Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Ear Hear ; 45(1): 250-256, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37677959

RESUMEN

OBJECTIVES: Attenuation of otoacoustic emissions over time has been reported for many patients with hearing impairment harboring mutations in the OTOF gene. In this study, the time course of changes of distortion product otoacoustic emissions (DPOAEs) has been analyzed in a cohort of patients in the light of tympanometry results. DESIGN: The changes of DPOAEs in 16 patients with OTOF -related hearing impairment were retrospectively analyzed. RESULTS: All but one subject showed DPOAEs bilaterally at the time of diagnosis. Three patients diagnosed as adults still had DPOAEs at ages of 27, 31, and 47 years, respectively. Follow-up was available for 7 children diagnosed at the age of 1 to 3 years, who still showed preservation of DPOAEs at ages of 5 to 16 years. The responses were absent or attenuated in amplitude at some follow-up appointments in association with type B or C tympanograms. CONCLUSIONS: DPOAEs are preserved much longer than expected in a cohort of patients with OTOF -related hearing impairment. The previously reported loss of DPOAEs may have been caused in some children by increased middle ear impedance due to otitis media.


Asunto(s)
Pérdida Auditiva , Adulto , Niño , Humanos , Lactante , Preescolar , Estudios Retrospectivos , Pérdida Auditiva/diagnóstico , Emisiones Otoacústicas Espontáneas/fisiología , Pruebas de Impedancia Acústica , Oído Medio , Audiometría de Tonos Puros , Umbral Auditivo/fisiología , Proteínas de la Membrana
2.
Genes (Basel) ; 13(1)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35052489

RESUMEN

Pathogenic variants in the PJVK gene cause the DFNB59 type of autosomal recessive non-syndromic hearing impairment (AR-NSHI). Phenotypes are not homogeneous, as a few subjects show auditory neuropathy spectrum disorder (ANSD), while others show cochlear hearing loss. The numbers of reported cases and pathogenic variants are still small to establish accurate genotype-phenotype correlations. We investigated a cohort of 77 Spanish familial cases of AR-NSHI, in whom DFNB1 had been excluded, and a cohort of 84 simplex cases with isolated ANSD in whom OTOF variants had been excluded. All seven exons and exon-intron boundaries of the PJVK gene were sequenced. We report three novel DFNB59 cases, one from the AR-NSHI cohort and two from the ANSD cohort, with stable, severe to profound NSHI. Two of the subjects received unilateral cochlear implantation, with apparent good outcomes. Our study expands the spectrum of PJVK mutations, as we report four novel pathogenic variants: p.Leu224Arg, p.His294Ilefs*43, p.His294Asp and p.Phe317Serfs*20. We review the reported cases of DFNB59, summarize the clinical features of this rare subtype of AR-NSHI and discuss the involvement of PJVK in ANSD.


Asunto(s)
Pérdida Auditiva Central/patología , Pérdida Auditiva/patología , Mutación , Proteínas del Tejido Nervioso/genética , Adolescente , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Pérdida Auditiva/complicaciones , Pérdida Auditiva/genética , Pérdida Auditiva Central/complicaciones , Pérdida Auditiva Central/genética , Humanos , Lactante , Masculino , Linaje
3.
Audiol Res ; 11(4): 639-652, 2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34940017

RESUMEN

Auditory Neuropathy (AN) is characterized by disruption of temporal coding of acoustic signals in auditory nerve fibers resulting in alterations of auditory perceptions. Mutations in several genes have been associated to the most forms of AN. Underlying mechanisms include both pre-synaptic and post-synaptic damage involving inner hair cell (IHC) depolarization, neurotransmitter release, spike initiation in auditory nerve terminals, loss of auditory fibers and impaired conduction. In contrast, outer hair cell (OHC) activities (otoacoustic emissions [OAEs] and cochlear microphonic [CM]) are normal. Disordered synchrony of auditory nerve activity has been suggested as the basis of both the alterations of auditory brainstem responses (ABRs) and reduction of speech perception. We will review how electrocochleography (ECochG) recordings provide detailed information to help objectively define the sites of auditory neural dysfunction and their effect on receptor summating potential (SP) and neural compound action potential (CAP), the latter reflecting disorders of ribbon synapses and auditory nerve fibers.

4.
Int J Pediatr Otorhinolaryngol ; 149: 110876, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34385039

RESUMEN

BACKGROUND: Little is known regarding fitting parameters and receptive and expressive language development in cochlear-implanted children (CCI) with profound sensorineural hearing loss (SHL) who are diagnosed with Autism Spectrum Disorder (ASD). The aim of the study was to evaluate a group of ASD CCI users in order to describe their ASD clinical features and CCI outcomes; report on the average electrical charge requirements; and evaluate the possible correlations between electrical and psychophysical outcomes with ASD characteristics. MATERIALS AND METHODS: A multicentre observational study of 22 ASD children implanted in four cochlear implant (CI) centers. Data concerning profound SHL diagnosis, ASD diagnosis, CI timing and CI compliance were collected. Sound Field (SF) was assessed through repeated behavioural measurements. Categories of Auditory Perception (CAP) and Categories of Language (CL) were used to evaluate speech perception and language skills at short (≤2 yrs), medium (5 yrs) and long term (>10 yrs) follow-up. Fitting parameters such as comfortable thresholds, pulse-width (pw, µsec) and clinical units converted into units of charge/phase were collected. The diagnosis of ASD was acquired by the referral neuropsychiatric department and severity was assessed through the Diagnostic and Statistical Manual of Mental Disorders (DSM-V) and the Childhood Autism Rating Scale (CARS). RESULTS: At the final follow-up session the median SF threshold for CI outcomes was 30 dB HL (min 15 - max 60). CAP score was extremely variable: 45.5% showed no improvement over time and only 22% of children reached CAP scores of 5-7. CL 45.5% showed no improvement over time and score was 1-2 in the majority of ASD children (72.7%), while only 18.2% reached the highest level of language skills. There were no statistically significant differences at each follow-up between subjects with or without comorbidities. CAP and CL were inversely correlated with DSM-V A and B domains, corresponding to lower speech and language scores in children with more severe ASD symptoms, and maintained their correlation at mid and long follow-ups whilst controlling for age at CI. Electrical charge requirements did not correlate with SF or age at implant but did inversely correlate with ASD severity. With regards to CI compliance: only 13.6% children (3) with severe DSM-V A/B levels and CARS score were partial/intermittent users. CONCLUSION: The present study is a targeted contribution to the current literature to support clinical procedures for CI fitting and audiological follow-up in children with ASD. The findings indicate that the outcomes of CI use and the fitting procedures are both influenced by the severity of the ASD symptoms rather than the demographic variables or associated disorders.


Asunto(s)
Trastorno del Espectro Autista , Implantación Coclear , Implantes Cocleares , Sordera , Percepción del Habla , Trastorno del Espectro Autista/complicaciones , Trastorno del Espectro Autista/diagnóstico , Niño , Sordera/cirugía , Humanos
5.
Ear Hear ; 42(6): 1627-1639, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33908410

RESUMEN

OBJECTIVES: Congenital profound hearing loss with preserved cochlear outer hair cell activity (otoacoustic emissions and cochlear microphonic) is the most common phenotype associated with mutations in the OTOF gene. The aim of this study was to investigate the pathophysiological mechanisms behind the auditory dysfunction in five patients (2 adults and 3 children) carrying biallelic mutations in OTOF, who showed an uncommon phenotype of mild hearing impairment associated with severe difficulties in speech perception and delay of language development. DESIGN: Patients underwent audiometric assessment with pure-tone and speech perception evaluation, and otoacoustic emissions and auditory brainstem response recording. Cochlear potentials were recorded in all subjects through transtympanic electrocochleography in response to clicks delivered in the free field from 120 to 60 dB peak equivalent SPL and were compared to recordings obtained from 20 normally hearing controls and from eight children with profound deafness due to mutations in the OTOF gene. Three patients out of five underwent unilateral cochlear implantation. Speech perception measures and electrically evoked auditory nerve potentials were obtained within 1 year of cochlear implant use. RESULTS: Pathogenic mutations in the two alleles of OTOF were found in all five patients, and five novel mutations were identified. Hearing thresholds indicated mild hearing loss in four patients and moderate hearing loss in one. Distortion product otoacoustic emissions were recorded in all subjects, whereas auditory brainstem responses were absent in all but two patients, who showed a delayed wave V in one ear. In electrocochleography recordings, cochlear microphonics and summating potentials showed normal latency and peak amplitude, consistently with preservation of both outer and inner hair cell activity. In contrast, the neural compound action potential recorded in normally hearing controls was replaced by a prolonged, low-amplitude negative response. No differences in cochlear potentials were found between OTOF subjects showing mild or profound hearing loss. Electrical stimulation through the cochlear implant improved speech perception and restored synchronized auditory nerve responses in all cochlear implant recipients. CONCLUSIONS: These findings indicate that disordered synchrony in auditory fiber activity underlies the impairment of speech perception in patients carrying biallelic mutations in OTOF gene who show a stable phenotype of mild hearing loss. Abnormal nerve synchrony with preservation of hearing sensitivity is consistent with selective impairment of vesicle replenishment at the ribbon synapses with relative preservation of synaptic exocytosis. Cochlear implants are effective in restoring speech perception and synchronous activation of the auditory pathway by directly stimulating auditory fibers.


Asunto(s)
Pérdida Auditiva , Proteínas de la Membrana , Percepción del Habla , Umbral Auditivo/fisiología , Cóclea , Nervio Coclear , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Humanos , Proteínas de la Membrana/genética , Mutación , Emisiones Otoacústicas Espontáneas/fisiología , Percepción del Habla/fisiología
7.
J Clin Med ; 9(6)2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32517138

RESUMEN

In recent years there has been an increasing percentage of cochlear implant (CI) users who have usable residual hearing in the contralateral, nonimplanted ear, typically aided by acoustic amplification. This raises the issue of the extent to which the signal presented through the cochlear implant may influence how listeners process information in the acoustically stimulated ear. This multicenter retrospective study examined pre- to postoperative changes in speech perception in the nonimplanted ear, the implanted ear, and both together. Results in the latter two conditions showed the expected increases, but speech perception in the nonimplanted ear showed a modest yet meaningful decrease that could not be completely explained by changes in unaided thresholds, hearing aid malfunction, or several other demographic variables. Decreases in speech perception in the nonimplanted ear were more likely in individuals who had better levels of speech perception in the implanted ear, and in those who had better speech perception in the implanted than in the nonimplanted ear. This raises the possibility that, in some cases, bimodal listeners may rely on the higher quality signal provided by the implant and may disregard or even neglect the input provided by the nonimplanted ear.

8.
J Hum Genet ; 65(10): 855-864, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32467589

RESUMEN

Non-syndromic hearing loss (NSHL) is characterized by a vast genetic heterogeneity; some syndromic forms as Usher syndrome (USH) have onset as isolated deafness and then evolve later in life. We developed an NGS targeted gene-panel containing 59 genes and a customized bioinformatic pipeline for the analysis of DNA samples from clinically highly selected subjects with sensorineural hearing loss, previously resulted negative for GJB2 mutations/GJB6 deletions. Among the 217 tested subjects, 24 (11.1%) were found to carry mutations in genes involved both in NSHL and USH. For 6 out of 24 patients a diagnosis of USH was performed. Eleven subjects out of 24 had hearing loss without vestibular or ocular dysfunction and, due to their young age, it was not possible to establish whether their phenotype could be NSHL or USH. Seven subjects were diagnosed with NSHL, due to their age and phenotype. A total of 41 likely pathogenic/pathogenic mutations were identified, among which 17 novel ones. We report a high frequency of mutations in genes involved both in NSHL and in USH in a cohort of individuals tested for seemingly isolated deafness. Our data also highlight a wider than expected phenotypic variability in the USH phenotype.


Asunto(s)
Sordera/genética , Heterogeneidad Genética , Mutación , Síndromes de Usher/genética , Adolescente , Adulto , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Niño , Preescolar , Conexina 26/genética , Conexina 30/genética , Femenino , Estudios de Asociación Genética , Pérdida Auditiva Sensorineural/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Fenotipo , Adulto Joven
9.
Front Neurosci ; 13: 501, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191217

RESUMEN

Hidden auditory neuropathy is characterized by reduced performances in challenging auditory tasks with the preservation of hearing thresholds, resulting from diffuse loss of cochlear inner hair cell (IHC) synapses following primary degeneration of unmyelinated terminals of auditory fibers. We report the audiological and electrophysiological findings collected from 10 members (4 males, 6 females) of a large Italian family affected by dominant optic atrophy, associated with the OPA8 locus, who complained of difficulties in understanding speech in the presence of noise. The patients were pooled into two groups, one consisting of 4 young adults (19-50 years) with normal hearing thresholds, and the other made up of 6 patients of an older age (55-72 years) showing mild hearing loss. Speech perception scores were normal in the first group and decreased in the second. Otoacoustic emissions (OAEs) and cochlear microphonics (CMs) recordings were consistent with preservation of outer hair cell (OHC) function in all patients, whereas auditory brainstem responses (ABRs) showed attenuated amplitudes in the first group and severe abnormalities in the second. Middle ear acoustic reflexes had delayed peak latencies in all patients in comparison with normally hearing individuals. Transtympanic electrocochleography (ECochG) recordings in response to 0.1 ms clicks at intensities from 120 to 60 dB peak equivalent SPL showed a reduction in amplitude of both summating potential (SP) and compound action potential (CAP) together with delayed CAP peak latencies and prolonged CAP duration in all patients in comparison with a control group of 20 normally hearing individuals. These findings indicate that underlying the hearing impairment in OPA8 patients is hidden AN resulting from diffuse loss of IHCs synapses. At an early stage the functional alterations only consist of abnormalities of ABR and ECochG potentials with increased latencies of acoustic reflexes, whereas reduction in speech perception scores become apparent with progression of the disease. Central mechanisms increasing the cortical gain are likely to compensate for the reduction of cochlear input.

10.
Int J Pediatr Otorhinolaryngol ; 104: 88-93, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29287889

RESUMEN

OBJECTIVES: This work was aimed at establishing the molecular etiology of hearing loss in a 9-year old girl with post-lingual non-syndromic mild sensorineural hearing loss with a complex family history of clinically heterogeneous deafness. METHODS: The proband's DNA was subjected to NGS analysis of a 59-targeted gene panel, with the use of the Ion Torrent PGM platform. Conventional Sanger sequencing was used for segregation analysis in all the affected relatives. The proband and all the other hearing impaired members of the family underwent a thorough clinical and audiological evaluation. RESULTS: A new likely pathogenic mutation in the EYA4 gene (c.1154C > T; p.Ser385Leu) was identified in the proband and in her 42-year-old father with post-lingual non-syndromic profound sensorineural hearing loss. The EYA4 mutation was also found in the proband's grandfather and uncle, both showing clinical features of Waardenburg syndrome type 1. A novel pathogenic splice-site mutation (c.321+1G > A) of the PAX3 gene was found to co-segregate with the EYA4 mutation in these two subjects. CONCLUSION: The identified novel EYA4 mutation can be considered responsible of the hearing loss observed in the proband and her father, while a dual molecular diagnosis was reached in the relatives co-segregating the EYA4 and the PAX3 mutations. In these two subjects the DFNA10 phenotype was masked by Waardenburg syndrome. The use of NGS targeted gene-panel, in combination with an extensive clinical and audiological examination led us to identify the genetic cause of the hearing loss in members of a family in which different forms of autosomal dominant deafness segregate. These results provide precise and especially important prognostic and follow-up information for the future audiologic management in the youngest affected member.


Asunto(s)
Sordera/genética , Pérdida Auditiva Sensorineural/genética , Factor de Transcripción PAX3/genética , Transactivadores/genética , Síndrome de Waardenburg/genética , Adulto , Audiometría , Niño , Familia , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación , Linaje , Fenotipo
12.
Hear Res ; 330(Pt B): 200-12, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26188103

RESUMEN

Mutations in the OTOF gene encoding otoferlin result in a disrupted function of the ribbon synapses with impairment of the multivesicular glutamate release. Most affected subjects present with congenital hearing loss and abnormal auditory brainstem potentials associated with preserved cochlear hair cell activities (otoacoustic emissions, cochlear microphonics [CMs]). Transtympanic electrocochleography (ECochG) has recently been proposed for defining the details of potentials arising in both the cochlea and auditory nerve in this disorder, and with a view to shedding light on the pathophysiological mechanisms underlying auditory dysfunction. We review the audiological and electrophysiological findings in children with congenital profound deafness carrying two mutant alleles of the OTOF gene. We show that cochlear microphonic (CM) amplitude and summating potential (SP) amplitude and latency are normal, consistently with a preserved outer and inner hair cell function. In the majority of OTOF children, the SP component is followed by a markedly prolonged low-amplitude negative potential replacing the compound action potential (CAP) recorded in normally-hearing children. This potential is identified at intensities as low as 90 dB below the behavioral threshold. In some ears, a synchronized CAP is superimposed on the prolonged responses at high intensity. Stimulation at high rates reduces the amplitude and duration of the prolonged potentials, consistently with their neural generation. In some children, however, the ECochG response only consists of the SP, with no prolonged potential. Cochlear implants restore hearing sensitivity, speech perception and neural CAP by electrically stimulating the auditory nerve fibers. These findings indicate that an impaired multivesicular glutamate release in OTOF-related disorders leads to abnormal auditory nerve fiber activation and a consequent impairment of spike generation. The magnitude of these effects seems to vary, ranging from no auditory nerve fiber activation to an abnormal generation of EPSPs that occasionally trigger a synchronized electrical activity, resulting in high-threshold CAPs.


Asunto(s)
Cóclea/inervación , Nervio Coclear/fisiopatología , Señales (Psicología) , Pérdida Auditiva/genética , Audición , Proteínas de la Membrana/genética , Mutación , Percepción del Habla , Transmisión Sináptica , Estimulación Acústica , Animales , Audiometría de Respuesta Evocada , Vías Auditivas/metabolismo , Vías Auditivas/fisiopatología , Umbral Auditivo , Implantación Coclear , Potenciales Microfónicos de la Cóclea , Nervio Coclear/metabolismo , Predisposición Genética a la Enfermedad , Ácido Glutámico/metabolismo , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/metabolismo , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/psicología , Pérdida Auditiva/rehabilitación , Humanos , Percepción Sonora , Proteínas de la Membrana/metabolismo , Personas con Deficiencia Auditiva/psicología , Fenotipo , Tiempo de Reacción , Inteligibilidad del Habla , Factores de Tiempo
13.
Neurodegener Dis ; 15(2): 114-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25791504

RESUMEN

BACKGROUND: Friedreich's ataxia (FRDA) is a degenerative disorder caused by mutations of the FXN gene. Sensorineural hearing loss is one of the clinical features of FRDA, and the majority of hearing-impaired patients have shown evidence of auditory neuropathy. OBJECTIVE: This study characterizes the cochlear receptor and auditory nerve potentials in a patient with FRDA who had the clinical profile of auditory neuropathy. The aim was to investigate the site of the lesion and the pathophysiological mechanisms behind the hearing dysfunction. METHODS: Using transtympanic electrocochleography, both receptor (cochlear microphonic, CM, and summating potential, SP) and auditory nerve potentials were recorded in response to trains of clicks with stimulation intensities from 60 to 120 dB SPL. The results were compared with recordings obtained from two groups of subjects, i.e. 20 normally hearing controls and 19 subjects with cochlear hearing loss. RESULTS: The results showed that the synchronized neural response seen in both normally hearing and hearing-impaired subjects was lacking in our patient, replaced by a prolonged, low-amplitude negative potential that decreased in both amplitude and duration for rapid stimulation rates, consistent with adaptation of neural sources. CMs were recorded with a normal amplitude, consistent with preserved outer hair cell function. SP peak latency was within normal limits, whereas SP amplitude was comparable with that of subjects with cochlear hearing loss, consistent with inner hair cell loss. CONCLUSION: These findings suggest that underlying auditory neuropathy in FRDA is a disordered synchrony in auditory nerve fiber discharge, possibly resulting from auditory nerve fiber degeneration and inner hair cell loss.


Asunto(s)
Nervio Coclear/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Ataxia de Friedreich/patología , Estimulación Acústica , Adulto , Audiometría de Respuesta Evocada , Lateralidad Funcional , Humanos , Masculino , Psicoacústica , Percepción del Habla/fisiología
14.
Brain ; 138(Pt 3): 563-76, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25564500

RESUMEN

Hearing impairment is the second most prevalent clinical feature after optic atrophy in dominant optic atrophy associated with mutations in the OPA1 gene. In this study we characterized the hearing dysfunction in OPA1-linked disorders and provided effective rehabilitative options to improve speech perception. We studied two groups of OPA1 subjects, one comprising 11 patients (seven males; age range 13-79 years) carrying OPA1 mutations inducing haploinsufficiency, the other, 10 subjects (three males; age range 5-58 years) carrying OPA1 missense mutations. Both groups underwent audiometric assessment with pure tone and speech perception evaluation, and otoacoustic emissions and auditory brainstem response recording. Cochlear potentials were recorded through transtympanic electrocochleography from the group of patients harbouring OPA1 missense mutations and were compared to recordings obtained from 20 control subjects with normal hearing and from 19 subjects with cochlear hearing loss. Eight patients carrying OPA1 missense mutations underwent cochlear implantation. Speech perception measures and electrically-evoked auditory nerve and brainstem responses were obtained after 1 year of cochlear implant use. Nine of 11 patients carrying OPA1 mutations inducing haploinsufficiency had normal hearing function. In contrast, all but one subject harbouring OPA1 missense mutations displayed impaired speech perception, abnormal brainstem responses and presence of otoacoustic emissions consistent with auditory neuropathy. In electrocochleography recordings, cochlear microphonic had enhanced amplitudes while summating potential showed normal latency and peak amplitude consistent with preservation of both outer and inner hair cell activities. After cancelling the cochlear microphonic, the synchronized neural response seen in both normally-hearing controls and subjects with cochlear hearing loss was replaced by a prolonged, low-amplitude negative potential that decreased in both amplitude and duration during rapid stimulation consistent with neural generation. The use of cochlear implant improved speech perception in all but one patient. Brainstem potentials were recorded in response to electrical stimulation in five of six subjects, whereas no compound action potential was evoked from the auditory nerve through the cochlear implant. These findings indicate that underlying the hearing impairment in patients carrying OPA1 missense mutations is a disordered synchrony in auditory nerve fibre activity resulting from neural degeneration affecting the terminal dendrites. Cochlear implantation improves speech perception and synchronous activation of auditory pathways by bypassing the site of lesion.


Asunto(s)
Implantación Coclear , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , GTP Fosfohidrolasas/genética , Pérdida Auditiva Central , Mutación/genética , Adolescente , Adulto , Anciano , Análisis de Varianza , Audiometría de Respuesta Evocada , Umbral Auditivo/fisiología , Niño , Preescolar , Femenino , Pérdida Auditiva Central/genética , Pérdida Auditiva Central/fisiopatología , Pérdida Auditiva Central/terapia , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Emisiones Otoacústicas Espontáneas , Percepción del Habla/fisiología , Tomógrafos Computarizados por Rayos X , Resultado del Tratamiento , Adulto Joven
15.
Proc Natl Acad Sci U S A ; 109(35): 14013-8, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891314

RESUMEN

Phosphatidylinositol phosphate kinase type 1γ (PIPKIγ) is a key enzyme in the generation of phosphatidylinositol 4,5-bisphosphate [PI(4,5)P(2)] and is expressed at high levels in the nervous system. Homozygous knockout mice lacking this enzyme die postnatally within 24 h, whereas PIPKIγ(+/-) siblings breed normally and have no reported phenotype. Here we show that adult PIPKIγ(+/-) mice have dramatically elevated hearing thresholds for high-frequency sounds. During the first postnatal week we observed a reduction of ATP-dependent Ca(2+) signaling activity in cochlear nonsensory cells. Because Ca(2+) signaling under these conditions depends on inositol-1,4,5-trisphosphate generation from phospholipase C (PLC)-dependent hydrolysis of PI(4,5)P(2), we conclude that (i) PIPKIγ is primarily responsible for the synthesis of the receptor-regulated PLC-sensitive PI(4,5)P(2) pool in the cell syncytia that supports auditory hair cells; (ii) spatially graded impairment of this signaling pathway in cochlear nonsensory cells causes a selective alteration in the acquisition of hearing in PIPKIγ(+/-) mice. This mouse model also suggests that PIPKIγ may determine the level of gap junction contribution to cochlear development.


Asunto(s)
Señalización del Calcio/fisiología , Sordera/genética , Sordera/metabolismo , Órgano Espiral/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Factores de Edad , Animales , Animales Recién Nacidos , Conexinas/genética , Conexinas/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Uniones Comunicantes/metabolismo , Células Ciliadas Auditivas/metabolismo , Audición/fisiología , Mecanotransducción Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Órgano Espiral/crecimiento & desarrollo , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Percepción de la Altura Tonal/fisiología
16.
PLoS One ; 6(8): e23279, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21876744

RESUMEN

The deafness locus DFNB1 contains GJB2, the gene encoding connexin26 and GJB6, encoding connexin30, which appear to be coordinately regulated in the inner ear. In this work, we investigated the expression and function of connexin26 and connexin30 from postnatal day 5 to adult age in double transgenic Cx26(Sox10Cre) mice, which we obtained by crossing connexin26 floxed mice with a deleter Sox10-Cre line. Cx26(Sox10Cre) mice presented with complete connexin26 ablation in the epithelial gap junction network of the cochlea, whereas connexin30 expression was developmentally delayed; immunolabeling patterns for both connexins were normal in the cochlear lateral wall. In vivo electrophysiological measurements in Cx26(Sox10Cre) mice revealed profound hearing loss accompanied by reduction of endocochlear potential, and functional experiments performed in postnatal cochlear organotypic cultures showed impaired gap junction coupling. Transduction of these cultures with a bovine adeno associated virus vector restored connexin26 protein expression and rescued gap junction coupling. These results suggest that restoration of normal connexin levels by gene delivery via recombinant adeno associated virus could be a way to rescue hearing function in DFNB1 mouse models and, in future, lead to the development of therapeutic interventions in humans.


Asunto(s)
Cóclea/patología , Conexinas/genética , Conexinas/uso terapéutico , Sordera/terapia , Dependovirus/metabolismo , Uniones Comunicantes/metabolismo , Terapia Genética , Animales , Bovinos , Cóclea/fisiopatología , Conexina 26 , Sordera/patología , Sordera/fisiopatología , Fenómenos Electrofisiológicos , Recuperación de Fluorescencia tras Fotoblanqueo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Integrasas/metabolismo , Ratones , Técnicas de Cultivo de Órganos , Órgano Espiral/patología , Permeabilidad , Proteínas Recombinantes de Fusión , Factores de Transcripción SOXE/metabolismo , Factores de Tiempo , Transducción Genética
17.
Hum Mol Genet ; 19(24): 4759-73, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20858605

RESUMEN

Mutations in the GJB2 and GJB6 genes, respectively, coding for connexin26 (Cx26) and connexin30 (Cx30) proteins, are the most common cause for prelingual non-syndromic deafness in humans. In the inner ear, Cx26 and Cx30 are expressed in different non-sensory cell types, where they largely co-localize and may form heteromeric gap junction channels. Here, we describe the generation and characterization of a mouse model for human bilateral middle/high-frequency hearing loss based on the substitution of an evolutionarily conserved threonine by a methionine residue at position 5 near the N-terminus of Cx30 (Cx30T5M). The mutation was inserted in the mouse genome by homologous recombination in mouse embryonic stem cells. Expression of the mutated Cx30T5M protein in these transgenic mice is under the control of the endogenous Cx30 promoter and was analysed via activation of the lacZ reporter gene. When probed by auditory brainstem recordings, Cx30(T5M/T5M) mice exhibited a mild, but significant increase in their hearing thresholds of about 15 dB at all frequencies. Immunolabelling with antibodies to Cx26 or Cx30 suggested normal location of these proteins in the adult inner ear, but western blot analysis showed significantly down-regulated the expression levels of Cx26 and Cx30. In the developing cochlea, electrical coupling, probed by dual patch-clamp recordings, was normal. However, transfer of the fluorescent tracer calcein between cochlear non-sensory cells was reduced, as was intercellular Ca(2+) signalling due to spontaneous ATP release from connexin hemichannels. Our findings link hearing loss to decreased biochemical coupling due to the point-mutated Cx30 in mice.


Asunto(s)
Cóclea/patología , Cóclea/fisiopatología , Conexinas/genética , Sordera/genética , Pérdida Auditiva Bilateral/genética , Mutación/genética , Adenosina Trifosfato/metabolismo , Envejecimiento/patología , Animales , Señalización del Calcio , Cóclea/crecimiento & desarrollo , Conexina 26 , Conexina 30 , Sordera/complicaciones , Sordera/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Recuperación de Fluorescencia tras Fotoblanqueo , Técnicas de Sustitución del Gen , Pérdida Auditiva Bilateral/complicaciones , Pérdida Auditiva Bilateral/fisiopatología , Humanos , Immunoblotting , Ratones , Órgano Espiral/metabolismo , Órgano Espiral/patología , Órgano Espiral/fisiopatología , Permeabilidad , Recombinación Genética/genética
18.
J Biol Chem ; 285(48): 37693-703, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20826782

RESUMEN

The mechanotransduction process in hair cells in the inner ear is associated with the influx of calcium from the endolymph. Calcium is exported back to the endolymph via the splice variant w/a of the PMCA2 of the stereocilia membrane. To further investigate the role of the pump, we have identified and characterized a novel ENU-induced mouse mutation, Tommy, in the PMCA2 gene. The mutation causes a non-conservative E629K change in the second intracellular loop of the pump that harbors the active site. Tommy mice show profound hearing impairment from P18, with significant differences in hearing thresholds between wild type and heterozygotes. Expression of mutant PMCA2 in CHO cells shows calcium extrusion impairment; specifically, the long term, non-stimulated calcium extrusion activity of the pump is inhibited. Calcium extrusion was investigated directly in neonatal organotypic cultures of the utricle sensory epithelium in Tommy mice. Confocal imaging combined with flash photolysis of caged calcium showed impairment of calcium export in both Tommy heterozygotes and homozygotes. Immunofluorescence studies of the organ of Corti in homozygous Tommy mice showed a progressive base to apex degeneration of hair cells after P40. Our results on the Tommy mutation along with previously observed interactions between cadherin-23 and PMCA2 mutations in mouse and humans underline the importance of maintaining the appropriate calcium concentrations in the endolymph to control the rigidity of cadherin and ensure the function of interstereocilia links, including tip links, of the stereocilia bundle.


Asunto(s)
Calcio/metabolismo , Citosol/metabolismo , Sordera/genética , Sordera/metabolismo , Células Ciliadas Auditivas/metabolismo , Mutación Missense , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Secuencia de Aminoácidos , Animales , Citosol/química , Modelos Animales de Enfermedad , Oído Interno/metabolismo , Células Ciliadas Auditivas/química , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Datos de Secuencia Molecular , ATPasas Transportadoras de Calcio de la Membrana Plasmática/química , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Alineación de Secuencia
19.
J Assoc Res Otolaryngol ; 10(4): 545-56, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19636622

RESUMEN

Otoferlin is involved in neurotransmitter release at the synapse between inner hair cells (IHCs) and auditory nerve fibres, and mutations in the OTOF gene result in severe to profound hearing loss. Abnormal sound-evoked cochlear potentials were recorded with transtympanic electrocochleography from four children with otoferlin (OTOF) mutations to evaluate physiological effects in humans of abnormal neurotransmitter release from IHCs. The subjects were profoundly deaf with absent auditory brainstem responses and preserved otoacoustic emissions consistent with auditory neuropathy. Two children were compound heterozygotes for mutations c.2732_2735dupAGCT and p.Ala964Glu; one subject was homozygous for mutation p.Phe1795Cys, and one was compound heterozygote for two novel mutations c.1609delG in exon 16 and c.1966delC in exon 18. Cochlear potentials evoked by clicks from 60 to 120 dB peak equivalent sound pressure level were compared to recordings obtained from 16 normally hearing children. Cochlear microphonic (CM) was recorded with normal amplitudes from all but one ear. After cancelling CM, cochlear potentials were of negative polarity with reduced amplitude and prolonged duration compared to controls. These cochlear potentials were recorded as low as 50-90 dB below behavioural thresholds in contrast to the close correlation in controls between cochlear potentials and behavioural threshold. Summating potential was identified in five out of eight ears with normal latency whilst auditory nerve compound action potentials were either absent or of low amplitude. Stimulation at high rates reduced amplitude and duration of the prolonged potentials, consistent with neural generation. This study suggests that mechano-electrical transduction and cochlear amplification are normal in patients with OTOF mutations. The low-amplitude prolonged negative potentials are consistent with decreased neurotransmitter release resulting in abnormal dendritic activation and impairment of auditory nerve firing.


Asunto(s)
Potenciales Microfónicos de la Cóclea , Sordera/genética , Sordera/fisiopatología , Proteínas de la Membrana/genética , Mutación , Estimulación Acústica , Audiometría de Tonos Puros , Umbral Auditivo , Nervio Coclear/metabolismo , Nervio Coclear/fisiopatología , Sordera/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Lactante , Masculino , Mecanotransducción Celular
20.
Eur Arch Otorhinolaryngol ; 265(1): 43-51, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17701047

RESUMEN

We recorded cochlear potentials by transtympanic electrocochleography (ECochG) in three hearing-impaired children with GJB2 mutation who showed otoacoustic emissions. Pure tone thresholds, distortion product otoacoustic emissions (DPOAEs) and, auditory brainstem responses (ABRs) were also obtained. Subjects 1 (35delG/35delG) and 3 (M34T/wt) had profound hearing loss and showed the picture of auditory neuropathy (AN) as DPOAEs were detected with absent ABRs in both ears. The hearing impairment found in subject 2 (35delG/35delG) was profound in the right ear and moderate in the left ear. Both DPOAEs and ABRs with normal latencies and morphology were recorded only from the left ear. On the ECochG recording the cochlear microphonic was obtained from all children. No compound action potential (CAP) was detected in subject 1. A neural response was recorded only from the left ear in subject 2 with a threshold corresponding to the audiometric threshold while no CAP was detected on the right side. The ECochG obtained from subject 3 showed a low-amplitude broad negative deflection which was identifiable down to low stimulus levels. This response decreased in amplitude and duration when utilizing a high-rate stimulation paradigm. The amount of amplitude reduction was close to that calculated for normal ears, thus revealing the presence of an adapting neural component. These findings indicate that patients with GJB2 mutations and preserved outer hair cells function could present with the picture of AN. The hearing impairment is underlain by a selective inner hair cell loss or a lesion involving the synapses and/or the auditory nerve terminals. We suggest that neonatal hyperbilirubinemia may play a role in protecting outer hair cells against the damage induced by GJB2 mutations.


Asunto(s)
Conexinas/genética , Pérdida Auditiva Sensorineural/fisiopatología , Mutación , Emisiones Otoacústicas Espontáneas , Potenciales de Acción , Audiometría de Respuesta Evocada , Umbral Auditivo , Preescolar , Potenciales Microfónicos de la Cóclea , Conexina 26 , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Pérdida Auditiva Sensorineural/genética , Humanos , Lactante , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...