Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Lancet ; 403(10441): 2317-2325, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38280389

RESUMEN

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.


Asunto(s)
Dependovirus , Terapia Genética , Humanos , Terapia Genética/métodos , Dependovirus/genética , Niño , Masculino , Preescolar , Femenino , Adolescente , Lactante , Vectores Genéticos , Resultado del Tratamiento , Sordera/genética , Sordera/terapia , Mutación , Proteínas de la Membrana
2.
Mol Ther ; 31(4): 934-950, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36755494

RESUMEN

Gene therapy focuses on genetic modification to produce therapeutic effects or treat diseases by repairing or reconstructing genetic material, thus being expected to be the most promising therapeutic strategy for genetic disorders. Due to the growing attention to hearing impairment, an increasing amount of research is attempting to utilize gene therapy for hereditary hearing loss (HHL), an important monogenic disease and the most common type of congenital deafness. Several gene therapy clinical trials for HHL have recently been approved, and, additionally, CRISPR-Cas tools have been attempted for HHL treatment. Therefore, in order to further advance the development of inner ear gene therapy and promote its broad application in other forms of genetic disease, it is imperative to review the progress of gene therapy for HHL. Herein, we address three main gene therapy strategies (gene replacement, gene suppression, and gene editing), summarizing the strategy that is most appropriate for particular monogenic diseases based on different pathogenic mechanisms, and then focusing on their successful applications for HHL in preclinical trials. Finally, we elaborate on the challenges and outlooks of gene therapy for HHL.


Asunto(s)
Sordera , Pérdida Auditiva , Humanos , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Terapia Genética , Edición Génica
3.
Cell Mol Life Sci ; 80(6): 148, 2023 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-37178259

RESUMEN

Mutations in GJB2 (Gap junction protein beta 2) are the most common genetic cause of non-syndromic hereditary deafness in humans, especially the 35delG and 235delC mutations. Owing to the homozygous lethality of Gjb2 mutations in mice, there are currently no perfect mouse models carrying Gjb2 mutations derived from patients for mimicking human hereditary deafness and for unveiling the pathogenesis of the disease. Here, we successfully constructed heterozygous Gjb2+/35delG and Gjb2+/235delC mutant mice through advanced androgenic haploid embryonic stem cell (AG-haESC)-mediated semi-cloning technology, and these mice showed normal hearing at postnatal day (P) 28. A homozygous mutant mouse model, Gjb235delG/35delG, was then generated using enhanced tetraploid embryo complementation, demonstrating that GJB2 plays an indispensable role in mouse placenta development. These mice exhibited profound hearing loss similar to human patients at P14, i.e., soon after the onset of hearing. Mechanistic analyses showed that Gjb2 35delG disrupts the function and formation of intercellular gap junction channels of the cochlea rather than affecting the survival and function of hair cells. Collectively, our study provides ideal mouse models for understanding the pathogenic mechanism of DFNB1A-related hereditary deafness and opens up a new avenue for investigating the treatment of this disease.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Humanos , Ratones , Animales , Conexinas/genética , Conexina 26/genética , Sordera/genética , Pérdida Auditiva Sensorineural/genética , Mutación , Audición
4.
Hum Genet ; 142(2): 289-304, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36383253

RESUMEN

Mutations to the OTOF gene are among the most common reasons for auditory neuropathy. Although cochlear implants are often effective in restoring sound transduction, there are currently no biological treatments for individuals with variants of OTOF. Previous studies have reported the rescue of hearing in DFNB9 mice using OTOF gene replacement although the efficacy needs improvement. Here, we developed a novel dual-AAV-mediated gene therapy system based on the principles of protein trans-splicing, and we show that this system can reverse bilateral deafness in Otof -/- mice after a single unilateral injection. The system effectively expressed exogenous mouse or human otoferlin after injection on postnatal day 0-2. Human otoferlin restored hearing to near wild-type levels for at least 6 months and restored the release of synaptic vesicles in inner hair cells. Our study not only provides a preferential clinical strategy for the treatment of OTOF-related auditory neuropathies, but also describes a route of development for other large-gene therapies and protein engineering techniques.


Asunto(s)
Pérdida Auditiva Central , Pérdida Auditiva Sensorineural , Humanos , Animales , Ratones , Trans-Empalme , Audición , Pérdida Auditiva Sensorineural/genética , Mutación , Proteínas de la Membrana/genética
5.
PLoS Biol ; 18(3): e3000686, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32226015

RESUMEN

Compact CRISPR/Cas9 systems that can be packaged into an adeno-associated virus (AAV) hold great promise for gene therapy. Unfortunately, currently available small Cas9 nucleases either display low activity or require a long protospacer adjacent motif (PAM) sequence, limiting their extensive applications. Here, we screened a panel of Cas9 nucleases and identified a small Cas9 ortholog from Staphylococcus auricularis (SauriCas9), which recognizes a simple NNGG PAM, displays high activity for genome editing, and is compact enough to be packaged into an AAV for genome editing. Moreover, the conversion of adenine and cytosine bases can be achieved by fusing SauriCas9 to the cytidine and adenine deaminase. Therefore, SauriCas9 holds great potential for both basic research and clinical applications.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , ADN/metabolismo , Edición Génica/métodos , Staphylococcus/enzimología , Secuencia de Aminoácidos , Proteína 9 Asociada a CRISPR/química , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , ADN/química , ADN/genética , Dependovirus/genética , Células HEK293 , Humanos , Motivos de Nucleótidos , Ingeniería de Proteínas , Staphylococcus/genética , Especificidad por Sustrato
6.
Mol Ther ; 30(1): 105-118, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34174443

RESUMEN

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.


Asunto(s)
Edición Génica , Pérdida Auditiva , Animales , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Audición , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Humanos , Ratones , ARN Guía de Kinetoplastida
7.
Clin Genet ; 102(2): 149-154, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35599357

RESUMEN

The pathogenic variants in KCNQ4 cause DFNA2 nonsyndromic hearing loss. However, the understanding of genotype-phenotype correlations between KCNQ4 and hearing is limited. Here, we identified a novel KCNQ4 mutation p.G228D from a Chinese family, including heterozygotes characterized by high-frequency hearing loss that is progressive across all frequencies and homozygotes with more severe hearing loss. We constructed a novel murine model with humanized homologous Kcnq4 mutation. The heterozygotes had mid-frequency and high-frequency hearing loss at 4 weeks, and moved toward all frequencies hearing loss at 12 weeks, while the homozygotes had severe-to-profound hearing loss at 8 weeks. The degeneration of outer hair cells (OHCs) was observed from basal to apical turn of cochlea. The reduced K+ currents and depolarized resting potentials were revealed in OHCs. Remarkably, we observed the loss of inner hair cells (IHCs) in the region corresponding to the frequency above 32 kHz at 8-12 weeks. The results suggest the degeneration of OHCs and IHCs may contribute to high-frequency hearing loss in DFNA2 over time. Our findings broaden the variants of KCNQ4 and provide a novel mouse model of progressive hearing loss, which contributes to an understanding of pathogenic mechanism and eventually treatment of DFNA2 progressive hearing loss.


Asunto(s)
Pérdida Auditiva de Alta Frecuencia , Canales de Potasio KCNQ , Animales , China , Modelos Animales de Enfermedad , Pérdida Auditiva de Alta Frecuencia/genética , Humanos , Canales de Potasio KCNQ/genética , Ratones , Mutación
8.
Anal Chem ; 91(19): 12156-12161, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31460749

RESUMEN

A rapid and sensitive method is crucial for nucleic acid detection. Recently, RNA-guided CRISPR/Cas12a nuclease-based methods present great promise for nucleic acid detection. In the present methods, however, DNA amplification and subsequent Cas12a cleavage is separated and the whole process takes as long as 2 h. Most importantly, the uncapping operation increases the risk of aerosol contamination. In this study, we propose a CRISPR/Cas12a-based method named "Cas12aVDet" for rapid nucleic acid detection. By integrating recombinase polymerase amplification (RPA) with Cas12a cleavage in a single reaction system, the detection can be accomplished in 30 min and uncapping contamination can be avoided. The detection signal can be observed by the naked eye under blue light. This method could detect DNA at single molecule level and demonstrated 100% accuracy for mycoplasma contamination detection, presenting great potential for a variety of nucleic acid detection applications.


Asunto(s)
Sistemas CRISPR-Cas , ADN/análisis , Técnicas de Amplificación de Ácido Nucleico/métodos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Tampones (Química) , Proteínas Asociadas a CRISPR/química , Proteínas Asociadas a CRISPR/genética , Electroforesis en Gel de Agar , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/genética , Mycoplasma/genética
9.
Inorg Chem ; 58(23): 16241-16249, 2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31710224

RESUMEN

In the design and construction of porous materials, these with exceptional structure and composition are often highly expected, as they may offer unique nanopore space for desired applications. Here, a new family of quasi-3D and a 3D porous coordination polymers (PCPs) (termed NTU-43 to NTU-50) were constructed via an evolution strategy from a layered structure (termed NTU-42). Single gas adsorption isotherms of CO2, N2, and CH4 display the dependency of gas capacity on optimized effects of pore size, functionality, and charged framework of these quasi-3D PCPs, where NTU-45 and NTU-46, the two with NH2-BDC and OH-BDC bidentate linkers (NH2-BDC = 2-aminoterephthalic acid and OH-BDC = 2-hydroxyterephthalic acid) have demonstrated outstanding ability for selective CO2 uptake. To the best of our knowledge, this is the first time to well explore the synergistic effects toward gas adsorption on a platform of quasi-3D frameworks. More importantly, the efficient CO2 capture from CO2/CH4 and CO2/N2 mixtures has been also validated by breakthrough experiments under continuous and dynamic conditions at 298 K.

10.
Inorg Chem ; 58(16): 11066-11075, 2019 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-31369245

RESUMEN

Multiphoton excitation allows one to access high energy excited states and perform valuable tasks in biological systems using tissue penetrating near-infrared (NIR) light. Here, we describe new photoactive manganese tricarbonyl complexes incorporating the ligand 4'-p-N,N-bis(2-hydroxyethyl)amino-benzyl-2,2':6',2″-terpyridine (TPYOH), which can serve as an antenna for two photon NIR excitation. Solutions of Mn(CO)3(TPYOH)X (X = Br- or CF3SO3-) complexes are very photoactive toward CO release under visible light excitation (405 nm, 451 nm). The same responses were also triggered by multiphoton excitation at 750 and 800 nm. In this context, we discuss the potential applications of these complexes as visible/NIR light photoactivated carbon monoxide releasing moieties (photoCORMs). We also report the isolation and crystal structures of the TPYOH complexes Mn(TPYOH)Cl2 and [Mn(TPYOH)2](CF3SO3)2, to illustrate a possible photolysis product(s).

11.
Inorg Chem ; 54(23): 11450-6, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26562721

RESUMEN

New trinuclear RuPt2 and heptanuclear RuPt6 complex salts are prepared by attaching Pt(II) 2,2':6',2"-terpyridine (tpy) moieties to Ru(II) 4,4':2',2":4",4"'-quaterpyridine (qpy) complexes. Characterization includes single crystal X-ray structures for both polymetallic species. The visible absorption bands are primarily due to Ru(II) → qpy metal-to-ligand charge-transfer (MLCT) transitions, according to time-dependent density functional theory (TD-DFT) calculations. These spectra change only slightly on Pt coordination, while the orange-red emission from the complexes shows corresponding small red-shifts, accompanied by decreases in intensity. Cubic molecular nonlinear optical behavior has been assessed by using Z-scan measurements. These reveal relatively high two-photon absorption (2PA) cross sections σ2, with maximal values of 301 GM at 834 nm (RuPt2) and 523 GM at 850 nm (RuPt6) when dissolved in methanol or acetone, respectively. Attaching Pt(II)(tpy) moieties triples or quadruples the 2PA activities when compared with the Ru(II)-based cores.

12.
Mol Ther Nucleic Acids ; 35(1): 102135, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38404504

RESUMEN

Adeno-associated viral (AAV) vectors are increasingly used as vehicles for gene delivery to treat hearing loss. However, lack of specificity of the transgene expression may lead to overexpression of the transgene in nontarget tissues. In this study, we evaluated the expression efficiency and specificity of transgene delivered by AAV-PHP.eB under the inner ear sensory cell-specific Myo15 promoter. Compared with the ubiquitous CAG promoter, the Myo15 promoter initiates efficient expression of the GFP fluorescence reporter in hair cells, while minimizing non-specific expression in other cell types of the inner ear and CNS. Furthermore, using the Myo15 promoter, we constructed an AAV-mediated therapeutic system with the coding sequence of OTOF gene. After inner ear injection, we observed apparent hearing recovery in Otof-/- mice, highly efficient expression of exogenous otoferlin, and significant improvement in the exocytosis function of inner hair cells. Overall, our results indicate that gene therapy mediated by the hair cell-specific Myo15 promoter has potential clinical application for the treatment of autosomal recessive deafness and yet for other hereditary hearing loss related to dysfunction of hair cells.

13.
Research (Wash D C) ; 7: 0341, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38665848

RESUMEN

Adeno-associated virus (AAV)-mediated gene therapy is widely applied to treat numerous hereditary diseases in animal models and humans. The specific expression of AAV-delivered transgenes driven by cell type-specific promoters should further increase the safety of gene therapy. However, current methods for screening cell type-specific promoters are labor-intensive and time-consuming. Herein, we designed a "multiple vectors in one AAV" strategy for promoter construction in vivo. Through this strategy, we truncated a native promoter for Myo15 expression in hair cells (HCs) in the inner ear, from 1,611 bp down to 1,157 bp, and further down to 956 bp. Under the control of these 2 promoters, green fluorescent protein packaged in AAV-PHP.eB was exclusively expressed in the HCs. The transcription initiation ability of the 2 promoters was further verified by intein-mediated otoferlin recombination in a dual-AAV therapeutic system. Driven by these 2 promoters, human otoferlin was selectively expressed in HCs, resulting in the restoration of hearing in treated Otof -/- mice for at least 52 weeks. In summary, we developed an efficient screening strategy for cell type-specific promoter engineering and created 2 truncated Myo15 promoters that not only restored hereditary deafness in animal models but also show great potential for treating human patients in future.

14.
Nat Med ; 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839897

RESUMEN

Gene therapy is a promising approach for hereditary deafness. We recently showed that unilateral AAV1-hOTOF gene therapy with dual adeno-associated virus (AAV) serotype 1 carrying human OTOF transgene is safe and associated with functional improvements in patients with autosomal recessive deafness 9 (DFNB9). The protocol was subsequently amended and approved to allow bilateral gene therapy administration. Here we report an interim analysis of the single-arm trial investigating the safety and efficacy of binaural therapy in five pediatric patients with DFNB9. The primary endpoint was dose-limiting toxicity at 6 weeks, and the secondary endpoint included safety (adverse events) and efficacy (auditory function and speech perception). No dose-limiting toxicity or serious adverse event occurred. A total of 36 adverse events occurred. The most common adverse events were increased lymphocyte counts (6 out of 36) and increased cholesterol levels (6 out of 36). All patients had bilateral hearing restoration. The average auditory brainstem response threshold in the right (left) ear was >95 dB (>95 dB) in all patients at baseline, and the average auditory brainstem response threshold in the right (left) ear was restored to 58 dB (58 dB) in patient 1, 75 dB (85 dB) in patient 2, 55 dB (50 dB) in patient 3 at 26 weeks, and 75 dB (78 dB) in patient 4 and 63 dB (63 dB) in patient 5 at 13 weeks. The speech perception and the capability of sound source localization were restored in all five patients. These results provide preliminary insights on the safety and efficacy of binaural AAV gene therapy for hereditary deafness. The trial is ongoing with longer follow-up to confirm the safety and efficacy findings. Chinese Clinical Trial Registry registration: ChiCTR2200063181 .

15.
Acta Crystallogr Sect E Struct Rep Online ; 69(Pt 6): m313, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23794980

RESUMEN

In the title compound, [Sn(CH3)3(C7H7N2O2S)(H2O)], the Sn(IV) atom has a distorted trigonal-bipyramidal coordination geometry, with one carboxyl-ate O atom of the 2-(4-methyl-pyrimidine-2-sulfan-yl)acetate ligand and the O atom of a water mol-ecule in axial positions, and three methyl groups in equatorial positions. In the crystal, mol-ecules are linked via O-H⋯O and O-H⋯N hydrogen bonds, forming double-stranded chains propagating along [010].

16.
Front Cell Dev Biol ; 11: 1305433, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38259515

RESUMEN

Aminoglycosides are commonly used for the treatment of life-threatening bacterial infections, however, aminoglycosides may cause irreversible hearing loss with a long-term clinical therapy. The mechanism and prevention of the ototoxicity of aminoglycosides are still limited although amounts of studies explored widely. Specifically, advancements in programmed cell death (PCD) provide more new perspectives. This review summarizes the general signal pathways in programmed cell death, including apoptosis, autophagy, and ferroptosis, as well as the mechanisms of aminoglycoside-induced ototoxicity. Additionally, novel interventions, especially gene therapy strategies, are also investigated for the prevention or treatment of aminoglycoside-induced hearing loss with prospective clinical applications.

17.
Mol Ther Methods Clin Dev ; 31: 101154, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38027066

RESUMEN

Pathogenic mutations in the OTOF gene cause autosomal recessive hearing loss (DFNB9), one of the most common forms of auditory neuropathy. There is no biological treatment for DFNB9. Here, we designed an OTOF gene therapy agent by dual-adeno-associated virus 1 (AAV1) carrying human OTOF coding sequences with the expression driven by the hair cell-specific promoter Myo15, AAV1-hOTOF. To develop a clinical application of AAV1-hOTOF gene therapy, we evaluated its efficacy and safety in animal models using pharmacodynamics, behavior, and histopathology. AAV1-hOTOF inner ear delivery significantly improved hearing in Otof-/- mice without affecting normal hearing in wild-type mice. AAV1 was predominately distributed to the cochlea, although it was detected in other organs such as the CNS and the liver, and no obvious toxic effects of AAV1-hOTOF were observed in mice. To further evaluate the safety of Myo15 promoter-driven AAV1-transgene, AAV1-GFP was delivered into the inner ear of Macaca fascicularis via the round window membrane. AAV1-GFP transduced 60%-94% of the inner hair cells along the cochlear turns. AAV1-GFP was detected in isolated organs and no significant adverse effects were detected. These results suggest that AAV1-hOTOF is well tolerated and effective in animals, providing critical support for its clinical translation.

18.
Biomolecules ; 13(1)2022 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-36671423

RESUMEN

Inner ear gene therapy using adeno-associated viral vectors (AAVs) in neonatal mice can alleviate hearing loss in mouse models of deafness. However, efficient and safe transgene delivery to the adult mouse cochlea is critical for the effectiveness of AAV-mediated therapy. Here, we examined three gene delivery approaches including posterior semicircular canal (PSCC) canalostomy, round window membrane (RWM) injection, and tubing-RWM+PSCC (t-RP) in adult mice. Transduction rates and survival rates of cochlear hair cells were analyzed, hearing function was recorded, AAV distribution in the sagittal brain sections was evaluated, and cochlear histopathologic images were appraised. We found that an injection volume of 1 µL AAV through the PSCC is safe and highly efficient and does not impair hearing function in adult mice, but local injection allows AAV vectors to spread slightly into the brain. We then tested five AAV serotypes (PHP.eB, IE, Anc80L65, AAV2, and PHP.s) in parallel and observed the most robust eGFP expression in inner hair cells, outer hair cells, and spiral ganglion neurons throughout the cochlea after AAV-Anc80L65 injection. Thus, PSCC-injected Anc80L65 provides a foundation for gene therapy in the adult cochlea and will facilitate the development of inner ear gene therapy.


Asunto(s)
Vectores Genéticos , Pérdida Auditiva , Animales , Ratones , Vectores Genéticos/genética , Cóclea/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Pérdida Auditiva/terapia
19.
Mol Ther Nucleic Acids ; 28: 280-292, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35433113

RESUMEN

A wide spectrum of SLC26A4 mutations causes Pendred syndrome and enlarged vestibular aqueduct, both associated with sensorineural hearing loss (SNHL). A splice-site mutation, c.919-2A>G (A-2G), which is common in Asian populations, impairs the 3' splice site of intron 7, resulting in exon 8 skipping during pre-mRNA splicing and a subsequent frameshift that creates a premature termination codon in the following exon. Currently, there is no effective drug treatment for SHNL. For A-2G-triggered SNHL, molecules that correct mis-splicing of the mutant hold promise to treat the disease. Antisense oligonucleotides (ASOs) can promote exon inclusion when targeting specific splicing silencers. Here, we systematically screened a large number of ASOs in a minigene system and identified a few that markedly repressed exon 8 skipping. A lead ASO, which targets a heterogeneous nuclear ribonucleoprotein (hnRNP) A1/A2 intronic splicing silencer (ISS) in intron 8, promoted efficient exon 8 inclusion in cultured peripheral blood mononuclear cells derived from two homozygous patients. In a partially humanized Slc26a4 A-2G mouse model, two subcutaneous injections of the ASO at 160 mg/kg significantly rescued exon 8 splicing in the liver. Our results demonstrate that the ISS-targeting ASO has therapeutic potential to treat genetic hearing loss caused by the A-2G mutation in SLC26A4.

20.
Signal Transduct Target Ther ; 7(1): 79, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35283480

RESUMEN

CRISPR/RfxCas13d (CasRx) editing system can specifically and precisely cleave single-strand RNAs, which is a promising treatment for various disorders by downregulation of related gene expression. Here, we tested this RNA-editing approach on Beethoven (Bth) mice, an animal model for human DFNA36 due to a point mutation in Tmc1. We first screened 30 sgRNAs in cell cultures and found that CasRx with sgRNA3 reduced the Tmc1Bth transcript by 90.8%, and the Tmc1 wild type transcript (Tmc1+) by 44.3%. We then injected a newly developed AAV vector (AAV-PHP.eB) based CasRx into the inner ears of neonatal Bth mice, and we found that Tmc1Bth was reduced by 70.2% in 2 weeks with few off-target effects in the whole transcriptome. Consistently, we found improved hair cell survival, rescued hair bundle degeneration, and reduced mechanoelectrical transduction current. Importantly, the hearing performance, measured in both ABR and DPOAE thresholds, was improved significantly in all ages over 8 weeks. We, therefore, have validated the CRISPR/CasRx-based RNA editing strategy in treating autosomal-dominant hearing loss, paving way for its further application in many other hereditary diseases in hearing and beyond.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Ratones , Edición de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA