Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
Am J Hum Genet ; 108(7): 1330-1341, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34102099

RESUMEN

Adaptor protein (AP) complexes mediate selective intracellular vesicular trafficking and polarized localization of somatodendritic proteins in neurons. Disease-causing alleles of various subunits of AP complexes have been implicated in several heritable human disorders, including intellectual disabilities (IDs). Here, we report two bi-allelic (c.737C>A [p.Pro246His] and c.1105A>G [p.Met369Val]) and eight de novo heterozygous variants (c.44G>A [p.Arg15Gln], c.103C>T [p.Arg35Trp], c.104G>A [p.Arg35Gln], c.229delC [p.Gln77Lys∗11], c.399_400del [p.Glu133Aspfs∗37], c.747G>T [p.Gln249His], c.928-2A>C [p.?], and c.2459C>G [p.Pro820Arg]) in AP1G1, encoding gamma-1 subunit of adaptor-related protein complex 1 (AP1γ1), associated with a neurodevelopmental disorder (NDD) characterized by mild to severe ID, epilepsy, and developmental delay in eleven families from different ethnicities. The AP1γ1-mediated adaptor complex is essential for the formation of clathrin-coated intracellular vesicles. In silico analysis and 3D protein modeling simulation predicted alteration of AP1γ1 protein folding for missense variants, which was consistent with the observed altered AP1γ1 levels in heterologous cells. Functional studies of the recessively inherited missense variants revealed no apparent impact on the interaction of AP1γ1 with other subunits of the AP-1 complex but rather showed to affect the endosome recycling pathway. Knocking out ap1g1 in zebrafish leads to severe morphological defect and lethality, which was significantly rescued by injection of wild-type AP1G1 mRNA and not by transcripts encoding the missense variants. Furthermore, microinjection of mRNAs with de novo missense variants in wild-type zebrafish resulted in severe developmental abnormalities and increased lethality. We conclude that de novo and bi-allelic variants in AP1G1 are associated with neurodevelopmental disorder in diverse populations.


Asunto(s)
Complejo 1 de Proteína Adaptadora/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Alelos , Animales , Análisis Mutacional de ADN , Femenino , Células HEK293 , Humanos , Masculino , Linaje , Ratas , Pez Cebra/genética
2.
Am J Hum Genet ; 107(3): 499-513, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32721402

RESUMEN

Signal transduction through the RAF-MEK-ERK pathway, the first described mitogen-associated protein kinase (MAPK) cascade, mediates multiple cellular processes and participates in early and late developmental programs. Aberrant signaling through this cascade contributes to oncogenesis and underlies the RASopathies, a family of cancer-prone disorders. Here, we report that de novo missense variants in MAPK1, encoding the mitogen-activated protein kinase 1 (i.e., extracellular signal-regulated protein kinase 2, ERK2), cause a neurodevelopmental disease within the RASopathy phenotypic spectrum, reminiscent of Noonan syndrome in some subjects. Pathogenic variants promote increased phosphorylation of the kinase, which enhances translocation to the nucleus and boosts MAPK signaling in vitro and in vivo. Two variant classes are identified, one of which directly disrupts binding to MKP3, a dual-specificity protein phosphatase negatively regulating ERK function. Importantly, signal dysregulation driven by pathogenic MAPK1 variants is stimulus reliant and retains dependence on MEK activity. Our data support a model in which the identified pathogenic variants operate with counteracting effects on MAPK1 function by differentially impacting the ability of the kinase to interact with regulators and substrates, which likely explains the minor role of these variants as driver events contributing to oncogenesis. After nearly 20 years from the discovery of the first gene implicated in Noonan syndrome, PTPN11, the last tier of the MAPK cascade joins the group of genes mutated in RASopathies.


Asunto(s)
Carcinogénesis/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Trastornos del Neurodesarrollo/genética , Síndrome de Noonan/genética , Preescolar , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Mutación Missense/genética , Trastornos del Neurodesarrollo/patología , Síndrome de Noonan/fisiopatología , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Transducción de Señal , Secuenciación del Exoma , Proteínas ras/genética
3.
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
4.
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
5.
Int J Mol Sci ; 24(6)2023 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-36982896

RESUMEN

Idiosyncratic Drug-Induced Liver Injury (iDILI) represents an actual health challenge, accounting for more than 40% of hepatitis cases in adults over 50 years and more than 50% of acute fulminant hepatic failure cases. In addition, approximately 30% of iDILI are cholestatic (drug-induced cholestasis (DIC)). The liver's metabolism and clearance of lipophilic drugs depend on their emission into the bile. Therefore, many medications cause cholestasis through their interaction with hepatic transporters. The main canalicular efflux transport proteins include: 1. the bile salt export pump (BSEP) protein (ABCB11); 2. the multidrug resistance protein-2 (MRP2, ABCC2) regulating the bile salts' independent flow by excretion of glutathione; 3. the multidrug resistance-1 protein (MDR1, ABCB1) that transports organic cations; 4. the multidrug resistance-3 protein (MDR3, ABCB4). Two of the most known proteins involved in bile acids' (BAs) metabolism and transport are BSEP and MDR3. BSEP inhibition by drugs leads to reduced BAs' secretion and their retention within hepatocytes, exiting in cholestasis, while mutations in the ABCB4 gene expose the biliary epithelium to the injurious detergent actions of BAs, thus increasing susceptibility to DIC. Herein, we review the leading molecular pathways behind the DIC, the links with the other clinical forms of familial intrahepatic cholestasis, and, finally, the main cholestasis-inducing drugs.


Asunto(s)
Colestasis Intrahepática , Colestasis , Adulto , Humanos , Colestasis/inducido químicamente , Colestasis/genética , Colestasis/metabolismo , Hepatocitos/metabolismo , Bilis/metabolismo , Ácidos y Sales Biliares/metabolismo , Colestasis Intrahepática/inducido químicamente , Colestasis Intrahepática/genética , Colestasis Intrahepática/metabolismo
6.
Hum Mol Genet ; 29(9): 1489-1497, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32307552

RESUMEN

Despite the wide use of genomics to investigate the molecular basis of rare congenital malformations, a significant fraction of patients remains bereft of diagnosis. As part of our continuous effort to recruit and perform genomic and functional studies on such cohorts, we investigated the genetic and mechanistic cause of disease in two independent consanguineous families affected by overlapping craniofacial, cardiac, laterality and neurodevelopmental anomalies. Using whole exome sequencing, we identified homozygous frameshift CCDC32 variants in three affected individuals. Functional analysis in a zebrafish model revealed that ccdc32 depletion recapitulates the human phenotypes. Because some of the patient phenotypes overlap defects common to ciliopathies, we asked if loss of CCDC32 might contribute to the dysfunction of this organelle. Consistent with this hypothesis, we show that ccdc32 is required for normal cilia formation in zebrafish embryos and mammalian cell culture, arguing that ciliary defects are at least partially involved in the pathomechanism of this disorder.


Asunto(s)
Ciliopatías/genética , Anomalías Congénitas/genética , Cardiopatías Congénitas/genética , Trastornos del Neurodesarrollo/genética , Animales , Sistemas CRISPR-Cas/genética , Cilios/genética , Cilios/patología , Ciliopatías/complicaciones , Ciliopatías/patología , Anomalías Congénitas/patología , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Exoma/genética , Femenino , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/patología , Homocigoto , Humanos , Mutación con Pérdida de Función/genética , Masculino , Trastornos del Neurodesarrollo/complicaciones , Trastornos del Neurodesarrollo/patología , Linaje , Fenotipo , Secuenciación del Exoma , Pez Cebra/genética
7.
Am J Hum Genet ; 105(4): 689-705, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31495489

RESUMEN

Sphingomyelinases generate ceramide from sphingomyelin as a second messenger in intracellular signaling pathways involved in cell proliferation, differentiation, or apoptosis. Children from 12 unrelated families presented with microcephaly, simplified gyral pattern of the cortex, hypomyelination, cerebellar hypoplasia, congenital arthrogryposis, and early fetal/postnatal demise. Genomic analysis revealed bi-allelic loss-of-function variants in SMPD4, coding for the neutral sphingomyelinase-3 (nSMase-3/SMPD4). Overexpression of human Myc-tagged SMPD4 showed localization both to the outer nuclear envelope and the ER and additionally revealed interactions with several nuclear pore complex proteins by proteomics analysis. Fibroblasts from affected individuals showed ER cisternae abnormalities, suspected for increased autophagy, and were more susceptible to apoptosis under stress conditions, while treatment with siSMPD4 caused delayed cell cycle progression. Our data show that SMPD4 links homeostasis of membrane sphingolipids to cell fate by regulating the cross-talk between the ER and the outer nuclear envelope, while its loss reveals a pathogenic mechanism in microcephaly.


Asunto(s)
Artrogriposis/genética , Microcefalia/genética , Trastornos del Neurodesarrollo/genética , Esfingomielina Fosfodiesterasa/genética , Artrogriposis/patología , Linaje de la Célula , Niño , Retículo Endoplásmico/metabolismo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Microcefalia/patología , Mitosis , Trastornos del Neurodesarrollo/patología , Linaje , Empalme del ARN
8.
Bioinformatics ; 37(5): 723-725, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32805025

RESUMEN

MOTIVATION: Next-generation sequencing is increasingly adopted in the clinical practice largely thanks to concurrent advancements in bioinformatic tools for variant detection and annotation. However, the need to assess sequencing quality at the base-pair level still poses challenges for diagnostic accuracy. One of the most popular quality parameters is the percentage of targeted bases characterized by low depth of coverage (DoC). These regions potentially 'hide' clinically relevant variants, but no annotation is usually returned with them. However, visualizing low-DoC data with their potential functional and clinical consequences may be useful to prioritize inspection of specific regions before re-sequencing all coverage gaps or making assertions about completeness of the diagnostic test. To meet this need, we have developed unCOVERApp, an interactive application for graphical inspection and clinical annotation of low-DoC genomic regions containing genes. RESULTS: unCOVERApp interactive plots allow to display gene sequence coverage down to the base-pair level, and functional and clinical annotations of sites below a user-defined DoC threshold can be downloaded in a user-friendly spreadsheet format. Moreover, unCOVERApp provides a simple statistical framework to evaluate if DoC is sufficient for the detection of somatic variants. A maximum credible allele frequency calculator is also available allowing users to set allele frequency cut-offs based on assumptions about the genetic architecture of the disease. In conclusion, unCOVERApp is an original tool designed to identify sites of potential clinical interest that may be 'hidden' in diagnostic sequencing data. AVAILABILITYAND IMPLEMENTATION: unCOVERApp is a free application developed with Shiny packages and available in GitHub (https://github.com/Manuelaio/uncoverappLib). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Programas Informáticos , Genoma , Genómica
9.
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
10.
Int J Mol Sci ; 23(21)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36361767

RESUMEN

The advent of Whole Genome Sequencing (WGS) broadened the genetic variation detection range, revealing the presence of variants even in non-coding regions of the genome, which would have been missed using targeted approaches. One of the most challenging issues in WGS analysis regards the interpretation of annotated variants. This review focuses on tools suitable for the functional annotation of variants falling into non-coding regions. It couples the description of non-coding genomic areas with the results and performance of existing tools for a functional interpretation of the effect of variants in these regions. Tools were tested in a controlled genomic scenario, representing the ground-truth and allowing us to determine software performance.


Asunto(s)
Genómica , Programas Informáticos , Humanos , Genómica/métodos , Secuenciación Completa del Genoma/métodos , Genoma , Genoma Humano
11.
J Cell Mol Med ; 25(5): 2459-2470, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33476483

RESUMEN

Autism spectrum disorder (ASD) is characterized by a complex polygenic background, but with the unique feature of a subset of cases (~15%-30%) presenting a rare large-effect variant. However, clinical interpretation in these cases is often complicated by incomplete penetrance, variable expressivity and different neurodevelopmental trajectories. NRXN1 intragenic deletions represent the prototype of such ASD-associated susceptibility variants. From chromosomal microarrays analysis of 104 ASD individuals, we identified an inherited NRXN1 deletion in a trio family. We carried out whole-exome sequencing and deep sequencing of mitochondrial DNA (mtDNA) in this family, to evaluate the burden of rare variants which may contribute to the phenotypic outcome in NRXN1 deletion carriers. We identified an increased burden of exonic rare variants in the ASD child compared to the unaffected NRXN1 deletion-transmitting mother, which remains significant if we restrict the analysis to potentially deleterious rare variants only (P = 6.07 × 10-5 ). We also detected significant interaction enrichment among genes with damaging variants in the proband, suggesting that additional rare variants in interacting genes collectively contribute to cross the liability threshold for ASD. Finally, the proband's mtDNA presented five low-level heteroplasmic mtDNA variants that were absent in the mother, and two maternally inherited variants with increased heteroplasmic load. This study underlines the importance of a comprehensive assessment of the genomic background in carriers of large-effect variants, as penetrance modulation by additional interacting rare variants to might represent a widespread mechanism in neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/etiología , Proteínas de Unión al Calcio/genética , Predisposición Genética a la Enfermedad , Heterocigoto , Moléculas de Adhesión de Célula Nerviosa/genética , Penetrancia , Eliminación de Secuencia , Adulto , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/psicología , Hibridación Genómica Comparativa , Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Exones , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Estudios de Asociación Genética , Variación Genética , Genoma Mitocondrial , Genómica/métodos , Humanos , Lactante , Masculino , Fenotipo , Secuenciación del Exoma
12.
Am J Hum Genet ; 102(2): 309-320, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29394990

RESUMEN

Exome sequencing has markedly enhanced the discovery of genes implicated in Mendelian disorders, particularly for individuals in whom a known clinical entity could not be assigned. This has led to the recognition that phenotypic heterogeneity resulting from allelic mutations occurs more commonly than previously appreciated. Here, we report that missense variants in CDC42, a gene encoding a small GTPase functioning as an intracellular signaling node, underlie a clinically heterogeneous group of phenotypes characterized by variable growth dysregulation, facial dysmorphism, and neurodevelopmental, immunological, and hematological anomalies, including a phenotype resembling Noonan syndrome, a developmental disorder caused by dysregulated RAS signaling. In silico, in vitro, and in vivo analyses demonstrate that mutations variably perturb CDC42 function by altering the switch between the active and inactive states of the GTPase and/or affecting CDC42 interaction with effectors, and differentially disturb cellular and developmental processes. These findings reveal the remarkably variable impact that dominantly acting CDC42 mutations have on cell function and development, creating challenges in syndrome definition, and exemplify the importance of functional profiling for syndrome recognition and delineation.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Craneofaciales/genética , Heterogeneidad Genética , Atrofia Muscular/genética , Mutación Missense , Trastornos del Neurodesarrollo/genética , Síndrome de Noonan/genética , Proteína de Unión al GTP cdc42/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Adolescente , Adulto , Niño , Preescolar , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Femenino , Expresión Génica , Humanos , Lactante , Masculino , Modelos Moleculares , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Síndrome de Noonan/metabolismo , Síndrome de Noonan/patología , Fenotipo , Estructura Secundaria de Proteína , Índice de Severidad de la Enfermedad , Proteína de Unión al GTP cdc42/química , Proteína de Unión al GTP cdc42/metabolismo
13.
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
14.
Am J Med Genet A ; 185(4): 1204-1210, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33442900

RESUMEN

Feingold Syndrome type 1 (FS1) is an autosomal dominant disorder due to a loss of function mutations in the MYCN gene. FS1 is generally clinically characterized by mild learning disability, microcephaly, short palpebral fissures, short stature, brachymesophalangy, hypoplastic thumbs, as well as syndactyly of toes, variably associated with organ abnormalities, the most common being gastrointestinal atresia. In current literature, more than 120 FS1 patients have been described, but diagnostic criteria are not well agreed upon, likewise the genotype-phenotype correlations are not well understood. Here, we describe 11 FS1 patients, belonging to six distinct families, where we have identified three novel MYCN mutations along with three pathogenetic variants, the latter which have already been reported. Several patients presented a mild phenotype of the condition and they have been diagnosed as being affected only after segregation analyses of the MYCN mutation identified in the propositus. We also describe here the first ever FS1 patient with severe intellectual disability having a maternally inherited MYCN variant together with an additional GNAO1 mutation inherited paternally. Mutations in the GNAO1 gene are associated with a specific form of intellectual disability and epilepsy, thus the finding of two different rare diseases in the same patient could explain his severe phenotype. Therein, a thorough investigation is merited into the possibility that additional variants in patients with a MYCN mutation and severe phenotype do exist. Finally, in order to guarantee a more reliable diagnosis of FS1, we suggest using both major and minor clinical-molecular diagnostic criteria.


Asunto(s)
Párpados/anomalías , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Deformidades Congénitas de las Extremidades/genética , Microcefalia/genética , Proteína Proto-Oncogénica N-Myc/genética , Fístula Traqueoesofágica/genética , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Adolescente , Niño , Preescolar , Párpados/patología , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Genotipo , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Deformidades Congénitas de las Extremidades/complicaciones , Deformidades Congénitas de las Extremidades/patología , Masculino , Microcefalia/complicaciones , Microcefalia/patología , Fenotipo , Sindactilia/complicaciones , Sindactilia/genética , Sindactilia/patología , Fístula Traqueoesofágica/complicaciones , Fístula Traqueoesofágica/patología
15.
J Genet Couns ; 30(3): 665-675, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33142017

RESUMEN

Exome/genome sequencing (ES/GS) is increasingly becoming routine in clinical genetic diagnosis, yet issues regarding how to disclose and manage secondary findings (SFs) remain to be addressed, and limited evidence is available on patients' preferences. We carried out semi-structured interviews with 307 individuals undergoing clinical genetic testing to explore their preferences for return of SFs in the hypothetical scenario that their test would be performed using ES/GS. Participants were 254 females (82.7%) and 53 males (17.3%), aged 18-86 years; 73.9% (81.1% of those with lower education levels) reported no prior knowledge of ES/GS. Prior knowledge of ES/GS was more common among patients tested for Mendelian conditions (34.5%), compared to those undergoing cancer genetic testing (22.3%) or carrier screening (7.4%). Despite this reported lack of knowledge, most participants (213, 69.6%) stated they would prefer to be informed of all possible results. Reasons in favor of disclosure included wanting to be aware of any risks (168; 83.6%) and to help relatives (23; 11.4%), but also hope that preventive measures might become available in the future (10, 5%). Conversely, potential negative impact on quality of life was the commonest motivation against disclosure. Among 179 participants seen for cancer genetic counseling who were interviewed again after test disclosure, 81.9% had not heard about ES/GS in the meantime; however, the proportion of participants opting for disclosure of any variants was lower (116; 64.8%), with 36 (20.1%) changing opinion compared to the first interview. Based on these findings, we conclude that genetic counseling for ES/GS should involve enhanced education and decision-making support to enable informed consent to SFs disclosure.


Asunto(s)
Exoma , Calidad de Vida , Femenino , Pruebas Genéticas , Humanos , Italia , Masculino , Secuenciación del Exoma
16.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34281261

RESUMEN

Inherited retinal diseases (IRDs) are a heterogeneous group of conditions that include retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA) and early-onset severe retinal dystrophy (EO[S]RD), which differ in severity and age of onset. IRDs are caused by mutations in >250 genes. Variants in the RPE65 gene account for 0.6-6% of RP and 3-16% of LCA/EORD cases. Voretigene neparvovec is a gene therapy approved for the treatment of patients with an autosomal recessive retinal dystrophy due to confirmed biallelic RPE65 variants (RPE65-IRDs). Therefore, the accurate molecular diagnosis of RPE65-IRDs is crucial to identify 'actionable' genotypes-i.e., genotypes that may benefit from the treatment-and is an integral part of patient management. To date, hundreds of RPE65 variants have been identified, some of which are classified as pathogenic or likely pathogenic, while the significance of others is yet to be established. In this review, we provide an overview of the genetic diagnostic workup needed to select patients that could be eligible for voretigene neparvovec treatment. Careful clinical characterization of patients by multidisciplinary teams of experts, combined with the availability of next-generation sequencing approaches, can accelerate patients' access to available therapeutic options.


Asunto(s)
Enfermedades Hereditarias del Ojo/genética , Enfermedades de la Retina/genética , cis-trans-Isomerasas/genética , Enfermedades Hereditarias del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo/terapia , Asesoramiento Genético , Pruebas Genéticas/métodos , Terapia Genética , Variación Genética , Genotipo , Humanos , Mutación , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/terapia
17.
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
19.
Genet Med ; 22(5): 867-877, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31949313

RESUMEN

PURPOSE: To investigate if specific exon 38 or 39 KMT2D missense variants (MVs) cause a condition distinct from Kabuki syndrome type 1 (KS1). METHODS: Multiple individuals, with MVs in exons 38 or 39 of KMT2D that encode a highly conserved region of 54 amino acids flanked by Val3527 and Lys3583, were identified and phenotyped. Functional tests were performed to study their pathogenicity and understand the disease mechanism. RESULTS: The consistent clinical features of the affected individuals, from seven unrelated families, included choanal atresia, athelia or hypoplastic nipples, branchial sinus abnormalities, neck pits, lacrimal duct anomalies, hearing loss, external ear malformations, and thyroid abnormalities. None of the individuals had intellectual disability. The frequency of clinical features, objective software-based facial analysis metrics, and genome-wide peripheral blood DNA methylation patterns in these patients were significantly different from that of KS1. Circular dichroism spectroscopy indicated that these MVs perturb KMT2D secondary structure through an increased disordered to ɑ-helical transition. CONCLUSION: KMT2D MVs located in a specific region spanning exons 38 and 39 and affecting highly conserved residues cause a novel multiple malformations syndrome distinct from KS1. Unlike KMT2D haploinsufficiency in KS1, these MVs likely result in disease through a dominant negative mechanism.


Asunto(s)
Anomalías Múltiples , Enfermedades Hematológicas , Enfermedades Vestibulares , Anomalías Múltiples/genética , Cara/anomalías , Enfermedades Hematológicas/diagnóstico , Enfermedades Hematológicas/genética , Humanos , Mutación , Enfermedades Vestibulares/diagnóstico , Enfermedades Vestibulares/genética
20.
Clin Genet ; 98(5): 477-485, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32725632

RESUMEN

Developmental and epileptic encephalopathies (DEE) encompass rare, sporadic neurodevelopmental disorders and usually with pediatric onset. As these conditions are characterized by marked clinical and genetic heterogeneity, whole-exome sequencing (WES) represents the strategy of choice for the molecular diagnosis. While its usefulness is well established in pediatric DEE cohorts, our study is aimed at assessing the WES feasibility in adult DEE patients who experienced a diagnostic odyssey prior to the advent of this technique. We analyzed exomes from 71 unrelated adult DEE patients, consecutively recruited from an Italian cohort for the EPI25 Project. All patients underwent accurate clinical and electrophysiological characterization. An overwhelming percentage (90.1%) had already undergone negative genetic testing. Variants were classified according to the American College of Medical Genetics and Genomics guidelines. WES disclosed 24 (likely) pathogenic variants among 18 patients in epilepsy-related genes with either autosomal dominant, recessive or X-linked inheritance. Ten of these were novel. We obtained a diagnostic yield of 25.3%, higher among patients with brain malformations, early-onset epilepsy and dysmorphisms. Despite a median diagnostic delay of 38.7 years, WES analysis provided the long-awaited diagnosis for 18 adult patients, which also had an impact on the clinical management of 50% of them.


Asunto(s)
Arginino-ARNt Ligasa/genética , Proteínas del Citoesqueleto/genética , Epilepsia/genética , Predisposición Genética a la Enfermedad , Trastornos del Neurodesarrollo/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Diagnóstico Tardío , Epilepsia/diagnóstico , Epilepsia/patología , Exoma/genética , Femenino , Pruebas Genéticas , Genómica , Humanos , Lactante , Masculino , Persona de Mediana Edad , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/patología , Secuenciación del Exoma/métodos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA