Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(7): 1184-1192, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36355422

RESUMEN

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.


Asunto(s)
Mecanotransducción Celular , Pez Cebra , Adulto , Humanos , Animales , Ratones , Pez Cebra/genética , Proteínas Qa-SNARE/genética , Audición/genética , Células Ciliadas Auditivas Externas
2.
Am J Hum Genet ; 108(12): 2368-2384, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34800363

RESUMEN

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.


Asunto(s)
Ataxia/genética , Epilepsia/genética , Pérdida Auditiva/genética , Complejo Cetoglutarato Deshidrogenasa/genética , Mutación , Trastornos del Neurodesarrollo/genética , Trastornos de la Visión/genética , Alelos , Animales , Células Cultivadas , Niño , Estudios de Cohortes , Análisis Mutacional de ADN , Drosophila melanogaster/genética , Salud de la Familia , Femenino , Fibroblastos , Humanos , Masculino , Empalme del ARN
3.
Am J Hum Genet ; 108(9): 1578-1589, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34265237

RESUMEN

Thoracic aortic aneurysm (TAA) is characterized by dilation of the aortic root or ascending/descending aorta. TAA is a heritable disease that can be potentially life threatening. While 10%-20% of TAA cases are caused by rare, pathogenic variants in single genes, the origin of the majority of TAA cases remains unknown. A previous study implicated common variants in FBN1 with TAA disease risk. Here, we report a genome-wide scan of 1,351 TAA-affected individuals and 18,295 control individuals from the Cardiovascular Health Improvement Project and Michigan Genomics Initiative at the University of Michigan. We identified a genome-wide significant association with TAA for variants within the third intron of TCF7L2 following replication with meta-analysis of four additional independent cohorts. Common variants in this locus are the strongest known genetic risk factor for type 2 diabetes. Although evidence indicates the presence of different causal variants for TAA and type 2 diabetes at this locus, we observed an opposite direction of effect. The genetic association for TAA colocalizes with an aortic eQTL of TCF7L2, suggesting a functional relationship. These analyses predict an association of higher expression of TCF7L2 with TAA disease risk. In vitro, we show that upregulation of TCF7L2 is associated with BCL2 repression promoting vascular smooth muscle cell apoptosis, a key driver of TAA disease.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Diabetes Mellitus Tipo 2/genética , Células Endoteliales/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Sitios de Carácter Cuantitativo , Proteína 2 Similar al Factor de Transcripción 7/genética , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta Torácica/metabolismo , Aneurisma de la Aorta Torácica/patología , Estudios de Casos y Controles , Caspasa 3/genética , Caspasa 3/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Endoteliales/patología , Regulación de la Expresión Génica , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Intrones , Michigan , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Mutación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
4.
Am J Med Genet A ; 194(4): e63478, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37975178

RESUMEN

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.


Asunto(s)
Anomalías Múltiples , Encefalopatías , Discapacidad Intelectual , Malformaciones del Desarrollo Cortical , Polimicrogiria , Femenino , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/complicaciones , Polimicrogiria/genética , Polimicrogiria/complicaciones , Anomalías Múltiples/genética , Malformaciones del Desarrollo Cortical/diagnóstico , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/complicaciones , Encefalopatías/complicaciones , Proteínas Adaptadoras Transductoras de Señales/genética
5.
BMC Genomics ; 24(1): 303, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37277705

RESUMEN

BACKGROUND: Analysis of imputed genotypes is an important and routine component of genome-wide association studies and the increasing size of imputation reference panels has facilitated the ability to impute and test low-frequency variants for associations. In the context of genotype imputation, the true genotype is unknown and genotypes are inferred with uncertainty using statistical models. Here, we present a novel method for integrating imputation uncertainty into statistical association tests using a fully conditional multiple imputation (MI) approach which is implemented using the Substantive Model Compatible Fully Conditional Specification (SMCFCS). We compared the performance of this method to an unconditional MI and two additional approaches that have been shown to demonstrate excellent performance: regression with dosages and a mixture of regression models (MRM). RESULTS: Our simulations considered a range of allele frequencies and imputation qualities based on data from the UK Biobank. We found that the unconditional MI was computationally costly and overly conservative across a wide range of settings. Analyzing data with Dosage, MRM, or MI SMCFCS resulted in greater power, including for low frequency variants, compared to unconditional MI while effectively controlling type I error rates. MRM andl MI SMCFCS are both more computationally intensive then using Dosage. CONCLUSIONS: The unconditional MI approach for association testing is overly conservative and we do not recommend its use in the context of imputed genotypes. Given its performance, speed, and ease of implementation, we recommend using Dosage for imputed genotypes with MAF [Formula: see text] 0.001 and Rsq [Formula: see text] 0.3.


Asunto(s)
Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Estudio de Asociación del Genoma Completo/métodos , Genotipo , Frecuencia de los Genes , Modelos Estadísticos
6.
Am J Hum Genet ; 106(2): 234-245, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31928709

RESUMEN

Germline pathogenic variants in chromatin-modifying enzymes are a common cause of pediatric developmental disorders. These enzymes catalyze reactions that regulate epigenetic inheritance via histone post-translational modifications and DNA methylation. Cytosine methylation (5-methylcytosine [5mC]) of DNA is the quintessential epigenetic mark, yet no human Mendelian disorder of DNA demethylation has yet been delineated. Here, we describe in detail a Mendelian disorder caused by the disruption of DNA demethylation. TET3 is a methylcytosine dioxygenase that initiates DNA demethylation during early zygote formation, embryogenesis, and neuronal differentiation and is intolerant to haploinsufficiency in mice and humans. We identify and characterize 11 cases of human TET3 deficiency in eight families with the common phenotypic features of intellectual disability and/or global developmental delay; hypotonia; autistic traits; movement disorders; growth abnormalities; and facial dysmorphism. Mono-allelic frameshift and nonsense variants in TET3 occur throughout the coding region. Mono-allelic and bi-allelic missense variants localize to conserved residues; all but one such variant occur within the catalytic domain, and most display hypomorphic function in an assay of catalytic activity. TET3 deficiency and other Mendelian disorders of the epigenetic machinery show substantial phenotypic overlap, including features of intellectual disability and abnormal growth, underscoring shared disease mechanisms.


Asunto(s)
Desmetilación del ADN , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Dioxigenasas/deficiencia , Adulto , Secuencia de Aminoácidos , Trastorno Autístico/genética , Trastorno Autístico/patología , Niño , Preescolar , Dioxigenasas/química , Dioxigenasas/genética , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/patología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Linaje , Conformación Proteica , Homología de Secuencia , Adulto Joven
7.
Clin Genet ; 104(4): 499-501, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37311648

RESUMEN

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.


Asunto(s)
Personas con Discapacidad , Pérdida Auditiva , Discapacidad Intelectual , Trastornos Motores , Humanos , Consanguinidad , Pérdida Auditiva/genética , Discapacidad Intelectual/genética , Proteínas de Microfilamentos/genética , Linaje , Fenotipo , Síndrome
8.
Clin Genet ; 103(6): 704-708, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36861389

RESUMEN

Exome sequencing of genes associated with heritable thoracic aortic disease (HTAD) failed to identify a pathogenic variant in a large family with Marfan syndrome (MFS). A genome-wide linkage analysis for thoracic aortic disease identified a peak at 15q21.1, and genome sequencing identified a novel deep intronic FBN1 variant that segregated with thoracic aortic disease in the family (LOD score 2.7) and was predicted to alter splicing. RT-PCR and bulk RNA sequencing of RNA harvested from fibroblasts explanted from the affected proband revealed an insertion of a pseudoexon between exons 13 and 14 of the FBN1 transcript, predicted to lead to nonsense mediated decay (NMD). Treating the fibroblasts with an NMD inhibitor, cycloheximide, greatly improved the detection of the pseudoexon-containing transcript. Family members with the FBN1 variant had later onset aortic events and fewer MFS systemic features than typical for individuals with haploinsufficiency of FBN1. Variable penetrance of the phenotype and negative genetic testing in MFS families should raise the possibility of deep intronic FBN1 variants and the need for additional molecular studies.


Asunto(s)
Enfermedades de la Aorta , Síndrome de Marfan , Humanos , Síndrome de Marfan/genética , Fibrilina-1/genética , Mutación , Fenotipo
9.
Clin Genet ; 103(6): 699-703, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36807241

RESUMEN

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.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Miopía , Distrofias Retinianas , Animales , Ratones , Humanos , Pérdida Auditiva Sensorineural/genética , Sordera/genética , Miopía/genética , Mutación , Linaje , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética
10.
J Med Genet ; 59(7): 669-677, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34321324

RESUMEN

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.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Ubiquitina-Proteína Ligasas , Genotipo , Humanos , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Hipotonía Muscular/patología , Trastornos del Neurodesarrollo/genética , Fenotipo , Convulsiones/genética , Ubiquitina-Proteína Ligasas/genética
11.
Ann Hum Genet ; 86(6): 291-296, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36039988

RESUMEN

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.


Asunto(s)
Ciliopatías , Síndromes Orofaciodigitales , Humanos , Síndromes Orofaciodigitales/genética , Ciliopatías/diagnóstico , Mutación , Homocigoto , Linaje
12.
Hum Genet ; 141(3-4): 413-430, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34291353

RESUMEN

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.


Asunto(s)
Sordera , Pérdida Auditiva , Consanguinidad , Sordera/genética , Genes Recesivos , Pérdida Auditiva/genética , Humanos , Linaje
13.
Am J Hum Genet ; 105(4): 822-835, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585107

RESUMEN

To analyze family-based whole-genome sequence (WGS) data for complex traits, we developed a rare variant (RV) non-parametric linkage (NPL) analysis method, which has advantages over association methods. The RV-NPL differs from the NPL in that RVs are analyzed, and allele sharing among affected relative-pairs is estimated only for minor alleles. Analyzing families can increase power because causal variants with familial aggregation usually have larger effect sizes than those underlying sporadic diseases. Differing from association analysis, for NPL only affected individuals are analyzed, which can increase power, since unaffected family members can be susceptibility variant carriers. RV-NPL is robust to population substructure and admixture, inclusion of nonpathogenic variants, as well as allelic and locus heterogeneity and can readily be applied outside of coding regions. In contrast to analyzing common variants using NPL, where loci localize to large genomic regions (e.g., >50 Mb), mapped regions are well defined for RV-NPL. Using simulation studies, we demonstrate that RV-NPL is substantially more powerful than applying traditional NPL methods to analyze RVs. The RV-NPL was applied to analyze 107 late-onset Alzheimer disease (LOAD) pedigrees of Caribbean Hispanic and European ancestry with WGS data, and statistically significant linkage (LOD ≥ 3.8) was found with RVs in PSMF1 and PTPN21 which have been shown to be involved in LOAD etiology. Additionally, nominally significant linkage was observed with RVs in ABCA7, ACE, EPHA1, and SORL1, genes that were previously reported to be associated with LOAD. RV-NPL is an ideal method to elucidate the genetic etiology of complex familial diseases.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Ligamiento Genético , Secuenciación Completa del Genoma , Femenino , Humanos , Masculino , Linaje
14.
J Med Genet ; 58(11): 743-751, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-32978268

RESUMEN

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.


Asunto(s)
Distrofina/genética , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Adolescente , Biopsia , Niño , Preescolar , Exones , Pruebas Genéticas/métodos , Humanos , Intrones , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Distrofias Musculares/diagnóstico por imagen , Adulto Joven
15.
J Med Genet ; 58(7): 442-452, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32709676

RESUMEN

BACKGROUND: Otitis media (OM) susceptibility has significant heritability; however, the role of rare variants in OM is mostly unknown. Our goal is to identify novel rare variants that confer OM susceptibility. METHODS: We performed exome and Sanger sequencing of >1000 DNA samples from 551 multiethnic families with OM and unrelated individuals, RNA-sequencing and microbiome sequencing and analyses of swabs from the outer ear, middle ear, nasopharynx and oral cavity. We also examined protein localisation and gene expression in infected and healthy middle ear tissues. RESULTS: A large, intermarried pedigree that includes 81 OM-affected and 53 unaffected individuals cosegregates two known rare A2ML1 variants, a common FUT2 variant and a rare, novel pathogenic variant c.1682A>G (p.Glu561Gly) within SPINK5 (LOD=4.09). Carriage of the SPINK5 missense variant resulted in increased relative abundance of Microbacteriaceae in the middle ear, along with occurrence of Microbacteriaceae in the outer ear and oral cavity but not the nasopharynx. Eight additional novel SPINK5 variants were identified in 12 families and individuals with OM. A role for SPINK5 in OM susceptibility is further supported by lower RNA counts in variant carriers, strong SPINK5 localisation in outer ear skin, faint localisation to middle ear mucosa and eardrum and increased SPINK5 expression in human cholesteatoma. CONCLUSION: SPINK5 variants confer susceptibility to non-syndromic OM. These variants potentially contribute to middle ear pathology through breakdown of mucosal and epithelial barriers, immunodeficiency such as poor vaccination response, alteration of head and neck microbiota and facilitation of entry of opportunistic pathogens into the middle ear.


Asunto(s)
Microbiota , Otitis Media/genética , Otitis Media/microbiología , Inhibidor de Serinpeptidasas Tipo Kazal-5/genética , Adulto , Animales , Bacterias/clasificación , Bacterias/genética , Niño , Susceptibilidad a Enfermedades/microbiología , Oído Externo/microbiología , Oído Medio/microbiología , Exoma , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Boca/microbiología , Nasofaringe/microbiología , Linaje , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
16.
Hum Genet ; 140(7): 1011-1029, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33710394

RESUMEN

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.


Asunto(s)
Exoma/genética , Discapacidad Intelectual/genética , Familia , Femenino , Finlandia , Genes Recesivos/genética , Predisposición Genética a la Enfermedad/genética , Genotipo , Homocigoto , Humanos , Masculino , Linaje , Secuenciación del Exoma/métodos
17.
Am J Hum Genet ; 102(4): 706-712, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29625025

RESUMEN

The major diseases affecting the thoracic aorta are aneurysms and acute dissections, and pathogenic variants in 11 genes are confirmed to lead to heritable thoracic aortic disease. However, many families in which multiple members have thoracic aortic disease do not have alterations in the known aortopathy genes. Genes highly expressed in the aorta were assessed for rare variants in exome sequencing data from such families, and compound rare heterozygous variants (p.Pro45Argfs∗25 and p.Glu750∗) in LTBP3 were identified in affected members of one family. A homozygous variant (p.Asn678_Gly681delinsThrCys) that introduces an additional cysteine into an epidermal growth factor (EGF)-like domain in the corresponding protein, latent TGF-ß binding protein (LTBP-3), was identified in a second family. Individuals with compound heterozygous or homozygous variants in these families have aneurysms and dissections of the thoracic aorta, as well as aneurysms of the abdominal aorta and other arteries, along with dental abnormalities and short stature. Heterozygous carriers of the p.Asn678_Gly681delinsThrCys variant have later onset of thoracic aortic disease, as well as dental abnormalities. In these families, LTBP3 variants segregated with thoracic aortic disease with a combined LOD score of 3.9. Additionally, heterozygous rare LTBP3 variants were found in individuals with early onset of acute aortic dissections, and some of these variants disrupted LTBP-3 levels or EGF-like domains. When compared to wild-type mice, Ltbp3-/- mice have enlarged aortic roots and ascending aortas. In summary, homozygous LTBP3 pathogenic variants predispose individuals to thoracic aortic aneurysms and dissections, along with the previously described skeletal and dental abnormalities.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Disección Aórtica/genética , Predisposición Genética a la Enfermedad , Proteínas de Unión a TGF-beta Latente/genética , Mutación/genética , Adulto , Anciano de 80 o más Años , Animales , Presión Sanguínea/genética , Femenino , Homocigoto , Humanos , Masculino , Ratones , Persona de Mediana Edad , Linaje
18.
Genet Med ; 23(11): 2122-2137, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34345025

RESUMEN

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.


Asunto(s)
Epilepsia , N-Metiltransferasa de Histona-Lisina , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Epilepsia/diagnóstico , Epilepsia/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Masculino , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/genética , Fenotipo , Convulsiones/diagnóstico , Convulsiones/genética
19.
J Hum Genet ; 66(12): 1169-1175, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34226616

RESUMEN

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.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/genética , Mutación , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Alelos , Secuencia de Aminoácidos , Femenino , Genotipo , Humanos , Masculino , Modelos Moleculares , Linaje , Fenotipo , Análisis de Secuencia de ADN , Miembro 2 de la Familia de Transportadores de Soluto 12/química , Relación Estructura-Actividad , Secuenciación del Exoma
20.
J Hum Genet ; 66(10): 1009-1018, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33879837

RESUMEN

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.


Asunto(s)
Proteína Quinasa CDC2/genética , Sordera/genética , Predisposición Genética a la Enfermedad , Atrofia Óptica/genética , Síndrome de Wolfram/genética , Adolescente , Adulto , Enfermedad de la Válvula Aórtica Bicúspide/genética , Enfermedad de la Válvula Aórtica Bicúspide/patología , Niño , Preescolar , Consanguinidad , Sordera/complicaciones , Sordera/patología , Diabetes Mellitus/genética , Femenino , Tracto Gastrointestinal/anomalías , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Pérdida Auditiva , Homocigoto , Humanos , Lactante , Masculino , Mutación Missense/genética , Atrofia Óptica/complicaciones , Atrofia Óptica/patología , Síndrome de Wolfram/complicaciones , Síndrome de Wolfram/epidemiología , Síndrome de Wolfram/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA