Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Hum Genet ; 143(3): 311-329, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38459354

RESUMEN

Identification of genes associated with nonsyndromic hearing loss is a crucial endeavor given the substantial number of individuals who remain without a diagnosis after even the most advanced genetic testing. PKHD1L1 was established as necessary for the formation of the cochlear hair-cell stereociliary coat and causes hearing loss in mice and zebrafish when mutated. We sought to determine if biallelic variants in PKHD1L1 also cause hearing loss in humans. Exome sequencing was performed on DNA of four families segregating autosomal recessive nonsyndromic sensorineural hearing loss. Compound heterozygous p.[(Gly129Ser)];p.[(Gly1314Val)] and p.[(Gly605Arg)];p[(Leu2818TyrfsTer5)], homozygous missense p.(His2479Gln) and nonsense p.(Arg3381Ter) variants were identified in PKHD1L1 that were predicted to be damaging using in silico pathogenicity prediction methods. In vitro functional analysis of two missense variants was performed using purified recombinant PKHD1L1 protein fragments. We then evaluated protein thermodynamic stability with and without the missense variants found in one of the families and performed a minigene splicing assay for another variant. In silico molecular modeling using AlphaFold2 and protein sequence alignment analysis were carried out to further explore potential variant effects on structure. In vitro functional assessment indicated that both engineered PKHD1L1 p.(Gly129Ser) and p.(Gly1314Val) mutant constructs significantly reduced the folding and structural stabilities of the expressed protein fragments, providing further evidence to support pathogenicity of these variants. Minigene assay of the c.1813G>A p.(Gly605Arg) variant, located at the boundary of exon 17, revealed exon skipping leading to an in-frame deletion of 48 amino acids. In silico molecular modeling exposed key structural features that might suggest PKHD1L1 protein destabilization. Multiple lines of evidence collectively associate PKHD1L1 with nonsyndromic mild-moderate to severe sensorineural hearing loss. PKHD1L1 testing in individuals with mild-moderate hearing loss may identify further affected families.


Asunto(s)
Sordera , Mutación Missense , Linaje , Receptores de Superficie Celular , Estereocilios , Animales , Femenino , Humanos , Masculino , Sordera/genética , Secuenciación del Exoma , Genes Recesivos , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Modelos Moleculares , Receptores de Superficie Celular/genética , Estereocilios/metabolismo , Estereocilios/patología , Estereocilios/genética
2.
Audiol Neurootol ; 28(6): 407-419, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37331337

RESUMEN

BACKGROUND: Mutations in TMPRSS3 are an important cause of autosomal recessive non-syndromic hearing loss. The hearing loss associated with mutations in TMPRSS3 is characterized by phenotypic heterogeneity, ranging from mild to profound hearing loss, and is generally progressive. Clinical presentation and natural history of TMPRSS3 mutations vary significantly based on the location and type of mutation in the gene. Understanding these genotype-phenotype relationships and associated natural disease histories is necessary for the successful development and application of gene-based therapies and precision medicine approaches to DFNB8/10. The heterogeneous presentation of TMPRSS3-associated disease makes it difficult to identify patients clinically. As the body of literature on TMPRSS3-associated deafness grows, there is need for better categorization of the hearing phenotypes associated with specific mutations in the gene. SUMMARY: In this review, we summarize TMPRSS3 genotype-phenotype relationships including a thorough description of the natural history of patients with TMPRSS3-associated hearing loss to lay the groundwork for the future of TMPRSS3 treatment using molecular therapy. KEY MESSAGES: TMPRSS3 mutation is a significant cause of genetic hearing loss. All patients with TMPRSS3 mutation display severe-to-profound prelingual (DFNB10) or a postlingual (DFNB8) progressive sensorineural hearing loss. Importantly, TMPRSS3 mutations have not been associated with middle ear or vestibular deficits. The c.916G>A (p.Ala306Thr) missense mutation is the most frequently reported mutation across populations and should be further explored as a target for molecular therapy.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Humanos , Serina Endopeptidasas/genética , Proteínas de la Membrana/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva/genética , Mutación , Estudios de Asociación Genética , Fenotipo , Proteínas de Neoplasias/genética
3.
Am J Hum Genet ; 103(4): 484-497, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30245029

RESUMEN

The classification of genetic variants represents a major challenge in the post-genome era by virtue of their extraordinary number and the complexities associated with ascribing a clinical impact, especially for disorders exhibiting exceptional phenotypic, genetic, and allelic heterogeneity. To address this challenge for hearing loss, we have developed the Deafness Variation Database (DVD), a comprehensive, open-access resource that integrates all available genetic, genomic, and clinical data together with expert curation to generate a single classification for each variant in 152 genes implicated in syndromic and non-syndromic deafness. We evaluate 876,139 variants and classify them as pathogenic or likely pathogenic (more than 8,100 variants), benign or likely benign (more than 172,000 variants), or of uncertain significance (more than 695,000 variants); 1,270 variants are re-categorized based on expert curation and in 300 instances, the change is of medical significance and impacts clinical care. We show that more than 96% of coding variants are rare and novel and that pathogenicity is driven by minor allele frequency thresholds, variant effect, and protein domain. The mutational landscape we define shows complex gene-specific variability, making an understanding of these nuances foundational for improved accuracy in variant interpretation in order to enhance clinical decision making and improve our understanding of deafness biology.


Asunto(s)
Sordera/genética , Mutación/genética , Bases de Datos Genéticas , Frecuencia de los Genes/genética , Genómica/métodos , Pérdida Auditiva/genética , Humanos
4.
Genet Med ; 21(11): 2614-2630, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31171844

RESUMEN

Early intervention for newborns who are deaf or hard-of-hearing leads to improved language, communication, and social-emotional outcomes. Universal physiologic newborn hearing screening has been widely implemented across the United States with the goal of identifying newborns who are deaf or hard-of-hearing, thereby reducing time to diagnosis and intervention. The current physiologic newborn hearing screen is generally successful in accomplishing its goals but improvements could be made. In the past ten years, genetic testing has emerged as the most important etiological diagnostic test for evaluation of children with deafness and congenital cytomegalovirus has been recognized as a major cause of childhood deafness that may be treatable. A comprehensive newborn hearing screen that includes physiologic, genetic, and cytomegalovirus testing would have multiple benefits, including (1) identifying newborns with deafness missed by the current physiologic screen, (2) providing etiologic information, and (3) possibly decreasing the number of children lost to follow up. We present a framework for integrating limited genetic testing and cytomegalovirus screening into the current physiologic newborn hearing screening. We identify needed areas of research and include an overview of genome sequencing, which we believe will become available over the next decade as a complement to universal physiologic newborn hearing screening.


Asunto(s)
Sordera/diagnóstico , Pruebas Auditivas/métodos , Tamizaje Neonatal/métodos , Sordera/genética , Femenino , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/genética , Humanos , Lactante , Recién Nacido , Masculino , Estados Unidos
5.
Genet Med ; 21(12): 2845-2846, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31213665

RESUMEN

In the original version of this Article, several individuals were erroneously acknowledged in the acknowledgements, they have been removed. The Acknowledgement section in the PDF and HTML versions of the Article has now been corrected to the following.

6.
PLoS Genet ; 11(3): e1005137, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25816005

RESUMEN

Hereditary hearing loss is a clinically and genetically heterogeneous disorder. More than 80 genes have been implicated to date, and with the advent of targeted genomic enrichment and massively parallel sequencing (TGE+MPS) the rate of novel deafness-gene identification has accelerated. Here we report a family segregating post-lingual progressive autosomal dominant non-syndromic hearing loss (ADNSHL). After first excluding plausible variants in known deafness-causing genes using TGE+MPS, we completed whole exome sequencing in three hearing-impaired family members. Only a single variant, p.Arg185Pro in HOMER2, segregated with the hearing-loss phenotype in the extended family. This amino acid change alters a highly conserved residue in the coiled-coil domain of HOMER2 that is essential for protein multimerization and the HOMER2-CDC42 interaction. As a scaffolding protein, HOMER2 is involved in intracellular calcium homeostasis and cytoskeletal organization. Consistent with this function, we found robust expression in stereocilia of hair cells in the murine inner ear and observed that over-expression of mutant p.Pro185 HOMER2 mRNA causes anatomical changes of the inner ear and neuromasts in zebrafish embryos. Furthermore, mouse mutants homozygous for the targeted deletion of Homer2 present with early-onset rapidly progressive hearing loss. These data provide compelling evidence that HOMER2 is required for normal hearing and that its sequence alteration in humans leads to ADNSHL through a dominant-negative mode of action.


Asunto(s)
Proteínas Portadoras/genética , Oído Interno/metabolismo , Exoma/genética , Pérdida Auditiva Sensorineural/genética , Animales , Proteínas Portadoras/biosíntesis , Cóclea/metabolismo , Cóclea/patología , Oído Interno/patología , Regulación de la Expresión Génica , Pérdida Auditiva Sensorineural/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Andamiaje Homer , Humanos , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Estereocilios/genética , Estereocilios/patología , Pez Cebra , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
7.
Hum Mol Genet ; 24(16): 4483-90, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25954030

RESUMEN

We studied a consanguineous Palestinian Arab family segregating an autosomal recessive progressive myoclonus epilepsy (PME) with early ataxia. PME is a rare, often fatal syndrome, initially responsive to antiepileptic drugs which over time becomes refractory and can be associated with cognitive decline. Linkage analysis was performed and the disease locus narrowed to chromosome 19p13.3. Fourteen candidate genes were screened by conventional Sanger sequencing and in one, LMNB2, a novel homozygous missense mutation was identified that segregated with the PME in the family. Whole exome sequencing excluded other likely pathogenic coding variants in the linked interval. The p.His157Tyr mutation is located in an evolutionarily highly conserved region of the alpha-helical rod of the lamin B2 protein. In vitro assembly analysis of mutant lamin B2 protein revealed a distinct defect in the assembly of the highly ordered fibrous arrays typically formed by wild-type lamin B2. Our data suggests that disruption of the organisation of the nuclear lamina in neurons, perhaps through abnormal neuronal migration, causes the epilepsy and early ataxia syndrome and extends the aetiology of PMEs to include dysfunction in nuclear lamin proteins.


Asunto(s)
Ataxia/genética , Cromosomas Humanos Par 19/genética , Epilepsias Mioclónicas/genética , Lamina Tipo B/genética , Mutación Missense , Sustitución de Aminoácidos , Niño , Familia , Femenino , Humanos , Masculino
9.
Am J Hum Genet ; 95(4): 445-53, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25262649

RESUMEN

Ethnic-specific differences in minor allele frequency impact variant categorization for genetic screening of nonsyndromic hearing loss (NSHL) and other genetic disorders. We sought to evaluate all previously reported pathogenic NSHL variants in the context of a large number of controls from ethnically distinct populations sequenced with orthogonal massively parallel sequencing methods. We used HGMD, ClinVar, and dbSNP to generate a comprehensive list of reported pathogenic NSHL variants and re-evaluated these variants in the context of 8,595 individuals from 12 populations and 6 ethnically distinct major human evolutionary phylogenetic groups from three sources (Exome Variant Server, 1000 Genomes project, and a control set of individuals created for this study, the OtoDB). Of the 2,197 reported pathogenic deafness variants, 325 (14.8%) were present in at least one of the 8,595 controls, indicating a minor allele frequency (MAF) > 0.00006. MAFs ranged as high as 0.72, a level incompatible with pathogenicity for a fully penetrant disease like NSHL. Based on these data, we established MAF thresholds of 0.005 for autosomal-recessive variants (excluding specific variants in GJB2) and 0.0005 for autosomal-dominant variants. Using these thresholds, we recategorized 93 (4.2%) of reported pathogenic variants as benign. Our data show that evaluation of reported pathogenic deafness variants using variant MAFs from multiple distinct ethnicities and sequenced by orthogonal methods provides a powerful filter for determining pathogenicity. The proposed MAF thresholds will facilitate clinical interpretation of variants identified in genetic testing for NSHL. All data are publicly available to facilitate interpretation of genetic variants causing deafness.


Asunto(s)
Etnicidad/genética , Evolución Molecular , Exoma/genética , Variación Genética/genética , Pérdida Auditiva/genética , Pérdida Auditiva/patología , Estudios de Casos y Controles , Conexina 26 , Conexinas , Frecuencia de los Genes , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Humanos , Filogenia
10.
Hum Genet ; 135(4): 441-450, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26969326

RESUMEN

Hearing loss is the most common sensory deficit in humans, affecting 1 in 500 newborns. Due to its genetic heterogeneity, comprehensive diagnostic testing has not previously been completed in a large multiethnic cohort. To determine the aggregate contribution inheritance makes to non-syndromic hearing loss, we performed comprehensive clinical genetic testing with targeted genomic enrichment and massively parallel sequencing on 1119 sequentially accrued patients. No patient was excluded based on phenotype, inheritance or previous testing. Testing resulted in identification of the underlying genetic cause for hearing loss in 440 patients (39%). Pathogenic variants were found in 49 genes and included missense variants (49%), large copy number changes (18%), small insertions and deletions (18%), nonsense variants (8%), splice-site alterations (6%), and promoter variants (<1%). The diagnostic rate varied considerably based on phenotype and was highest for patients with a positive family history of hearing loss or when the loss was congenital and symmetric. The spectrum of implicated genes showed wide ethnic variability. These findings support the more efficient utilization of medical resources through the development of evidence-based algorithms for the diagnosis of hearing loss.


Asunto(s)
Pruebas Genéticas , Pérdida Auditiva/genética , Adolescente , Niño , Preescolar , Femenino , Heterogeneidad Genética , Pérdida Auditiva/diagnóstico , Humanos , Lactante , Masculino
11.
Bioinformatics ; 30(23): 3438-9, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25123904

RESUMEN

UNLABELLED: Cordova is an out-of-the-box solution for building and maintaining an online database of genetic variations integrated with pathogenicity prediction results from popular algorithms. Our primary motivation for developing this system is to aid researchers and clinician-scientists in determining the clinical significance of genetic variations. To achieve this goal, Cordova provides an interface to review and manually or computationally curate genetic variation data as well as share it for clinical diagnostics and the advancement of research. AVAILABILITY AND IMPLEMENTATION: Cordova is open source under the MIT license and is freely available for download at https://github.com/clcg/cordova.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Variación Genética , Algoritmos , Humanos , Internet , Programas Informáticos
12.
Ann Otol Rhinol Laryngol ; 124 Suppl 1: 177S-83S, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25788561

RESUMEN

OBJECTIVES: We present a family with a mitochondrial DNA 3243A>G mutation resulting in mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS), of which some members have hearing loss in which a novel mutation in the P2RX2 gene was identified. METHODS: One hundred ninety-four (194) Japanese subjects from unrelated families were enrolled in the study. Targeted genomic enrichment and massively parallel sequencing of all known nonsyndromic hearing loss genes were performed to identify the genetic causes of hearing loss. RESULTS: A novel mutation in the P2RX2 gene that corresponded to c.601G>A (p.Asp201Tyr) was identified. Two patients carried the mutation and had severe sensorineural hearing loss, while other members with MELAS (who did not carry the P2RX2 mutation) had normal hearing. CONCLUSION: This is the first case report of a diagnosis of hearing loss caused by P2RX2 mutation in patients with MELAS. A potential explanation is that a decrease in adenosine triphosphate (ATP) production due to MELAS with a mitochondrial 3243A>G mutation might suppress activation of P2X2 receptors. We also suggest that hearing loss caused by the P2RX2 mutation might be influenced by the decrease in ATP production due to MELAS.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Síndrome MELAS/genética , Mitocondrias/genética , Receptores Purinérgicos P2X2/genética , Adenosina Trifosfato/metabolismo , Sordera/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndrome MELAS/metabolismo , Persona de Mediana Edad , Linaje , Análisis de Secuencia de ADN/métodos
13.
Ann Otol Rhinol Laryngol ; 124 Suppl 1: 184S-92S, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25788564

RESUMEN

OBJECTIVES: We present 3 patients with congenital sensorineural hearing loss (SNHL) caused by novel PTPRQ mutations, including clinical manifestations and phenotypic features. METHODS: Two hundred twenty (220) Japanese subjects with SNHL from unrelated and nonconsanguineous families were enrolled in the study. Targeted genomic enrichment with massively parallel DNA sequencing of all known nonsyndromic hearing loss genes was performed to identify the genetic cause of hearing loss. RESULTS: Four novel causative PTPRQ mutations were identified in 3 cases. Case 1 had progressive profound SNHL with a homozygous nonsense mutation. Case 2 had nonprogressive profound SNHL with a compound heterozygous mutation (nonsense and missense mutation). Case 3 had nonprogressive moderate SNHL with a compound heterozygous mutation (missense and splice site mutation). Caloric test and vestibular evoked myogenic potential (VEMP) test showed vestibular dysfunction in Case 1. CONCLUSION: Hearing loss levels and progression among the present cases were varied, and there seem to be no obvious correlations between genotypes and the phenotypic features of their hearing loss. The PTPRQ mutations appeared to be responsible for vestibular dysfunction.


Asunto(s)
Codón sin Sentido , Pérdida Auditiva Sensorineural/genética , Mutación Missense , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Pueblo Asiatico/genética , Audiometría de Tonos Puros , Análisis Mutacional de ADN/métodos , Sordera/genética , Potenciales Evocados Auditivos , Pérdida Auditiva Sensorineural/congénito , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Linaje
14.
Ann Otol Rhinol Laryngol ; 124 Suppl 1: 169S-76S, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25792666

RESUMEN

OBJECTIVES: In this report, we present a male patient with no family history of hearing loss, in whom we identified a novel de novo mutation in the POU3F4 gene. METHODS: One hundred ninety-four (194) Japanese subjects from unrelated and nonconsanguineous families were enrolled in this study. We used targeted genomic enrichment and massively parallel sequencing of all known nonsyndromic hearing loss genes for identifying the genetic causes of hearing loss. RESULTS: A novel de novo frameshift mutation of POU3F4 to c.727_728insA (p.N244KfsX26) was identified. The patient was a 7-year-old male with congenital progressive hearing loss and inner ear deformity. Although the patient had received a cochlear implant, auditory skills were still limited. The patient also exhibited developmental delays similar to those previously associated with POU3F4 mutation. CONCLUSION: This is the first report of a mutation in POU3F4 causing hearing loss in a Japanese patient without a family history of hearing loss. This study underscores the importance of comprehensive genetic testing of patients with hearing loss for providing accurate prognostic information and guiding the optimal management of patient rehabilitation.


Asunto(s)
Mutación del Sistema de Lectura , Factores del Dominio POU/genética , Pueblo Asiatico/genética , Niño , Análisis Mutacional de ADN , Sordera/genética , Discapacidades del Desarrollo/genética , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino
15.
Ann Otol Rhinol Laryngol ; 124 Suppl 1: 123S-8S, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25743181

RESUMEN

OBJECTIVE: We present 2 patients who were identified with mutations in the GPR98 gene that causes Usher syndrome type 2 (USH2). METHODS: One hundred ninety-four (194) Japanese subjects from unrelated families were enrolled in the study. Targeted genomic enrichment and massively parallel sequencing of all known nonsyndromic hearing loss genes were used to identify the genetic causes of hearing loss. RESULTS: We identified causative mutations in the GPR98 gene in 1 family (2 siblings). The patients had moderate sloping hearing loss, and no progression was observed over a period of 10 years. Fundus examinations were normal. However, electroretinograms revealed impaired responses in both patients. CONCLUSION: Early diagnosis of Usher syndrome has many advantages for patients and their families. This study supports the use of comprehensive genetic diagnosis for Usher syndrome, especially prior to the onset of visual symptoms, to provide the highest chance of diagnostic success in early life stages.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Receptores Acoplados a Proteínas G/genética , Síndromes de Usher/genética , Adolescente , Pueblo Asiatico/genética , Electrorretinografía , Femenino , Humanos , Mutación , Análisis de Secuencia de ADN/métodos
16.
Hum Mutat ; 35(7): 819-23, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24729539

RESUMEN

Hereditary hearing loss is extremely heterogeneous. Over 70 genes have been identified to date, and with the advent of massively parallel sequencing, the pace of novel gene discovery has accelerated. In a family segregating progressive autosomal-dominant nonsyndromic hearing loss (NSHL), we used OtoSCOPE® to exclude mutations in known deafness genes and then performed segregation mapping and whole-exome sequencing to identify a unique variant, p.Ser178Leu, in TBC1D24 that segregates with the hearing loss phenotype. TBC1D24 encodes a GTPase-activating protein expressed in the cochlea. Ser178 is highly conserved across vertebrates and its change is predicted to be damaging. Other variants in TBC1D24 have been associated with a panoply of clinical symptoms including autosomal recessive NSHL, syndromic hearing impairment associated with onychodystrophy, osteodystrophy, mental retardation, and seizures (DOORS syndrome), and a wide range of epileptic disorders.


Asunto(s)
Proteínas Portadoras/genética , Genes Dominantes , Mutación , Secuencia de Aminoácidos , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Análisis Mutacional de ADN , Sordera/genética , Exoma , Femenino , Proteínas Activadoras de GTPasa , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Proteínas de la Membrana , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso , Linaje , Alineación de Secuencia
17.
Am J Hum Genet ; 88(2): 127-37, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21255762

RESUMEN

By using homozygosity mapping in a consanguineous Pakistani family, we detected linkage of nonsyndromic hearing loss to a 7.6 Mb region on chromosome 3q13.31-q21.1 within the previously reported DFNB42 locus. Subsequent candidate gene sequencing identified a homozygous nonsense mutation (c.1135G>T [p.Glu379X]) in ILDR1 as the cause of hearing impairment. By analyzing additional consanguineous families with homozygosity at this locus, we detected ILDR1 mutations in the affected individuals of 10 more families from Pakistan and Iran. The identified ILDR1 variants include missense, nonsense, frameshift, and splice-site mutations as well as a start codon mutation in the family that originally defined the DFNB42 locus. ILDR1 encodes the evolutionarily conserved immunoglobulin-like domain containing receptor 1, a putative transmembrane receptor of unknown function. In situ hybridization detected expression of Ildr1, the murine ortholog, early in development in the vestibule and in hair cells and supporting cells of the cochlea. Expression in hair cell- and supporting cell-containing neurosensory organs is conserved in the zebrafish, in which the ildr1 ortholog is prominently expressed in the developing ear and neuromasts of the lateral line. These data identify loss-of-function mutations of ILDR1, a gene with a conserved expression pattern pointing to a conserved function in hearing in vertebrates, as underlying nonsyndromic prelingual sensorineural hearing impairment.


Asunto(s)
Codón sin Sentido/genética , Genes Recesivos/genética , Predisposición Genética a la Enfermedad , Pérdida Auditiva/genética , Receptores de Superficie Celular/genética , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 3/genética , Consanguinidad , Oído Interno , Femenino , Ligamiento Genético , Genotipo , Humanos , Hibridación in Situ , Escala de Lod , Masculino , Ratones , Linaje , Pez Cebra
18.
J Med Genet ; 50(9): 627-34, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804846

RESUMEN

BACKGROUND: Non-syndromic hearing loss (NSHL) is the most common sensory impairment in humans. Until recently its extreme genetic heterogeneity precluded comprehensive genetic testing. Using a platform that couples targeted genomic enrichment (TGE) and massively parallel sequencing (MPS) to sequence all exons of all genes implicated in NSHL, we tested 100 persons with presumed genetic NSHL and in so doing established sequencing requirements for maximum sensitivity and defined MPS quality score metrics that obviate Sanger validation of variants. METHODS: We examined DNA from 100 sequentially collected probands with presumed genetic NSHL without exclusions due to inheritance, previous genetic testing, or type of hearing loss. We performed TGE using post-capture multiplexing in variable pool sizes followed by Illumina sequencing. We developed a local Galaxy installation on a high performance computing cluster for bioinformatics analysis. RESULTS: To obtain maximum variant sensitivity with this platform 3.2-6.3 million total mapped sequencing reads per sample were required. Quality score analysis showed that Sanger validation was not required for 95% of variants. Our overall diagnostic rate was 42%, but this varied by clinical features from 0% for persons with asymmetric hearing loss to 56% for persons with bilateral autosomal recessive NSHL. CONCLUSIONS: These findings will direct the use of TGE and MPS strategies for genetic diagnosis for NSHL. Our diagnostic rate highlights the need for further research on genetic deafness focused on novel gene identification and an improved understanding of the role of non-exonic mutations. The unsolved families we have identified provide a valuable resource to address these areas.


Asunto(s)
Sordera/genética , Pruebas Genéticas/métodos , Genómica/métodos , Adolescente , Adulto , Femenino , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN
19.
Proc Natl Acad Sci U S A ; 108(10): 4218-23, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368133

RESUMEN

We report on a secreted protein found in mammalian cochlear outer hair cells (OHC) that is a member of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family of adhesion proteins. Ceacam16 mRNA is expressed in OHC, and its protein product localizes to the tips of the tallest stereocilia and the tectorial membrane (TM). This specific localization suggests a role in maintaining the integrity of the TM as well as in the connection between the OHC stereocilia and TM, a linkage essential for mechanical amplification. In agreement with this role, CEACAM16 colocalizes and coimmunoprecipitates with the TM protein α-tectorin. In addition, we show that mutation of CEACAM16 leads to autosomal dominant nonsyndromic deafness (ADNSHL) at the autosomal dominant hearing loss (DFNA4) locus. In aggregate, these data identify CEACAM16 as an α-tectorin-interacting protein that concentrates at the point of attachment of the TM to the stereocilia and, when mutated, results in ADNSHL at the DFNA4 locus.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Genes Dominantes , Pérdida Auditiva/metabolismo , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Secuencia de Aminoácidos , Animales , Western Blotting , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Proteínas Ligadas a GPI/metabolismo , Pérdida Auditiva/genética , Humanos , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética
20.
Int J Pediatr Otorhinolaryngol ; 182: 111999, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38850598

RESUMEN

OBJECTIVE: Atraumatic cerebrospinal fluid (CSF) rhinorrhea is uncommon in children and necessitates a multi-disciplinary evaluation for an etiology. Underlying osseous abnormality due to extensive or multifocal low flow vascular anomaly should be considered as a potential cause of spontaneous CSF leak. Treatment of multifocal low flow vascular anomalies may include medical and surgical approaches. In this series, we seek to determine the presenting signs and symptoms and medical and surgical treatment options for multifocal or extensive low flow vascular anomalies. METHODS: A retrospective case series at a quaternary care children's hospital was compiled. All children with CSF rhinorrhea diagnosed and treated for multifocal low flow vascular anomalies at our institution were included. A total of four patients were identified. RESULTS: All four patients had delay in initial diagnosis of underlying cause of meningitis and CSF rhinorrhea. Average age at diagnosis of multifocal low flow vascular anomaly was 7 years. This was on average 4 years after initial presentation for medical attention. Treatment approach was multidisciplinary and included medical management with sirolimus and bisphosphonates as well as surgical approaches to the skull base (lateral and anterior) to prevent CSF egress. CONCLUSION: Consideration of multifocal low flow vascular anomaly should be included in any pediatric patient presenting with CSF rhinorrhea.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda