Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 61(6): 590-594, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38228391

RESUMEN

Background Beckwith-Wiedemann syndrome (BWS) is an imprinting disorder caused by (epi)genetic alterations at 11p15. Because approximately 20% of patients test negative via molecular testing of peripheral blood leukocytes, the concept of Beckwith-Wiedemann spectrum (BWSp) was established to encompass a broader cohort with diverse and overlapping phenotypes. The prevalence of other overgrowth syndromes concealed within molecularly negative BWSp remains unexplored. Methods We conducted whole-exome sequencing (WES) on 69 singleton patients exhibiting molecularly negative BWSp. Variants were confirmed by Sanger sequencing or quantitative genomic PCR. We compared BWSp scores and clinical features between groups with classical BWS (cBWS), atypical BWS or isolated lateralised overgrowth (aBWS+ILO) and overgrowth syndromes identified via WES. Results Ten patients, one classified as aBWS and nine as cBWS, showed causative gene variants for Simpson-Golabi-Behmel syndrome (five patients), Sotos syndrome (two), Imagawa-Matsumoto syndrome (one), glycosylphosphatidylinositol biosynthesis defect 11 (one) or 8q duplication/9p deletion (one). BWSp scores did not distinguish between cBWS and other overgrowth syndromes. Birth weight and height in other overgrowth syndromes were significantly larger than in aBWS+ILO and cBWS, with varying intergroup frequencies of clinical features. Conclusion Molecularly negative BWSp encapsulates other syndromes, and considering both WES and clinical features may facilitate accurate diagnosis.


Asunto(s)
Síndrome de Beckwith-Wiedemann , Secuenciación del Exoma , Humanos , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/patología , Síndrome de Beckwith-Wiedemann/diagnóstico , Masculino , Femenino , Lactante , Preescolar , Niño , Fenotipo , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/patología , Variación Genética , Mutación/genética
2.
Am J Med Genet A ; : e63656, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760879

RESUMEN

KIF1A-related disorders (KRDs) encompass recessive and dominant variants with wide clinical variability. Recent genetic investigations have expanded the clinical phenotypes of heterozygous KIF1A variants. However, there have been a few long-term observational studies of patients with heterozygous KIF1A variants. A retrospective chart review of consecutive patients diagnosed with spastic paraplegia at Miyagi Children's Hospital from 2016 to 2020 identified six patients with heterozygous KIF1A variants. To understand the long-term changes in clinical symptoms, we examined these patients in terms of their characteristics, clinical symptoms, results of electrophysiological and neuroimaging studies, and genetic testing. The median follow-up period was 30 years (4-44 years). This long-term observational study showed that early developmental delay and equinus gait, or unsteady gait, are the first signs of disease onset, appearing with the commencement of independent walking. In addition, later age-related progression was observed in spastic paraplegia, and the appearance of axonal neuropathy and reduced visual acuity were characteristic features of the late disease phenotype. Brain imaging showed age-related progression of cerebellar atrophy and the appearance of hyperintensity of optic radiation on T2WI and FLAIR imaging. Long-term follow-up revealed a pattern of steady progression and a variety of clinical symptoms, including spastic paraplegia, peripheral neuropathy, reduced visual acuity, and some degree of cerebellar ataxia. Clinical variability between patients was observed to some extent, and therefore, further studies are required to determine the phenotype-genotype correlation.

3.
Am J Hum Genet ; 106(4): 549-558, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-32169168

RESUMEN

De novo variants (DNVs) cause many genetic diseases. When DNVs are examined in the whole coding regions of genes in next-generation sequencing analyses, pathogenic DNVs often cluster in a specific region. One such region is the last exon and the last 50 bp of the penultimate exon, where truncating DNVs cause escape from nonsense-mediated mRNA decay [NMD(-) region]. Such variants can have dominant-negative or gain-of-function effects. Here, we first developed a resource of rates of truncating DNVs in NMD(-) regions under the null model of DNVs. Utilizing this resource, we performed enrichment analysis of truncating DNVs in NMD(-) regions in 346 developmental and epileptic encephalopathy (DEE) trios. We observed statistically significant enrichment of truncating DNVs in semaphorin 6B (SEMA6B) (p value: 2.8 × 10-8; exome-wide threshold: 2.5 × 10-6). The initial analysis of the 346 individuals and additional screening of 1,406 and 4,293 independent individuals affected by DEE and developmental disorders collectively identified four truncating DNVs in the SEMA6B NMD(-) region in five individuals who came from unrelated families (p value: 1.9 × 10-13) and consistently showed progressive myoclonic epilepsy. RNA analysis of lymphoblastoid cells established from an affected individual showed that the mutant allele escaped NMD, indicating stable production of the truncated protein. Importantly, heterozygous truncating variants in the NMD(+) region of SEMA6B are observed in general populations, and SEMA6B is most likely loss-of-function tolerant. Zebrafish expressing truncating variants in the NMD(-) region of SEMA6B orthologs displayed defective development of brain neurons and enhanced pentylenetetrazole-induced seizure behavior. In summary, we show that truncating DNVs in the final exon of SEMA6B cause progressive myoclonic epilepsy.


Asunto(s)
Exoma/genética , Exones/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Epilepsias Mioclónicas Progresivas/genética , Semaforinas/genética , Adolescente , Adulto , Alelos , Animales , Femenino , Heterocigoto , Humanos , Masculino , Degradación de ARNm Mediada por Codón sin Sentido/genética , Convulsiones/genética , Adulto Joven , Pez Cebra/genética
4.
Am J Hum Genet ; 106(5): 596-610, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32243864

RESUMEN

Weaver syndrome (WS), an overgrowth/intellectual disability syndrome (OGID), is caused by pathogenic variants in the histone methyltransferase EZH2, which encodes a core component of the Polycomb repressive complex-2 (PRC2). Using genome-wide DNA methylation (DNAm) data for 187 individuals with OGID and 969 control subjects, we show that pathogenic variants in EZH2 generate a highly specific and sensitive DNAm signature reflecting the phenotype of WS. This signature can be used to distinguish loss-of-function from gain-of-function missense variants and to detect somatic mosaicism. We also show that the signature can accurately classify sequence variants in EED and SUZ12, which encode two other core components of PRC2, and predict the presence of pathogenic variants in undiagnosed individuals with OGID. The discovery of a functionally relevant signature with utility for diagnostic classification of sequence variants in EZH2, EED, and SUZ12 supports the emerging paradigm shift for implementation of DNAm signatures into diagnostics and translational research.


Asunto(s)
Anomalías Múltiples/genética , Hipotiroidismo Congénito/genética , Anomalías Craneofaciales/genética , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2/genética , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Mutación , Complejo Represivo Polycomb 2/genética , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Lactante , Masculino , Mosaicismo , Mutación Missense/genética , Proteínas de Neoplasias , Reproducibilidad de los Resultados , Factores de Transcripción , Adulto Joven
5.
J Hum Genet ; 68(9): 643-648, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37268768

RESUMEN

The RNA-binding motif protein 10, RBM10, is an RNA splicing regulator essential for development. Loss-of-function RBM10 variants are associated with TARP syndrome, a severe X-linked recessive condition in males. We report a 3-year-old male with a mild phenotype, consisting of cleft palate, hypotonia, developmental delay, and minor dysmorphisms, associated with a missense RBM10 variant, c.943T>C, p.Ser315Pro, affecting the RRM2 RNA-binding domain. His clinical features were similar to a previously reported case associated with a missense variant. The p.Ser315Pro mutant protein was expressed normally in the nucleus, but its expression level and protein stability were slightly reduced. Nuclear magnetic resonance spectroscopy showed that the structure and the RNA-binding ability of the RRM2 domain with the p.Ser315Pro were unaffected. However, it affects the alternative splicing regulations of downstream genes, NUMB and TNRC6A, and its splicing alteration patterns were variable depending on target transcripts. In summary, a novel germline missense RBM10 p.Ser315Pro variant that causes functional changes in the expression of its downstream genes results in a non-lethal phenotype associated with developmental delays. The functional alteration effects depend on the residues affected by missense variants. Our findings are expected to bring broader insights into the RBM10-associated genotype-phenotype relationships by delineating the molecular mechanism of RBM10 functions.


Asunto(s)
Empalme Alternativo , Trastornos del Neurodesarrollo , Masculino , Humanos , Empalme del ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Mutación Missense , Trastornos del Neurodesarrollo/genética
6.
Clin Genet ; 103(4): 383-391, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36645289

RESUMEN

The SUZ12 gene encodes a subunit of polycomb repressive complex 2 (PRC2) that is essential for development by silencing the expression of multiple genes. Germline heterozygous variants in SUZ12 have been found in Imagawa-Matsumoto syndrome (IMMAS) characterized by overgrowth and multiple dysmorphic features. Similarly, both EZH2 and EED also encode a subunit of PRC2 each and their pathogenic variants cause Weaver syndrome and Cohen-Gibson syndrome, respectively. Clinical manifestations of these syndromes significantly overlap, although their different prevalence rates have recently been noted: generalized overgrowth, intellectual disability, scoliosis, and excessive loose skin appear to be less prevalent in IMMAS than in the other two syndromes. We could not determine any apparent genotype-phenotype correlation in IMMAS. The phenotype of neurofibromatosis type 1 arising from NF1 deletion was also shown to be modified by the deletion of SUZ12, 560 kb away. This review deepens our understanding of the clinical and genetic characteristics of IMMAS together with other overgrowth syndromes related to PRC2. We also report on a novel IMMAS patient carrying a splicing variant (c.1023+1G>C) in SUZ12. This patient had a milder phenotype than other previously reported IMMAS cases, with no macrocephaly or overgrowth phenotypes, highlighting the clinical variation in IMMAS.


Asunto(s)
Anomalías Múltiples , Anomalías Craneofaciales , Discapacidad Intelectual , Humanos , Anomalías Múltiples/genética , Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Fenotipo , Complejo Represivo Polycomb 2/genética
7.
Hum Mol Genet ; 28(14): 2319-2329, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30985895

RESUMEN

Disorders of sex development (DSDs) are defined as congenital conditions in which chromosomal, gonadal or anatomical sex is atypical. In many DSD cases, genetic causes remain to be elucidated. Here, we performed a case-control exome sequencing study comparing gene-based burdens of rare damaging variants between 26 DSD cases and 2625 controls. We found exome-wide significant enrichment of rare heterozygous truncating variants in the MYRF gene encoding myelin regulatory factor, a transcription factor essential for oligodendrocyte development. All three variants occurred de novo. We identified an additional 46,XY DSD case of a de novo damaging missense variant in an independent cohort. The clinical symptoms included hypoplasia of Müllerian derivatives and ovaries in 46,XX DSD patients, defective development of Sertoli and Leydig cells in 46,XY DSD patients and congenital diaphragmatic hernia in one 46,XY DSD patient. As all of these cells and tissues are or partly consist of coelomic epithelium (CE)-derived cells (CEDC) and CEDC developed from CE via proliferaiton and migration, MYRF might be related to these processes. Consistent with this hypothesis, single-cell RNA sequencing of foetal gonads revealed high expression of MYRF in CE and CEDC. Reanalysis of public chromatin immunoprecipitation sequencing data for rat Myrf showed that genes regulating proliferation and migration were enriched among putative target genes of Myrf. These results suggested that MYRF is a novel causative gene of 46,XY and 46,XX DSD and MYRF is a transcription factor regulating CD and/or CEDC proliferation and migration, which is essential for development of multiple organs.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Trastorno del Desarrollo Sexual 46,XY/genética , Proteínas de la Membrana/genética , Factores de Transcripción/genética , Trastornos del Desarrollo Sexual 46, XX/patología , Adolescente , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Preescolar , Estudios de Cohortes , Biología Computacional , Trastorno del Desarrollo Sexual 46,XY/patología , Femenino , Ontología de Genes , Gónadas/crecimiento & desarrollo , Haploinsuficiencia , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Mutación , Mutación Missense , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Secuenciación del Exoma , Adulto Joven
8.
J Hum Genet ; 66(3): 321-325, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32908218

RESUMEN

Cytosolic PEPCK deficiency (PCKDC) is a rare autosomal recessive inborn error of metabolism, which can present with hypoglycemia, lactic acidosis, and liver failure. It is caused by biallelic pathogenic variants in the PCK1 gene. Only four PCK1 variants have been previously reported in seven patients with PCKDC, and their clinical course of this condition has not been well characterized. Here, we report a Hispanic male with novel biallelic PCK1 variants, p.(Gly430Asp) and p.(His496Gln), who had a unique clinical presentation. He presented with a new onset of growth failure, elevated blood lactate, transaminitis, and abnormal urine metabolites profile, but he has not had documented hypoglycemia. Growth restriction happened due to insufficient caloric intake, and it was improved with nutritional intervention. PCKDC is a manageable disorder and therefore appropriate nutritional and clinical suspicion from typical lab abnormalities which lead to molecular confirmation tests are essential to prevent poor clinical outcomes.


Asunto(s)
Codón sin Sentido , Ingestión de Energía/genética , Insuficiencia de Crecimiento/genética , Trastornos del Crecimiento/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Secuencia de Aminoácidos , Peso al Nacer , Preescolar , Ciclo del Ácido Cítrico , Citosol/enzimología , Insuficiencia de Crecimiento/sangre , Insuficiencia de Crecimiento/orina , Femenino , Preferencias Alimentarias , Genotipo , Trastornos del Crecimiento/sangre , Trastornos del Crecimiento/orina , Humanos , Alimentos Infantiles , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Microcefalia/genética , Linaje , Fosfoenolpiruvato Carboxiquinasa (GTP)/deficiencia , Embarazo , Complicaciones del Embarazo , Convulsiones , Alineación de Secuencia , Homología de Secuencia de Aminoácido
9.
Clin Genet ; 98(6): 606-612, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32812661

RESUMEN

RBM10, is an RNA binding protein that is important for development by regulating the expression of multiple genes. RBM10 is on the X chromosome, and nonsense and frameshift RBM10 variants cause TARP syndrome in males. In a 4-year-old male, we identified a novel maternally inherited missense RBM10 variant in the RRM2 RNA binding domain, c.965C>T, p.Pro322Leu. His clinical features included intellectual disability, developmental delay, growth restriction, hypotonia, and craniofacial malformations. These features were much milder than those described in previously reported cases of TARP syndrome. By in vitro assays, we found that the mutant p.Pro322Leu RBM10 protein retained its specific RNA binding capacity, while gaining a low-affinity nonspecific RNA binding. It was normally localized to the nucleus, but its expression level was significantly reduced with a significantly short half-life. These results indicated that the p.Pro322Leu missense variant causes a developmental disorder in humans through a unique loss-of-function mechanism.


Asunto(s)
Pie Equinovaro/genética , Discapacidades del Desarrollo/genética , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/genética , Síndrome de Pierre Robin/genética , Proteínas de Unión al ARN/genética , Preescolar , Pie Equinovaro/complicaciones , Pie Equinovaro/patología , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/patología , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/patología , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Anomalías Musculoesqueléticas/complicaciones , Anomalías Musculoesqueléticas/genética , Anomalías Musculoesqueléticas/patología , Mutación Missense/genética , Fenotipo , Síndrome de Pierre Robin/complicaciones , Síndrome de Pierre Robin/patología , Secuenciación del Exoma
10.
Am J Hum Genet ; 99(4): 950-961, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27666374

RESUMEN

We describe four families with affected siblings showing unique clinical features: early-onset (before 1 year of age) progressive diffuse brain atrophy with regression, postnatal microcephaly, postnatal growth retardation, muscle weakness/atrophy, and respiratory failure. By whole-exome sequencing, we identified biallelic TBCD mutations in eight affected individuals from the four families. TBCD encodes TBCD (tubulin folding co-factor D), which is one of five tubulin-specific chaperones playing a pivotal role in microtubule assembly in all cells. A total of seven mutations were found: five missense mutations, one nonsense, and one splice site mutation resulting in a frameshift. In vitro cell experiments revealed the impaired binding between most mutant TBCD proteins and ARL2, TBCE, and ß-tubulin. The in vivo experiments using olfactory projection neurons in Drosophila melanogaster indicated that the TBCD mutations caused loss of function. The wide range of clinical severity seen in this neurodegenerative encephalopathy may result from the residual function of mutant TBCD proteins. Furthermore, the autopsied brain from one deceased individual showed characteristic neurodegenerative findings: cactus and somatic sprout formations in the residual Purkinje cells in the cerebellum, which are also seen in some diseases associated with mitochondrial impairment. Defects of microtubule formation caused by TBCD mutations may underlie the pathomechanism of this neurodegenerative encephalopathy.


Asunto(s)
Alelos , Encefalopatías/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Enfermedades Neurodegenerativas/genética , Adolescente , Edad de Inicio , Secuencia de Aminoácidos , Animales , Encefalopatías/patología , Encefalopatías/fisiopatología , Niño , Preescolar , Drosophila melanogaster/genética , Exoma , Femenino , Mutación del Sistema de Lectura/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Linaje , Sitios de Empalme de ARN/genética , Tubulina (Proteína)/metabolismo , Adulto Joven
11.
Genet Med ; 21(7): 1629-1638, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30467404

RESUMEN

PURPOSE: The diagnostic rate for Mendelian diseases by exome sequencing (ES) is typically 20-40%. The low rate is partly because ES misses deep-intronic or synonymous variants leading to aberrant splicing. In this study, we aimed to apply RNA sequencing (RNA-seq) to efficiently detect the aberrant splicings and their related variants. METHODS: Aberrant splicing in biopsied muscles from six nemaline myopathy (NM) cases unresolved by ES were analyzed with RNA-seq. Variants related to detected aberrant splicing events were analyzed with Sanger sequencing. Detected variants were screened in NM patients unresolved by ES. RESULTS: We identified a novel deep-intronic NEB pathogenic variant, c.1569+339A>G in one case, and another novel synonymous NEB pathogenic variant, c.24684G>C (p.Ser8228Ser) in three cases. The c.24684G>C variant was observed to be the most frequent among all NEB pathogenic variants in normal Japanese populations with a frequency of 1 in 178 (20 alleles in 3552 individuals), but was previously unrecognized. Expanded screening of the variant identified it in a further four previously unsolved nemaline myopathy cases. CONCLUSION: These results indicated that RNA-seq may be able to solve a large proportion of previously undiagnosed muscle diseases.


Asunto(s)
Miopatías Nemalínicas/genética , Análisis de Secuencia de ARN , Humanos , Japón , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Mutación , Empalme del ARN
12.
Ann Neurol ; 84(6): 843-853, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30412317

RESUMEN

OBJECTIVE: Approximately 5% of cerebral small vessel diseases are hereditary, which include COL4A1/COL4A2-related disorders. COL4A1/COL4A2 encode type IV collagen α1/2 chains in the basement membranes of cerebral vessels. COL4A1/COL4A2 mutations impair the secretion of collagen to the extracellular matrix, thereby resulting in vessel fragility. The diagnostic yield for COL4A1/COL4A2 variants is around 20 to 30%, suggesting other mutated genes might be associated with this disease. This study aimed to identify novel genes that cause COL4A1/COL4A2-related disorders. METHODS: Whole exome sequencing was performed in 2 families with suspected COL4A1/COL4A2-related disorders. We validated the role of COLGALT1 variants by constructing a 3-dimensional structural model, evaluating collagen ß (1-O) galactosyltransferase 1 (ColGalT1) protein expression and ColGalT activity by Western blotting and collagen galactosyltransferase assays, and performing in vitro RNA interference and rescue experiments. RESULTS: Exome sequencing demonstrated biallelic variants in COLGALT1 encoding ColGalT1, which was involved in the post-translational modification of type IV collagen in 2 unrelated patients: c.452 T > G (p.Leu151Arg) and c.1096delG (p.Glu366Argfs*15) in Patient 1, and c.460G > C (p.Ala154Pro) and c.1129G > C (p.Gly377Arg) in Patient 2. Three-dimensional model analysis suggested that p.Leu151Arg and p.Ala154Pro destabilized protein folding, which impaired enzymatic activity. ColGalT1 protein expression and ColGalT activity in Patient 1 were undetectable. RNA interference studies demonstrated that reduced ColGalT1 altered COL4A1 secretion, and rescue experiments showed that mutant COLGALT1 insufficiently restored COL4A1 production in cells compared with wild type. INTERPRETATION: Biallelic COLGALT1 variants cause cerebral small vessel abnormalities through a common molecular pathogenesis with COL4A1/COL4A2-related disorders. Ann Neurol 2018;84:843-853.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales/genética , Colágeno Tipo IV/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Línea Celular Transformada , Enfermedades de los Pequeños Vasos Cerebrales/diagnóstico por imagen , Niño , Análisis Mutacional de ADN , Glucosiltransferasas/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Modelos Moleculares , Mutagénesis , ARN Mensajero/metabolismo , Transfección
13.
J Hum Genet ; 63(5): 673-676, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29491473

RESUMEN

SNAP25 is a core component of the soluble N-ethylmaleimide-sensitive factor attachment receptor complex, which plays a critical role in synaptic vesicle exocytosis. To date, six de novo SNAP25 mutations have been reported in patients with neurological features including seizures, intellectual disability, severe speech delay, and cerebellar ataxia. Here, we analyzed an Israeli family with two affected siblings showing seizures and cerebellar dysfunction by whole-exome sequencing, and identified a novel missense SNAP25 mutation (c.176G > C, p.Arg59Pro) inherited from their unaffected father. Two SNAP25 isoforms are known, SNAP25a and SNAP25b, which each contain a different exon 5. The c.176G > C mutation found in this study was specific to SNAP25b, while five previously reported mutations were identified in exons common to both isoforms. Another was previously reported to be specific to SNAP25b. Comparing clinical features of reported patients with SNAP25 mutations, the current patients demonstrated apparently milder clinical features with normal intelligence, and no magnetic resonance imaging abnormality or facial dysmorphism. Our results expand the clinical spectrum of SNAP25 mutations.


Asunto(s)
Ataxia Cerebelosa/diagnóstico , Ataxia Cerebelosa/genética , Mutación Missense , Convulsiones/diagnóstico , Convulsiones/genética , Hermanos , Proteína 25 Asociada a Sinaptosomas/genética , Alelos , Niño , Preescolar , Análisis Mutacional de ADN , Electroencefalografía , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Israel , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Secuenciación del Exoma
14.
J Hum Genet ; 63(12): 1223-1229, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30258207

RESUMEN

Biallelic mutations in IBA57 cause a mitochondrial disorder with a broad phenotypic spectrum that ranges from severe intellectual disability to adolescent-onset spastic paraplegia. Only 21 IBA57 mutations have been reported, therefore the phenotypic spectrum of IBA57-related mitochondrial disease has not yet been fully elucidated. In this study, we performed whole-exome sequencing on a Sepharadi Jewish and Japanese family with leukodystrophy. We identified four novel biallelic variants in IBA57 in the two families: one frameshift insertion and three missense variants. The three missense variants were predicted to be disease-causing by multiple in silico tools. The 29-year-old Sepharadi Jewish male had infantile-onset optic atrophy with clinically asymptomatic leukodystrophy involving periventricular white matter. The 19-year-old younger brother, with the same compound heterozygous IBA57 variants, had a similar clinical course until 7 years of age. However, he then developed a rapidly progressive spastic paraparesis following a febrile illness. A 7-year-old Japanese girl had developmental regression, spastic quadriplegia, and abnormal periventricular white matter signal on brain magnetic resonance imaging performed at 8 months of age. She had febrile convulsions at the age of 18 months and later developed epilepsy. In summary, we have identified four novel IBA57 mutations in two unrelated families. Consequently, we describe a patient with infantile-onset optic atrophy and asymptomatic white matter involvement, thus broadening the phenotypic spectrum of biallelic IBA57 mutations.


Asunto(s)
Proteínas Portadoras/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Mutación , Fenotipo , Adulto , Femenino , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico por imagen , Humanos , Masculino
15.
Clin Genet ; 94(5): 461-466, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30019515

RESUMEN

SUZ12 is a core component of polycomb repressive complex 2 (PRC2) along with EZH2 and EED. Recently, germline mutations in the SUZ12, EZH2 and EED genes have been reported in Weaver syndrome (WS) or Weaver-like syndrome, suggesting a functional link between PRC2 deficits and WS. However, only one case of a SUZ12 mutation presenting with Weaver-like syndrome has been reported. Here, we report a missense and a frameshift mutation in SUZ12 (c.1797A>C; p.Gln599His and c.844_845del; p.Ala282Glnfs*7), both of which are novel, in two individuals. Their clinical features included postnatal overgrowth, increased bifrontal diameter, large ears, round face, horizontal chin crease and skeletal anomalies, but did not fulfill the WS diagnostic criteria. These data provide strong evidence that SUZ12 mutations cause Weaver-like syndrome.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Hipotiroidismo Congénito/diagnóstico , Hipotiroidismo Congénito/genética , Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Deformidades Congénitas de la Mano/diagnóstico , Deformidades Congénitas de la Mano/genética , Mutación , Fenotipo , Complejo Represivo Polycomb 2/genética , Alelos , Sustitución de Aminoácidos , Facies , Femenino , Genotipo , Humanos , Masculino , Proteínas de Neoplasias , Linaje , Factores de Transcripción
16.
Clin Genet ; 94(6): 548-553, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30051457

RESUMEN

We report a patient with thrombocytopenia from a Japanese family with hemophilia A spanning four generations. Various etiologies of thrombocytopenia, including genetic, immunological, and hematopoietic abnormalities, determine the prognosis for this disease. In this study, we identified a novel heterozygous mutation in a gene encoding cytochrome c, somatic (CYCS, MIM123970) using whole exome sequencing. This variant (c.301_303del:p.Lys101del) is located in the α-helix of the cytochrome c (CYCS) C-terminal domain. In silico structural analysis suggested that this mutation results in protein folding instability. CYCS is one of the key factors regulating the intrinsic apoptotic pathway and the mitochondrial respiratory chain. Using the yeast model system, we clearly demonstrated that this one amino acid deletion (in-frame) resulted in significantly reduced cytochrome c protein expression and functional defects in the mitochondrial respiratory chain, indicating that the loss of function of cytochrome c underlies thrombocytopenia. The clinical features of known CYCS variants have been reported to be confined to mild or asymptomatic thrombocytopenia, as was observed for the patient in our study. This study clearly demonstrates that thrombocytopenia can result from CYCS loss-of-function variants.


Asunto(s)
Citocromos c/química , Citocromos c/genética , Predisposición Genética a la Enfermedad , Mutación , Dominios Proteicos/genética , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Sustitución de Aminoácidos , Biomarcadores , Citocinas/sangre , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Humanos , Japón , Masculino , Persona de Mediana Edad , Linaje , Conformación Proteica en Hélice alfa , Relación Estructura-Actividad , Trombocitopenia/sangre
17.
Hum Mutat ; 38(6): 637-648, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28229514

RESUMEN

Weaver syndrome (WS) is a rare congenital overgrowth disorder caused by heterozygous mutations in EZH2 (enhancer of zeste homolog 2) or EED (embryonic ectoderm development). EZH2 and EED are core components of the polycomb repressive complex 2 (PRC2), which possesses histone methyltransferase activity and catalyzes trimethylation of histone H3 at lysine 27. Here, we analyzed eight probands with clinically suspected WS by whole-exome sequencing and identified three mutations: a 25.4-kb deletion partially involving EZH2 and CUL1 (individual 1), a missense mutation (c.707G>C, p.Arg236Thr) in EED (individual 2), and a missense mutation (c.1829A>T, p.Glu610Val) in SUZ12 (suppressor of zeste 12 homolog) (individual 3) inherited from her father (individual 4) with a mosaic mutation. SUZ12 is another component of PRC2 and germline mutations in SUZ12 have not been previously reported in humans. In vitro functional analyses demonstrated that the identified EED and SUZ12 missense mutations cause decreased trimethylation of lysine 27 of histone H3. These data indicate that loss-of-function mutations of PRC2 components are an important cause of WS.


Asunto(s)
Anomalías Múltiples/genética , Hipotiroidismo Congénito/genética , Anomalías Craneofaciales/genética , Proteínas Cullin/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Deformidades Congénitas de la Mano/genética , Complejo Represivo Polycomb 2/genética , Anomalías Múltiples/patología , Adulto , Niño , Preescolar , Hipotiroidismo Congénito/patología , Anomalías Craneofaciales/patología , Proteínas de Unión al ADN/genética , Femenino , Deformidades Congénitas de la Mano/patología , Heterocigoto , Histonas/genética , Humanos , Masculino , Metilación , Mutación , Proteínas de Neoplasias , Linaje , Mapas de Interacción de Proteínas , Factores de Transcripción
18.
Hum Mutat ; 38(3): 317-323, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28054739

RESUMEN

Congenital scoliosis (CS) occurs as a result of vertebral malformations and has an incidence of 0.5-1/1,000 births. Recently, TBX6 on chromosome 16p11.2 was reported as a disease gene for CS; about 10% of Chinese CS patients were compound heterozygotes for rare null mutations and a common haplotype defined by three SNPs in TBX6. All patients had hemivertebrae. We recruited 94 Japanese CS patients, investigated the TBX6 locus for both mutations and the risk haplotype, examined transcriptional activities of mutant TBX6 in vitro, and evaluated clinical and radiographic features. We identified TBX6 null mutations in nine patients, including a missense mutation that had a loss of function in vitro. All had the risk haplotype in the opposite allele. One of the mutations showed dominant negative effect. Although all Chinese patients had one or more hemivertebrae, two Japanese patients did not have hemivertebra. The compound heterozygosity of null mutations and the common risk haplotype in TBX6 also causes CS in Japanese patients with similar incidence. Hemivertebra was not a specific type of spinal malformation in TBX6-associated CS (TACS). A heterozygous TBX6 loss-of-function mutation has been reported in a family with autosomal-dominant spondylocostal dysostosis, but it may represent a spectrum of the same disease with TACS.


Asunto(s)
Anomalías Congénitas/genética , Haplotipos , Heterocigoto , Mutación con Pérdida de Función , Escoliosis/genética , Proteínas de Dominio T Box/genética , Adolescente , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 16 , Anomalías Congénitas/diagnóstico , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Linaje , Fenotipo , Radiografía , Escoliosis/diagnóstico
19.
J Hum Genet ; 61(2): 177-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26490182

RESUMEN

Cataract is defined as opacity in the crystalline lens and congenital cataract occurs during the first year of life. Until now, mutations of more than 50 genes in congenital cataract have been reported with various modes of inheritance. Among them, HSF4 mutations have been reported in autosomal dominant, autosomal recessive and age-related forms of cataract. The inheritance patterns of these mutations depend on their mutational positions in HSF4: autosomal dominant or recessive mutations are respectively found either in a DNA-binding domain or in (or downstream of) hydrophobic repeats. Here we report a novel homozygous HSF4 mutation (c.521T>C, p.Leu174Pro) in two affected sibs of an Iranian consanguineous family using whole exome sequencing. The mutation is predicted as highly pathogenic by in silico analysis (SIFT, Polyphen2 and MutationTaster) and is not found in any of control databases. This mutation is located in a hydrophobic repeat of the HSF4 protein, which is consistent with the mode of inheritance as an autosomal recessive trait.


Asunto(s)
Catarata/congénito , Catarata/genética , Proteínas de Unión al ADN/genética , Mutación Missense , Factores de Transcripción/genética , Femenino , Genes Recesivos , Factores de Transcripción del Choque Térmico , Homocigoto , Humanos , Masculino
20.
J Neurol Neurosurg Psychiatry ; 87(2): 212-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25995486

RESUMEN

BACKGROUND: Leigh syndrome (LS) is an early-onset progressive neurodegenerative disorder associated with mitochondrial dysfunction. LS is characterised by elevated lactate and pyruvate and bilateral symmetric hyperintense lesions in the basal ganglia, thalamus, brainstem, cerebral white matter or spinal cord on T2-weighted MRI. LS is a genetically heterogeneous disease, and to date mutations in approximately 40 genes related to mitochondrial function have been linked to the disorder. METHODS: We investigated a pair of female monozygotic twins diagnosed with LS from consanguineous healthy parents of Indian origin. Their common clinical features included optic atrophy, ophthalmoplegia, spastic paraparesis and mild intellectual disability. High-blood lactate and high-intensity signal in the brainstem on T2-weighted MRI were consistent with a clinical diagnosis of LS. To identify the genetic cause of their condition, we performed whole exome sequencing. RESULTS: We identified a homozygous nonsense mutation in C12orf65 (NM_001143905; c.346delG, p.V116*) in the affected twins. Interestingly, the identical mutation was previously reported in an Indian family with Charcot-Marie Tooth disease type 6, which displayed some overlapping clinical features with the twins. CONCLUSIONS: We demonstrate that the identical nonsense mutation in C12orf65 can result in different clinical features, suggesting the involvement of unknown modifiers.


Asunto(s)
Enfermedad de Leigh/genética , Proteínas Mitocondriales/genética , Factores de Terminación de Péptidos/genética , Adolescente , Edad de Inicio , Pueblo Asiatico , Encéfalo/patología , Enfermedad de Charcot-Marie-Tooth/genética , Niño , Codón sin Sentido/genética , Consanguinidad , Exoma/genética , Femenino , Humanos , Judíos , Enfermedad de Leigh/patología , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/patología , Linaje , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA