Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 31(8): 2439-2453, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37312453

RESUMO

Usher syndrome type 1F (USH1F), characterized by congenital lack of hearing and balance and progressive loss of vision, is caused by mutations in the PCDH15 gene. In the Ashkenazi population, a recessive truncation mutation accounts for a large proportion of USH1F cases. The truncation is caused by a single C→T mutation, which converts an arginine codon to a stop (R245X). To test the potential for base editors to revert this mutation, we developed a humanized Pcdh15R245X mouse model for USH1F. Mice homozygous for the R245X mutation were deaf and exhibited profound balance deficits, while heterozygous mice were unaffected. Here we show that an adenine base editor (ABE) is capable of reversing the R245X mutation to restore the PCDH15 sequence and function. We packaged a split-intein ABE into dual adeno-associated virus (AAV) vectors and delivered them into cochleas of neonatal USH1F mice. Hearing was not restored in a Pcdh15 constitutive null mouse despite base editing, perhaps because of early disorganization of cochlear hair cells. However, injection of vectors encoding the split ABE into a late-deletion conditional Pcdh15 knockout rescued hearing. This study demonstrates the ability of an ABE to correct the PCDH15 R245X mutation in the cochlea and restore hearing.


Assuntos
Síndromes de Usher , Camundongos , Animais , Síndromes de Usher/genética , Síndromes de Usher/terapia , Edição de Genes , Mutação , Audição/genética , Caderinas/genética
2.
Nat Commun ; 14(1): 2400, 2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37100771

RESUMO

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.


Assuntos
Orelha Interna , Síndromes de Usher , Animais , Camundongos , Caderinas/metabolismo , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Audição/genética , Síndromes de Usher/genética , Síndromes de Usher/terapia , Proteínas Relacionadas a Caderinas/metabolismo
3.
Elife ; 102021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34665127

RESUMO

Excess noise damages sensory hair cells, resulting in loss of synaptic connections with auditory nerves and, in some cases, hair-cell death. The cellular mechanisms underlying mechanically induced hair-cell damage and subsequent repair are not completely understood. Hair cells in neuromasts of larval zebrafish are structurally and functionally comparable to mammalian hair cells but undergo robust regeneration following ototoxic damage. We therefore developed a model for mechanically induced hair-cell damage in this highly tractable system. Free swimming larvae exposed to strong water wave stimulus for 2 hr displayed mechanical injury to neuromasts, including afferent neurite retraction, damaged hair bundles, and reduced mechanotransduction. Synapse loss was observed in apparently intact exposed neuromasts, and this loss was exacerbated by inhibiting glutamate uptake. Mechanical damage also elicited an inflammatory response and macrophage recruitment. Remarkably, neuromast hair-cell morphology and mechanotransduction recovered within hours following exposure, suggesting severely damaged neuromasts undergo repair. Our results indicate functional changes and synapse loss in mechanically damaged lateral-line neuromasts that share key features of damage observed in noise-exposed mammalian ear. Yet, unlike the mammalian ear, mechanical damage to neuromasts is rapidly reversible.


Assuntos
Sistema da Linha Lateral/lesões , Mecanorreceptores/fisiologia , Mecanotransdução Celular , Sinapses/fisiologia , Peixe-Zebra/lesões , Animais , Fenômenos Biomecânicos , Células Ciliadas Auditivas/fisiologia , Sistema da Linha Lateral/fisiologia , Peixe-Zebra/fisiologia
4.
Nat Commun ; 10(1): 5530, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31797926

RESUMO

The adult mammalian inner ear lacks the capacity to divide or regenerate. Damage to inner ear generally leads to permanent hearing loss in humans. Here, we present that reprogramming of the adult inner ear induces renewed proliferation and regeneration of inner ear cell types. Co-activation of cell cycle activator Myc and inner ear progenitor gene Notch1 induces robust proliferation of diverse adult cochlear sensory epithelial cell types. Transient MYC and NOTCH activities enable adult supporting cells to respond to transcription factor Atoh1 and efficiently transdifferentiate into hair cell-like cells. Furthermore, we uncover that mTOR pathway participates in MYC/NOTCH-mediated proliferation and regeneration. These regenerated hair cell-like cells take up the styryl dye FM1-43 and are likely to form connections with adult spiral ganglion neurons, supporting that Myc and Notch1 co-activation is sufficient to reprogram fully mature supporting cells to proliferate and regenerate hair cell-like cells in adult mammalian auditory organs.


Assuntos
Proliferação de Células/fisiologia , Cóclea/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Regeneração/fisiologia , Animais , Proliferação de Células/genética , Cóclea/citologia , Cóclea/metabolismo , Orelha Interna/citologia , Orelha Interna/metabolismo , Orelha Interna/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Gânglios Sensitivos/citologia , Gânglios Sensitivos/metabolismo , Gânglios Sensitivos/fisiologia , Regulação da Expressão Gênica , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Regeneração/genética
5.
Mol Ther Methods Clin Dev ; 13: 1-13, 2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-30581889

RESUMO

Hereditary hearing loss often results from mutation of genes expressed by cochlear hair cells. Gene addition using AAV vectors has shown some efficacy in mouse models, but clinical application requires two additional advances. First, new AAV capsids must mediate efficient transgene expression in both inner and outer hair cells of the cochlea. Second, to have the best chance of clinical translation, these new vectors must also transduce hair cells in non-human primates. Here, we show that an AAV9 capsid variant, PHP.B, produces efficient transgene expression of a GFP reporter in both inner and outer hair cells of neonatal mice. We show also that AAV9-PHP.B mediates almost complete transduction of inner and outer HCs in a non-human primate. In a mouse model of Usher syndrome type 3A deafness (gene CLRN1), we use AAV9-PHP.B encoding Clrn1 to partially rescue hearing. Thus, we have identified a vector with promise for clinical treatment of hereditary hearing disorders, and we demonstrate, for the first time, viral transduction of the inner ear of a primate with an AAV vector.

6.
Nat Biotechnol ; 35(3): 264-272, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28165476

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Terapia Genética/métodos , Perda Auditiva Neurossensorial/terapia , Síndromes de Usher/genética , Síndromes de Usher/terapia , Doenças Vestibulares/terapia , Animais , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Feminino , Técnicas de Introdução de Genes , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/administração & dosagem , Plasmídeos/genética , Recuperação de Função Fisiológica/genética , Resultado do Tratamento , Doenças Vestibulares/diagnóstico , Doenças Vestibulares/genética
7.
Mol Ther ; 25(2): 379-391, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28082074

RESUMO

Adeno-associated virus (AAV) is a safe and effective vector for gene therapy for retinal disorders. Gene therapy for hearing disorders is not as advanced, in part because gene delivery to sensory hair cells of the inner ear is inefficient. Although AAV transduces the inner hair cells of the mouse cochlea, outer hair cells remain refractory to transduction. Here, we demonstrate that a vector, exosome-associated AAV (exo-AAV), is a potent carrier of transgenes to all inner ear hair cells. Exo-AAV1-GFP is more efficient than conventional AAV1-GFP, both in mouse cochlear explants in vitro and with direct cochlear injection in vivo. Exo-AAV shows no toxicity in vivo, as assayed by tests of auditory and vestibular function. Finally, exo-AAV1 gene therapy partially rescues hearing in a mouse model of hereditary deafness (lipoma HMGIC fusion partner-like 5/tetraspan membrane protein of hair cell stereocilia [Lhfpl5/Tmhs-/-]). Exo-AAV is a powerful gene delivery system for hair cell research and may be useful for gene therapy for deafness.


Assuntos
Dependovirus/genética , Exossomos/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células Ciliadas Auditivas Internas/metabolismo , Audição/genética , Animais , Células Cultivadas , Dependovirus/classificação , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Feminino , Expressão Gênica , Genes Reporter , Terapia Genética , Vetores Genéticos/administração & dosagem , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Transdução Genética , Transgenes
8.
Elife ; 42015 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-26302205

RESUMO

The precise assembly of inner ear hair cell stereocilia into rows of increasing height is critical for mechanotransduction and the sense of hearing. Yet, how the lengths of actin-based stereocilia are regulated remains poorly understood. Mutations of the molecular motor myosin 15 stunt stereocilia growth and cause deafness. We found that hair cells express two isoforms of myosin 15 that differ by inclusion of an 133-kDa N-terminal domain, and that these isoforms can selectively traffic to different stereocilia rows. Using an isoform-specific knockout mouse, we show that hair cells expressing only the small isoform remarkably develop normal stereocilia bundles. However, a critical subset of stereocilia with active mechanotransducer channels subsequently retracts. The larger isoform with the 133-kDa N-terminal domain traffics to these specialized stereocilia and prevents disassembly of their actin core. Our results show that myosin 15 isoforms can navigate between functionally distinct classes of stereocilia, and are independently required to assemble and then maintain the intricate hair bundle architecture.


Assuntos
Audição , Miosinas/genética , Miosinas/metabolismo , Estereocílios/metabolismo , Estereocílios/fisiologia , Animais , Orelha Interna/fisiologia , Células Ciliadas Auditivas/fisiologia , Camundongos , Camundongos Knockout , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico
9.
J Assoc Res Otolaryngol ; 12(6): 729-40, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21879401

RESUMO

Aminoglycoside ototoxicity involves the accumulation of antibiotic molecules in the inner ear hair cells and the subsequent degeneration of these cells. The exact route of entry of aminoglycosides into the hair cells in vivo is still unknown. Similar to other small organic cations, aminoglycosides could be brought into the cell by endocytosis or permeate through large non-selective cation channels, such as mechanotransduction channels or ATP-gated P2X channels. Here, we show that the aminoglycoside antibiotic gentamicin can enter mouse outer hair cells (OHCs) via TRPA1, non-selective cation channels activated by certain pungent compounds and by endogenous products of lipid peroxidation. Using conventional and perforated whole-cell patch clamp recordings, we found that application of TRPA1 agonists initiates inward current responses in wild-type OHCs, but not in OHCs of homozygous Trpa1 knockout mice. Similar responses consistent with the activation of non-selective cation channels were observed in heterologous cells transfected with mouse Trpa1. Upon brief activation with TRPA1 agonists, Trpa1-transfected cells become loaded with fluorescent gentamicin-Texas Red conjugate (GTTR). This uptake was not observed in mock-transfected or non-transfected cells. In mouse organ of Corti explants, TRPA1 activation resulted in the rapid entry of GTTR and another small cationic dye, FM1-43, in OHCs and some supporting cells, even when hair cell mechanotransduction was disrupted by pre-incubation in calcium-free solution. This TRPA1-mediated entry of GTTR and FM1-43 into OHCs was observed in wild-type but not in Trpa1 knockout mice and was not blocked by PPADS, a non-selective blocker of P2X channels. Notably, TRPA1 channels in mouse OHCs were activated by 4-hydroxynonenal, an endogenous molecule that is known to be generated during episodes of oxidative stress and accumulate in the cochlea after noise exposure. We concluded that TRPA1 channels may provide a novel pathway for the entry of aminoglycosides into OHCs.


Assuntos
Aminoglicosídeos/farmacocinética , Aminoglicosídeos/toxicidade , Gentamicinas/farmacocinética , Gentamicinas/toxicidade , Células Ciliadas Auditivas Externas , Canais de Potencial de Receptor Transitório/metabolismo , Aldeídos/farmacologia , Animais , Células COS , Cátions/farmacocinética , Chlorocebus aethiops , Inibidores de Cisteína Proteinase/farmacologia , Corantes Fluorescentes/farmacocinética , Genótipo , Células HEK293 , Células Ciliadas Auditivas Externas/efeitos dos fármacos , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Técnicas de Patch-Clamp , Compostos de Piridínio/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/genética , Xantenos/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA