Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 2400, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37100771

RESUMEN

Usher syndrome type 1 F (USH1F), caused by mutations in the protocadherin-15 gene (PCDH15), is characterized by congenital deafness, lack of balance, and progressive blindness. In hair cells, the receptor cells of the inner ear, PCDH15 is a component of tip links, fine filaments which pull open mechanosensory transduction channels. A simple gene addition therapy for USH1F is challenging because the PCDH15 coding sequence is too large for adeno-associated virus (AAV) vectors. We use rational, structure-based design to engineer mini-PCDH15s in which 3-5 of the 11 extracellular cadherin repeats are deleted, but which still bind a partner protein. Some mini-PCDH15s can fit in an AAV. An AAV encoding one of these, injected into the inner ears of mouse models of USH1F, produces a mini-PCDH15 which properly forms tip links, prevents the degeneration of hair cell bundles, and rescues hearing. Mini-PCDH15s may be a useful therapy for the deafness of USH1F.


Asunto(s)
Oído Interno , Síndromes de Usher , Animales , Ratones , Cadherinas/metabolismo , Oído Interno/metabolismo , Células Ciliadas Auditivas/metabolismo , Audición/genética , Síndromes de Usher/genética , Síndromes de Usher/terapia , Proteínas Relacionadas con las Cadherinas/metabolismo
2.
Sci Adv ; 8(28): eabo1126, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35857511

RESUMEN

The transmembrane (TM) channel-like 1 (TMC1) and TMC2 proteins play a central role in auditory transduction, forming ion channels that convert sound into electrical signals. However, the molecular mechanism of their gating remains unknown. Here, using predicted structural models as a guide, we probed the effects of 12 mutations on the mechanical gating of the transduction currents in native hair cells of Tmc1/2-null mice expressing virally introduced TMC1 variants. Whole-cell electrophysiological recordings revealed that mutations within the pore-lining TM4 and TM6 helices modified gating, reducing the force sensitivity or shifting the open probability of the channels, or both. For some of the mutants, these changes were accompanied by a change in single-channel conductance. Our observations are in line with a model wherein conformational changes in the TM4 and TM6 helices are involved in the mechanical gating of the transduction channel.

3.
Sci Total Environ ; 834: 155166, 2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-35413348

RESUMEN

The Anoxic/Oxic (A/O) process involves recirculating mixed liquor between its A and O tanks so that nitrate produced in the O tank can be used to for denitrification with influent COD in the A tank. Because biomass is recirculated along with nitrate, A/O operation leads to similar microbial communities in the A and O tanks, which may decrease the rates of denitrification and nitrification in each tank. Here, bench-scale experiments simulated this aspect of the A/O process by exchanging biomass between an anoxic flask and an oxic cylinder at exchange ratios of 0%, 20%, 30%, and 50%. Nitrification and denitrification rates were only 40% and 19% for 50% biomass exchange of that for no biomass exchange. Phylogenetic analysis documented that the microbial communities became much more similar with biomass exchange, and the finding was consistent with community composition in a full-scale A/O process in a municipal wastewater treatment plant. A two-stage vertical baffled bioreactor (VBBR) realized efficient total­nitrogen removal in recirculation without biomass exchange. Average removals of COD and TN were respectively 6% and 22% higher for the two-stage VBBR than the conventional A/O process, but its hydraulic retention time (HRT) was 55% to 70% of the volume of a conventional A/O process treating the same influent wastewater. The VBBR was more efficient because its anoxic biofilm was enriched in denitrifying bacteria, while its oxic biofilm was enriched in nitrifying bacteria. For example, the phylum Chloroflexi was greater in the An-VBBR, while the phylum Proteobacteria was greater in the Ox-VBBR.


Asunto(s)
Desnitrificación , Nitratos , Biomasa , Reactores Biológicos , Nitrificación , Nitrógeno/análisis , Filogenia , Aguas del Alcantarillado , Aguas Residuales
4.
Mol Ther ; 28(12): 2662-2676, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-32818431

RESUMEN

Usher syndrome is a syndromic form of hereditary hearing impairment that includes sensorineural hearing loss and delayed-onset retinitis pigmentosa (RP). Type 1 Usher syndrome (USH1) is characterized by congenital profound sensorineural hearing impairment and vestibular areflexia, with adolescent-onset RP. Systemic treatment with antisense oligonucleotides (ASOs) targeting the human USH1C c.216G>A splicing mutation in a knockin mouse model of USH1 restores hearing and balance. Herein, we explore the effect of delivering ASOs locally to the ear to treat hearing and vestibular dysfunction associated with Usher syndrome. Three localized delivery strategies were investigated in USH1C mice: inner ear injection, trans-tympanic membrane injection, and topical tympanic membrane application. We demonstrate, for the first time, that ASOs delivered directly to the ear correct Ush1c expression in inner ear tissue, improve cochlear hair cell transduction currents, restore vestibular afferent irregularity, spontaneous firing rate, and sensitivity to head rotation, and successfully recover hearing thresholds and balance behaviors in USH1C mice. We conclude that local delivery of ASOs to the middle and inner ear reach hair cells and can rescue both hearing and balance. These results also demonstrate the therapeutic potential of ASOs to treat hearing and balance deficits associated with Usher syndrome and other ear diseases.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Oído Medio/efectos de los fármacos , Terapia Genética/métodos , Células Ciliadas Auditivas/efectos de los fármacos , Mutación , Oligonucleótidos Antisentido/administración & dosificación , Síndromes de Usher/genética , Síndromes de Usher/terapia , Vestíbulo del Laberinto/efectos de los fármacos , Administración Tópica , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Células Ciliadas Auditivas/metabolismo , Audición/efectos de los fármacos , Inyecciones , Masculino , Ratones , Ratones Endogámicos C57BL , Membrana Timpánica/efectos de los fármacos , Vestíbulo del Laberinto/metabolismo
5.
Sci Transl Med ; 12(546)2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493795

RESUMEN

Most genetic diseases arise from recessive point mutations that require correction, rather than disruption, of the pathogenic allele to benefit patients. Base editing has the potential to directly repair point mutations and provide therapeutic restoration of gene function. Mutations of transmembrane channel-like 1 gene (TMC1) can cause dominant or recessive deafness. We developed a base editing strategy to treat Baringo mice, which carry a recessive, loss-of-function point mutation (c.A545G; resulting in the substitution p.Y182C) in Tmc1 that causes deafness. Tmc1 encodes a protein that forms mechanosensitive ion channels in sensory hair cells of the inner ear and is required for normal auditory function. We found that sensory hair cells of Baringo mice have a complete loss of auditory sensory transduction. To repair the mutation, we tested several optimized cytosine base editors (CBEmax variants) and guide RNAs in Baringo mouse embryonic fibroblasts. We packaged the most promising CBE, derived from an activation-induced cytidine deaminase (AID), into dual adeno-associated viruses (AAVs) using a split-intein delivery system. The dual AID-CBEmax AAVs were injected into the inner ears of Baringo mice at postnatal day 1. Injected mice showed up to 51% reversion of the Tmc1 c.A545G point mutation to wild-type sequence (c.A545A) in Tmc1 transcripts. Repair of Tmc1 in vivo restored inner hair cell sensory transduction and hair cell morphology and transiently rescued low-frequency hearing 4 weeks after injection. These findings provide a foundation for a potential one-time treatment for recessive hearing loss and support further development of base editing to correct pathogenic point mutations.


Asunto(s)
Sordera , Proteínas de la Membrana , Animales , Sordera/genética , Sordera/terapia , Fibroblastos , Células Ciliadas Auditivas , Audición/genética , Humanos , Ratones
6.
Hear Res ; 394: 107882, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31980281

RESUMEN

Viral delivery of exogenous coding sequences into the inner ear has the potential for therapeutic benefit for patients suffering genetic or acquired hearing loss. To devise improved strategies for viral delivery, we investigated two injection techniques, round window membrane injection or a novel utricle injection method, for their ability to safely and efficiently transduce sensory hair cells and neurons of the mouse inner ear. In addition, we evaluated three synthetic AAV vectors (Anc80L65, AAV9-PHP.B, AAV2.7m8) encoding enhanced green fluorescent protein (eGFP) and three promoters (Cmv, Synapsin, Gfap) for their ability to transduce and drive expression in desired cell types. We found the utricle injection method with AAV9-PHP.B and a Cmv promoter was the most efficient combination for driving robust eGFP expression in both inner and outer hair cells. We found eGFP expression levels rose over 3-5 days post-injection, a viral dose of 1.5 × 109 gc yielded half maximal eGFP expression and that the utricle injection method yielded transduced hair cells even when delivered as late as postnatal day 16. Sensory transduction and auditory thresholds were unaltered in injected mice relative to uninjected wild-type controls. Vestibular end organs were also transduced without affecting balance behavior. The Synapsin promoter and the Gfap promoter drove strong eGFP expression in inner ear neurons and supporting cells, respectively. We conclude the AAV9-PHP.B vector and the utricle injection method are well-suited for delivery of exogenous gene constructs into inner ears of mouse models of auditory and vestibular dysfunction.


Asunto(s)
Oído Interno , Animales , Infecciones por Citomegalovirus , Dependovirus/genética , Terapia Genética , Vectores Genéticos , Células Ciliadas Auditivas Externas , Ratones , Sáculo y Utrículo , Sinapsinas/genética
7.
Nat Med ; 25(7): 1123-1130, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270503

RESUMEN

Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.


Asunto(s)
Alelos , Edición Génica , Pérdida Auditiva Sensorineural/prevención & control , Proteínas de la Membrana/genética , Animales , Proteína 9 Asociada a CRISPR/fisiología , Línea Celular , Células Cultivadas , Dependovirus/genética , Modelos Animales de Enfermedad , Pérdida Auditiva Sensorineural/genética , Humanos , Ratones , Ratones Endogámicos C57BL
8.
Nat Commun ; 10(1): 734, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30737404

RESUMEN

The original version of this Article contained errors in Fig. 5. In panels i and j the three rightmost x-axis labels inadvertently read 'Tmc1' instead of 'Tmc2'. These errors have been corrected in both the PDF and HTML versions of the Article.

9.
Nat Commun ; 10(1): 236, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670701

RESUMEN

Fifty percent of inner ear disorders are caused by genetic mutations. To develop treatments for genetic inner ear disorders, we designed gene replacement therapies using synthetic adeno-associated viral vectors to deliver the coding sequence for Transmembrane Channel-Like (Tmc) 1 or 2 into sensory hair cells of mice with hearing and balance deficits due to mutations in Tmc1 and closely related Tmc2. Here we report restoration of function in inner and outer hair cells, enhanced hair cell survival, restoration of cochlear and vestibular function, restoration of neural responses in auditory cortex and recovery of behavioral responses to auditory and vestibular stimulation. Secondarily, we find that inner ear Tmc gene therapy restores breeding efficiency, litter survival and normal growth rates in mouse models of genetic inner ear dysfunction. Although challenges remain, the data suggest that Tmc gene therapy may be well suited for further development and perhaps translation to clinical application.


Asunto(s)
Sordera/genética , Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Pérdida Auditiva/genética , Enfermedades del Laberinto/genética , Proteínas de la Membrana/genética , Animales , Sordera/terapia , Células Ciliadas Auditivas/fisiología , Células Ciliadas Vestibulares/fisiología , Pérdida Auditiva/terapia , Enfermedades del Laberinto/terapia , Ratones , Ratones Mutantes
10.
Neuron ; 99(4): 736-753.e6, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30138589

RESUMEN

The proteins that form the permeation pathway of mechanosensory transduction channels in inner-ear hair cells have not been definitively identified. Genetic, anatomical, and physiological evidence support a role for transmembrane channel-like protein (TMC) 1 in hair cell sensory transduction, yet the molecular function of TMC proteins remains unclear. Here, we provide biochemical evidence suggesting TMC1 assembles as a dimer, along with structural and sequence analyses suggesting similarity to dimeric TMEM16 channels. To identify the pore region of TMC1, we used cysteine mutagenesis and expressed mutant TMC1 in hair cells of Tmc1/2-null mice. Cysteine-modification reagents rapidly and irreversibly altered permeation properties of mechanosensory transduction. We propose that TMC1 is structurally similar to TMEM16 channels and includes ten transmembrane domains with four domains, S4-S7, that line the channel pore. The data provide compelling evidence that TMC1 is a pore-forming component of sensory transduction channels in auditory and vestibular hair cells.


Asunto(s)
Células Ciliadas Auditivas Internas/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/química , Proteínas de la Membrana/fisiología , Porinas/química , Porinas/fisiología , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Estructura Secundaria de Proteína
11.
Sci Rep ; 8(1): 12125, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30108230

RESUMEN

Mouse Tmc1 and Tmc2 are required for sensory transduction in cochlear and vestibular hair cells. Homozygous Tmc1∆/∆ mice are deaf, Tmc2∆/∆ mice have normal hearing, and double homozygous Tmc1∆/∆; Tmc2∆/∆ mice have deafness and profound vestibular dysfunction. These phenotypes are consistent with their different spatiotemporal expression patterns. Tmc1 expression is persistent in cochlear and vestibular hair cells, whereas Tmc2 expression is transient in cochlear hair cells but persistent in vestibular hair cells. On the basis of these findings, we hypothesized that persistent Tmc2 expression in mature cochlear hair cells could restore auditory function in Tmc1∆/∆ mice. To express Tmc2 in mature cochlear hair cells, we generated a transgenic mouse line, Tg[PTmc1::Tmc2], in which Tmc2 cDNA is expressed under the control of the Tmc1 promoter. The Tg[PTmc1::Tmc2] transgene slightly but significantly restored hearing in young Tmc1∆/∆ mice, though hearing thresholds were elevated with age. The elevation of hearing thresholds was associated with deterioration of sensory transduction in inner hair cells and loss of outer hair cell function. Although sensory transduction was retained in outer hair cells, their stereocilia eventually degenerated. These results indicate distinct roles and requirements for Tmc1 and Tmc2 in mature cochlear hair cells.


Asunto(s)
Células Ciliadas Auditivas/patología , Pérdida Auditiva Sensorineural/patología , Proteínas de la Membrana/metabolismo , Estereocilios/patología , Animales , Modelos Animales de Enfermedad , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/ultraestructura , Células Ciliadas Vestibulares/metabolismo , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Pruebas Auditivas , Homocigoto , Humanos , Mecanotransducción Celular , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Mutación , Técnicas de Placa-Clamp , Regiones Promotoras Genéticas/genética , Estereocilios/ultraestructura
12.
Sci Rep ; 8(1): 12124, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30108254

RESUMEN

Recent work has demonstrated that transmembrane channel-like 1 protein (TMC1) is an essential component of the sensory transduction complex in hair cells of the inner ear. A closely related homolog, TMC2, is expressed transiently in the neonatal mouse cochlea and can enable sensory transduction in Tmc1-null mice during the first postnatal week. Both TMC1 and TMC2 are expressed at adult stages in mouse vestibular hair cells. The extent to which TMC1 and TMC2 can substitute for each other is unknown. Several biophysical differences between TMC1 and TMC2 suggest these proteins perform similar but not identical functions. To investigate these differences, and whether TMC2 can substitute for TMC1 in mature hair cells, we generated a knock-in mouse model allowing Cre-inducible expression of Tmc2. We assayed for changes in hair cell sensory transduction and auditory and vestibular function in Tmc2 knockin mice (Tm[Tmc2]) in the presence or absence of endogenous Tmc1, Tmc2 or both. Our results show that expression of Tm[TMC2] restores sensory transduction in vestibular hair cells and transiently in cochlear hair cells in the absence of TMC1. The cellular rescue leads to recovery of balance but not auditory function. We conclude that TMC1 provides some additional necessary function, not provided by TMC2.


Asunto(s)
Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Vestibulares/metabolismo , Proteínas de la Membrana/metabolismo , Equilibrio Postural/fisiología , Animales , Técnicas de Sustitución del Gen , Audición/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Modelos Animales , Transgenes/genética
13.
Front Cell Neurosci ; 12: 41, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29515374

RESUMEN

Hair cells of the inner ear transduce mechanical stimuli like sound or head movements into electrical signals, which are propagated to the central nervous system. The hair-cell mechanotransduction channel remains unidentified. We tested whether three transient receptor channel (TRP) family members, TRPV6, TRPM6 and TRPM7, were necessary for transduction. TRPV6 interacted with USH1C (harmonin), a scaffolding protein that participates in transduction. Using a cysteine-substitution knock-in mouse line and methanethiosulfonate (MTS) reagents selective for this allele, we found that inhibition of TRPV6 had no effect on transduction in mouse cochlear hair cells. TRPM6 and TRPM7 each interacted with the tip-link component PCDH15 in cultured eukaryotic cells, which suggested they might be part of the transduction complex. Cochlear hair cell transduction was not affected by manipulations of Mg2+, however, which normally perturbs TRPM6 and TRPM7. To definitively examine the role of these two channels in transduction, we showed that deletion of either or both of their genes selectively in hair cells had no effect on auditory function. We suggest that TRPV6, TRPM6 and TRPM7 are unlikely to be the pore-forming subunit of the hair-cell transduction channel.

14.
Nature ; 553(7687): 217-221, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29258297

RESUMEN

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.


Asunto(s)
Proteínas Asociadas a CRISPR/administración & dosificación , Edición Génica/métodos , Genes Dominantes/genética , Terapia Genética/métodos , Pérdida Auditiva/genética , Estimulación Acústica , Alelos , Animales , Animales Recién Nacidos , Umbral Auditivo , Secuencia de Bases , Proteínas Asociadas a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/uso terapéutico , Sistemas CRISPR-Cas , Supervivencia Celular , Cóclea/citología , Cóclea/metabolismo , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Fibroblastos , Células Ciliadas Auditivas/citología , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/prevención & control , Humanos , Liposomas , Masculino , Proteínas de la Membrana/genética , Ratones , Reflejo de Sobresalto
15.
Nat Biotechnol ; 35(3): 280-284, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28165475

RESUMEN

Efforts to develop gene therapies for hearing loss have been hampered by the lack of safe, efficient, and clinically relevant delivery modalities. Here we demonstrate the safety and efficiency of Anc80L65, a rationally designed synthetic vector, for transgene delivery to the mouse cochlea. Ex vivo transduction of mouse organotypic explants identified Anc80L65 from a set of other adeno-associated virus (AAV) vectors as a potent vector for the cochlear cell targets. Round window membrane injection resulted in highly efficient transduction of inner and outer hair cells in mice, a substantial improvement over conventional AAV vectors. Anc80L65 round window injection was well tolerated, as indicated by sensory cell function, hearing and vestibular function, and immunologic parameters. The ability of Anc80L65 to target outer hair cells at high rates, a requirement for restoration of complex auditory function, may enable future gene therapies for hearing and balance disorders.


Asunto(s)
Cóclea/fisiología , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Plásmidos/genética , Transducción Genética/métodos , Animales , Cóclea/virología , Ratones , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación
16.
Nat Biotechnol ; 35(3): 264-272, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28165476

RESUMEN

Because there are currently no biological treatments for hearing loss, we sought to advance gene therapy approaches to treat genetic deafness. We focused on Usher syndrome, a devastating genetic disorder that causes blindness, balance disorders and profound deafness, and studied a knock-in mouse model, Ush1c c.216G>A, for Usher syndrome type IC (USH1C). As restoration of complex auditory and balance function is likely to require gene delivery systems that target auditory and vestibular sensory cells with high efficiency, we delivered wild-type Ush1c into the inner ear of Ush1c c.216G>A mice using a synthetic adeno-associated viral vector, Anc80L65, shown to transduce 80-90% of sensory hair cells. We demonstrate recovery of gene and protein expression, restoration of sensory cell function, rescue of complex auditory function and recovery of hearing and balance behavior to near wild-type levels. The data represent unprecedented recovery of inner ear function and suggest that biological therapies to treat deafness may be suitable for translation to humans with genetic inner ear disorders.


Asunto(s)
Proteínas Portadoras/genética , Terapia Genética/métodos , Pérdida Auditiva Sensorineural/terapia , Síndromes de Usher/genética , Síndromes de Usher/terapia , Enfermedades Vestibulares/terapia , Animales , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Femenino , Técnicas de Sustitución del Gen , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación , Plásmidos/genética , Recuperación de la Función/genética , Resultado del Tratamiento , Enfermedades Vestibulares/diagnóstico , Enfermedades Vestibulares/genética
17.
Am J Hum Genet ; 98(6): 1101-1113, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27236922

RESUMEN

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.


Asunto(s)
Vías Auditivas , Pérdida Auditiva/prevención & control , Proteínas de la Membrana/fisiología , MicroARNs/genética , Mutación Missense/genética , Animales , Dependovirus/genética , Pérdida Auditiva/etiología , Pérdida Auditiva/patología , Humanos , Mecanotransducción Celular , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , MicroARNs/administración & dosificación , Interferencia de ARN
18.
Cell Rep ; 12(10): 1606-17, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26321635

RESUMEN

Mechanosensitive ion channels at stereocilia tips mediate mechanoelectrical transduction (MET) in inner ear sensory hair cells. Transmembrane channel-like 1 and 2 (TMC1 and TMC2) are essential for MET and are hypothesized to be components of the MET complex, but evidence for their predicted spatiotemporal localization in stereocilia is lacking. Here, we determine the stereocilia localization of the TMC proteins in mice expressing TMC1-mCherry and TMC2-AcGFP. Functionality of the tagged proteins was verified by transgenic rescue of MET currents and hearing in Tmc1(Δ/Δ);Tmc2(Δ/Δ) mice. TMC1-mCherry and TMC2-AcGFP localize along the length of immature stereocilia. However, as hair cells develop, the two proteins localize predominantly to stereocilia tips. Both TMCs are absent from the tips of the tallest stereocilia, where MET activity is not detectable. This distribution was confirmed for the endogenous proteins by immunofluorescence. These data are consistent with TMC1 and TMC2 being components of the stereocilia MET channel complex.


Asunto(s)
Cilios/metabolismo , Células Ciliadas Auditivas Internas/fisiología , Proteínas de la Membrana/metabolismo , Animales , Cilios/ultraestructura , Femenino , Expresión Génica , Células Ciliadas Auditivas Internas/ultraestructura , Masculino , Mecanotransducción Celular , Proteínas de la Membrana/genética , Ratones Transgénicos , Transporte de Proteínas
19.
Curr Opin Neurobiol ; 34: 165-71, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26218316

RESUMEN

Years of searching and researching have finally yielded a few leads in the quest to identify molecules required for mechanosensory transduction in the mammalian inner ear. Studies of human and mouse genetics have raised the profile of several molecules that are crucial for the function sensory hair cells. Follow up studies have begun to define the molecular function and biochemical interactions of several key proteins. These studies have exposed a sensory transduction apparatus that is more complex than originally envisioned and have reinvigorated the search for additional molecular components required for normal inner ear function.


Asunto(s)
Oído Interno/citología , Audición/fisiología , Mastodinia , Células Receptoras Sensoriales/fisiología , Transducción de Señal/fisiología , Animales , Audición/genética , Humanos , Transducción de Señal/genética
20.
Sci Transl Med ; 7(295): 295ra108, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26157030

RESUMEN

Genetic hearing loss accounts for up to 50% of prelingual deafness worldwide, yet there are no biologic treatments currently available. To investigate gene therapy as a potential biologic strategy for restoration of auditory function in patients with genetic hearing loss, we tested a gene augmentation approach in mouse models of genetic deafness. We focused on DFNB7/11 and DFNA36, which are autosomal recessive and dominant deafnesses, respectively, caused by mutations in transmembrane channel-like 1 (TMC1). Mice that carry targeted deletion of Tmc1 or a dominant Tmc1 point mutation, known as Beethoven, are good models for human DFNB7/11 and DFNA36. We screened several adeno-associated viral (AAV) serotypes and promoters and identified AAV2/1 and the chicken ß-actin (Cba) promoter as an efficient combination for driving the expression of exogenous Tmc1 in inner hair cells in vivo. Exogenous Tmc1 or its closely related ortholog, Tmc2, were capable of restoring sensory transduction, auditory brainstem responses, and acoustic startle reflexes in otherwise deaf mice, suggesting that gene augmentation with Tmc1 or Tmc2 is well suited for further development as a strategy for restoration of auditory function in deaf patients who carry TMC1 mutations.


Asunto(s)
Sordera/terapia , Terapia Genética , Audición , Proteínas de la Membrana/genética , Animales , Sordera/genética , Dependovirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA