Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Nature ; 592(7853): 195-204, 2021 04.
Article in English | MEDLINE | ID: mdl-33828315

ABSTRACT

The move from reading to writing the human genome offers new opportunities to improve human health. The United States National Institutes of Health (NIH) Somatic Cell Genome Editing (SCGE) Consortium aims to accelerate the development of safer and more-effective methods to edit the genomes of disease-relevant somatic cells in patients, even in tissues that are difficult to reach. Here we discuss the consortium's plans to develop and benchmark approaches to induce and measure genome modifications, and to define downstream functional consequences of genome editing within human cells. Central to this effort is a rigorous and innovative approach that requires validation of the technology through third-party testing in small and large animals. New genome editors, delivery technologies and methods for tracking edited cells in vivo, as well as newly developed animal models and human biological systems, will be assembled-along with validated datasets-into an SCGE Toolkit, which will be disseminated widely to the biomedical research community. We visualize this toolkit-and the knowledge generated by its applications-as a means to accelerate the clinical development of new therapies for a wide range of conditions.


Subject(s)
Cells/metabolism , Gene Editing/methods , Genome, Human/genetics , National Institutes of Health (U.S.)/organization & administration , Animals , Genetic Therapy , Goals , Humans , United States
2.
Proc Natl Acad Sci U S A ; 120(17): e2215253120, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37068229

ABSTRACT

Strategies to overcome irreversible cochlear hair cell (HC) damage and loss in mammals are of vital importance to hearing recovery in patients with permanent hearing loss. In mature mammalian cochlea, co-activation of Myc and Notch1 reprograms supporting cells (SC) and promotes HC regeneration. Understanding of the underlying mechanisms may aid the development of a clinically relevant approach to achieve HC regeneration in the nontransgenic mature cochlea. By single-cell RNAseq, we show that MYC/NICD "rejuvenates" the adult mouse cochlea by activating multiple pathways including Wnt and cyclase activator of cyclic AMP (cAMP), whose blockade suppresses HC-like cell regeneration despite Myc/Notch activation. We screened and identified a combination (the cocktail) of drug-like molecules composing of small molecules and small interfering RNAs to activate the pathways of Myc, Notch1, Wnt and cAMP. We show that the cocktail effectively replaces Myc and Notch1 transgenes and reprograms fully mature wild-type (WT) SCs for HC-like cells regeneration in vitro. Finally, we demonstrate the cocktail is capable of reprogramming adult cochlea for HC-like cells regeneration in WT mice with HC loss in vivo. Our study identifies a strategy by a clinically relevant approach to reprogram mature inner ear for HC-like cells regeneration, laying the foundation for hearing restoration by HC regeneration.


Subject(s)
Ear, Inner , Hair Cells, Auditory , Mice , Animals , Cell Proliferation/physiology , Hair Cells, Auditory/physiology , Ear, Inner/metabolism , Cochlea/physiology , Regeneration/physiology , Mammals
3.
Lancet ; 403(10441): 2317-2325, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38280389

ABSTRACT

BACKGROUND: Autosomal recessive deafness 9, caused by mutations of the OTOF gene, is characterised by congenital or prelingual, severe-to-complete, bilateral hearing loss. However, no pharmacological treatment is currently available for congenital deafness. In this Article, we report the safety and efficacy of gene therapy with an adeno-associated virus (AAV) serotype 1 carrying a human OTOF transgene (AAV1-hOTOF) as a treatment for children with autosomal recessive deafness 9. METHODS: This single-arm, single-centre trial enrolled children (aged 1-18 years) with severe-to-complete hearing loss and confirmed mutations in both alleles of OTOF, and without bilateral cochlear implants. A single injection of AAV1-hOTOF was administered into the cochlea through the round window. The primary endpoint was dose-limiting toxicity at 6 weeks after injection. Auditory function and speech were assessed by appropriate auditory perception evaluation tools. All analyses were done according to the intention-to-treat principle. This trial is registered with Chinese Clinical Trial Registry, ChiCTR2200063181, and is ongoing. FINDINGS: Between Oct 19, 2022, and June 9, 2023, we screened 425 participants for eligibility and enrolled six children for AAV1-hOTOF gene therapy (one received a dose of 9 × 1011 vector genomes [vg] and five received 1·5 × 1012 vg). All participants completed follow-up visits up to week 26. No dose-limiting toxicity or serious adverse events occurred. In total, 48 adverse events were observed; 46 (96%) were grade 1-2 and two (4%) were grade 3 (decreased neutrophil count in one participant). Five children had hearing recovery, shown by a 40-57 dB reduction in the average auditory brainstem response (ABR) thresholds at 0·5-4·0 kHz. In the participant who received the 9 × 1011 vg dose, the average ABR threshold was improved from greater than 95 dB at baseline to 68 dB at 4 weeks, 53 dB at 13 weeks, and 45 dB at 26 weeks. In those who received 1·5 × 1012 AAV1-hOTOF, the average ABR thresholds changed from greater than 95 dB at baseline to 48 dB, 38 dB, 40 dB, and 55 dB in four children with hearing recovery at 26 weeks. Speech perception was improved in participants who had hearing recovery. INTERPRETATION: AAV1-hOTOF gene therapy is safe and efficacious as a novel treatment for children with autosomal recessive deafness 9. FUNDING: National Natural Science Foundation of China, National Key R&D Program of China, Science and Technology Commission of Shanghai Municipality, and Shanghai Refreshgene Therapeutics.


Subject(s)
Dependovirus , Genetic Therapy , Humans , Genetic Therapy/methods , Dependovirus/genetics , Child , Male , Child, Preschool , Female , Adolescent , Infant , Genetic Vectors , Treatment Outcome , Deafness/genetics , Deafness/therapy , Mutation , Membrane Proteins
4.
Nature ; 553(7687): 217-221, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29258297

ABSTRACT

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.


Subject(s)
CRISPR-Associated Proteins/administration & dosage , Gene Editing/methods , Genes, Dominant/genetics , Genetic Therapy/methods , Hearing Loss/genetics , Acoustic Stimulation , Alleles , Animals , Animals, Newborn , Auditory Threshold , Base Sequence , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/therapeutic use , CRISPR-Cas Systems , Cell Survival , Cochlea/cytology , Cochlea/metabolism , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Fibroblasts , Hair Cells, Auditory/cytology , Hearing Loss/physiopathology , Hearing Loss/prevention & control , Humans , Liposomes , Male , Membrane Proteins/genetics , Mice , Reflex, Startle
5.
Mol Ther ; 31(9): 2796-2810, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37244253

ABSTRACT

Patients with mutations in the TMPRSS3 gene suffer from recessive deafness DFNB8/DFNB10. For these patients, cochlear implantation is the only treatment option. Poor cochlear implantation outcomes are seen in some patients. To develop biological treatment for TMPRSS3 patients, we generated a knockin mouse model with a frequent human DFNB8 TMPRSS3 mutation. The Tmprss3A306T/A306T homozygous mice display delayed onset progressive hearing loss similar to human DFNB8 patients. Using AAV2 as a vector to carry a human TMPRSS3 gene, AAV2-hTMPRSS3 injection in the adult knockin mouse inner ear results in TMPRSS3 expression in the hair cells and the spiral ganglion neurons. A single AAV2-hTMPRSS3 injection in Tmprss3A306T/A306T mice of an average age of 18.5 months leads to sustained rescue of the auditory function to a level similar to wild-type mice. AAV2-hTMPRSS3 delivery rescues the hair cells and the spiral ganglions neurons. This study demonstrates successful gene therapy in an aged mouse model of human genetic deafness. It lays the foundation to develop AAV2-hTMPRSS3 gene therapy to treat DFNB8 patients, as a standalone therapy or in combination with cochlear implantation.


Subject(s)
Deafness , Serine Endopeptidases , Adult , Humans , Mice , Animals , Infant , Serine Endopeptidases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Hearing , Deafness/genetics , Deafness/therapy , Genetic Therapy , Neoplasm Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34050020

ABSTRACT

Genes that are primarily expressed in cochlear glia-like supporting cells (GLSs) have not been clearly associated with progressive deafness. Herein, we present a deafness locus mapped to chromosome 3p25.1 and an auditory neuropathy spectrum disorder (ANSD) gene, TMEM43, mainly expressed in GLSs. We identify p.(Arg372Ter) of TMEM43 by linkage analysis and exome sequencing in two large Asian families segregating ANSD, which is characterized by inability to discriminate speech despite preserved sensitivity to sound. The knock-in mouse with the p.(Arg372Ter) variant recapitulates a progressive hearing loss with histological abnormalities in GLSs. Mechanistically, TMEM43 interacts with the Connexin26 and Connexin30 gap junction channels, disrupting the passive conductance current in GLSs in a dominant-negative fashion when the p.(Arg372Ter) variant is introduced. Based on these mechanistic insights, cochlear implant was performed on three subjects, and speech discrimination was successfully restored. Our study highlights a pathological role of cochlear GLSs by identifying a deafness gene and its causal relationship with ANSD.


Subject(s)
Codon, Nonsense , Connexins/metabolism , Genes, Dominant , Hearing Loss, Central/genetics , Membrane Proteins/genetics , Animals , Cochlear Implantation , Female , Hearing Loss, Central/metabolism , Hearing Loss, Central/physiopathology , Hearing Loss, Central/surgery , Humans , Male , Mice , Mice, Inbred C57BL , Pedigree , Speech Perception
7.
Hum Mol Genet ; 30(11): 985-995, 2021 05 31.
Article in English | MEDLINE | ID: mdl-33791800

ABSTRACT

P2RX2 encodes the P2X2 receptor, which is an adenosine triphosphate (ATP) gated (purinoreceptor) ion channel. P2RX2 c. 178G > T (p.V60L) mutation was previously identified in two unrelated Chinese families, as the cause of human DFNA41, a form of dominant, early-onset and progressive sensorineural hearing loss. We generated and characterized a knock-in mouse model based on human p.V60L mutation that recapitulates the human phenotype. Heterozygous KI mice started to exhibit hearing loss at 21-day-old and progressed to deafness by 6-month-old. Vestibular dysfunction was also observed in mutant mice. Abnormal morphology of the inner hair cells and ribbon synapses was progressively observed in KI animals suggesting that P2rx2 plays a role in the membrane spatial location of the ribbon synapses. These results suggest that P2rx2 is essential for acoustic information transfer, which can be the molecular mechanism related to hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Receptors, Purinergic P2X2/genetics , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Gene Knock-In Techniques , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Sensorineural/pathology , Heterozygote , Humans , Mice , Mutation/genetics , Pedigree , Phenotype , Synapses/genetics , Synapses/pathology , Vestibular Diseases/genetics , Vestibular Diseases/pathology
8.
Mol Ther ; 30(1): 105-118, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34174443

ABSTRACT

Myosin VI(MYO6) is an unconventional myosin that is vital for auditory and vestibular function. Pathogenic variants in the human MYO6 gene cause autosomal-dominant or -recessive forms of hearing loss. Effective treatments for Myo6 mutation causing hearing loss are limited. We studied whether adeno-associated virus (AAV)-PHP.eB vector-mediated in vivo delivery of Staphylococcus aureus Cas9 (SaCas9-KKH)-single-guide RNA (sgRNA) complexes could ameliorate hearing loss in a Myo6WT/C442Y mouse model that recapitulated the phenotypes of human patients. The in vivo editing efficiency of the AAV-SaCas9-KKH-Myo6-g2 system on Myo6C442Y is 4.05% on average in Myo6WT/C442Y mice, which was ∼17-fold greater than editing efficiency of Myo6WT alleles. Rescue of auditory function was observed up to 5 months post AAV-SaCas9-KKH-Myo6-g2 injection in Myo6WT/C442Y mice. Meanwhile, shorter latencies of auditory brainstem response (ABR) wave I, lower distortion product otoacoustic emission (DPOAE) thresholds, increased cell survival rates, more regular hair bundle morphology, and recovery of inward calcium levels were also observed in the AAV-SaCas9-KKH-Myo6-g2-treated ears compared to untreated ears. These findings provide further reference for in vivo genome editing as a therapeutic treatment for various semi-dominant forms of hearing loss and other semi-dominant diseases.


Subject(s)
Gene Editing , Hearing Loss , Animals , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing , Hearing Loss/genetics , Hearing Loss/therapy , Humans , Mice , RNA, Guide, Kinetoplastida
9.
Adv Exp Med Biol ; 1185: 91-96, 2019.
Article in English | MEDLINE | ID: mdl-31884594

ABSTRACT

Mutations in USH2A gene account for most cases of Usher syndrome type II (USH2), characterized by a combination of congenital hearing loss and progressive vision loss. In particular, approximately 30% of USH2A patients harbor a single base pair deletion, c.2299delG, in exon 13 that creates a frameshift and premature stop codon, leading to a nonfunctional USH2A protein. The USH2A protein, also known as usherin, is an extremely large transmembrane protein (5202 aa), which limits the use of conventional AAV-mediated gene therapy; thus development of alternative approaches is required for the treatment of USH2A patients. As usherin contains multiple repetitive domains, we hypothesize that removal of one or more of those domains encoded by mutant exon(s) in the USH2A gene may reconstitute the reading frame and restore the production of a shortened yet adequately functional protein. In this study, we deleted the exon 12 of mouse Ush2a gene (corresponding to exon 13 of human USH2A) using CRISPR/Cas9-based exon-skipping approach and revealed that a shortened form of Ush2a that lacks exon 12 (Ush2a-∆Ex12) is produced and localized correctly in the cochlea. When the Ush2a-∆Ex12 allele is expressed on an Ush2a null background, the Ush2a-∆Ex12 protein can successfully restore the impaired hair cell structure and the auditory function in the Ush2a-/- mice. These results demonstrate that CRISPR/Cas9-based exon-skipping strategy holds a great therapeutic potential for the treatment of USH2A patients.


Subject(s)
Extracellular Matrix Proteins/genetics , Usher Syndromes/therapy , Animals , CRISPR-Cas Systems , Exons , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Usher Syndromes/genetics
10.
Proc Natl Acad Sci U S A ; 112(1): 166-71, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535395

ABSTRACT

The activation of cochlear progenitor cells is a promising approach for hair cell (HC) regeneration and hearing recovery. The mechanisms underlying the initiation of proliferation of postnatal cochlear progenitor cells and their transdifferentiation to HCs remain to be determined. We show that Notch inhibition initiates proliferation of supporting cells (SCs) and mitotic regeneration of HCs in neonatal mouse cochlea in vivo and in vitro. Through lineage tracing, we identify that a majority of the proliferating SCs and mitotic-generated HCs induced by Notch inhibition are derived from the Wnt-responsive leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5(+)) progenitor cells. We demonstrate that Notch inhibition removes the brakes on the canonical Wnt signaling and promotes Lgr5(+) progenitor cells to mitotically generate new HCs. Our study reveals a new function of Notch signaling in limiting proliferation and regeneration potential of postnatal cochlear progenitor cells, and provides a new route to regenerate HCs from progenitor cells by interrupting the interaction between the Notch and Wnt pathways.


Subject(s)
Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Mitosis , Receptors, Notch/metabolism , Wnt Signaling Pathway , Animals , Cell Lineage , Cell Proliferation , Labyrinth Supporting Cells/metabolism , Mice , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/antagonists & inhibitors , SOXB1 Transcription Factors/metabolism
11.
Neural Plast ; 2018: 7513258, 2018.
Article in English | MEDLINE | ID: mdl-30410537

ABSTRACT

Inner ear formation requires that a series of cell fate decisions and morphogenetic events occur in a precise temporal and spatial pattern. Previous studies have shown that transcription factors, including Pax2, Sox2, and Prox1, play important roles during the inner ear development. However, the temporospatial expression patterns among these transcription factors are poorly understood. In the current study, we present a comprehensive description of the temporal and spatial expression profiles of Pax2, Sox2, and Prox1 during auditory and vestibular sensory organ development in mice. Using immunohistochemical analyses, we show that Sox2 and Pax2 are both expressed in the prosensory cells (the developing hair cells), but Sox2 is later restricted to only the supporting cells of the organ of Corti. In the vestibular sensory organ, however, the Pax2 expression is localized in hair cells at postnatal day 7, while Sox2 is still expressed in both the hair cells and supporting cells at that time. Prox1 was transiently expressed in the presumptive hair cells and developing supporting cells, and lower Prox1 expression was observed in the vestibular sensory organ compared to the organ of Corti. The different expression patterns of these transcription factors in the developing auditory and vestibular sensory organs suggest that they play different roles in the development of the sensory epithelia and might help to shape the respective sensory structures.


Subject(s)
Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Vestibular/metabolism , Homeodomain Proteins/biosynthesis , PAX2 Transcription Factor/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Animals , Animals, Newborn , Cell Differentiation/physiology , Cochlea/growth & development , Cochlea/metabolism , Ear, Inner , Female , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , PAX2 Transcription Factor/genetics , Pregnancy , SOXB1 Transcription Factors/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
12.
Hum Genet ; 136(11-12): 1463-1475, 2017 11.
Article in English | MEDLINE | ID: mdl-29094203

ABSTRACT

Human Waardenburg syndrome 2A (WS2A) is a dominant hearing loss (HL) syndrome caused by mutations in the microphthalmia-associated transcription factor (MITF) gene. In mouse models with MITF mutations, WS2A is transmitted in a recessive pattern, which limits the study of hearing loss (HL) pathology. In the current study, we performed ENU (ethylnitrosourea) mutagenesis that resulted in substituting a conserved lysine with a serine (p. L247S) in the DNA-binding domain of the MITF gene to generate a novel miniature pig model of WS2A. The heterozygous mutant pig (MITF +/L247S) exhibits a dominant form of profound HL and hypopigmentation in skin, hair, and iris, accompanied by degeneration of stria vascularis (SV), fused hair cells, and the absence of endocochlear potential, which indicate the pathology of human WS2A. Besides hypopigmentation and bilateral HL, the homozygous mutant pig (MITF L247S/L247S) and CRISPR/Cas9-mediated MITF bi-allelic knockout pigs both exhibited anophthalmia. Three WS2 patients carrying MITF mutations adjacent to the corresponding region were also identified. The pig models resemble the clinical symptom and molecular pathology of human WS2A patients perfectly, which will provide new clues for better understanding the etiology and development of novel treatment strategies for human HL.


Subject(s)
Disease Models, Animal , Ethylnitrosourea/toxicity , Hearing Loss/genetics , Microphthalmia-Associated Transcription Factor/genetics , Mutation , Waardenburg Syndrome/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Female , Hearing Loss/chemically induced , Hearing Loss/pathology , Humans , Male , Microphthalmia-Associated Transcription Factor/antagonists & inhibitors , Mutagenesis , Mutagens/toxicity , Sequence Homology , Swine , Swine, Miniature , Waardenburg Syndrome/chemically induced , Waardenburg Syndrome/pathology
13.
J Neurosci ; 35(16): 6366-80, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904789

ABSTRACT

Hair cells of the inner ear are essential for hearing and balance. As a consequence, pathogenic variants in genes specifically expressed in hair cells often cause hereditary deafness. Hair cells are few in number and not easily isolated from the adjacent supporting cells, so the biochemistry and molecular biology of hair cells can be difficult to study. To study gene expression in hair cells, we developed a protocol for hair cell isolation by FACS. With nearly pure hair cells and surrounding cells, from cochlea and utricle and from E16 to P7, we performed a comprehensive cell type-specific RNA-Seq study of gene expression during mouse inner ear development. Expression profiling revealed new hair cell genes with distinct expression patterns: some are specific for vestibular hair cells, others for cochlear hair cells, and some are expressed just before or after maturation of mechanosensitivity. We found that many of the known hereditary deafness genes are much more highly expressed in hair cells than surrounding cells, suggesting that genes preferentially expressed in hair cells are good candidates for unknown deafness genes.


Subject(s)
Gene Expression Regulation, Developmental , Hair Cells, Auditory, Inner/metabolism , Animals , Cell Separation , Flow Cytometry , Gene Expression Profiling , Hair Cells, Auditory, Inner/cytology , Mice , Mice, Transgenic , Saccule and Utricle/cytology , Saccule and Utricle/growth & development , Saccule and Utricle/metabolism
14.
Proc Natl Acad Sci U S A ; 110(6): 2228-33, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23345450

ABSTRACT

Age-related hearing loss and noise-induced hearing loss are major causes of human morbidity. Here we used genetics and functional studies to show that a shared cause of these disorders may be loss of function of the ATP-gated P2X(2) receptor (ligand-gated ion channel, purinergic receptor 2) that is expressed in sensory and supporting cells of the cochlea. Genomic analysis of dominantly inherited, progressive sensorineural hearing loss DFNA41 in a six-generation kindred revealed a rare heterozygous allele, P2RX2 c.178G > T (p.V60L), at chr12:133,196,029, which cosegregated with fully penetrant hearing loss in the index family, and also appeared in a second family with the same phenotype. The mutation was absent from more than 7,000 controls. P2RX2 p.V60L abolishes two hallmark features of P2X(2) receptors: ATP-evoked inward current response and ATP-stimulated macropore permeability, measured as loss of ATP-activated FM1-43 fluorescence labeling. Coexpression of mutant and WT P2X(2) receptor subunits significantly reduced ATP-activated membrane permeability. P2RX2-null mice developed severe progressive hearing loss, and their early exposure to continuous moderate noise led to high-frequency hearing loss as young adults. Similarly, among family members heterozygous for P2RX2 p.V60L, noise exposure exacerbated high-frequency hearing loss in young adulthood. Our results suggest that P2X(2) function is required for life-long normal hearing and for protection from exposure to noise.


Subject(s)
Hearing Loss, Noise-Induced/genetics , Hearing Loss, Sensorineural/genetics , Mutation, Missense , Receptors, Purinergic P2X2/genetics , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Disease Models, Animal , Evoked Potentials, Auditory , Female , Genes, Dominant , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Humans , Ion Channel Gating , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Pedigree , Penetrance , Receptors, Purinergic P2X2/deficiency , Receptors, Purinergic P2X2/physiology , Sequence Homology, Amino Acid , Young Adult
15.
Neural Plast ; 2016: 9409846, 2016.
Article in English | MEDLINE | ID: mdl-28116172

ABSTRACT

Mammalian inner ear harbors diverse cell types that are essential for hearing and balance. Adenovirus is one of the major vectors to deliver genes into the inner ear for functional studies and hair cell regeneration. To identify adenovirus vectors that target specific cell subtypes in the inner ear, we studied three adenovirus vectors, carrying a reporter gene encoding green fluorescent protein (GFP) from two vendors or with a genome editing gene Cre recombinase (Cre), by injection into postnatal days 0 (P0) and 4 (P4) mouse cochlea through scala media by cochleostomy in vivo. We found three adenovirus vectors transduced mouse inner ear cells with different specificities and expression levels, depending on the type of adenoviral vectors and the age of mice. The most frequently targeted region was the cochlear sensory epithelium, including auditory hair cells and supporting cells. Adenovirus with GFP transduced utricular supporting cells as well. This study shows that adenovirus vectors are capable of efficiently and specifically transducing different cell types in the mammalian inner ear and provides useful tools to study inner ear gene function and to evaluate gene therapy to treat hearing loss and vestibular dysfunction.


Subject(s)
Adenoviridae/genetics , Ear, Inner/cytology , Ear, Inner/physiology , Gene Targeting/methods , Genetic Vectors/genetics , Animals , Animals, Newborn , Cochlea/cytology , Cochlea/drug effects , Cochlea/physiology , Ear, Inner/drug effects , Genetic Vectors/administration & dosage , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/genetics , Mice
16.
J Am Chem Soc ; 137(44): 14084-93, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26465072

ABSTRACT

The inefficient delivery of proteins into mammalian cells remains a major barrier to realizing the therapeutic potential of many proteins. We and others have previously shown that superpositively charged proteins are efficiently endocytosed and can bring associated proteins and nucleic acids into cells. The vast majority of cargo delivered in this manner, however, remains in endosomes and does not reach the cytosol. In this study we designed and implemented a screen to discover peptides that enhance the endosomal escape of proteins fused to superpositively charged GFP (+36 GFP). From a screen of peptides previously reported to disrupt microbial membranes without known mammalian cell toxicity, we discovered a 13-residue peptide, aurein 1.2, that substantially increases cytosolic protein delivery by up to ∼5-fold in a cytosolic fractionation assay in cultured cells. Four additional independent assays for nonendosomal protein delivery collectively suggest that aurein 1.2 enhances endosomal escape of associated endocytosed protein cargo. Structure-function studies clarified peptide sequence and protein conjugation requirements for endosomal escape activity. When applied to the in vivo delivery of +36 GFP-Cre recombinase fusions into the inner ear of live mice, fusion with aurein 1.2 dramatically increased nonendosomal Cre recombinase delivery potency, resulting in up to 100% recombined inner hair cells and 96% recombined outer hair cells, compared to 0-4% recombined hair cells from +36-GFP-Cre without aurein 1.2. Collectively, these findings describe a genetically encodable, endosome escape-enhancing peptide that can substantially increase the cytoplasmic delivery of cationic proteins in vitro and in vivo.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Drug Delivery Systems , Ear, Inner/cytology , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , Integrases/metabolism , Animals , Antimicrobial Cationic Peptides/chemistry , Cells, Cultured , Cytoplasm/metabolism , Ear, Inner/metabolism , Endosomes/chemistry , Mice
17.
Am J Hum Genet ; 91(5): 872-82, 2012 11 02.
Article in English | MEDLINE | ID: mdl-23122586

ABSTRACT

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Here we present OTOGL mutations, a homozygous one base pair deletion (c.1430 delT) causing a frameshift (p.Val477Glufs(∗)25) in a large consanguineous family and two compound heterozygous mutations, c.547C>T (p.Arg183(∗)) and c.5238+5G>A, in a nonconsanguineous family with moderate nonsyndromic sensorineural hearing loss. OTOGL maps to the DFNB84 locus at 12q21.31 and encodes otogelin-like, which has structural similarities to the epithelial-secreted mucin protein family. We demonstrate that Otogl is expressed in the inner ear of vertebrates with a transcription level that is high in embryonic, lower in neonatal, and much lower in adult stages. Otogelin-like is localized to the acellular membranes of the cochlea and the vestibular system and to a variety of inner ear cells located underneath these membranes. Knocking down of otogl with morpholinos in zebrafish leads to sensorineural hearing loss and anatomical changes in the inner ear, supporting that otogelin-like is essential for normal inner ear function. We propose that OTOGL mutations affect the production and/or function of acellular structures of the inner ear, which ultimately leads to sensorineural hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Mutation , Adolescent , Animals , Child, Preschool , Chromosome Aberrations , Cochlea/metabolism , Cochlea/pathology , Exome , Gene Expression Profiling , Gene Knockdown Techniques , Hearing Loss, Sensorineural/diagnosis , Humans , INDEL Mutation , Male , Mice , Polymorphism, Single Nucleotide , Rats , Zebrafish
18.
J Neurosci ; 33(38): 15086-94, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048839

ABSTRACT

Isl1 is a LIM-homeodomain transcription factor that is critical in the development and differentiation of multiple tissues. In the mouse inner ear, Isl1 is expressed in the prosensory region of otocyst, in young hair cells and supporting cells, and is no longer expressed in postnatal auditory hair cells. To evaluate how continuous Isl1 expression in postnatal hair cells affects hair cell development and cochlear function, we created a transgenic mouse model in which the Pou4f3 promoter drives Isl1 overexpression specifically in hair cells. Isl1 overexpressing hair cells develop normally, as seen by morphology and cochlear functions (auditory brainstem response and otoacoustic emissions). As the mice aged to 17 months, wild-type (WT) controls showed the progressive threshold elevation and outer hair cell loss characteristic of the age-related hearing loss (ARHL) in the background strain (C57BL/6J). In contrast, the Isl1 transgenic mice showed significantly less threshold elevation with survival of hair cells. Further, the Isl1 overexpression protected the ear from noise-induced hearing loss (NIHL): both ABR threshold shifts and hair cell death were significantly reduced when compared with WT littermates. Our model suggests a common mechanism underlying ARHL and NIHL, and provides evidence that hair cell-specific Isl1 expression can promote hair cell survival and therefore minimize the hearing impairment that normally occurs with aging and/or acoustic overexposure.


Subject(s)
Aging , Gene Expression Regulation/physiology , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/pathology , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Acoustic Stimulation , Analysis of Variance , Animals , Cochlea/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hearing Loss, Noise-Induced/metabolism , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otoacoustic Emissions, Spontaneous , RNA, Messenger/metabolism , Rats , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics
19.
Am J Hum Genet ; 89(1): 56-66, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21722859

ABSTRACT

SMAC/DIABLO is a mitochondrial proapoptotic protein that is released from mitochondria during apoptosis and counters the inhibitory activities of inhibitor of apoptosis proteins, IAPs. By linkage analysis and candidate screening, we identified a heterozygous SMAC/DIABLO mutation, c.377C>T (p.Ser126Leu, refers to p.Ser71Leu in the mature protein) in a six-generation Chinese kindred characterized by dominant progressive nonsyndromic hearing loss, designated as DFNA64. SMAC/DIABLO is highly expressed in human embryonic ears and is enriched in the developing mouse inner-ear hair cells, suggesting it has a role in the development and homeostasis of hair cells. We used a functional study to demonstrate that the SMAC/DIABLO(S71L) mutant, while retaining the proapoptotic function, triggers significant degradation of both wild-type and mutant SMAC/DIABLO and renders host mitochondria susceptible to calcium-induced loss of the membrane potential. Our work identifies DFNA64 as the human genetic disorder associated with SMAC/DIABLO malfunction and suggests that mutant SMAC/DIABLO(S71L) might cause mitochondrial dysfunction.


Subject(s)
Apoptosis/genetics , Hearing Loss/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mitochondrial Proteins/genetics , Mutation, Missense , Adolescent , Adult , Age of Onset , Aged , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Asian People , DNA Mutational Analysis , Down-Regulation , Female , Gene Expression Regulation, Developmental , Genetic Linkage , HeLa Cells , Hearing Loss/pathology , Humans , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Potentials/genetics , Mice , Middle Aged , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Pedigree , Polymorphism, Single Nucleotide , RNA, Small Interfering/metabolism , Up-Regulation , Young Adult
20.
Sci Transl Med ; 16(755): eadn0689, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985856

ABSTRACT

Mutations in microRNA-96 (MIR96) cause autosomal dominant deafness-50 (DFNA50), a form of delayed-onset hearing loss. Genome editing has shown efficacy in hearing recovery through intervention in neonatal mice, yet editing in the adult inner ear is necessary for clinical applications, which has not been done. Here, we developed a genome editing therapy for the MIR96 mutation 14C>A by screening different CRISPR systems and optimizing Cas9 expression and the sgRNA scaffold for efficient and specific mutation editing. AAV delivery of the KKH variant of Staphylococcus aureus Cas9 (SaCas9-KKH) and sgRNA to the cochleae of presymptomatic (3-week-old) and symptomatic (6-week-old) adult Mir9614C>A/+ mutant mice improved hearing long term, with efficacy increased by injection at a younger age. Adult inner ear delivery resulted in transient Cas9 expression without evidence of AAV genomic integration, indicating the good safety profile of our in vivo genome editing strategy. We developed a dual-AAV system, including an AAV-sgmiR96-master carrying sgRNAs against all known human MIR96 mutations. Because mouse and human MIR96 sequences share 100% homology, our approach and sgRNA selection for efficient and specific hair cell editing for long-term hearing recovery lay the foundation for the development of treatment for patients with DFNA50 caused by MIR96 mutations.


Subject(s)
Dependovirus , Gene Editing , Hearing Loss , MicroRNAs , Mutation , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Editing/methods , Humans , Mutation/genetics , Hearing Loss/genetics , Hearing Loss/therapy , Dependovirus/genetics , Mice , CRISPR-Cas Systems/genetics , Cochlea/metabolism , Genetic Therapy/methods , RNA, Guide, CRISPR-Cas Systems/genetics , Base Sequence , Hearing
SELECTION OF CITATIONS
SEARCH DETAIL