Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ann Hum Genet ; 86(1): 1-13, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34374074

RESUMO

The study was conducted between 2018 and 2020. From a cohort of 113 hearing impaired (HI), five non-DFNB12 probands identified with heterozygous CDH23 variants were subjected to exome analysis. This resolved the etiology of hearing loss (HL) in four South Indian assortative mating families. Six variants, including three novel ones, were identified in four genes: PNPT1 p.(Ala46Gly) and p.(Asn540Ser), MYO15A p.(Leu1485Pro) and p.(Tyr1891Ter), PTPRQ p.(Gln1336Ter), and SLC12A2 p.(Pro988Ser). Compound heterozygous PNPT1 variants were associated with DFNB70 causing prelingual profound sensorineural hearing loss (SNHL), vestibular dysfunction, and unilateral progressive vision loss in one family. In the second family, MYO15A variants in the myosin motor domain, including a novel variant, causing DFNB3, were found to be associated with prelingual profound SNHL. A novel PTPRQ variant was associated with postlingual progressive sensorineural/mixed HL and vestibular dysfunction in the third family with DFNB84A. In the fourth family, the SLC12A2 novel variant was found to segregate with severe-to-profound HL causing DFNA78, across three generations. Our results suggest a high level of allelic, genotypic, and phenotypic heterogeneity of HL in these families. This study is the first to report the association of PNPT1, PTPRQ, and SLC12A2 variants with HL in the Indian population.


Assuntos
Perda Auditiva Neurossensorial , Perda Auditiva , Exorribonucleases/genética , Audição , Perda Auditiva Neurossensorial/genética , Humanos , Índia , Mutação , Miosinas/genética , Linhagem , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Membro 2 da Família 12 de Carreador de Soluto/genética
2.
Otol Neurotol ; 42(8): e1143-e1151, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34049328

RESUMO

OBJECTIVES: To better distinguish NOG-related-symphalangism spectrum disorder (NOG-SSD) from chromosomal 17q22 microdeletion syndromes and to inform surgical considerations in stapes surgery for patients with NOG-SSD. BACKGROUND: Mutations in NOG cause a variety of skeletal syndromes that often include conductive hearing loss. Several microdeletions of chromosome 17q22 lead to severe syndromes with clinical characteristics that overlap NOG-SSD. Isolated deletion of NOG has not been described, and therefore the contribution of NOG deletion in these syndromes is unknown. METHODS: Two families with autosomal dominant NOG-SSD exhibited stapes ankylosis, facial dysmorphisms, and skeletal and joint anomalies. In each family, NOG was evaluated by genomic sequencing and candidate mutations confirmed as damaging by in vitro assays. Temporal bone histology of a patient with NOG-SSD was compared with temporal bones of 40 patients diagnosed with otosclerosis. RESULTS: Family 1 harbors a 555 kb chromosomal deletion encompassing only NOG and ANKFN1. Family 2 harbors a missense mutation in NOG leading to absence of noggin protein. The incus-footplate distance of the temporal bone was significantly longer in a patient with NOG-SSD than in patients with otosclerosis. CONCLUSION: The chromosomal microdeletion of family 1 led to a phenotype comparable to that due to a NOG point mutation and much milder than the phenotypes due to other chromosome 17q22 microdeletions. Severe clinical findings in other microdeletion cases are likely due to deletion of genes other than NOG. Based on temporal bone findings, we recommend that surgeons obtain longer stapes prostheses before stapes surgery in individuals with NOG-SSD stapes ankylosis.


Assuntos
Deformidades Congênitas do Pé , Deformidades Congênitas da Mão , Sinostose , Ossos do Carpo/anormalidades , Heterogeneidade Genética , Humanos , Estribo/anormalidades , Sinostose/genética , Ossos do Tarso/anormalidades
3.
Eur J Hum Genet ; 29(6): 988-997, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33398081

RESUMO

Nonsyndromic hearing loss is genetically heterogeneous. Despite comprehensive genetic testing, many cases remain unsolved because the clinical significance of identified variants is uncertain or because biallelic pathogenic variants are not identified for presumed autosomal recessive cases. Common synonymous variants are often disregarded. Determining the pathogenicity of synonymous variants may improve genetic diagnosis. We report a synonymous variant c.9861 C > T/p.(Gly3287=) in MYO15A in homozygosity or compound heterozygosity with another pathogenic or likely pathogenic MYO15A variant in 10 unrelated families with nonsyndromic sensorineural hearing loss. Biallelic variants in MYO15A were identified in 21 affected and were absent in 22 unaffected siblings. A mini-gene assay confirms that the synonymous variant leads to abnormal splicing. The variant is enriched in the Ashkenazi Jewish population. Individuals carrying biallelic variants involving c.9861 C > T often exhibit progressive post-lingual hearing loss distinct from the congenital profound deafness typically associated with biallelic loss-of-function MYO15A variants. This study establishes the pathogenicity of the c.9861 C > T variant in MYO15A and expands the phenotypic spectrum of MYO15A-related hearing loss. Our work also highlights the importance of multicenter collaboration and data sharing to establish the pathogenicity of a relatively common synonymous variant for improved diagnosis and management of hearing loss.


Assuntos
Frequência do Gene , Perda Auditiva/genética , Miosinas/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Genes Recessivos , Perda Auditiva/etnologia , Perda Auditiva/patologia , Humanos , Lactente , Judeus/genética , Masculino , Mutação , Linhagem , Splicing de RNA
4.
EMBO Mol Med ; 13(2): e13259, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33350593

RESUMO

Genetic variants account for approximately half the cases of congenital and early-onset deafness. Methods and technologies for viral delivery of genes into the inner ear have evolved over the past decade to render gene therapy a viable and attractive approach for treatment. Variants in SYNE4, encoding the protein nesprin-4, a member of the linker of nucleoskeleton and cytoskeleton (LINC), lead to DFNB76 human deafness. Syne4-/- mice have severe-to-profound progressive hearing loss and exhibit mislocalization of hair cell nuclei and hair cell degeneration. We used AAV9-PHP.B, a recently developed synthetic adeno-associated virus, to deliver the coding sequence of Syne4 into the inner ears of neonatal Syne4-/- mice. Here we report rescue of hair cell morphology and survival, nearly complete recovery of auditory function, and restoration of auditory-associated behaviors, without observed adverse effects. Uncertainties remain regarding the durability of the treatment and the time window for intervention in humans, but our results suggest that gene therapy has the potential to prevent hearing loss in humans with SYNE4 mutations.


Assuntos
Surdez , Perda Auditiva , Animais , Surdez/genética , Surdez/terapia , Dependovirus/genética , Terapia Genética , Audição/genética , Perda Auditiva/genética , Perda Auditiva/terapia , Camundongos
5.
FASEB J ; 33(4): 4729-4740, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30592649

RESUMO

The adherens junctions (AJs) and tight junctions (TJs) provide critical adhesive contacts between neighboring epithelial cells and are crucial for epithelial adhesion, integrity, and barrier functions in a wide variety of tissues and organisms. The striatin protein family, which are part of the striatin interaction phosphatases and kinases complex, are multidomain scaffolding proteins that play important biologic roles. We have previously shown that striatin colocalizes with the tumor suppressor protein adenomatous polyposis coli in the TJs of epithelial cells. Here we show that striatin affects junction integrity and cell migration, probably through a mechanism that involves the adhesion molecule E-cadherin. Cells engaged in cell-cell adhesion expressed a high MW-modified form of striatin that forms stable associations with detergent-insoluble, membrane-bound cellular fractions. In addition, striatin has recently been suggested to be a target of the poly (ADP-ribose) polymerases Tankyrase 1, and we have found that striatin interacts with Tankyrase 1 and is subsequently poly-ADP-ribosylated. Taken together, our results suggest that striatin is a novel cell-cell junctional protein that functions to maintain correct cell adhesion and may have a role in establishing the relationship between AJs and TJs that is fundamental for epithelial cell-cell adhesion.-Lahav-Ariel, L., Caspi, M., Nadar-Ponniah, P. T., Zelikson, N., Hofmann, I., Hanson, K. K., Franke, W. W., Sklan, E. H., Avraham, K. B., Rosin-Arbesfeld, R. Striatin is a novel modulator of cell adhesion.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Adesão Celular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Junções Aderentes/metabolismo , Animais , Western Blotting , Células COS , Células CACO-2 , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ligação a Calmodulina/genética , Adesão Celular/genética , Chlorocebus aethiops , Cães , Células Hep G2 , Humanos , Imunoprecipitação , Células MCF-7 , Células Madin Darby de Rim Canino , Proteínas de Membrana/genética , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/genética , Tanquirases/metabolismo , Junções Íntimas/metabolismo
6.
Eur J Hum Genet ; 26(12): 1840-1847, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30139988

RESUMO

For multiple generations, much of the Arab population of Northern Israel has lived in communities with consanguineous marriages and large families. These communities have been particularly cooperative and informative for understanding the genetics of recessive traits. We studied the genetics of hearing loss in this population, evaluating 168 families from 46 different villages. All families were screened for founder variants by Sanger sequencing and 13 families were further evaluated by sequencing all known genes for hearing loss using our targeted gene panel HEar-Seq. Deafness in 34 of 168 families (20%) was explained by founder variants in GJB2, SLC26A4, or OTOF. In 6 of 13 families (46%) evaluated using HEar-Seq, deafness was explained by damaging alleles of SLC26A4, MYO15A, OTOG, LOXHD1, and TBC1D24. In some genes critical to hearing, it is particularly difficult to interpret variants that might affect splicing, because the genes are not expressed in accessible tissue. To address this problem for possible splice-altering variants of MYO15A, we evaluated minigenes transfected into HEK293 cells. Results revealed exon skipping in the message of MYO15A c.9083+6T>A, and intron retention in the message of MYO15A c.8340G>A, in each case leading to a premature stop and consistent with co-segregation of homozygosity for each variant with hearing loss. The profile of genetics of hearing loss in this population reflects the genetic heterogeneity of hearing loss and the usefulness of synthetic technologies to evaluate potentially causal variants in genes not expressed in accessible tissues.


Assuntos
Árabes/genética , Perda Auditiva/genética , Taxa de Mutação , Proteínas de Transporte/genética , Conexina 26 , Conexinas/genética , Feminino , Proteínas Ativadoras de GTPase , Células HEK293 , Perda Auditiva/epidemiologia , Humanos , Israel , Masculino , Proteínas de Membrana/genética , Miosinas/genética , Proteínas do Tecido Nervoso , Linhagem , Transportadores de Sulfato/genética
7.
BMC Genomics ; 18(1): 305, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28420336

RESUMO

BACKGROUND: The quantitative relations between RNA and protein are fundamental to biology and are still not fully understood. Across taxa, it was demonstrated that the protein-to-mRNA ratio in steady state varies in a direction that lessens the change in protein levels as a result of changes in the transcript abundance. Evidence for this behavior in tissues is sparse. We tested this phenomenon in new data that we produced for the mouse auditory system, and in previously published tissue datasets. A joint analysis of the transcriptome and proteome was performed across four datasets: inner-ear mouse tissues, mouse organ tissues, lymphoblastoid primate samples and human cancer cell lines. RESULTS: We show that the protein levels are more conserved than the mRNA levels in all datasets, and that changes in transcription are associated with translational changes that exert opposite effects on the final protein level, in all tissues except cancer. Finally, we observe that some functions are enriched in the inner ear on the mRNA level but not in protein. CONCLUSIONS: We suggest that partial buffering between transcription and translation ensures that proteins can be made rapidly in response to a stimulus. Accounting for the buffering can improve the prediction of protein levels from mRNA levels.


Assuntos
Neoplasias/genética , Proteoma/genética , RNA Mensageiro/genética , Transcriptoma/genética , Animais , Proliferação de Células , Orelha Interna/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Especificidade de Órgãos/genética , Primatas/genética , RNA Mensageiro/biossíntese
8.
Am J Otolaryngol ; 37(2): 162-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26954875

RESUMO

PURPOSE: The mechanism and the type of hearing loss induced by cochlear implants are mostly unknown. Therefore, this study evaluated the impact and type of hearing loss induced by each stage of cochlear implantation surgery in an animal model. STUDY DESIGN: Original basic research animal study. SETTING: The study was conducted in a tertiary, university-affiliated medical center in accordance with the guidelines of the Institutional Animal Care and Use Committee. SUBJECTS AND METHODS: Cochlear implant electrode array was inserted via the round window membrane in 17 ears of 9 adult-size fat sand rats. In 7 ears of 5 additional animals round window incision only was performed, followed by patching with a small piece of periosteum (control). Hearing thresholds to air (AC) and bone conduction (BC), clicks, 1 kHz and 6 kHz tone bursts were measured by auditory brainstem evoked potential, before, during each stage of surgery and one week post-operatively. In addition, inner ear histology was performed. RESULTS: The degree of hearing loss increased significantly from baseline throughout the stages of cochlear implantation surgery and up to one week after (p<0.0001). In both operated groups, the greatest deterioration was noted after round window incision. Overall, threshold shift to air-conduction clicks, reached 61 dB SPL and the bone conduction threshold deteriorated by 19 dB SPL only. Similar losses were found for 1-kHz and 6-kHz frequencies. The hearing loss was not associated with significant changes in inner ear histology. CONCLUSIONS: Hearing loss following cochlear implantation in normal hearing animals is progressive and of mixed type, but mainly conductive. Changes in the inner-ear mechanism are most likely responsible for the conductive hearing loss.


Assuntos
Limiar Auditivo/fisiologia , Condução Óssea/fisiologia , Cóclea/cirurgia , Implantes Cocleares/efeitos adversos , Perda Auditiva/etiologia , Janela da Cóclea/cirurgia , Animais , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Perda Auditiva/fisiopatologia , Ratos
9.
Eur J Hum Genet ; 22(6): 768-75, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24105371

RESUMO

Hereditary hearing loss is genetically heterogeneous, with a large number of genes and mutations contributing to this sensory, often monogenic, disease. This number, as well as large size, precludes comprehensive genetic diagnosis of all known deafness genes. A combination of targeted genomic capture and massively parallel sequencing (MPS), also referred to as next-generation sequencing, was applied to determine the deafness-causing genes in hearing-impaired individuals from Israeli Jewish and Palestinian Arab families. Among the mutations detected, we identified nine novel mutations in the genes encoding myosin VI, myosin VIIA and myosin XVA, doubling the number of myosin mutations in the Middle East. Myosin VI mutations were identified in this population for the first time. Modeling of the mutations provided predicted mechanisms for the damage they inflict in the molecular motors, leading to impaired function and thus deafness. The myosin mutations span all regions of these molecular motors, leading to a wide range of hearing phenotypes, reinforcing the key role of this family of proteins in auditory function. This study demonstrates that multiple mutations responsible for hearing loss can be identified in a relatively straightforward manner by targeted-gene MPS technology and concludes that this is the optimal genetic diagnostic approach for identification of mutations responsible for hearing loss.


Assuntos
Predisposição Genética para Doença/genética , Perda Auditiva/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação , Cadeias Pesadas de Miosina/genética , Miosinas/genética , Árabes/genética , Sequência de Bases , Saúde da Família , Feminino , Genômica/métodos , Humanos , Israel , Judeus/genética , Masculino , Miosina VIIa , Linhagem , Fenótipo
10.
Hear Res ; 309: 124-35, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24333301

RESUMO

Mutations in the connexin 26 gene (GJB2) are the most common genetic cause of deafness, leading to congenital bilateral non-syndromic sensorineural hearing loss. Here we report the generation of a mouse model for a connexin 26 (Cx26) mutation, in which cre-Sox10 drives excision of the Cx26 gene from non-sensory cells flanking the auditory epithelium. We determined that these conditional knockout mice, designated Gjb2-CKO, have a severe hearing loss. Immunocytochemistry of the auditory epithelium confirmed absence of Cx26 in the non-sensory cells. Histology of the organ of Corti and the spiral ganglion neurons (SGNs) performed at ages 1, 3, or 6 months revealed that in Gjb2-CKO mice, the organ of Corti began to degenerate in the basal cochlear turn at an early stage, and the degeneration rapidly spread to the apex. In addition, the density of SGNs in Rosenthal's canal decreased rapidly along a gradient from the base of the cochlea to the apex, where some SGNs survived until at least 6 months of age. Surviving neurons often clustered together and formed clumps of cells in the canal. We then assessed the influence of brain derived neurotrophic factor (BDNF) gene therapy on the SGNs of Gjb2-CKO mice by inoculating Adenovirus with the BDNF gene insert (Ad.BDNF) into the base of the cochlea via the scala tympani or scala media. We determined that over-expression of BDNF beginning around 1 month of age resulted in a significant rescue of neurons in Rosenthal's canal of the cochlear basal turn but not in the middle or apical portions. This data may be used to design therapies for enhancing the SGN physiological status in all GJB2 patients and especially in a sub-group of GJB2 patients where the hearing loss progresses due to ongoing degeneration of the auditory nerve, thereby improving the outcome of cochlear implant therapy in these ears.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Conexinas/deficiência , Terapia Genética/métodos , Perda Auditiva Neurossensorial/terapia , Neurônios/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Adenoviridae/genética , Fatores Etários , Animais , Limiar Auditivo , Fator Neurotrófico Derivado do Encéfalo/genética , Conexina 26 , Conexinas/genética , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Perda Auditiva Neurossensorial/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural , Neurônios/patologia , Órgão Espiral/metabolismo , Órgão Espiral/patologia , Gânglio Espiral da Cóclea/patologia , Gânglio Espiral da Cóclea/fisiopatologia
11.
Front Mol Neurosci ; 6: 52, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24391537

RESUMO

Non-coding microRNAs (miRNAs) have a fundamental role in gene regulation and expression in almost every multicellular organism. Only discovered in the last decade, miRNAs are already known to play a leading role in many aspects of disease. In the vertebrate inner ear, miRNAs are essential for controlling development and survival of hair cells. Moreover, dysregulation of miRNAs has been implicated in sensorineural hearing impairment, as well as in other ear diseases such as cholesteatomas, vestibular schwannomas, and otitis media. Due to the inaccessibility of the ear in humans, animal models have provided the optimal tools to study miRNA expression and function, in particular mice and zebrafish. A major focus of current research has been to discover the targets of the miRNAs expressed in the inner ear, in order to determine the regulatory pathways of the auditory and vestibular systems. The potential for miRNAs manipulation in development of therapeutic tools for hearing impairment is as yet unexplored, paving the way for future work in the field.

12.
EMBO Mol Med ; 4(9): 849-59, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22745034

RESUMO

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression through the RNA interference (RNAi) pathway and by inhibition of mRNA translation. miRNAs first made their appearance in the auditory and vestibular systems in 2005, with the discovery of a triad of hair cell-specific miRNAs later found to be involved in both human and mouse deafness. Since then, miRNAs have been implicated in other medical conditions related to these systems, such as cholesteatomas, vestibular schwannomas and otitis media. Due to the limitations in studying miRNAs and their targets derived from human inner ears, animal models are vital in this field of research. Therefore their role in inner ear development and function has been demonstrated by studies in zebrafish and mice. Transcriptomic and proteomic approaches have been undertaken to identify miRNAs and their targets. Finally, it has been suggested that miRNAs may be used in the future in regeneration of inner ear hair cells and ultimately play a role in therapeutics.


Assuntos
Colesteatoma da Orelha Média/patologia , Surdez/patologia , Inativação Gênica , MicroRNAs/metabolismo , Neuroma Acústico/patologia , Otite Média/patologia , Animais , Colesteatoma da Orelha Média/genética , Colesteatoma da Orelha Média/fisiopatologia , Surdez/genética , Surdez/fisiopatologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos , MicroRNAs/genética , Neuroma Acústico/genética , Neuroma Acústico/fisiopatologia , Otite Média/genética , Otite Média/fisiopatologia , Proteômica/métodos , Peixe-Zebra
13.
Genome Biol ; 12(9): R89, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917145

RESUMO

BACKGROUND: Identification of genes responsible for medically important traits is a major challenge in human genetics. Due to the genetic heterogeneity of hearing loss, targeted DNA capture and massively parallel sequencing are ideal tools to address this challenge. Our subjects for genome analysis are Israeli Jewish and Palestinian Arab families with hearing loss that varies in mode of inheritance and severity. RESULTS: A custom 1.46 MB design of cRNA oligonucleotides was constructed containing 246 genes responsible for either human or mouse deafness. Paired-end libraries were prepared from 11 probands and bar-coded multiplexed samples were sequenced to high depth of coverage. Rare single base pair and indel variants were identified by filtering sequence reads against polymorphisms in dbSNP132 and the 1000 Genomes Project. We identified deleterious mutations in CDH23, MYO15A, TECTA, TMC1, and WFS1. Critical mutations of the probands co-segregated with hearing loss. Screening of additional families in a relevant population was performed. TMC1 p.S647P proved to be a founder allele, contributing to 34% of genetic hearing loss in the Moroccan Jewish population. CONCLUSIONS: Critical mutations were identified in 6 of the 11 original probands and their families, leading to the identification of causative alleles in 20 additional probands and their families. The integration of genomic analysis into early clinical diagnosis of hearing loss will enable prediction of related phenotypes and enhance rehabilitation. Characterization of the proteins encoded by these genes will enable an understanding of the biological mechanisms involved in hearing loss.


Assuntos
Testes Genéticos/métodos , Perda Auditiva/genética , Judeus/genética , Alelos , Animais , Árabes/genética , Proteínas Relacionadas a Caderinas , Caderinas/genética , Biologia Computacional , Análise Mutacional de DNA/métodos , Éxons , Proteínas da Matriz Extracelular/genética , Efeito Fundador , Proteínas Ligadas por GPI/genética , Frequência do Gene , Biblioteca Gênica , Predisposição Genética para Doença , Genética Populacional , Genoma Humano , Perda Auditiva/epidemiologia , Humanos , Mutação INDEL , Padrões de Herança , Proteínas de Membrana/genética , Camundongos , Oriente Médio/epidemiologia , Miosinas/genética , Linhagem
14.
J Med Genet ; 47(9): 643-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20472657

RESUMO

BACKGROUND: Moderate to severe prelingual hearing impairment (DFNB84) was observed in an extended consanguineous Palestinian kindred. All affected relatives shared a 12.5 MB homozygous haplotype on chromosome 12q21 with lod score 4.30. This homozygous region harbours the protein tyrosine phosphatase receptor Q gene PTPRQ, which is known to be essential to hearing in mouse. METHODS: Candidate genes in the 12.5 MB homozygous region were characterized genomically and sequenced in deaf and hearing relatives in the family. RESULTS: Sequence of PTPRQ in affected individuals in the extended kindred revealed c.1285C-->T, leading to p.Gln429Stop. This nonsense mutation co-segregated with hearing loss in the family and was homozygous in all affected relatives. The mutation did not appear among 288 Palestinian controls (576 chromosomes), all adults with normal hearing. No homozygous mutations in PTPRQ appeared in any of 218 other probands with hearing loss. CONCLUSION: Identification of the DFNB84 gene represents the first identification of PTPRQ mutation in human hearing loss.


Assuntos
Cílios/genética , Códon sem Sentido/genética , Loci Gênicos/genética , Perda Auditiva/enzimologia , Perda Auditiva/genética , Proteínas de Membrana/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Análise Mutacional de DNA , Família , Feminino , Humanos , Padrões de Herança/genética , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem
15.
Brain Res ; 1317: 157-64, 2010 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-20043895

RESUMO

Anxiety disorders are among the most common mental disorders, and are comorbid with balance disorders in a significant proportion of these individuals. Presently, it is unclear whether anxiety and balance disorders are causally related, and what direction this causality may take. We argue that balance disorders may predispose an individual to anxiety and that demonstration of such causality may be informative to the development of preferred treatment for such individuals. To demonstrate that balance disorders may predispose to anxiety, we studied headbanger (Hdb) mutant mice in which the balance disorder is due to progressive vestibular impairment and wildtype (Wt) mice. Balance was assessed by swim and tail-hang tests that demonstrated clear behavioral balance deficits in the Hdb mice. Anxiety was assessed by open-field and elevated plus-maze tests, which confirmed elevated anxiety in the Hdb mice. These findings demonstrate that congenital vestibular genotype predisposes the animal to elevated levels of anxiety in space-related tests. Similar causality in clinics may redirect treatment strategies in afflicted patients.


Assuntos
Ansiedade/genética , Doenças Vestibulares/genética , Doenças Vestibulares/psicologia , Envelhecimento , Análise de Variância , Animais , Predisposição Genética para Doença , Genótipo , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Mutantes , Mutação , Miosina VIIa , Miosinas/genética , Testes Neuropsicológicos , Equilíbrio Postural/genética , Natação , Análise e Desempenho de Tarefas , Fatores de Tempo
16.
PLoS Genet ; 4(10): e1000207, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18833301

RESUMO

Myosin VI, found in organisms from Caenorhabditis elegans to humans, is essential for auditory and vestibular function in mammals, since genetic mutations lead to hearing impairment and vestibular dysfunction in both humans and mice. Here, we show that a missense mutation in this molecular motor in an ENU-generated mouse model, Tailchaser, disrupts myosin VI function. Structural changes in the Tailchaser hair bundles include mislocalization of the kinocilia and branching of stereocilia. Transfection of GFP-labeled myosin VI into epithelial cells and delivery of endocytic vesicles to the early endosome revealed that the mutant phenotype displays disrupted motor function. The actin-activated ATPase rates measured for the D179Y mutation are decreased, and indicate loss of coordination of the myosin VI heads or 'gating' in the dimer form. Proper coordination is required for walking processively along, or anchoring to, actin filaments, and is apparently destroyed by the proximity of the mutation to the nucleotide-binding pocket. This loss of myosin VI function may not allow myosin VI to transport its cargoes appropriately at the base and within the stereocilia, or to anchor the membrane of stereocilia to actin filaments via its cargos, both of which lead to structural changes in the stereocilia of myosin VI-impaired hair cells, and ultimately leading to deafness.


Assuntos
Surdez/genética , Surdez/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Mutação de Sentido Incorreto , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Linhagem Celular , Mapeamento Cromossômico , Feminino , Células Ciliadas Auditivas Internas/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Cadeias Pesadas de Miosina/química , Estrutura Terciária de Proteína , Transporte Proteico , Vesículas Transportadoras/química , Vesículas Transportadoras/metabolismo
17.
Arch Otolaryngol Head Neck Surg ; 134(4): 403-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18427006

RESUMO

OBJECTIVES: To identify mutations in the SLC26A4 gene in individuals with nonsyndromic hearing loss and enlarged vestibular aqueduct, to design a predicted model of the pendrin protein, and to characterize novel mutations by means of localization in mammalian cells and effect of the mutation on the predicted model. DESIGN: Validation of the mutation by its exclusion in more than 300 individuals with normal hearing. SETTING: A laboratory of genetics of hearing loss research, clinical genetics laboratories, an otolaryngology department at Tel Aviv University, and medical centers in Israel. PATIENTS: A patient with nonsyndromic hearing loss and enlarged vestibular aqueduct, 203 deaf probands, and 310 controls with normal hearing. INTERVENTIONS: Sequencing the SLC26A4 gene in the patient with nonsyndromic hearing loss and enlarged vestibular aqueduct. Transfection of yellow fluorescent protein (YFP) constructs into mammalian COS7 cells. Designing a computational model of the human SLC26A4 protein. MAIN OUTCOME MEASURE: Detection of a novel c.1458_1459insT SLC26A4 mutation. RESULTS: A computational model of the human pendrin protein suggests that the novel c.1458_1459insT mutation leads to a prematurely truncated protein, p.Ile487TyrfsX39. Mammalian COS7 cells transfected with the YFP-1458_1459insT construct showed mislocalization of the mutant protein. CONCLUSIONS: A novel SLC26A4 mutation was detected in Israel. Because current estimates demonstrate that SLC26A4 mutations are involved in up to 4% of nonsyndromic deafness, our findings emphasize the importance of adding a molecular test for the SLC26A4 gene in the diagnosis of deafness, particularly when bone abnormalities are involved, to the list of genes screened in Israel and elsewhere in the world.


Assuntos
Surdez/genética , Retículo Endoplasmático/genética , Perda Auditiva Neurossensorial/genética , Proteínas de Membrana Transportadoras/genética , Mutação/genética , Aqueduto Vestibular/anormalidades , Adulto , Estudos de Casos e Controles , Criança , Surdez/etnologia , Feminino , Perda Auditiva Neurossensorial/etnologia , Humanos , Irã (Geográfico)/epidemiologia , Israel , Judeus/genética , Masculino , Transportadores de Sulfato
18.
Arch Otolaryngol Head Neck Surg ; 132(5): 495-500, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16702564

RESUMO

OBJECTIVE: To compare performance after cochlear implantation in children with mutations in connexin (Cx) 26 (GJB2) or Cx30 (GJB6) and children with deafness of unknown etiology. DESIGN: Genetic analysis and speech perception evaluation was performed in the children with and without Cx mutations who had undergone cochlear implantation. Speech perception performance was retrospectively analyzed 6, 12, 24, 36, and 48 months after implantation. Test material was selected according to the child's age and cognitive and language abilities. SETTING: The study took place at speech and hearing and genetic centers of a hospital in the central part of Israel and the genetics departments of 3 additional centrally located hospitals. PATIENTS: A total of 30 children who had undergone cochlear implantation were selected for the study, with control patients matched according to age at implantation, duration of implant use, and mode of communication. There was no evidence for additional disabilities or handicaps in either group. MAIN OUTCOME MEASURES: Speech perception measurements included a questionnaire, as well as closed and open-set tests. RESULTS: Overall, the 2 groups showed significant improvement in speech perception results after implantation. Four years after implantation, both groups achieved mean open-set speech perception scores of approximately 60%, 75%, and 90% for monosyllabic, 2 syllables, and words in sentences tests, respectively. CONCLUSIONS: There were no apparent differences in speech perception performance after implantation between the children with Cx mutations and children with deafness of unknown etiology. These data have important implications as a prognostic indicator when counseling candidates for cochlear implantation.


Assuntos
Implantes Cocleares , Conexinas/genética , Análise Mutacional de DNA , Surdez/genética , Percepção da Fala , Criança , Pré-Escolar , Deleção Cromossômica , Conexina 26 , Conexina 30 , Surdez/reabilitação , Feminino , Triagem de Portadores Genéticos , Homozigoto , Humanos , Lactente , Masculino , Avaliação de Resultados em Cuidados de Saúde , Reação em Cadeia da Polimerase , Estudos Retrospectivos , Teste do Limiar de Recepção da Fala
19.
Arch Otolaryngol Head Neck Surg ; 132(4): 416-24, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16618911

RESUMO

OBJECTIVE: To perform chromosomal mapping and clinical analysis of hereditary otosclerosis linked to the fourth locus for otosclerosis (OTSC4) in an Israeli family. DESIGN: Pedigree study. SETTING: A genetics of hearing loss research laboratory, a clinical genetics laboratory, a center for speech and hearing, and an otolaryngology department at a university and medical centers in Israel. SUBJECTS: An Israeli family of which 24 members were ascertained and a pedigree was constructed; 12 members had otosclerosis. INTERVENTIONS: Confirmation of otosclerosis by surgery (3 subjects) and by audiologic evaluation, medical history, and family history (9 subjects), and whole-genome scanning to identify the chromosomal region of the mutant locus. MAIN OUTCOME MEASURES: Chromosomal location of the otosclerosis locus. RESULTS: Linkage to the 16q21-23.2 interval was identified and confirmed with a logarithm of odds (LOD) score of 3.97 at theta = 0. The new locus for otosclerosis was designated OTSC4. The OTSC4 interval of 9 to 10 megabase includes several genes involved in the immune system and bone homeostasis that may be good candidates for genes otosclerosis. CONCLUSION: The elucidation of the OTSC4 gene may disclose the etiology of the disorder, and the functional and structural analysis of the protein may open new options for diagnosis, treatment, and prevention of otosclerosis.


Assuntos
Cromossomos Humanos Par 16/genética , Ligação Genética , Perda Auditiva Condutiva/genética , Otosclerose/genética , Fenótipo , Adulto , Idoso , Audiometria , Mapeamento Cromossômico , Feminino , Humanos , Israel , Escore Lod , Masculino , Pessoa de Meia-Idade , Linhagem , Penetrância
20.
EMBO Rep ; 7(3): 326-33, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16397623

RESUMO

Gfi1 is a transcriptional repressor essential for haematopoiesis and inner ear development. It shares with its paralogue Gfi1b an amino-terminal SNAG repressor domain and six carboxy-terminal zinc-finger motifs, but differs from Gfi1b in sequences separating these domains. Here, we describe two knock-in mouse models, in which the N-terminal SNAG repressor domain was mutated or in which the Gfi1 coding region was replaced by Gfi1b. Mouse mutants without an intact SNAG domain show the full phenotype of Gfi1 null mice. However, Gfi1:Gfi1b knock-in mice show almost normal pre-T-cell and neutrophil development, but lack properly formed inner ear hair cells. Hence, our findings show that an intact SNAG domain is essential for all functions of Gfi1 and that Gfi1b can replace Gfi1 functionally in haematopoiesis but, surprisingly, not in inner ear hair cell development, demonstrating that Gfi1 and Gfi1b have equivalent and domain-dependent, cell type-specific functions.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Orelha Interna/crescimento & desenvolvimento , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Proteínas de Ligação a DNA/genética , Orelha Interna/citologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Camundongos , Camundongos Knockout , Mutação , Fenótipo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA