Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Otol Neurotol ; 42(4): e399-e407, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33710989

RESUMO

OBJECTIVE: To study the genotype and phenotype of a Dutch family with autosomal dominantly inherited hearing loss. STUDY DESIGN: Genotype-phenotype correlation study. Genetic analysis consisted of linkage analysis, variable number of tandem repeats analysis, and Sanger sequencing. Audiovestibular function was examined. Regression analysis was performed on pure tone audiometry and speech recognition scores and correlated with the age and/or level of hearing loss. SETTING: Tertiary referral center. PATIENTS: A large Dutch family presenting with sensorineural hearing loss. MAIN OUTCOME MEASURES: Identification of the underlying genetic defect of the hearing loss in this family. Results of pure tone and speech audiometry, onset age, progression of hearing loss and vestibular (dys)function. RESULTS: A novel mutation in COCH, c.1312C > T p.(Arg438Cys), cosegregates with hearing loss and a variable degree of vestibular (dys)function in this family. The reported mean age of onset of hearing loss is 33 years (range, 18-49 yr). Hearing loss primarily affects higher frequencies and its progression is relatively mild (0.8 dB/yr). Speech perception is remarkably well preserved in affected family members when compared with other DFNA9 families with different COCH mutations. CONCLUSION: These findings expand the genotypic and phenotypic spectrum of DFNA9. The c.1312C > T mutation, which affects the vWFA2 domain, causes a relatively mild audiovestibular phenotype when compared with other COCH mutations.


Assuntos
Proteínas da Matriz Extracelular , Perda Auditiva Neurossensorial , Adolescente , Adulto , Análise Mutacional de DNA , Proteínas da Matriz Extracelular/genética , Perda Auditiva Neurossensorial/genética , Humanos , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Adulto Jovem
2.
Hum Genet ; 138(1): 61-72, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30535804

RESUMO

ATP2B2 encodes the PMCA2 Ca2+ pump that plays an important role in maintaining ion homeostasis in hair cells among others by extrusion of Ca2+ from the stereocilia to the endolymph. Several mouse models have been described for this gene; mice heterozygous for loss-of-function defects display a rapidly progressive high-frequency hearing impairment. Up to now ATP2B2 has only been reported as a modifier, or in a digenic mechanism with CDH23 for hearing impairment in humans. Whole exome sequencing in hearing impaired index cases of Dutch and Polish origins revealed five novel heterozygous (predicted to be) loss-of-function variants of ATP2B2. Two variants, c.1963G>T (p.Glu655*) and c.955delG (p.Ala319fs), occurred de novo. Three variants c.397+1G>A (p.?), c.1998C>A (p.Cys666*), and c.2329C>T (p.Arg777*), were identified in families with an autosomal dominant inheritance pattern of hearing impairment. After normal newborn hearing screening, a rapidly progressive high-frequency hearing impairment was diagnosed at the age of about 3-6 years. Subjects had no balance complaints and vestibular testing did not yield abnormalities. There was no evidence for retrocochlear pathology or structural inner ear abnormalities. Although a digenic inheritance pattern of hearing impairment has been reported for heterozygous missense variants of ATP2B2 and CDH23, our findings indicate a monogenic cause of hearing impairment in cases with loss-of-function variants of ATP2B2.


Assuntos
Biomarcadores/análise , Predisposição Genética para Doença , Perda Auditiva/genética , Mutação , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Seguimentos , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Prognóstico , Adulto Jovem
3.
PLoS One ; 13(8): e0201713, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30157177

RESUMO

Hearing and balance depend upon the precise morphogenesis and mechanosensory function of stereocilia, the specialized structures on the apical surface of sensory hair cells in the inner ear. Previous studies of Grxcr1 mutant mice indicated a critical role for this gene in control of stereocilia dimensions during development. In this study, we analyzed expression of the paralog Grxcr2 in the mouse and evaluated auditory and vestibular function of strains carrying targeted mutations of the gene. Peak expression of Grxcr2 occurs during early postnatal development of the inner ear and GRXCR2 is localized to stereocilia in both the cochlea and in vestibular organs. Homozygous Grxcr2 deletion mutants exhibit significant hearing loss by 3 weeks of age that is associated with developmental defects in stereocilia bundle orientation and organization. Despite these bundle defects, the mechanotransduction apparatus assembles in relatively normal fashion as determined by whole cell electrophysiological evaluation and FM1-43 uptake. Although Grxcr2 mutants do not exhibit overt vestibular dysfunction, evaluation of vestibular evoked potentials revealed subtle defects of the mutants in response to linear accelerations. In addition, reduced Grxcr2 expression in a hypomorphic mutant strain is associated with progressive hearing loss and bundle defects. The stereocilia localization of GRXCR2, together with the bundle pathologies observed in the mutants, indicate that GRXCR2 plays an intrinsic role in bundle orientation, organization, and sensory function in the inner ear during development and at maturity.


Assuntos
Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Glutarredoxinas/metabolismo , Morfogênese , Estereocílios/metabolismo , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos/genética , Glutarredoxinas/química , Glutarredoxinas/genética , Perda Auditiva/genética , Perda Auditiva/patologia , Humanos , Mecanotransdução Celular , Camundongos , Modelos Moleculares , Mutação , Conformação Proteica , Especificidade da Espécie
4.
Am J Hum Genet ; 103(1): 74-88, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29961571

RESUMO

In a Dutch consanguineous family with recessively inherited nonsyndromic hearing impairment (HI), homozygosity mapping combined with whole-exome sequencing revealed a MPZL2 homozygous truncating variant, c.72del (p.Ile24Metfs∗22). By screening a cohort of phenotype-matched subjects and a cohort of HI subjects in whom WES had been performed previously, we identified two additional families with biallelic truncating variants of MPZL2. Affected individuals demonstrated symmetric, progressive, mild to moderate sensorineural HI. Onset of HI was in the first decade, and high-frequency hearing was more severely affected. There was no vestibular involvement. MPZL2 encodes myelin protein zero-like 2, an adhesion molecule that mediates epithelial cell-cell interactions in several (developing) tissues. Involvement of MPZL2 in hearing was confirmed by audiometric evaluation of Mpzl2-mutant mice. These displayed early-onset progressive sensorineural HI that was more pronounced in the high frequencies. Histological analysis of adult mutant mice demonstrated an altered organization of outer hair cells and supporting cells and degeneration of the organ of Corti. In addition, we observed mild degeneration of spiral ganglion neurons, and this degeneration was most pronounced at the cochlear base. Although MPZL2 is known to function in cell adhesion in several tissues, no phenotypes other than HI were found to be associated with MPZL2 defects. This indicates that MPZL2 has a unique function in the inner ear. The present study suggests that deleterious variants of Mplz2/MPZL2 affect adhesion of the inner-ear epithelium and result in loss of structural integrity of the organ of Corti and progressive degeneration of hair cells, supporting cells, and spiral ganglion neurons.


Assuntos
Moléculas de Adesão Celular/genética , Células Ciliadas Auditivas/patologia , Perda Auditiva Neurossensorial/genética , Audição/genética , Animais , Adesão Celular/genética , Cóclea/patologia , Surdez/genética , Epitélio/patologia , Feminino , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Neurônios/patologia , Gânglio Espiral da Cóclea/patologia
5.
Hum Genet ; 137(5): 389-400, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29754270

RESUMO

Unraveling the causes and pathomechanisms of progressive disorders is essential for the development of therapeutic strategies. Here, we identified heterozygous pathogenic missense variants of LMX1A in two families of Dutch origin with progressive nonsyndromic hearing impairment (HI), using whole exome sequencing. One variant, c.721G > C (p.Val241Leu), occurred de novo and is predicted to affect the homeodomain of LMX1A, which is essential for DNA binding. The second variant, c.290G > C (p.Cys97Ser), predicted to affect a zinc-binding residue of the second LIM domain that is involved in protein-protein interactions. Bi-allelic deleterious variants of Lmx1a are associated with a complex phenotype in mice, including deafness and vestibular defects, due to arrest of inner ear development. Although Lmx1a mouse mutants demonstrate neurological, skeletal, pigmentation and reproductive system abnormalities, no syndromic features were present in the participating subjects of either family. LMX1A has previously been suggested as a candidate gene for intellectual disability, but our data do not support this, as affected subjects displayed normal cognition. Large variability was observed in the age of onset (a)symmetry, severity and progression rate of HI. About half of the affected individuals displayed vestibular dysfunction and experienced symptoms thereof. The late-onset progressive phenotype and the absence of cochleovestibular malformations on computed tomography scans indicate that heterozygous defects of LMX1A do not result in severe developmental abnormalities in humans. We propose that a single LMX1A wild-type copy is sufficient for normal development but insufficient for maintenance of cochleovestibular function. Alternatively, minor cochleovestibular developmental abnormalities could eventually lead to the progressive phenotype seen in the families.


Assuntos
Perda Auditiva/genética , Heterozigoto , Proteínas com Homeodomínio LIM/genética , Mutação de Sentido Incorreto , Fatores de Transcrição/genética , Doenças Vestibulares/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Substituição de Aminoácidos , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
6.
Hear Res ; 347: 56-62, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28089734

RESUMO

DFNB28 is characterized by prelingual, severe to profound sensorineural hearing impairment (HI). It is associated with mutations in exon 6 and 7 of TRIOBP and has not been reported in the European population. Here, we describe two isolated cases of Dutch origin with congenital, moderate HI and compound heterozygous mutations in TRIOBP. Three of the mutations are novel, one nonsense mutation (c.5014G>T (p.Gly1672*)) and two frameshift mutations (c.2653del (p.Arg885Alafs*120) and c.3460_3461del (p.Leu1154Alafs*29)). The fourth mutation is the known c.3232dup (p.Arg1078Profs*6) mutation. Longitudinal audiometric analyses in one of the subjects revealed that HI was stable over a period of 15 years. Vestibular function was normal. Predicted effects of the mutations do not explain the relatively mild phenotype in the presented subjects, whereas location of the mutation might well contribute to the milder HI in one of the subjects. It is known that isoform classes TRIOBP-4 and TRIOBP-5 are important for stereocilia stability and rigidity. To our knowledge, p.Gly1672* is the first pathogenic variant identified in DFNB28 that does not affect isoform class TRIOBP-4. This suggests that a single TRIOBP copy to encode wildtype TRIOBP-4 is insufficient for normal hearing, and that at least one TRIOBP copy to encode TRIOBP-5 is indispensable for normal inner ear function. Furthermore, this study demonstrates that DFNB28 can be milder than reported so far and that mutations in TRIOBP are thus associated with a heterogeneous phenotype.


Assuntos
Códon sem Sentido , Mutação da Fase de Leitura , Perda Auditiva Neurossensorial/genética , Audição/genética , Proteínas dos Microfilamentos/genética , Limiar Auditivo , Análise Mutacional de DNA , Marcadores Genéticos , Predisposição Genética para Doença , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/fisiopatologia , Perda Auditiva Neurossensorial/psicologia , Testes Auditivos , Hereditariedade , Humanos , Linhagem , Fenótipo , Fatores de Risco , Índice de Gravidade de Doença
7.
Dis Model Mech ; 10(2): 105-118, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28067622

RESUMO

A consanguineous family from Pakistan was ascertained to have a novel deafness-dystonia syndrome with motor regression, ichthyosis-like features and signs of sensory neuropathy. By applying a combined strategy of linkage analysis and whole-exome sequencing in the presented family, a homozygous nonsense mutation, c.4G>T (p.Glu2*), in FITM2 was identified. FITM2 and its paralog FITM1 constitute an evolutionary conserved protein family involved in partitioning of triglycerides into cellular lipid droplets. Despite the role of FITM2 in neutral lipid storage and metabolism, no indications for lipodystrophy were observed in the affected individuals. In order to obtain independent evidence for the involvement of FITM2 in the human pathology, downregulation of the single Fitm ortholog, CG10671, in Drosophila melanogaster was pursued using RNA interference. Characteristics of the syndrome, including progressive locomotor impairment, hearing loss and disturbed sensory functions, were recapitulated in Drosophila, which supports the causative nature of the FITM2 mutation. Mutation-based genetic counseling can now be provided to the family and insight is obtained into the potential impact of genetic variation in FITM2.


Assuntos
Surdocegueira/genética , Proteínas de Drosophila/genética , Distonia/genética , Ictiose/genética , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Atividade Motora , Mutação/genética , Atrofia Óptica/genética , Células Receptoras Sensoriais/patologia , Adiposidade , Animais , Audiometria de Tons Puros , Sequência de Bases , Criança , Códon sem Sentido/genética , Surdocegueira/sangue , Surdocegueira/fisiopatologia , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Distonia/sangue , Distonia/fisiopatologia , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Perda Auditiva/genética , Homozigoto , Humanos , Ictiose/complicações , Ictiose/fisiopatologia , Deficiência Intelectual/sangue , Deficiência Intelectual/fisiopatologia , Gotículas Lipídicas/metabolismo , Fígado/metabolismo , Locomoção , Masculino , Proteínas de Membrana/metabolismo , Atrofia Óptica/sangue , Atrofia Óptica/fisiopatologia , Linhagem , Sequenciamento do Exoma , Adulto Jovem
8.
Eur J Hum Genet ; 25(3): 308-314, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28000701

RESUMO

Hearing impairment (HI) is genetically heterogeneous which hampers genetic counseling and molecular diagnosis. Testing of several single HI-related genes is laborious and expensive. In this study, we evaluate the diagnostic utility of whole-exome sequencing (WES) targeting a panel of HI-related genes. Two hundred index patients, mostly of Dutch origin, with presumed hereditary HI underwent WES followed by targeted analysis of an HI gene panel of 120 genes. We found causative variants underlying the HI in 67 of 200 patients (33.5%). Eight of these patients have a large homozygous deletion involving STRC, OTOA or USH2A, which could only be identified by copy number variation detection. Variants of uncertain significance were found in 10 patients (5.0%). In the remaining 123 cases, no potentially causative variants were detected (61.5%). In our patient cohort, causative variants in GJB2, USH2A, MYO15A and STRC, and in MYO6 were the leading causes for autosomal recessive and dominant HI, respectively. Segregation analysis and functional analyses of variants of uncertain significance will probably further increase the diagnostic yield of WES.


Assuntos
Exoma , Testes Genéticos/estatística & dados numéricos , Perda Auditiva/genética , Análise de Sequência de DNA/estatística & dados numéricos , Conexina 26 , Conexinas/genética , Variações do Número de Cópias de DNA , Proteínas da Matriz Extracelular/genética , Proteínas Ligadas por GPI/genética , Testes Genéticos/normas , Perda Auditiva/diagnóstico , Perda Auditiva/epidemiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana/genética , Mutação , Cadeias Pesadas de Miosina/genética , Miosinas/genética , Países Baixos , Análise de Sequência de DNA/normas
9.
Eur J Hum Genet ; 24(4): 542-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26173970

RESUMO

Variants in CIB2 can underlie either Usher syndrome type I (USH1J) or nonsyndromic hearing impairment (NSHI) (DFNB48). Here, a novel homozygous missense variant c.196C>T and compound heterozygous variants, c.[97C>T];[196C>T], were found, respectively, in two unrelated families of Dutch origin. Besides, the previously reported c.272 T>C functional missense variant in CIB2 was identified in two families of Pakistani origin. The missense variants are demonstrated not to affect subcellular localization of CIB2 in vestibular hair cells in ex vivo expression experiments. Furthermore, these variants do not affect the ATP-induced calcium responses in COS-7 cells. However, based on the residues affected, the variants are suggested to alter αIIß integrin binding. HI was nonsyndromic in all four families. However, deafness segregating with the c.272T>C variant in one Pakistani family is remarkably less severe than that in all other families with this mutation. Our results contribute to the insight in genotype-phenotype correlations of CIB2 mutations.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Cálcio/metabolismo , Surdez/genética , Células Ciliadas Auditivas/metabolismo , Adolescente , Adulto , Animais , Células COS , Proteínas de Ligação ao Cálcio/metabolismo , Criança , Chlorocebus aethiops , Surdez/metabolismo , Feminino , Humanos , Integrina alfa2beta1/metabolismo , Masculino , Mutação de Sentido Incorreto , Linhagem , Ligação Proteica
10.
Am J Hum Genet ; 97(5): 647-60, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26522471

RESUMO

Linkage analysis combined with whole-exome sequencing in a large family with congenital and stable non-syndromic unilateral and asymmetric hearing loss (NS-UHL/AHL) revealed a heterozygous truncating mutation, c.286_303delinsT (p.Ser96Ter), in KITLG. This mutation co-segregated with NS-UHL/AHL as a dominant trait with reduced penetrance. By screening a panel of probands with NS-UHL/AHL, we found an additional mutation, c.200_202del (p.His67_Cys68delinsArg). In vitro studies revealed that the p.His67_Cys68delinsArg transmembrane isoform of KITLG is not detectable at the cell membrane, supporting pathogenicity. KITLG encodes a ligand for the KIT receptor. Also, KITLG-KIT signaling and MITF are suggested to mutually interact in melanocyte development. Because mutations in MITF are causative of Waardenburg syndrome type 2 (WS2), we screened KITLG in suspected WS2-affected probands. A heterozygous missense mutation, c.310C>G (p.Leu104Val), that segregated with WS2 was identified in a small family. In vitro studies revealed that the p.Leu104Val transmembrane isoform of KITLG is located at the cell membrane, as is wild-type KITLG. However, in culture media of transfected cells, the p.Leu104Val soluble isoform of KITLG was reduced, and no soluble p.His67_Cys68delinsArg and p.Ser96Ter KITLG could be detected. These data suggest that mutations in KITLG associated with NS-UHL/AHL have a loss-of-function effect. We speculate that the mechanism of the mutation underlying WS2 and leading to membrane incorporation and reduced secretion of KITLG occurs via a dominant-negative or gain-of-function effect. Our study unveils different phenotypes associated with KITLG, previously associated with pigmentation abnormalities, and will thereby improve the genetic counseling given to individuals with KITLG variants.


Assuntos
Ligação Genética , Perda Auditiva Unilateral/genética , Mutação/genética , Fator de Células-Tronco/genética , Síndrome de Waardenburg/genética , Alelos , Animais , Feminino , Imunofluorescência , Perda Auditiva Unilateral/metabolismo , Perda Auditiva Unilateral/patologia , Humanos , Masculino , Camundongos , Células NIH 3T3 , Linhagem , Fenótipo , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome de Waardenburg/metabolismo , Síndrome de Waardenburg/patologia
11.
Ear Hear ; 36(2): 205-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25255398

RESUMO

OBJECTIVE: Currently, six genes are known to be associated with Usher syndrome type I, and mutations in most of these genes can also cause nonsyndromic hearing loss. The one exception is USH1G, which is currently only known to be involved in Usher syndrome type I and atypical Usher syndrome. DESIGN: A Dutch family with autosomal recessively inherited hearing loss was examined. Audiometric, ophthalmic, and vestibular evaluations were performed besides the genetic analysis. RESULTS: The hearing loss had an early onset with a downsloping audiogram configuration. Slight progression of the hearing loss was seen in both affected individuals. Compound heterozygous mutations in USH1G were found to segregate with the hearing loss in this family, a missense (c.310A>G, p.Met104Val) and a frameshift mutation (c.780insGCAC, p.Tyr261Alafs*96). Extensive ophthalmic and vestibular examinations demonstrated no abnormalities that are usually associated with Usher syndrome type I. CONCLUSIONS: This is the first family presented with nonsyndromic hearing loss caused by mutations in USH1G. Our findings expand the phenotypic spectrum of mutations in USH1G.


Assuntos
Perda Auditiva Neurossensorial/genética , Proteínas do Tecido Nervoso/genética , Audiometria de Tons Puros , Feminino , Mutação da Fase de Leitura , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Masculino , Mutação de Sentido Incorreto , Linhagem , Síndromes de Usher/genética
12.
Eur J Hum Genet ; 23(2): 189-94, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24781754

RESUMO

In a consanguineous Turkish family diagnosed with autosomal recessive nonsyndromic hearing impairment (arNSHI), a homozygous region of 47.4 Mb was shared by the two affected siblings on chromosome 6p21.1-q15. This region contains 247 genes including the known deafness gene MYO6. No pathogenic variants were found in MYO6, neither with sequence analysis of the coding region and splice sites nor with mRNA analysis. Subsequent candidate gene evaluation revealed CLIC5 as an excellent candidate gene. The orthologous mouse gene is mutated in the jitterbug mutant that exhibits progressive hearing impairment and vestibular dysfunction. Mutation analysis of CLIC5 revealed a homozygous nonsense mutation c.96T>A (p.(Cys32Ter)) that segregated with the hearing loss. Further analysis of CLIC5 in 213 arNSHI patients from mostly Dutch and Spanish origin did not reveal any additional pathogenic variants. CLIC5 mutations are thus not a common cause of arNSHI in these populations. The hearing loss in the present family had an onset in early childhood and progressed from mild to severe or even profound before the second decade. Impaired hearing is accompanied by vestibular areflexia and in one of the patients with mild renal dysfunction. Although we demonstrate that CLIC5 is expressed in many other human tissues, no additional symptoms were observed in these patients. In conclusion, our results show that CLIC5 is a novel arNSHI gene involved in progressive hearing impairment, vestibular and possibly mild renal dysfunction in a family of Turkish origin.


Assuntos
Canais de Cloreto/genética , Códon sem Sentido , Homozigoto , Proteínas dos Microfilamentos/genética , Doenças Vestibulares/genética , Adolescente , Linhagem Celular , Criança , Canais de Cloreto/metabolismo , Surdez/diagnóstico , Surdez/genética , Feminino , Humanos , Lactente , Masculino , Proteínas dos Microfilamentos/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido , Linhagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Doenças Vestibulares/diagnóstico
13.
J Hum Genet ; 59(12): 683-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25296581

RESUMO

With homozygosity mapping we have identified two large homozygous regions on chromosome 3q13.11-q13.31 and chromosome 19p13.3-q31.32 in a large Pakistani family suffering from autosomal recessive nonsyndromic hearing impairment (arNSHI). The region on chromosome 19 overlaps with the previously described deafness loci DFNB15, DFNB72 and DFNB95. Mutations in GIPC3 have been shown to underlie the nonsyndromic hearing impairment linked to these loci. Sequence analysis of all exons and exon-intron boundaries of GIPC3 revealed a homozygous canonical splice site mutation, c.226-1G>T, in GIPC3. This is the first mutation described in GIPC3 that affects splicing. The c.226-1G>T mutation is located in the acceptor splice site of intron 1 and is predicted to affect the normal splicing of exon 2. With a minigene assay it was shown to result in the use of an alternative acceptor site in exon 2, resulting in a frameshift and a premature stop codon. This study expands the mutational spectrum of GIPC3 in arNSHI.


Assuntos
Proteínas de Transporte/genética , Perda Auditiva Neurossensorial/genética , Sítios de Splice de RNA/genética , Proteínas Adaptadoras de Transdução de Sinal , Ligação Genética , Perda Auditiva Neurossensorial/patologia , Humanos , Mutação , Paquistão , Linhagem
14.
PLoS One ; 9(6): e100146, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24949729

RESUMO

The frequency of inherited bilateral autosomal recessive non-syndromic hearing loss (ARNSHL) in Pakistan is 1.6/1000 individuals. More than 50% of the families carry mutations in GJB2 while mutations in MYO15A account for about 5% of recessive deafness. In the present study a cohort of 30 ARNSHL families was initially screened for mutations in GJB2 and MYO15A. Homozygosity mapping was performed by employing whole genome single nucleotide polymorphism (SNP) genotyping in the families that did not carry mutations in GJB2 or MYO15A. Mutation analysis was performed for the known ARNSHL genes present in the homozygous regions to determine the causative mutations. This allowed the identification of a causative mutation in all the 30 families including 9 novel mutations, which were identified in 9 different families (GJB2 (c.598G>A, p.Gly200Arg); MYO15A (c.9948G>A, p.Gln3316Gln; c.3866+1G>A; c.8767C>T, p.Arg2923* and c.8222T>C, p.Phe2741Ser), TMC1 (c.362+18A>G), BSND (c.97G>C, p.Val33Leu), TMPRSS3 (c.726C>G, p.Cys242Trp) and MSRB3 (c.20T>G, p.Leu7Arg)). Furthermore, 12 recurrent mutations were detected in 21 other families. The 21 identified mutations included 10 (48%) missense changes, 4 (19%) nonsense mutations, 3 (14%) intronic mutations, 2 (9%) splice site mutations and 2 (9%) frameshift mutations. GJB2 accounted for 53% of the families, while mutations in MYO15A were the second most frequent (13%) cause of ARNSHL in these 30 families. The identification of novel as well as recurrent mutations in the present study increases the spectrum of mutations in known deafness genes which could lead to the identification of novel founder mutations and population specific mutated deafness genes causative of ARNSHL. These results provide detailed genetic information that has potential diagnostic implication in the establishment of cost-efficient allele-specific analysis of frequently occurring variants in combination with other reported mutations in Pakistani populations.


Assuntos
Genes Recessivos/genética , Genômica , Perda Auditiva/genética , Linhagem , Sequência de Bases , Conexina 26 , Conexinas/genética , Análise Mutacional de DNA , Feminino , Homozigoto , Humanos , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/genética , Modelos Moleculares , Dados de Sequência Molecular , Miosinas/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Paquistão , Conformação Proteica , Serina Endopeptidases/química , Serina Endopeptidases/genética
15.
Ear Hear ; 35(3): e84-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24378291

RESUMO

OBJECTIVES: Recently, OTOG and OTOGL were identified as human deafness genes. Currently, only four families are known to have autosomal recessive hearing loss based on mutations in these genes. Because the two genes code for proteins (otogelin and otogelin-like) that are strikingly similar in structure and localization in the inner ear, this study is focused on characterizing and comparing the hearing loss caused by mutations in these genes. DESIGN: To evaluate this type of hearing, an extensive set of audiometric and vestibular examinations was performed in the 13 patients from four families. RESULTS: All families show a flat to downsloping configuration of the audiogram with mild to moderate sensorineural hearing loss. Speech recognition scores remain good (>90%). Hearing loss is not significantly different in the four families and the psychophysical test results also do not differ among the families. Vestibular examinations show evidence for vestibular hyporeflexia. CONCLUSION: Because otogelin and otogelin-like are localized in the tectorial membrane, one could expect a cochlear conductive hearing loss, as was previously shown in DFNA13 (COL11A2) and DFNA8/12 (TECTA) patients. Results of psychophysical examinations, however, do not support this. Furthermore, the authors conclude that there are no phenotypic differences between hearing loss based on mutations in OTOG or OTOGL. This phenotype description will facilitate counseling of hearing loss caused by defects in either of these two genes.


Assuntos
Perda Auditiva Neurossensorial/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Emissões Otoacústicas Espontâneas/genética , Reflexo Anormal/genética , Reflexo Vestíbulo-Ocular/genética , Adolescente , Adulto , Audiometria de Tons Puros , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Masculino , Mutação , Fenótipo , Reflexo Acústico/genética , Teste do Limiar de Recepção da Fala , Testes de Função Vestibular , Adulto Jovem
16.
J Hum Genet ; 58(12): 819-21, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24172246

RESUMO

Bjørnstad syndrome is an extremely rare condition characterized by pilitorti and nerve deafness. Only few large families have been reported worldwide. Here we describe a large Pakistani family with five affected individuals. The hair fibers of all the patients were twisted around their axis and devoid of any pigment. In addition the patients had a moderate-to-severe degree of hearing impairment. Genotyping with high-density single-nucleotide polymorphism arrays showed homozygosity in two intervals on chromosome 2. Linkage with one of these regions (genomic position 218745685-221025443, hg19) was confirmed. This region encompasses the BCS1L gene. Mutations in this gene have previously been associated with Bjørnstad's syndrome. We sequenced the BCS1L gene for identification of the causative mutation in the family. A novel homozygous missense mutation c.901T>A was identified, which segregated with the disease in the family. This mutation results in the amino acid change p.Tyr301Asn and was predicted to be pathogenic by bioinformatics tools.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons/genética , Doenças do Cabelo/genética , Perda Auditiva Neurossensorial/genética , Doenças Mitocondriais/congênito , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , ATPases Associadas a Diversas Atividades Celulares , Criança , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Doenças Mitocondriais/genética , Paquistão
17.
Am J Hum Genet ; 91(5): 872-82, 2012 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-23122586

RESUMO

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Here we present OTOGL mutations, a homozygous one base pair deletion (c.1430 delT) causing a frameshift (p.Val477Glufs(∗)25) in a large consanguineous family and two compound heterozygous mutations, c.547C>T (p.Arg183(∗)) and c.5238+5G>A, in a nonconsanguineous family with moderate nonsyndromic sensorineural hearing loss. OTOGL maps to the DFNB84 locus at 12q21.31 and encodes otogelin-like, which has structural similarities to the epithelial-secreted mucin protein family. We demonstrate that Otogl is expressed in the inner ear of vertebrates with a transcription level that is high in embryonic, lower in neonatal, and much lower in adult stages. Otogelin-like is localized to the acellular membranes of the cochlea and the vestibular system and to a variety of inner ear cells located underneath these membranes. Knocking down of otogl with morpholinos in zebrafish leads to sensorineural hearing loss and anatomical changes in the inner ear, supporting that otogelin-like is essential for normal inner ear function. We propose that OTOGL mutations affect the production and/or function of acellular structures of the inner ear, which ultimately leads to sensorineural hearing loss.


Assuntos
Perda Auditiva Neurossensorial/genética , Proteínas de Membrana/genética , Mutação , Adolescente , Animais , Pré-Escolar , Aberrações Cromossômicas , Cóclea/metabolismo , Cóclea/patologia , Exoma , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Perda Auditiva Neurossensorial/diagnóstico , Humanos , Mutação INDEL , Masculino , Camundongos , Polimorfismo de Nucleotídeo Único , Ratos , Peixe-Zebra
18.
Am J Hum Genet ; 91(5): 883-9, 2012 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-23122587

RESUMO

Already 40 genes have been identified for autosomal-recessive nonsyndromic hearing impairment (arNSHI); however, many more genes are still to be identified. In a Dutch family segregating arNSHI, homozygosity mapping revealed a 2.4 Mb homozygous region on chromosome 11 in p15.1-15.2, which partially overlapped with the previously described DFNB18 locus. However, no putative pathogenic variants were found in USH1C, the gene mutated in DFNB18 hearing impairment. The homozygous region contained 12 additional annotated genes including OTOG, the gene encoding otogelin, a component of the tectorial membrane. It is thought that otogelin contributes to the stability and strength of this membrane through interaction or stabilization of its constituent fibers. The murine orthologous gene was already known to cause hearing loss when defective. Analysis of OTOG in the Dutch family revealed a homozygous 1 bp deletion, c.5508delC, which leads to a shift in the reading frame and a premature stop codon, p.Ala1838ProfsX31. Further screening of 60 unrelated probands from Spanish arNSHI families detected compound heterozygous OTOG mutations in one family, c.6347C>T (p.Pro2116Leu) and c. 6559C>T (p.Arg2187X). The missense mutation p.Pro2116Leu affects a highly conserved residue in the fourth von Willebrand factor type D domain of otogelin. The subjects with OTOG mutations have a moderate hearing impairment, which can be associated with vestibular dysfunction. The flat to shallow "U" or slightly downsloping shaped audiograms closely resembled audiograms of individuals with recessive mutations in the gene encoding α-tectorin, another component of the tectorial membrane. This distinctive phenotype may represent a clue to orientate the molecular diagnosis.


Assuntos
Genes Recessivos , Perda Auditiva Neurossensorial/genética , Glicoproteínas de Membrana/genética , Mutação , Homozigoto , Humanos , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Irmãos
19.
Nat Genet ; 44(5): 581-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22522421

RESUMO

Walker-Warburg syndrome (WWS) is an autosomal recessive multisystem disorder characterized by complex eye and brain abnormalities with congenital muscular dystrophy (CMD) and aberrant a-dystroglycan glycosylation. Here we report mutations in the ISPD gene (encoding isoprenoid synthase domain containing) as the second most common cause of WWS. Bacterial IspD is a nucleotidyl transferase belonging to a large glycosyltransferase family, but the role of the orthologous protein in chordates is obscure to date, as this phylum does not have the corresponding non-mevalonate isoprenoid biosynthesis pathway. Knockdown of ispd in zebrafish recapitulates the human WWS phenotype with hydrocephalus, reduced eye size, muscle degeneration and hypoglycosylated a-dystroglycan. These results implicate ISPD in a-dystroglycan glycosylation in maintaining sarcolemma integrity in vertebrates.


Assuntos
Distroglicanas/metabolismo , Mutação/genética , Síndrome de Walker-Warburg/genética , Peixe-Zebra/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Pré-Escolar , Embrião não Mamífero , Olho/metabolismo , Olho/patologia , Glicosilação , Humanos , Manosiltransferases/genética , Manosiltransferases/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Peixe-Zebra/embriologia
20.
J Assoc Res Otolaryngol ; 12(6): 753-66, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21786053

RESUMO

In the present study, genotype-phenotype correlations in eight Dutch DFNB8/10 families with compound heterozygous mutations in TMPRSS3 were addressed. We compared the phenotypes of the families by focusing on the mutation data. The compound heterozygous variants in the TMPRSS3 gene in the present families included one novel variant, p.Val199Met, and four previously described pathogenic variants, p.Ala306Thr, p.Thr70fs, p.Ala138Glu, and p.Cys107Xfs. In addition, the p.Ala426Thr variant, which had previously been reported as a possible polymorphism, was found in one family. All affected family members reported progressive bilateral hearing impairment, with variable onset ages and progression rates. In general, the hearing impairment affected the high frequencies first, and sooner or later, depending on the mutation, the low frequencies started to deteriorate, which eventually resulted in a flat audiogram configuration. The ski-slope audiogram configuration is suggestive for the involvement of TMPRSS3. Our data suggest that not only the protein truncating mutation p.T70fs has a severe effect but also the amino acid substitutions p.Ala306Thr and p.Val199Met. A combination of two of these three mutations causes prelingual profound hearing impairment. However, in combination with the p.Ala426Thr or p.Ala138Glu mutations, a milder phenotype with postlingual onset of the hearing impairment is seen. Therefore, the latter mutations are likely to be less detrimental for protein function. Further studies are needed to distinguish possible phenotypic differences between different TMPRSS3 mutations. Evaluation of performance of patients with a cochlear implant indicated that this is a good treatment option for patients with TMPRSS3 mutations as satisfactory speech reception was reached after implantation.


Assuntos
Estudos de Associação Genética , Perda Auditiva Bilateral/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Serina Endopeptidases/química , Serina Endopeptidases/genética , Adolescente , Adulto , Substituição de Aminoácidos/genética , Audiometria de Tons Puros , Audiometria da Fala , Criança , Pré-Escolar , Implante Coclear , Saúde da Família , Feminino , Ligação Genética , Perda Auditiva Bilateral/diagnóstico , Perda Auditiva Bilateral/terapia , Humanos , Lactente , Masculino , Linhagem , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...