Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Am J Hum Genet ; 108(6): 1069-1082, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34022130

RESUMEN

BCAS3 microtubule-associated cell migration factor (BCAS3) is a large, highly conserved cytoskeletal protein previously proposed to be critical in angiogenesis and implicated in human embryogenesis and tumorigenesis. Here, we established BCAS3 loss-of-function variants as causative for a neurodevelopmental disorder. We report 15 individuals from eight unrelated families with germline bi-allelic loss-of-function variants in BCAS3. All probands share a global developmental delay accompanied by pyramidal tract involvement, microcephaly, short stature, strabismus, dysmorphic facial features, and seizures. The human phenotype is less severe compared with the Bcas3 knockout mouse model and cannot be explained by angiogenic defects alone. Consistent with being loss-of-function alleles, we observed absence of BCAS3 in probands' primary fibroblasts. By comparing the transcriptomic and proteomic data based on probands' fibroblasts with those of the knockout mouse model, we identified similar dysregulated pathways resulting from over-representation analysis, while the dysregulation of some proposed key interactors could not be confirmed. Together with the results from a tissue-specific Drosophila loss-of-function model, we demonstrate a vital role for BCAS3 in neural tissue development.


Asunto(s)
Mutación con Pérdida de Función , Pérdida de Heterocigocidad , Proteínas de Neoplasias/genética , Trastornos del Neurodesarrollo/etiología , Adolescente , Adulto , Animales , Movimiento Celular , Niño , Preescolar , Drosophila , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Lactante , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Linaje , Proteoma/análisis , Adulto Joven
2.
Mov Disord ; 39(1): 141-151, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37964426

RESUMEN

BACKGROUND: The ITPR1 gene encodes the inositol 1,4,5-trisphosphate (IP3 ) receptor type 1 (IP3 R1), a critical player in cerebellar intracellular calcium signaling. Pathogenic missense variants in ITPR1 cause congenital spinocerebellar ataxia type 29 (SCA29), Gillespie syndrome (GLSP), and severe pontine/cerebellar hypoplasia. The pathophysiological basis of the different phenotypes is poorly understood. OBJECTIVES: We aimed to identify novel SCA29 and GLSP cases to define core phenotypes, describe the spectrum of missense variation across ITPR1, standardize the ITPR1 variant nomenclature, and investigate disease progression in relation to cerebellar atrophy. METHODS: Cases were identified using next-generation sequencing through the Deciphering Developmental Disorders study, the 100,000 Genomes project, and clinical collaborations. ITPR1 alternative splicing in the human cerebellum was investigated by quantitative polymerase chain reaction. RESULTS: We report the largest, multinational case series of 46 patients with 28 unique ITPR1 missense variants. Variants clustered in functional domains of the protein, especially in the N-terminal IP3 -binding domain, the carbonic anhydrase 8 (CA8)-binding region, and the C-terminal transmembrane channel domain. Variants outside these domains were of questionable clinical significance. Standardized transcript annotation, based on our ITPR1 transcript expression data, greatly facilitated analysis. Genotype-phenotype associations were highly variable. Importantly, while cerebellar atrophy was common, cerebellar volume loss did not correlate with symptom progression. CONCLUSIONS: This dataset represents the largest cohort of patients with ITPR1 missense variants, expanding the clinical spectrum of SCA29 and GLSP. Standardized transcript annotation is essential for future reporting. Our findings will aid in diagnostic interpretation in the clinic and guide selection of variants for preclinical studies. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Aniridia , Anhidrasas Carbónicas , Ataxia Cerebelosa , Discapacidad Intelectual , Trastornos del Movimiento , Degeneraciones Espinocerebelosas , Humanos , Ataxia Cerebelosa/genética , Mutación Missense/genética , Trastornos del Movimiento/complicaciones , Atrofia , Receptores de Inositol 1,4,5-Trifosfato/química , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética
3.
J Med Genet ; 59(4): 393-398, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33879512

RESUMEN

PURPOSE: The increased adoption of genomic strategies in the clinic makes it imperative for diagnostic laboratories to improve the efficiency of variant interpretation. Clinical exome sequencing (CES) is becoming a valuable diagnostic tool, capable of meeting the diagnostic demand imposed by the vast array of different rare monogenic disorders. We have assessed a clinician-led and phenotype-based approach for virtual gene panel generation for analysis of targeted CES in patients with rare disease in a single institution. METHODS: Retrospective survey of 400 consecutive cases presumed by clinicians to have rare monogenic disorders, referred on singleton basis for targeted CES. We evaluated diagnostic yield and variant workload to characterise the usefulness of a clinician-led approach for generation of virtual gene panels that can incorporate up to three different phenotype-driven gene selection methods. RESULTS: Abnormalities of the nervous system (54.5%), including intellectual disability, head and neck (19%), skeletal system (16%), ear (15%) and eye (15%) were the most common clinical features reported in referrals. Combined phenotype-driven strategies for virtual gene panel generation were used in 57% of cases. On average, 7.3 variants (median=5) per case were retained for clinical interpretation. The overall diagnostic rate of proband-only CES using personalised phenotype-driven virtual gene panels was 24%. CONCLUSIONS: Our results show that personalised virtual gene panels are a cost-effective approach for variant analysis of CES, maintaining diagnostic yield and optimising the use of resources for clinical genomic sequencing in the clinic.


Asunto(s)
Exoma , Enfermedades Raras , Exoma/genética , Humanos , Enfermedades Raras/genética , Estudios Retrospectivos , Secuenciación del Exoma , Carga de Trabajo
4.
Am J Hum Genet ; 100(3): 523-536, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28190456

RESUMEN

Phosphoinositides are small phospholipids that control diverse cellular downstream signaling events. Their spatial and temporal availability is tightly regulated by a set of specific lipid kinases and phosphatases. Congenital muscular dystrophies are hereditary disorders characterized by hypotonia and weakness from birth with variable eye and central nervous system involvement. In individuals exhibiting congenital muscular dystrophy, early-onset cataracts, and mild intellectual disability but normal cranial magnetic resonance imaging, we identified bi-allelic mutations in INPP5K, encoding inositol polyphosphate-5-phosphatase K. Mutations impaired phosphatase activity toward the phosphoinositide phosphatidylinositol (4,5)-bisphosphate or altered the subcellular localization of INPP5K. Downregulation of INPP5K orthologs in zebrafish embryos disrupted muscle fiber morphology and resulted in abnormal eye development. These data link congenital muscular dystrophies to defective phosphoinositide 5-phosphatase activity that is becoming increasingly recognized for its role in mediating pivotal cellular mechanisms contributing to disease.


Asunto(s)
Catarata/genética , Disfunción Cognitiva/genética , Distrofia Muscular de Cinturas/genética , Anomalías Musculoesqueléticas/genética , Monoéster Fosfórico Hidrolasas/genética , Adolescente , Adulto , Alelos , Animales , Encéfalo/patología , Niño , Preescolar , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Discapacidad Intelectual/genética , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/patología , Mutación , Linaje , Adulto Joven , Pez Cebra/embriología , Pez Cebra/genética
5.
Genet Med ; 22(7): 1215-1226, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32376980

RESUMEN

PURPOSE: Somatic variants in tumor necrosis factor receptor-associated factor 7 (TRAF7) cause meningioma, while germline variants have recently been identified in seven patients with developmental delay and cardiac, facial, and digital anomalies. We aimed to define the clinical and mutational spectrum associated with TRAF7 germline variants in a large series of patients, and to determine the molecular effects of the variants through transcriptomic analysis of patient fibroblasts. METHODS: We performed exome, targeted capture, and Sanger sequencing of patients with undiagnosed developmental disorders, in multiple independent diagnostic or research centers. Phenotypic and mutational comparisons were facilitated through data exchange platforms. Whole-transcriptome sequencing was performed on RNA from patient- and control-derived fibroblasts. RESULTS: We identified heterozygous missense variants in TRAF7 as the cause of a developmental delay-malformation syndrome in 45 patients. Major features include a recognizable facial gestalt (characterized in particular by blepharophimosis), short neck, pectus carinatum, digital deviations, and patent ductus arteriosus. Almost all variants occur in the WD40 repeats and most are recurrent. Several differentially expressed genes were identified in patient fibroblasts. CONCLUSION: We provide the first large-scale analysis of the clinical and mutational spectrum associated with the TRAF7 developmental syndrome, and we shed light on its molecular etiology through transcriptome studies.


Asunto(s)
Discapacidad Intelectual , Transcriptoma , Exoma , Células Germinativas , Humanos , Discapacidad Intelectual/genética , Mutación Missense , Fenotipo , Transcriptoma/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral
6.
Hum Mol Genet ; 26(3): 519-526, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28053047

RESUMEN

Haploinsufficiency in DYRK1A is associated with a recognizable developmental syndrome, though the mechanism of action of pathogenic missense mutations is currently unclear. Here we present 19 de novo mutations in this gene, including five missense mutations, identified by the Deciphering Developmental Disorder study. Protein structural analysis reveals that the missense mutations are either close to the ATP or peptide binding-sites within the kinase domain, or are important for protein stability, suggesting they lead to a loss of the protein's function mechanism. Furthermore, there is some correlation between the magnitude of the change and the severity of the resultant phenotype. A comparison of the distribution of the pathogenic mutations along the length of DYRK1A with that of natural variants, as found in the ExAC database, confirms that mutations in the N-terminal end of the kinase domain are more disruptive of protein function. In particular, pathogenic mutations occur in significantly closer proximity to the ATP and the substrate peptide than the natural variants. Overall, we suggest that de novo dominant mutations in DYRK1A account for nearly 0.5% of severe developmental disorders due to substantially reduced kinase function.


Asunto(s)
Trastorno Autístico/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Trastorno Autístico/patología , Discapacidades del Desarrollo/fisiopatología , Femenino , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual/patología , Masculino , Mutación , Mutación Missense , Linaje , Fenotipo , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Tirosina Quinasas/química , Relación Estructura-Actividad , Quinasas DyrK
7.
Hum Mutat ; 38(4): 426-438, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28058752

RESUMEN

Calcium (Ca2+ ) is a physiological key factor, and the precise modulation of free cytosolic Ca2+ levels regulates multiple cellular functions. Store-operated Ca2+ entry (SOCE) is a major mechanism controlling Ca2+ homeostasis, and is mediated by the concerted activity of the Ca2+ sensor STIM1 and the Ca2+ channel ORAI1. Dominant gain-of-function mutations in STIM1 or ORAI1 cause tubular aggregate myopathy (TAM) or Stormorken syndrome, whereas recessive loss-of-function mutations are associated with immunodeficiency. Here, we report the identification and functional characterization of novel ORAI1 mutations in TAM patients. We assess basal activity and SOCE of the mutant ORAI1 channels, and we demonstrate that the G98S and V107M mutations generate constitutively permeable ORAI1 channels, whereas T184M alters the channel permeability only in the presence of STIM1. These data indicate a mutation-dependent pathomechanism and a genotype/phenotype correlation, as the ORAI1 mutations associated with the most severe symptoms induce the strongest functional cellular effect. Examination of the non-muscle features of our patients strongly suggests that TAM and Stormorken syndrome are spectra of the same disease. Overall, our results emphasize the importance of SOCE in skeletal muscle physiology, and provide new insights in the pathomechanisms involving aberrant Ca2+ homeostasis and leading to muscle dysfunction.


Asunto(s)
Activación del Canal Iónico/genética , Mutación Missense , Miopatías Estructurales Congénitas/genética , Proteína ORAI1/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Calcio/metabolismo , Células Cultivadas , Dislexia/genética , Dislexia/metabolismo , Eritrocitos Anormales/metabolismo , Femenino , Células HEK293 , Humanos , Ictiosis/genética , Ictiosis/metabolismo , Masculino , Ratones Noqueados , Microscopía Fluorescente/métodos , Trastornos Migrañosos/genética , Trastornos Migrañosos/metabolismo , Miosis/genética , Miosis/metabolismo , Fatiga Muscular/genética , Miopatías Estructurales Congénitas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Linaje , Homología de Secuencia de Aminoácido , Bazo/anomalías , Bazo/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
8.
Am J Hum Genet ; 92(4): 605-13, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23541340

RESUMEN

Perrault syndrome is a genetically and clinically heterogeneous autosomal-recessive condition characterized by sensorineural hearing loss and ovarian failure. By a combination of linkage analysis, homozygosity mapping, and exome sequencing in three families, we identified mutations in CLPP as the likely cause of this phenotype. In each family, affected individuals were homozygous for a different pathogenic CLPP allele: c.433A>C (p.Thr145Pro), c.440G>C (p.Cys147Ser), or an experimentally demonstrated splice-donor-site mutation, c.270+4A>G. CLPP, a component of a mitochondrial ATP-dependent proteolytic complex, is a highly conserved endopeptidase encoded by CLPP and forms an element of the evolutionarily ancient mitochondrial unfolded-protein response (UPR(mt)) stress signaling pathway. Crystal-structure modeling suggests that both substitutions would alter the structure of the CLPP barrel chamber that captures unfolded proteins and exposes them to proteolysis. Together with the previous identification of mutations in HARS2, encoding mitochondrial histidyl-tRNA synthetase, mutations in CLPP expose dysfunction of mitochondrial protein homeostasis as a cause of Perrault syndrome.


Asunto(s)
Proteasas ATP-Dependientes/genética , Endopeptidasa Clp/genética , Exoma/genética , Genes Recesivos , Disgenesia Gonadal 46 XX/etiología , Pérdida Auditiva Sensorineural/etiología , Mitocondrias/enzimología , Mutación/genética , Proteasas ATP-Dependientes/metabolismo , Adenosina Trifosfato/metabolismo , Adolescente , Adulto , Femenino , Homocigoto , Humanos , Hibridación in Situ , Masculino , Mitocondrias/genética , Linaje , Fenotipo , Adulto Joven
9.
Nat Genet ; 39(9): 1134-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17676042

RESUMEN

Centronuclear myopathies are characterized by muscle weakness and abnormal centralization of nuclei in muscle fibers not secondary to regeneration. The severe neonatal X-linked form (myotubular myopathy) is due to mutations in the phosphoinositide phosphatase myotubularin (MTM1), whereas mutations in dynamin 2 (DNM2) have been found in some autosomal dominant cases. By direct sequencing of functional candidate genes, we identified homozygous mutations in amphiphysin 2 (BIN1) in three families with autosomal recessive inheritance. Two missense mutations affecting the BAR (Bin1/amphiphysin/RVS167) domain disrupt its membrane tubulation properties in transfected cells, and a partial truncation of the C-terminal SH3 domain abrogates the interaction with DNM2 and its recruitment to the membrane tubules. Our results suggest that mutations in BIN1 cause centronuclear myopathy by interfering with remodeling of T tubules and/or endocytic membranes, and that the functional interaction between BIN1 and DNM2 is necessary for normal muscle function and positioning of nuclei.


Asunto(s)
Dinamina II/genética , Enfermedades Musculares/genética , Mutación , Proteínas del Tejido Nervioso/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión/genética , Células COS , Línea Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Dinamina II/metabolismo , Femenino , Genes Recesivos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Haplotipos , Humanos , Masculino , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Proteínas del Tejido Nervioso/metabolismo , Polimorfismo de Nucleótido Simple , Unión Proteica , Homología de Secuencia de Aminoácido , Transfección
10.
Ann Rheum Dis ; 74(6): 1249-56, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24442880

RESUMEN

OBJECTIVES: Leri's pleonosteosis (LP) is an autosomal dominant rheumatic condition characterised by flexion contractures of the interphalangeal joints, limited motion of multiple joints, and short broad metacarpals, metatarsals and phalanges. Scleroderma-like skin thickening can be seen in some individuals with LP. We undertook a study to characterise the phenotype of LP and identify its genetic basis. METHODS AND RESULTS: Whole-genome single-nucleotide polymorphism genotyping in two families with LP defined microduplications of chromosome 8q22.1 as the cause of this condition. Expression analysis of dermal fibroblasts from affected individuals showed overexpression of two genes, GDF6 and SDC2, within the duplicated region, leading to dysregulation of genes that encode proteins of the extracellular matrix and downstream players in the transforming growth factor (TGF)-ß pathway. Western blot analysis revealed markedly decreased inhibitory SMAD6 levels in patients with LP. Furthermore, in a cohort of 330 systemic sclerosis cases, we show that the minor allele of a missense SDC2 variant, p.Ser71Thr, could confer protection against disease (p<1×10(-5)). CONCLUSIONS: Our work identifies the genetic cause of LP in these two families, demonstrates the phenotypic range of the condition, implicates dysregulation of extracellular matrix homoeostasis genes in its pathogenesis, and highlights the link between TGF-ß/SMAD signalling, growth/differentiation factor 6 and syndecan-2. We propose that LP is an additional member of the growing 'TGF-ß-pathies' group of musculoskeletal disorders, which includes Myhre syndrome, acromicric dysplasia, geleophysic dysplasias, Weill-Marchesani syndromes and stiff skin syndrome. Identification of a systemic sclerosis-protective SDC2 variant lays the foundation for exploration of the role of syndecan-2 in systemic sclerosis in the future.


Asunto(s)
Cromosomas Humanos Par 8/genética , Duplicación de Gen , Factor 6 de Diferenciación de Crecimiento/genética , Deformidades Congénitas de la Mano/genética , Artropatías/congénito , Osificación Heterotópica/genética , Esclerodermia Sistémica/genética , Sindecano-2/genética , Adulto , Anciano , Preescolar , Matriz Extracelular/metabolismo , Facies , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Factor 6 de Diferenciación de Crecimiento/metabolismo , Deformidades Congénitas de la Mano/metabolismo , Deformidades Congénitas de la Mano/fisiopatología , Humanos , Lactante , Artropatías/genética , Artropatías/metabolismo , Artropatías/fisiopatología , Masculino , Persona de Mediana Edad , Osificación Heterotópica/metabolismo , Osificación Heterotópica/fisiopatología , Fenotipo , Transducción de Señal , Sindecano-2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
11.
J Med Genet ; 51(10): 659-68, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25125236

RESUMEN

BACKGROUND: Cornelia de Lange syndrome (CdLS) is a multisystem disorder with distinctive facial appearance, intellectual disability and growth failure as prominent features. Most individuals with typical CdLS have de novo heterozygous loss-of-function mutations in NIPBL with mosaic individuals representing a significant proportion. Mutations in other cohesin components, SMC1A, SMC3, HDAC8 and RAD21 cause less typical CdLS. METHODS: We screened 163 affected individuals for coding region mutations in the known genes, 90 for genomic rearrangements, 19 for deep intronic variants in NIPBL and 5 had whole-exome sequencing. RESULTS: Pathogenic mutations [including mosaic changes] were identified in: NIPBL 46 [3] (28.2%); SMC1A 5 [1] (3.1%); SMC3 5 [1] (3.1%); HDAC8 6 [0] (3.6%) and RAD21 1 [0] (0.6%). One individual had a de novo 1.3 Mb deletion of 1p36.3. Another had a 520 kb duplication of 12q13.13 encompassing ESPL1, encoding separase, an enzyme that cleaves the cohesin ring. Three de novo mutations were identified in ANKRD11 demonstrating a phenotypic overlap with KBG syndrome. To estimate the number of undetected mosaic cases we used recursive partitioning to identify discriminating features in the NIPBL-positive subgroup. Filtering of the mutation-negative group on these features classified at least 18% as 'NIPBL-like'. A computer composition of the average face of this NIPBL-like subgroup was also more typical in appearance than that of all others in the mutation-negative group supporting the existence of undetected mosaic cases. CONCLUSIONS: Future diagnostic testing in 'mutation-negative' CdLS thus merits deeper sequencing of multiple DNA samples derived from different tissues.


Asunto(s)
Síndrome de Cornelia de Lange/genética , Heterogeneidad Genética , Mosaicismo , Cara/patología , Estudios de Asociación Genética , Humanos , Mutación , Fenotipo
12.
Health Expect ; 18(2): 177-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23094806

RESUMEN

BACKGROUND: Progressive ataxias are neurological disorders affecting balance, co-ordination of movement and speech. OBJECTIVE: A qualitative study was undertaken to discover patients' experiences of ataxia and its symptoms. PARTICIPANTS: Thirty-eight people with ataxia recruited from patient support groups and two hospital outpatients departments. DESIGN: Cross-sectional qualitative study with thematic analysis. RESULTS: These accounts highlight the limits of medicine in the context of a rare, incurable and disabling disorder, and the embodied uncertainties brought by slowly progressive diseases that lie at the boundaries of mainstream medical knowledge. The existential crises faced by people with ataxia are seemingly magnified by sometimes idiopathic aetiologies and the limited number of inherited conditions identifiable by the available genetic tests. Interviewees were drawn into a medical system that was focused mainly on the diagnosis process, with widely varying results. However, when asked, most had rather valued the provision of disability aids and physical therapies. Only one informant reported overcoming the myriad uncertainties of progressive ataxia, and their account supported the notion of 'biographical repair' in chronic illness. CONCLUSIONS: Clinical uncertainties in ataxia constrained people's attempts to deal with their condition. The construction of the proactive, informed, medical consumer who is assumed to be a partner in care is problematic in the context of a rare and difficult-to-diagnose disease for which there is usually no cure. Service providers should be mindful of the need to manage patient expectations in relation to diagnosis and cure. More focus might usefully be placed on the provision of physical therapies and disability aids.


Asunto(s)
Ataxia/psicología , Ataxia/terapia , Adulto , Anciano , Enfermedad Crónica , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Aceptación de la Atención de Salud , Modalidades de Fisioterapia , Dispositivos de Autoayuda , Índice de Severidad de la Enfermedad , Incertidumbre
13.
Am J Hum Genet ; 89(1): 148-53, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21737058

RESUMEN

3-M syndrome, a primordial growth disorder, is associated with mutations in CUL7 and OBSL1. Exome sequencing now identifies mutations in CCDC8 as a cause of 3-M syndrome. CCDC8 is a widely expressed gene that is transcriptionally associated to CUL7 and OBSL1, and coimmunoprecipitation indicates a physical interaction between CCDC8 and OBSL1 but not CUL7. We propose that CUL7, OBSL1, and CCDC8 are members of a pathway controlling mammalian growth.


Asunto(s)
Proteínas Cullin/genética , Proteínas del Citoesqueleto/genética , Enanismo/genética , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Línea Celular , Preescolar , Proteínas Cullin/metabolismo , Proteínas del Citoesqueleto/metabolismo , Femenino , Expresión Génica , Homocigoto , Humanos , Lactante , Masculino , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Columna Vertebral/anomalías , Factores de Transcripción
14.
Brain ; 136(Pt 12): 3634-44, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24176978

RESUMEN

Marinesco-Sjögren syndrome is a rare autosomal recessive multisystem disorder featuring cerebellar ataxia, early-onset cataracts, chronic myopathy, variable intellectual disability and delayed motor development. More recently, mutations in the SIL1 gene, which encodes an endoplasmic reticulum resident co-chaperone, were identified as the main cause of Marinesco-Sjögren syndrome. Here we describe the results of SIL1 mutation analysis in 62 patients presenting with early-onset ataxia, cataracts and myopathy or combinations of at least two of these. We obtained a mutation detection rate of 60% (15/25) among patients with the characteristic Marinesco-Sjögren syndrome triad (ataxia, cataracts, myopathy) whereas the detection rate in the group of patients with more variable phenotypic presentation was below 3% (1/37). We report 16 unrelated families with a total of 19 different SIL1 mutations. Among these mutations are 15 previously unreported changes, including single- and multi-exon deletions. Based on data from our screening cohort and data compiled from the literature we found that SIL1 mutations are invariably associated with the combination of a cerebellar syndrome and chronic myopathy. Cataracts were observed in all patients beyond the age of 7 years, but might be missing in infants. Six patients with SIL1 mutations had no intellectual disability, extending the known wide range of cognitive capabilities in Marinesco-Sjögren syndrome to include normal intelligence. Modestly constant features were somatic growth retardation, skeletal abnormalities and pyramidal tract signs. Examination of mutant SIL1 expression in cultured patient lymphoblasts suggested that SIL1 mutations result in severely reduced SIL1 protein levels irrespective of the type and position of mutations. Our data broaden the SIL1 mutation spectrum and confirm that SIL1 is the major Marinesco-Sjögren syndrome gene. SIL1 patients usually present with the characteristic triad but cataracts might be missing in young children. As cognitive impairment is not obligatory, patients without intellectual disability but a Marinesco-Sjögren syndrome-compatible phenotype should receive SIL1 mutation analysis. Despite allelic heterogeneity and many families with private mutations, the phenotype related to SIL1 mutations is relatively homogenous. Based on SIL1 expression studies we speculate that this may arise from a uniform effect of different mutations on protein expression.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Mutación/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Linfocitos B , Encéfalo/patología , Encéfalo/ultraestructura , Células Cultivadas , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Estudios Retrospectivos , Degeneraciones Espinocerebelosas/patología , Degeneraciones Espinocerebelosas/fisiopatología
15.
Eur J Med Genet ; 67: 104907, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38141875

RESUMEN

Genetic variants in ATP7A are associated with a spectrum of X-linked disorders. In descending order of severity, these are Menkes disease, occipital horn syndrome, and X-linked distal spinal muscular atrophy. After 30 years of diagnostic investigation, we identified a deep intronic ATP7A variant in four males from a family affected to variable degrees by a predominantly skeletal phenotype, featuring bowing of long bones, elbow joints with restricted mobility which dislocate frequently, coarse curly hair, chronic diarrhoea, and motor coordination difficulties. Analysis of whole genome sequencing data from the Genomics England 100,000 Genomes Project following clinical re-evaluation identified a deep intronic ATP7A variant, which was predicted by SpliceAI to have a modest splicing effect. Using a mini-gene splicing assay, we determined that the intronic variant results in aberrant splicing. Sanger sequencing of patient cDNA revealed ATP7A transcripts with exon 5 skipping, or inclusion of a novel intron 4 pseudoexon. In both instances, frameshift leading to premature termination are predicted. Quantification of ATP7A mRNA transcripts using a qPCR assay indicated that the majority of transcripts (86.1 %) have non-canonical splicing, with 68.0 % featuring exon 5 skipping, and 18.1 % featuring the novel pseudoexon. We suggest that the variability of the phenotypes within the affected males results from the stochastic effects of splicing. This deep intronic variant, resulting in aberrant ATP7A splicing, expands the understanding of intronic variation on the ATP7A-related disease spectrum.


Asunto(s)
Cutis Laxo , Síndrome de Ehlers-Danlos , Humanos , Masculino , ATPasas Transportadoras de Cobre/genética , Cutis Laxo/genética , Síndrome de Ehlers-Danlos/genética , Mutación , Fragmentos de Péptidos/genética , Fenotipo
16.
Hum Mutat ; 33(6): 981-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473935

RESUMEN

Ryanodine receptor 1 (RYR1) mutations are a common cause of congenital myopathies associated with both dominant and recessive inheritance. Histopathological findings frequently feature central cores or multi-minicores, more rarely, type 1 predominance/uniformity, fiber-type disproportion, increased internal nucleation, and fatty and connective tissue. We describe 71 families, 35 associated with dominant RYR1 mutations and 36 with recessive inheritance. Five of the dominant mutations and 35 of the 55 recessive mutations have not been previously reported. Dominant mutations, typically missense, were frequently located in recognized mutational hotspot regions, while recessive mutations were distributed throughout the entire coding sequence. Recessive mutations included nonsense and splice mutations expected to result in reduced RyR1 protein. There was wide clinical variability. As a group, dominant mutations were associated with milder phenotypes; patients with recessive inheritance had earlier onset, more weakness, and functional limitations. Extraocular and bulbar muscle involvement was almost exclusively observed in the recessive group. In conclusion, our study reports a large number of novel RYR1 mutations and indicates that recessive variants are at least as frequent as the dominant ones. Assigning pathogenicity to novel mutations is often difficult, and interpretation of genetic results in the context of clinical, histological, and muscle magnetic resonance imaging findings is essential.


Asunto(s)
Mutación , Miopatías Estructurales Congénitas/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Niño , Preescolar , Femenino , Genes Dominantes , Genes Recesivos , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Linaje
17.
Muscle Nerve ; 46(5): 698-704, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22996176

RESUMEN

INTRODUCTION: Although unmyelinated nerve fibers are affected in Charcot-Marie-Tooth type 1A (CMT1A) disease, they have not been studied in detail due to the invasive nature of the techniques needed to study them. We established alterations in C-fiber bundles of the cornea in patients with CMT1A using non-invasive corneal confocal microscopy (CCM). METHODS: Twelve patients with CMT1A and 12 healthy control subjects underwent assessment of neuropathic symptoms and deficits, electrophysiology, quantitative sensory testing, corneal sensitivity, and corneal confocal microscopy. RESULTS: Corneal sensitivity, corneal nerve fiber density, corneal nerve branch density, corneal nerve fiber length, and corneal nerve fiber tortuosity were significantly reduced in CMT1A patients compared with controls. There was a significant correlation between corneal sensation and CCM parameters with the severity of painful neuropathic symptoms, cold and warm thresholds, and median nerve CMAP amplitude. CONCLUSIONS: CCM demonstrates significant damage to C-fiber bundles, which relates to some measures of neuropathy in CMT1A patients.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/patología , Córnea/patología , Fibras Nerviosas Amielínicas/patología , Adulto , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Femenino , Humanos , Masculino , Microscopía Confocal/métodos , Persona de Mediana Edad , Dimensión del Dolor/métodos
18.
Am J Med Genet A ; 155A(12): 2910-5, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22002932

RESUMEN

We present a newly recognized, likely autosomal recessive, pleiotropic disorder seen in four individuals (three siblings and their nephew) from a consanguineous family of Pakistani origin. The condition is characterized by hypogonadotropic hypogonadism, severe microcephaly, sensorineural deafness, moderate learning disability, and distinctive facial dysmorphic features. Autozygosity mapping using SNP array genotyping defined a single, large autozygous region of 13.1 Mb on chromosome 3p21 common to the affected individuals. The critical region contains 227 genes and initial sequence analysis of a functional candidate gene has not identified causative mutations.


Asunto(s)
Cromosomas Humanos Par 3 , Anomalías Craneofaciales/genética , Genes Recesivos , Pérdida Auditiva Sensorineural/genética , Hipogonadismo/genética , Microcefalia/genética , Adulto , Encéfalo/patología , Niño , Mapeo Cromosómico , Consanguinidad , Anomalías Craneofaciales/diagnóstico , Femenino , Pérdida Auditiva Sensorineural/diagnóstico , Homocigoto , Humanos , Hipogonadismo/diagnóstico , Imagen por Resonancia Magnética , Masculino , Microcefalia/diagnóstico , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Síndrome , Adulto Joven
19.
J Clin Invest ; 130(3): 1431-1445, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31794431

RESUMEN

Epigenetic integrity is critical for many eukaryotic cellular processes. An important question is how different epigenetic regulators control development and influence disease. Lysine acetyltransferase 8 (KAT8) is critical for acetylation of histone H4 at lysine 16 (H4K16), an evolutionarily conserved epigenetic mark. It is unclear what roles KAT8 plays in cerebral development and human disease. Here, we report that cerebrum-specific knockout mice displayed cerebral hypoplasia in the neocortex and hippocampus, along with improper neural stem and progenitor cell (NSPC) development. Mutant cerebrocortical neuroepithelia exhibited faulty proliferation, aberrant neurogenesis, massive apoptosis, and scant H4K16 propionylation. Mutant NSPCs formed poor neurospheres, and pharmacological KAT8 inhibition abolished neurosphere formation. Moreover, we describe KAT8 variants in 9 patients with intellectual disability, seizures, autism, dysmorphisms, and other anomalies. The variants altered chromobarrel and catalytic domains of KAT8, thereby impairing nucleosomal H4K16 acetylation. Valproate was effective for treating epilepsy in at least 2 of the individuals. This study uncovers a critical role of KAT8 in cerebral and NSPC development, identifies 9 individuals with KAT8 variants, and links deficient H4K16 acylation directly to intellectual disability, epilepsy, and other developmental anomalies.


Asunto(s)
Hipocampo/enzimología , Histona Acetiltransferasas/metabolismo , Discapacidad Intelectual/enzimología , Neocórtex/enzimología , Células-Madre Neurales/enzimología , Acetilación , Animales , Células HEK293 , Hipocampo/patología , Histona Acetiltransferasas/genética , Humanos , Discapacidad Intelectual/patología , Ratones , Ratones Noqueados , Neocórtex/patología , Células-Madre Neurales/patología , Nucleosomas/genética , Nucleosomas/metabolismo
20.
Hum Mutat ; 30(3): 334-41, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19006240

RESUMEN

Beals-Hecht syndrome or congenital contractural arachnodactyly (CCA) is a rare, autosomal dominant connective tissue disorder characterized by crumpled ears, arachnodactyly, contractures, and scoliosis. Recent reports also mention aortic root dilatation, a finding previously thought to differentiate the condition from Marfan syndrome (MFS). In many cases, the condition is caused by mutations in the fibrillin 2 gene (FBN2) with 26 mutations reported so far, all located in the middle region of the gene (exons 23-34). We directly sequenced the entire FBN2 gene in 32 probands clinically diagnosed with CCA. In 14 probands, we found 13 new and one previously described FBN2 mutation including a mutation in exon 17, expanding the region in which FBN2 mutations occur in CCA. Review of the literature showed that the phenotype of the FBN2 positive patients was comparable to all previously published FBN2-positive patients. In our FBN2-positive patients, cardiovascular involvement included mitral valve prolapse in two adult patients and aortic root enlargement in three patients. Whereas the dilatation regressed in one proband, it remained marked in a child proband (z-score: 4.09) and his father (z-score: 2.94), warranting echocardiographic follow-up. We confirm paradoxical patellar laxity and report keratoconus, shoulder muscle hypoplasia, and pyeloureteral junction stenosis as new features. In addition, we illustrate large intrafamilial variability. Finally, the FBN2-negative patients in this cohort were clinically indistinguishable from all published FBN2-positive patients harboring a FBN2 mutation, suggesting locus heterogeneity.


Asunto(s)
Anomalías Múltiples/genética , Aracnodactilia/patología , Contractura/patología , Proteínas de Microfilamentos/genética , Mutación , Anomalías Múltiples/patología , Niño , Contractura/congénito , Análisis Mutacional de ADN , Femenino , Fibrilina-2 , Fibrilinas , Humanos , Masculino , Linaje , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA