Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 172(5): 924-936.e11, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474920

RESUMEN

Certain mutations can cause proteins to accumulate in neurons, leading to neurodegeneration. We recently showed, however, that upregulation of a wild-type protein, Ataxin1, caused by haploinsufficiency of its repressor, the RNA-binding protein Pumilio1 (PUM1), also causes neurodegeneration in mice. We therefore searched for human patients with PUM1 mutations. We identified eleven individuals with either PUM1 deletions or de novo missense variants who suffer a developmental syndrome (Pumilio1-associated developmental disability, ataxia, and seizure; PADDAS). We also identified a milder missense mutation in a family with adult-onset ataxia with incomplete penetrance (Pumilio1-related cerebellar ataxia, PRCA). Studies in patient-derived cells revealed that the missense mutations reduced PUM1 protein levels by ∼25% in the adult-onset cases and by ∼50% in the infantile-onset cases; levels of known PUM1 targets increased accordingly. Changes in protein levels thus track with phenotypic severity, and identifying posttranscriptional modulators of protein expression should identify new candidate disease genes.


Asunto(s)
Discapacidades del Desarrollo/genética , Predisposición Genética a la Enfermedad , Haploinsuficiencia/genética , Mutación/genética , Proteínas de Unión al ARN/genética , Convulsiones/genética , Adolescente , Adulto , Edad de Inicio , Anciano de 80 o más Años , Animales , Secuencia de Bases , Niño , Preescolar , Discapacidades del Desarrollo/diagnóstico por imagen , Evolución Molecular , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Lactante , Masculino , Ratones , Persona de Mediana Edad , Mutación Missense/genética , Neuronas/metabolismo , Neuronas/patología , Linaje , Estabilidad Proteica , Convulsiones/diagnóstico por imagen
2.
J Med Genet ; 60(6): 587-596, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36379543

RESUMEN

BACKGROUND: SHROOM4 is thought to play an important role in cytoskeletal modification and development of the early nervous system. Previously, single-nucleotide variants (SNVs) or copy number variations (CNVs) in SHROOM4 have been associated with the neurodevelopmental disorder Stocco dos Santos syndrome, but not with congenital anomalies of the urinary tract and the visceral or the cardiovascular system. METHODS: Here, exome sequencing and CNV analyses besides expression studies in zebrafish and mouse and knockdown (KD) experiments using a splice blocking morpholino in zebrafish were performed to study the role of SHROOM4 during embryonic development. RESULTS: In this study, we identified putative disease-causing SNVs and CNVs in SHROOM4 in six individuals from four families with congenital anomalies of the urinary tract and the anorectal, cardiovascular and central nervous systems (CNS). Embryonic mouse and zebrafish expression studies showed Shroom4 expression in the upper and lower urinary tract, the developing cloaca, the heart and the cerebral CNS. KD studies in zebrafish larvae revealed pronephric cysts, anomalies of the cloaca and the heart, decreased eye-to-head ratio and higher mortality compared with controls. These phenotypes could be rescued by co-injection of human wild-type SHROOM4 mRNA and morpholino. CONCLUSION: The identified SNVs and CNVs in affected individuals with congenital anomalies of the urinary tract, the anorectal, the cardiovascular and the central nervous systems, and subsequent embryonic mouse and zebrafish studies suggest SHROOM4 as a developmental gene for different organ systems.


Asunto(s)
Sistema Cardiovascular , Sistema Urinario , Embarazo , Femenino , Humanos , Animales , Ratones , Pez Cebra/genética , Variaciones en el Número de Copia de ADN , Morfolinos , Sistema Urinario/anomalías , Sistema Nervioso Central
3.
J Med Genet ; 60(6): 597-607, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36328423

RESUMEN

BACKGROUND: Heterozygous disruptions of FOXP2 were the first identified molecular cause for severe speech disorder: childhood apraxia of speech (CAS), and yet few cases have been reported, limiting knowledge of the condition. METHODS: Here we phenotyped 28 individuals from 17 families with pathogenic FOXP2-only variants (12 loss-of-function, five missense variants; 14 males; aged 2 to 62 years). Health and development (cognitive, motor, social domains) were examined, including speech and language outcomes with the first cross-linguistic analysis of English and German. RESULTS: Speech disorders were prevalent (23/25, 92%) and CAS was most common (22/25, 88%), with similar speech presentations across English and German. Speech was still impaired in adulthood, and some speech sounds (eg, 'th', 'r', 'ch', 'j') were never acquired. Language impairments (21/25, 84%) ranged from mild to severe. Comorbidities included feeding difficulties in infancy (10/26, 38%), fine (13/26, 50%) and gross (13/26, 50%) motor impairment, anxiety (5/27, 19%), depression (6/27, 22%) and sleep disturbance (10/24, 42%). Physical features were common (22/27, 81%) but with no consistent pattern. Cognition ranged from average to mildly impaired and was incongruent with language ability; for example, seven participants with severe language disorder had average non-verbal cognition. CONCLUSIONS: Although we identify an increased prevalence of conditions like anxiety, depression and sleep disturbance, we confirm that the consequences of FOXP2 dysfunction remain relatively specific to speech disorder, as compared with other recently identified monogenic conditions associated with CAS. Thus, our findings reinforce that FOXP2 provides a valuable entry point for examining the neurobiological bases of speech disorder.


Asunto(s)
Apraxias , Trastornos del Lenguaje , Masculino , Humanos , Niño , Trastornos del Habla/genética , Trastornos del Lenguaje/epidemiología , Trastornos del Lenguaje/genética , Habla , Apraxias/genética , Mutación Missense/genética , Factores de Transcripción Forkhead/genética
4.
Am J Med Genet A ; 185(4): 1261-1265, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33577136

RESUMEN

Haploinsufficiency of AUTS2 has been associated with neurodevelopmental disorders and dysmorphic features (MIM # 615834). More than 50 patients have been described, mostly carrying de novo deletions of one or more exons, including eight patients with exon 6 deletions. We report on two siblings, a girl and a boy aged 11 and 13 years, in whom the same pathogenic 85 kb deletion on 7q11.22 encompassing exon 6 of AUTS2 by SNP array analysis was identified. Both children had typical symptoms of AUTS2 syndrome such as intellectual impairment and behavioral problems, but with markedly different expression. SNP array analysis excluded the deletion in blood samples of both parents and a healthy brother. Conventional karyotyping of both parents and additional FISH analyses, marking the flanking regions of the deletion, did not show any structural rearrangements involving 7q11.22. A germ cell mosaicism was suggested as the most probable explanation for occurrence of the same deletion in these two siblings. To our knowledge this is the first report of germ cell mosaicism for AUTS2 syndrome. It additionally provides further evidence of intrafamilial phenotypic variability in AUTS2 syndrome and adds clinical information to the phenotypic spectrum of patients with AUTS2 exon 6 deletions.


Asunto(s)
Anomalías Múltiples/genética , Proteínas del Citoesqueleto/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Adolescente , Niño , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/patología , Exones/genética , Femenino , Células Germinativas/metabolismo , Células Germinativas/patología , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Mosaicismo , Eliminación de Secuencia/genética
5.
Am J Med Genet A ; 185(2): 549-554, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33191647

RESUMEN

Silver-Russell syndrome (SRS) is characterized by pre- and postnatal growth deficiency. It is most often caused by hypomethylation of the paternal imprinting center 1 of chromosome 11p15.5. In contrast, Sotos syndrome is an overgrowth syndrome that results either from pathogenic NSD1 gene variants or copy number variations affecting the NSD1 gene. Here, we report on a 6 month-old boy with severe short stature, relative macrocephaly, severe feeding difficulties with underweight, muscular hypotonia, motor delay, medullary nephrocalcinosis, bilateral sensorineural hearing impairment and facial dysmorphisms. SNP array revealed a 2.1 Mb de novo interstitial deletion of 5q35.2q35.3 encompassing the NSD1 gene. As Sotos syndrome could not satisfactorily explain his symptoms, diagnostic testing for SRS was initiated. It demonstrated hypomethylation of the imprinting center 1 of chromosome 11p15.5 confirming the clinically suspected SRS. We compared the symptoms of our patient with the typical clinical features of individuals with SRS and Sotos syndrome, respectively. To our knowledge, this is the first study reporting the very unusual coincidence of both Sotos syndrome and SRS in the same patient.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Síndrome de Silver-Russell/genética , Síndrome de Sotos/genética , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Impresión Genómica/genética , Humanos , Lactante , Recién Nacido , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Síndrome de Silver-Russell/complicaciones , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/patología , Síndrome de Sotos/complicaciones , Síndrome de Sotos/diagnóstico , Síndrome de Sotos/patología
6.
Neurogenetics ; 21(2): 121-133, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31940116

RESUMEN

Biallelic variants in POLR3A cause 4H leukodystrophy, characterized by hypomyelination in combination with cerebellar and pyramidal signs and variable non-neurological manifestations. Basal ganglia are spared in 4H leukodystrophy, and dystonia is not prominent. Three patients with variants in POLR3A, an atypical presentation with dystonia, and MR involvement of putamen and caudate nucleus (striatum) and red nucleus have previously been reported. Genetic, clinical findings and 18 MRI scans from nine patients with homozygous or compound heterozygous POLR3A variants and predominant striatal changes were retrospectively reviewed in order to characterize the striatal variant of POLR3A-associated disease. Prominent extrapyramidal involvement was the predominant clinical sign in all patients. The three youngest children were severely affected with muscle hypotonia, impaired head control, and choreic movements. Presentation of the six older patients was milder. Two brothers diagnosed with juvenile parkinsonism were homozygous for the c.1771-6C > G variant in POLR3A; the other seven either carried c.1771-6C > G (n = 1) or c.1771-7C > G (n = 7) together with another variant (missense, synonymous, or intronic). Striatal T2-hyperintensity and atrophy together with involvement of the superior cerebellar peduncles were characteristic. Additional MRI findings were involvement of dentate nuclei, hila, or peridentate white matter (3, 6, and 4/9), inferior cerebellar peduncles (6/9), red nuclei (2/9), and abnormal myelination of pyramidal and visual tracts (6/9) but no frank hypomyelination. Clinical and MRI findings in patients with a striatal variant of POLR3A-related disease are distinct from 4H leukodystrophy and associated with one of two intronic variants, c.1771-6C > G or c.1771-7C > G, in combination with another POLR3A variant.


Asunto(s)
Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Mutación , Neostriado/patología , ARN Polimerasa III/genética , Adulto , Ganglios Basales/patología , Encéfalo/patología , Preescolar , Femenino , Humanos , Lactante , Masculino , Sustancia Blanca/patología , Adulto Joven
7.
Genet Med ; 22(3): 610-621, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31761904

RESUMEN

PURPOSE: Pathogenic variants in neuroblastoma-amplified sequence (NBAS) cause an autosomal recessive disorder with a wide range of symptoms affecting liver, skeletal system, and brain, among others. There is a continuously growing number of patients but a lack of systematic and quantitative analysis. METHODS: Individuals with biallelic variants in NBAS were recruited within an international, multicenter study, including novel and previously published patients. Clinical variables were analyzed with log-linear models and visualized by mosaic plots; facial profiles were investigated via DeepGestalt. The structure of the NBAS protein was predicted using computational methods. RESULTS: One hundred ten individuals from 97 families with biallelic pathogenic NBAS variants were identified, including 26 novel patients with 19 previously unreported variants, giving a total number of 86 variants. Protein modeling redefined the ß-propeller domain of NBAS. Based on the localization of missense variants and in-frame deletions, three clinical subgroups arise that differ significantly regarding main clinical features and are directly related to the affected region of the NBAS protein: ß-propeller (combined phenotype), Sec39 (infantile liver failure syndrome type 2/ILFS2), and C-terminal (short stature, optic atrophy, and Pelger-Huët anomaly/SOPH). CONCLUSION: We define clinical subgroups of NBAS-associated disease that can guide patient management and point to domain-specific functions of NBAS.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Predisposición Genética a la Enfermedad , Proteínas de Neoplasias/genética , Alelos , Encéfalo/patología , Niño , Preescolar , Femenino , Enfermedades Genéticas Congénitas/patología , Humanos , Lactante , Hígado/patología , Trasplante de Hígado/efectos adversos , Masculino , Músculo Esquelético/patología , Mutación Missense/genética , Fenotipo
8.
Hum Mol Genet ; 26(23): 4689-4698, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973399

RESUMEN

The rare recessive developmental disorder Trichothiodystrophy (TTD) is characterized by brittle hair and nails. Patients also present a variable set of poorly explained additional clinical features, including ichthyosis, impaired intelligence, developmental delay and anemia. About half of TTD patients are photosensitive due to inherited defects in the DNA repair and transcription factor II H (TFIIH). The pathophysiological contributions of unrepaired DNA lesions and impaired transcription have not been dissected yet. Here, we functionally characterize the consequence of a homozygous missense mutation in the general transcription factor II E, subunit 2 (GTF2E2/TFIIEß) of two unrelated non-photosensitive TTD (NPS-TTD) families. We demonstrate that mutant TFIIEß strongly reduces the total amount of the entire TFIIE complex, with a remarkable temperature-sensitive transcription defect, which strikingly correlates with the phenotypic aggravation of key clinical symptoms after episodes of high fever. We performed induced pluripotent stem (iPS) cell reprogramming of patient fibroblasts followed by in vitro erythroid differentiation to translate the intriguing molecular defect to phenotypic expression in relevant tissue, to disclose the molecular basis for some specific TTD features. We observed a clear hematopoietic defect during late-stage differentiation associated with hemoglobin subunit imbalance. These new findings of a DNA repair-independent transcription defect and tissue-specific malfunctioning provide novel mechanistic insight into the etiology of TTD.


Asunto(s)
Factores de Transcripción TFII/genética , Síndromes de Tricotiodistrofia/genética , Diferenciación Celular/genética , Reprogramación Celular/genética , ADN Helicasas/genética , Reparación del ADN , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Mutación , Mutación Missense , Especificidad de Órganos , Linaje , Factores de Transcripción TFII/metabolismo , Transcripción Genética , Síndromes de Tricotiodistrofia/metabolismo , Síndromes de Tricotiodistrofia/patología
9.
Am J Hum Genet ; 98(3): 579-587, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26942290

RESUMEN

Encephalocraniocutaneous lipomatosis (ECCL) is a sporadic condition characterized by ocular, cutaneous, and central nervous system anomalies. Key clinical features include a well-demarcated hairless fatty nevus on the scalp, benign ocular tumors, and central nervous system lipomas. Seizures, spasticity, and intellectual disability can be present, although affected individuals without seizures and with normal intellect have also been reported. Given the patchy and asymmetric nature of the malformations, ECCL has been hypothesized to be due to a post-zygotic, mosaic mutation. Despite phenotypic overlap with several other disorders associated with mutations in the RAS-MAPK and PI3K-AKT pathways, the molecular etiology of ECCL remains unknown. Using exome sequencing of DNA from multiple affected tissues from five unrelated individuals with ECCL, we identified two mosaic mutations, c.1638C>A (p.Asn546Lys) and c.1966A>G (p.Lys656Glu) within the tyrosine kinase domain of FGFR1, in two affected individuals each. These two residues are the most commonly mutated residues in FGFR1 in human cancers and are associated primarily with CNS tumors. Targeted resequencing of FGFR1 in multiple tissues from an independent cohort of individuals with ECCL identified one additional individual with a c.1638C>A (p.Asn546Lys) mutation in FGFR1. Functional studies of ECCL fibroblast cell lines show increased levels of phosphorylated FGFRs and phosphorylated FRS2, a direct substrate of FGFR1, as well as constitutive activation of RAS-MAPK signaling. In addition to identifying the molecular etiology of ECCL, our results support the emerging overlap between mosaic developmental disorders and tumorigenesis.


Asunto(s)
Oftalmopatías/genética , Lipomatosis/genética , Síndromes Neurocutáneos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Adolescente , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/diagnóstico , Neoplasias del Sistema Nervioso Central/genética , Preescolar , Exoma , Ojo/fisiopatología , Oftalmopatías/diagnóstico , Femenino , Humanos , Lactante , Lipomatosis/diagnóstico , Masculino , Mutación , Mutación Missense , Síndromes Neurocutáneos/diagnóstico , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Convulsiones/genética , Análisis de Secuencia de ADN
10.
J Appl Res Intellect Disabil ; 32(5): 1129-1137, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30983121

RESUMEN

BACKGROUND: Caring for a child with intellectual disability (ID) has been associated with increased social and psychological burdens. Diagnostic and prognostic uncertainty may enhance emotional stress in families. METHOD: The present authors assessed the motivations, expectations, mental health, physical health and the quality of life of 194 parents whose children with intellectual disability were undergoing a genetic diagnostic workup. RESULTS: Most parents considered a diagnosis highly relevant for their own emotional relief, their child's therapies and education, or family planning. Parental mental health was significantly lower compared with the normative sample, but physical health was not different. The severity of the child's intellectual disability correlated negatively with their parents' mental and physical health, quality of life, and positively with parental anxiety. CONCLUSION: Healthcare providers should be aware of the disadvantages facing families with intellectually disabled children. Receiving practical, social and psychological support as well as genetic testing might be particularly relevant for families with severely disabled children.


Asunto(s)
Ansiedad/psicología , Discapacidades del Desarrollo/diagnóstico , Niños con Discapacidad , Pruebas Genéticas , Estado de Salud , Discapacidad Intelectual/diagnóstico , Padres/psicología , Calidad de Vida/psicología , Adolescente , Adulto , Niño , Preescolar , Discapacidades del Desarrollo/genética , Femenino , Humanos , Lactante , Discapacidad Intelectual/genética , Masculino , Persona de Mediana Edad , Adulto Joven
11.
Am J Med Genet C Semin Med Genet ; 178(4): 414-422, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30580480

RESUMEN

Oculocerebrocutaneous syndrome (OCCS) is a rare disorder characterized primarily by congenital skin, eye, and brain anomalies. The most distinctive findings are hypoplastic or aplastic skin defects; pedunculated, typically hamartomatous, or nodular skin appendages; cystic microphthalmia; and a combination of forebrain anomalies and a specific mid-hindbrain malformation. Based on a review of 40 patients with OCCS, existing clinical criteria have been revised. Because of the asymmetric and patchy distribution of features, lack of recurrence in families, male preponderance and completely skewed X-inactivation in one female, OCCS is hypothesized to result from postzygotic mosaic variants in an X-linked gene. Whole exome and genome sequencing on blood DNA in two patients failed to identify pathogenic variants so far. In view of the overlapping features, in particular of the brain, of OCCS and Aicardi syndrome, both may be pathogenetically related or even result from different variants in the same gene. For the elucidation of the cause of OCCS, exome or genome sequencing on multiple lesional tissues is the primary goal.


Asunto(s)
Anomalías Múltiples , Quistes del Sistema Nervioso Central/patología , Anomalías del Ojo/patología , Dedos/anomalías , Anomalías Cutáneas/patología , Dedos/patología , Humanos , Fenotipo
12.
Brain ; 140(10): 2610-2622, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28969385

RESUMEN

Mutations of genes within the phosphatidylinositol-3-kinase (PI3K)-AKT-MTOR pathway are well known causes of brain overgrowth (megalencephaly) as well as segmental cortical dysplasia (such as hemimegalencephaly, focal cortical dysplasia and polymicrogyria). Mutations of the AKT3 gene have been reported in a few individuals with brain malformations, to date. Therefore, our understanding regarding the clinical and molecular spectrum associated with mutations of this critical gene is limited, with no clear genotype-phenotype correlations. We sought to further delineate this spectrum, study levels of mosaicism and identify genotype-phenotype correlations of AKT3-related disorders. We performed targeted sequencing of AKT3 on individuals with these phenotypes by molecular inversion probes and/or Sanger sequencing to determine the type and level of mosaicism of mutations. We analysed all clinical and brain imaging data of mutation-positive individuals including neuropathological analysis in one instance. We performed ex vivo kinase assays on AKT3 engineered with the patient mutations and examined the phospholipid binding profile of pleckstrin homology domain localizing mutations. We identified 14 new individuals with AKT3 mutations with several phenotypes dependent on the type of mutation and level of mosaicism. Our comprehensive clinical characterization, and review of all previously published patients, broadly segregates individuals with AKT3 mutations into two groups: patients with highly asymmetric cortical dysplasia caused by the common p.E17K mutation, and patients with constitutional AKT3 mutations exhibiting more variable phenotypes including bilateral cortical malformations, polymicrogyria, periventricular nodular heterotopia and diffuse megalencephaly without cortical dysplasia. All mutations increased kinase activity, and pleckstrin homology domain mutants exhibited enhanced phospholipid binding. Overall, our study shows that activating mutations of the critical AKT3 gene are associated with a wide spectrum of brain involvement ranging from focal or segmental brain malformations (such as hemimegalencephaly and polymicrogyria) predominantly due to mosaic AKT3 mutations, to diffuse bilateral cortical malformations, megalencephaly and heterotopia due to constitutional AKT3 mutations. We also provide the first detailed neuropathological examination of a child with extreme megalencephaly due to a constitutional AKT3 mutation. This child has one of the largest documented paediatric brain sizes, to our knowledge. Finally, our data show that constitutional AKT3 mutations are associated with megalencephaly, with or without autism, similar to PTEN-related disorders. Recognition of this broad clinical and molecular spectrum of AKT3 mutations is important for providing early diagnosis and appropriate management of affected individuals, and will facilitate targeted design of future human clinical trials using PI3K-AKT pathway inhibitors.


Asunto(s)
Discapacidades del Desarrollo/genética , Megalencefalia/genética , Mutación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Encéfalo/diagnóstico por imagen , Niño , Discapacidades del Desarrollo/diagnóstico por imagen , Discapacidades del Desarrollo/patología , Femenino , Estudios de Asociación Genética , Células HEK293 , Humanos , Inmunoprecipitación , Imagen por Resonancia Magnética , Masculino , Megalencefalia/diagnóstico por imagen , Megalencefalia/patología , Mutagénesis Sitio-Dirigida/métodos , Fosfatidilinositoles/metabolismo , Transfección
13.
J Med Genet ; 54(1): 64-72, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27572252

RESUMEN

BACKGROUND: Disruptions of the FOXP2 gene, encoding a forkhead transcription factor, are the first known monogenic cause of a speech and language disorder. So far, mainly chromosomal rearrangements such as translocations or larger deletions affecting FOXP2 have been reported. Intragenic deletions or convincingly pathogenic point mutations in FOXP2 have up to date only been reported in three families. We thus aimed at a further characterisation of the mutational and clinical spectrum. METHODS: Chromosomal microarray testing, trio exome sequencing, multigene panel sequencing and targeted sequencing of FOXP2 were performed in individuals with variable developmental disorders, and speech and language deficits. RESULTS: We identified four different truncating mutations, two novel missense mutations within the forkhead domain and an intragenic deletion in FOXP2 in 14 individuals from eight unrelated families. Mutations occurred de novo in four families and were inherited from an affected parent in the other four. All index patients presented with various manifestations of language and speech impairment. Apart from two individuals with normal onset of speech, age of first words was between 4 and 7 years. Articulation difficulties such as slurred speech, dyspraxia, stuttering and poor pronunciation were frequently noted. Motor development was normal or only mildly delayed. Mild cognitive impairment was reported for most individuals. CONCLUSIONS: By identifying intragenic deletions or mutations in 14 individuals from eight unrelated families with variable developmental delay/cognitive impairment and speech and language deficits, we considerably broaden the mutational and clinical spectrum associated with aberrations in FOXP2.


Asunto(s)
Factores de Transcripción Forkhead/genética , Trastornos del Lenguaje/genética , Mutación Missense/genética , Mutación Puntual/genética , Eliminación de Secuencia/genética , Trastornos del Habla/genética , Discapacidades del Desarrollo/genética , Humanos , Masculino , Linaje , Habla/fisiología
14.
Mol Genet Metab ; 121(4): 297-307, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28688840

RESUMEN

Whole exome sequencing (WES) is well established in research and is now being introduced into clinically indicated diagnostics (so-called clinical exomes). We evaluated the diagnostic yield and clinical implications of WES in 72 patients from 60 families with undiagnosed neurodevelopmental disorders (NDD), neurometabolic disorders, and dystonias. Pathogenic or likely pathogenic variants leading to a molecular diagnosis could be identified in 21 of the 60 families (overall 35%, in 36% of patients with NDD, in 43% of patients with neurometabolic disorders, in 25% of patients with dystonias). In one family two coexisting autosomal recessive diseases caused by homozygous pathogenic variants in two different genes were diagnosed. In another family, a homozygous frameshift variant in STRADA was found to cause a severe NDD with early onset epilepsy, brain anomalies, hypotonia, heart defect, nephrocalcinosis, macrocephaly and distinctive facies so far designated as PMSE (polyhydramnios, megalencephaly, symptomatic epilepsy) syndrome. In 7 of the 21 families with a molecular diagnosis the pathogenic variants were only identified by clinical follow-up, manual reevaluation of the literature, a change of filter setting, and/or reconsideration of inheritance pattern. Most importantly, clinical implications included management changes in 8 cases and impact on family planning in 20 families with a molecular diagnosis. This study shows that reevaluation and follow-up can improve the diagnostic rate and that WES results have important implications on medical management and family planning. Furthermore, we could confirm STRADA as a gene associated with syndromic ID but find it questionable if the current designation as PMSE depicts the most important clinical features.


Asunto(s)
Exoma , Técnicas de Diagnóstico Molecular/métodos , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/genética , Servicios de Planificación Familiar , Femenino , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Masculino , Trastornos del Neurodesarrollo/fisiopatología , Linaje , Embarazo , Análisis de Secuencia de ADN , Secuenciación del Exoma
15.
Am J Med Genet A ; 173(7): 1878-1886, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28489334

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) is a group of neurodegenerative disorders characterized by iron accumulation in the basal ganglia. Recently, mutations in CoA synthase (COASY) have been identified as a cause of a novel NBIA subtype (COASY Protein-Associated Neurodegeneration, CoPAN) in two patients with dystonic paraparesis, parkinsonian features, cognitive impairment, behavior abnormalities, and axonal neuropathy. COASY encodes an enzyme required for Coenzyme A (CoA) biosynthesis. Using whole exome sequencing (WES) we identified compound heterozygous COASY mutations in two siblings with intellectual disability, ataxic gait, progressive spasticity, and obsessive-compulsive behavior. The "eye-of-the tiger-sign," a characteristic hypointense spot within the hyperintense globi pallidi on MRI found in the most common subtype of NBIA (Pantothenate Kinase-Associated Neurodegeneration, PKAN), was not present. Instead, bilateral hyperintensity and swelling of caudate nucleus, putamen, and thalamus were found. In addition, our patients showed a small corpus callosum and frontotemporal and parietal white matter changes, expanding the brain phenotype of patients with CoPAN. Metabolic investigations showed increased free carnitine and decreased acylcarnitines in the patients dried blood samples. Carnitine palmitoyl transferase 1 (CPT1) deficiency was excluded by further enzymatic and metabolic investigations. As CoA and its derivate Acetyl-CoA play an essential role in fatty acid metabolism, we assume that abnormal acylcarnitine profiles are a result of the COASY mutations. This report not only illustrates that WES is a powerful tool to elucidate the etiology of rare genetic diseases, but also identifies unique neuroimaging and metabolic findings that may be key features for an early diagnosis of CoPAN.

16.
Am J Med Genet A ; 173(5): 1369-1373, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28371085

RESUMEN

Recently, de novo heterozygous variants in DDX3X have been reported in about 1.5% of 2659 females with previously unexplained intellectual disability (ID). We report on the identification of DDX3X variants in two unrelated girls with clinical features of Toriello-Carey Syndrome (T-CS). In patient 1, the recurrent variant c.1703C>T; p.(P568L) was identified when reconsidering X-linked de novo heterozygous variants in exome sequencing data. In patient 2, the DDX3X variant c.1600C>G; p.(R534G) was also detected by exome sequencing. Based on these data, de novo heterozygous DDX3X variants should be considered not only in females with unexplained ID, but also in individuals with a clinical diagnosis of T-CS.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Anomalías Craneofaciales/genética , ARN Helicasas DEAD-box/genética , Cardiopatías Congénitas/genética , Discapacidad Intelectual/genética , Deformidades Congénitas de las Extremidades/genética , Síndrome de Pierre Robin/genética , Anomalías Urogenitales/genética , Agenesia del Cuerpo Calloso/diagnóstico , Agenesia del Cuerpo Calloso/fisiopatología , Niño , Preescolar , Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/fisiopatología , Exoma/genética , Femenino , Genes Ligados a X , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/fisiopatología , Heterocigoto , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/fisiopatología , Deformidades Congénitas de las Extremidades/diagnóstico , Deformidades Congénitas de las Extremidades/fisiopatología , Mutación , Fenotipo , Síndrome de Pierre Robin/diagnóstico , Síndrome de Pierre Robin/fisiopatología , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/fisiopatología
17.
J Med Genet ; 53(2): 98-110, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26502894

RESUMEN

BACKGROUND: Orodental diseases include several clinically and genetically heterogeneous disorders that can present in isolation or as part of a genetic syndrome. Due to the vast number of genes implicated in these disorders, establishing a molecular diagnosis can be challenging. We aimed to develop a targeted next-generation sequencing (NGS) assay to diagnose mutations and potentially identify novel genes mutated in this group of disorders. METHODS: We designed an NGS gene panel that targets 585 known and candidate genes in orodental disease. We screened a cohort of 101 unrelated patients without a molecular diagnosis referred to the Reference Centre for Oro-Dental Manifestations of Rare Diseases, Strasbourg, France, for a variety of orodental disorders including isolated and syndromic amelogenesis imperfecta (AI), isolated and syndromic selective tooth agenesis (STHAG), isolated and syndromic dentinogenesis imperfecta, isolated dentin dysplasia, otodental dysplasia and primary failure of tooth eruption. RESULTS: We discovered 21 novel pathogenic variants and identified the causative mutation in 39 unrelated patients in known genes (overall diagnostic rate: 39%). Among the largest subcohorts of patients with isolated AI (50 unrelated patients) and isolated STHAG (21 unrelated patients), we had a definitive diagnosis in 14 (27%) and 15 cases (71%), respectively. Surprisingly, COL17A1 mutations accounted for the majority of autosomal-dominant AI cases. CONCLUSIONS: We have developed a novel targeted NGS assay for the efficient molecular diagnosis of a wide variety of orodental diseases. Furthermore, our panel will contribute to better understanding the contribution of these genes to orodental disease. TRIAL REGISTRATION NUMBERS: NCT01746121 and NCT02397824.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación , Anomalías Dentarias/genética , Amelogénesis Imperfecta/genética , Autoantígenos/genética , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 11/genética , Estudios de Cohortes , Coloboma/genética , Displasia de la Dentina/genética , Francia , Pérdida Auditiva Sensorineural/genética , Humanos , Colágenos no Fibrilares/genética , Reproducibilidad de los Resultados , Colágeno Tipo XVII
18.
Am J Med Genet A ; 170(6): 1502-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27016154

RESUMEN

Intellectual disability (ID) with cerebellar ataxia comprises a genetically heterogeneous group of neurodevelopmental disorders. We identified a homozygous frameshift mutation in CWF19L1 (c.467delC; p.(P156Hfs*33)) by a combination of linkage analysis and Whole Exome Sequencing in a consanguineous Turkish family with a 9-year-old boy affected by early onset cerebellar ataxia and mild ID. Serial MRI showed mildly progressive cerebellar atrophy. Absent C19L1 protein expression in lymphoblastoid cell lines strongly suggested that c.467delC is a disease-causing alteration. One further pregnancy of the mother had been terminated at 22 weeks of gestation because of a small cerebellum and agenesis of corpus callosum. The homozygous CWF19L1 variant was also present in the fetus. Postmortem examination of the fetus in addition showed unilateral hexadactyly and vertebral malformations. These features have not been reported and may represent an expansion of the CWF19L1-related phenotypic spectrum, but could also be due to another, possibly autosomal recessive disorder. The exact function of the evolutionarily highly conserved C19L1 protein is unknown. So far, homozygous or compound heterozygous mutations in CWF19L1 have been identified in two Turkish siblings and a Dutch girl, respectively, affected by cerebellar ataxia and ID. A zebrafish model showed that CWF19L1 loss-of-function mutations result in abnormal cerebellar morphology and movement disorders. Our report corroborates that loss-of-function mutations in CWF19Ll lead to early onset cerebellar ataxia and (progressive) cerebellar atrophy. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cerebelo/anomalías , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Mutación , Malformaciones del Sistema Nervioso/diagnóstico , Malformaciones del Sistema Nervioso/genética , Encéfalo/anomalías , Niño , Hibridación Genómica Comparativa , Consanguinidad , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Estudios de Asociación Genética , Ligamiento Genético , Humanos , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Radiografía
19.
Am J Med Genet A ; 167A(3): 553-62, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25691408

RESUMEN

In females, large duplications in Xp often lead to preferential inactivation of the aberrant X chromosome and a normal phenotype. Recently, a recurrent ∼4.5 Mb microduplication of Xp11.22-p11.23 was found in females with developmental delay/intellectual disability and other neurodevelopmental disorders (speech development disorder, epilepsy or EEG anomalies, autism spectrum disorder, or behavioral disorder). Unexpectedly, most of them showed preferential inactivation of the normal X chromosome. We describe five female patients carrying de novo Xp duplications encompassing p11.23. Patient 1 carried the recurrent microduplication Xp11.22-p11.23, her phenotype and X-chromosome inactivation (XI) pattern was consistent with previous reports. The other four patients had novel Xp duplications. Two were monozygotic twins with a similar phenotype to Patient 1 and unfavorable XI skewing carrying an overlapping ∼5 Mb duplication of Xp11.23-p11.3. Patient 4 showed a duplication of ∼5.5 Mb comparable to the twins but had a more severe phenotype and unskewed XI. Patient 5 had a ∼8.5 Mb duplication Xp11.23-p11.4 and presented with mild ID, epilepsy, behavioral problems, and inconsistent results of XI analysis. A comparison of phenotype, size and location of the duplications and XI patterns in Patients 1-5 and previously reported females with overlapping duplications provides further evidence that microduplications encompassing Xp11.23 are associated with ID and other neurodevelopmental disorders in females. To further assess the implication of XI for female carriers, we recommend systematic analysis of XI pattern in any female with X imbalances that are known or suspected to be pathogenic.


Asunto(s)
Duplicación Cromosómica , Cromosomas Humanos X , Trastornos de los Cromosomas Sexuales/genética , Inactivación del Cromosoma X , Adolescente , Adulto , Niño , Preescolar , Hibridación Genómica Comparativa , Facies , Femenino , Estudios de Asociación Genética , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple , Trastornos de los Cromosomas Sexuales/diagnóstico , Adulto Joven
20.
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA