Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
2.
Development ; 150(19)2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37796037

RESUMO

Inner ear development requires the coordination of cell types from distinct epithelial, mesenchymal and neuronal lineages. Although we have learned much from animal models, many details about human inner ear development remain elusive. We recently developed an in vitro model of human inner ear organogenesis using pluripotent stem cells in a 3D culture, fostering the growth of a sensorineural circuit, including hair cells and neurons. Despite previously characterizing some cell types, many remain undefined. This study aimed to chart the in vitro development timeline of the inner ear organoid to understand the mechanisms at play. Using single-cell RNA sequencing at ten stages during the first 36 days of differentiation, we tracked the evolution from pluripotency to various ear cell types after exposure to specific signaling modulators. Our findings showcase gene expression that influences differentiation, identifying a plethora of ectodermal and mesenchymal cell types. We also discern aspects of the organoid model consistent with in vivo development, while highlighting potential discrepancies. Our study establishes the Inner Ear Organoid Developmental Atlas (IODA), offering deeper insights into human biology and improving inner ear tissue differentiation.


Assuntos
Orelha Interna , Animais , Humanos , Orelha Interna/metabolismo , Células Ciliadas Auditivas , Organoides , Células Cultivadas , Diferenciação Celular/genética
3.
Cell Rep ; 42(6): 112623, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37289589

RESUMO

Inner ear disorders are among the most common congenital abnormalities; however, current tissue culture models lack the cell type diversity to study these disorders and normal otic development. Here, we demonstrate the robustness of human pluripotent stem cell-derived inner ear organoids (IEOs) and evaluate cell type heterogeneity by single-cell transcriptomics. To validate our findings, we construct a single-cell atlas of human fetal and adult inner ear tissue. Our study identifies various cell types in the IEOs including periotic mesenchyme, type I and type II vestibular hair cells, and developing vestibular and cochlear epithelium. Many genes linked to congenital inner ear dysfunction are confirmed to be expressed in these cell types. Additional cell-cell communication analysis within IEOs and fetal tissue highlights the role of endothelial cells on the developing sensory epithelium. These findings provide insights into this organoid model and its potential applications in studying inner ear development and disorders.


Assuntos
Células Endoteliais , Vestíbulo do Labirinto , Humanos , Cóclea/metabolismo , Epitélio/metabolismo , Organoides/metabolismo
4.
Curr Opin Genet Dev ; 76: 101954, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35853286

RESUMO

The vertebrate inner ear contains a diversity of unique cell types arranged in a particularly complex 3D cytoarchitecture. Both of these features are integral to the proper development, function, and maintenance of hearing and balance. Since the elucidation of the timing and delivery of signaling molecules to produce inner ear sensory cells, supporting cells, and neurons from human induced pluripotent stem cells, we have entered a revolution using organ-like 'otic organoid' cultures to explore inner ear specific genetic programs, developmental rules, and potential therapeutics. This review aims to highlight a selection of reviews and primary research papers from the past two years of particular merit that use otic organoids to investigate the broadly defined topics of cell reprogramming, regeneration, and repair.


Assuntos
Orelha Interna , Células-Tronco Pluripotentes Induzidas , Diferenciação Celular/genética , Humanos , Organogênese/fisiologia , Organoides
5.
Sci Adv ; 8(12): eabm1568, 2022 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-35333573

RESUMO

Human TMEM175, a noncanonical potassium (K+) channel in endolysosomes, contributes to their pH stability and is implicated in the pathogenesis of Parkinson's disease (PD). Structurally, the TMEM175 family exhibits an architecture distinct from canonical potassium channels, as it lacks the typical TVGYG selectivity filter. Here, we show that human TMEM175 not only exhibits pH-dependent structural changes that reduce K+ permeation at acidic pH but also displays proton permeation. TMEM175 constitutively conducts K+ at pH 7.4 but displays reduced K+ permeation at lower pH. In contrast, proton current through TMEM175 increases with decreasing pH because of the increased proton gradient. Molecular dynamics simulation, structure-based mutagenesis, and electrophysiological analysis suggest that K+ ions and protons share the same permeation pathway. The M393T variant of human TMEM175 associated with PD shows reduced function in both K+ and proton permeation. Together, our structural and electrophysiological analysis reveals a mechanism of TMEM175 regulation by pH.

6.
Sci Adv ; 7(51): eabi7629, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34910522

RESUMO

Hearing loss affects an estimated 466 million people worldwide, with a substantial fraction due to genetic causes. Approximately 16% of genetic hearing loss is caused by pathogenic mutations in STRC, a gene that encodes the protein stereocilin. To develop gene therapy strategies for patients with STRC hearing loss, we generated a mouse model with a targeted deletion in the Strc gene. We devised a novel dual-vector approach to circumvent the size limitation of AAV vectors and drive expression of full-length STRC protein. To target outer hair cells, which are difficult to transduce, we used synthetic AAV9-PHP.B vectors for efficient dual-vector transduction. We report robust recovery of exogenous STRC expression in outer hair cells of Strc-deficient mice, recovery of hair bundle morphology, substantially improved cochlear amplification, and enhanced auditory sensitivity. The data raise the prospect that our strategy could benefit ~2.3 million patients worldwide affected by STRC mutations.

7.
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
8.
Mol Ther ; 28(12): 2662-2676, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-32818431

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas do Citoesqueleto/genética , Orelha Média/efeitos dos fármacos , Terapia Genética/métodos , Células Ciliadas Auditivas/efeitos dos fármacos , Mutação , Oligonucleotídeos Antissenso/administração & dosagem , Síndromes de Usher/genética , Síndromes de Usher/terapia , Vestíbulo do Labirinto/efeitos dos fármacos , Administração Tópica , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Células Ciliadas Auditivas/metabolismo , Audição/efeitos dos fármacos , Injeções , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Membrana Timpânica/efeitos dos fármacos , Vestíbulo do Labirinto/metabolismo
9.
Hear Res ; 394: 107882, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31980281

RESUMO

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.


Assuntos
Orelha Interna , Animais , Infecções por Citomegalovirus , Dependovirus/genética , Terapia Genética , Vetores Genéticos , Células Ciliadas Auditivas Externas , Camundongos , Sáculo e Utrículo , Sinapsinas/genética
10.
Nat Med ; 25(7): 1123-1130, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31270503

RESUMO

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.


Assuntos
Alelos , Edição de Genes , Perda Auditiva Neurossensorial/prevenção & controle , Proteínas de Membrana/genética , Animais , Proteína 9 Associada à CRISPR/fisiologia , Linhagem Celular , Células Cultivadas , Dependovirus/genética , Modelos Animais de Doenças , Perda Auditiva Neurossensorial/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL
11.
Nat Commun ; 10(1): 734, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30737404

RESUMO

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.

12.
Nat Commun ; 10(1): 236, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670701

RESUMO

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.


Assuntos
Surdez/genética , Predisposição Genética para Doença , Terapia Genética/métodos , Perda Auditiva/genética , Doenças do Labirinto/genética , Proteínas de Membrana/genética , Animais , Surdez/terapia , Células Ciliadas Auditivas/fisiologia , Células Ciliadas Vestibulares/fisiologia , Perda Auditiva/terapia , Doenças do Labirinto/terapia , Camundongos , Camundongos Mutantes
13.
Sci Rep ; 8(1): 12124, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30108254

RESUMO

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.


Assuntos
Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Vestibulares/metabolismo , Proteínas de Membrana/metabolismo , Equilíbrio Postural/fisiologia , Animais , Técnicas de Introdução de Genes , Audição/fisiologia , Mecanotransdução Celular/fisiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Modelos Animais , Transgenes/genética
14.
Neuron ; 99(4): 736-753.e6, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30138589

RESUMO

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.


Assuntos
Células Ciliadas Auditivas Internas/fisiologia , Mecanotransdução Celular/fisiologia , Proteínas de Membrana/química , Proteínas de Membrana/fisiologia , Porinas/química , Porinas/fisiologia , Animais , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA