1-/- megakaryocytes, leading to a decrease in intracellular COL6 polypeptide retention, endoplasmic reticulum stress and apoptosis. Furthermore, megakaryocytes cultured from peripheral blood hematopoietic progenitors of patients affected by Bethlem myopathy and Ullrich congenital muscular dystrophy, two COL6-related disorders, displayed increased apoptosis, endoplasmic reticulum stress and impaired autophagy. These data demonstrate that genetic disorders of collagens, endoplasmic reticulum stress and autophagy regulation in megakaryocytes may be interrelated.Abbreviations: 7-AAD: 7-amino-actinomycin D; ATF: activating transcriptional factor; BAX: BCL2 associated X protein; BCL2: B cell leukemia/lymphoma 2; BCL2L1/Bcl-xL: BCL2-like 1; BM: bone marrow; COL6: collagen, type VI; col6a1-/-: mice that are null for Col6a1; DDIT3/CHOP/GADD153: DNA-damage inducible transcript 3; EGFP: enhanced green fluorescent protein; ER: endoplasmic reticulum; reticulophagy: endoplasmic reticulum-selective autophagy; HSPA5/Bip: heat shock protein 5; HSP90B1/GRP94: heat shock protein 90, beta (Grp94), member 1; LAMP2: lysosomal associated membrane protein 2; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; Mk: megakaryocytes; MTOR: mechanistic target of rapamycin kinase; NIMV: noninvasive mechanical ventilation; PI3K: phosphoinositide 3-kinase; PPP1R15A/GADD34: protein phosphatase 1, regulatory subunit 15A; RT-qPCR: reverse transcription-quantitative real-time PCR; ROS: reactive oxygen species; SERPINH1/HSP47: serine (or cysteine) peptidase inhibitor, clade H, member 1; sh-RNA: short hairpin RNA; SOCE: store operated calcium entry; UCMD: Ullrich congenital muscular dystrophy; UPR: unfolded protein response; WIPI2: WD repeat domain, phosphoinositide-interacting 2; WT: wild type; XBP1: X-box binding protein 1.


Asunto(s)
Autofagia , Fosfatidilinositol 3-Quinasas , Ratones , Animales , Autofagia/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Megacariocitos/metabolismo , Colágeno Tipo VI , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Estrés del Retículo Endoplásmico , Chaperón BiP del Retículo Endoplásmico , Proteínas Proto-Oncogénicas c-bcl-2 , Sirolimus
14.
Blood ; 116(26): 5832-7, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20844233

RESUMEN

Platelet transfusion is currently the primary medical treatment for reducing thrombocytopenia in patients with inherited thrombocytopenias. To evaluate whether stimulating megakaryopoiesis could increase platelet count in these conditions, we treated patients with a severe thrombocytopenia induced by MYH9 mutations (MYH9-related disease) with a nonpeptide thrombopoietin receptor agonist, eltrombopag. Twelve adult patients with MYH9-RD and platelet counts of less than 50 × 10(9)/L received 50 mg of eltrombopag orally per day for 3 weeks. Patients who achieved a platelet count higher than 150 × 10(9)/L stopped therapy, those with 100 to 150 platelets × 10(9)/L continued treatment at the same eltrombopag dose for 3 additional weeks, while those with less than 100 platelets × 10(9)/L increased the eltrombopag dose to 75 mg for 3 weeks. Major responses (platelet count of at least 100 × 10(9)/L or 3 times the baseline value) were obtained in 8 patients, minor responses (platelet counts at least twice the baseline value) in 3. One patient did not respond. Bleeding tendency disappeared in 8 of 10 patients with bleeding symptoms at baseline. Mild adverse events were reported in 2 patients. The availability of thrombopoietin mimetics opened new prospects in the treatment of inherited thrombocytopenias. This study is registered at www.clinicaltrials.gov as NCT01133860 (European Union Drug Regulating Authorities Clinical Trials number 2008-001903-42).


Asunto(s)
Benzoatos/administración & dosificación , Predisposición Genética a la Enfermedad , Hidrazinas/administración & dosificación , Proteínas Motoras Moleculares/genética , Mutación/genética , Cadenas Pesadas de Miosina/genética , Pirazoles/administración & dosificación , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/genética , Administración Oral , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Agregación Plaquetaria , Recuento de Plaquetas , Receptores de Trombopoyetina/agonistas , Tasa de Supervivencia , Resultado del Tratamiento , Adulto Joven
15.
Haematologica ; 96(3): 417-23, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21173099

RESUMEN

BACKGROUND: Bernard-Soulier syndrome is a severe bleeding disease due to a defect of GPIb/IX/V, a platelet complex that binds the von Willebrand factor. Due to the rarity of the disease, there are reports only on a few cases compromising any attempt to establish correlations between genotype and phenotype. In order to identify any associations, we describe the largest case series ever reported, which was evaluated systematically at the same center. DESIGN AND METHODS: Thirteen patients with the disease and seven obligate carriers were enrolled. We collected clinical aspects and determined platelet features, including number and size, expression of membrane glycoproteins, and ristocetin induced platelet aggregation. Mutations were identified by direct sequencing of the GP1BA, GP1BB, and GP9 genes and their effect was shown by molecular modeling analyses. RESULTS: Patients all had a moderate thrombocytopenia with giant platelets and a bleeding tendency whose severity varied among individuals. Consistent with expression levels of GPIbα always lower than 10% of control values, platelet aggregation was absent or severely reduced. Homozygous mutations were identified in the GP1BA, GP1BB and GP9 genes; six were novel alterations expected to destabilize the conformation of the respective protein. Except for obligate carriers of a GP9 mutation with a reduced GPIb/IX/V expression and defective aggregation, all the other carriers had no obvious anomalies. CONCLUSIONS: Regardless of mutations identified, the patients' bleeding diathesis did not correlate with thrombocytopenia, which was always moderate, and platelet GPIbα expression, which was always severely impaired. Obligate carriers had features similar to controls though their GPIb/IX/V expression showed discrepancies. Aware of the limitations of our cohort, we cannot define any correlations. However, further investigations should be encouraged to better understand the causes of this rare and underestimated disease.


Asunto(s)
Síndrome de Bernard-Soulier/fisiopatología , Glicoproteínas de Membrana/análisis , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Síndrome de Bernard-Soulier/sangre , Síndrome de Bernard-Soulier/genética , Plaquetas/patología , Forma de la Célula , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Marcadores Genéticos , Hemorragia , Homocigoto , Humanos , Italia , Masculino , Glicoproteínas de Membrana/genética , Persona de Mediana Edad , Datos de Secuencia Molecular , Agregación Plaquetaria/efectos de los fármacos , Recuento de Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Mutación Puntual , Reacción en Cadena de la Polimerasa , Ristocetina/farmacología , Trombocitopenia/sangre , Adulto Joven , Factor de von Willebrand/metabolismo
16.
Eur J Haematol ; 84(4): 291-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20002731

RESUMEN

MYH9-related disease (MYH9-RD) is a rare autosomal dominant disorder caused by mutations in MYH9, the gene encoding the heavy chain of non-muscle myosin IIA. All patients present congenital macrothrombocytopenia and inclusion bodies in neutrophils. Some of them can also develop sensorineural deafness, presenile cataract, and/or progressive nephropathy leading to end-stage renal failure. We report four families, each with a novel mutation: two missense mutations, in exons 31 and 32, and two out of frame deletions in exon 40. They were associated with no bleeding diathesis, normal, or only slightly reduced platelet count and no extra-hematological manifestations, confirming that alterations of the tail domain cause a mild form of MYH9-RD with no clinically relevant defects.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Enfermedades Hematológicas/genética , Cuerpos de Inclusión Intranucleares , Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/genética , Neutrófilos , Adulto , Anciano , Anciano de 80 o más Años , Exones/genética , Familia , Femenino , Enfermedades Genéticas Congénitas/sangre , Enfermedades Hematológicas/sangre , Humanos , Masculino , Persona de Mediana Edad
17.
Thromb Haemost ; 102(1): 90-6, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19572073

RESUMEN

MYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations of MYH9, the gene for the heavy chain of myosin-IIA. Pathogenesis of thrombocytopenia of MYH9-RD is unknown. Recent studies in mice demonstrated that myosin-IIA is an inhibitor of proplatelet formation (PPF), and suggested that it could be involved in the suppression of PPF exerted by megakaryocyte adhesion to type I collagen, which regulates the timing of platelet release within bone marrow. However, the consequences on PPF of the heterozygous mutations causative of the MYH9-RD have never been investigated. We studied the in-vitro PPF by megakaryocytes obtained from four patients carrying the p.D1424N or the p.R1933X mutations. We demonstrated that MYH9-RD megakaryocytes completely lose the physiologic suppression of proplatelet extension exerted by interaction with type I collagen, thus supporting the hypothesis that a premature platelet release within bone marrow contributes to pathogenesis of MYH9-related thrombocytopenia. Moreover, proplatelets extended by MYH9-RD megakaryocytes presented a significant defect in branching in secondary processes (p=0.001) and formed a significantly lower number of proplatelet tips (p=0.005). Since platelets are assembled at the level of proplatelet tips, this defect could further contribute to pathogenesis of thrombocytopenia of MYH9-RD patients.


Asunto(s)
Plaquetas/patología , Megacariocitos/patología , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Células Madre/patología , Trombocitopenia/genética , Trombocitopenia/patología , Adulto , Diferenciación Celular , Células Cultivadas , Femenino , Heterocigoto , Humanos , Masculino , Mutación Puntual
18.
Platelets ; 20(8): 598-602, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19860543

RESUMEN

MYH9-related disease (MYH9-RD) is a rare autosomal dominant disorder caused by mutations in MYH9, the gene encoding the heavy chain of non-muscle myosin IIA. Patients present with congenital macrothrombocytopenia and inclusion bodies in neutrophils and might develop sensorineural deafness, presenile cataract, and/or progressive nephritis leading to end-stage renal failure. In a family with eight individuals suffering from macrothrombocytopenia and hearing impairment we identified a novel c.Ala95Asp mutation. Affecting the motor domain of the protein, the mutation is likely to be associated with a severe phenotype. Therefore, this family should be carefully monitored to follow-up the renal status even though the affected members do not seem to be at risk of early kidney disease.


Asunto(s)
Proteínas Motoras Moleculares/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Adulto , Alanina/genética , Secuencia de Aminoácidos , Animales , Ácido Aspártico/genética , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Modelos Moleculares , Proteínas Motoras Moleculares/química , Datos de Secuencia Molecular , Cadenas Pesadas de Miosina/química , Linaje , Estructura Terciaria de Proteína , Alineación de Secuencia
19.
Hamostaseologie ; 39(1): 87-94, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29996171

RESUMEN

MYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations in the gene for non-muscle myosin heavy chain IIA (NMMHC-IIA). Patients present congenital macrothrombocytopenia and inclusions of NMMHC-IIA in leukocytes, and have a variable risk of developing kidney damage, sensorineural deafness, presenile cataracts and/or liver enzymes abnormalities. The spectrum of mutations found in MYH9-RD patients is limited and the incidence and severity of the non-congenital features are predicted by the causative MYH9 variant. In particular, different alterations of the C-terminal tail domain of NMMHC-IIA associate with remarkably different disease evolution. We report four novel MYH9 mutations affecting the tail domain of NMMHC-IIA and responsible for MYH9-RD in four families. Two variants cause amino acid substitutions in the coiled-coil region of NMMHC-IIA, while the other two are a splicing variant and a single nucleotide deletion both resulting in frameshift alterations of the short non-helical tailpiece. Characterization of phenotypes of affected individuals shows that all of these novel variants are associated with a mild clinical evolution of the disease.


Asunto(s)
Trastornos de los Cromosomas/genética , Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/genética , Trombocitopenia/congénito , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Rotura Cromosómica , Trastornos de los Cromosomas/patología , Femenino , Mutación del Sistema de Lectura , Humanos , Masculino , Persona de Mediana Edad , Proteínas Motoras Moleculares/química , Cadenas Pesadas de Miosina/química , Linaje , Fenotipo , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Trombocitopenia/genética , Trombocitopenia/patología , Adulto Joven
20.
Hum Mutat ; 29(3): 409-17, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18059020

RESUMEN

MYH9-related disease (MYH9-RD) is a rare autosomal-dominant disorder caused by mutations in MYH9, the gene for the heavy chain of nonmuscle myosin IIA (NMMHC-IIA). All patients present from birth with macrothrombocytopenia, but in infancy or adult life, some of them develop sensorineural deafness, presenile cataracts, and/or progressive nephritis leading to end-stage renal failure. No consistent correlations have been identified between the 27 different MYH9 mutations identified so far and the variable clinical evolution of the disease. We have evaluated 108 consecutive MYH9-RD patients belonging to 50 unrelated pedigrees. The risk of noncongenital manifestations associated with different genotypes was estimated over time by event-free survival analysis. We demonstrated that all subjects with mutations in the motor domain of NMMHC-IIA present with severe thrombocytopenia and develop nephritis and deafness before the age of 40 years, while those with mutations in the tail domain have a much lower risk of noncongenital complications and significantly higher platelet counts. We also evaluated the clinical course of patients with mutations in the four most frequently affected residues of NMMHC-IIA (responsible for 70% of MYH9-RD cases). We concluded that mutations at residue 1933 do not induce kidney damage or cataracts and cause deafness only in the elderly, those in position 702 result in severe thrombocytopenia and produce nephritis and deafness at a juvenile age, while alterations at residue 1424 or 1841 result in intermediate clinical pictures. These findings are relevant not only to patients' clinical management but also to the elucidation of the pathogenesis of the disease.


Asunto(s)
Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/genética , Mutación , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Trombocitopenia/genética , Adulto , Catarata/genética , Femenino , Genes Dominantes , Genotipo , Pérdida Auditiva Sensorineural/genética , Humanos , Lactante , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Nefritis/genética , Fenotipo , Recuento de Plaquetas , Estructura Terciaria de Proteína , Síndrome , Trombocitopenia/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA