Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 553(7687): 217-221, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29258297

RESUMO

Although genetic factors contribute to almost half of all cases of deafness, treatment options for genetic deafness are limited. We developed a genome-editing approach to target a dominantly inherited form of genetic deafness. Here we show that cationic lipid-mediated in vivo delivery of Cas9-guide RNA complexes can ameliorate hearing loss in a mouse model of human genetic deafness. We designed and validated, both in vitro and in primary fibroblasts, genome editing agents that preferentially disrupt the dominant deafness-associated allele in the Tmc1 (transmembrane channel-like gene family 1) Beethoven (Bth) mouse model, even though the mutant Tmc1Bth allele differs from the wild-type allele at only a single base pair. Injection of Cas9-guide RNA-lipid complexes targeting the Tmc1Bth allele into the cochlea of neonatal Tmc1Bth/+ mice substantially reduced progressive hearing loss. We observed higher hair cell survival rates and lower auditory brainstem response thresholds in injected ears than in uninjected ears or ears injected with control complexes that targeted an unrelated gene. Enhanced acoustic startle responses were observed among injected compared to uninjected Tmc1Bth/+ mice. These findings suggest that protein-RNA complex delivery of target gene-disrupting agents in vivo is a potential strategy for the treatment of some types of autosomal-dominant hearing loss.


Assuntos
Proteínas Associadas a CRISPR/administração & dosagem , Edição de Genes/métodos , Genes Dominantes/genética , Terapia Genética/métodos , Perda Auditiva/genética , Estimulação Acústica , Alelos , Animais , Animais Recém-Nascidos , Limiar Auditivo , Sequência de Bases , Proteínas Associadas a CRISPR/metabolismo , Proteínas Associadas a CRISPR/uso terapêutico , Sistemas CRISPR-Cas , Sobrevivência Celular , Cóclea/citologia , Cóclea/metabolismo , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Fibroblastos , Células Ciliadas Auditivas/citologia , Perda Auditiva/fisiopatologia , Perda Auditiva/prevenção & controle , Humanos , Lipossomos , Masculino , Proteínas de Membrana/genética , Camundongos , Reflexo de Sobressalto
2.
Audiol Neurootol ; 28(6): 407-419, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37331337

RESUMO

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


Assuntos
Perda Auditiva Neurossensorial , Perda Auditiva , Humanos , Serina Endopeptidases/genética , Proteínas de Membrana/genética , Perda Auditiva Neurossensorial/genética , Perda Auditiva/genética , Mutação , Estudos de Associação Genética , Fenótipo , Proteínas de Neoplasias/genética
3.
Mol Ther ; 29(3): 973-988, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33212302

RESUMO

AAV-mediated gene therapy is a promising approach for treating genetic hearing loss. Replacement or editing of the Tmc1 gene, encoding hair cell mechanosensory ion channels, is effective for hearing restoration in mice with some limitations. Efficient rescue of outer hair cell function and lack of hearing recovery with later-stage treatment remain issues to be solved. Exogenous genes delivered with the adeno-associated virus (AAV)9-PHP.B capsid via the utricle transduce both inner and outer hair cells of the mouse cochlea with high efficacy. Here, we demonstrate that AAV9-PHP.B gene therapy can promote hair cell survival and successfully rescues hearing in three distinct mouse models of hearing loss. Tmc1 replacement with AAV9-PHP.B in a Tmc1 knockout mouse rescues hearing and promotes hair cell survival with equal efficacy in inner and outer hair cells. The same treatment in a recessive Tmc1 hearing-loss model, Baringo, partially recovers hearing even with later-stage treatment. Finally, dual delivery of Streptococcus pyogenes Cas9 (SpCas9) and guide RNA (gRNA) in separate AAV9-PHP.B vectors selectively disrupts a dominant Tmc1 allele and preserves hearing in Beethoven mice, a model of dominant, progressive hearing loss. Tmc1-targeted gene therapies using single or dual AAV9-PHP.B vectors offer potent and versatile approaches for treating dominant and recessive deafness.


Assuntos
Dependovirus/genética , Modelos Animais de Doenças , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Perda Auditiva/terapia , Proteínas de Membrana/fisiologia , RNA Guia de Cinetoplastídeos/genética , Animais , Feminino , Vetores Genéticos/genética , Perda Auditiva/genética , Perda Auditiva/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
J Neurophysiol ; 122(5): 1962-1974, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31533018

RESUMO

Optogenetics comprise a promising alternative to electrical stimulation for characterization of neural circuits and for the next generation of neural prostheses. Optogenetic stimulation relies on expression of photosensitive microbial proteins in animal cells to initiate a flow of ions into the cells in response to visible light. Here, we generated a novel transgenic mouse model in which we studied the optogenetic activation of spiral ganglion neurons, the primary afferent neurons of the auditory system, and showed a strong optogenetic response, with a similar amplitude as the acoustically evoked response. A twofold increase in the level of channelrhodopsin expression significantly increased the photosensitivity at both the single cell and organismal levels but also partially compromised the native electrophysiological properties of the neurons. The importance of channelrhodopsin expression level to optogenetic stimulation, revealed by these quantitative measurements, will be significant for the characterization of neural circuitry and for the use of optogenetics in neural prostheses.NEW & NOTEWORTHY This study reveals a dose-response relationship between channelrhodopsin expression and optogenetic excitation. Both single cell and organismal responses depend on the expression level of the heterologous protein. Expression level of the opsin is thus an important variable in determining the outcome of an optogenetic experiment. These results are key to the implementation of neural prostheses based on optogenetics, such as next generation cochlear implants, which would use light to elicit a neural response to sound.


Assuntos
Channelrhodopsins/fisiologia , Cóclea/fisiologia , Fenômenos Eletrofisiológicos , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Neurônios Aferentes/fisiologia , Optogenética , Gânglio Espiral da Cóclea/fisiologia , Animais , Camundongos , Camundongos Transgênicos , Modelos Animais
5.
Am J Hum Genet ; 98(6): 1101-1113, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27236922

RESUMO

Hearing impairment is the most common sensory deficit. It is frequently caused by the expression of an allele carrying a single dominant missense mutation. Herein, we show that a single intracochlear injection of an artificial microRNA carried in a viral vector can slow progression of hearing loss for up to 35 weeks in the Beethoven mouse, a murine model of non-syndromic human deafness caused by a dominant gain-of-function mutation in Tmc1 (transmembrane channel-like 1). This outcome is noteworthy because it demonstrates the feasibility of RNA-interference-mediated suppression of an endogenous deafness-causing allele to slow progression of hearing loss. Given that most autosomal-dominant non-syndromic hearing loss in humans is caused by this mechanism of action, microRNA-based therapeutics might be broadly applicable as a therapy for this type of deafness.


Assuntos
Vias Auditivas , Perda Auditiva/prevenção & controle , Proteínas de Membrana/fisiologia , MicroRNAs/genética , Mutação de Sentido Incorreto/genética , Animais , Dependovirus/genética , Perda Auditiva/etiologia , Perda Auditiva/patologia , Humanos , Mecanotransdução Celular , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , MicroRNAs/administração & dosagem , Interferência de RNA
6.
J Neurosci ; 36(43): 10921-10926, 2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27798174

RESUMO

Sensory transduction in vertebrate hair cells and the molecules that mediate it have long been of great interest. Some components of the mechanotransduction apparatus have been identified, most as deafness gene products. Although prior candidates for the mechanotransduction channel have been proposed, each has faded with new evidence. Now, two strong candidates, TMC1 and TMC2 (transmembrane channel-like), have emerged from discovery of deafness genes in humans and mice. They are expressed at the right time during development: exactly at the onset of mechanosensitivity. They are expressed in the right place: in hair cells but not surrounding cells. Fluorescently tagged TMCs localize to the tips of stereocilia, the site of the transduction channels. TMCs bind other proteins essential for mechanosensation, suggesting a larger transduction complex. Although TMC1 and TMC2 can substitute for each other, genetic deletion of both renders mouse hair cells mechanically insensitive. Finally, the conductance and Ca2+ selectivity of the transduction channels depend on the TMC proteins, differing when hair cells express one or the other TMC, and differing if TMC1 harbors a point mutation. Some contrary evidence has emerged: a current activated in hair cells by negative pressure, with some similarity to the transduction current, persists in TMC knock-outs. But it is not clear that this anomalous current is carried by the same proteins. Further evidence is desired, such as production of a mechanically gated conductance by pure TMCs. But the great majority of evidence is consistent with these TMCs as pore-forming subunits of the long-sought hair-cell transduction channel.


Assuntos
Cálcio/metabolismo , Células Ciliadas Auditivas/fisiologia , Mecanotransdução Celular/fisiologia , Proteínas de Membrana/metabolismo , Modelos Biológicos , Animais , Sinalização do Cálcio/fisiologia , Medicina Baseada em Evidências , Audição , Humanos , Ativação do Canal Iônico/fisiologia , Camundongos
7.
Neurogenetics ; 17(2): 115-123, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26879195

RESUMO

TMC1 encodes a protein required for the normal function of mechanically activated channels that enable sensory transduction in auditory and vestibular hair cells. TMC1 protein is localized at the tips of the hair cell stereocilia, the site of conventional mechanotransduction. In many populations, loss-of-function recessive mutations of TMC1 are associated with profound deafness across all frequencies tested. In six families reported here, variable moderate-to-severe or moderate-to-profound hearing loss co-segregated with STR (short tandem repeats) markers at the TMC1 locus DFNB7/11. Massively parallel and Sanger sequencing of genomic DNA revealed each family co-segregating hearing loss with a homozygous TMC1 mutation: two reported mutations (p.R34X and p.R389Q) and three novel mutations (p.S596R, p.N199I, and c.1404 + 1G > T). TMC1 cDNA sequence from affected subjects homozygous for the donor splice site transversion c.1404 + 1G > T revealed skipping of exon 16, deleting 60 amino acids from the TMC1 protein. Since the mutations in our study cause less than profound hearing loss, we speculate that there is hypo-functional TMC1 mechanotransduction channel activity and that other even less damaging variants of TMC1 may be associated with more common mild-to-severe sensorineural hearing loss.


Assuntos
Perda Auditiva/genética , Proteínas de Membrana/genética , Adolescente , Criança , Genes Recessivos , Perda Auditiva/fisiopatologia , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Linhagem , Adulto Jovem
8.
Hum Mol Genet ; 23(9): 2374-90, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24334608

RESUMO

Usher syndrome type 2 (USH2) is the predominant form of USH, a leading genetic cause of combined deafness and blindness. PDZD7, a paralog of two USH causative genes, USH1C and USH2D (WHRN), was recently reported to be implicated in USH2 and non-syndromic deafness. It encodes a protein with multiple PDZ domains. To understand the biological function of PDZD7 and the pathogenic mechanism caused by PDZD7 mutations, we generated and thoroughly characterized a Pdzd7 knockout mouse model. The Pdzd7 knockout mice exhibit congenital profound deafness, as assessed by auditory brainstem response, distortion product otoacoustic emission and cochlear microphonics tests, and normal vestibular function, as assessed by their behaviors. Lack of PDZD7 leads to the disorganization of stereocilia bundles and a reduction in mechanotransduction currents and sensitivity in cochlear outer hair cells. At the molecular level, PDZD7 determines the localization of the USH2 protein complex, composed of USH2A, GPR98 and WHRN, to ankle links in developing cochlear hair cells, likely through its direct interactions with these three proteins. The localization of PDZD7 to the ankle links of cochlear hair bundles also relies on USH2 proteins. In photoreceptors of Pdzd7 knockout mice, the three USH2 proteins largely remain unchanged at the periciliary membrane complex. The electroretinogram responses of both rod and cone photoreceptors are normal in knockout mice at 1 month of age. Therefore, although the organization of the USH2 complex appears different in photoreceptors, it is clear that PDZD7 plays an essential role in organizing the USH2 complex at ankle links in developing cochlear hair cells. GenBank accession numbers: KF041446, KF041447, KF041448, KF041449, KF041450, KF041451.


Assuntos
Deleção de Genes , Células Ciliadas Auditivas/metabolismo , Perda Auditiva/genética , Perda Auditiva/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Síndromes de Usher/metabolismo , Animais , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Síndromes de Usher/genética
9.
Pflugers Arch ; 467(1): 85-94, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25074487

RESUMO

Mutations of the transmembrane channel-like 1 (TMC1) gene can cause dominant and recessive forms of deafness in humans and mice. TMC1 is one of eight mammalian TMC genes of unknown function. The multi-pass transmembrane topologic structure of the proteins they encode suggests roles as a receptor, transporter, channel, or pump. Tmc1 and the closely related Tmc2 gene are expressed in neurosensory hair cells of the auditory and vestibular end organs of the mouse inner ear. Recent studies have demonstrated that Tmc1 and Tmc2 are specifically required for mechanoelectrical transduction in hair cells. The exact role of these proteins in mechanoelectrical transduction is unknown. TMC1 and TMC2 are viable candidates for the mechanoelectrical transduction channel of hair cells, whose component molecules have eluded identification for over 30 years. We expect that studies of TMC proteins will yield insights into molecular components and mechanisms of mechanosensation in auditory and vestibular hair cells, as well as in other tissues and organs.


Assuntos
Células Ciliadas Auditivas/fisiologia , Células Ciliadas Vestibulares/fisiologia , Audição/fisiologia , Canais Iônicos/fisiologia , Mecanotransdução Celular/fisiologia , Equilíbrio Postural/fisiologia , Animais , Humanos , Ativação do Canal Iônico/fisiologia
10.
Front Neurol ; 15: 1356614, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38638308

RESUMO

Tmc1 and Tmc2 are essential pore-forming subunits of mechanosensory transduction channels localized to the tips of stereovilli in auditory and vestibular hair cells of the inner ear. To investigate expression and function of Tmc1 and Tmc2 in vestibular organs, we used quantitative polymerase chain reaction (qPCR), fluorescence in situ hybridization - hairpin chain reaction (FISH-HCR), immunostaining, FM1-43 uptake and we measured vestibular evoked potentials (VsEPs) and vestibular ocular reflexes (VORs). We found that Tmc1 and Tmc2 showed dynamic developmental changes, differences in regional expression patterns, and overall expression levels which differed between the utricle and saccule. These underlying changes contributed to unanticipated phenotypic loss of VsEPs and VORs in Tmc1 KO mice. In contrast, Tmc2 KO mice retained VsEPs despite the loss of the calcium buffering protein calretinin, a characteristic biomarker of mature striolar calyx-only afferents. Lastly, we found that neonatal Tmc1 gene replacement therapy is sufficient to restore VsEP in Tmc1 KO mice for up to six months post-injection.

11.
J Neurosci ; 32(28): 9485-98, 2012 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-22787034

RESUMO

Mutation in the clarin-1 gene (Clrn1) results in loss of hearing and vision in humans (Usher syndrome III), but the role of clarin-1 in the sensory hair cells is unknown. Clarin-1 is predicted to be a four transmembrane domain protein similar to members of the tetraspanin family. Mice carrying null mutation in the clarin-1 gene (Clrn1(-/-)) show loss of hair cell function and a possible defect in ribbon synapse. We investigated the role of clarin-1 using various in vitro and in vivo approaches. We show by immunohistochemistry and patch-clamp recordings of Ca(2+) currents and membrane capacitance from inner hair cells that clarin-1 is not essential for formation or function of ribbon synapse. However, reduced cochlear microphonic potentials, FM1-43 [N-(3-triethylammoniumpropyl)-4-(4-(dibutylamino)styryl) pyridinium dibromide] loading, and transduction currents pointed to diminished cochlear hair bundle function in Clrn1(-/-) mice. Electron microscopy of cochlear hair cells revealed loss of some tall stereocilia and gaps in the v-shaped bundle, although tip links and staircase arrangement of stereocilia were not primarily affected by Clrn1(-/-) mutation. Human clarin-1 protein expressed in transfected mouse cochlear hair cells localized to the bundle; however, the pathogenic variant p.N48K failed to localize to the bundle. The mouse model generated to study the in vivo consequence of p.N48K in clarin-1 (Clrn1(N48K)) supports our in vitro and Clrn1(-/-) mouse data and the conclusion that CLRN1 is an essential hair bundle protein. Furthermore, the ear phenotype in the Clrn1(N48K) mouse suggests that it is a valuable model for ear disease in CLRN1(N48K), the most prevalent Usher syndrome III mutation in North America.


Assuntos
Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Células Ciliadas Auditivas/fisiologia , Mecanorreceptores/fisiologia , Proteínas de Membrana/genética , Síndromes de Usher/genética , Estimulação Acústica , Fatores Etários , Oxirredutases do Álcool/metabolismo , Animais , Animais Recém-Nascidos , Asparagina/genética , Bário/farmacologia , Fenômenos Biofísicos/genética , Caderinas/genética , Linhagem Celular Transformada , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células Ciliadas Auditivas/ultraestrutura , Humanos , Lisina/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Varredura/métodos , Mutação/genética , Fibras Nervosas/patologia , Fibras Nervosas/ultraestrutura , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Estimulação Física/métodos , Psicoacústica , Compostos de Piridínio/metabolismo , Compostos de Amônio Quaternário/metabolismo , Receptores de AMPA/metabolismo , Sinapses/patologia , Sinapses/ultraestrutura , Transfecção , Síndromes de Usher/patologia , Síndromes de Usher/fisiopatologia
12.
Otol Neurotol ; 44(1): 21-25, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36509434

RESUMO

OBJECTIVE: Investigate hearing preservation and spatial hearing outcomes in children with TMPRSS3 mutations who received bilateral cochlear implantation. STUDY DESIGN AND METHODS: Longitudinal case series report. Two siblings (ages, 7 and 4 yr) with TMPRSS3 mutations with down-sloping audiograms received sequential bilateral cochlear implantation with hearing preservation with low-frequency acoustic amplification and high-frequency electrical stimulation. Spatial hearing, including speech perception and localization, was assessed at three time points: preoperative, postoperative of first and second cochlear implant (CI). RESULTS: Both children showed low-frequency hearing preservation in unaided, acoustic-only audiograms. Both children demonstrated improvements in speech perception in both quiet and noise after CI activations. The emergence of spatial hearing was observed. Each child's overall speech perception and spatial hearing when listening with bilateral CIs were within the range or better than published group data from children with bilateral CIs of other etiology. CONCLUSION: Bilateral cochlear implantation with hearing preservation is a viable option for managing hearing loss for pediatric patients with TMPRSS3 mutations.


Assuntos
Implante Coclear , Implantes Cocleares , Surdez , Percepção da Fala , Humanos , Criança , Percepção da Fala/fisiologia , Audição/genética , Surdez/reabilitação , Proteínas de Membrana , Proteínas de Neoplasias , Serina Endopeptidases/genética
13.
Neuron ; 111(20): 3195-3210.e7, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37543036

RESUMO

OSCA/TMEM63s form mechanically activated (MA) ion channels in plants and animals, respectively. OSCAs and related TMEM16s and transmembrane channel-like (TMC) proteins form homodimers with two pores. Here, we uncover an unanticipated monomeric configuration of TMEM63 proteins. Structures of TMEM63A and TMEM63B (referred to as TMEM63s) revealed a single highly restricted pore. Functional analyses demonstrated that TMEM63s are bona fide mechanosensitive ion channels, characterized by small conductance and high thresholds. TMEM63s possess evolutionary variations in the intracellular linker IL2, which mediates dimerization in OSCAs. Replacement of OSCA1.2 IL2 with TMEM63A IL2 or mutations to key variable residues resulted in monomeric OSCA1.2 and MA currents with significantly higher thresholds. Structural analyses revealed substantial conformational differences in the mechano-sensing domain IL2 and gating helix TM6 between TMEM63s and OSCA1.2. Our studies reveal that mechanosensitivity in OSCA/TMEM63 channels is affected by oligomerization and suggest gating mechanisms that may be shared by OSCA/TMEM63, TMEM16, and TMC channels.


Assuntos
Interleucina-2 , Canais Iônicos , Animais , Interleucina-2/genética , Interleucina-2/metabolismo , Canais Iônicos/metabolismo , Mutação/genética
14.
Sci Transl Med ; 15(702): eabq3916, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37379370

RESUMO

Inner ear gene therapy has recently effectively restored hearing in neonatal mice, but it is complicated in adulthood by the structural inaccessibility of the cochlea, which is embedded within the temporal bone. Alternative delivery routes may advance auditory research and also prove useful when translated to humans with progressive genetic-mediated hearing loss. Cerebrospinal fluid flow via the glymphatic system is emerging as a new approach for brain-wide drug delivery in rodents as well as humans. The cerebrospinal fluid and the fluid of the inner ear are connected via a bony channel called the cochlear aqueduct, but previous studies have not explored the possibility of delivering gene therapy via the cerebrospinal fluid to restore hearing in adult deaf mice. Here, we showed that the cochlear aqueduct in mice exhibits lymphatic-like characteristics. In vivo time-lapse magnetic resonance imaging, computed tomography, and optical fluorescence microscopy showed that large-particle tracers injected into the cerebrospinal fluid reached the inner ear by dispersive transport via the cochlear aqueduct in adult mice. A single intracisternal injection of adeno-associated virus carrying solute carrier family 17, member 8 (Slc17A8), which encodes vesicular glutamate transporter-3 (VGLUT3), rescued hearing in adult deaf Slc17A8-/- mice by restoring VGLUT3 protein expression in inner hair cells, with minimal ectopic expression in the brain and none in the liver. Our findings demonstrate that cerebrospinal fluid transport comprises an accessible route for gene delivery to the adult inner ear and may represent an important step toward using gene therapy to restore hearing in humans.


Assuntos
Orelha Interna , Adulto , Animais , Humanos , Camundongos , Orelha Interna/patologia , Cóclea , Audição , Terapia Genética/métodos , Técnicas de Transferência de Genes
15.
bioRxiv ; 2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37398045

RESUMO

Calcium and integrin-binding protein 2 (CIB2) and CIB3 bind to transmembrane channel-like 1 (TMC1) and TMC2, the pore-forming subunits of the inner-ear mechanoelectrical transduction (MET) apparatus. Whether these interactions are functionally relevant across mechanosensory organs and vertebrate species is unclear. Here we show that both CIB2 and CIB3 can form heteromeric complexes with TMC1 and TMC2 and are integral for MET function in mouse cochlea and vestibular end organs as well as in zebrafish inner ear and lateral line. Our AlphaFold 2 models suggest that vertebrate CIB proteins can simultaneously interact with at least two cytoplasmic domains of TMC1 and TMC2 as validated using nuclear magnetic resonance spectroscopy of TMC1 fragments interacting with CIB2 and CIB3. Molecular dynamics simulations of TMC1/2 complexes with CIB2/3 predict that TMCs are structurally stabilized by CIB proteins to form cation channels. Overall, our work demonstrates that intact CIB2/3 and TMC1/2 complexes are integral to hair-cell MET function in vertebrate mechanosensory epithelia.

16.
J Neurosci ; 31(46): 16814-25, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22090507

RESUMO

HCN1-4 subunits form Na+/K+-permeable ion channels that are activated by hyperpolarization and carry the current known as I(h). I(h) has been characterized in vestibular hair cells of the inner ear, but its molecular correlates and functional contributions have not been elucidated. We examined Hcn mRNA expression and immunolocalization of HCN protein in the mouse utricle, a mechanosensitive organ that contributes to the sense of balance. We found that HCN1 is the most highly expressed subunit, localized to the basolateral membranes of type I and type II hair cells. We characterized I(h) using the whole-cell, voltage-clamp technique and found the current expressed in 84% of the cells with a mean maximum conductance of 4.4 nS. I(h) was inhibited by ZD7288, cilobradine, and by adenoviral expression of a dominant-negative form of HCN2. To determine which HCN subunits carried I(h), we examined hair cells from mice deficient in Hcn1, 2, or both. I(h) was completely abolished in hair cells of Hcn1⁻/⁻ mice and Hcn1/2⁻/⁻ mice but was similar to wild-type in Hcn2⁻/⁻ mice. To examine the functional contributions of I(h), we recorded hair cell membrane responses to small hyperpolarizing current steps and found that activation of I(h) evoked a 5-10 mV sag depolarization and a subsequent 15-20 mV rebound upon termination. The sag and rebound were nearly abolished in Hcn1-deficient hair cells. We also found that Hcn1-deficient mice had deficits in vestibular-evoked potentials and balance assays. We conclude that HCN1 contributes to vestibular hair cell function and the sense of balance.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Orelha Interna/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Equilíbrio Postural/genética , Canais de Potássio/metabolismo , Sáculo e Utrículo/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Benzazepinas/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/deficiência , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Orelha Interna/efeitos dos fármacos , Estimulação Elétrica , Feminino , Fatores de Transcrição Forkhead/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Knockout , Movimento (Física) , Proteínas do Tecido Nervoso/genética , Proteínas de Neurofilamentos/metabolismo , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Canais de Potássio/deficiência , Canais de Potássio/genética , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Teste de Desempenho do Rota-Rod , Sáculo e Utrículo/citologia , Potenciais Evocados Miogênicos Vestibulares/genética , Potenciais Evocados Miogênicos Vestibulares/fisiologia
17.
J Neurosci ; 31(34): 12241-50, 2011 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-21865467

RESUMO

The polycystic kidney disease-1 (Pkd1) gene encodes a large transmembrane protein (polycystin-1, or PC-1) that is reported to function as a fluid flow sensor in the kidney. As a member of the transient receptor potential family, PC-1 has also been hypothesized to play a role in the elusive mechanoelectrical transduction (MET) channel in inner ear hair cells. Here, we analyze two independent mouse models of PC-1, a knock-in (KI) mutant line and a hair cell-specific inducible Cre-mediated knock-out line. Both models exhibit normal MET channel function at neonatal ages despite hearing loss and ultrastructural abnormalities of sterecilia that remain properly polarized at adult ages. These findings demonstrate that PC-1 plays an essential role in stereocilia structure and maintenance but not directly in MET channel function or planar cell polarity. We also demonstrate that PC-1 is colocalized with F-actin in hair cell stereocilia in vivo, using a hemagglutinin-tagged PC-1 KI mouse model, and in renal epithelial cell microvilli in vitro. These results not only demonstrate a novel role for PC-1 in the cochlea, but also suggest insight into the development of polycystic kidney disease.


Assuntos
Cílios/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Mecanotransdução Celular/fisiologia , Órgão Espiral/fisiologia , Canais de Cátion TRPP/fisiologia , Animais , Animais Recém-Nascidos , Cílios/genética , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Células Ciliadas Auditivas Internas/citologia , Células HeLa , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Humanos , Masculino , Mecanotransdução Celular/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Canais de Cátion TRPP/deficiência , Canais de Cátion TRPP/genética
18.
J Neurophysiol ; 108(1): 175-86, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22496522

RESUMO

Inner ear hair cells respond to mechanical stimuli with graded receptor potentials. These graded responses are modulated by a host of voltage-dependent currents that flow across the basolateral membrane. Here, we examine the molecular identity and the function of a class of voltage-dependent ion channels that carries the potassium-selective inward rectifier current known as I(K1). I(K1) has been identified in vestibular hair cells of various species, but its molecular composition and functional contributions remain obscure. We used quantitative RT-PCR to show that the inward rectifier gene, Kir2.1, is highly expressed in mouse utricle between embryonic day 15 and adulthood. We confirmed Kir2.1 protein expression in hair cells by immunolocalization. To examine the molecular composition of I(K1), we recorded voltage-dependent currents from type II hair cells in response to 50-ms steps from -124 to -54 in 10-mV increments. Wild-type cells had rapidly activating inward currents with reversal potentials close to the K(+) equilibrium potential and a whole-cell conductance of 4.8 ± 1.5 nS (n = 46). In utricle hair cells from Kir2.1-deficient (Kir2.1(-/-)) mice, I(K1) was absent at all stages examined. To identify the functional contribution of Kir2.1, we recorded membrane responses in current-clamp mode. Hair cells from Kir2.1(-/-) mice had significantly (P < 0.001) more depolarized resting potentials and larger, slower membrane responses than those of wild-type cells. These data suggest that Kir2.1 is required for I(K1) in type II utricle hair cells and contributes to hyperpolarized resting potentials and fast, small amplitude receptor potentials in response to current inputs, such as those evoked by hair bundle deflections.


Assuntos
Orelha Interna/citologia , Células Ciliadas Vestibulares/metabolismo , Ativação do Canal Iônico/genética , Potenciais da Membrana/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Compostos de Bário/farmacologia , Biofísica , Cloretos/farmacologia , Estimulação Elétrica , Embrião de Mamíferos , Vetores Genéticos/fisiologia , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Técnicas In Vitro , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Mutação Puntual/genética , Canais de Potássio Corretores do Fluxo de Internalização/deficiência , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção/métodos
19.
Proc Natl Acad Sci U S A ; 106(13): 5252-7, 2009 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-19270079

RESUMO

Deafness is the most common form of sensory impairment in humans and is frequently caused by single gene mutations. Interestingly, different mutations in a gene can cause syndromic and nonsyndromic forms of deafness, as well as progressive and age-related hearing loss. We provide here an explanation for the phenotypic variability associated with mutations in the cadherin 23 gene (CDH23). CDH23 null alleles cause deaf-blindness (Usher syndrome type 1D; USH1D), whereas missense mutations cause nonsyndromic deafness (DFNB12). In a forward genetic screen, we have identified salsa mice, which suffer from hearing loss due to a Cdh23 missense mutation modeling DFNB12. In contrast to waltzer mice, which carry a CDH23 null allele mimicking USH1D, hair cell development is unaffected in salsa mice. Instead, tip links, which are thought to gate mechanotransduction channels in hair cells, are progressively lost. Our findings suggest that DFNB12 belongs to a new class of disorder that is caused by defects in tip links. We propose that mutations in other genes that cause USH1 and nonsyndromic deafness may also have distinct effects on hair cell development and function.


Assuntos
Caderinas/genética , Surdez/genética , Células Ciliadas Auditivas , Mutação de Sentido Incorreto , Animais , Modelos Animais de Doenças , Mecanotransdução Celular/genética , Camundongos , Síndromes de Usher/genética
20.
Biomolecules ; 12(7)2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35883470

RESUMO

Gene therapy for genetic hearing loss is an emerging therapeutic modality for hearing restoration. However, the approach has not yet been translated into clinical application. To further develop inner-ear gene therapy, we engineered a novel mouse model bearing a human mutation in the transmembrane channel-1 gene (Tmc1) and characterized the auditory phenotype of the mice. TMC1 forms the mechanosensory transduction channel in mice and humans and is necessary for auditory function. We found that mice harboring the equivalent of the human p.N199I mutation (p.N193I) had profound congenital hearing loss due to loss of hair cell sensory transduction. Next, we optimized and screened viral payloads packaged into AAV9-PHP.B capsids. The vectors were injected into the inner ears of Tmc1Δ/Δ mice and the new humanized Tmc1-p.N193I mouse model. Auditory brainstem responses (ABRs), distortion product otoacoustic emissions (DPOAEs), cell survival, and biodistribution were evaluated in the injected mice. We found broad-spectrum, durable recovery of auditory function in Tmc1-p.N193I mice injected with AAV9-PHP.B-CB6-hTMC1-WPRE. ABR and DPOAE thresholds were equivalent to those of wild-type mice across the entire frequency range. Biodistribution analysis revealed viral DNA/RNA in the contralateral ear, brain, and liver but no overt toxicity. We conclude that the AAV9-PHP.B-CB6-hTMC1-WPRE construct may be suitable for further development as a gene therapy reagent for treatment of humans with genetic hearing loss due to recessive TMC1 mutations.


Assuntos
Surdez , Perda Auditiva , Animais , Surdez/genética , Modelos Animais de Doenças , Terapia Genética , Perda Auditiva/genética , Perda Auditiva/terapia , Perda Auditiva Neurossensorial , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA