1
|
Ji RJ, Wang MY, Zhang Y. Precision epitope editing: A path to advanced immunotherapies. CELL INSIGHT 2025; 4:100226. [PMID: 39906754 PMCID: PMC11791281 DOI: 10.1016/j.cellin.2024.100226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025]
Abstract
The ability to recognize antigen epitope is crucial for generating an effective immune response. By engineering these epitopes, researchers can reduce on-target/off-tumor toxicity associated with targeted immunotherapy. Recent studies indicate that employing various gene editing tools to modify the epitopes of healthy hematopoietic stem and progenitor cells (HSPCs) can protect these cells from toxicity during tumor eradication, all while preserving their differentiation and function. This advancement greatly enhances the safety and efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Rui-Jin Ji
- Esophagus, Mediastinum and Lymphatic Oncology Department, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Mu-Yao Wang
- Esophagus, Mediastinum and Lymphatic Oncology Department, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Ying Zhang
- Esophagus, Mediastinum and Lymphatic Oncology Department, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Centre for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, 430071, Hubei, China
- State Key Laboratory of Virology, Wuhan University, Wuhan, 430071, Hubei, China
| |
Collapse
|
2
|
Konishi CT, Mulaiese N, Butola T, Zhang Q, Kagan D, Yang Q, Pressler M, Dirvin BG, Devinsky O, Basu J, Long C. Modeling and correction of protein conformational disease in iPSC-derived neurons through personalized base editing. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102441. [PMID: 39877004 PMCID: PMC11773622 DOI: 10.1016/j.omtn.2024.102441] [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: 03/22/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Altered protein conformation can cause incurable neurodegenerative disorders. Mutations in SERPINI1, the gene encoding neuroserpin, can alter protein conformation resulting in cytotoxic aggregation leading to neuronal death. Familial encephalopathy with neuroserpin inclusion bodies (FENIB) is a rare autosomal dominant progressive myoclonic epilepsy that progresses to dementia and premature death. We developed HEK293T and induced pluripotent stem cell (iPSC) models of FENIB, harboring a patient-specific pathogenic SERPINI1 variant or stably overexpressing mutant neuroserpin fused to GFP (MUT NS-GFP). Here, we utilized a personalized adenine base editor (ABE)-mediated approach to correct the pathogenic variant efficiently and precisely to restore neuronal dendritic morphology. ABE-treated MUT NS-GFP cells demonstrated reduced inclusion size and number. Using an inducible MUT NS-GFP neuron system, we identified early prevention of toxic protein expression allowed aggregate clearance, while late prevention halted further aggregation. To address several challenges for clinical applications of gene correction, we developed a neuron-specific engineered virus-like particle to optimize neuronal ABE delivery, resulting in higher correction efficiency. Our findings provide a targeted strategy that may treat FENIB and potentially other neurodegenerative diseases due to altered protein conformation such as Alzheimer's and Huntington's diseases.
Collapse
Affiliation(s)
- Colin T. Konishi
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Nancy Mulaiese
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Tanvi Butola
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Qinkun Zhang
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Dana Kagan
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Qiaoyan Yang
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Mariel Pressler
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Brooke G. Dirvin
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Orrin Devinsky
- Department of Neurology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Jayeeta Basu
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 100016, USA
| | - Chengzu Long
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY 100016, USA
- Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY 100016, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 100016, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY 100016, USA
| |
Collapse
|
3
|
Dou B, Li Y, Wang F, Chen L, Zhang W. Chassis engineering for high light tolerance in microalgae and cyanobacteria. Crit Rev Biotechnol 2025; 45:257-275. [PMID: 38987975 DOI: 10.1080/07388551.2024.2357368] [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: 03/25/2024] [Revised: 04/21/2024] [Accepted: 05/05/2024] [Indexed: 07/12/2024]
Abstract
Oxygenic photosynthesis in microalgae and cyanobacteria is considered an important chassis to accelerate energy transition and mitigate global warming. Currently, cultivation systems for photosynthetic microbes for large-scale applications encountered excessive light exposure stress. High light stress can: affect photosynthetic efficiency, reduce productivity, limit cell growth, and even cause cell death. Deciphering photoprotection mechanisms and constructing high-light tolerant chassis have been recent research focuses. In this review, we first briefly introduce the self-protection mechanisms of common microalgae and cyanobacteria in response to high light stress. These mechanisms mainly include: avoiding excess light absorption, dissipating excess excitation energy, quenching excessive high-energy electrons, ROS detoxification, and PSII repair. We focus on the species-specific differences in these mechanisms as well as recent advancements. Then, we review engineering strategies for creating high-light tolerant chassis, such as: reducing the size of the light-harvesting antenna, optimizing non-photochemical quenching, optimizing photosynthetic electron transport, and enhancing PSII repair. Finally, we propose a comprehensive exploration of mechanisms: underlying identified high light tolerant chassis, identification of new genes pertinent to high light tolerance using innovative methodologies, harnessing CRISPR systems and artificial intelligence for chassis engineering modification, and introducing plant photoprotection mechanisms as future research directions.
Collapse
Affiliation(s)
- Biyun Dou
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, P.R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P.R. China
| | - Yang Li
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, P.R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P.R. China
| | - Fangzhong Wang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, P.R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P.R. China
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin, P.R. China
| | - Lei Chen
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, P.R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P.R. China
| | - Weiwen Zhang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, P.R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, P.R. China
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin, P.R. China
| |
Collapse
|
4
|
Zhang D, Parth F, da Silva LM, Ha TC, Schambach A, Boch J. Engineering a bacterial toxin deaminase from the DYW-family into a novel cytosine base editor for plants and mammalian cells. Genome Biol 2025; 26:18. [PMID: 39901278 PMCID: PMC11789416 DOI: 10.1186/s13059-025-03478-w] [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: 05/24/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Base editors are precise editing tools that employ deaminases to modify target DNA bases. The DYW-family of cytosine deaminases is structurally and phylogenetically distinct and might be harnessed for genome editing tools. We report a novel CRISPR/Cas9-cytosine base editor using SsdA, a DYW-like deaminase and bacterial toxin. A G103S mutation in SsdA enhances C-to-T editing efficiency while reducing its toxicity. Truncations result in an extraordinarily small enzyme. The SsdA-base editor efficiently converts C-to-T in rice and barley protoplasts and induces mutations in rice plants and mammalian cells. The engineered SsdA is a highly efficient genome editing tool.
Collapse
Affiliation(s)
- Dingbo Zhang
- Institute of Plant Genetics, Leibniz Universität Hannover, Herrenhäuser Str. 2, Hannover, 30419, Germany
- Research Institute of Biology and Agriculture, University of Science and Technology, Beijing, 100083, China
| | - Fiona Parth
- Institute of Plant Genetics, Leibniz Universität Hannover, Herrenhäuser Str. 2, Hannover, 30419, Germany
| | - Laura Matos da Silva
- Institute of Plant Genetics, Leibniz Universität Hannover, Herrenhäuser Str. 2, Hannover, 30419, Germany
| | - Teng-Cheong Ha
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jens Boch
- Institute of Plant Genetics, Leibniz Universität Hannover, Herrenhäuser Str. 2, Hannover, 30419, Germany.
| |
Collapse
|
5
|
Clarissa EM, Karmacharya M, Choi H, Kumar S, Cho YK. Nature Inspired Delivery Vehicles for CRISPR-Based Genome Editing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409353. [PMID: 39901476 DOI: 10.1002/smll.202409353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/16/2025] [Indexed: 02/05/2025]
Abstract
The advent of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based genome editing technologies has opened up groundbreaking possibilities for treating a wide spectrum of genetic disorders and diseases. However, the success of these technologies relies heavily on the development of efficient and safe delivery systems. Among the most promising approaches are natural and synthetic nanocarrier-mediated delivery systems, including viral vectors, extracellular vesicles (EVs), engineered cellular membrane particles, liposomes, and various nanoparticles. These carriers enhance the efficacy of the CRISPR system by providing a unique combination of efficiency, specificity, and reduced immunogenicity. Synthetic carriers such as liposomes and nanoparticles facilitate CRISPR delivery with high reproducibility and customizable functions. Viral vectors, renowned for their high transduction efficiency and broad tropism, serve as powerful vehicles for delivering CRISPR components to various cell types. EVs, as natural carriers of RNA and proteins, offer a stealth mechanism to evade immune detection, allowing for the targeted delivery of genome editors with minimal off-target effects. Engineered cellular membrane particles further improve delivery by simulating the cellular environment, enhancing uptake, and minimizing immune response. This review explores the innovative integration of CRISPR genome editors with various nanocarrier systems, focusing on recent advancements, applications, and future directions in therapeutic genome editing.
Collapse
Affiliation(s)
- Elizabeth Maria Clarissa
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Mamata Karmacharya
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Hyunmin Choi
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Sumit Kumar
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Yoon-Kyoung Cho
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| |
Collapse
|
6
|
Hu L, Han J, Wang HD, Cheng ZH, Lv CC, Liu DF, Yu HQ. A universal and wide-range cytosine base editor via domain-inlaid and fidelity-optimized CRISPR-FrCas9. Nat Commun 2025; 16:1260. [PMID: 39893181 PMCID: PMC11787337 DOI: 10.1038/s41467-025-56655-7] [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: 07/04/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
CRISPR-based base editor (BE) offer diverse editing options for genetic engineering of microorganisms, but its application is limited by protospacer adjacent motif (PAM) sequences, context preference, editing window, and off-target effects. Here, a series of iteratively improved cytosine base editors (CBEs) are constructed using the FrCas9 nickase (FrCas9n) with the unique PAM palindromic structure (NNTA) to alleviate these challenges. The deaminase domain-inlaid FrCas9n exhibits an editing range covering 38 nucleotides upstream and downstream of the palindromic PAM, without context preference, which is 6.3 times larger than that of traditional CBEs. Additionally, lower off-target editing is achieved when incorporating high-fidelity mutations at R61A and Q964A in FrCas9n, while maintaining high editing efficiency. The final CBE, HF-ID824-evoCDA-FrCas9n demonstrates broad applicability across different microbes such as Escherichia coli MG1655, Shewanella oneidensis MR-1, and Pseudomonas aeruginosa PAO1. Collectively, this tool offers robust gene editing for facilitating mechanistic studies, functional exploration, and protein evolution in microbes.
Collapse
Affiliation(s)
- Lan Hu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Jing Han
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Hao-Da Wang
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Zhou-Hua Cheng
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| | - Chang-Ce Lv
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Dong-Feng Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| | - Han-Qing Yu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
7
|
Rimskaya B, Shebanov N, Entelis N, Mazunin I. Enzymatic tools for mitochondrial genome manipulation. Biochimie 2025; 229:114-128. [PMID: 39426703 DOI: 10.1016/j.biochi.2024.10.013] [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: 06/27/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Mutations in mitochondrial DNA (mtDNA) can manifest phenotypically as a wide range of neuromuscular and neurodegenerative pathologies that are currently only managed symptomatically without addressing the root cause. A promising approach is the development of molecular tools aimed at mtDNA cutting or editing. Unlike nuclear DNA, a cell can have hundreds or even thousands of mitochondrial genomes, and mutations can be present either in all of them or only in a subset. Consequently, the developed tools are aimed at reducing the number of copies of mutant mtDNA or editing mutant nucleotides. Despite some progress in the field of mitochondrial genome editing in human cells, working with model animals is still limited due to the complexity of their creation. Furthermore, not all existing editing systems can be easily adapted to function within mitochondria. In this review, we evaluate the mtDNA editing tools available today, with a particular focus on specific mtDNA mutations linked to hereditary mitochondrial diseases, aiming to provide an in-depth understanding of both the opportunities and hurdles to the development of mitochondrial genome editing technologies.
Collapse
Affiliation(s)
- Beatrisa Rimskaya
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, 143026, Russian Federation; Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, 141700, Russian Federation; Department of Biology and Genetics, Petrovsky Medical University, Moscow, 117418, Russian Federation
| | - Nikita Shebanov
- UMR7156 Molecular Genetics, Genomics, Microbiology, University of Strasbourg - CNRS, Strasbourg, 67000, France
| | - Nina Entelis
- UMR7156 Molecular Genetics, Genomics, Microbiology, University of Strasbourg - CNRS, Strasbourg, 67000, France.
| | - Ilya Mazunin
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, 143026, Russian Federation; Department of Biology and Genetics, Petrovsky Medical University, Moscow, 117418, Russian Federation.
| |
Collapse
|
8
|
Liu Y, Bai X, Feng X, Liu S, Hu Y, Chu H, Zhang L, Cai B, Ma Y. Revolutionizing animal husbandry: Breakthroughs in gene editing delivery systems. Gene 2025; 935:149044. [PMID: 39490705 DOI: 10.1016/j.gene.2024.149044] [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: 08/20/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Gene editing technology has become an essential tool for advancing breeding practices, enhancing disease resistance, and boosting productivity in animal husbandry. Despite its potential, the delivery of gene editing reagents into cells faces several challenges, including low targeting efficiency, immunogenicity, and cytotoxicity, which have hindered its wider application in the field. This review discusses the evolution of gene editing technologies and highlights recent advancements in various delivery methods used in animal husbandry. It critically evaluates the strengths and weaknesses of these different delivery approaches while identifying potential directions for future development. The goal is to equip researchers with effective strategies to optimize delivery methods, ultimately facilitating the implementation and progress of gene editing technologies in animal husbandry.
Collapse
Affiliation(s)
- Yuan Liu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Xue Bai
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China
| | - Xue Feng
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Shuang Liu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Yamei Hu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Hongen Chu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Lingkai Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Bei Cai
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| | - Yun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
9
|
Yuan Y, Li Y, Li G, Lei L, Huang X, Li M, Yao Y. Intelligent Design of Lipid Nanoparticles for Enhanced Gene Therapeutics. Mol Pharm 2025. [PMID: 39878334 DOI: 10.1021/acs.molpharmaceut.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Lipid nanoparticles (LNPs) are an effective delivery system for gene therapeutics. By optimizing their formulation, the physiochemical properties of LNPs can be tailored to improve tissue penetration, cellular uptake, and precise targeting. The application of these targeted delivery strategies within the LNP framework ensures efficient delivery of therapeutic agents to specific organs or cell types, thereby maximizing therapeutic efficacy. In the realm of genome editing, LNPs have emerged as a potent vehicle for delivering CRISPR/Cas components, offering significant advantages such as high in vivo efficacy. The incorporation of machine learning into the optimization of LNP platforms for gene therapeutics represents a significant advancement, harnessing its predictive capabilities to substantially accelerate the research and development process. This review highlights the dynamic evolution of LNP technology, which is expected to drive transformative progress in the field of gene therapy.
Collapse
Affiliation(s)
- Yichen Yuan
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Ying Li
- Research Center for Space Computing System, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
| | - Liqun Lei
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Xingxu Huang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
- Zhejiang Key Laboratory of Intelligent Manufacturing for Functional Chemicals, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| |
Collapse
|
10
|
Ramezani M, Weisbart E, Bauman J, Singh A, Yong J, Lozada M, Way GP, Kavari SL, Diaz C, Leardini E, Jetley G, Pagnotta J, Haghighi M, Batista TM, Pérez-Schindler J, Claussnitzer M, Singh S, Cimini BA, Blainey PC, Carpenter AE, Jan CH, Neal JT. A genome-wide atlas of human cell morphology. Nat Methods 2025:10.1038/s41592-024-02537-7. [PMID: 39870862 DOI: 10.1038/s41592-024-02537-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/25/2024] [Indexed: 01/29/2025]
Abstract
A key challenge of the modern genomics era is developing empirical data-driven representations of gene function. Here we present the first unbiased morphology-based genome-wide perturbation atlas in human cells, containing three genome-wide genotype-phenotype maps comprising CRISPR-Cas9-based knockouts of >20,000 genes in >30 million cells. Our optical pooled cell profiling platform (PERISCOPE) combines a destainable high-dimensional phenotyping panel (based on Cell Painting) with optical sequencing of molecular barcodes and a scalable open-source analysis pipeline to facilitate massively parallel screening of pooled perturbation libraries. This perturbation atlas comprises high-dimensional phenotypic profiles of individual cells with sufficient resolution to cluster thousands of human genes, reconstruct known pathways and protein-protein interaction networks, interrogate subcellular processes and identify culture media-specific responses. Using this atlas, we identify the poorly characterized disease-associated TMEM251/LYSET as a Golgi-resident transmembrane protein essential for mannose-6-phosphate-dependent trafficking of lysosomal enzymes. In sum, this perturbation atlas and screening platform represents a rich and accessible resource for connecting genes to cellular functions at scale.
Collapse
Affiliation(s)
- Meraj Ramezani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Erin Weisbart
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Bauman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanford University, Stanford, CA, USA
| | - Avtar Singh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech Department of Cellular and Tissue Genomics, South San Francisco, CA, USA
| | - John Yong
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Maria Lozada
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gregory P Way
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sanam L Kavari
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Celeste Diaz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanford University, Stanford, CA, USA
| | - Eddy Leardini
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gunjan Jetley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jenlu Pagnotta
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Thiago M Batista
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
| | - Joaquín Pérez-Schindler
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
| | - Melina Claussnitzer
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Beth A Cimini
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paul C Blainey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Research, MIT, Cambridge, MA, USA
| | | | - Calvin H Jan
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - James T Neal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
11
|
Tang J, Du K. Mitochondrial base editing: from principle, optimization to application. Cell Biosci 2025; 15:9. [PMID: 39856740 PMCID: PMC11762502 DOI: 10.1186/s13578-025-01351-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
In recent years, mitochondrial DNA (mtDNA) base editing systems have emerged as bioengineering tools. DddA-derived cytosine base editors (DdCBEs) have been developed to specifically induce C-to-T conversion in mtDNA by the fusion of sequence-programmable transcription activator-like effector nucleases (TALENs) or zinc-finger nucleases (ZFNs), and split deaminase derived from interbacterial toxins. Similar to DdCBEs, mtDNA adenine base editors have been developed with the ability to introduce targeted A-to-G conversions into human mtDNA. In this review, we summarize the principles of mtDNA base-editing systems and elaborate on the evolution of different platforms of mtDNA base editors, including their deaminase replacement, engineering of DddAtox variants, structure optimization and editing outcomes. Finally, we highlight their applications in animal models and human embroys and discuss the future developmental direction and challenges of mtDNA base editors.
Collapse
Affiliation(s)
- Jinling Tang
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Kunzhao Du
- Jinshan Hospital Center for Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
12
|
Perez AR, Mavrothalassitis O, Chen JS, Hellman J, Gropper MA. CRISPR: fundamental principles and implications for anaesthesia. Br J Anaesth 2025:S0007-0912(24)00760-8. [PMID: 39855935 DOI: 10.1016/j.bja.2024.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 01/27/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-based medical therapies are increasingly gaining regulatory approval worldwide. Consequently, patients receiving CRISPR therapy will come under the care of anaesthesiologists. An understanding of CRISPR, its technological implementations, and the characteristics of patients likely to receive this therapy will be essential to caring for this patient population. However, the role of CRISPR in anaesthesiology extends beyond simply caring for patients with prior CRISPR therapy. CRISPR has multiple direct potential applications in anaesthesia, particularly for managing chronic pain and critical illness. Additionally, given the unique skills anaesthesiologists possess, CRISPR potentially allows new roles for anaesthesiologists in the field of oncology. Consequently, CRISPR technology could enable new domains of anaesthetic practice. This review provides a primer on CRISPR for anaesthesiologists and an overview on how the technology could impact the field.
Collapse
Affiliation(s)
- Alexendar R Perez
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Silico Therapeutics, Inc., San Jose, CA, USA.
| | - Orestes Mavrothalassitis
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | | | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Gropper
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Marx N, Otte AM, Leitner K, Sitepu R, Berger T, Schäpertöns V, Huber CG, Zhu Q, Nema S, Higgins JJ, Borth N. Characterization of large transgene integrations in Chinese hamster ovary cells using a bioengineered mammalian transposase. Biotechnol Prog 2025:e3524. [PMID: 39846713 DOI: 10.1002/btpr.3524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 01/24/2025]
Abstract
We present the first use of a bioengineered mammalian transposase system derived from Myotis lucifugus (bMLT) for integration of expression vectors into the CHO genome, focusing on GFP and trastuzumab production. Initially, CHO-K1 cells are transfected with a GFP reporter and varying amounts of bMLT DNA or mRNA. GFP expression is monitored over 17 weeks without selective pressure. Transfection efficiency shows around 90% GFP-positive cells, but in control cultures GFP expression disappears after 10 days. In contrast, bMLT-treated cultures maintain stable GFP expression, with a dose-dependent integration efficiency of up to 60%. The highest GFP expression per cell is observed with lower bMLT amounts. Next-generation sequencing analysis reveals multiple integration sites, with 85% correctly integrated sequences. Next, CHO-GS-/- cells are transfected with trastuzumab and bMLT DNA or mRNA. Cells are selected in glutamine-free medium with varying methionine sulfoximine (MSX) concentrations. Recovery is faster without MSX, and no difference is observed between bMLT DNA and mRNA transfections. bMLT-treated cultures show a higher percentage of trastuzumab-secreting cells (40%-55%) compared with random integration (0.3%-0.5%). The absence of insulators in the trastuzumab plasmid likely affects selection behavior, as integration in heterochromatic regions results in gene repression. Overall, bMLT-mediated integration proves efficient, generating stable cell pools with high expression profiles without selective pressure. The integration sites' genomic location significantly impacts productivity, with favorable regions supporting higher expression. This method shows promise for the rapid and efficient generation of high-producing cell lines and for rapid evaluation of long-term effects of different cell engineering approaches.
Collapse
Affiliation(s)
- Nicolas Marx
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Anna-Maria Otte
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Klaus Leitner
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Rehmadanta Sitepu
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Thomas Berger
- Bioanalytical Research Labs, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Veronika Schäpertöns
- Bioanalytical Research Labs, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Christian G Huber
- Bioanalytical Research Labs, Paris-Lodron University Salzburg, Salzburg, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Quan Zhu
- SalioGen Therapeutics, Lexington, Massachusetts, USA
| | - Sandeep Nema
- SalioGen Therapeutics, Lexington, Massachusetts, USA
| | | | - Nicole Borth
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
14
|
Zhao W, Zhu X, Huang G, Gu H, Bi Y, Tang D, Ren H. Application of Multiple Base-Editing Mediated by Polycistronic tRNA-gRNA-Processing System in Pig Cells. Biotechnol Bioeng 2025. [PMID: 39844444 DOI: 10.1002/bit.28931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
Gene edited pigs have extensive and important application value in the fields of agriculture and biomedicine. With the increasing demand in medical research and agricultural markets, more and more application scenarios require gene edited pigs to possess two or even more advantageous phenotypes simultaneously. The current production of multi gene edited pigs is inefficient, time-consuming, and costly, and there is an urgent need to develop efficient and accurate multi gene editing application technologies. The polycistronic tRNA-gRNA-processing system (PTG), developed based on endogenous tRNA self-processing systems, has been shown to exhibit efficient multi gene editing in plants. This study aims to combine a PTG strategy with multiple gRNA production functions with an adenine base editor (ABE) to test its feasibility for efficient and precise multi gene base editing in pig cells. The results indicate that the PTG based integrated ABE plasmid can perform efficient base editing at multiple gene loci in pig cells. And while the gene editing efficiency was significantly improved, no indel and sgRNA dependent off target effects caused by DSB were detected. This work permit will provide a solid foundation for the production of multi gene edited pigs with agricultural and medical applications.
Collapse
Affiliation(s)
- Wudi Zhao
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Gene Editing Technology Center of Guangdong Province, Foshan University, Foshan, China
| | - Xiangxing Zhu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Gene Editing Technology Center of Guangdong Province, Foshan University, Foshan, China
| | - Guobin Huang
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Gene Editing Technology Center of Guangdong Province, Foshan University, Foshan, China
| | - Hao Gu
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Yanzhen Bi
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Dongsheng Tang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Gene Editing Technology Center of Guangdong Province, Foshan University, Foshan, China
| | - Hongyan Ren
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
15
|
Wang J, Gao T, Zhang D, Tang Y, Gu J. Phospholipase C epsilon 1 as a therapeutic target in cardiovascular diseases. J Adv Res 2025:S2090-1232(25)00051-7. [PMID: 39855298 DOI: 10.1016/j.jare.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Phospholipase C epsilon 1 (PLCε1) can hydrolyze phosphatidylinositol-4,5-bisphosphate and phosphatidylinositol-4-phosphate at the plasma membrane and perinuclear membrane in the cardiovascular system, producing lipid-derived second messengers. These messengers are considered prominent triggers for various signal transduction processes. Notably, diverse cardiac phenotypes have been observed in cardiac-specific and global Plce1 knockout mice under conditions of pathological stress. It is well established that the cardiac-specific Plce1 knockout confers cardioprotective benefits. Therefore, the development of tissue/cell-specific targeting approaches is critical for advancing therapeutic interventions. AIM OF REVIEW This review aims to distill the foundational biology and functional significance of PLCε1 in cardiovascular diseases, as well as to explore potential avenues for research and the development of novel therapeutic strategies targeting PLCε1. KEY SCIENTIFIC CONCEPTS OF REVIEW Cardiovascular diseases remain the leading cause of morbidity and mortality worldwide, with incidence rates escalating annually. A comprehensive understanding of the multifaceted role of PLCε1 is essential for enhancing the diagnosis, management, and prognostic assessment of patients suffering from cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Dongmei Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
16
|
Zhang X, Qiu M, Han B, Liao L, Peng X, Lin J, Zhang N, Hai L, Liang L, Ma Y, Li W, Liu M. Generation and propagation of high fecundity gene edited fine wool sheep by CRISPR/Cas9. Sci Rep 2025; 15:2557. [PMID: 39833409 PMCID: PMC11747170 DOI: 10.1038/s41598-025-86592-w] [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: 09/27/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
CRISPR/Cas9 technology has been widely utilized to enhance productive performance, increase disease resistance and generate medical models in livestock. The FecB allele in sheep is a mutation in the BMPRIB gene, recognized as the first major gene responsible for the high fecundity trait in sheep, leading to an increased ovulation rate in ewe. In this study, we employed CRISPR/Cas9-mediated homologous-directed repair (HDR) to introduce a defined point mutation (c.746 A > G) using single-stranded oligonucleotides (ssODN) and the ligase IV inhibitor (SCR7) into the BMPRIB gene of fine wool sheep. A total of nine gene-edited sheep were produced, six of which carried the targeted point mutation, with a precise base substitution efficiency (A > G) of 31.6%. Based on the six targeted founders (F0), we expanded the BMPRIB-targeted population, which included F1 heterozygous (B+) and F2 homozygous(BB) or heterozygous offspring. The average litter size of F1 ewes carrying the B + allele reached 170%, comparable to that of heterozygous native Australian Booroola sheep. Gene-edited ewes with B + and BB genotype produced 0.62 and 0.42 more lambs, respectively, compared to wide-type ewes (p < 0.01). Our results also indicated that the parity signification, our data demonstrate that highly efficient introduction of the intended base mutation into the sheep genome can be achieved by combining the CRISPR/Cas9 system with ssODN and SCR7. The offspring of BMPR/B edited sheep with the defined mutation exhibited high fecundity performance. Compared to conventional sheep breeding strategies, genetic improvement through gene editing offered significant advantages without compromising the fine wool traits of Merino sheep, which are often affected by routine cross-breeding methods.
Collapse
Affiliation(s)
- Xuemei Zhang
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Meiyu Qiu
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Bing Han
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Li Liao
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Xinrong Peng
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Jiapeng Lin
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Ning Zhang
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Lati Hai
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Long Liang
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Yila Ma
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Wenrong Li
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China
| | - Mingjun Liu
- Key Laboratory of Genetics, Breeding and Reproduction of Grass-Feeding Livestock, Key Laboratory of Animal Biotechnology of Xinjiang, Ministry of Agriculture(MOA), Urumqi, 830026, Xinjiang, China.
- Institute of Animal Biotechnology, Xinjiang Academy of Animal Science, Urumqi, 830026, Xinjiang, China.
| |
Collapse
|
17
|
Wang Z, Wu J, Lv Z, Liang P, Li Q, Li Y, Guo Y. LMNA-related cardiomyopathy: From molecular pathology to cardiac gene therapy. J Adv Res 2025:S2090-1232(25)00001-3. [PMID: 39827909 DOI: 10.1016/j.jare.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The genetic variants of LMNA cause an array of diseases that often affect the heart. LMNA-related cardiomyopathy exhibits high-penetrance and early-onset phenotypes that lead to late-stage heart failure or lethal arrhythmia. As a subtype of dilated cardiomyopathy and arrhythmogenic cardiomyopathy, LMNA-related cardiac dysfunction is resistant to existing cardiac therapeutic strategies, leaving a major unmet clinical need in cardiomyopathy management. AIM OF REVIEW Here we comprehensively summarize current knowledge about the genetic basis, disease models and pathological mechanisms of LMNA-related cardiomyopathy. Recent translational studies were highlighted to indicate new therapeutic modalities such as gene supplementation, gene silencing and genome editing therapy, which offer potential opportunities to overcome the difficulties in the development of specific drugs for this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW LMNA-related cardiomyopathy involves many diverse disease mechanisms that preclude small-molecule drugs that target only a small fraction of the mechanisms. Agreeing to this notion, the first-in-human clinical trial for this disease recently reported futility. By contrast, gene therapy offers the new hope to directly intervene LMNA variants and demonstrates a tremendous potential for breakthrough therapy for this disease. Concepts in this review are also applicable to studies of other genetic diseases that lack effective therapeutics.
Collapse
Affiliation(s)
- Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiahao Wu
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyuan Lv
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China.
| | - Qirui Li
- Department of Cardiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Yifei Li
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
18
|
Wang Q, You J, Li Y, Zhang J, Wang Y, Xu M, Rao Z. Continuous Evolution of Protein through T7 RNA Polymerase-Guided Base Editing in Corynebacterium glutamicum. ACS Synth Biol 2025; 14:216-229. [PMID: 39700484 DOI: 10.1021/acssynbio.4c00606] [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] [Indexed: 12/21/2024]
Abstract
In vivo targeted mutagenesis technologies are the basis for the continuous directed evolution of specific proteins. Here, an efficient mutagenesis system (CgMutaT7) for continuous evolution of the targeted gene in Corynebacterium glutamicum was developed. First, cytosine deaminase and uracil-DNA glycosylase inhibitor were sequentially fused to T7 RNA polymerase using flexible linkers to build the CgMutaT7 system, which introduces mutations in targeted regions controlled by the T7 promoter. After a series of optimizations, the resulting targeted mutagenesis system (CgMutaT74) can increase the mutant frequency of the target gene by 1.12 × 104-fold, with low off-target mutant frequency. Subsequently, high-throughput sequencing further revealed that the CgMutaT74 system performs efficient and uniform C → T transitions in at least a 1.8 kb DNA region. Finally, the xylose isomerase was successfully continuously evolved by CgMutaT74 to improve the xylose utilization, indicating that the CgMutaT7 system has great potential for applications in the continuous evolution of protein function and expression components.
Collapse
Affiliation(s)
- Qing Wang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Jiajia You
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Yichen Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Jie Zhang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Yi Wang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Meijuan Xu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Zhiming Rao
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| |
Collapse
|
19
|
Dong Y, Chen Z. Accelerated Metabolic Engineering for Industrial Strain Development via the Construction of a Large-Scale Genome Library. ACS Synth Biol 2025; 14:41-56. [PMID: 39680725 DOI: 10.1021/acssynbio.4c00620] [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] [Indexed: 12/18/2024]
Abstract
Production of chemicals via metabolic engineering of microbes is becoming highly important for sustainable bioeconomy. Conventional metabolic engineering methodologies typically involve labor-intensive and time-consuming processes of iterative genetic modifications, which are inefficient in identifying new genetic targets for the construction of robust industrial strains on a large scale. To accelerate the creation of efficient microbial cell factories and enhance our insights into cellular metabolism, diverse large-scale genome libraries are emerging as powerful tools, which can be established through multiplex or parallel genome editing, gene expression regulation, and incorporation of evolutionary strategies. In this review, we discuss the latest advancements in the construction of genome-scale libraries as well as their applications within the domain of metabolic engineering. We also address the limitations of various techniques and provide insights into future prospects for the field.
Collapse
Affiliation(s)
- Yufei Dong
- Key Laboratory of Industrial Biocatalysis (Ministry of Education), Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Zhen Chen
- Key Laboratory of Industrial Biocatalysis (Ministry of Education), Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
20
|
Bhakta S, Kodama H, Mimaki M, Tsukahara T. Restoration of Genetic Code in Macular Mouse Fibroblasts via APOBEC1-Mediated RNA Editing. Biomolecules 2025; 15:136. [PMID: 39858530 PMCID: PMC11762822 DOI: 10.3390/biom15010136] [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: 11/26/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
RNA editing is a significant mechanism underlying genetic variation and protein molecule alteration; C-to-U RNA editing, specifically, is important in the regulation of mammalian genetic diversity. The ability to define and limit accesses of enzymatic machinery to avoid the modification of unintended targets is key to the success of RNA editing. Identification of the core component of the apoB RNA editing holoenzyme, APOBEC, and investigation into new candidate genes encoding other elements of the complex could reveal further details regarding APOBEC-mediated mRNA editing. Menkes disease is a recessive X-chromosome-linked hereditary syndrome in humans, caused by defective copper metabolism due to mutations in the ATP7A gene, which encodes a copper transport protein. Here, we generated plasmids encoding the MS2 system and the APOBEC1 deaminase domain and used a guide RNA with flanking MS2 sites to restore mutated Atp7a in fibroblasts from a macular mouse model of Menkes disease withs T>C mutation. Around 35% of the mutated C nucleotide (nt) was restored to U, demonstrating that our RNA editing system is reliable and has potential for therapeutic clinical application. RNA base editing via human RNA-guided cytidine deaminases is a potentially attractive approach for in vivo therapeutic application and provides opportunities for new developments in this field.
Collapse
Affiliation(s)
- Sonali Bhakta
- Bioscience, Biotechnology and Biomedical Engineering Research Area, Japan Advanced Institute of Science and Technology, Nomi 923-1211, Japan;
- Department of Anatomy and Histology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Hiroko Kodama
- General Medical Education and Research Center, Teikyo University School of Medicine, Tokyo 173-0003, Japan
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo 173-0003, Japan
| | - Masakazu Mimaki
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo 173-0003, Japan
| | - Toshifumi Tsukahara
- Bioscience, Biotechnology and Biomedical Engineering Research Area, Japan Advanced Institute of Science and Technology, Nomi 923-1211, Japan;
- GeCoRT Co., Ltd., Nishi-ku, Yokohama 220-0011, Japan
| |
Collapse
|
21
|
An M, Davis JR, Levy JM, Serack FE, Harvey JW, Brauer PP, Pirtle CP, Berríos KN, Newby GA, Yeh WH, Kamath N, Mortberg M, Lian Y, Howard M, DeSouza-Lenz K, Guzman K, Thai A, Graffam S, Laversenne V, Coffey AA, Frei J, Pierce SE, Safar JG, Deverman BE, Minikel EV, Vallabh SM, Liu DR. In vivo base editing extends lifespan of a humanized mouse model of prion disease. Nat Med 2025:10.1038/s41591-024-03466-w. [PMID: 39810005 DOI: 10.1038/s41591-024-03466-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
Prion disease is a fatal neurodegenerative disease caused by the misfolding of prion protein (PrP) encoded by the PRNP gene. While there is currently no cure for the disease, depleting PrP in the brain is an established strategy to prevent or stall templated misfolding of PrP. Here we developed in vivo cytosine and adenine base strategies delivered by adeno-associated viruses to permanently modify the PRNP locus to achieve PrP knockdown in the mouse brain. Systemic injection of dual-adeno-associated virus PHP.eB encoding BE3.9max and single guide RNA installing PRNP R37X resulted in 37% average installation of the desired edit, 50% reduction of PrP in the mouse brain and 52% extension of lifespan in transgenic human PRNP mice inoculated with pathogenic human prion isolates representing the most common sporadic and genetic subtypes of prion disease. We further engineered base editing systems to achieve improved in vivo potency and reduced base editor expression in nontargeting tissues, resulting in 63% average PrP reduction in the mouse brain from a 6.7-fold lower viral dose, with no detected off-target editing of anticipated clinical significance observed in either human cells or mouse tissues. These findings support the potential of in vivo base editing as one-time treatment for prion disease.
Collapse
Affiliation(s)
- Meirui An
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jessie R Davis
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jonathan M Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Fiona E Serack
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John W Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pamela P Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine P Pirtle
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiara N Berríos
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Wei-Hsi Yeh
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Nikita Kamath
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Meredith Mortberg
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yuan Lian
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Howard
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Kenia Guzman
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron Thai
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samantha Graffam
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vanessa Laversenne
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alissa A Coffey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeannine Frei
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sarah E Pierce
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jiri G Safar
- Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin E Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Prion Alliance, Cambridge, MA, USA.
| | - Sonia M Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Prion Alliance, Cambridge, MA, USA.
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
22
|
Youssef E, Fletcher B, Palmer D. Enhancing precision in cancer treatment: the role of gene therapy and immune modulation in oncology. Front Med (Lausanne) 2025; 11:1527600. [PMID: 39871848 PMCID: PMC11769984 DOI: 10.3389/fmed.2024.1527600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 01/29/2025] Open
Abstract
Gene therapy has long been a cornerstone in the treatment of rare diseases and genetic disorders, offering targeted solutions to conditions once considered untreatable. As the field advances, its transformative potential is now expanding into oncology, where personalized therapies address the genetic and immune-related complexities of cancer. This review highlights innovative therapeutic strategies, including gene replacement, gene silencing, oncolytic virotherapy, CAR-T cell therapy, and CRISPR-Cas9 gene editing, with a focus on their application in both hematologic malignancies and solid tumors. CRISPR-Cas9, a revolutionary tool in precision medicine, enables precise editing of cancer-driving mutations, enhancing immune responses and disrupting tumor growth mechanisms. Additionally, emerging approaches target ferroptosis-a regulated, iron-dependent form of cell death-offering new possibilities for selectively inducing tumor cell death in resistant cancers. Despite significant breakthroughs, challenges such as tumor heterogeneity, immune evasion, and the immunosuppressive tumor microenvironment (TME) remain. To overcome these barriers, novel approaches like dual-targeting, armored CAR-T cells, and combination therapies with immune checkpoint inhibitors and ferroptosis inducers are being explored. Additionally, the rise of allogeneic "off-the-shelf" CAR-T therapies offers scalable and more accessible treatment options. The regulatory landscape is evolving to accommodate these advancements, with frameworks like RMAT (Regenerative Medicine Advanced Therapy) in the U.S. and ATMP (Advanced Therapy Medicinal Products) in Europe fast-tracking the approval of gene therapies. However, ethical considerations surrounding CRISPR-based gene editing-such as off-target effects, germline editing, and ensuring equitable access-remain at the forefront, requiring ongoing ethical oversight. Advances in non-viral delivery systems, such as lipid nanoparticles (LNPs) and exosomes, are improving the safety and efficacy of gene therapies. By integrating these innovations with combination therapies and addressing regulatory and ethical concerns, gene therapy is poised to revolutionize cancer treatment, providing durable, effective, and personalized solutions for both hematologic and solid tumors.
Collapse
|
23
|
Su-Tobon Q, Fan J, Goldstein M, Feeney K, Ren H, Autissier P, Wang P, Huang Y, Mohanty U, Niu J. CRISPR-Hybrid: A CRISPR-Mediated Intracellular Directed Evolution Platform for RNA Aptamers. Nat Commun 2025; 16:595. [PMID: 39799111 PMCID: PMC11724954 DOI: 10.1038/s41467-025-55957-0] [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: 08/24/2023] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
Recent advances in gene editing and precise regulation of gene expression based on CRISPR technologies have provided powerful tools for the understanding and manipulation of gene functions. Fusing RNA aptamers to the sgRNA of CRISPR can recruit cognate RNA-binding protein (RBP) effectors to target genomic sites, and the expression of sgRNA containing different RNA aptamers permit simultaneous multiplexed and multifunctional gene regulations. Here, we report an intracellular directed evolution platform for RNA aptamers against intracellularly expressed RBPs. We optimize a bacterial CRISPR-hybrid system coupled with FACS, and identified high affinity RNA aptamers orthogonal to existing aptamer-RBP pairs. Application of orthogonal aptamer-RBP pairs in multiplexed CRISPR allows effective simultaneous transcriptional activation and repression of endogenous genes in mammalian cells.
Collapse
Affiliation(s)
- Qiwen Su-Tobon
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Jiayi Fan
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | | | - Kevin Feeney
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Hongyuan Ren
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | | | - Peiyi Wang
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Yingzi Huang
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Udayan Mohanty
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Jia Niu
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA.
| |
Collapse
|
24
|
Hazel K, Singh D, He S, Guertin Z, Husser MC, Helfield B. Focused ultrasound and microbubble-mediated delivery of CRISPR-Cas9 ribonucleoprotein to human induced pluripotent stem cells. Mol Ther 2025:S1525-0016(25)00017-6. [PMID: 39797397 DOI: 10.1016/j.ymthe.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
CRISPR-Cas9 ribonucleoproteins (RNPs) have been heavily considered for gene therapy due to their high on-target efficiency, rapid activity, and lack of insertional mutagenesis relative to other CRISPR-Cas9 delivery formats. Genetic diseases such as hypertrophic cardiomyopathy currently lack effective treatment strategies and are prime targets for CRISPR-Cas9 gene editing technology. However, current in vivo delivery strategies for Cas9 pose risks of unwanted immunogenic responses. This proof-of-concept study aimed to demonstrate that focused ultrasound (FUS) in combination with microbubbles can be used to deliver Cas9-sgRNA (single-guide RNA) RNPs and functionally edit human induced pluripotent stem cells (hiPSCs) in vitro, a model system that can be expanded to cardiovascular research via hiPSC-derived cardiomyocytes. Here, we first determine acoustic conditions suitable for the viable delivery of large proteins to hiPSCs with clinical Definity microbubble agents using our customized experimental platform. From here, we delivered Cas9-sgRNA RNP complexes targeting the EGFP (enhanced green fluorescent protein) gene to EGFP-expressing hiPSCs for EGFP knockout. Simultaneous acoustic cavitation detection during treatment confirmed a strong correlation between microbubble disruption and viable FUS-mediated protein delivery in hiPSCs. This study shows for the first time the potential for an FUS-mediated technique for targeted and precise CRISPR-Cas9 gene editing in human stem cells.
Collapse
Affiliation(s)
- Kyle Hazel
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada
| | - Davindra Singh
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada
| | - Stephanie He
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada
| | - Zakary Guertin
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada
| | - Mathieu C Husser
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada
| | - Brandon Helfield
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada; Department of Physics, Concordia University, 7141 Sherbrooke St. W, H4B 1R6 Montreal, QC, Canada.
| |
Collapse
|
25
|
Wang H, Dong Z, Shi J, Chen L, Sun T, Zhang W. Genome-Wide A → G and C → T Mutations Induced by Functional TadA Variants in Escherichia coli. ACS Synth Biol 2025. [PMID: 39787000 DOI: 10.1021/acssynbio.4c00597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The fusion expression of deoxyribonucleic acid (DNA) replication-related proteins with nucleotide deaminase enzymes promotes random mutations in bacterial genomes, thereby increasing genetic diversity among the population. Most previous studies have focused on cytosine deaminase, which produces only C → T mutations, significantly limiting the variety of mutation types. In this study, we developed a fusion expression system by combining DnaG (RNA primase) with adenine deaminase TadA-8e (DnaG-TadA) in Escherichia coli, which is capable of rapidly introducing A → G mutations into the E. coli genome, resulting in a 664-fold increase in terms of mutation rate. Additionally, we tested a dual-functional TadA variant, TadAD, and then fused it with DnaG. This construct introduced both C → T and A → G mutations into the E. coli genome, with the mutation rate increased by 370-fold upon coexpression with a uracil glycosylase inhibitor (DnaG-TadAD-UGI). We applied DnaG-TadA and DnaG-TadAD-UGI systems to the adaptive laboratory evolution for Cd2+ and kanamycin resistance, achieving an 8.0 mM Cd2+ and 200 μg/mL kanamycin tolerance within just 17 days and 132 h, respectively. Compared to conventional evolution methods, the final tolerance levels were increased by 320 and 266%, respectively. Our work offers a novel strategy for random mutagenesis in E. coli and potentially other prokaryotic species.
Collapse
Affiliation(s)
- Hao Wang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, P. R. China
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin 300072, P. R. China
| | - Zhengxin Dong
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, P. R. China
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin 300072, P. R. China
| | - Jingyi Shi
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, P. R. China
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin 300072, P. R. China
| | - Lei Chen
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, P. R. China
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin 300072, P. R. China
| | - Tao Sun
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, P. R. China
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin 300072, P. R. China
| | - Weiwen Zhang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, P. R. China
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin 300072, P. R. China
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
26
|
Wang Y, Lin RZ, Harris M, Lavayen B, Diwanji N, McCreedy B, Hofmeister R, Getts D. CRISPR-Enabled Autonomous Transposable Element (CREATE) for RNA-based gene editing and delivery. EMBO Rep 2025:10.1038/s44319-024-00364-7. [PMID: 39789389 DOI: 10.1038/s44319-024-00364-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
To address a wide range of genetic diseases, genome editing tools that can achieve targeted delivery of large genes without causing double-strand breaks (DSBs) or requiring DNA templates are necessary. Here, we introduce CRISPR-Enabled Autonomous Transposable Element (CREATE), a genome editing system that combines the programmability and precision of CRISPR/Cas9 with the RNA-mediated gene insertion capabilities of the human LINE-1 (L1) element. CREATE employs a modified L1 mRNA to carry a payload gene, and a Cas9 nickase to facilitate targeted editing by L1-mediated reverse transcription and integration without relying on DSBs or DNA templates. Using this system, we demonstrate programmable insertion of a 1.1 kb gene expression cassette into specific genomic loci of human cell lines and primary T cells. Mechanistic studies reveal that CREATE editing is highly specific with no observed off-target events. Together, these findings establish CREATE as a programmable, RNA-based gene delivery technology with broad therapeutic potential.
Collapse
Affiliation(s)
- Yuxiao Wang
- Myeloid Therapeutics Inc., Cambridge, MA, 02139, USA.
| | - Ruei-Zeng Lin
- Myeloid Therapeutics Inc., Cambridge, MA, 02139, USA
| | - Meghan Harris
- Myeloid Therapeutics Inc., Cambridge, MA, 02139, USA
| | | | - Neha Diwanji
- Myeloid Therapeutics Inc., Cambridge, MA, 02139, USA
| | - Bruce McCreedy
- Myeloid Therapeutics Inc., Cambridge, MA, 02139, USA
- ONK Therapeutics Ltd., Galway, Co. Galway, Ireland
| | | | - Daniel Getts
- Myeloid Therapeutics Inc., Cambridge, MA, 02139, USA.
| |
Collapse
|
27
|
Muller A, Sullivan J, Schwarzer W, Wang M, Park-Windhol C, Hasler PW, Janeschitz-Kriegl L, Duman M, Klingler B, Matsell J, Hostettler SM, Galliker P, Hou Y, Balmer P, Virág T, Barrera LA, Young L, Xu Q, Magda DP, Kilin F, Khadka A, Moreau PH, Fellmann L, Azoulay T, Quinodoz M, Karademir D, Leppert J, Fratzl A, Kosche G, Sharma R, Montford J, Cattaneo M, Croyal M, Cronin T, Picelli S, Grison A, Cowan CS, Kusnyerik Á, Anders P, Renner M, Nagy ZZ, Szabó A, Bharti K, Rivolta C, Scholl HPN, Bryson D, Ciaramella G, Roska B, György B. High-efficiency base editing in the retina in primates and human tissues. Nat Med 2025:10.1038/s41591-024-03422-8. [PMID: 39779923 DOI: 10.1038/s41591-024-03422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/14/2024] [Indexed: 01/11/2025]
Abstract
Stargardt disease is a currently untreatable, inherited neurodegenerative disease that leads to macular degeneration and blindness due to loss-of-function mutations in the ABCA4 gene. We have designed a dual adeno-associated viral vector encoding a split-intein adenine base editor to correct the most common mutation in ABCA4 (c.5882G>A, p.Gly1961Glu). We optimized ABCA4 base editing in human models, including retinal organoids, induced pluripotent stem cell-derived retinal pigment epithelial (RPE) cells, as well as adult human retinal explants and RPE/choroid explants in vitro. The resulting gene therapy vectors achieved high levels of gene correction in mutation-carrying mice and in female nonhuman primates, with average editing of 75% of cones and 87% of RPE cells in vivo, which has the potential to translate to a clinical benefit. No off-target editing was detectable in human retinal explants and RPE/choroid explants. The high editing rates in primates show promise for efficient gene editing in other ocular diseases that are targetable by base editing.
Collapse
Affiliation(s)
- Alissa Muller
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | | | - Wibke Schwarzer
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Mantian Wang
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | | | - Pascal W Hasler
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Lucas Janeschitz-Kriegl
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Mert Duman
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Beryll Klingler
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Jane Matsell
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Simon Manuel Hostettler
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Patricia Galliker
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Yanyan Hou
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Pierre Balmer
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | | | | | | | - Quan Xu
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Dániel Péter Magda
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Ferenc Kilin
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | | | | | - Lyne Fellmann
- SILABE, Université de Strasbourg, Niederhausbergen, France
| | | | - Mathieu Quinodoz
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Duygu Karademir
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Juna Leppert
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Alex Fratzl
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Georg Kosche
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jair Montford
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marco Cattaneo
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Mikaël Croyal
- Nantes Université, CNRS, INSERM, L'institut du thorax, Nantes, France
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Therese Cronin
- Université de Nantes, CHU de Nantes, INSERM UMR 1089, Translational Gene Therapy for Genetic Diseases, Nantes, France
| | - Simone Picelli
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Alice Grison
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Cameron S Cowan
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Ákos Kusnyerik
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Philipp Anders
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Magdalena Renner
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Zoltán Zsolt Nagy
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Arnold Szabó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Hendrik P N Scholl
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- European Vision Institute, Basel, Switzerland
- Medical University of Vienna, Department of Clinical Pharmacology, Vienna, Austria
| | | | | | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
- Department of Ophthalmology, University of Basel, Basel, Switzerland.
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.
| | - Bence György
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
- Department of Ophthalmology, University of Basel, Basel, Switzerland.
| |
Collapse
|
28
|
Yin S, Gao L, Sun X, Zhang M, Gao H, Chen X, Zhang D, Ming X, Yang L, Hu Y, Chen X, Liu M, Zhan X, Guan Y, Wang L, Han L, Zhu P, Li D. Amelioration of metabolic and behavioral defects through base editing in the Pah R408W phenylketonuria mouse model. Mol Ther 2025; 33:119-132. [PMID: 39600089 PMCID: PMC11764323 DOI: 10.1016/j.ymthe.2024.11.032] [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: 05/21/2024] [Revised: 09/16/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Phenylketonuria (PKU) is a liver metabolic disorder mainly caused by a deficiency of the hepatic phenylalanine hydroxylase (PAH) enzyme activity, often leading to severe brain function impairment in patients if untreated or if treatment is delayed. In this study, we utilized dual-AAV8 vectors to deliver a near PAM-less adenine base editor variant, known as ABE8e-SpRY, to treat the PahR408W PKU mouse model carrying a frequent R408W mutation in the Pah gene. Our findings revealed that a single intravenous injection in adult mice and a single intraperitoneal injection in neonatal mice resulted in 19.1%-34.6% A-to-G editing efficiency at the pathogenic mutation site with minimal bystander edits. Furthermore, the dual-AAV8-treated mice exhibited reduced blood Phe levels to below the therapeutic threshold of 360 μmol L-1 and restored weight and fur color to normal levels. Importantly, the brain function of the mice was restored after the treatment, particularly when administered during the neonatal stage, as levels of monoamine neurotransmitters and metabolites in the brain returned to normal and near-normal levels. Our study demonstrated that ABE8e-SpRY-based base editing could effectively correct the point mutation in the PahR408W PKU mouse model, indicating potential clinical applications for PKU and other genetic diseases.
Collapse
Affiliation(s)
- Shuming Yin
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510100, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Liangcai Gao
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaoyue Sun
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mei Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Hongyi Gao
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaoqing Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dan Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xinyu Ming
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lei Yang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yaqiang Hu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xi Chen
- BRL Medicine, Inc., Shanghai 200241, China
| | - Meizhen Liu
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xia Zhan
- Department of Pediatric Endocrinology and Genetics Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yuting Guan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Liren Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lianshu Han
- Department of Pediatric Endocrinology and Genetics Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510100, China; Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong 510080, China.
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
29
|
Shi W, Jin E, Fang L, Sun Y, Fan Z, Zhu J, Liang C, Zhang YP, Zhang YQ, Wang GD, Zhao W. VDGE: a data repository of variation database for gene-edited animals across multiple species. Nucleic Acids Res 2025; 53:D1250-D1260. [PMID: 39470732 PMCID: PMC11701559 DOI: 10.1093/nar/gkae956] [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: 08/19/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024] Open
Abstract
Gene-edited animals are crucial for addressing fundamental questions in biology and medicine and hold promise for practical applications. In light of the rapid advancement of gene editing technologies over the past decade, a dramatically increased number of gene-edited animals have been generated. Genome editing at off-target sites can, however, introduce genomic variations, potentially leading to unintended functional consequences in these animals. So, there is an urgent need to systematically collect and collate these variations in gene-edited animals to aid data mining and integrative in-depth analyses. However, existing databases are currently insufficient to meet this need. Here, we present the Variation Database of Gene-Edited animals (VDGE, https://ngdc.cncb.ac.cn/vdge), the first open-access repository to present genomic variations and annotations in gene-edited animals, with a particular focus on larger animals such as monkeys. At present, VDGE houses 151 on-target mutations from 210 samples, and 115,710 variations identified from 107 gene-edited and wild-type animal trios through unified and standardized analysis and concurrently provides comprehensive annotation details for each variation, thus facilitating the assessment of their functional consequences and promoting mechanistic studies and practical applications for gene-edited animals.
Collapse
Affiliation(s)
- Wenwen Shi
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Enhui Jin
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Lu Fang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Yanling Sun
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Cambridge Street, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Cambridge Street, Houston, TX 77030, USA
| | - Zhuojing Fan
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Junwei Zhu
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Chengzhi Liang
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Ya-Ping Zhang
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Panlong District, Kunming 650201, China
| | - Yong Q Zhang
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- School of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Guo-Dong Wang
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Panlong District, Kunming 650201, China
| | - Wenming Zhao
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| |
Collapse
|
30
|
Liang Z, Wu Y, Deng S, Wei S, Zhang K, Guo Y. Enhanced Genome Editing Activity with Novel Chimeric ScCas9 Variants in Rice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411549. [PMID: 39755933 DOI: 10.1002/advs.202411549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/15/2024] [Indexed: 01/06/2025]
Abstract
The Streptococcus canis Cas9 protein (ScCas9) recognizes the NNG protospacer adjacent motif (PAM), offering a wider range of targets than that offered by the commonly used S. pyogenes Cas9 protein (SpCas9). However, both ScCas9 and its evolved Sc++ variant still exhibit low genome editing efficiency in plants, particularly at the less preferred NTG and NCG PAM targets. In this study, a chimeric SpcRN++ variant is engineered by grafting the recognition (REC) domain of SpCas9 into the Sc++ variant, incorporating the R221K/N394K mutations, and retaining the positively charged loop of S. anginosus Cas9. The SpcRN++ variant exhibits a higher genome editing capacity and wider target range than the Sc++ variant in rice protoplasts and stable transgenic plants. Further evidence indicates that nSpcRN++-based A3A/Y130F and TadA8e exhibit enhanced cytosine and adenine editing efficiency in plants. Finally, herbicide-resistant rice germplasms are produced by targeting the OsACC gene using nSpcRN++-based adenine base editors. These results demonstrate that SpcRN++ is a powerful tool for genome editing in plants, and this integrative protein engineering strategy holds promise for engineering other Cas9 proteins.
Collapse
Affiliation(s)
- Zhen Liang
- School of Life Science, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Yuqing Wu
- School of Life Science, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Shuke Deng
- School of Life Science, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Sha Wei
- School of Life Science, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Kai Zhang
- School of Life Science, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Yingjie Guo
- Research Institute of Big Data Science and Industry, Shanxi University, Taiyuan, Shanxi, 030006, China
| |
Collapse
|
31
|
Xiang T, Feng H, Xing XH, Zhang C. GLiDe: a web-based genome-scale CRISPRi sgRNA design tool for prokaryotes. BMC Bioinformatics 2025; 26:1. [PMID: 39754035 PMCID: PMC11699761 DOI: 10.1186/s12859-024-06012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND CRISPRi screening has become a powerful approach for functional genomic research. However, the off-target effects resulting from the mismatch tolerance between sgRNAs and their intended targets is a primary concern in CRISPRi applications. RESULTS We introduce Guide Library Designer (GLiDe), a web-based tool specifically created for the genome-scale design of sgRNA libraries tailored for CRISPRi screening in prokaryotic organisms. GLiDe incorporates a robust quality control framework, rooted in prior experimental knowledge, ensuring the accurate identification of off-target hits. It boasts an extensive built-in database, encompassing 1,397 common prokaryotic species as a comprehensive design resource. It also provides the capability to design sgRNAs for newly discovered organisms by accepting uploaded design resource. We further demonstrated that GLiDe exhibits enhanced precision in identifying off-target binding sites for the CRISPRi system. CONCLUSIONS We present a web server that allows the construction of genome-scale CRISPRi sgRNA libraries for prokaryotes. It mitigates off-target effects through a robust quality control framework, leveraging prior experimental knowledge within an end-to-end, user-friendly pipeline.
Collapse
Affiliation(s)
- Tongjun Xiang
- MOE Key Laboratory for Industrial Biocatalysis, Institute of Biochemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Huibao Feng
- MOE Key Laboratory for Industrial Biocatalysis, Institute of Biochemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Xin-Hui Xing
- MOE Key Laboratory for Industrial Biocatalysis, Institute of Biochemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, China
| | - Chong Zhang
- MOE Key Laboratory for Industrial Biocatalysis, Institute of Biochemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
32
|
Dewars ER, Landstrom AP. The Genetic Basis of Sudden Cardiac Death: From Diagnosis to Emerging Genetic Therapies. Annu Rev Med 2025; 76:283-299. [PMID: 39499917 DOI: 10.1146/annurev-med-042423-042903] [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] [Indexed: 01/28/2025]
Abstract
Sudden cardiac death (SCD) is an abrupt, tragic manifestation of a number of cardiovascular diseases, primarily ion channelopathies and heritable cardiomyopathies. Because these diseases are heritable, genetics play a key role in the diagnosis and management of SCD-predisposing diseases. Historically, genetics have been used to confirm a diagnosis and identify at-risk family members, but a deeper understanding of the genetic causes of SCD could pave the way for individualized therapy, early risk detection, and a transformative shift toward genetically informed therapies. This review focuses on the evolving genetic landscape of SCD-predisposing diseases, the current state of gene therapy and therapeutic development, and the promise of using predictive genetics to identify individuals at risk of SCD.
Collapse
Affiliation(s)
- Enya R Dewars
- Developmental and Stem Cell Biology Program and Cell and Molecular Biology Program, Duke University School of Medicine, Durham, North Carolina, USA
- Division of Pediatric Cardiology, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA;
| | - Andrew P Landstrom
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
- Division of Pediatric Cardiology, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA;
| |
Collapse
|
33
|
Smith DJ, Lunj S, Adamson AD, Nagarajan S, Smith TAD, Reeves KJ, Hoskin PJ, Choudhury A. CRISPR-Cas9 potential for identifying novel therapeutic targets in muscle-invasive bladder cancer. Nat Rev Urol 2025; 22:55-65. [PMID: 38951705 DOI: 10.1038/s41585-024-00901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2024] [Indexed: 07/03/2024]
Abstract
Gene editing technologies help identify the genetic perturbations driving tumour initiation, growth, metastasis and resistance to therapeutics. This wealth of information highlights tumour complexity and is driving cancer research towards precision medicine approaches based on an individual's tumour genetics. Bladder cancer is the 11th most common cancer in the UK, with high rates of relapse and low survival rates in patients with muscle-invasive bladder cancer (MIBC). MIBC is highly heterogeneous and encompasses multiple molecular subtypes, each with different responses to therapeutics. This evidence highlights the need to identify innovative therapeutic targets to address the challenges posed by this heterogeneity. CRISPR-Cas9 technologies have been used to advance our understanding of MIBC and determine novel drug targets through the identification of drug resistance mechanisms, targetable cell-cycle regulators, and novel tumour suppressor and oncogenes. However, the use of these technologies in the clinic remains a substantial challenge and will require careful consideration of dosage, safety and ethics. CRISPR-Cas9 offers considerable potential for revolutionizing bladder cancer therapies, but substantial research is required for validation before these technologies can be used in the clinical setting.
Collapse
Affiliation(s)
- Danielle J Smith
- Division of Cancer Sciences, University of Manchester, Manchester, UK.
| | - Sapna Lunj
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Antony D Adamson
- Faculty of Biology, Medicine and Health Research and Innovation, University of Manchester, Manchester, UK
| | - Sankari Nagarajan
- Division of Molecular and Cellular Function, University of Manchester, Manchester, UK
| | - Tim A D Smith
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Nuclear Futures Institute, Bangor University, Bangor, UK
| | | | - Peter J Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
34
|
Cirincione A, Simpson D, Yan W, McNulty R, Ravisankar P, Solley SC, Yan J, Lim F, Farley EK, Singh M, Adamson B. A benchmarked, high-efficiency prime editing platform for multiplexed dropout screening. Nat Methods 2025; 22:92-101. [PMID: 39562753 PMCID: PMC11725502 DOI: 10.1038/s41592-024-02502-4] [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: 03/12/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024]
Abstract
Prime editing installs precise edits into the genome with minimal unwanted byproducts, but low and variable editing efficiencies have complicated application of the approach to high-throughput functional genomics. Here we assembled a prime editing platform capable of high-efficiency substitution editing suitable for functional interrogation of small genetic variants. We benchmarked this platform for pooled, loss-of-function screening using a library of ~240,000 engineered prime editing guide RNAs (epegRNAs) targeting ~17,000 codons with 1-3 bp substitutions. Comparing the abundance of these epegRNAs across screen samples identified negative selection phenotypes for 7,996 nonsense mutations targeted to 1,149 essential genes and for synonymous mutations that disrupted splice site motifs at 3' exon boundaries. Rigorous evaluation of codon-matched controls demonstrated that these phenotypes were highly specific to the intended edit. Altogether, we established a prime editing approach for multiplexed, functional characterization of genetic variants with simple readouts.
Collapse
Affiliation(s)
- Ann Cirincione
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Danny Simpson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Weihao Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Ryan McNulty
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Purnima Ravisankar
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sabrina C Solley
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jun Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Fabian Lim
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Emma K Farley
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Mona Singh
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Britt Adamson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
35
|
György B. A base editor for correcting deafness. Nat Biomed Eng 2025; 9:3-4. [PMID: 39134682 DOI: 10.1038/s41551-024-01234-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Affiliation(s)
- Bence György
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
- Department of Ophthalmology, University of Basel, Basel, Switzerland.
| |
Collapse
|
36
|
Cui C, Wang S, Wang D, Zhao J, Huang B, Zhu B, Chen Y, Tang H, Han Y, Ye C, Mu D, Zhang C, Yang Y, Bao Y, Lv J, Han S, Li GL, Li H, Shu Y. A base editor for the long-term restoration of auditory function in mice with recessive profound deafness. Nat Biomed Eng 2025; 9:40-56. [PMID: 39134683 DOI: 10.1038/s41551-024-01235-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/18/2024] [Indexed: 01/24/2025]
Abstract
A prevalent recessive mutation (c.2485C>T, p.Q829X) within the OTOF gene leads to profound prelingual hearing loss. Here we show that in Otof mice harbouring a mutation (c.2482C>T, p.Q828X) homozygous to human OTOF that faithfully mimics the hearing-loss phenotype, a base editor (consisting of the deaminase ABE7.10max and the Cas9 variant SpCas9-NG) packaged in adeno-associated viruses and injected into the inner ear of the mice via the round-window membrane effectively corrected the pathogenic mutation, with no apparent off-target effects. The treatment restored the levels of the otoferlin protein in 88% of the inner hair cells and stably rescued the auditory function of the mice to near-wild-type levels for over 1.5 years while improving synaptic exocytosis in the inner hair cells. We also show that an adenine base editor that targets the prevalent human OTOF mutation restored hearing in humanized mice to levels comparable to those of the wild-type counterparts. Base editors may be effective for the treatment of hereditary deafness.
Collapse
Affiliation(s)
- Chong Cui
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Institutes of Biomedical Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shengyi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, Finland
| | - Daqi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Jingjing Zhao
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Bowei Huang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Institutes of Biomedical Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Biyun Zhu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Yuxin Chen
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Honghai Tang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Yu Han
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Cheng Ye
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Institutes of Biomedical Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Dan Mu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Chengdong Zhang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Yang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihan Bao
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- Department of Otolaryngology-Head and Neck Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Lv
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Institutes of Biomedical Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shuang Han
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Geng-Lin Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Huawei Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Institutes of Biomedical Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China.
- Institutes of Biomedical Science, Fudan University, Shanghai, China.
- NHC Key Laboratory of Hearing Medicine, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Li X, Wei Y, Wang SY, Wang SG, Xia PF. One-for-all gene inactivation via PAM-independent base editing in bacteria. J Biol Chem 2025; 301:108113. [PMID: 39706269 PMCID: PMC11782819 DOI: 10.1016/j.jbc.2024.108113] [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: 11/06/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
Base editing is preferable for bacterial gene inactivation without generating double-strand breaks, requiring homology recombination, or highly efficient DNA delivery capability. However, the potential of base editing is limited by the adjoined dependence on the editing window and protospacer adjacent motif. Herein, we report an unconstrained base-editing system to enable the inactivation of any genes of interest in bacteria. We employed a dCas9 derivative, dSpRY, and activation-induced cytidine deaminase to build a protospacer adjacent motif-independent base editor. Then, we programmed the base editor to exclude the START codon of a gene of interest instead of introducing premature STOP codons to obtain a universal approach for gene inactivation, namely XSTART, with an overall efficiency approaching 100%. By using XSTART, we successfully manipulated the amino acid metabolisms in Escherichia coli, generating glutamine, arginine, and aspartate auxotrophic strains. While we observed a high frequency of off-target events as a trade-off for increased efficiency, refining the regulatory system of XSTART to limit expression levels reduced off-target events by over 60% without sacrificing efficiency, aligning our results with previously reported levels. Finally, the effectiveness of XSTART was also demonstrated in probiotic E. coli Nissle 1917 and photoautotrophic cyanobacterium Synechococcus elongatus, illustrating its potential in reprogramming diverse bacteria.
Collapse
Affiliation(s)
- Xin Li
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| | - Ying Wei
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| | - Shu-Yan Wang
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| | - Shu-Guang Wang
- School of Environmental Science and Engineering, Shandong University, Qingdao, China; Sino-French Research Institute for Ecology and Environment, Shandong University, Qingdao, China; Weihai Research Institute of Industrial Technology, Shandong University, Weihai, China
| | - Peng-Fei Xia
- School of Environmental Science and Engineering, Shandong University, Qingdao, China.
| |
Collapse
|
38
|
He K, Xue Q, Zhou W, Wang P, Hu X, Lin T, Chen N, Wang B, Ma T, Ding S. Extended pegRNAs enhance the editing capability of Prime editing. Trends Biotechnol 2025; 43:206-219. [PMID: 39341743 DOI: 10.1016/j.tibtech.2024.09.004] [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: 03/22/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
Genome editing is highly valuable in biomedical research. Despite their versatility, current Prime editing (PE) techniques are limited to short sequence alterations [up to ~44 base pairs (bp)], and exhibit inconsistent or low efficiency across genomic loci, particularly when faced with poly-T sequences. To address these challenges, we developed an extended PE (exPE) technique that can potentially execute any precise genome editing. By harnessing RNA polymerase II (Pol II) promoters to transcribe extended PE guide RNAs (expegRNAs), exPE substantially improves editing efficiency and overcomes the challenges posed by poly-T sequences. Compared with conventional PE, exPE achieves up to a 14-fold increase in the efficiency of base conversions and short insertions, and, remarkably, up to a 259-fold improvement in regions with poly-T sequences. Uniquely, exPE enables seamless insertion of gene-sized DNA fragments into genomes, potentially correcting nearly 90% of human genetic variants, thereby broadening its applications in genetic research and therapy.
Collapse
Affiliation(s)
- Kezhang He
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Qiaomei Xue
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Zhou
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Pengqi Wang
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaodan Hu
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Tongtong Lin
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Nan Chen
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Bowen Wang
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Tianhua Ma
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| | - Sheng Ding
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
39
|
Endo M, Negishi K, Toki S. Precise Base Substitution Using CRISPR/Cas-Mediated Base Editor in Rice. Methods Mol Biol 2025; 2869:101-111. [PMID: 39499471 DOI: 10.1007/978-1-0716-4204-7_11] [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] [Indexed: 11/07/2024]
Abstract
Base editors, CRISPR/Cas-based precise genome editing tools, enable base conversion at a target site without inducing DNA double-strand breaks. The genome editing targetable range is restricted by the requirement for protospacer adjacent motif (PAM) sequence. Cas9 derived from Streptococcus pyogenes (SpCas9)-most widely used for genome editing in many organisms-requires an NGG sequence adjacent to the target site as a PAM. Then, engineered and natural Cas variants with altered PAM recognition are used for base editor to expand the flexibility of base substitution position. In this chapter, we describe a protocol for base editing based on SpCas9-NG, which is a rationally engineered SpCas9 variant that can recognize relaxed NG PAMs.
Collapse
Affiliation(s)
- Masaki Endo
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan.
| | - Katsuya Negishi
- Institute of Fruit Tree and Tea Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Seiichi Toki
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
- Kihara Institute for Biological Research, Yokohama City University, Yokohama, Japan
- Faculty of Agriculture, Department of Life Science, Ryukoku University, Kyoto, Japan
| |
Collapse
|
40
|
Corydon TJ, Bek T. Multiple gene therapy as a tool for regulating the expression of molecules involved in neovascular age-related macular degeneration. Prog Retin Eye Res 2025; 104:101323. [PMID: 39672501 DOI: 10.1016/j.preteyeres.2024.101323] [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: 08/27/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Anti-vascular endothelial growth factor (VEGF) therapies have revolutionized the treatment of neovascular age-related macular degeneration (nAMD) and other retinal diseases. However, the necessity for repeated intravitreal injections and the observation of variable treatment responses calls for new treatment modalities where fewer and more effective interventions can result in a clinical effect. Gene therapy might be such an alternative, and therefore the development and clinical application of gene therapy aimed at modifying gene expression has received considerable attention. The article reviews current knowledge of the background, pathophysiological mechanisms, technologies, limitations, and future directions for gene therapy aimed at modifying the synthesis of compounds involved in acquired and senescent retinal disease. The authors have contributed to the field by developing gene therapy to reduce the expression of vascular endothelial growth factor (VEGF), as well as multiple gene therapy for simultaneous downregulation of the synthesis of VEGF and upregulation of pigment epithelium-derived factor (PEDF) using adeno-associated virus (AAV) vectors. It is suggested that such multi-target gene therapy might be included in future treatments of retinal diseases where the underlying mechanisms are complex and cannot be attributed to one specific mediator. Such diseases might include dry AMD (dAMD) with geographic atrophy, but also diabetic macular edema (DME) and retinal vein occlusion (RVO). Gene therapy can be expected to be most beneficial for the patients in need of multiple intra-vitreal injections and in whom the therapeutic response is insufficient. It is concluded, that in parallel with basic research, there is a need for clinical studies aimed at identifying factors that can be used to identify patients who will benefit from gene therapy already at the time of diagnosis of the retinal disease.
Collapse
Affiliation(s)
- Thomas J Corydon
- Department of Biomedicine, Hoegh Guldbergs Gade 10, Aarhus University, 8000, Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
41
|
Feng S, Li J, Yan A, Zhu X, Zhang L, Tang D, Liu L. Application of Gene Editing in Triple-Negative Breast Cancer Research. Cell Biochem Funct 2025; 43:e70044. [PMID: 39844394 DOI: 10.1002/cbf.70044] [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: 10/17/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
With the rapid development of gene editing technology, its application in breast cancer has gradually become the focus of research. This article reviews the application of gene editing technology in the treatment of breast cancer, and discusses its challenges and future development directions. The key application areas of gene editing technology in the treatment of breast cancer will be outlined, including the discovery of new therapeutic targets and the development of drugs related to the pathway. Gene editing technology has played an important role in the discovery of new therapeutic targets. Through the use of gene editing technology, breast cancer-related genes are systematically edited to regulate key regulatory factors on related pathways or key tumor suppressor genes such as FOXC1 and BRCA, and the results are analyzed in cell or animal experiments, and the target is obtained from the experimental results, which provides important clues for the development of new drugs. This approach provides an innovative way to find more effective treatment strategies and inhibit tumor growth. In addition, gene editing technology has also promoted the personalization of breast cancer treatment. By analyzing a patient's genomic information, researchers can pinpoint key genetic mutations in a patient's tumor and design personalized treatments. This personalized treatment approach is expected to improve the therapeutic effect and reduce adverse reactions. Finally, the application of gene editing technology also provides support for the development of breast cancer immunotherapy. By editing immune cells to make them more potent against tumors, researchers are trying to develop more effective immunotherapies to bring new treatment options to breast cancer patients.
Collapse
Affiliation(s)
- Shuying Feng
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jixia Li
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Aifen Yan
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xiangxing Zhu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Ligang Zhang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Dongsheng Tang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Lian Liu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| |
Collapse
|
42
|
Ngoi NYL, Gallo D, Torrado C, Nardo M, Durocher D, Yap TA. Synthetic lethal strategies for the development of cancer therapeutics. Nat Rev Clin Oncol 2025; 22:46-64. [PMID: 39627502 DOI: 10.1038/s41571-024-00966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Synthetic lethality is a genetic phenomenon whereby the simultaneous presence of two different genetic alterations impairs cellular viability. Importantly, targeting synthetic lethal interactions offers potential therapeutic strategies for cancers with alterations in pathways that might otherwise be considered undruggable. High-throughput screening methods based on modern CRISPR-Cas9 technologies have emerged and become crucial for identifying novel synthetic lethal interactions with the potential for translation into biologically rational cancer therapeutic strategies as well as associated predictive biomarkers of response capable of guiding patient selection. Spurred by the clinical success of PARP inhibitors in patients with BRCA-mutant cancers, novel agents targeting multiple synthetic lethal interactions within DNA damage response pathways are in clinical development, and rational strategies targeting synthetic lethal interactions spanning alterations in epigenetic, metabolic and proliferative pathways have also emerged and are in late preclinical and/or early clinical testing. In this Review, we provide a comprehensive overview of established and emerging technologies for synthetic lethal drug discovery and development and discuss promising therapeutic strategies targeting such interactions.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Gallo
- Repare Therapeutics, Inc., Montreal, Quebec, Canada
| | - Carlos Torrado
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirella Nardo
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
43
|
Hołubowicz R, Du SW, Felgner J, Smidak R, Choi EH, Palczewska G, Menezes CR, Dong Z, Gao F, Medani O, Yan AL, Hołubowicz MW, Chen PZ, Bassetto M, Risaliti E, Salom D, Workman JN, Kiser PD, Foik AT, Lyon DC, Newby GA, Liu DR, Felgner PL, Palczewski K. Safer and efficient base editing and prime editing via ribonucleoproteins delivered through optimized lipid-nanoparticle formulations. Nat Biomed Eng 2025; 9:57-78. [PMID: 39609561 PMCID: PMC11754100 DOI: 10.1038/s41551-024-01296-2] [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: 12/19/2023] [Accepted: 10/25/2024] [Indexed: 11/30/2024]
Abstract
Delivering ribonucleoproteins (RNPs) for in vivo genome editing is safer than using viruses encoding for Cas9 and its respective guide RNA. However, transient RNP activity does not typically lead to optimal editing outcomes. Here we show that the efficiency of delivering RNPs can be enhanced by cell-penetrating peptides (covalently fused to the protein or as excipients) and that lipid nanoparticles (LNPs) encapsulating RNPs can be optimized for enhanced RNP stability, delivery efficiency and editing potency. Specifically, after screening for suitable ionizable cationic lipids and by optimizing the concentration of the synthetic lipid DMG-PEG 2000, we show that the encapsulation, via microfluidic mixing, of adenine base editor and prime editor RNPs within LNPs using the ionizable lipid SM102 can result in in vivo editing-efficiency enhancements larger than 300-fold (with respect to the delivery of the naked RNP) without detectable off-target edits. We believe that chemically defined LNP formulations optimized for RNP-encapsulation stability and delivery efficiency will lead to safer genome editing.
Collapse
Grants
- F30 EY033642 NEI NIH HHS
- FENG.02.01-IP.05-T005/23 Fundacja na rzecz Nauki Polskiej (Foundation for Polish Science)
- R01 EY032948 NEI NIH HHS
- R01EY032948, R21NS113264 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- RM1 HG009490 NHGRI NIH HHS
- R00 HL163805 NHLBI NIH HHS
- R21 NS113264 NINDS NIH HHS
- R01 EY030873 NEI NIH HHS
- U01 AI142756 NIAID NIH HHS
- UG3AI150551, U01AI142756, R35GM118062, RM1HG009490 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 EY034501 NEI NIH HHS
- N66001-21-C-4013 United States Department of Defense | Defense Threat Reduction Agency (DTRA)
- T32GM008620, F30EY033642 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- T32GM148383 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- P30EY034070 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- I01BX004939 U.S. Department of Veterans Affairs (Department of Veterans Affairs)
- UG3 AI150551 NIAID NIH HHS
- 75N93022C00054 NIAID NIH HHS
- R01EY009339, R01EY030873, P30EY034070, P30CA062203 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 EY009339 NEI NIH HHS
- P30 EY034070 NEI NIH HHS
- T32 GM008620 NIGMS NIH HHS
- R00HL163805 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- I01 BX004939 BLRD VA
- R35 GM118062 NIGMS NIH HHS
- T32 GM148383 NIGMS NIH HHS
- P30 CA062203 NCI NIH HHS
- 2022/47/B/NZ5/03023, 2020/39/D/NZ4/01881, 2019/34/E/NZ5/00434 Narodowe Centrum Nauki (National Science Centre)
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- Knights Templar Eye Foundation (Knights Templar Eye Foundation, Inc.)
- Howard Hughes Medical Institute (HHMI)
Collapse
Affiliation(s)
- Rafał Hołubowicz
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Samuel W Du
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Jiin Felgner
- Adeline Yen Mah Vaccine Center, Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Roman Smidak
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Elliot H Choi
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Grazyna Palczewska
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Carolline Rodrigues Menezes
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Zhiqian Dong
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Fangyuan Gao
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Omar Medani
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Alexander L Yan
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Program in Neuroscience, Amherst College, Amherst, MA, USA
| | - Maria W Hołubowicz
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Paul Z Chen
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marco Bassetto
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
- Research Service, Tibor Rubin VA Long Beach Medical Center, Long Beach, CA, USA
| | - Eleonora Risaliti
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - David Salom
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - J Noah Workman
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Philip D Kiser
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
- Research Service, Tibor Rubin VA Long Beach Medical Center, Long Beach, CA, USA
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Andrzej T Foik
- International Centre for Translational Eye Research (ICTER), Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - David C Lyon
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| | - Philip L Felgner
- Adeline Yen Mah Vaccine Center, Department of Physiology and Biophysics, University of California, Irvine, CA, USA.
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, USA.
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
- Department of Chemistry, University of California, Irvine, CA, USA.
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
44
|
Xiao Q, Li G, Han D, Wang H, Yao M, Ma T, Zhou J, Zhang Y, Zhang X, He B, Yuan Y, Shi L, Li T, Yang H, Huang J, Zhang H. Engineered IscB-ωRNA system with expanded target range for base editing. Nat Chem Biol 2025; 21:100-108. [PMID: 39147927 PMCID: PMC11666462 DOI: 10.1038/s41589-024-01706-1] [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: 12/02/2023] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
As the evolutionary ancestor of Cas9 nuclease, IscB proteins serve as compact RNA-guided DNA endonucleases and nickases, making them strong candidates for base editing. Nevertheless, the narrow targeting scope limits the application of IscB systems; thus, it is necessary to find more IscBs that recognize different target-adjacent motifs (TAMs). Here, we identified 10 of 19 uncharacterized IscB proteins from uncultured microbes with activity in mammalian cells. Through protein and ωRNA engineering, we further enhanced the activity of IscB ortholog IscB.m16 and expanded its TAM scope from MRNRAA to NNNGNA, resulting in a variant named IscB.m16*. By fusing the deaminase domains with IscB.m16* nickase, we generated IscB.m16*-derived base editors that exhibited robust base-editing efficiency in mammalian cells and effectively restored Duchenne muscular dystrophy proteins in diseased mice through single adeno-associated virus delivery. Thus, this study establishes a set of compact base-editing tools for basic research and therapeutic applications.
Collapse
Affiliation(s)
- Qingquan Xiao
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai, China
| | - Guoling Li
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
| | - Dingyi Han
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Mingyu Yao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, China
| | - Tingting Ma
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
| | | | - Yu Zhang
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
| | - Xiumei Zhang
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
| | - Bingbing He
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Yuan
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
| | - Linyu Shi
- HuidaGene Therapeutics Co. Ltd., Shanghai, China
| | - Tong Li
- HuidaGene Therapeutics Co. Ltd., Shanghai, China.
| | - Hui Yang
- HuidaGene Therapeutics Co. Ltd., Shanghai, China.
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai, China.
| | - Jinhai Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.
- NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, China.
| | - Hainan Zhang
- HuidaGene Therapeutics Co. Ltd., Shanghai, China.
| |
Collapse
|
45
|
Singh N, Kizhatil K, Duraikannu D, Choquet H, Saidas Nair K. Structural framework to address variant-gene relationship in primary open-angle glaucoma. Vision Res 2025; 226:108505. [PMID: 39520803 DOI: 10.1016/j.visres.2024.108505] [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: 06/17/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Primary open-angle glaucoma (POAG) is a complex, multifactorial disease leading to progressive optic neuropathy and irreversible vision loss. Genome-Wide Association Studies (GWAS) have significantly advanced our understanding of the genetic loci associated with POAG. Expanding on these findings, Exome-Wide Association Studies (ExWAS) refine the genetic landscape by identifying rare coding variants with potential functional relevance. Post-GWAS in silico analyses, including fine-mapping, gene-based association testing, and pathway analysis, offer insights into target genes and biological mechanisms underlying POAG. This review aims to provide a comprehensive roadmap for the post-GWAS characterization of POAG genes. We integrate current knowledge from GWAS, ExWAS, and post-GWAS analyses, highlighting key genetic variants and pathways implicated in POAG. Recent advancements in genomics, such as ATAC-seq, CUT&RUN, and Hi-C, are crucial for identifying disease-relevant gene regulatory elements by profiling chromatin accessibility, histone modifications, and three-dimensional chromatin architecture. These approaches help pinpoint regulatory elements that influence gene expression in POAG. Expression Quantitative Trait Loci (eQTL) analysis and Transcriptome-Wide Association Studies (TWAS) elucidate the impact of these elements on gene expression and disease risk, while functional validations like enhancer reporter assays confirm their relevance. The integration of high-resolution genomics with functional assays and the characterization of genes in vivo using animal models provides a robust framework for unraveling the complex genetic architecture of POAG. This roadmap is essential for advancing our understanding and identification of genes and regulatory networks involved in POAG pathogenesis.
Collapse
Affiliation(s)
- Nivedita Singh
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA.
| | - Krishnakumar Kizhatil
- Department of Ophthalmology and Visual Sciences, The Ohio State University Medical Center, Columbus, OH 43210, USA.
| | - Durairaj Duraikannu
- Departments of Ophthalmology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Hélène Choquet
- Kaiser Permanente, Division of Research, Pleasanton, CA 94588, USA; Department of Health Systems Science Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA 91101, USA.
| | - K Saidas Nair
- Departments of Ophthalmology and Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
46
|
Thangaraj A, Kaul R, Sharda S, Kaul T. Revolutionizing cotton cultivation: A comprehensive review of genome editing technologies and their impact on breeding and production. Biochem Biophys Res Commun 2025; 742:151084. [PMID: 39637703 DOI: 10.1016/j.bbrc.2024.151084] [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: 07/28/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Cotton (Gossypium hirsutum L.), a vital global cash crop, significantly impacts both the agricultural and industrial sectors, providing essential fiber for textiles and valuable byproducts such as cottonseed oil and animal feed. The cultivation of cotton supports millions of livelihoods worldwide, particularly in developing regions, making it a cornerstone of rural economies. Despite its importance, cotton production faces numerous challenges, including biotic stresses from pests and diseases, and abiotic stresses like drought, salinity, and extreme temperatures. These challenges necessitate innovative solutions to ensure sustainable production. Genome editing technologies, particularly CRISPR/Cas9, have revolutionized cotton breeding by enabling precise genetic modifications. These advancements hold promise for developing cotton varieties with enhanced resistance to pests, diseases, and environmental stresses. Early genome editing tools like ZFNs and TALENs paved the way for more precise modifications but were limited by complexity and cost. The introduction of CRISPR/Cas-based technology with its simplicity and efficiency, has dramatically transformed the field, making it the preferred tool for genome editing in crops. Improved version of the technology like CRISPR/Cas12a, CRISPR/Cas13, base and prime editing, developed from CRISPR/Cas systems, provide additional tools with distinct mechanisms, further expanding their potential applications in crop improvement. This comprehensive review explores the impact of genome editing on cotton breeding and production. It discusses the technical challenges, including off-target effects and delivery methods for genome editing components, and highlights ongoing research efforts to overcome these hurdles. The review underscores the potential of genome editing technologies to revolutionize cotton cultivation, enhancing yield, quality, and resilience, ultimately contributing to a sustainable future for the cotton industry.
Collapse
Affiliation(s)
- Arulprakash Thangaraj
- Nutritional Improvement of Crops Group, Plant Molecular Biology Division, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India; Centre for Cellular & Molecular Biology, Amity Institute of Biotechnology, Amity University, Noida, UP, India
| | - Rashmi Kaul
- Nutritional Improvement of Crops Group, Plant Molecular Biology Division, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India; Centre for Cellular & Molecular Biology, Amity Institute of Biotechnology, Amity University, Noida, UP, India
| | - Shivani Sharda
- Centre for Cellular & Molecular Biology, Amity Institute of Biotechnology, Amity University, Noida, UP, India
| | - Tanushri Kaul
- Nutritional Improvement of Crops Group, Plant Molecular Biology Division, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India.
| |
Collapse
|
47
|
Tsuji‐Hosokawa A, Tsuchiya I, Shimizu K, Terao M, Yasuhara M, Miyamoto N, Kikuchi S, Ogawa Y, Nakamura K, Matsubara Y, Takada S. Genetically humanized phenylketonuria mouse model as a testing tool for human genome editing in fertilized eggs. J Inherit Metab Dis 2025; 48:e12803. [PMID: 39380247 PMCID: PMC11729594 DOI: 10.1002/jimd.12803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Targeted genome editing has made significant advancements; however, safety and ethical issues have not been fully elucidated, resulting in strict control of the technique. We tested genome editing tools on gametes from a genetically humanized mouse model using a phenylketonuria (PKU) mouse model to gain insights into genome editing in human embryos. The human PKU mouse model PahhR111X mice was generated. The junctional region between exon 3 and intron 3 of Pah was replaced with a 120 bp corresponding human PAH sequence, including the pathogenic common variant c.331C > T in PahhR111X mice. PahhR111X mice successfully recapitulated the PKU phenotype and showed cognitive dysfunction and depressive-like behavior, which are observed in human patients with PKU. Genome editing was applied to fertilized eggs of PahhR111X mice utilizing sgRNA that targets the human sequence. Mice with the corrected allele exhibited normal serum phenylalanine levels. Through genome editing, we validated the utility of sgRNA. The genetically humanized mouse model suggested that germ-line genome editing of the pathogenic variant may be feasible for monogenic disorders by revealing the recovery of the phenotype; however, there are remaining issues with the tool, including its efficiency and accuracy. This genome editing protocol using a genetically humanized mouse model will provide insights for improving current issues and contribute to the establishment of heritable human genome editing protocols.
Collapse
Affiliation(s)
- Atsumi Tsuji‐Hosokawa
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Iku Tsuchiya
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of NCCHD, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Kie Shimizu
- Department of PharmacologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Miho Terao
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Mito Yasuhara
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of NCCHD, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Natsuho Miyamoto
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Saki Kikuchi
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Yuya Ogawa
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of NCCHD, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Kazuaki Nakamura
- Department of PharmacologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Yoichi Matsubara
- National Center for Child Health and DevelopmentSetagayaTokyoJapan
| | - Shuji Takada
- Department of Systems BioMedicineNational Research Institute for Child Health and DevelopmentTokyoJapan
| |
Collapse
|
48
|
Zhong Z, Hu X, Zhang R, Liu X, Chen W, Zhang S, Sun J, Zhong TP. Improving precision base editing of the zebrafish genome by Rad51DBD-incorporated single-base editors. J Genet Genomics 2025; 52:105-115. [PMID: 39428086 DOI: 10.1016/j.jgg.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024]
Abstract
Single-base editors, including cytosine base editors (CBEs) and adenine base editors (ABEs), facilitate accurate C⋅G to T⋅A and A⋅T to G⋅C, respectively, holding promise for the precise modeling and treatment of human hereditary disorders. Efficient base editing and expanded base conversion range have been achieved in human cells through base editors fusing with Rad51 DNA binding domain (Rad51DBD), such as hyA3A-BE4max. Here, we show that hyA3A-BE4max catalyzes C-to-T substitution in the zebrafish genome and extends editing positions (C12-C16) proximal to the protospacer adjacent motif. We develop a codon-optimized counterpart zhyA3A-CBE5, which exhibits substantially high C-to-T conversion with 1.59- to 3.50-fold improvement compared with the original hyA3A-BE4max. With these tools, disease-relevant hereditary mutations can be more efficaciously generated in zebrafish. We introduce human genetic mutation rpl11Q42∗ and abcc6aR1463C by zhyA3A-CBE5 in zebrafish, mirroring Diamond-Blackfan anemia and Pseudoxanthoma Elasticum, respectively. Our study expands the base editing platform targeting the zebrafish genomic landscape and the application of single-base editors for disease modeling and gene function study.
Collapse
Affiliation(s)
- Zhilin Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xueli Hu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Renjie Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xu Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wenqi Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shubin Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jianjian Sun
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong 510100, China.
| | - Tao P Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
49
|
Niu K, Zheng R, Zhang M, Chen MQ, Kong YM, Liu ZQ, Zheng YG. Adjustment of the main biosynthesis modules to enhance the production of l-homoserine in Escherichia coli W3110. Biotechnol Bioeng 2025; 122:223-232. [PMID: 39425492 DOI: 10.1002/bit.28861] [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: 07/13/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
l-homoserine is an important platform compound of many valuable products. Construction of microbial cell factory for l-homoserine production from glucose has attracted a great deal of attention. In this study, l-homoserine biosynthesis pathway was divided into three modules, the glucose uptake and upstream pathway, the downstream pathway, and the energy supply module. Metabolomics of the chassis strain HS indicated that the supply of ATP was inadequate, therefore, the energy supply module was firstly modified. By balancing the ATP supply module, the l-homoserine production increased by 66% to 12.55 g/L. Further, the results indicated that the upstream pathway was blocked, and increasing the culture temperature to 37°C could solve this problem and the l-homoserine production reached 21.38 g/L. Then, the downstream synthesis pathways were further strengthened to balance the fluxes, and the l-homoserine production reached the highest reported level of 32.55 g/L in shake flasks. Finally, fed-batch fermentation in a 5-L bioreactor was conducted, and l-homoserine production could reach to 119.96 g/L after 92 h cultivation, with the yield of 0.41 g/g glucose and productivity of 1.31 g/L/h. The study provides a well research foundation for l-homoserine production by microbial fermentation with the capacity for industrial application.
Collapse
Affiliation(s)
- Kun Niu
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Rui Zheng
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Miao Zhang
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Mao-Qin Chen
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yi-Ming Kong
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Zhi-Qiang Liu
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yu-Guo Zheng
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
50
|
Roth JF, Sánchez-Rivera FJ. Precision mutational scanning: your multipass to the future of genetics. Nat Methods 2025; 22:13-15. [PMID: 39562755 DOI: 10.1038/s41592-024-02522-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Affiliation(s)
- Jonathan F Roth
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Francisco J Sánchez-Rivera
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|