Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
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.
Br J Haematol ; 204(6): 2453-2457, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38594875

RESUMEN

ACTN1-related thrombocytopenia is a rare disorder caused by heterozygous variants in the ACTN1 gene characterized by macrothrombocytopenia and mild bleeding tendency. We describe for the first time two patients affected with ACTN1-RT caused by a homozygous variant in ACTN1 (c.982G>A) with mild heart valve defects unexplained by any other genetic variants investigated by WES. Within the reported family, the homozygous sisters have moderate thrombocytopenia and marked platelet macrocytosis with giant platelets, revealing a more severe haematological phenotype compared to their heterozygous relatives and highlighting a significant effect of allelic burden on platelet size. Moreover, we hypothesize that some ACTN1 variants, especially when present in the homozygous state, may also contribute to the cardiac abnormalities.


Asunto(s)
Actinina , Homocigoto , Fenotipo , Trombocitopenia , Humanos , Trombocitopenia/genética , Actinina/genética , Femenino , Masculino , Linaje , Mutación , Adulto
3.
Sci Rep ; 14(1): 1117, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212351

RESUMEN

DNA polymerase eta (Polη) is the only translesion synthesis polymerase capable of error-free bypass of UV-induced cyclobutane pyrimidine dimers. A deficiency in Polη function is associated with the human disease Xeroderma pigmentosum variant (XPV). We hereby report the case of a 60-year-old woman known for XPV and carrying a Polη Thr191Pro variant in homozygosity. We further characterize the variant in vitro and in vivo, providing molecular evidence that the substitution abrogates polymerase activity and results in UV sensitivity through deficient damage bypass. This is the first functional molecular characterization of a missense variant of Polη, whose reported pathogenic variants have thus far been loss of function truncation or frameshift mutations. Our work allows the upgrading of Polη Thr191Pro from 'variant of uncertain significance' to 'likely pathogenic mutant', bearing direct impact on molecular diagnosis and genetic counseling. Furthermore, we have established a robust experimental approach that will allow a precise molecular analysis of further missense mutations possibly linked to XPV. Finally, it provides insight into critical Polη residues that may be targeted to develop small molecule inhibitors for cancer therapeutics.


Asunto(s)
Xerodermia Pigmentosa , Humanos , Persona de Mediana Edad , Daño del ADN , Mutación Missense , Prolina/genética , Dímeros de Pirimidina , Rayos Ultravioleta , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/patología , Femenino
4.
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
5.
Brain ; 145(7): 2313-2331, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35786744

RESUMEN

Epilepsy is one of the most frequent neurological diseases, with focal epilepsy accounting for the largest number of cases. The genetic alterations involved in focal epilepsy are far from being fully elucidated. Here, we show that defective lipid signalling caused by heterozygous ultra-rare variants in PIK3C2B, encoding for the class II phosphatidylinositol 3-kinase PI3K-C2ß, underlie focal epilepsy in humans. We demonstrate that patients' variants act as loss-of-function alleles, leading to impaired synthesis of the rare signalling lipid phosphatidylinositol 3,4-bisphosphate, resulting in mTORC1 hyperactivation. In vivo, mutant Pik3c2b alleles caused dose-dependent neuronal hyperexcitability and increased seizure susceptibility, indicating haploinsufficiency as a key driver of disease. Moreover, acute mTORC1 inhibition in mutant mice prevented experimentally induced seizures, providing a potential therapeutic option for a selective group of patients with focal epilepsy. Our findings reveal an unexpected role for class II PI3K-mediated lipid signalling in regulating mTORC1-dependent neuronal excitability in mice and humans.


Asunto(s)
Fosfatidilinositol 3-Quinasas Clase II , Epilepsias Parciales , Animales , Fosfatidilinositol 3-Quinasas Clase II/genética , Epilepsias Parciales/genética , Humanos , Lípidos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Convulsiones
6.
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
7.
Clin Genet ; 100(3): 329-333, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34037256

RESUMEN

Arthrogryposis describes the presence of multiple joint-contractures. Clinical severity of this phenotype is variable, and more than 400 causative genes have been proposed. Among these, ERGIC1 is a recently reported candidate encoding a putative transmembrane protein of the ER-Golgi interface. Two homozygous missense variants have been reported in patients with relatively mild non-syndromic arthrogryposis. In a consanguineous family with two affected siblings presenting congenital arthrogryposis and some facial dysmorphism we performed prenatal array-CGH, postnatal targeted exome and genome sequencing. Genome sequencing identified a homozygous 22.6 Kb deletion encompassing the promoter and first exon of ERGIC1. mRNA quantification showed the complete absence of ERGIC1 expression in the two affected siblings and a decrease in heterozygous parents. Our observations validate the pathogenic role of ERGIC1 in congenital arthrogryposis and demonstrate that complete loss of function causes a relatively mild phenotype. These findings will contribute to improve genetic counseling of ERGIC1 mutations.


Asunto(s)
Artrogriposis/genética , Proteínas de Transporte Vesicular/genética , Consanguinidad , Homocigoto , Humanos , Lactante , Mutación con Pérdida de Función , Pérdida de Heterocigocidad , Masculino , Regiones Promotoras Genéticas/genética , Análisis por Matrices de Proteínas , ARN Mensajero , Secuenciación del Exoma
9.
Mol Diagn Ther ; 24(5): 571-577, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32772316

RESUMEN

BACKGROUND: Formalin-fixed, paraffin-embedded brain specimens are a potentially rich resource to identify somatic variants, but their DNA is characterised by low yield and extensive degradation, and matched peripheral samples are usually unavailable for analysis. METHODS: We designed single-molecule molecular inversion probes to target 18 MTOR somatic mutational hot-spots in unmatched, histologically proven focal cortical dysplasias from formalin-fixed, paraffin-embedded tissues of 50 patients. RESULTS: We achieved adequate DNA and sequencing quality in 28 focal cortical dysplasias, mostly extracted within 2 years from fixation, showing a statistically significant effect of time from fixation as a major determinant for successful genetic analysis. We identified and validated seven encompassing hot-spot residues (found in 14% of all patients and in 25% of those sequenced and analysed). The allele fraction had a range of 2-5% and variants were absent in available neighbouring non-focal cortical dysplasia specimens. We computed an alternate allele threshold for calling true variants, based on an experiment-wise mismatch count distribution, well predicting call reliability. CONCLUSIONS: Single-molecule molecular inversion probes are experimentally simple, cost effective and scalable, accurately detecting clinically relevant somatic variants in challenging brain formalin-fixed, paraffin-embedded tissues.


Asunto(s)
Alelos , Pruebas Genéticas , Malformaciones del Desarrollo Cortical/diagnóstico , Malformaciones del Desarrollo Cortical/genética , Mutación , Serina-Treonina Quinasas TOR/genética , Análisis Mutacional de ADN/métodos , Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Malformaciones del Desarrollo Cortical/cirugía , Sondas Moleculares , Reproducibilidad de los Resultados , Imagen Individual de Molécula , Serina-Treonina Quinasas TOR/metabolismo
10.
Eur J Hum Genet ; 28(11): 1602-1614, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32681121

RESUMEN

In December 2019, an initial cluster of interstitial bilateral pneumonia emerged in Wuhan, China. A human-to-human transmission was assumed and a previously unrecognized entity, termed coronavirus disease-19 (COVID-19) due to a novel coronavirus (SARS-CoV-2) was described. The infection has rapidly spread out all over the world and Italy has been the first European country experiencing the endemic wave with unexpected clinical severity in comparison with Asian countries. It has been shown that SARS-CoV-2 utilizes angiotensin converting enzyme 2 (ACE2) as host receptor and host proteases for cell surface binding and internalization. Thus, a predisposing genetic background can give reason for interindividual disease susceptibility and/or severity. Taking advantage of the Network of Italian Genomes (NIG), here we mined whole-exome sequencing data of 6930 Italian control individuals from five different centers looking for ACE2 variants. A number of variants with a potential impact on protein stability were identified. Among these, three more common missense changes, p.(Asn720Asp), p.(Lys26Arg), and p.(Gly211Arg) were predicted to interfere with protein structure and stabilization. Rare variants likely interfering with the internalization process, namely p.(Leu351Val) and p.(Pro389His), predicted to interfere with SARS-CoV-2 spike protein binding, were also observed. Comparison of ACE2 WES data between a cohort of 131 patients and 258 controls allowed identifying a statistically significant (P value < 0.029) higher allelic variability in controls compared with patients. These findings suggest that a predisposing genetic background may contribute to the observed interindividual clinical variability associated with COVID-19, allowing an evidence-based risk assessment leading to personalized preventive measures and therapeutic options.


Asunto(s)
Infecciones por Coronavirus/genética , Peptidil-Dipeptidasa A/genética , Neumonía Viral/genética , Anciano , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/química , COVID-19 , Estudios de Cohortes , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Bases de Datos Genéticas , Femenino , Mutación del Sistema de Lectura , Predisposición Genética a la Enfermedad , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Mutación Missense , Pandemias , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/epidemiología , Neumonía Viral/virología , Estabilidad Proteica , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo , Secuenciación del Exoma
11.
Hum Genet ; 139(11): 1429-1441, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32488467

RESUMEN

Autozygosity-driven exome analysis has been shown effective for identification of genes underlying recessive diseases especially in countries of the so-called Greater Middle East (GME), where high consanguinity unravels the phenotypic effects of recessive alleles and large family sizes facilitate homozygosity mapping. In Italy, as in most European countries, consanguinity is estimated low. Nonetheless, consanguineous Italian families are not uncommon in publications of genetic findings and are often key to new associations of genes with rare diseases. We collected 52 patients from 47 consanguineous families with suspected recessive diseases, 29 originated in GME countries and 18 of Italian descent. We performed autozygosity-driven exome analysis by detecting long runs of homozygosity (ROHs > 1.5 Mb) and by prioritizing candidate clinical variants within. We identified a pathogenic synonymous variant that had been previously missed in NARS2 and we increased an initial high diagnostic rate (47%) to 55% by matchmaking our candidate genes and including in the analysis shorter ROHs that may also happen to be autozygous. GME and Italian families contributed to diagnostic yield comparably. We found no significant difference either in the extension of the autozygous genome, or in the distribution of candidate clinical variants between GME and Italian families, while we showed that the average autozygous genome was larger and the mean number of candidate clinical variants was significantly higher (p = 0.003) in mutation-positive than in mutation-negative individuals, suggesting that these features influence the likelihood that the disease is autozygosity-related. We highlight the utility of autozygosity-driven genomic analysis also in countries and/or communities, where consanguinity is not widespread cultural tradition.


Asunto(s)
Pruebas Genéticas/métodos , Genoma Humano/genética , Mapeo Cromosómico/métodos , Consanguinidad , Exoma/genética , Familia , Femenino , Genes Recesivos/genética , Humanos , Italia , Masculino , Medio Oriente , Mutación/genética , Linaje
12.
Epileptic Disord ; 21(2): 185-191, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30977726

RESUMEN

Epilepsy with auditory features (EAF) is a focal epilepsy syndrome characterized by prominent auditory ictal manifestations. Two main genes, LGI1 and RELN, have been implicated in EAF, but the genetic aetiology remains unknown in half of families and most sporadic cases. We previously described a pathogenic SCN1A missense variant (p.Thr956Met) segregating in a large family in which the proband and her affected daughter had EAF, thus satisfying the minimum requirement for diagnosis of autosomal dominant EAF (ADEAF). However, the remaining eight affected family members had clinical manifestations typically found in families with genetic epilepsy with febrile seizures plus (GEFS+). We aimed to investigate the role/impact of SCN1A mutations in EAF. We detailed the phenotype of this family and report on SCN1A screening in a cohort of 29 familial and 52 sporadic LGI1 variant-negative EAF patients. We identified two possibly pathogenic missense variants (p.Tyr790Phe and p.Thr140Ile) in sporadic patients (3.8%) showing typical EAF and no antecedent febrile seizures. Both p.Thr956Met and p.Tyr790Phe were previously described in unrelated patients with epilepsies within the GEFS+ spectrum. SCN1A mutations may be involved in EAF within the GEFS+ spectrum, however, the role of SCN1A in EAF without features that lead to a suspicion of underlying GEFS+ remains unclear and should be elucidated in future studies.


Asunto(s)
Trastornos de la Percepción Auditiva , Epilepsias Parciales , Epilepsia Generalizada , Canal de Sodio Activado por Voltaje NAV1.1/genética , Convulsiones Febriles , Adulto , Anciano , Trastornos de la Percepción Auditiva/etiología , Trastornos de la Percepción Auditiva/genética , Trastornos de la Percepción Auditiva/fisiopatología , Epilepsias Parciales/complicaciones , Epilepsias Parciales/genética , Epilepsias Parciales/fisiopatología , Epilepsia Generalizada/complicaciones , Epilepsia Generalizada/genética , Epilepsia Generalizada/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Proteína Reelina , Convulsiones Febriles/complicaciones , Convulsiones Febriles/genética , Convulsiones Febriles/fisiopatología
13.
Ann Clin Transl Neurol ; 6(3): 475-485, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30911571

RESUMEN

Objective: We investigated the contribution to sporadic focal epilepsies (FE) of ultrarare variants in genes coding for the components of complexes regulating mechanistic Target Of Rapamycin (mTOR)complex 1 (mTORC1). Methods: We collected genetic data of 121 Italian isolated FE cases and 512 controls by Whole Exome Sequencing (WES) and single-molecule Molecular Inversion Probes (smMIPs) targeting 10 genes of the GATOR1, GATOR2, and TSC complexes. We collapsed "qualifying" variants (ultrarare and predicted to be deleterious or loss of function) across the examined genes and sought to identify their enrichment in cases compared to controls. Results: We found eight qualifying variants in cases and nine in controls, demonstrating enrichment in FE patients (P = 0.006; exact unconditional test, one-tailed). Pathogenic variants were identified in DEPDC5 and TSC2, both major genes for Mendelian FE syndromes. Interpretation: Our findings support the contribution of ultrarare variants in genes in the mTOR pathway complexes GATOR and TSC to the risk of sporadic FE and a shared genetic basis between rare and common epilepsies. The identification of a monogenic etiology in isolated cases, most typically encountered in clinical practice, may offer to a broader community of patients the perspective of precision therapies directed by the underlying genetic cause.


Asunto(s)
Epilepsias Parciales/genética , Variación Estructural del Genoma/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Proteínas Activadoras de GTPasa/genética , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Persona de Mediana Edad , Complejos Multiproteicos/genética , Transducción de Señal , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Secuenciación del Exoma
14.
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
15.
Br J Haematol ; 183(2): 276-288, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30351444

RESUMEN

The inherited thrombocytopenias (IT) are a heterogeneous group of diseases resulting from mutations in more than 30 different genes. Among them, ACTN1-related thrombocytopenia (ACTN1-RT; Online Mendelian Inheritance in Man: 615193) is one of the most recently identified forms. It has been described as a mild autosomal dominant macrothrombocytopenia caused by mutations in ACTN1, a gene encoding for one of the two non-muscle isoforms of α-actinin. We recently identified seven new unrelated families with ACTN1-RT caused by different mutations. Two of them are novel missense variants (p.Trp128Cys and p.Pro233Leu), whose pathogenic role has been confirmed by in vitro studies. Together with the 10 families we have previously described, our cohort of ACTN1-RT now consists of 49 individuals carrying ACTN1 mutations. This is the largest case series ever collected and enabled a critical evaluation of the clinical aspects of the disease. We concluded that ACTN1-RT is the fourth most frequent form of IT worldwide and it is characterized by platelet macrocytosis in all affected subjects and mild thrombocytopenia in less than 80% of cases. The risk of bleeding, either spontaneous or upon haemostatic challenge, is negligible and there are no other associated defects, either congenital or acquired. Therefore, ACTN1-RT is a benign form of IT, whose diagnosis provides affected individuals and their families with a good prognosis.


Asunto(s)
Actinina/genética , Enfermedades Hematológicas/genética , Mutación , Trombocitopenia/genética , Adulto , Anciano , Recuento de Células Sanguíneas , Plaquetas/patología , Niño , Análisis Mutacional de ADN/métodos , Eritrocitos Anormales/patología , Femenino , Enfermedades Hematológicas/sangre , Enfermedades Hematológicas/patología , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Agregación Plaquetaria , Trombocitopenia/sangre
17.
Interact Cardiovasc Thorac Surg ; 25(5): 813-817, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29049801

RESUMEN

OBJECTIVES: Mutations in ACTA2 have been reported as a cause of familiar thoracic aortic aneurysm (TAA) with associated bicuspid aortic valve (BAV) in some individuals. Our aim is to investigate the role of ACTA2 mutations in BAV associated with TAA in 20 patients. METHODS: We recruited 20 patients who underwent surgery for BAV and TAA; clinical genetic evaluation and ACTA2 mutation analysis were performed on each patient, along with next-generation sequencing analysis of BAV-related genes. Available first-degree relatives were enrolled and evaluated with echocardiography and clinical genetic examination. RESULTS: No mutations were found in ACTA2 or in BAV-related genes in our probands nor any common clinical signs possibly related to their heart disease. One-third of probands did not have any cardiovascular risk factor. Surgery was required at a young age (mean age 47.2 years) and at relatively small ascending aortic diameters (mean size 49.7 mm). In 77 first-degree relatives, 1 new diagnosis of TAA requiring surgery was made and 8 previous BAV/TAA diagnoses (9/77 = 11.7%) were confirmed. The phenotype BAV ± TAA segregated in 25% of our families. CONCLUSIONS: Although based on a small cohort, our results seemed to justify the conclusion that ACTA2 did not play a significant role in the pathogenesis of BAV aortopathy. The underlying genetic factors of this condition remain elusive and both large association studies and exome or genome sequencing could represent promising tools to unravel its pathogenesis. Aortic resection of TAA at elective surgery in these patients should be recommended as well as echocardiography in their first-degree relatives.


Asunto(s)
Actinas/genética , Aneurisma de la Aorta Torácica/genética , Válvula Aórtica/anomalías , ADN/genética , Enfermedades de las Válvulas Cardíacas/genética , Mutación , Actinas/metabolismo , Adolescente , Adulto , Aneurisma de la Aorta Torácica/complicaciones , Aneurisma de la Aorta Torácica/metabolismo , Enfermedad de la Válvula Aórtica Bicúspide , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Marcadores Genéticos/genética , Enfermedades de las Válvulas Cardíacas/complicaciones , Enfermedades de las Válvulas Cardíacas/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Factores de Riesgo , Adulto Joven
19.
J Hematol Oncol ; 10(1): 18, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28100250

RESUMEN

Thrombocytopenia 2 (THC2) is an inherited disorder caused by monoallelic single nucleotide substitutions in the 5'UTR of the ANKRD26 gene. Patients have thrombocytopenia and increased risk of myeloid malignancies, in particular, acute myeloid leukemia (AML). Given the association of variants in the ANKRD26 5'UTR with myeloid neoplasms, we investigated whether, and to what extent, mutations in this region contribute to apparently sporadic AML. To this end, we studied 250 consecutive, non-familial, adult AML patients and screened the first exon of ANKRD26 including the 5'UTR. We found variants in four patients. One patient had the c.-125T>G substitution in the 5'UTR, while three patients carried two different variants in the 5' end of the ANKRD26 coding region (c.3G>A or c.105C>G). Review of medical history showed that the patient carrying the c.-125T>G was actually affected by typical but unrecognized THC2, highlighting that some apparently sporadic AML cases represent the evolution of a well-characterized familial predisposition disorder. As regards the c.3G>A and the c.105C>G, we found that both variants result in the synthesis of N-terminal truncated ANKRD26 isoforms, which are stable and functional in cells, in particular, have a strong ability to activate the MAPK/ERK signaling pathway. Moreover, investigation of one patient with the c.3G>A showed that mutation was associated with strong ANKRD26 overexpression in vivo, which is the proposed mechanism for predisposition to AML in THC2 patients. These data provide evidence that N-terminal ANKRD26 truncating mutations play a potential pathogenetic role in AML. Recognition of AML patients with germline ANKRD26 pathogenetic variants is mandatory for selection of donors for bone marrow transplantation.


Asunto(s)
Leucemia Mieloide Aguda/genética , Mutación , Proteínas Nucleares/genética , Regiones no Traducidas 5'/genética , Transformación Celular Neoplásica , Rotura Cromosómica , Trastornos de los Cromosomas/complicaciones , Trastornos de los Cromosomas/genética , Predisposición Genética a la Enfermedad , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intercelular , Leucemia Mieloide Aguda/etiología , ARN Mensajero/sangre , Trombocitopenia/complicaciones , Trombocitopenia/congénito , Trombocitopenia/genética , Transfección
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...