Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Hum Mol Genet ; 32(7): 1184-1192, 2023 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-36355422

RESUMO

Congenital hearing impairment (HI) is a genetically highly heterogeneous disorder in which prompt recognition and intervention are crucial to optimize outcomes. In this study, we used exome sequencing to investigate a large consanguineous Pakistani family with eight affected individuals showing bilateral severe-to-profound HI. This identified a homozygous splice region variant in STX4 (c.232 + 6T>C), which causes exon skipping and a frameshift, that segregated with HI (two-point logarithm of odds (LOD) score = 5.9). STX4, a member of the syntaxin family, is a component of the SNARE machinery involved in several vesicle transport and recycling pathways. In silico analysis showed that murine orthologue Stx4a is highly and widespread expressed in the developing and adult inner ear. Immunofluorescent imaging revealed localization of STX4A in the cell body, cell membrane and stereocilia of inner and outer hair cells. Furthermore, a morpholino-based knockdown of stx4 in zebrafish showed an abnormal startle response, morphological and developmental defects, and a disrupted mechanotransduction function in neuromast hair cells measured via FM1-43 uptake. Our findings indicate that STX4 dysfunction leads to HI in humans and zebrafish and supports the evolutionary conserved role of STX4 in inner ear development and hair cell functioning.


Assuntos
Mecanotransdução Celular , Peixe-Zebra , Adulto , Humanos , Animais , Camundongos , Peixe-Zebra/genética , Proteínas Qa-SNARE/genética , Audição/genética , Células Ciliadas Auditivas Externas
2.
Am J Hum Genet ; 108(12): 2368-2384, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34800363

RESUMO

The 2-oxoglutarate dehydrogenase-like (OGDHL) protein is a rate-limiting enzyme in the Krebs cycle that plays a pivotal role in mitochondrial metabolism. OGDHL expression is restricted mainly to the brain in humans. Here, we report nine individuals from eight unrelated families carrying bi-allelic variants in OGDHL with a range of neurological and neurodevelopmental phenotypes including epilepsy, hearing loss, visual impairment, gait ataxia, microcephaly, and hypoplastic corpus callosum. The variants include three homozygous missense variants (p.Pro852Ala, p.Arg244Trp, and p.Arg299Gly), three compound heterozygous single-nucleotide variants (p.Arg673Gln/p.Val488Val, p.Phe734Ser/p.Ala327Val, and p.Trp220Cys/p.Asp491Val), one homozygous frameshift variant (p.Cys553Leufs∗16), and one homozygous stop-gain variant (p.Arg440Ter). To support the pathogenicity of the variants, we developed a novel CRISPR-Cas9-mediated tissue-specific knockout with cDNA rescue system for dOgdh, the Drosophila ortholog of human OGDHL. Pan-neuronal knockout of dOgdh led to developmental lethality as well as defects in Krebs cycle metabolism, which was fully rescued by expression of wild-type dOgdh. Studies using the Drosophila system indicate that p.Arg673Gln, p.Phe734Ser, and p.Arg299Gly are severe loss-of-function alleles, leading to developmental lethality, whereas p.Pro852Ala, p.Ala327Val, p.Trp220Cys, p.Asp491Val, and p.Arg244Trp are hypomorphic alleles, causing behavioral defects. Transcript analysis from fibroblasts obtained from the individual carrying the synonymous variant (c.1464T>C [p.Val488Val]) in family 2 showed that the synonymous variant affects splicing of exon 11 in OGDHL. Human neuronal cells with OGDHL knockout exhibited defects in mitochondrial respiration, indicating the essential role of OGDHL in mitochondrial metabolism in humans. Together, our data establish that the bi-allelic variants in OGDHL are pathogenic, leading to a Mendelian neurodevelopmental disease in humans.


Assuntos
Ataxia/genética , Epilepsia/genética , Perda Auditiva/genética , Complexo Cetoglutarato Desidrogenase/genética , Mutação , Transtornos do Neurodesenvolvimento/genética , Transtornos da Visão/genética , Alelos , Animais , Células Cultivadas , Criança , Estudos de Coortes , Análise Mutacional de DNA , Drosophila melanogaster/genética , Saúde da Família , Feminino , Fibroblastos , Humanos , Masculino , Splicing de RNA
3.
Am J Med Genet A ; 194(4): e63478, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37975178

RESUMO

Bilateral perisylvian polymicrogyria (BPP) is a structural malformation of the cerebral cortex that can be caused by several genetic abnormalities. The most common clinical manifestations of BPP include intellectual disability and epilepsy. Cytoplasmic FMRP-interacting protein 2 (CYFIP2) is a protein that interacts with the fragile X mental retardation protein (FMRP). CYFIP2 variants can cause various brain structural abnormalities with the most common clinical manifestations of intellectual disability, epileptic encephalopathy and dysmorphic features. We present a girl with multiple disabilities and BPP caused by a heterozygous, novel, likely pathogenic variant (c.1651G>C: p.(Val551Leu) in the CYFIP2 gene. Our case report broadens the spectrum of genetic diversity associated with BPP by incorporating CYFIP2.


Assuntos
Anormalidades Múltiplas , Encefalopatias , Deficiência Intelectual , Malformações do Desenvolvimento Cortical , Polimicrogiria , Feminino , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/complicações , Polimicrogiria/genética , Polimicrogiria/complicações , Anormalidades Múltiplas/genética , Malformações do Desenvolvimento Cortical/diagnóstico , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/complicações , Encefalopatias/complicações , Proteínas Adaptadoras de Transdução de Sinal/genética
4.
Clin Genet ; 104(4): 499-501, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37311648

RESUMO

A short report with two affected siblings from consanguineous family born with intellectual disability, motor disability, language deficit, and hearing impairment and found to carry biallelic nonsense variant in KPTN gene known to be associated with KPTN gene related syndrome.


Assuntos
Pessoas com Deficiência , Perda Auditiva , Deficiência Intelectual , Transtornos Motores , Humanos , Consanguinidade , Perda Auditiva/genética , Deficiência Intelectual/genética , Proteínas dos Microfilamentos/genética , Linhagem , Fenótipo , Síndrome
5.
Clin Genet ; 103(6): 699-703, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36807241

RESUMO

Hereditary deafness and retinal dystrophy are each genetically heterogenous and clinically variable. Three small unrelated families segregating the combination of deafness and retinal dystrophy were studied by exome sequencing (ES). The proband of Family 1 was found to be compound heterozygous for NM_004525.3: LRP2: c.5005A > G, p.(Asn1669Asp) and c.149C > G, p.(Thr50Ser). In Family 2, two sisters were found to be compound heterozygous for LRP2 variants, p.(Tyr3933Cys) and an experimentally confirmed c.7715 + 3A > T consensus splice-altering variant. In Family 3, the proband is compound heterozygous for a consensus donor splice site variant LRP2: c.8452_8452 + 1del and p.(Cys3150Tyr). In mouse cochlea, Lrp2 is expressed abundantly in the stria vascularis marginal cells demonstrated by smFISH, single-cell and single-nucleus RNAseq, suggesting that a deficiency of LRP2 may compromise the endocochlear potential, which is required for hearing. LRP2 variants have been associated with Donnai-Barrow syndrome and other multisystem pleiotropic phenotypes different from the phenotypes of the four cases reported herein. Our data expand the phenotypic spectrum associated with pathogenic variants in LRP2 warranting their consideration in individuals with a combination of hereditary hearing loss and retinal dystrophy.


Assuntos
Surdez , Perda Auditiva Neurossensorial , Perda Auditiva , Miopia , Distrofias Retinianas , Animais , Camundongos , Humanos , Perda Auditiva Neurossensorial/genética , Surdez/genética , Miopia/genética , Mutação , Linhagem , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética
6.
Cerebellum ; 22(5): 1029-1033, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35987951

RESUMO

To the best of our knowledge, this is the first case to address episodic ataxia (EA) as a possible phenotypic feature of HECW2-related disorder. This single case study describes a 26-year-old female born at term with mild intellectual disability, neonatal hypotonia, and a history of febrile seizures who presented with paroxysmal events since the age of 2. These episodes include frequent falls due to imbalance, dilated pupils, vertigo, diaphoresis, nausea, vomiting, and nystagmus. Brain imaging was normal. A prolonged electroencephalogram (EEG) revealed interictal epileptiform discharges but failed to capture her clinical events. For several years, she was treated for presumed focal seizures with preserved awareness and trialed on adequate dosing of several antiepileptic medications without improvement. After 25 years, given the more prolonged nature of her episodes and the mild interictal cerebellar signs, empiric treatment with acetazolamide was initiated for a presumed diagnosis of EA. Acetazolamide treatment led to a dramatic reduction in event frequency and severity. The initial EA genetic panel was negative. Clinical exome sequence analysis revealed a novel pathogenic de novo missense variant in the HECW2 gene [c.3829 T > C;(p.Tyr1277His)], located in the HECT domain. HECW2 variants are associated with neurodevelopmental delay, hypotonia, and epilepsy. This study expands the genetic and clinical spectrum of HECW2-related disorder and adds EA to the phenotypic spectrum in affected individuals.


Assuntos
Acetazolamida , Ataxia , Adulto , Feminino , Humanos , Recém-Nascido , Acetazolamida/uso terapêutico , Anticonvulsivantes/uso terapêutico , Ataxia/diagnóstico por imagem , Ataxia/tratamento farmacológico , Ataxia/genética , Epilepsia , Hipotonia Muscular/genética , Ubiquitina-Proteína Ligases/genética
7.
J Med Genet ; 59(7): 669-677, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34321324

RESUMO

BACKGROUND: Variants in HECW2 have recently been reported to cause a neurodevelopmental disorder with hypotonia, seizures and impaired language; however, only six variants have been reported and the clinical characteristics have only broadly been defined. METHODS: Molecular and clinical data were collected from clinical and research cohorts. Massive parallel sequencing was performed and identified individuals with a HECW2-related neurodevelopmental disorder. RESULTS: We identified 13 novel missense variants in HECW2 in 22 unpublished cases, of which 18 were confirmed to have a de novo variant. In addition, we reviewed the genotypes and phenotypes of previously reported and new cases with HECW2 variants (n=35 cases). All variants identified are missense, and the majority of likely pathogenic and pathogenic variants are located in or near the C-terminal HECT domain (88.2%). We identified several clustered variants and four recurrent variants (p.(Arg1191Gln);p.(Asn1199Lys);p.(Phe1327Ser);p.(Arg1330Trp)). Two variants, (p.(Arg1191Gln);p.(Arg1330Trp)), accounted for 22.9% and 20% of cases, respectively. Clinical characterisation suggests complete penetrance for hypotonia with or without spasticity (100%), developmental delay/intellectual disability (100%) and developmental language disorder (100%). Other common features are behavioural problems (88.9%), vision problems (83.9%), motor coordination/movement (75%) and gastrointestinal issues (70%). Seizures were present in 61.3% of individuals. Genotype-phenotype analysis shows that HECT domain variants are more frequently associated with cortical visual impairment and gastrointestinal issues. Seizures were only observed in individuals with variants in or near the HECT domain. CONCLUSION: We provide a comprehensive review and expansion of the genotypic and phenotypic spectrum of HECW2 disorders, aiding future molecular and clinical diagnosis and management.


Assuntos
Deficiência Intelectual , Transtornos do Neurodesenvolvimento , Ubiquitina-Proteína Ligases , Genótipo , Humanos , Deficiência Intelectual/genética , Hipotonia Muscular/genética , Hipotonia Muscular/patologia , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Convulsões/genética , Ubiquitina-Proteína Ligases/genética
8.
Ann Hum Genet ; 86(6): 291-296, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36039988

RESUMO

Orofaciodigital syndrome (OFD) is clinically heterogeneous and is characterized by abnormalities in the oral cavity, facial features, digits, and central nervous system. At least 18 subtypes of the condition have been described in the literature. OFD is caused by variants in several genes with overlapping phenotypes. We studied a consanguineous Pakistani family with two affected siblings with an atypical form of OFD type 4 (OFD4). In addition to the typical features of OFD4 that include limb defects and growth retardation, the siblings displayed rare features of scaphocephaly and seizures. Exome sequencing analysis revealed a novel homozygous splice site variant c.257-1G>A in TCTN3 that segregated with disease. This homozygous splice site variant in TCTN3 is most likely the underlying cause of the atypical form of OFD4 observed in this family. Our results contribute to the phenotypic spectrum of TCTN3 associated ciliopathies and will facilitate better clinical diagnosis.


Assuntos
Ciliopatias , Síndromes Orofaciodigitais , Humanos , Síndromes Orofaciodigitais/genética , Ciliopatias/diagnóstico , Mutação , Homozigoto , Linhagem
9.
Hum Genet ; 141(3-4): 413-430, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34291353

RESUMO

Hearing impairment (HI) is one of the most common sensory disabilities with exceptionally high genetic heterogeneity. Of genetic HI cases, 30% are syndromic and 70% are nonsyndromic. For nonsyndromic (NS) HI, 77% of the cases are due to autosomal recessive (AR) inheritance. ARNSHI is usually congenital/prelingual, severe-to-profound, affects all frequencies and is not progressive. Thus far, 73 ARNSHI genes have been identified. Populations with high rates of consanguinity have been crucial in the identification of ARNSHI genes, and 92% (67/73) of these genes were identified in consanguineous families. Recent changes in genomic technologies and analyses have allowed a shift towards ARNSHI gene discovery in outbred populations. The latter is crucial towards understanding the genetic architecture of ARNSHI in diverse and understudied populations. We present an overview of the 73 ARNSHI genes, the methods used to identify them, including next-generation sequencing which revolutionized the field, and new technologies that show great promise in advancing ARNSHI discoveries.


Assuntos
Surdez , Perda Auditiva , Consanguinidade , Surdez/genética , Genes Recessivos , Perda Auditiva/genética , Humanos , Linhagem
10.
Hum Genet ; 141(3-4): 951-963, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34410490

RESUMO

In this study, we investigated the association of ACAN variants with otosclerosis, a frequent cause of hearing loss among young adults. We sequenced the coding, 5'-UTR and 3'-UTR regions of ACAN in 1497 unrelated otosclerosis cases and 1437 matched controls from six different subpopulations. The association between variants in ACAN and the disease risk was tested through single variant and gene-based association tests. After correction for multiple testing, 14 variants were significantly associated with otosclerosis, ten of which represented independent association signals. Eight variants showed a consistent association across all subpopulations. Allelic odds ratios of the variants identified four predisposing and ten protective variants. Gene-based tests showed an association of very rare variants in the 3'-UTR with the phenotype. The associated exonic variants are all located in the CS domain of ACAN and include both protective and predisposing variants with a broad spectrum of effect sizes and population frequencies. This includes variants with strong effect size and low frequency, typical for monogenic diseases, to low effect size variants with high frequency, characteristic for common complex traits. This single-gene allelic spectrum with both protective and predisposing alleles is unique in the field of complex diseases. In conclusion, these findings are a significant advancement to the understanding of the etiology of otosclerosis.


Assuntos
Otosclerose , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Agrecanas/genética , Suscetibilidade a Doenças , Frequência do Gene , Predisposição Genética para Doença , Humanos , Otosclerose/genética , Fenótipo , Polimorfismo de Nucleotídeo Único
11.
J Med Genet ; 58(11): 743-751, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-32978268

RESUMO

OBJECTIVE: To investigate the diagnostic value of implementing a stepwise genetic testing strategy (SGTS) in genetically unsolved cases with dystrophinopathies. METHODS: After routine genetic testing in 872 male patients with highly suspected dystrophinopathies, we identified 715 patients with a pathogenic DMD variant. Of the 157 patients who had no pathogenic DMD variants and underwent a muscle biopsy, 142 patients were confirmed to have other myopathies, and 15 suspected dystrophinopathies remained genetically undiagnosed. These 15 patients underwent a more comprehensive evaluation as part of the SGTS pipeline, which included the stepwise analysis of dystrophin mRNA, short-read whole-gene DMD sequencing, long-read whole-gene DMD sequencing and in silico bioinformatic analyses. RESULTS: SGTS successfully yielded a molecular diagnosis of dystrophinopathy in 11 of the 15 genetically unsolved cases. We identified 8 intronic and 2 complex structural variants (SVs) leading to aberrant splicing in 10 of 11 patients, of which 9 variants were novel. In one case, a molecular defect was detected on mRNA and protein level only. Aberrant splicing mechanisms included 6 pseudoexon inclusions and 4 alterations of splice sites and splicing regulatory elements. We showed for the first time the exonisation of a MER48 element as a novel pathogenic mechanism in dystrophinopathies. CONCLUSION: Our study highlights the high diagnostic utility of implementing a SGTS pipeline in dystrophinopathies with intronic variants and complex SVs.


Assuntos
Distrofina/genética , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Adolescente , Biópsia , Criança , Pré-Escolar , Éxons , Testes Genéticos/métodos , Humanos , Íntrons , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/patologia , Distrofias Musculares/diagnóstico por imagem , Adulto Jovem
12.
BMC Ophthalmol ; 22(1): 69, 2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35148716

RESUMO

BACKGROUND: The aim of this study is to identify the genetic defects in a Chinese family with fundus albipunctatus. METHODS: Complete ophthalmic examinations, including slit-lamp biomicroscopy, dilated indirect ophthalmoscopy, fundus photography, autofluorescence, swept source optical coherence tomography (SS-OCT) and full-field electroretinography (ffERG) were performed. Genomic DNA was extracted from blood samples and whole genome sequencing was performed. Variants were validated with Sanger sequencing. RESULTS: Six members in this Chinese family, including three affected individuals and three controls, were recruited in this study. The ophthalmic examination of three recruited patients was consistent with fundus albipunctatus. Three variants, a novel frameshift deletion c.39delA [p.(Val14CysfsX47] and a haplotype of two rare missense variants, c.683G > A [p.(Arg228Gln)] along with c.710A > G [p.(Tyr237Cys], within the retinal dehydrogenase 5 (RDH5) gene were found to segregate with fundus albipunctatus in this family in an autosomal recessive matter. CONCLUSION: We identified novel compound heterozygous variants in RDH5 responsible for fundus albipunctatus in a large Chinese family. The results of our study further broaden the genetic defects of RDH5 associated with fundus albipunctatus.


Assuntos
Oxirredutases do Álcool , Cegueira Noturna , Distrofias Retinianas , Oxirredutases do Álcool/genética , China , Eletrorretinografia , Humanos , Mutação , Linhagem , Reação em Cadeia da Polimerase , Retinal Desidrogenase , Doenças Retinianas , Tomografia de Coerência Óptica
13.
Hum Genet ; 140(7): 1011-1029, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33710394

RESUMO

The genetics of autosomal recessive intellectual disability (ARID) has mainly been studied in consanguineous families, however, founder populations may also be of interest to study intellectual disability (ID) and the contribution of ARID. Here, we used a genotype-driven approach to study the genetic landscape of ID in the founder population of Finland. A total of 39 families with syndromic and non-syndromic ID were analyzed using exome sequencing, which revealed a variant in a known ID gene in 27 families. Notably, 75% of these variants in known ID genes were de novo or suspected de novo (64% autosomal dominant; 11% X-linked) and 25% were inherited (14% autosomal recessive; 7% X-linked; and 4% autosomal dominant). A dual molecular diagnosis was suggested in two families (5%). Via additional analysis and molecular testing, we identified three cases with an abnormal molecular karyotype, including chr21q22.12q22.2 uniparental disomy with a mosaic interstitial 2.7 Mb deletion covering DYRK1A and KCNJ6. Overall, a pathogenic or likely pathogenic variant was identified in 64% (25/39) of the families. Last, we report an alternate inheritance model for 3 known ID genes (UBA7, DDX47, DHX58) and discuss potential candidate genes for ID, including SYPL1 and ERGIC3 with homozygous founder variants and de novo variants in POLR2F and DNAH3. In summary, similar to other European populations, de novo variants were the most common variants underlying ID in the studied Finnish population, with limited contribution of ARID to ID etiology, though mainly driven by founder and potential founder variation in the latter case.


Assuntos
Exoma/genética , Deficiência Intelectual/genética , Família , Feminino , Finlândia , Genes Recessivos/genética , Predisposição Genética para Doença/genética , Genótipo , Homozigoto , Humanos , Masculino , Linhagem , Sequenciamento do Exoma/métodos
14.
Genet Med ; 23(11): 2122-2137, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34345025

RESUMO

PURPOSE: Pathogenic variants in SETD1B have been associated with a syndromic neurodevelopmental disorder including intellectual disability, language delay, and seizures. To date, clinical features have been described for 11 patients with (likely) pathogenic SETD1B sequence variants. This study aims to further delineate the spectrum of the SETD1B-related syndrome based on characterizing an expanded patient cohort. METHODS: We perform an in-depth clinical characterization of a cohort of 36 unpublished individuals with SETD1B sequence variants, describing their molecular and phenotypic spectrum. Selected variants were functionally tested using in vitro and genome-wide methylation assays. RESULTS: Our data present evidence for a loss-of-function mechanism of SETD1B variants, resulting in a core clinical phenotype of global developmental delay, language delay including regression, intellectual disability, autism and other behavioral issues, and variable epilepsy phenotypes. Developmental delay appeared to precede seizure onset, suggesting SETD1B dysfunction impacts physiological neurodevelopment even in the absence of epileptic activity. Males are significantly overrepresented and more severely affected, and we speculate that sex-linked traits could affect susceptibility to penetrance and the clinical spectrum of SETD1B variants. CONCLUSION: Insights from this extensive cohort will facilitate the counseling regarding the molecular and phenotypic landscape of newly diagnosed patients with the SETD1B-related syndrome.


Assuntos
Epilepsia , Histona-Lisina N-Metiltransferase , Deficiência Intelectual , Transtornos do Neurodesenvolvimento , Epilepsia/diagnóstico , Epilepsia/genética , Histona-Lisina N-Metiltransferase/genética , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Masculino , Transtornos do Neurodesenvolvimento/diagnóstico , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Convulsões/diagnóstico , Convulsões/genética
15.
J Hum Genet ; 66(12): 1169-1175, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34226616

RESUMO

Congenital hearing impairment (HI) is genetically heterogeneous making its genetic diagnosis challenging. Investigation of novel HI genes and variants will enhance our understanding of the molecular mechanisms and to aid genetic diagnosis. We performed exome sequencing and analysis using DNA samples from affected members of two large families from Ghana and Pakistan, segregating autosomal-dominant (AD) non-syndromic HI (NSHI). Using in silico approaches, we modeled and evaluated the effect of the likely pathogenic variants on protein structure and function. We identified two likely pathogenic variants in SLC12A2, c.2935G>A:p.(E979K) and c.2939A>T:p.(E980V), which segregate with NSHI in a Ghanaian and Pakistani family, respectively. SLC12A2 encodes an ion transporter crucial in the homeostasis of the inner ear endolymph and has recently been reported to be implicated in syndromic and non-syndromic HI. Both variants were mapped to alternatively spliced exon 21 of the SLC12A2 gene. Exon 21 encodes for 17 residues in the cytoplasmatic tail of SLC12A2, is highly conserved between species, and preferentially expressed in cochlear tissues. A review of previous studies and our current data showed that out of ten families with either AD non-syndromic or syndromic HI, eight (80%) had variants within the 17 amino acid residue region of exon 21 (48 bp), suggesting that this alternate domain is critical to the transporter activity in the inner ear. The genotypic spectrum of SLC12A2 was expanded and the involvement of SLC12A2 in ADNSHI was confirmed. These results also demonstrate the role that SLC12A2 plays in ADNSHI in diverse populations including sub-Saharan Africans.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Perda Auditiva/diagnóstico , Perda Auditiva/genética , Mutação , Membro 2 da Família 12 de Carreador de Soluto/genética , Alelos , Sequência de Aminoácidos , Feminino , Genótipo , Humanos , Masculino , Modelos Moleculares , Linhagem , Fenótipo , Análise de Sequência de DNA , Membro 2 da Família 12 de Carreador de Soluto/química , Relação Estrutura-Atividade , Sequenciamento do Exoma
16.
J Hum Genet ; 66(10): 1009-1018, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33879837

RESUMO

BACKGROUND: Wolfram syndrome (WFS) is characterized by deafness, diabetes mellitus, and diabetes insipidus along with optic atrophy. WFS has an autosomal recessive mode of inheritance and is due to variants in WFS1 and CISD2. METHODS: We evaluated the underlying molecular etiology of three affected members of a consanguineous family with hearing impairment, bicuspid aortic valve, diabetes mellitus and insipidus, clinodactyly, and gastrointestinal tract abnormalities via exome sequencing approach. We correlated clinical and imaging data with the genetic findings and their associated phenotypes. RESULTS: We identified a homozygous missense variant p.(Asn1097Lys) in CDK13, a gene previously associated with autosomal dominant congenital heart defects, dysmorphic facial features, clinodactyly, gastrointestinal tract abnormalities, intellectual developmental disorder, and seizures with variable phenotypic features. CONCLUSION: We report a homozygous variant in CDK13 and suggest that this gene causes an autosomal recessive disorder with hearing impairment, bicuspid aortic valve, diabetes mellitus and insipidus, clinodactyly, and gastrointestinal tract abnormalities.


Assuntos
Proteína Quinase CDC2/genética , Surdez/genética , Predisposição Genética para Doença , Atrofia Óptica/genética , Síndrome de Wolfram/genética , Adolescente , Adulto , Doença da Válvula Aórtica Bicúspide/genética , Doença da Válvula Aórtica Bicúspide/patologia , Criança , Pré-Escolar , Consanguinidade , Surdez/complicações , Surdez/patologia , Diabetes Mellitus/genética , Feminino , Trato Gastrointestinal/anormalidades , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Perda Auditiva , Homozigoto , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto/genética , Atrofia Óptica/complicações , Atrofia Óptica/patologia , Síndrome de Wolfram/complicações , Síndrome de Wolfram/epidemiologia , Síndrome de Wolfram/patologia , Adulto Jovem
17.
BMC Ophthalmol ; 21(1): 353, 2021 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-34610801

RESUMO

BACKGROUND: The aim of this study is to identify the genetic defect in a Chinese family with congenital aniridia combined with cataract and nystagmus. METHODS: Complete ophthalmic examinations, including slit-lamp biomicroscopy, dilated indirect ophthalmoscopy, anterior segment photography, and anterior segment optical coherence tomography (OCT) were performed. Blood samples were collected from all family members and genomic DNA was extracted. Genome sequencing was performed in all family members and Sanger sequencing was used to verify variant breakpoints. RESULTS: All the thirteen members in this Chinese family, including seven patients and six normal people, were recruited in this study. The ophthalmic examination of affected patients in this family was consistent with congenital aniridia combined with cataract and nystagmus. A novel heterozygous deletion (NC_000011.10:g.31802307_31806556del) containing the 5' region of PAX6 gene was detected that segregated with the disease. CONCLUSION: We detected a novel deletion in PAX6 responsible for congenital aniridia in the affected individuals of this Chinese family. The novel 4.25 kb deletion in PAX6 gene of our study would further broaden the genetic defects of PAX6 associated with congenital aniridia.


Assuntos
Aniridia , Catarata , Proteínas do Olho , Fator de Transcrição PAX6 , Aniridia/complicações , Aniridia/genética , Catarata/complicações , Catarata/genética , China , Proteínas do Olho/genética , Humanos , Fator de Transcrição PAX6/genética , Linhagem
18.
Hum Mutat ; 41(2): 412-419, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31660686

RESUMO

We report a likely pathogenic splice-altering AP4S1 intronic variant in two sisters with progressive spastic paraplegia, global developmental delay, shy character, and foot deformities. Sequencing was completed on whole-blood messenger RNA (mRNA) and analyzed for gene expression outliers after exome sequencing analysis failed to identify a causative variant. AP4S1 was identified as an outlier and contained a rare homozygous variant located three bases upstream of exon 5 (NC_000014.8(NM_007077.4):c.295-3C>A). Confirmed by additional RNA-seq, reverse-transcription polymerase chain reaction, and Sanger sequencing, this variant corresponded with exon 5, including skipping, altered isoform usage, and loss of expression from the canonical isoform 2 (NM_001128126.3). Previously, loss-of-function variants within AP4S1 were associated with a quadriplegic cerebral palsy-6 phenotype, AP-4 Deficiency Syndrome. In this study, the inclusion of mRNA-seq allowed for the identification of a previously missed splice-altering variant, and thereby expands the mutational spectrum of AP-4 Deficiency Syndrome to include impacts to some tissue-dependent isoforms.


Assuntos
Complexo 4 de Proteínas Adaptadoras/genética , Processamento Alternativo , Estudos de Associação Genética , Predisposição Genética para Doença , Íntrons , Irmãos , Paraplegia Espástica Hereditária/diagnóstico , Paraplegia Espástica Hereditária/genética , Alelos , Feminino , Estudos de Associação Genética/métodos , Genótipo , Humanos , Linhagem , Fenótipo
19.
J Hum Genet ; 65(2): 187-192, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31656313

RESUMO

Autosomal-recessive (AR) nonsyndromic hearing impairment (NSHI) displays a high degree of genetic heterogeneity with >100 genes identified. Recently, TMEM132E, which is highly expressed in inner hair cells, was suggested as a novel ARNSHI gene for DFNB99. A missense variant c.1259G>A: p.(Arg420Gln) in TMEM132E was identified that segregated with ARNSHI in a single Chinese family with two affected members. In the present study, a family of Pakistani origin with prelingual profound sensorineural hearing impairment displaying AR mode of inheritance was investigated via exome and Sanger sequencing. Compound heterozygous variants c.382G>T: p.(Ala128Ser) and c.2204C>T: p.(Pro735Leu) in TMEM132E were observed in affected but not in unaffected family members. TMEM132E variants identified in this and the previously reported ARNSHI family are located in the extracellular domain. In conclusion, we present a second ARNSHI family with TMEM132E variants which strengthens the evidence of the involvement of this gene in the etiology of ARNSHI.


Assuntos
Surdez/genética , Perda Auditiva Neurossensorial/genética , Proteínas de Membrana/genética , Povo Asiático , Surdez/diagnóstico , Exoma/genética , Feminino , Genes Recessivos , Perda Auditiva Neurossensorial/diagnóstico , Heterozigoto , Humanos , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Linhagem
20.
Exp Mol Pathol ; 115: 104471, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32446860

RESUMO

Megalencephaly-polymicrogyria-polydactyly-hydrocephalus (MPPH) syndrome is a developmental brain disorder characterized by an enlarged brain size with bilateral perisylvian polymicrogyria and a variable degree of ventriculomegaly. MPPH syndrome is associated with oromotor dysfunction, epilepsy, intellectual disability and postaxial hexadactyly. The molecular diagnosis of this disorder is established by the identification of a pathogenic variant in either AKT3, CCND2 or PIK3R2. Previously reported AKT3 variants are associated with various brain abnormalities and may lead to megalencephaly. MPPH syndrome is usually due to germline pathogenic AKT3 variants. Somatic mosaic pathogenic variants associated with hemimegalencephaly, which is similar to MPPH, have also been observed. A Hungarian Roma family with two half-siblings, which present with intellectual disability, dysmorphic features, epilepsy, brain malformations, and megalencephaly was studied. Whole exome sequencing (WES) analysis was performed. WES analysis revealed a heterozygous c.1393C > T p.(Arg465Trp) pathogenic missense AKT3 variant in both affected half-siblings. The variant was verified via Sanger sequencing and was not present in the DNA sample from the healthy mother, which was derived from peripheral blood, suggesting maternal germline mosaicism. In conclusion, this is the first report in which maternal germline mosaicism of a rare pathogenic AKT3 variant leads to autosomal dominantly inherited MPPH syndrome.


Assuntos
Dedos/anormalidades , Células Germinativas/metabolismo , Hidrocefalia/congênito , Padrões de Herança/genética , Megalencefalia/genética , Mosaicismo , Polidactilia/genética , Polimicrogiria/genética , Proteínas Proto-Oncogênicas c-akt/genética , Dedos do Pé/anormalidades , Adolescente , Criança , Feminino , Dedos/diagnóstico por imagem , Humanos , Hidrocefalia/diagnóstico por imagem , Hidrocefalia/genética , Imageamento por Ressonância Magnética , Masculino , Megalencefalia/diagnóstico por imagem , Linhagem , Fenótipo , Polidactilia/diagnóstico por imagem , Polimicrogiria/diagnóstico por imagem , Irmãos , Síndrome , Dedos do Pé/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA