Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 111(1): 119-132, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38141607

RESUMEN

Cyclin D2 (CCND2) stabilization underpins a range of macrocephaly-associated disorders through mutation of CCND2 or activating mutations in upstream genes encoding PI3K-AKT pathway components. Here, we describe three individuals with overlapping macrocephaly-associated phenotypes who carry the same recurrent de novo c.179G>A (p.Arg60Gln) variant in Myc-associated factor X (MAX). The mutation, located in the b-HLH-LZ domain, causes increased intracellular CCND2 through increased transcription but it does not cause stabilization of CCND2. We show that the purified b-HLH-LZ domain of MAXArg60Gln (Max∗Arg60Gln) binds its target E-box sequence with a lower apparent affinity. This leads to a more efficient heterodimerization with c-Myc resulting in an increase in transcriptional activity of c-Myc in individuals carrying this mutation. The recent development of Omomyc-CPP, a cell-penetrating b-HLH-LZ-domain c-Myc inhibitor, provides a possible therapeutic option for MAXArg60Gln individuals, and others carrying similar germline mutations resulting in dysregulated transcriptional c-Myc activity.


Asunto(s)
Megalencefalia , Proteínas Proto-Oncogénicas c-myc , Humanos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Dimerización , Megalencefalia/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
2.
Am J Hum Genet ; 111(3): 487-508, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38325380

RESUMEN

Pathogenic variants in multiple genes on the X chromosome have been implicated in syndromic and non-syndromic intellectual disability disorders. ZFX on Xp22.11 encodes a transcription factor that has been linked to diverse processes including oncogenesis and development, but germline variants have not been characterized in association with disease. Here, we present clinical and molecular characterization of 18 individuals with germline ZFX variants. Exome or genome sequencing revealed 11 variants in 18 subjects (14 males and 4 females) from 16 unrelated families. Four missense variants were identified in 11 subjects, with seven truncation variants in the remaining individuals. Clinical findings included developmental delay/intellectual disability, behavioral abnormalities, hypotonia, and congenital anomalies. Overlapping and recurrent facial features were identified in all subjects, including thickening and medial broadening of eyebrows, variations in the shape of the face, external eye abnormalities, smooth and/or long philtrum, and ear abnormalities. Hyperparathyroidism was found in four families with missense variants, and enrichment of different tumor types was observed. In molecular studies, DNA-binding domain variants elicited differential expression of a small set of target genes relative to wild-type ZFX in cultured cells, suggesting a gain or loss of transcriptional activity. Additionally, a zebrafish model of ZFX loss displayed an altered behavioral phenotype, providing additional evidence for the functional significance of ZFX. Our clinical and experimental data support that variants in ZFX are associated with an X-linked intellectual disability syndrome characterized by a recurrent facial gestalt, neurocognitive and behavioral abnormalities, and an increased risk for congenital anomalies and hyperparathyroidism.


Asunto(s)
Hiperparatiroidismo , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Masculino , Femenino , Animales , Humanos , Discapacidad Intelectual/patología , Pez Cebra/genética , Mutación Missense/genética , Factores de Transcripción/genética , Fenotipo , Trastornos del Neurodesarrollo/genética
3.
Clin Genet ; 101(2): 255-259, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34713892

RESUMEN

The developmental disorder Burn-McKeown Syndrome (BMKS) is characterised by choanal atresia and specific craniofacial features. BMKS is caused by biallelic variants in the pre-messenger RNA splicing factor TXNL4A. Most patients have a loss-of-function variant in trans with a 34-base pair (bp) deletion (type 1 Δ34) in the promoter region. Here, we identified two patients with BMKS. One individual has a TXNL4A c.93_94delCC, p.His32Argfs *21 variant combined with a type 1 Δ34 promoter deletion. The other has an intronic TXNL4A splice site variant (c.258-3C>G) and a type 1 Δ34 promoter deletion. We show the c.258-3C>G variant and a previously reported c.258-2A>G variant, cause skipping of the final exon of TXNL4A in a minigene splicing assay. Furthermore, we identify putative transcription factor binding sites within the 56 bp of the TXNL4A promoter affected by the type 1 and type 2 Δ34 and use dual luciferase assays to identify a 22 bp repeated motif essential for TXNL4A expression within this promoter region. We propose that additional variants affecting critical transcription factor binding nucleotides within the 22 bp repeated motif could be relevant to BMKS aetiology. Finally, our data emphasises the need to analyse the non-coding sequence in individuals where a single likely pathogenic coding variant is identified in an autosomal recessive disorder consistent with the clinical presentation.


Asunto(s)
Atresia de las Coanas/diagnóstico , Atresia de las Coanas/genética , Sordera/congénito , Genotipo , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Mutación , Ribonucleoproteína Nuclear Pequeña U5/genética , Alelos , Sitios de Unión , Sordera/diagnóstico , Sordera/genética , Facies , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Linaje , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U5/metabolismo , Factores de Transcripción/metabolismo
4.
J Med Genet ; 58(5): 334-341, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32571899

RESUMEN

BACKGROUND: The HERC2 gene encodes a 527 kDa E3 ubiquitin protein ligase that has key roles in cell cycle regulation, spindle formation during mitosis, mitochondrial functions and DNA damage responses. It has essential roles during embryonic development, particularly for neuronal and muscular functions. To date, missense mutations in HERC2 have been associated with an autosomal recessive neurodevelopmental disorder with some phenotypical similarities to Angelman syndrome, and a homozygous deletion spanning HERC2 and OCA2 causing a more severe neurodevelopmental phenotype. METHODS AND RESULTS: We ascertained a consanguineous family with a presumed autosomal recessive severe neurodevelopmental disorder that leads to paediatric lethality. In affected individuals, we identified a homozygous HERC2 frameshift variant that results in a premature stop codon and complete loss of HERC2 protein. Functional characterisation of this variant in fibroblasts, from one living affected individual, revealed impaired mitochondrial network and function as well as disrupted levels of known interacting proteins such as XPA. CONCLUSION: This study extends the genotype-phenotype correlation for HERC2 variants to include a distinct lethal neurodevelopmental disorder, highlighting the importance of further characterisation for HERC2-related disorders.


Asunto(s)
Genes Letales , Mutación con Pérdida de Función , Trastornos del Neurodesarrollo/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Adulto , Células Cultivadas , Niño , Mortalidad del Niño , Consanguinidad , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Linaje , Adulto Joven
5.
Genet Med ; 23(4): 653-660, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33299146

RESUMEN

PURPOSE: This study aims to provide a comprehensive description of the phenotypic and genotypic spectrum of SNAP25 developmental and epileptic encephalopathy (SNAP25-DEE) by reviewing newly identified and previously reported individuals. METHODS: Individuals harboring heterozygous missense or loss-of-function variants in SNAP25 were assembled through collaboration with international colleagues, matchmaking platforms, and literature review. For each individual, detailed phenotyping, classification, and structural modeling of the identified variant were performed. RESULTS: The cohort comprises 23 individuals with pathogenic or likely pathogenic de novo variants in SNAP25. Intellectual disability and early-onset epilepsy were identified as the core symptoms of SNAP25-DEE, with recurrent findings of movement disorders, cerebral visual impairment, and brain atrophy. Structural modeling for all variants predicted possible functional defects concerning SNAP25 or impaired interaction with other components of the SNARE complex. CONCLUSION: We provide a comprehensive description of SNAP25-DEE with intellectual disability and early-onset epilepsy mostly occurring before the age of two years. These core symptoms and additional recurrent phenotypes show an overlap to genes encoding other components or associated proteins of the SNARE complex such as STX1B, STXBP1, or VAMP2. Thus, these findings advance the concept of a group of neurodevelopmental disorders that may be termed "SNAREopathies."


Asunto(s)
Encefalopatías , Epilepsia , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Proteína 25 Asociada a Sinaptosomas/genética , Preescolar , Epilepsia/genética , Humanos , Trastornos del Neurodesarrollo/genética , Fenotipo
6.
Am J Med Genet A ; 185(8): 2445-2454, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34032352

RESUMEN

Smith-Kingsmore Syndrome (SKS) is a rare genetic syndrome associated with megalencephaly, a variable intellectual disability, autism spectrum disorder, and MTOR gain of function variants. Only 30 patients with MTOR missense variants are published, including 14 (47%) with the MTOR c.5395G>A p.(Glu1799Lys) variant. Limited phenotypic data impacts the quality of information delivered to families and the robustness of interpretation of novel MTOR missense variation. This study aims to improve our understanding of the SKS phenotype through the investigation of 16 further patients with the MTOR c.5395G>A p.(Glu1799Lys) variant. Through the careful phenotypic evaluation of these 16 patients and integration with data from 14 previously reported patients, we have defined major (100% patients) and frequent (>15%) SKS clinical characteristics and, using these data, proposed guidance for evidence-based management. In addition, in the absence of functional studies, we suggest that the combination of the SKS major clinical features of megalencephaly (where the head circumference is at least 3SD) and an intellectual disability with a de novo MTOR missense variant (absent from population databases) should be considered diagnostic for SKS.


Asunto(s)
Alelos , Estudios de Asociación Genética , Mutación Missense , Fenotipo , Serina-Treonina Quinasas TOR/genética , Adolescente , Sustitución de Aminoácidos , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Niño , Preescolar , Facies , Femenino , Sitios Genéticos , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Masculino , Megalencefalia/diagnóstico , Megalencefalia/genética , Síndrome
7.
Genome Res ; 27(10): 1704-1714, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28855261

RESUMEN

Structural mosaic abnormalities are large post-zygotic mutations present in a subset of cells and have been implicated in developmental disorders and cancer. Such mutations have been conventionally assessed in clinical diagnostics using cytogenetic or microarray testing. Modern disease studies rely heavily on exome sequencing, yet an adequate method for the detection of structural mosaicism using targeted sequencing data is lacking. Here, we present a method, called MrMosaic, to detect structural mosaic abnormalities using deviations in allele fraction and read coverage from next-generation sequencing data. Whole-exome sequencing (WES) and whole-genome sequencing (WGS) simulations were used to calculate detection performance across a range of mosaic event sizes, types, clonalities, and sequencing depths. The tool was applied to 4911 patients with undiagnosed developmental disorders, and 11 events among nine patients were detected. For eight of these 11 events, mosaicism was observed in saliva but not blood, suggesting that assaying blood alone would miss a large fraction, possibly >50%, of mosaic diagnostic chromosomal rearrangements.


Asunto(s)
Exoma , Genoma Humano , Mosaicismo , Análisis de Secuencia de ADN/métodos , Femenino , Humanos , Masculino , Análisis de Secuencia de ADN/instrumentación
8.
Am J Hum Genet ; 98(2): 373-81, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26833328

RESUMEN

Mutations in more than a hundred genes have been reported to cause X-linked recessive intellectual disability (ID) mainly in males. In contrast, the number of identified X-linked genes in which de novo mutations specifically cause ID in females is limited. Here, we report 17 females with de novo loss-of-function mutations in USP9X, encoding a highly conserved deubiquitinating enzyme. The females in our study have a specific phenotype that includes ID/developmental delay (DD), characteristic facial features, short stature, and distinct congenital malformations comprising choanal atresia, anal abnormalities, post-axial polydactyly, heart defects, hypomastia, cleft palate/bifid uvula, progressive scoliosis, and structural brain abnormalities. Four females from our cohort were identified by targeted genetic testing because their phenotype was suggestive for USP9X mutations. In several females, pigment changes along Blaschko lines and body asymmetry were observed, which is probably related to differential (escape from) X-inactivation between tissues. Expression studies on both mRNA and protein level in affected-female-derived fibroblasts showed significant reduction of USP9X level, confirming the loss-of-function effect of the identified mutations. Given that some features of affected females are also reported in known ciliopathy syndromes, we examined the role of USP9X in the primary cilium and found that endogenous USP9X localizes along the length of the ciliary axoneme, indicating that its loss of function could indeed disrupt cilium-regulated processes. Absence of dysregulated ciliary parameters in affected female-derived fibroblasts, however, points toward spatiotemporal specificity of ciliary USP9X (dys-)function.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Mutación , Ubiquitina Tiolesterasa/genética , Adolescente , Secuencia de Bases , Niño , Preescolar , Atresia de las Coanas/diagnóstico , Atresia de las Coanas/genética , Discapacidades del Desarrollo/diagnóstico , Femenino , Genes Ligados a X , Pruebas Genéticas , Humanos , Discapacidad Intelectual/diagnóstico , Datos de Secuencia Molecular , Fenotipo , Ubiquitina Tiolesterasa/metabolismo , Inactivación del Cromosoma X , Adulto Joven
9.
Am J Med Genet A ; 179(3): 344-349, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30637921

RESUMEN

Primrose syndrome is a rare autosomal dominant condition caused by heterozygous missense variants within ZBTB20. Through an exome sequencing approach (as part of the Deciphering Developmental Disorders [DDD] study) we have identified five unrelated individuals with previously unreported, de novo ZBTB20 pathogenic missense variants. All five missense variants targeted the C2H2 zinc finger domains. This genotype-up approach has allowed further refinement of the Primrose syndrome phenotype. Major characteristics (>90% individuals) include an intellectual disability (most frequently in the moderate range), a recognizable facial appearance and brain MRI abnormalities, particularly abnormalities of the corpus callosum. Other frequent clinical associations (in 50-90% individuals) include sensorineural hearing loss (83%), hypotonia (78%), cryptorchidism in males (75%), macrocephaly (72%), behavioral issues (56%), and dysplastic/hypoplastic nails (57%). Based upon these clinical data we discuss our current management of patients with Primrose syndrome.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Calcinosis/diagnóstico , Calcinosis/genética , Enfermedades del Oído/diagnóstico , Enfermedades del Oído/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Atrofia Muscular/diagnóstico , Atrofia Muscular/genética , Proteínas del Tejido Nervioso/genética , Fenotipo , Factores de Transcripción/genética , Niño , Preescolar , Facies , Femenino , Sitios Genéticos , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Mutación
10.
J Med Genet ; 55(1): 28-38, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021403

RESUMEN

INTRODUCTION: Recent evidence has emerged linking mutations in CDK13 to syndromic congenital heart disease. We present here genetic and phenotypic data pertaining to 16 individuals with CDK13 mutations. METHODS: Patients were investigated by exome sequencing, having presented with developmental delay and additional features suggestive of a syndromic cause. RESULTS: Our cohort comprised 16 individuals aged 4-16 years. All had developmental delay, including six with autism spectrum disorder. Common findings included feeding difficulties (15/16), structural cardiac anomalies (9/16), seizures (4/16) and abnormalities of the corpus callosum (4/11 patients who had undergone MRI). All had craniofacial dysmorphism, with common features including short, upslanting palpebral fissures, hypertelorism or telecanthus, medial epicanthic folds, low-set, posteriorly rotated ears and a small mouth with thin upper lip vermilion. Fifteen patients had predicted missense mutations, including five identical p.(Asn842Ser) substitutions and two p.(Gly717Arg) substitutions. One patient had a canonical splice acceptor site variant (c.2898-1G>A). All mutations were located within the protein kinase domain of CDK13. The affected amino acids are highly conserved, and in silico analyses including comparative protein modelling predict that they will interfere with protein function. The location of the missense mutations in a key catalytic domain suggests that they are likely to cause loss of catalytic activity but retention of cyclin K binding, resulting in a dominant negative mode of action. Although the splice-site mutation was predicted to produce a stable internally deleted protein, this was not supported by expression studies in lymphoblastoid cells. A loss of function contribution to the underlying pathological mechanism therefore cannot be excluded, and the clinical significance of this variant remains uncertain. CONCLUSIONS: These patients demonstrate that heterozygous, likely dominant negative mutations affecting the protein kinase domain of the CDK13 gene result in a recognisable, syndromic form of intellectual disability, with or without congenital heart disease.


Asunto(s)
Proteína Quinasa CDC2/química , Proteína Quinasa CDC2/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Mutación/genética , Adolescente , Niño , Secuencia Conservada , Femenino , Heterocigoto , Humanos , Masculino , Modelos Moleculares , Mutación Missense/genética , Dominios Proteicos , Síndrome , Termodinámica
12.
Am J Med Genet A ; 170(12): 3150-3156, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27576954

RESUMEN

Osteogenesis Imperfecta (OI) is an inherited bone fragility disorder most commonly associated with autosomal dominant mutations in the type I collagen genes. Autosomal recessive mutations in a number of genes have also been described, including the BMP1 gene that encodes the mammalian Tolloid (mTLD) and its shorter isoform bone morphogenic protein-1 (BMP1). To date, less than 20 individuals with OI have been identified with BMP1 mutations, with skeletal phenotypes ranging from mild to severe and progressively deforming. In the majority of patients, bone fragility was associated with increased bone mineral density (BMD); however, the full range of phenotypes associated with BMP1 remains unclear. Here, we describe three children with mutations in BMP1 associated with a highly variable phenotype: a sibship homozygous for the c.2188delC mutation that affects only the shorter BMP1 isoform and a further patient who is compound heterozygous for a c.1293C>G nonsense mutation and a c.1148G>A missense mutation in the CUB1 domain. These individuals had recurrent fractures from early childhood, are hypermobile and have no evidence of dentinogenesis imperfecta. The homozygous siblings with OI had normal areal BMD by dual energy X-ray absorptiometry whereas the third patient presented with a high bone mass phenotype. Intravenous bisphosphonate therapy was started in all patients, but discontinued in two patients and reduced in another due to concerns about increasing bone stiffness leading to chalk-stick fractures. Given the association of BMP1-related OI with very high bone material density, concerns remain whether anti-resorptive therapy is indicated in this ultra-rare form of OI.© 2016 Wiley Periodicals, Inc.


Asunto(s)
Densidad Ósea/genética , Proteína Morfogenética Ósea 1/genética , Colágeno Tipo I/genética , Osteogénesis Imperfecta/genética , Adolescente , Huesos/fisiopatología , Niño , Difosfonatos/administración & dosificación , Femenino , Homocigoto , Humanos , Masculino , Mutación , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/fisiopatología , Fenotipo
13.
PLoS Genet ; 8(11): e1002945, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144622

RESUMEN

A homozygous mutational change in the Ataxia-Telangiectasia and RAD3 related (ATR) gene was previously reported in two related families displaying Seckel Syndrome (SS). Here, we provide the first identification of a Seckel Syndrome patient with mutations in ATRIP, the gene encoding ATR-Interacting Protein (ATRIP), the partner protein of ATR required for ATR stability and recruitment to the site of DNA damage. The patient has compound heterozygous mutations in ATRIP resulting in reduced ATRIP and ATR expression. A nonsense mutational change in one ATRIP allele results in a C-terminal truncated protein, which impairs ATR-ATRIP interaction; the other allele is abnormally spliced. We additionally describe two further unrelated patients native to the UK with the same novel, heterozygous mutations in ATR, which cause dramatically reduced ATR expression. All patient-derived cells showed defective DNA damage responses that can be attributed to impaired ATR-ATRIP function. Seckel Syndrome is characterised by microcephaly and growth delay, features also displayed by several related disorders including Majewski (microcephalic) osteodysplastic primordial dwarfism (MOPD) type II and Meier-Gorlin Syndrome (MGS). The identification of an ATRIP-deficient patient provides a novel genetic defect for Seckel Syndrome. Coupled with the identification of further ATR-deficient patients, our findings allow a spectrum of clinical features that can be ascribed to the ATR-ATRIP deficient sub-class of Seckel Syndrome. ATR-ATRIP patients are characterised by extremely severe microcephaly and growth delay, microtia (small ears), micrognathia (small and receding chin), and dental crowding. While aberrant bone development was mild in the original ATR-SS patient, some of the patients described here display skeletal abnormalities including, in one patient, small patellae, a feature characteristically observed in Meier-Gorlin Syndrome. Collectively, our analysis exposes an overlapping clinical manifestation between the disorders but allows an expanded spectrum of clinical features for ATR-ATRIP Seckel Syndrome to be defined.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Enanismo/genética , Trastornos del Crecimiento , Micrognatismo , Proteínas Serina-Treonina Quinasas , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Codón sin Sentido , Microtia Congénita , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Enanismo/patología , Oído/anomalías , Oído/patología , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/patología , Regulación de la Expresión Génica , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/patología , Heterocigoto , Humanos , Masculino , Microcefalia/genética , Microcefalia/patología , Micrognatismo/genética , Micrognatismo/patología , Osteocondrodisplasias/genética , Osteocondrodisplasias/patología , Rótula/anomalías , Rótula/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Empalme del ARN , Transducción de Señal/genética
14.
J Biol Chem ; 288(47): 33745-33759, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24108130

RESUMEN

Hyperekplexia is a syndrome of readily provoked startle responses, alongside episodic and generalized hypertonia, that presents within the first month of life. Inhibitory glycine receptors are pentameric ligand-gated ion channels with a definitive and clinically well stratified linkage to hyperekplexia. Most hyperekplexia cases are caused by mutations in the α1 subunit of the human glycine receptor (hGlyR) gene (GLRA1). Here we analyzed 68 new unrelated hyperekplexia probands for GLRA1 mutations and identified 19 mutations, of which 9 were novel. Electrophysiological analysis demonstrated that the dominant mutations p.Q226E, p.V280M, and p.R414H induced spontaneous channel activity, indicating that this is a recurring mechanism in hGlyR pathophysiology. p.Q226E, at the top of TM1, most likely induced tonic activation via an enhanced electrostatic attraction to p.R271 at the top of TM2, suggesting a structural mechanism for channel activation. Receptors incorporating p.P230S (which is heterozygous with p.R65W) desensitized much faster than wild type receptors and represent a new TM1 site capable of modulating desensitization. The recessive mutations p.R72C, p.R218W, p.L291P, p.D388A, and p.E375X precluded cell surface expression unless co-expressed with α1 wild type subunits. The recessive p.E375X mutation resulted in subunit truncation upstream of the TM4 domain. Surprisingly, on the basis of three independent assays, we were able to infer that p.E375X truncated subunits are incorporated into functional hGlyRs together with unmutated α1 or α1 plus ß subunits. These aberrant receptors exhibit significantly reduced glycine sensitivity. To our knowledge, this is the first suggestion that subunits lacking TM4 domains might be incorporated into functional pentameric ligand-gated ion channel receptors.


Asunto(s)
Regulación de la Expresión Génica , Rigidez Muscular/metabolismo , Mutación Missense , Receptores de Glicina/metabolismo , Sustitución de Aminoácidos , Femenino , Humanos , Masculino , Rigidez Muscular/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Glicina/genética
15.
Am J Med Genet A ; 164A(4): 907-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24459067

RESUMEN

We report on a family in which four males over three generations are affected with X-linked recessive developmental delay, learning difficulties, severe behavioral difficulties and mild dysmorphic features. Plasma sterol analysis in three of the four affected males demonstrated increased concentrations of 8-dehydrocholesterol (8-DHC) and cholest-8(9)-enol. All four affected males had a novel hemizygous missense mutation, p.W47R (c.139T>C), in EBP. Functional studies showed raised levels of cholest-8(9)-enol in patient's cultured fibroblast cells, which were suppressed when the cells were incubated with simvastatin. EBP encodes 3ß-hydroxysteroid-delta8, delta7-isomerase, a key enzyme involved in the cholesterol biosynthesis pathway. Mutations in EBP have previously been associated with Conradi-Hunermann-Happle syndrome (CHH), an X-linked dominant disorder characterized by skeletal dysplasia, skin, and ocular abnormalities, which is usually lethal in males. Four previous reports describe X-linked recessive multiple anomaly syndromes associated with non-mosaic EBP mutations in males, two at the same amino acid position, p.W47C. This phenotype has previously been described as "MEND" syndrome (male EBP disorder with neurological defects). The family reported herein represent either a novel phenotype, or an expansion of the MEND phenotype, characterized by extreme behavioral difficulties and a scarcity of structural anomalies. Simvastatin therapy is being evaluated in two males from this family.


Asunto(s)
Discapacidades del Desarrollo/genética , Genes Ligados a X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Trastornos Mentales/genética , Mutación , Esteroide Isomerasas/genética , Adulto , Niño , Colestadienoles/sangre , Discapacidades del Desarrollo/sangre , Enfermedades Genéticas Ligadas al Cromosoma X/sangre , Humanos , Lactante , Masculino , Trastornos Mentales/sangre , Linaje , Fenotipo , Adulto Joven
16.
Eur J Hum Genet ; 30(1): 95-100, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34645992

RESUMEN

White-Sutton syndrome (WHSUS) is a neurodevelopmental disorder caused by heterozygous loss-of-function variants in POGZ. Through the Deciphering Developmental Disorders study and clinical testing, we identified 12 individuals from 10 families with pathogenic or likely pathogenic variants in POGZ (eight de novo and two inherited). Most individuals had delayed development and/or intellectual disability. We analyzed the clinical findings in our series and combined it with data from 89 previously reported individuals. The results demonstrate WHSUS is associated with variable developmental delay or intellectual disability, increased risk of obesity, visual defects, craniofacial dysmorphism, sensorineural hearing loss, feeding problems, seizures, and structural brain malformations. Our series includes further individuals with rod-cone dystrophy, cleft lip and palate, congenital diaphragmatic hernia, and duplicated renal drainage system, suggesting these are rare complications of WHSUS. In addition, we describe an individual with a novel, de novo missense variant in POGZ and features of WHSUS. Our work further delineates the phenotypic spectrum of WHSUS highlighting the variable severity of this disorder and the observation of familial pathogenic POGZ variants.


Asunto(s)
Anomalías Múltiples/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Fenotipo , Transposasas/genética , Anomalías Múltiples/patología , Adolescente , Adulto , Niño , Preescolar , Discapacidades del Desarrollo/diagnóstico , Femenino , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Masculino , Mutación Missense , Linaje , Síndrome
17.
Neuromuscul Disord ; 31(4): 359-366, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33558124

RESUMEN

Pathogenic variants in TPM2 have been associated with a variable clinical spectrum, including congenital myopathies and distal arthrogryposis, all but one with dominant inheritance. We report the second case of recessively inherited TPM2-related Escobar variant of multiple pterygium syndrome and congenital myopathy in a patient from a consanguineous family. Ultra-structural examination of the biopsy revealed few cores/mini-cores and sparse nemaline rods. We found a novel homozygous intronic sequence variant, c.564-2A>C in TPM2. This variant is predicted to abolish the consensus acceptor splice site for exon 6b of TPM2 gene. Parents of the proband, both healthy adults with no clinical features, were heterozygous for the variant. Here we establish a homozygous intronic variant in TPM2 as the likely cause of Escobar variant of multiple pterygium syndrome and congenital myopathy, with sparse nemaline rods.


Asunto(s)
Anomalías Múltiples/genética , Hipertermia Maligna/genética , Miotonía Congénita/genética , Anomalías Cutáneas/genética , Tropomiosina/genética , Artrogriposis/genética , Preescolar , Consanguinidad , Heterocigoto , Homocigoto , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Fenotipo , Sitios de Empalme de ARN
18.
Hum Mutat ; 31(3): 279-83, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20020533

RESUMEN

We performed molecular, enzyme, and metabolic studies in 50 patients with D-2-hydroxyglutaric aciduria (D-2-HGA) who accumulated D-2-hydroxyglutarate (D-2-HG) in physiological fluids. Presumed pathogenic mutations were detected in 24 of 50 patients in the D-2-hydroxyglutarate dehydrogenase (D2HGDH) gene, which encodes D-2-hydroxyglutarate dehydrogenase (D-2-HGDH). Enzyme assay of D-2-HGDH confirmed that all patients with mutations had impaired enzyme activity, whereas patients with D-2-HGA whose enzyme activity was normal did not have mutations. Significantly lower D-2-HG concentrations in body fluids were observed in mutation-positive D-2-HGA patients than in mutation-negative patients. These results imply that multiple genetic loci may be associated with hyperexcretion of D-2-HG. Accordingly, we suggest a new classification: D-2-HGA Type I associates with D-2-HGDH deficiency, whereas idiopathic D-2-HGA manifests with normal D-2-HGDH activity and higher D-2-HG levels in body fluids compared with Type I patients. It remains possible that several classifications for idiopathic D-2-HGA patients with diverse genetic loci will be revealed in future studies.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Glutaratos/sangre , Glutaratos/orina , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Algoritmos , Líquidos Corporales , Análisis Mutacional de ADN , Genotipo , Glutaratos/líquido cefalorraquídeo , Homocigoto , Humanos , Modelos Genéticos , Mutación , Reproducibilidad de los Resultados , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/genética
19.
Mol Genet Genomic Med ; 7(12): e912, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31568717

RESUMEN

BACKGROUND: With increased access to genetic testing, variants of uncertain significance (VUS) where pathogenicity is uncertain are being increasingly identified. More than 85% Osteogenesis Imperfecta (OI) patients have pathogenic variants in COL1A1/A2. However, when a VUS is identified, there are no pathways in place for determining significance. OBJECTIVE: Define a diagnostic pathway to confirm pathogenicity, providing patients with definitive genetic diagnosis, accurate recurrence risks, and prenatal testing options. METHODS: Functional studies on collagen secretion from cultured patient fibroblasts combined with detailed phenotyping and segregation family studies. RESULTS: We demonstrate data from a family with a VUS identified in type I collagen. FAMILY-1: Six-year-old boy with failure-to-gain weight, talipes, fractures, on and off treatment with Pamidronate as diagnosis of OI uncertain. Transiliac bone biopsy at 2 years of age demonstrated active new bone formation within periosteum; bone cortices were normal thickness but increased porosity. Trabecular bone showed features of advanced osteoporosis. Genetic testing identified a de novo COL1A1 c.206_208delTGT, p.Leu69del variant. Sibling with similar phenotype but no fractures as yet, tested positive for variant raising concerns regarding her diagnosis, and management. Results from three independent experiments (cell immunofluorescence, collagen secretion assay by Western Blot, and unbiased proteomics) from cultured patient fibroblasts demonstrate COL1A1 c.206_208delTGT, p.Leu69del variant causing a substantial defect to collagen extracellular matrix assembly confirming variant pathogenicity. CONCLUSION: Access to genetic testing in OI is increasing as advances in genetic technologies decreases cost; a clinical diagnostic pathway needs to be implemented for managing variants identified by such testing.


Asunto(s)
Colágeno Tipo I/genética , Pruebas Genéticas/métodos , Osteogénesis Imperfecta/genética , Adulto , Niño , Preescolar , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Familia , Femenino , Fibroblastos , Fracturas Óseas/genética , Pruebas Genéticas/ética , Variación Genética , Humanos , Hallazgos Incidentales , Lactante , Masculino , Mutación , Osteogénesis Imperfecta/patología , Fenotipo , Cultivo Primario de Células , Virulencia
20.
Eur J Med Genet ; 62(4): 243-247, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30048822

RESUMEN

GPC3 and GPC4 are the only two genes in which mutations are known to cause Simpson-Golabi-Behmel syndrome type 1 (SGBS1). The majority of SGBS1 patients have point mutations or deletions in GPC3. Only one SGBS1 family has been reported with duplication of both GPC3 and GPC4. Although clinical presentation of SGBS1 in affected males is well defined, the phenotype in female carriers is less clear. In total, six female carriers with clinical expression of SGBS1 have been reported to date. In this study, we provide description of two families with rare duplications in both GPC3 and GPC4. These imbalances resulted in SGBS1 in males, while female carriers with skewed X-inactivation exhibited significant features of SGBS1 including congenital heart defect, hernias, intellectual disability and coarse facial features. In family 2, a SGBS diagnosis was not considered in the father until after the diagnosis had been first considered and made in the affected daughter. We emphasize on the importance of testing at risk females and careful examination of those who are found to be carriers of SGBS1. We also discuss and provide supportive evidence for the role of skewed X-inactivation in clinical expression of SGBS1 in female carriers.


Asunto(s)
Arritmias Cardíacas/genética , Duplicación de Gen , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Gigantismo/genética , Glipicanos/genética , Cardiopatías Congénitas/genética , Heterocigoto , Discapacidad Intelectual/genética , Inactivación del Cromosoma X , Adulto , Arritmias Cardíacas/patología , Niño , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Gigantismo/patología , Cardiopatías Congénitas/patología , Humanos , Discapacidad Intelectual/patología , Masculino , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA