1
|
Yu W, Hill SF, Huang Y, Zhu L, Demetriou Y, Ziobro J, Reger F, Jia X, Mattis J, Meisler MH. Allele-Specific Editing of a Dominant SCN8A Epilepsy Variant Protects against Seizures and Lethality in a Murine Model. Ann Neurol 2024; 96:958-969. [PMID: 39158034 PMCID: PMC11496010 DOI: 10.1002/ana.27053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 07/26/2024] [Indexed: 08/20/2024]
Abstract
OBJECTIVE Developmental and epileptic encephalopathies (DEEs) can result from dominant, gain of function variants of neuronal ion channels. More than 450 de novo missense variants of the sodium channel gene SCN8A have been identified in individuals with DEE. METHODS We studied a mouse model carrying the patient Scn8a variant p.Asn1768Asp. An AAV-PHP.eB virus carrying an allele-specific single guide RNA (sgRNA) was administered by intracerebroventricular injection. Cas9 was provided by an inherited transgene. RESULTS Allele-specific disruption of the reading frame of the pathogenic transcript generated out-of-frame indels in 1/4 to 1/3 of transcripts throughout the brain. This editing efficiency was sufficient to rescue lethality and seizures. Neuronal hyperexcitability was reduced in cells expressing the virus. INTERPRETATION The data demonstrate efficient allele-specific editing of a dominant missense variant and support the feasibility of allele-specific therapy for DEE epilepsy. ANN NEUROL 2024;96:958-969.
Collapse
Affiliation(s)
- Wenxi Yu
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Sophie F Hill
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
| | - Yumei Huang
- Center for Genomic Technologies, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Limei Zhu
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | | | - Julie Ziobro
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Faith Reger
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Xiaoyan Jia
- Center for Genomic Technologies, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Joanna Mattis
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
| |
Collapse
|
2
|
Jc L, R M, T M. Gene therapy for hearing loss: Current status and future prospects of non-viral vector delivery systems. Hear Res 2024; 453:109130. [PMID: 39427589 DOI: 10.1016/j.heares.2024.109130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Current therapeutic options for hearing loss rely on hearing aids, ossiculoplasty or cochlear implants. These devices have limitations, particularly in noisy acoustic environments. Therefore, interest in exploring aetiological treatments to improve not only auditory perception but also the quality of life of those affected is increasing. Gene therapy is a promising aetiological treatment that can fully restore auditory function. The success of gene therapy relies on the efficient delivery of therapeutic genes or genetic modifications to the cells of the inner ear that are designed to repair or replace defective genes and restore normal hearing function. Two main strategies for gene therapy involve the use of recombinant viral vectors and nonviral delivery vehicles. Owing to their excellent diffusion properties and compatibility with sensory cells, recombinant viral vectors, particularly adeno-associated viruses (AAVs), have dominated gene therapy in the cochlea. However, recombinant viral vectors have several drawbacks, such as limited transgene size, immunogenicity (particularly in neonates), and potential need for repeat administration. Nonviral vectors, such as cationic lipids and polymeric nanoparticles, are potential attractive alternatives. Nonviral vectors have several advantages, including low immunogenicity and unlimited transgene size. Recent studies have demonstrated significant auditory recovery in vivo using nonviral vectors in murine models. However, nonviral vectors are not as efficient as viral vectors in transferring genetic material. An alternative to nanoparticles is the use of other methods, such as electroporation. The main advantage of electroporation is that it can be used in combination with cochlear implantation and can target surface cells, but this method has a risk of cell damage. The goal of this review is to provide valuable insights into the current state of research on nonviral vectors for inner ear gene therapy and propose the exploration of innovative and effective gene therapy strategies for the treatment of hearing loss.
Collapse
Affiliation(s)
- Leclère Jc
- CHU de Brest, Service d'ORL et chirurgie cervico-faciale, 29200 Brest, France; Univ Brest, LIEN, 29200 Brest, France.
| | - Marianowski R
- CHU de Brest, Service d'ORL et chirurgie cervico-faciale, 29200 Brest, France; Univ Brest, LIEN, 29200 Brest, France
| | - Montier T
- Univ Brest, INSERM, EFS, UMR 1078 GGB-GTCA team, 29200 Brest, France; CHU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Centre de Référence des Maladies Rares "Maladies Neuromusculaires", 29200 Brest, France.
| |
Collapse
|
3
|
Carufe KEW, Economos NG, Glazer PM. Peptide Nucleic Acid-Mediated Regulation of CRISPR-Cas9 Specificity. Nucleic Acid Ther 2024; 34:245-256. [PMID: 39037032 DOI: 10.1089/nat.2024.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Although CRISPR-Cas9 gene therapies have proven to be a powerful tool across many applications, improvements are necessary to increase the specificity of this technology. Cas9 cutting in off-target sites remains an issue that limits CRISPR's application in human-based therapies. Treatment of autosomal dominant diseases also remains a challenge when mutant alleles differ from the wild-type sequence by only one base pair. Here, we utilize synthetic peptide nucleic acids (PNAs) that bind selected spacer sequences in the guide RNA (gRNA) to increase Cas9 specificity up to 10-fold. We interrogate variations in PNA length, binding position, and degree of homology with the gRNA. Our findings reveal that PNAs bound in the region distal to the protospacer adjacent motif (PAM) site effectively enhance specificity in both on-target/off-target and allele-specific scenarios. In addition, we demonstrate that introducing deliberate mismatches between PNAs bound in the PAM-proximal region of the gRNA can modulate Cas9 activity in an allele-specific manner. These advancements hold promise for addressing current limitations and expanding the therapeutic potential of CRISPR technology.
Collapse
Affiliation(s)
- Kelly E W Carufe
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nicholas G Economos
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
4
|
Wang L, Zhang R, Jiang L, Gao S, Wu J, Jiao Y. Biomaterials as a new option for treating sensorineural hearing loss. Biomater Sci 2024; 12:4006-4023. [PMID: 38979939 DOI: 10.1039/d4bm00518j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensorineural hearing loss (SNHL) usually involves damage to complex auditory pathways such as inner ear cells and auditory nerves. The highly intricate and nuanced characteristics of these cells render their repair and regeneration extremely challenging, making it difficult to restore hearing to normal levels once it has been compromised. The effectiveness of traditional drugs is so minimal that they provide little help with the treatment. Fortunately, extensive experiments have demonstrated that combining biomaterials with conventional techniques significantly enhances drug effectiveness. This article reviews the research progress of biomaterials in protecting hair cells and the auditory nerve, repairing genes related to hearing, and developing artificial cochlear materials. By organizing the knowledge presented in this article, perhaps new insights can be provided for the clinical management of SNHL.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| | - Ruhe Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Linlan Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| | - Shuyi Gao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| | - Jun Wu
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China.
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuenong Jiao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou 510620, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, Guangzhou Medical University, Guangzhou 510620, China.
| |
Collapse
|
5
|
Masarwy R, Stotsky-Oterin L, Elisha A, Hazan-Halevy I, Peer D. Delivery of nucleic acid based genome editing platforms via lipid nanoparticles: Clinical applications. Adv Drug Deliv Rev 2024; 211:115359. [PMID: 38857763 DOI: 10.1016/j.addr.2024.115359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/17/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
CRISPR/Cas technology presents a promising approach for treating a wide range of diseases, including cancer and genetic disorders. Despite its potential, the translation of CRISPR/Cas into effective in-vivo gene therapy encounters challenges, primarily due to the need for safe and efficient delivery mechanisms. Lipid nanoparticles (LNPs), FDA-approved for RNA delivery, show potential for delivering also CRISPR/Cas, offering the capability to efficiently encapsulate large mRNA molecules with single guide RNAs. However, achieving precise targeting in-vivo remains a significant obstacle, necessitating further research into optimizing LNP formulations. Strategies to enhance specificity, such as modifying LNP structures and incorporating targeting ligands, are explored to improve organ and cell type targeting. Furthermore, the development of base and prime editing technology presents a potential breakthrough, offering precise modifications without generating double-strand breaks (DSBs). Prime editing, particularly when delivered via targeted LNPs, holds promise for treating diverse diseases safely and precisely. This review assesses both the progress made and the persistent challenges faced in using LNP-encapsulated CRISPR-based technologies for therapeutic purposes, with a particular focus on clinical translation.
Collapse
Affiliation(s)
- Razan Masarwy
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
| | - Dan Peer
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
6
|
Zhu W, Du W, Rameshbabu AP, Armstrong AM, Silver S, Kim Y, Wei W, Shu Y, Liu X, Lewis MA, Steel KP, Chen ZY. Targeted genome editing restores auditory function in adult mice with progressive hearing loss caused by a human microRNA mutation. Sci Transl Med 2024; 16:eadn0689. [PMID: 38985856 PMCID: PMC7616320 DOI: 10.1126/scitranslmed.adn0689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
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.
Collapse
Affiliation(s)
- Wenliang Zhu
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Wan Du
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Arun Prabhu Rameshbabu
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Ariel Miura Armstrong
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Stewart Silver
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Yehree Kim
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Wei Wei
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai200031, China
| | - Xuezhong Liu
- Department of Otolaryngology, University of Miami School of Medicine, Miami, FL 33136, USA
| | - Morag A. Lewis
- Wolfson Sensory, Pain and Regeneration Centre, King’s College London, LondonWC2R 2LS, UK
| | - Karen P. Steel
- Wolfson Sensory, Pain and Regeneration Centre, King’s College London, LondonWC2R 2LS, UK
| | - Zheng-Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody laboratory, Massachusetts Eye and Ear, Boston, MA02114, USA
| |
Collapse
|
7
|
Shan H, Yu N, Chen M, Sun Q, Sun X, Du C, Shang W, Li Z, Wei X, Lin Q, Jiang Z, Chen Z, Zhu B, Zhao S, Chen Z, Chen X. Cavitation-on-a-Chip Enabled Size-Specific Liposomal Drugs for Selective Pharmacokinetics and Pharmacodynamics. NANO LETTERS 2024; 24:8151-8161. [PMID: 38912914 DOI: 10.1021/acs.nanolett.4c02114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
The size of liposomal drugs has been demonstrated to strongly correlate with their pharmacokinetics and pharmacodynamics. While the microfluidic method successfully achieves the production of liposomes with well-controlled sizes across various buffer/lipid flow rate ratio (FRR) settings, any adjustments to the FRR inevitably influence the concentration, encapsulation efficiency (EE), and stability of liposomal drugs. Here we describe a controllable cavitation-on-a-chip (CCC) strategy that facilitates the precise regulation of liposomal drug size at any desired FRR. The CCC-enabled size-specific liposomes exhibited striking differences in uptake and biodistribution behaviors, thereby demonstrating distinct antitumor efficacy in both tumor-bearing animal and melanoma patient-derived organoid (PDO) models. Intriguingly, as the liposome size decreased to approximately 80 nm, the preferential accumulation of liposomal drugs in the liver transitioned to a predominant enrichment in the kidneys. These findings underscore the considerable potential of our CCC approach in influencing the pharmacokinetics and pharmacodynamics of liposomal nanomedicines.
Collapse
Affiliation(s)
- Han Shan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Nianzhou Yu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Maike Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Qi Sun
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xin Sun
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Changsheng Du
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wansong Shang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhaoxi Li
- The School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Xiongwei Wei
- The School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Qibo Lin
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zixi Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Ziyan Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Benpeng Zhu
- School of Integrated Circuits, Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Zeyu Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| |
Collapse
|
8
|
Yi H, Yun Y, Choi WH, Hwang HY, Cha JH, Seok H, Song JJ, Lee JH, Lee SY, Kim D. CRISPR-based editing strategies to rectify EYA1 complex genomic rearrangement linked to haploinsufficiency. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102199. [PMID: 38766525 PMCID: PMC11101721 DOI: 10.1016/j.omtn.2024.102199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/21/2024] [Indexed: 05/22/2024]
Abstract
Pathogenic structure variations (SVs) are associated with various types of cancer and rare genetic diseases. Recent studies have used Cas9 nuclease with paired guide RNAs (gRNAs) to generate targeted chromosomal rearrangements, focusing on producing fusion proteins that cause cancer, whereas research on precision genome editing for rectifying SVs is limited. In this study, we identified a novel complex genomic rearrangement (CGR), specifically an EYA1 inversion with a deletion, implicated in branchio-oto-renal/branchio-oto syndrome. To address this, two CRISPR-based approaches were tested. First, we used Cas9 nuclease and paired gRNAs tailored to the patient's genome. The dual CRISPR-Cas9 system induced efficient correction of paracentric inversion in patient-derived fibroblast, and effectively restored the expression of EYA1 mRNA and protein, along with its transcriptional activity required to regulate the target gene expression. Additionally, we used CRISPR activation (CRISPRa), which leads to the upregulation of EYA1 mRNA expression in patient-derived fibroblasts. Moreover, CRISPRa significantly improved EYA1 protein expression and transcriptional activity essential for target gene expression. This suggests that CRISPRa-based gene therapies could offer substantial translational potential for approximately 70% of disease-causing EYA1 variants responsible for haploinsufficiency. Our findings demonstrate the potential of CRISPR-guided genome editing for correcting SVs, including those with EYA1 CGR linked to haploinsufficiency.
Collapse
Affiliation(s)
- Hwalin Yi
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Yejin Yun
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, South Korea
| | - Won Hoon Choi
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, South Korea
| | - Hye-Yeon Hwang
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Ju Hyuen Cha
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, South Korea
| | - Heeyoung Seok
- Department of Transdisciplinary Research and Collaboration, Genomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea
| | - Jae-Jin Song
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, South Korea
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, South Korea
- Department of Genomic Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Daesik Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| |
Collapse
|
9
|
Chen X, Zhong S, Zhan Y, Zhang X. CRISPR-Cas9 applications in T cells and adoptive T cell therapies. Cell Mol Biol Lett 2024; 29:52. [PMID: 38609863 PMCID: PMC11010303 DOI: 10.1186/s11658-024-00561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
T cell immunity is central to contemporary cancer and autoimmune therapies, encompassing immune checkpoint blockade and adoptive T cell therapies. Their diverse characteristics can be reprogrammed by different immune challenges dependent on antigen stimulation levels, metabolic conditions, and the degree of inflammation. T cell-based therapeutic strategies are gaining widespread adoption in oncology and treating inflammatory conditions. Emerging researches reveal that clustered regularly interspaced palindromic repeats-associated protein 9 (CRISPR-Cas9) genome editing has enabled T cells to be more adaptable to specific microenvironments, opening the door to advanced T cell therapies in preclinical and clinical trials. CRISPR-Cas9 can edit both primary T cells and engineered T cells, including CAR-T and TCR-T, in vivo and in vitro to regulate T cell differentiation and activation states. This review first provides a comprehensive summary of the role of CRISPR-Cas9 in T cells and its applications in preclinical and clinical studies for T cell-based therapies. We also explore the application of CRISPR screen high-throughput technology in editing T cells and anticipate the current limitations of CRISPR-Cas9, including off-target effects and delivery challenges, and envisioned improvements in related technologies for disease screening, diagnosis, and treatment.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Cardiology, Cardiovascular Institute of Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Shuhan Zhong
- Department of Hematology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, 310003, China
| | - Yonghao Zhan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| | - Xuepei Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
10
|
Pan X, Li Y, Huang P, Staecker H, He M. Extracellular vesicles for developing targeted hearing loss therapy. J Control Release 2024; 366:460-478. [PMID: 38182057 DOI: 10.1016/j.jconrel.2023.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
Substantial efforts have been made for local administration of small molecules or biologics in treating hearing loss diseases caused by either trauma, genetic mutations, or drug ototoxicity. Recently, extracellular vesicles (EVs) naturally secreted from cells have drawn increasing attention on attenuating hearing impairment from both preclinical studies and clinical studies. Highly emerging field utilizing diverse bioengineering technologies for developing EVs as the bioderived therapeutic materials, along with artificial intelligence (AI)-based targeting toolkits, shed the light on the unique properties of EVs specific to inner ear delivery. This review will illuminate such exciting research field from fundamentals of hearing protective functions of EVs to biotechnology advancement and potential clinical translation of functionalized EVs. Specifically, the advancements in assessing targeting ligands using AI algorithms are systematically discussed. The overall translational potential of EVs is reviewed in the context of auditory sensing system for developing next generation gene therapy.
Collapse
Affiliation(s)
- Xiaoshu Pan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Yanjun Li
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| | - Peixin Huang
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States
| | - Hinrich Staecker
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States.
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States.
| |
Collapse
|
11
|
De Rosa MA, Bernardi MT, Kleppe S, Walz K. Hearing Loss: Genetic Testing, Current Advances and the Situation in Latin America. Genes (Basel) 2024; 15:178. [PMID: 38397168 PMCID: PMC10888486 DOI: 10.3390/genes15020178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Congenital hearing loss is the most common birth defect, estimated to affect 2-3 in every 1000 births, with ~50-60% of those related to genetic causes. Technological advances enabled the identification of hundreds of genes related to hearing loss (HL), with important implications for patients, their families, and the community. Despite these advances, in Latin America, the population with hearing loss remains underdiagnosed, with most studies focusing on a single locus encompassing the GJB2/GJB6 genes. Here we discuss how current and emerging genetic knowledge has the potential to alter the approach to diagnosis and management of hearing loss, which is the current situation in Latin America, and the barriers that still need to be overcome.
Collapse
Affiliation(s)
- Maria Agustina De Rosa
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (M.A.D.R.); (M.T.B.)
| | - Maria T. Bernardi
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (M.A.D.R.); (M.T.B.)
| | - Soledad Kleppe
- Department of Clinical Pediatrics, Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires C1199ABB, Argentina;
| | - Katherina Walz
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (M.A.D.R.); (M.T.B.)
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, 1501 NW 10th Avenue, BRB-418 (M-860), Miami, FL 33136, USA
| |
Collapse
|
12
|
Pan X, Huang P, Ali SS, Renslo B, Hutchinson TE, Erwin N, Greenberg Z, Ding Z, Li Y, Warnecke A, Fernandez NE, Staecker H, He M. CRISPR-Cas9 Engineered Extracellular Vesicles for the Treatment of Dominant Progressive Hearing Loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557853. [PMID: 38168224 PMCID: PMC10760051 DOI: 10.1101/2023.09.14.557853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Clinical translation of gene therapy has been challenging, due to limitations in current delivery vehicles such as traditional viral vectors. Herein, we report the use of gRNA:Cas9 ribonucleoprotein (RNP) complexes engineered extracellular vesicles (EVs) for in vivo gene therapy. By leveraging a novel high-throughput microfluidic droplet-based electroporation system (μDES), we achieved 10-fold enhancement of loading efficiency and more than 1000-fold increase in processing throughput on loading RNP complexes into EVs (RNP-EVs), compared with conventional bulk electroporation. The flow-through droplets serve as enormous bioreactors for offering millisecond pulsed, low-voltage electroporation in a continuous-flow and scalable manner, which minimizes the Joule heating influence and surface alteration to retain natural EV stability and integrity. In the Shaker-1 mouse model of dominant progressive hearing loss, we demonstrated the effective delivery of RNP-EVs into inner ear hair cells, with a clear reduction of Myo7ash1 mRNA expression compared to RNP-loaded lipid-like nanoparticles (RNP-LNPs), leading to significant hearing recovery measured by auditory brainstem responses (ABR).
Collapse
Affiliation(s)
- Xiaoshu Pan
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Peixin Huang
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States
| | - Samantha S. Ali
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Bryan Renslo
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States
| | - Tarun E Hutchinson
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Nina Erwin
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Zachary Greenberg
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Zuo Ding
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Yanjun Li
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida, 32610, United States
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, 30625 Hannover, Germany
| | - Natalia E. Fernandez
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| | - Hinrich Staecker
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States
| | - Mei He
- College of Pharmacy, Department of Pharmaceutics, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
13
|
Zhu W, Li M, Zou J, Zhang D, Fang M, Sun Y, Li C, Tang M, Wang Y, Zhou Q, Zhao T, Li W, Hu Z, Hu B. Induction of local immunosuppression in allogeneic cell transplantation by cell-type-specific expression of PD-L1 and CTLA4Ig. Stem Cell Reports 2023; 18:2344-2355. [PMID: 37995700 PMCID: PMC10724073 DOI: 10.1016/j.stemcr.2023.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
Immune rejection has long hindered allogeneic cell transplantation therapy. Current genetic modification approaches, including direct targeting of major histocompatibility complex or constitutive expression of immune inhibitory molecules, exhibit drawbacks such as severe adverse effects or elevated tumorigenesis risks. To overcome these limitations, we introduce an innovative approach to induce cell-type-specific immune tolerance in differentiated cells. By engineering human embryonic stem cells, we ensure the exclusive production of the immune inhibitory molecules PD-L1/CTLA4Ig in differentiated cells. Using this strategy, we generated hepatocyte-like cells expressing PD-L1 and CTLA4Ig, which effectively induced local immunotolerance. This approach was evaluated in a humanized mouse model that mimics the human immune system dynamics. We thus demonstrate a robust and selective induction of immunotolerance specific to hepatocytes, improving graft survival without observed tumorigenesis. This precise immune tolerance strategy holds great promise for advancing the development of stem cell-based therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Wenliang Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Mengqi Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Jun Zou
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Jilin, China; National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin 130061, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Minghui Fang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Jilin, China; National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin 130061, China
| | - Yun Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Can Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Mingming Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Zheng Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Jilin, China; National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin 130061, China.
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
14
|
Zhu W, Du W, Rameshbabu AP, Armstrong AM, Silver S, Kim Y, Wei W, Shu Y, Liu X, Lewis MA, Steel KP, Chen ZY. Targeted genome editing restores auditory function in adult mice with progressive hearing loss caused by a human microRNA mutation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564008. [PMID: 37961137 PMCID: PMC10634841 DOI: 10.1101/2023.10.26.564008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mutations in microRNA-96 ( MIR96 ) cause dominant delayed onset hearing loss DFNA50 without treatment. Genome editing has shown efficacy in hearing recovery by intervention in neonatal mice, yet editing in the adult inner ear is necessary for clinical applications. Here, we developed an editing therapy for a C>A point mutation in the seed region of the Mir96 gene, Mir96 14C>A associated with hearing loss by screening gRNAs for genome editors and optimizing Cas9 and sgRNA scaffold for efficient and specific mutation editing in vitro. By AAV delivery in pre-symptomatic (3-week-old) and symptomatic (6-week-old) adult Mir96 14C>A mutant mice, hair cell on-target editing significantly improved hearing long-term, with an efficacy inversely correlated with injection age. We achieved transient Cas9 expression without the evidence of AAV genomic integration to significantly reduce the safety concerns associated with editing. We developed an AAV-sgmiR96-master system capable of targeting all known human MIR96 mutations. As 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 lays the foundation for future treatment of DFNA50 caused by MIR96 mutations.
Collapse
|