1
|
Wu X, Wan X, Yu H, Liu H. Recent advances in CRISPR-Cas system for Saccharomyces cerevisiae engineering. Biotechnol Adv 2025; 81:108557. [PMID: 40081781 DOI: 10.1016/j.biotechadv.2025.108557] [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: 12/04/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Yeast Saccharomyces cerevisiae (S. cerevisiae) is a crucial industrial platform for producing a wide range of chemicals, fuels, pharmaceuticals, and nutraceutical ingredients. It is also commonly used as a model organism for fundamental research. In recent years, the CRISPR-Cas (clustered regularly interspaced short palindromic repeats-CRISPR-associated proteins) system has become the preferred technology for genetic manipulation in S. cerevisiae owing to its high efficiency, precision, and user-friendliness. This system, along with its extensive toolbox, has significantly accelerated the construction of pathways, enzyme optimization, and metabolic engineering in S. cerevisiae. Furthermore, it has allowed researchers to accelerate phenotypic evolution and gain deeper insights into fundamental biological questions, such as genotype-phenotype relationships. In this review, we summarize the latest advancements in the CRISPR-Cas toolbox for S. cerevisiae and highlight its applications in yeast cell factory construction and optimization, enzyme and phenotypic evolution, genome-scale functional interrogation, gene drives, and the advancement of biotechnologies. Finally, we discuss the challenges and potential for further optimization and applications of the CRISPR-Cas system in S. cerevisiae.
Collapse
Affiliation(s)
- Xinxin Wu
- Frontiers Science Center of Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Xiaowen Wan
- State Key Laboratory of Biotherapy and Cancer Centre/Collaborative Innovation Centre for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongbin Yu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huayi Liu
- Frontiers Science Center of Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; State Key Laboratory of Biotherapy and Cancer Centre/Collaborative Innovation Centre for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Hematology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Rothgangl T, Tálas A, Ioannidi EI, Weber Y, Böck D, Matsushita M, Villiger EA, Schmidheini L, Moon WJ, Lin PJC, Fan SHY, Marquart KF, Schwerdel C, Rimann N, Faccin E, Villiger L, Muramatsu H, Vadovics M, Cremonesi A, Kulcsár PI, Thöny B, Kopf M, Häberle J, Pardi N, Tam YK, Schwank G. Treatment of a metabolic liver disease in mice with a transient prime editing approach. Nat Biomed Eng 2025:10.1038/s41551-025-01399-4. [PMID: 40394220 DOI: 10.1038/s41551-025-01399-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/11/2025] [Indexed: 05/22/2025]
Abstract
Prime editing is a versatile genome editing technology that circumvents the need for DNA double-strand break formation and homology-directed repair, making it particularly suitable for in vivo correction of pathogenic mutations. Here we developed liver-specific prime editing approaches with temporally restricted prime editor (PE) expression. We first established a dual-delivery approach where the prime editor guide RNA is continuously expressed from adeno-associated viral vectors and only the PE is transiently delivered as nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNP). This strategy achieved 26.2% editing with PEmax and 47.4% editing with PE7 at the Dnmt1 locus using a single 2 mg kg-1 dose of mRNA-LNP. When targeting the pathogenic Pahenu2 mutation in a phenylketonuria mouse model, gene correction rates reached 4.3% with PEmax and 20.7% with PE7 after three doses of 2 mg kg-1 mRNA-LNP, effectively reducing blood L-phenylalanine levels from over 1,500 µmol l-1 to below the therapeutic threshold of 360 µmol l-1. Encouraged by the high efficiency of PE7, we next explored a simplified approach where PE7 mRNA was co-delivered with synthetic prime editor guide RNAs encapsulated in LNP. This strategy yielded 35.9% editing after two doses of RNA-LNP at the Dnmt1 locus and 8.0% editing after three doses of RNA-LNP at the Pahenu2 locus, again reducing L-phenylalanine levels below 360 µmol l-1. These findings highlight the therapeutic potential of mRNA-LNP-based prime editing for treating phenylketonuria and other genetic liver diseases, offering a scalable and efficient platform for future clinical translation.
Collapse
Affiliation(s)
- Tanja Rothgangl
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - András Tálas
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Eleonora I Ioannidi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Yanik Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Desirée Böck
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Mai Matsushita
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Lukas Schmidheini
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Woohyun J Moon
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Paulo J C Lin
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Steven H Y Fan
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Kim F Marquart
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Schwerdel
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Nicole Rimann
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Erica Faccin
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Lukas Villiger
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Máté Vadovics
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Péter István Kulcsár
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Beat Thöny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying K Tam
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Dacquay LC, Antoniou P, Mentani A, Selfjord N, Mårtensson H, Hsieh PP, Mustfa S, Thom G, Wimberger S, Firth M, Akrap N, Maresca M, Peterka M. Dual inhibition of DNA-PK and Polϴ boosts precision of diverse prime editing systems. Nat Commun 2025; 16:4290. [PMID: 40341582 PMCID: PMC12062269 DOI: 10.1038/s41467-025-59708-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 05/01/2025] [Indexed: 05/10/2025] Open
Abstract
Prime editing is a genome engineering tool that allows installation of various small edits with high precision. However, prime editing efficiency and purity can vary widely across different edits, genomic targets, and cell types. Prime editing typically offers more precise editing outcomes compared to other genome editing methods such as homology-directed repair. However, it can still result in significant rates of unintended editing outcomes, such as indels or imprecise prime edits. This issue is particularly notable in systems utilizing a second nicking gRNA, such as PE3 and PE5, as well as in dual pegRNA systems and fully active nuclease systems such as PEn, which increase efficiency but compromise precision. In this work, we show that pharmacological inhibition of DNA-PK and Polϴ, two major mediators of mutagenic DNA repair pathways, improves precision of PEn, PE3, PE5, PE7, and dual pegRNA editing systems, including TwinPE, HOPE, and Bi-PE, across multiple genomic loci and edit types. We show that co-inhibition of DNA-PK and Polϴ mitigates both prime editing-unrelated indels and prime editing by-products such as template duplications. Moreover, in the case of PEn, this strategy also substantially improved the off-target editing profile. Together, our data establish small molecule modulation of DNA repair pathways as a general strategy to maximize the precision of diverse prime editing systems.
Collapse
Affiliation(s)
- Louis C Dacquay
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Promega Corporation, Madison, WI, USA
| | - Panagiotis Antoniou
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Astrid Mentani
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Niklas Selfjord
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Hanna Mårtensson
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Pei-Pei Hsieh
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Salman Mustfa
- RNA Therapy, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - George Thom
- RNA Therapy, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sandra Wimberger
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Nina Akrap
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Marcello Maresca
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Martin Peterka
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
4
|
Matuszek Z, Brown BL, Yrigollen CM, Keiser MS, Davidson BL. Current trends in gene therapy to treat inherited disorders of the brain. Mol Ther 2025; 33:1988-2014. [PMID: 40181540 DOI: 10.1016/j.ymthe.2025.03.057] [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: 03/17/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
Gene therapy development, re-engineering, and application to patients hold promise to revolutionize medicine, including therapies for disorders of the brain. Advances in delivery modalities, expression regulation, and improving safety profiles are of critical importance. Additionally, each inherited disorder has its own unique characteristics as to regions and cell types impacted and the temporal dynamics of that impact that are essential for the design of therapeutic design strategies. Here, we review the current state of the art in gene therapies for inherited brain disorders, summarizing key considerations for vector delivery, gene addition, gene silencing, gene editing, and epigenetic editing. We provide examples from animal models, human cell lines, and, where possible, clinical trials. This review also highlights the various tools available to researchers for basic research questions and discusses our views on the current limitations in the field.
Collapse
Affiliation(s)
- Zaneta Matuszek
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Brandon L Brown
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carolyn M Yrigollen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Megan S Keiser
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Osborn MJ, Panda S, Reineke TM, Tolar J, Nyström A. Progress in skin gene therapy: From the inside and out. Mol Ther 2025; 33:2065-2081. [PMID: 40077969 DOI: 10.1016/j.ymthe.2025.03.017] [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: 01/31/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
The skin is the largest organ of the body and forms and serves as the barrier for preventing external material from accessing and damaging internal organs. As the outward interface to the environment, it is accessible for the application of therapeutic agents and cellular and gene therapy represent attractive and promising options to treat severe genetic conditions for which palliation has long been the main stay. However, because of its barrier function, transit across and to the subdermal compartment can be challenging. This commentary examines the current approaches of cell and gene therapies for genetic skin disorders. We write this from a local and systemic "outside and inside." perspective. Delivery from the outside encompasses topical, intradermal, and transdermal strategies for cell and vector delivery and ex vivo cell expansion and grafting. The inside approach details systemic delivery via infusion of cells or agents toward providing benefit to the skin. We use recessive dystrophic epidermolysis bullosa (RDEB) as a representative and paradigmatic disease to showcase these approaches as a means to highlight potential broader applicability to other conditions.
Collapse
Affiliation(s)
- Mark J Osborn
- Medical School, Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular and Gene Therapy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Sidharth Panda
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jakub Tolar
- Medical School, Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular and Gene Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
6
|
Butt H, Sathish S, London E, Lee Johnson T, Essawi K, Leonard A, Tisdale JF, Demirci S. Genome editing strategies for targeted correction of β-globin mutation in sickle cell disease: From bench to bedside. Mol Ther 2025; 33:2154-2171. [PMID: 40165374 DOI: 10.1016/j.ymthe.2025.03.047] [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: 03/06/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025] Open
Abstract
Sickle cell disease (SCD) includes a range of genotypes that result in a clinical syndrome, where abnormal red blood cell (RBC) physiology leads to widespread complications affecting nearly every organ system. Treatment strategies for SCD can be broadly categorized into disease-modifying therapies and those aimed toward a cure. Although several disease-modifying drugs have been approved, they do not fully address the complexity and severity of SCD. Recent advances in allogeneic transplantation and autologous gene therapy show promising outcomes in terms of efficacy and safety. While these approaches have improved the lives of many patients, achieving a durable and comprehensive cure for all remains challenging. To address this, gene-editing technologies, including zinc-finger nucleases, TALENs, CRISPR-Cas, base editing, and prime editing, have been explored both ex vivo and in vivo for targeted correction of the β-globin gene (HBB) in SCD. However, direct correction of HBB and its translation from the laboratory to the clinic presents ongoing limitations, with challenges involved in achieving robust mutation-correction efficiency, off-target effects, and high costs of therapies. The optimal strategy for curing SCD remains uncertain, but several promising approaches are emerging. This review touches on past, present, and future developments in HBB correction.
Collapse
Affiliation(s)
- Henna Butt
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Shruti Sathish
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Evan London
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Taylor Lee Johnson
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Khaled Essawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan 45142, Saudi Arabia
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| |
Collapse
|
7
|
Marnis H, Syahputra K. Advancing fish disease research through CRISPR-Cas genome editing: Recent developments and future perspectives. FISH & SHELLFISH IMMUNOLOGY 2025; 160:110220. [PMID: 39988220 DOI: 10.1016/j.fsi.2025.110220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
CRISPR-Cas genome editing technology has transformed genetic research, by enabling unprecedented precision in modifying DNA sequences across various organisms, including fish. This review explores the significant advancements and potential uses of CRISPR-Cas technology in the study and management of fish diseases, which pose serious challenges to aquaculture and wild fish populations. Fish diseases cause significant economic losses and environmental impacts, therefore effective disease control a top priority. The review highlights the pivotal role of CRISPR-Cas in identifying disease-associated genes, which is critical to comprehending the genetic causes of disease susceptibility and resistance. Some studies have reported key genetic factors that influence disease outcomes, using targeted gene knockouts and modifications to pave the way for the development of disease-resistant fish strains. The creation of such genetically engineered fish holds great promise for enhancing aquaculture sustainability by reducing the reliance on antibiotics and other conventional disease control measures. In addition, CRISPR-Cas has facilitated in-depth studies of pathogen-host interactions, offering new insights into the mechanisms by which pathogens infect and proliferate within their hosts. By manipulating both host and pathogen genes, this technology provides a powerful tool for uncovering the molecular underpinnings of these interactions, leading to the development of more effective treatment strategies. While CRISPR-Cas has shown great promise in fish research, its application remains limited to a few species, primarily model organisms and some freshwater fish. In addition, challenges such as off-target effects, ecological risks, and ethical concerns regarding the release of genetically modified organisms into the environment must be carefully addressed. This review also discusses these challenges and emphasizes the need for robust regulatory frameworks and ongoing research to mitigate risks. Looking forward, the integration of CRISPR-Cas with other emerging technologies, such as multi-omics approaches, promises to further advance our understanding and management of fish diseases. This review concludes by envisioning the future directions of CRISPR-Cas applications in fish health, underscoring its potential to its growing in the field.
Collapse
Affiliation(s)
- Huria Marnis
- Research Center for Fishery, National Research and Innovation Agency (BRIN), Cibinong, 16911, Indonesia.
| | - Khairul Syahputra
- Research Center for Fishery, National Research and Innovation Agency (BRIN), Cibinong, 16911, Indonesia; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute for Fish and Wildlife Health, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Lu P, Zuo E, Yan J. Developing a multi-modular assembled prime editing (mPE) system improved precise multi-base insertion efficiency in dicots. J Adv Res 2025; 71:81-92. [PMID: 38942381 DOI: 10.1016/j.jare.2024.06.021] [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: 02/19/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
INTRODUCTION The Prime Editing (PE) system is a precise and versatile genome editing tool with great potential in plant breeding and plant synthetic biology. However, low PE efficiency severely restricts its application, especially in dicots. PE can introduce small tags to trace target protein or cis-element to regulate gene transcription which is an expertise superior to other gene editing tools. Owing to low efficiency, PE adaption in stably transformed Arabidopsis is lacking. OBJECTIVES This study aimed to investigate the issue of low PE efficiency in dicots and develop systematic solutions to improve it. Currently, PE in dicots is undetectable and inconsistent, and this study seeks to address it. Split PE into several parts showed better performance in some target sites in mammal cells. We plan to discover the optimal split PE combination in dicot. METHODS We conducted large-scale transformation experiments in dicot model plants Arabidopsis thaliana (At) and Nicotiana benthamiana (Nb) by Agrobacterium-mediated transformation with deep amplicon sequencing (0.2-0.5 million clean total reads). RESULTS The editing efficiency decreased upon using a fused reverse transcriptase (RT) or an extended pegRNA separately and further decreased dramatically when these were used together. With the help of the pol II strategy to express PE gRNA (pegRNA), we named the most effective split PE combination as a multi-modular assembled prime editing system (mPE). mPE exhibited improved precise editing efficiency on most gene sites with various editing types, ranging from 1.3-fold to 1288.5-fold and achieved PE on some sites that could not be edited by original PE2. Especially, mPE showed superiority for multi-base insertion with an average improvement of 197.9-fold. CONCLUSION The original PE architecture strongly inhibited the cleavage activity of Cas9. Split PE improved PE efficiency extensively and was in favor of introducing small insertions in dicot plants, indicating that different PE variants might have their own expertise.
Collapse
Affiliation(s)
- Pengjun Lu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Erwei Zuo
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Jianbin Yan
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China.
| |
Collapse
|
9
|
James JS, Dai J, Chew WL, Cai Y. The design and engineering of synthetic genomes. Nat Rev Genet 2025; 26:298-319. [PMID: 39506144 DOI: 10.1038/s41576-024-00786-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: 09/23/2024] [Indexed: 11/08/2024]
Abstract
Synthetic genomics seeks to design and construct entire genomes to mechanistically dissect fundamental questions of genome function and to engineer organisms for diverse applications, including bioproduction of high-value chemicals and biologics, advanced cell therapies, and stress-tolerant crops. Recent progress has been fuelled by advancements in DNA synthesis, assembly, delivery and editing. Computational innovations, such as the use of artificial intelligence to provide prediction of function, also provide increasing capabilities to guide synthetic genome design and construction. However, translating synthetic genome-scale projects from idea to implementation remains highly complex. Here, we aim to streamline this implementation process by comprehensively reviewing the strategies for design, construction, delivery, debugging and tailoring of synthetic genomes as well as their potential applications.
Collapse
Affiliation(s)
- Joshua S James
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Junbiao Dai
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Shenzhen Key Laboratory of Agricultural Synthetic Biology, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Leong Chew
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Yizhi Cai
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK.
| |
Collapse
|
10
|
Mathis N, Allam A, Tálas A, Kissling L, Benvenuto E, Schmidheini L, Schep R, Damodharan T, Balázs Z, Janjuha S, Ioannidi EI, Böck D, van Steensel B, Krauthammer M, Schwank G. Machine learning prediction of prime editing efficiency across diverse chromatin contexts. Nat Biotechnol 2025; 43:712-719. [PMID: 38907037 PMCID: PMC7617539 DOI: 10.1038/s41587-024-02268-2] [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: 10/08/2023] [Accepted: 05/01/2024] [Indexed: 06/23/2024]
Abstract
The success of prime editing depends on the prime editing guide RNA (pegRNA) design and target locus. Here, we developed machine learning models that reliably predict prime editing efficiency. PRIDICT2.0 assesses the performance of pegRNAs for all edit types up to 15 bp in length in mismatch repair-deficient and mismatch repair-proficient cell lines and in vivo in primary cells. With ePRIDICT, we further developed a model that quantifies how local chromatin environments impact prime editing rates.
Collapse
Affiliation(s)
- Nicolas Mathis
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Ahmed Allam
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - András Tálas
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Lucas Kissling
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Elena Benvenuto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Lukas Schmidheini
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ruben Schep
- Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tanav Damodharan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Zsolt Balázs
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Sharan Janjuha
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Eleonora I Ioannidi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Desirée Böck
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Bas van Steensel
- Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
11
|
Han X, Xu X, Xiong Y, Zhao G, He R, Su Y, Li S, Zhao C, Xi X, Zhao Y, Xu X, Xie S, Wang H, Li X, Zhao S, Ruan J. Enhancing Prime Editing Efficiency Through Modulation of Methylation on the Newly Synthesized DNA Strand and Prolonged Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417790. [PMID: 40051369 PMCID: PMC12061241 DOI: 10.1002/advs.202417790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Indexed: 05/10/2025]
Abstract
Prime editors (PEs) have emerged as transformative tools for precision genome engineering, yet their broader application remains constrained by incomplete understanding of repair mechanisms. In this study, it is found that an increase in the methylation level of the CpG sequence on the newly generated strand can increase PE efficiency and that de novo DNA methyltransferases (DNMT3A/3B) are involved in the PE repair pathway. On the basis of these novel findings, the development of an episomal element-driven PE system (epiPE) achieved through the use of EBNA1/oriP are presented, which increases methylation levels around target sites and prolongs PE expression. A comparative analysis with canonical PE systems, including PE2, lentiPE2, and PE4max, reveals that the epiPE2 system significantly enhances editing efficiency while maintaining minimal insertion and deletion (indels) rates. Specifically, comparing to PE2, the epiPE2 system demonstrated an efficiency enhancement of 2.0 to 38.2-fold. In addition, the epiPE2 system is capable of efficient multiplex precise gene editing at up to 10 genetic loci in human cells. In conclusion, this findings increase the understanding of the PE repair mechanism, and presents the epiPE2 system as an efficient and multiplex-capable prime editing tool with potential applications in both basic research and translational studies.
Collapse
Affiliation(s)
- Xiaosong Han
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Yazhouwan National LaboratorySanya572024P. R. China
| | - Xianghua Xu
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Youcai Xiong
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Guangxing Zhao
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Ruigao He
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Yinyu Su
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Sheng Li
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Changzhi Zhao
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Yazhouwan National LaboratorySanya572024P. R. China
| | - Xiaoning Xi
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Yunxia Zhao
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Yazhouwan National LaboratorySanya572024P. R. China
| | - Xuewen Xu
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Heng Wang
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionHuazhong Agricultural UniversityWuhan430070P. R. China
- Hubei Hongshan LaboratoryFrontiers Science Center for Animal Breeding and Sustainable ProductionWuhan430070P. R. China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Yazhouwan National LaboratorySanya572024P. R. China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionHuazhong Agricultural UniversityWuhan430070P. R. China
- Hubei Hongshan LaboratoryFrontiers Science Center for Animal Breeding and Sustainable ProductionWuhan430070P. R. China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070P. R. China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionHuazhong Agricultural UniversityWuhan430070P. R. China
| |
Collapse
|
12
|
Zhang P, Xu Z. The advancements in precision medicine for Leber congenital amaurosis: Breakthroughs from genetic diagnosis to therapy. Surv Ophthalmol 2025:S0039-6257(25)00070-0. [PMID: 40311816 DOI: 10.1016/j.survophthal.2025.04.005] [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: 12/17/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Leber congenital amaurosis (LCA) is a hereditary retinal disease, typically manifesting as severe vision impairment in infancy. With the advancement of precision medicine, genetic diagnosis and targeted therapies offer new hope for LCA patients, significantly improving both diagnostic accuracy and therapeutic efficacy. We summarize the epidemiological characteristics, clinical manifestations, and molecular genetics underlying LCA. It also highlights recent developments in precision treatment strategies, including gene replacement therapy, CRISPR/Cas9-mediated gene editing, and antisense oligonucleotide therapies. In addition, we discuss the applications of induced pluripotent stem cells and retinal organoids in LCA treatment research. Furthermore, we explore preventive strategies and future treatment directions for LCA, including the development of novel gene therapy vectors, the optimization of combinatorial treatment strategies, and the formulation of personalized treatment approaches. These advancements hold significant potential to offer improved treatment options and enhance the quality of life for LCA patients.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
13
|
Mu H, Liu Y, Chi Y, Wang F, Meng S, Zhang Y, Wang X, Zhao D. Systematic optimization of prime editing for enhanced efficiency and versatility in genome engineering across diverse cell types. Front Cell Dev Biol 2025; 13:1589034. [PMID: 40365017 PMCID: PMC12069386 DOI: 10.3389/fcell.2025.1589034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Prime editing offers remarkable versatility in genome editing, but its efficiency remains a major bottleneck. While continuous optimization of the prime editing enzymes and guide RNAs (pegRNAs) has improved editing outcomes, the method of delivery also plays a crucial role in overall performance. To maximize prime editing efficiency, we implemented a series of systematic optimizations, achieving up to 80% editing efficiency across multiple loci and cell lines. Beyond integrating the latest advancements in prime editing, our approach combined stable genomic integration of prime editors via the piggyBac transposon system, selection of integrated single clones, the use of an enhanced promoter, and lentiviral delivery of pegRNAs, ensuring robust, ubiquitous, and sustained expression of both prime editors and pegRNAs. To further assess its efficacy in challenging cell types, we validated our optimized system in human pluripotent stem cells (hPSCs) in both primed and naïve states, achieving substantial editing efficiencies of up to 50%. Collectively, our optimized prime editing strategy provides a highly efficient and versatile framework for genome engineering in vitro, serving as a roadmap for refining prime editing technologies and expanding their applications in genetic research and therapeutic development.
Collapse
Affiliation(s)
- Huiling Mu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yeyi Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Yijia Chi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Fei Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shuting Meng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, Henan University, Kaifeng, China
| | - Yi Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xunting Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Dongxin Zhao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Gao X, Zhou C, Feng Y, Ye B, Zhao Z, Qi L, Hu L, Deng Y, Lin C, Ding Q, Liu G, Wang C, Song C, Qian B, Wu T, Wang X, Liu Z, Lin Z, Zhang M. Research progress of gene editing technology in neurological diseases. Gene 2025:149534. [PMID: 40294708 DOI: 10.1016/j.gene.2025.149534] [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: 02/20/2025] [Revised: 04/12/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Gene editing (GE) technology is a genetic manipulation technique based on artificial nucleases that enables the precise modification of DNA or RNA. With the development of technology, GE in disease treatment is becoming increasingly widespread, playing an essential role in haematology, cancer, and neurological disorders (ND). This review describes the principles, advantages, and limitations of four GE technologies, focusing on the fourth generation of GE (next-generation GE). The next-generation GE technology breaks the limitations of traditional GE technology, makes GE more precise and stable, and broadens the scope of gene technology applications. Additionally, this review explores the latest gene therapy strategies for ND, focusing on the application of next-generation GE technologies to examine the prospects for the application of GE technologies. This study discusses and analyses the great advantages and potential of GE technology for treating ND and elucidates the shortcomings of GE in this field.
Collapse
Affiliation(s)
- Xiuying Gao
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunting Zhou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yani Feng
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bangming Ye
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziming Zhao
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lixin Qi
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Hu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixuan Deng
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Congying Lin
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiang Ding
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guanhao Liu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenyi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyu Song
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bo Qian
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianhao Wu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiming Liu
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Zhenlang Lin
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Min Zhang
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
15
|
Haider S, Mussolino C. Fine-Tuning Homology-Directed Repair (HDR) for Precision Genome Editing: Current Strategies and Future Directions. Int J Mol Sci 2025; 26:4067. [PMID: 40362308 PMCID: PMC12071731 DOI: 10.3390/ijms26094067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
CRISPR-Cas9 is a powerful genome-editing technology that can precisely target and cleave DNA to induce double-strand breaks (DSBs) at almost any genomic locus. While this versatility holds tremendous therapeutic potential, the predominant cellular pathway for DSB repair-non-homologous end-joining (NHEJ)-often introduces small insertions or deletions that disrupt the target site. In contrast, homology-directed repair (HDR) utilizes exogenous donor templates to enable precise gene modifications, including targeted insertions, deletions, and substitutions. However, HDR remains relatively inefficient compared to NHEJ, especially in postmitotic cells where cell cycle constraints further limit HDR. To address this challenge, numerous methodologies have been explored, ranging from inhibiting key NHEJ factors and optimizing donor templates to synchronizing cells in HDR-permissive phases and engineering HDR-enhancing fusion proteins. These strategies collectively aim to boost HDR efficiency and expand the clinical and research utility of CRISPR-Cas9. In this review, we discuss recent advances in manipulating the balance between NHEJ and HDR, examine the trade-offs and practical considerations of these approaches, and highlight promising directions for achieving high-fidelity genome editing in diverse cell types.
Collapse
Affiliation(s)
- Sibtain Haider
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany;
- Center for Chronic Immunodeficiency (CCI), Medical Center—University of Freiburg, 79106 Freiburg, Germany
| | - Claudio Mussolino
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany;
- Center for Chronic Immunodeficiency (CCI), Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
16
|
Rengifo-Gonzalez M, Mazzuoli MV, Janssen AB, Rueff AS, Burnier J, Liu X, Veening JW. Make-or-break prime editing for genome engineering in Streptococcus pneumoniae. Nat Commun 2025; 16:3796. [PMID: 40263274 PMCID: PMC12015366 DOI: 10.1038/s41467-025-59068-8] [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: 07/11/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
CRISPR-Cas9 has revolutionized genome engineering by allowing precise introductions of DNA double-strand breaks (DSBs). However, genome engineering in bacteria is still a complex, multi-step process requiring a donor DNA template for repair of DSBs. Prime editing circumvents this need as the repair template is indirectly provided within the prime editing guide RNA (pegRNA). Here, we developed make-or-break Prime Editing (mbPE) that allows for precise and effective genetic engineering in the opportunistic human pathogen Streptococcus pneumoniae. In contrast to traditional prime editing in which a nicking Cas9 is employed, mbPE harnesses wild type Cas9 in combination with a pegRNA that destroys the seed region or protospacer adjacent motif. Since most bacteria poorly perform template-independent end joining, correctly genome-edited clones are selectively enriched during mbPE. We show that mbPE is RecA-independent and can be used to introduce point mutations, deletions and targeted insertions, including protein tags such as a split luciferase, at selection efficiencies of over 93%. mbPE enables sequential genome editing, is scalable, and can be used to generate pools of mutants in a high-throughput manner. The mbPE system and pegRNA design guidelines described here will ameliorate future bacterial genome editing endeavors.
Collapse
Affiliation(s)
- Monica Rengifo-Gonzalez
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-, Lausanne, Switzerland
| | - Maria-Vittoria Mazzuoli
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-, Lausanne, Switzerland
| | - Axel B Janssen
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-, Lausanne, Switzerland
| | - Anne-Stéphanie Rueff
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-, Lausanne, Switzerland
| | - Jessica Burnier
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-, Lausanne, Switzerland
| | - Xue Liu
- Department of Pathogen Biology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, China.
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-, Lausanne, Switzerland.
| |
Collapse
|
17
|
Biber J, Gandor C, Becirovic E, Michalakis S. Retina-directed gene therapy: Achievements and remaining challenges. Pharmacol Ther 2025; 271:108862. [PMID: 40268248 DOI: 10.1016/j.pharmthera.2025.108862] [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: 10/07/2024] [Revised: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Gene therapy is an innovative medical approach that offers new treatment options for congenital and acquired diseases by transferring, correcting, inactivating or regulating genes to supplement, replace or modify a gene function. The approval of voretigene neparvovec (Luxturna), a gene therapy for RPE65-associated retinopathy, has marked a milestone for the field of retinal gene therapy, but has also helped to accelerate the development of gene therapies for genetic diseases affecting other organs. Voretigene neparvovec is a vector based on adeno-associated virus (AAV) that delivers a functional copy of RPE65 to supplement the missing function of this gene. The AAV-based gene delivery has proven to be versatile and safe for the transfer of genetic material to retinal cells. However, challenges remain in treating additional inherited as well as acquired retinopathies with this technology. Despite the high level of activity in this field, no other AAV gene therapy for retinal diseases has been approved since voretigene neparvovec. Ongoing research focuses on overcoming the current restraints through innovative strategies like AAV capsid engineering, dual-AAV vector systems, or CRISPR/Cas-mediated genome editing. Additionally, AAV gene therapy is being explored for the treatment of complex acquired diseases like age-related macular degeneration (AMD) and diabetic retinopathy (DR) by targeting molecules involved in the pathobiology of the degenerative processes. This review outlines the current state of retinal gene therapy, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Josef Biber
- Department of Ophthalmology, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Catharina Gandor
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Elvir Becirovic
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Stylianos Michalakis
- Department of Ophthalmology, LMU University Hospital, LMU Munich, 80336 Munich, Germany.
| |
Collapse
|
18
|
Sheng M, Zheng Y, Li K, Ye C, Cao G. Optimized electroporation buffer improves transfection and prime editing efficiency in adult bovine fibroblasts. Gene 2025; 946:149315. [PMID: 39922551 DOI: 10.1016/j.gene.2025.149315] [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: 11/27/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
For livestock breeding, using somatic cells from adult animals for gene editing and subsequent cloning allows the preservation and enhancement of superior traits from the parent directly in the offspring, while avoiding the loss of genetic gain that can occur through crossbreeding. However, primary cells generally more difficult to transfect and perform gene editing. To date, most related studies have used more vigorous fetal fibroblasts as donor cells, while using somatic cells from adult animals requires more post-editing screening efforts due to the low yield of edited cells. Here, we performed electroporation on adult bovine ear fibroblasts (BEFs) under various conditions such as electrical pulse settings, plasmid dosage, cell density, and concentration of electroporation buffer, and evaluated the transfect efficiency using flow cytometry analysis. We confirmed that the 270 V-10-10 program (270 V, 10 ms, 10 cycles) using 1.5 million cells and 5 µg of EGFP plasmid yielded the highest number of EGFP positive cells. Additionally, we used prime editor (PE) to edit the MSTN locus in BEFs. More importantly, we discovered that lowering the osmolarity of the electroporation buffer improves both electroporation and gene editing efficiency, which relate to the repression of cGAS-STING pathway. Our finding provides valuable references for using electroporation methods in adult bovine primary cell gene editing.
Collapse
Affiliation(s)
- Mingxuan Sheng
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Yuequan Zheng
- Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Kunlong Li
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Chongyuan Ye
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Gengsheng Cao
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| |
Collapse
|
19
|
He S, Liu Y, Zhang Z, Cai M, Hao Y, Hu H. Gene Editing in Ganoderma lucidum: Development, Challenges, and Future Prospects. J Fungi (Basel) 2025; 11:310. [PMID: 40278130 PMCID: PMC12029067 DOI: 10.3390/jof11040310] [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: 03/10/2025] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
As an emerging and innovative technology, gene-editing technology has been widely applied in crop breeding, human disease treatment, animal model research, drug and vaccine development, and microbial engineering. We mainly introduce the development of gene-editing technology, the application of clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) in Ganoderma lucidum breeding, the current challenges and optimization strategies in the use of gene-editing technology in Ganoderma breeding, as well as the current status of gene-editing technology in Ganoderma breeding. Finally, the future research directions and innovative strategies that gene editing may explore in Ganoderma breeding are prospects given the existing background, future research directions, and innovative strategies that gene editing may explore in Ganoderma breeding prospects.
Collapse
Affiliation(s)
- Shiqi He
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Yuanchao Liu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Zhi Zhang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
- Guangdong Yuewei Biotechnology Co., Ltd., Shaoguan 512029, China
| | - Manjun Cai
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Yufan Hao
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Huiping Hu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
- Guangdong Yuewei Biotechnology Co., Ltd., Shaoguan 512029, China
| |
Collapse
|
20
|
Almasoudi HH. Therapeutic promise of CRISPR-Cas9 gene editing in sickle cell disease and β-thalassemia: A current review. Curr Res Transl Med 2025; 73:103513. [PMID: 40252393 DOI: 10.1016/j.retram.2025.103513] [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: 01/19/2025] [Revised: 03/15/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
Sickle cell disease (SCD and β-thalassemia (BT) affects millions of people worldwide. In addition, around 500,000 infants are born with SCD and 60,000 people are diagnosed with BT every year. Mutations in the hemoglobin subunit beta (HBB) gene are responsible for causing both BT and SCD. Indeed, the diversity of potential mutations in the HBB gene elucidates the diversity in clinical severity observed in individuals with BT and related morbidities. On the other hand, SCD takes place because of the alteration in a single amino acid at position 6 in the beta-globin chain, where a base substitution occurs from glutamic acid to valine, which eventually results in abnormal sickle hemoglobin. Conventional therapies for BT and SCD including pharmaceutical drugs and blood transfusion might temporarily improve the clinical severity of these diseases, however these therapies cannot cure the diseases. CRISPR-Cas9 (CC9) is revolutionizing genome engineering, offering promising therapeutic avenues for genetic diseases. Therefore, CC9-mediated gene therapy provides great hope in the treatment of both BT and SCD. CC9-mediated gene therapy has already demonstrated its effectiveness in correcting both SCD and BT-causing mutations. Moreover, CC9-mediated gene editing was found to be effective in reactivating the expression of hemoglobin F (HbF) and regulating LRF and BCL11A. A number of clinical trials with CC9 gene-edited therapies are being carried out to elucidate their potential in treating BT and SCD. Genetics and pathophysiological mechanisms of SCD and BT, the mechanism of CC9-mediated gene editing, and common delivery methods of the CC9 system have been discussed in this review. Moreover, an in-depth discussion on applications and the current status of CC9-mediated gene editing in SCD and BT along with current challenges and future perspectives have been provided.
Collapse
Affiliation(s)
- Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia.
| |
Collapse
|
21
|
Petrova IO, Smirnikhina SA. Prime Editing in Dividing and Quiescent Cells. Int J Mol Sci 2025; 26:3596. [PMID: 40332080 PMCID: PMC12026808 DOI: 10.3390/ijms26083596] [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: 02/26/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Prime editing is a method of genome editing based on reverse transcription. Recent results have shown its elevated efficiency in dividing cells, which raises some questions regarding the mechanism of this effect, because prime editing does not employ homology-driven repair. This mini review aims to identify the reason for this phenomenon and find a possible solution to the problems that it poses. In dividing cells, prime editing takes advantage of high levels of dNTPs and active endonuclease and ligase machinery, such as FEN1 endonuclease and LIG1 ligase, but DNA mismatch repair, which is closely associated with replication, works against prime editing. Prime editing is a method which relies on retroviral reverse transcription, so mechanisms of intrinsic anti-retroviral defense should also work against editing. One of the factors which drastically reduce the efficiency of reverse translation is SAMHD1, which maintains low levels of dNTPs in non-dividing cells. Recent works aimed at the mitigation of SAMHD1 function demonstrated a significant increase in prime editing efficiency.
Collapse
Affiliation(s)
- Irina O. Petrova
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, 115478 Moscow, Russia
| | | |
Collapse
|
22
|
Antoniou P, Dacquay L, Mårtensson H, Madeyski-Bengtson K, Loyd AL, Shiriaeva A, Gordon E, Mustfa S, Thom G, Hsieh PP, Šviković S, Firth M, Akrap N, Maresca M, Peterka M. Modified pegRNAs mitigate scaffold-derived prime editing by-products. Nat Commun 2025; 16:3374. [PMID: 40204736 PMCID: PMC11982355 DOI: 10.1038/s41467-025-58653-1] [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: 08/07/2024] [Accepted: 03/20/2025] [Indexed: 04/11/2025] Open
Abstract
Prime editors (PEs) employ reverse transcriptase (RT) to install genomic edits using a template within the prime editing guide RNA (pegRNA). RT creates a 3' genomic flap containing the intended edit. However, reverse transcription can continue beyond the template, incorporating the pegRNA scaffold sequence into the 3' flap. These scaffold-derived by-products can be installed alongside the intended edit, reducing prime editing precision. Here, we develop a method that prevents RT from accessing the scaffold, thereby mitigating such by-products. We demonstrate that an internal abasic spacer or 2'-O-methylation within the pegRNAs terminates RT at the end of the template. This prevents scaffold-derived sequences from being incorporated into the target locus. We benchmark these pegRNAs in different cell types and demonstrate that they can be used with processive PEs such as PE6d or PE**. Our findings provide a simple approach to mitigate a common prime editing by-product and improve prime editing precision.
Collapse
Affiliation(s)
- Panagiotis Antoniou
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Louis Dacquay
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Promega Corporation, Madison, WI, USA
| | - Hanna Mårtensson
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Anna-Lena Loyd
- Translational Genomics, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Shiriaeva
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Promega Corporation, Madison, WI, USA
| | - Euan Gordon
- Protein Science, Structure and Biophysics, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Salman Mustfa
- RNA Therapy, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - George Thom
- RNA Therapy, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Pei-Pei Hsieh
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Saša Šviković
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Nina Akrap
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Marcello Maresca
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Martin Peterka
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
23
|
Herger M, Kajba CM, Buckley M, Cunha A, Strom M, Findlay GM. High-throughput screening of human genetic variants by pooled prime editing. CELL GENOMICS 2025; 5:100814. [PMID: 40120586 PMCID: PMC12008803 DOI: 10.1016/j.xgen.2025.100814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 01/10/2025] [Accepted: 02/13/2025] [Indexed: 03/25/2025]
Abstract
Multiplexed assays of variant effect (MAVEs) enable scalable functional assessment of human genetic variants. However, established MAVEs are limited by exogenous expression of variants or constraints of genome editing. Here, we introduce a pooled prime editing (PE) platform to scalably assay variants in their endogenous context. We first improve efficiency of PE in HAP1 cells, defining optimal prime editing guide RNA (pegRNA) designs and establishing enrichment of edited cells via co-selection. We next demonstrate negative selection screening by testing over 7,500 pegRNAs targeting SMARCB1 and observing depletion of efficiently installed loss-of-function (LoF) variants. We then screen for LoF variants in MLH1 via 6-thioguanine selection, testing 65.3% of all possible SNVs in a 200-bp region including exon 10 and 362 non-coding variants from ClinVar spanning a 60-kb region. The platform's overall accuracy for discriminating pathogenic variants indicates that it will be highly valuable for identifying new variants underlying diverse human phenotypes across large genomic regions.
Collapse
Affiliation(s)
- Michael Herger
- The Genome Function Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Christina M Kajba
- The Genome Function Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Megan Buckley
- The Genome Function Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Ana Cunha
- Viral Vector Core, Human Biology Facility, The Francis Crick Institute, London NW1 1AT, UK
| | - Molly Strom
- Viral Vector Core, Human Biology Facility, The Francis Crick Institute, London NW1 1AT, UK
| | - Gregory M Findlay
- The Genome Function Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
24
|
Wu K, Sánchez-Rivera FJ. It's prime time for multiplexed prime editing. CELL GENOMICS 2025; 5:100852. [PMID: 40209678 PMCID: PMC12008801 DOI: 10.1016/j.xgen.2025.100852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Prime editing screens allow precise and scalable studies of genetic variants in their native genomic context but are limited by variable editing efficiency. In this issue of Cell Genomics, Herger, Kajba, et al.1 overcome these challenges by optimizing and applying prime editing screens to investigate variants in SMARCB1 and MLH1.
Collapse
Affiliation(s)
- Ke Wu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Francisco J Sánchez-Rivera
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
25
|
Pincay J, da Costa BL, Quinn PMJ, Rodriguez M, Zhou A, Kong MD, Sparrow JR, Tsang SH. Phenotypic spectrum and theoretical prime editing analysis of WDR19-mediated retinal degeneration. Doc Ophthalmol 2025:10.1007/s10633-025-10016-3. [PMID: 40183892 DOI: 10.1007/s10633-025-10016-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE The ciliopathies are a broad category of pleiotropic disease with numerous genes involved in pathogenesis. One of the genes implicated in the ciliopathies is WDR19, which can lead to several syndromic diseases that may manifest with a form of retinal degeneration. There is a lack of reporting on the WDR19-mediated retinal phenotype, and therefore warrants more clinical investigation. With retinal degeneration being the most prevalent symptom among the ciliopathies, phenotypic reporting is needed to enhance understanding of pathogenesis. METHODS Clinical, imaging, and diagnostic records of patients with two variants in the WDR19 gene and a form of retinal degeneration were retrospectively reviewed. Two different individuals analyzed the variants in the studied patients using SnapGene (Version 4.3.11), employing both the canonical NGG PAM and the NGA PAM prime editors. RESULTS Four patients from three families each carrying biallelic variants the WDR19 gene were reviewed. Two of the six unique variants identified among the patients were novel. Two identical twin patients presented with a recessive Stargardt (STGD)-like phenotype while the other two patients presented with a clinical picture more characteristic of retinitis pigmentosa (RP). Three of four patients had thickened external limiting membrane (ELM) on spectral-domain optical coherence tomography (SD-OCT). Full-field electroretinograms (ffERG) performed on two patients with the STGD-like phenotype showed a cone-rod pattern of degeneration. Quantitative short-wave fundus autofluorescence (qAF) performed on the two STGD-like patients was within the 95th percentile of normal eyes. CONCLUSIONS WDR19-mediated retinal degeneration is heterogenous in presentation, and in some cases can phenocopy STGD. The foveal sparing phenotype was apparent in three of four patients with relatively preserved visual acuity, which may serve as a retinal prognostic factor in patients with pathogenic variants in WDR19. All six variants evaluated are correctable by prime editing, establishing a foundation for future research in therapeutic development.
Collapse
Affiliation(s)
- Jorge Pincay
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- State University of New York at Downstate Medical Center, Brooklyn, NY, USA
| | - Bruna Lopes da Costa
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University. New York, New York, NY, USA
| | - Peter M J Quinn
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Marilyn Rodriguez
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ashley Zhou
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maximilian D Kong
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
- State University of New York at Downstate Medical Center, Brooklyn, NY, USA
| | - Janet R Sparrow
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University. New York, New York, NY, USA.
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.
- Vanderbilt Clinic, Columbia University Irving Medical Center, 622 W 168Th St 3Rd Floor, New York, NY, 10032, USA.
| |
Collapse
|
26
|
Cabré-Romans JJ, Cuella-Martin R. CRISPR-dependent base editing as a therapeutic strategy for rare monogenic disorders. Front Genome Ed 2025; 7:1553590. [PMID: 40242216 PMCID: PMC12000063 DOI: 10.3389/fgeed.2025.1553590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Rare monogenic disorders are caused by mutations in single genes and have an incidence rate of less than 0.5%. Due to their low prevalence, these diseases often attract limited research and commercial interest, leading to significant unmet medical needs. In a therapeutic landscape where treatments are targeted to manage symptoms, gene editing therapy emerges as a promising approach to craft curative and lasting treatments for these patients, often referred to as "one-and-done" therapeutics. CRISPR-dependent base editing enables the precise correction of genetic mutations by direct modification of DNA bases without creating potentially deleterious DNA double-strand breaks. Base editors combine a nickase version of Cas9 with cytosine or adenine deaminases to convert C·G to T·A and A·T to G·C, respectively. Together, cytosine (CBE) and adenine (ABE) base editors can theoretically correct ∼95% of pathogenic transition mutations cataloged in ClinVar. This mini-review explores the application of base editing as a therapeutic approach for rare monogenic disorders. It provides an overview of the state of gene therapies and a comprehensive compilation of preclinical studies using base editing to treat rare monogenic disorders. Key considerations for designing base editing-driven therapeutics are summarized in a user-friendly guide for researchers interested in applying this technology to a specific rare monogenic disorder. Finally, we discuss the prospects and challenges for bench-to-bedside translation of base editing therapies for rare monogenic disorders.
Collapse
Affiliation(s)
- Júlia-Jié Cabré-Romans
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Raquel Cuella-Martin
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Levesque S, Cosentino A, Verma A, Genovese P, Bauer DE. Enhancing prime editing in hematopoietic stem and progenitor cells by modulating nucleotide metabolism. Nat Biotechnol 2025; 43:534-538. [PMID: 38806736 DOI: 10.1038/s41587-024-02266-4] [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: 10/16/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024]
Abstract
Therapeutic prime editing of hematopoietic stem and progenitor cells (HSPCs) holds great potential to remedy blood disorders. Quiescent cells have low nucleotide levels and resist retroviral infection, and it is possible that nucleotide metabolism could limit reverse transcription-mediated prime editing in HSPCs. We demonstrate that deoxynucleoside supplementation and Vpx-mediated degradation of SAMHD1 improve prime editing efficiency in HSPCs, especially when coupled with editing approaches that evade mismatch repair.
Collapse
Affiliation(s)
- Sébastien Levesque
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Andrea Cosentino
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Milano-Bicocca University, Milan, Italy
| | - Archana Verma
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Pietro Genovese
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Li M, Lin Y, Cheng Q, Wei T. Prime Editing: A Revolutionary Technology for Precise Treatment of Genetic Disorders. Cell Prolif 2025; 58:e13808. [PMID: 40014809 PMCID: PMC11969253 DOI: 10.1111/cpr.13808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/06/2024] [Accepted: 01/03/2025] [Indexed: 03/01/2025] Open
Abstract
Genetic diseases have long posed significant challenges, with limited breakthroughs in treatment. Recent advances in gene editing technologies offer new possibilities in gene therapy for the treatment of inherited disorders. However, traditional gene editing methods have limitations that hinder their potential for clinical use, such as limited editing capabilities and the production of unintended byproducts. To overcome these limitations, prime editing (PE) has been developed as a powerful tool for precise and efficient genome modification. In this review, we provide an overview of the latest advancements in PE and its potential applications in the treatment of inherited disorders. Furthermore, we examine the current delivery vehicles employed for delivering PE systems in vitro and in vivo, and analyze their respective benefits and limitations. Ultimately, we discuss the challenges that need to be addressed to fully unlock the potential of PE for the remission or cure of genetic diseases.
Collapse
Affiliation(s)
- Mengyao Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yi Lin
- Department of Biomedical Engineering, College of Future TechnologyPeking UniversityBeijingChina
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future TechnologyPeking UniversityBeijingChina
- Beijing Advanced Center of RNA BiologyPeking UniversityBeijingChina
| | - Tuo Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
29
|
Koodamvetty A, Thangavel S. Advancing Precision Medicine: Recent Innovations in Gene Editing Technologies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410237. [PMID: 40025867 PMCID: PMC11984848 DOI: 10.1002/advs.202410237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/13/2024] [Indexed: 03/04/2025]
Abstract
The advent of gene editing has significantly advanced the field of medicine, opening new frontiers in the treatment of genetic disorders, cancer, and infectious diseases. Gene editing technology remains a dynamic and promising area of research and development. Recent advancements in protein and RNA engineering within this field have addressed critical issues such as imprecise edits, poor editing efficiency, and off-target effects. Advancements in delivery methods have allowed the achievement of therapeutic or even selection-free gene editing efficiency with reduced toxicity in primary cells, thereby enhancing the safety and efficacy of gene manipulation. This progress paves the way for transformative changes in molecular biology, medicine, and other fields. This review provides a comprehensive overview of the advancements in gene editing techniques, focusing on prime editor proteins and their engineered variants. It also explores alternative systems that expand the toolkit for precise genomic modifications and highlights the potential of these innovations in treating hematological disorders, while also discussing the limitations and challenges that remain.
Collapse
Affiliation(s)
- Abhijith Koodamvetty
- Centre for Stem Cell Research (CSCR)A unit of InStem BengaluruChristian Medical College campusVelloreTamil Nadu632002India
- Manipal Academy of Higher EducationManipalKarnataka576104India
| | - Saravanabhavan Thangavel
- Centre for Stem Cell Research (CSCR)A unit of InStem BengaluruChristian Medical College campusVelloreTamil Nadu632002India
| |
Collapse
|
30
|
Mukhare R, Gandhi KA, Kadam A, Raja A, Singh A, Madhav M, Chaubal R, Pandey S, Gupta S. Integration of Organoids With CRISPR Screens: A Narrative Review. Biol Cell 2025; 117:e70006. [PMID: 40223602 PMCID: PMC11995251 DOI: 10.1111/boc.70006] [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: 03/05/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025]
Abstract
Organoids represent a significant advancement in disease modeling, demonstrated by their capacity to mimic the physiological/pathological structure and functional characteristics of the native tissue. Recently CRISPR/Cas9 technology has emerged as a powerful tool in combination with organoids for the development of novel therapies in preclinical settings. This review explores the current literature on applications of pooled CRISPR screening in organoids and the emerging role of these models in understanding cancer. We highlight the evolution of genome-wide CRISPR gRNA library screens in organoids, noting their increasing adoption in the field over the past decade. Noteworthy studies utilizing these screens to investigate oncogenic vulnerabilities and developmental pathways in various organoid systems are discussed. Despite the promise organoids hold, challenges such as standardization, reproducibility, and the complexity of data interpretation remain. The review also addresses the ideas of assessing tumor organoids (tumoroids) against established cancer hallmarks and the potential of studying intercellular cooperation within these models. Ultimately, we propose that organoids, particularly when personalized for patient-specific applications, could revolutionize drug screening and therapeutic approaches, minimizing the reliance on traditional animal models and enhancing the precision of clinical interventions.
Collapse
Affiliation(s)
- Rushikesh Mukhare
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Khushboo A. Gandhi
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Anushree Kadam
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Aishwarya Raja
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Ankita Singh
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Mrudula Madhav
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Rohan Chaubal
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Surgical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Shwetali Pandey
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Sudeep Gupta
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| |
Collapse
|
31
|
Loveless TB, Carlson CK, Dentzel Helmy CA, Hu VJ, Ross SK, Demelo MC, Murtaza A, Liang G, Ficht M, Singhai A, Pajoh-Casco MJ, Liu CC. Open-ended molecular recording of sequential cellular events into DNA. Nat Chem Biol 2025; 21:512-521. [PMID: 39543397 PMCID: PMC11952980 DOI: 10.1038/s41589-024-01764-5] [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: 07/07/2023] [Accepted: 09/29/2024] [Indexed: 11/17/2024]
Abstract
Genetically encoded DNA recorders noninvasively convert transient biological events into durable mutations in a cell's genome, allowing for the later reconstruction of cellular experiences by DNA sequencing. We present a DNA recorder, peCHYRON, that achieves high-information, durable, and temporally resolved multiplexed recording of multiple cellular signals in mammalian cells. In each step of recording, prime editor, a Cas9-reverse transcriptase fusion protein, inserts a variable triplet DNA sequence alongside a constant propagator sequence that deactivates the previous and activates the next step of insertion. Insertions accumulate sequentially in a unidirectional order, editing can continue indefinitely, and high information is achieved by coexpressing a variety of prime editing guide RNAs (pegRNAs), each harboring unique triplet DNA sequences. We demonstrate that the constitutive expression of pegRNA collections generates insertion patterns for the straightforward reconstruction of cell lineage relationships and that the inducible expression of specific pegRNAs results in the accurate recording of exposures to biological stimuli.
Collapse
Affiliation(s)
- Theresa B Loveless
- Department of Biomedical Engineering, University of California, Irvine, CA, USA.
- Center for Synthetic Biology, University of California, Irvine, CA, USA.
- NSF-Simons Center for Multiscale Cell Fate, University of California, Irvine, CA, USA.
- Department of BioSciences, Rice University, Houston, TX, USA.
| | - Courtney K Carlson
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
| | - Catalina A Dentzel Helmy
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Vincent J Hu
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
- Graduate Program in Mathematical, Computational and Systems Biology, University of California, Irvine, CA, USA
| | - Sara K Ross
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Matt C Demelo
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Ali Murtaza
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Guohao Liang
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
| | - Michelle Ficht
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
| | - Arushi Singhai
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
| | - Marcello J Pajoh-Casco
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
- Center for Synthetic Biology, University of California, Irvine, CA, USA
| | - Chang C Liu
- Department of Biomedical Engineering, University of California, Irvine, CA, USA.
- Center for Synthetic Biology, University of California, Irvine, CA, USA.
- NSF-Simons Center for Multiscale Cell Fate, 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
|
32
|
Liu P, Ponnienselvan K, Nyalile T, Oikemus S, Joynt AT, Iyer S, Kelly K, Guo D, Kyawe PP, Vanderleeden E, Redick SD, Huang L, Chen Z, Lee JM, Schiffer CA, Harlan DM, Wang JP, Emerson CP, Lawson ND, Watts JK, Sontheimer EJ, Luban J, Wolfe SA. Increasing intracellular dNTP levels improves prime editing efficiency. Nat Biotechnol 2025; 43:539-544. [PMID: 39322763 DOI: 10.1038/s41587-024-02405-x] [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: 10/09/2023] [Accepted: 08/27/2024] [Indexed: 09/27/2024]
Abstract
In primary cell types, intracellular deoxynucleotide triphosphate (dNTP) levels are tightly regulated in a cell cycle-dependent manner. We report that prime editing efficiency is increased by mutations that improve the enzymatic properties of Moloney murine leukemia virus reverse transcriptase and treatments that increase intracellular dNTP levels. In combination, these modifications produce substantial increases in precise editing rates.
Collapse
Affiliation(s)
- Pengpeng Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Karthikeyan Ponnienselvan
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Thomas Nyalile
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sarah Oikemus
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Anya T Joynt
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sukanya Iyer
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Karen Kelly
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dongsheng Guo
- Department of Neurology, Wellstone Program, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Pyae P Kyawe
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Emma Vanderleeden
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sambra D Redick
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lei Huang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zexiang Chen
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeong Min Lee
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - David M Harlan
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jennifer P Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Charles P Emerson
- Department of Neurology, Wellstone Program, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nathan D Lawson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jonathan K Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Erik J Sontheimer
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
33
|
Li F, Yu J, Wang P, Li T, Tang Q, Zhu J. Bridge RNA-Guided Genetic Recombination Tools for Treating Neurodegenerative Nucleotide Repeat Disorders. Neurosci Bull 2025; 41:734-736. [PMID: 39966309 PMCID: PMC11979011 DOI: 10.1007/s12264-025-01358-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/06/2024] [Indexed: 02/20/2025] Open
Affiliation(s)
- Fengshi Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, National Key Laboratory of Brain Function and Brain Diseases, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Institute of Stem Cell Research and Clinical Translation, Fudan University, Shanghai, 200040, China
| | - Jingyu Yu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, National Key Laboratory of Brain Function and Brain Diseases, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Institute of Stem Cell Research and Clinical Translation, Fudan University, Shanghai, 200040, China
| | - Peng Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, National Key Laboratory of Brain Function and Brain Diseases, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Institute of Stem Cell Research and Clinical Translation, Fudan University, Shanghai, 200040, China
| | - Tianwen Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, National Key Laboratory of Brain Function and Brain Diseases, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Institute of Stem Cell Research and Clinical Translation, Fudan University, Shanghai, 200040, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, National Key Laboratory of Brain Function and Brain Diseases, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Institute of Stem Cell Research and Clinical Translation, Fudan University, Shanghai, 200040, China
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, National Center for Neurological Disorders, National Key Laboratory of Brain Function and Brain Diseases, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Institute of Stem Cell Research and Clinical Translation, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
34
|
Li G, Chen G, Yuan GH, Wei J, Ni Q, Wu J, Yang B, Yang L, Chen J. Specific and efficient RNA A-to-I editing through cleavage of an ADAR inhibitor. Nat Biotechnol 2025:10.1038/s41587-025-02591-2. [PMID: 40140558 DOI: 10.1038/s41587-025-02591-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 02/13/2025] [Indexed: 03/28/2025]
Abstract
RNA editing can be a promising therapeutic approach. However, ectopic expression of RNA editing enzymes has been shown to trigger off-target editing. Here we identified adenosine deaminase acting on RNA (ADAR) inhibitors (ADIs) that suppress the activity of the fused ADAR2 deamination domain (ADAR2DD). Using these specific ADIs, we develop an RNA transformer adenosine base editor (RtABE) with high specificity. Fusing ADI to ADAR2DD, RtABE remains inactive until it binds to its target site. After binding to the target site, ADI is cleaved from ADAR2DD, and RtABE becomes active. RtABE can induce efficient editing in broad sequence contexts, including UAN, AAN, CAN and GAN. Using an adeno-associated virus for delivery of RtABE enables therapeutic RNA correction and restoration of α-L-iduronidase activity in Hurler syndrome mice with no substantial off-target editing. RtABE is a specific and efficient RNA editing system with a broad scope that may be a better alternative to existing RNA editing tools.
Collapse
Affiliation(s)
- Guangye Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guo Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Guo-Hua Yuan
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jia Wei
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qingyang Ni
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jing Wu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bei Yang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Li Yang
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Jia Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
35
|
Qu C, Koskinen Holm C. Impact of a Heterozygous C1R R301P/WT Mutation on Collagen Metabolism and Inflammatory Response in Human Gingival Fibroblasts. Cells 2025; 14:479. [PMID: 40214433 PMCID: PMC11987961 DOI: 10.3390/cells14070479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Periodontal Ehlers-Danlos syndrome arising from heterozygous pathogenic mutation in C1R and/or C1S genes is an autosomal-dominant disorder characterized by early-onset periodontitis. Due to the difficulties in obtaining and culturing the patient-derived gingival fibroblasts, we established a model system by introducing a heterozygous C1RR301P/WT mutation into human TERT-immortalized gingival fibroblasts (hGFBs) to investigate its specific effects on collagen metabolism and inflammatory responses. A heterozygous C1RR301P/WT mutation was introduced into hGFBs using engineered prime editing. The functional consequences of this mutation were assessed at cellular, molecular, and enzymatic levels using a variety of techniques, including cell growth analysis, collagen deposition quantification, immunocytochemistry, enzyme-linked immunosorbent assay, and quantitative real-time reverse transcription polymerase chain reaction. The C1RR301P/WT-mutated hGFBs (mhGFBs) exhibited normal morphology and growth rate compared to wild-type hGFBs. However, mhGFBs displayed upregulated procollagen α1(V), MMP-1, and IL-6 mRNA expression while simultaneously downregulating collagen deposition and C1r protein levels. A modest accumulation of unfolded collagens was observed in mhGFBs. The mhGFBs exhibited a heightened inflammatory response, with a more pronounced increase in MMP-1 and IL-6 mRNA expression compared to TNF-α/IL-1β-stimulated hGFBs. Unlike cytokine-stimulated hGFBs, cytokine-stimulated mhGFB did not increase C1R, C1S, procollagen α1(III), and procollagen α1(V) mRNA expression. Our results suggest that the C1RR301P/WT mutation specifically disrupts collagen metabolism and inflammatory pathways in hGFBs, highlighting the mutation's role in these processes. While other cellular functions appear largely unaffected, these findings underscore the potential of targeting collagen metabolism and inflammation for therapeutic interventions in pEDS.
Collapse
Affiliation(s)
- Chengjuan Qu
- Department of Odontology, Umeå University, 90185 Umeå, Sweden;
| | | |
Collapse
|
36
|
Godbout K, Dugas M, Reiken SR, Ramezani S, Falle A, Rousseau J, Wronska AE, Lamothe G, Canet G, Lu Y, Planel E, Marks AR, Tremblay JP. Universal Prime Editing Therapeutic Strategy for RyR1-Related Myopathies: A Protective Mutation Rescues Leaky RyR1 Channel. Int J Mol Sci 2025; 26:2835. [PMID: 40243436 PMCID: PMC11988564 DOI: 10.3390/ijms26072835] [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: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
RyR1-related myopathies (RyR1-RMs) include a wide range of genetic disorders that result from mutations in the RYR1 gene. Pathogenic variants lead to defective intracellular calcium homeostasis and muscle dysfunction. Fixing intracellular calcium leaks by stabilizing the RyR1 calcium channel has been identified as a promising therapeutic target. Gene therapy via prime editing also holds great promise as it can cure diseases by correcting genetic mutations. However, as more than 700 variants have been identified in the RYR1 gene, a universal treatment would be a more suitable solution for patients. Our investigation into the RyR1-S2843A mutation has yielded promising results. Using a calcium leak assay, we determined that the S2843A mutation was protective when combined with pathogenic mutations and significantly reduced the Ca2+ leak of the RyR1 channel. Our study demonstrated that prime editing can efficiently introduce the protective S2843A mutation. In vitro experiments using the RNA electroporation of the prime editing components in human myoblasts achieved a 31% introduction of this mutation. This article lays the foundation for a new therapeutic approach for RyR1-RM, where a unique once-in-a-lifetime prime editing treatment could potentially be universally applied to all patients with a leaky RyR1 channel.
Collapse
Affiliation(s)
- Kelly Godbout
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Mathieu Dugas
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Steven R. Reiken
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (S.R.R.); (A.E.W.); (A.R.M.)
| | - Sina Ramezani
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Alexia Falle
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Joël Rousseau
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Anetta E. Wronska
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (S.R.R.); (A.E.W.); (A.R.M.)
| | - Gabriel Lamothe
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Geoffrey Canet
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Yaoyao Lu
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Emmanuel Planel
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (S.R.R.); (A.E.W.); (A.R.M.)
| | - Jacques P. Tremblay
- Molecular Medicine Department, Laval University, Quebec, QC G1V 0A6, Canada; (M.D.); (S.R.); (A.F.); (G.L.); (G.C.); (Y.L.); (E.P.)
- CHU de Québec Research Center-Laval University, Quebec, QC G1V 4G2, Canada;
| |
Collapse
|
37
|
Liu D, Cao D, Han R. Recent advances in therapeutic gene-editing technologies. Mol Ther 2025:S1525-0016(25)00200-X. [PMID: 40119516 DOI: 10.1016/j.ymthe.2025.03.026] [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: 12/13/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The advent of gene-editing technologies, particularly CRISPR-based systems, has revolutionized the landscape of biomedical research and gene therapy. Ongoing research in gene editing has led to the rapid iteration of CRISPR technologies, such as base and prime editors, enabling precise nucleotide changes without the need for generating harmful double-strand breaks (DSBs). Furthermore, innovations such as CRISPR fusion systems with DNA recombinases, DNA polymerases, and DNA ligases have expanded the size limitations for edited sequences, opening new avenues for therapeutic development. Beyond the CRISPR system, mobile genetic elements (MGEs) and epigenetic editors are emerging as efficient alternatives for precise large insertions or stable gene manipulation in mammalian cells. These advances collectively set the stage for next-generation gene therapy development. This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.
Collapse
Affiliation(s)
- Dongqi Liu
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Di Cao
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Renzhi Han
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
38
|
Liu Y, Liu Y, Niu X, Chen A, Li Y, Yu Y, Mo B, Liu Z, Xu T, Cheng J, Wu Z, Wei W. Massively parallel interrogation of human functional variants modulating cancer immunosurveillance. Signal Transduct Target Ther 2025; 10:88. [PMID: 40102418 PMCID: PMC11920242 DOI: 10.1038/s41392-025-02171-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Anti-PD-1/PD-L1 immune checkpoint blockade (ICB) therapy has revolutionized clinical cancer treatment, while abnormal PD-L1 or HLA-I expression in patients can significantly impact the therapeutic efficacy. Somatic mutations in cancer cells that modulate these critical regulators are closely associated with tumor progression and ICB response. However, a systematic interpretation of cancer immune-related mutations is still lacking. Here, we harnessed the ABEmax system to establish a large-scale sgRNA library encompassing approximately 820,000 sgRNAs that target all feasible serine/threonine/tyrosine residues across the human genome, which systematically unveiled thousands of novel mutations that decrease or augment PD-L1 or HLA-I expression. Beyond residues associated with phosphorylation events, our screens also identified functional mutations that affect mRNA or protein stability, DNA binding capacity, protein-protein interactions, and enzymatic catalytic activity, leading to either gene inactivation or activation. Notably, we uncovered certain mutations that concurrently modulate PD-L1 and HLA-I expression, represented by the clinically relevant mutation SETD2_Y1666. We demonstrated that this mutation induces consistent phenotypic effects across multiple cancer cell lines and enhances the efficacy of immunotherapy in different tumor models. Our findings provide an unprecedented resource of functional residues that regulate cancer immunosurveillance, offering valuable guidance for clinical diagnosis, ICB therapy, and the development of innovative drugs for cancer treatment.
Collapse
Affiliation(s)
- Ying Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuran Niu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Ang Chen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yizhou Li
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Ying Yu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Binrui Mo
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Zhiheng Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Tao Xu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Jie Cheng
- Department of pathology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zeguang Wu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China.
- Changping Laboratory, Beijing, China.
| |
Collapse
|
39
|
Liu J, Zhang J, Zhao T, Zhao M, Su M, Chen Y, Huang Z, Wang Y, Zhong C, Hu Z, Zhou P, Tian R, He D. SunTag-PE: a modular prime editing system enables versatile and efficient genome editing. Commun Biol 2025; 8:452. [PMID: 40097588 PMCID: PMC11914589 DOI: 10.1038/s42003-025-07893-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Prime editing (PE) holds tremendous potential in the treatment of genetic diseases because it can install any desired base substitution or local insertion/deletion. However, the full-length PE effector size (6.3-kb) is beyond the packaging capacity of adeno-associated virus (AAV), hindering its clinical translation. Various splitting strategies have been used to improve its delivery, but always accompanied by compromised PE efficiency. Here, we developed a modular and efficient SunTag-PE system that splits PE effectors into GCN4-nCas9 and single-chain variable fragment (scFv) tethered reverse transcriptase (RT). We observed that SunTag-PEs with 1×GCN4 in the N terminus of nCas9 was the most efficient configuration rather than multiple copies of GCN4. This SunTag-PE strategy achieved editing levels comparable to canonical fused-PE (nCas9 and RT are linked together) and higher than other split-PE strategies (including sPE and MS2-PE) in both PE2 and PE3 forms with no increase in insertion and deletion (indel) byproducts. Moreover, we successfully validated the modularity of SunTag-PE system in the Cas9 orthologs of SauCas9 and FrCas9. Finally, we employed dual AAVs to deliver SunTag-ePE3 and efficiently corrected the pathogenic mutation in HBB mutant cell line. Collectively, our SunTag-PE system provides an efficient modular splitting strategy for prime editing and further facilitate its transformation in clinics.
Collapse
Affiliation(s)
- Jiashuo Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingjing Zhang
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | | | - Mengya Zhao
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Su
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ye Chen
- Department of Gynecological oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zheying Huang
- Department of Gynecological oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuyan Wang
- Department of Gynecological oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | | | - Zheng Hu
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Ping Zhou
- Department of Obstetrics and Gynecology, Dongguan Maternal and Child Health Care Hospital, Dongguan, China.
| | - Rui Tian
- Generulor Co., Ltd. Zhuhai, Guangdong, China.
| | - Dan He
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
40
|
Ramadoss GN, Namaganda SJ, Hamilton JR, Sharma R, Chow KG, Macklin BL, Sun M, Liu JC, Fellmann C, Watry HL, Jin J, Perez BS, Espinoza CRS, Matia MP, Lu SH, Judge LM, Nussenzweig A, Adamson B, Murthy N, Doudna JA, Kampmann M, Conklin BR. Neuronal DNA repair reveals strategies to influence CRISPR editing outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.25.600517. [PMID: 38979269 PMCID: PMC11230251 DOI: 10.1101/2024.06.25.600517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Genome editing is poised to revolutionize treatment of genetic diseases, but poor understanding and control of DNA repair outcomes hinders its therapeutic potential. DNA repair is especially understudied in nondividing cells like neurons, which must withstand decades of DNA damage without replicating. This lack of knowledge limits the efficiency and precision of genome editing in clinically relevant cells. To address this, we used induced pluripotent stem cells (iPSCs) and iPSC-derived neurons to examine how postmitotic human neurons repair Cas9-induced DNA damage. We discovered that neurons can take weeks to fully resolve this damage, compared to just days in isogenic iPSCs. Furthermore, Cas9-treated neurons upregulated unexpected DNA repair genes, including factors canonically associated with replication. Manipulating this response with chemical or genetic perturbations allowed us to direct neuronal repair toward desired editing outcomes. By studying DNA repair in postmitotic human cells, we uncovered unforeseen challenges and opportunities for precise therapeutic editing.
Collapse
Affiliation(s)
- Gokul N Ramadoss
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
| | | | - Jennifer R Hamilton
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Rohit Sharma
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | | | | | - Mengyuan Sun
- Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Jia-Cheng Liu
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Christof Fellmann
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
| | | | - Julianne Jin
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
| | - Barbara S Perez
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Cindy R Sandoval Espinoza
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
| | | | - Serena H Lu
- Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Luke M Judge
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA, 94158, USA
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Britt Adamson
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Niren Murthy
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Jennifer A Doudna
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, 94720, USA
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA
- MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, 94158, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
- Department of Medicine, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
41
|
Saxena S, Kabra M, Abdeen A, Sinha D, Zhu M, Xie R, Hanstad G, Zepeda MAF, Gamm DM, Pattnaik BR, Gong S, Saha K. Genome-Wide CRISPR Screening Identifies Cellular Factors Controlling Nonviral Genome Editing Efficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642795. [PMID: 40161775 PMCID: PMC11952466 DOI: 10.1101/2025.03.12.642795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
After administering genome editors, their efficiency is limited by a multi-step process involving cellular uptake, trafficking, and nuclear import of the vector and its payload. These processes vary widely across cell types and differ depending on the nature and structure of the vector, whether it is a lipid nanoparticle or a different synthetic material. We developed a novel genome-wide CRISPR screening strategy to better understand these limitations within human cells to identify genes modulating cellular uptake, payload delivery, and gene editing efficiency. Our screen interrogates the cellular processes controlling genome editing by Cas-based nuclease and base editing strategies in human cells. We designed a genome-wide screen targeting 19,114 genes in HEK293 cells, and we identified six genes whose knockout increased nonviral editing efficiency in human cells by up to five-fold. Further validation through arrayed knockouts of the top hits from our screen boosted the editing efficiency from 5% to 50% when Cas9 was delivered via lipid-based nanoparticles. By designing the guides to target the screen library cassette, we could accurately track the library sgRNA identity and the editing outcome on the same amplicon via short-read sequencing, enabling the identification of rare outcomes via 'computationally' sorting edited from unedited cells within a heterogenous pool of >200M cells. In patient-derived human retinal pigment epithelium cells derived from pluripotent stem cells, BET1L, GJB2, and MS4A13 gene knockouts increased targeted genome editing by over five-fold. We anticipate that this high-throughput screening approach will facilitate the systematic engineering of novel nonviral genome editing delivery methods, where the identified novel gene hits can be further used to increase editing efficiency for other therapeutically relevant cell types.
Collapse
|
42
|
Lu C, Li Y, Cummings JR, Banskota S. Delivery of genome editors with engineered virus-like particles. Methods Enzymol 2025; 712:475-516. [PMID: 40121085 DOI: 10.1016/bs.mie.2025.01.007] [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: 03/25/2025]
Abstract
Genome editing technologies have revolutionized biomedical sciences and biotechnology. However, their delivery in vivo remains one of the major obstacles for clinical translation. Here, we introduce various emerging genome editing systems and review different delivery systems have been developed to realize the promise of in vivo gene editing therapies. In particular, we focus on virus-like particles (VLPs), an emerging delivery platform and provide in depth analysis on recent advancements to improve VLPs delivery potential and highlight opportunities for future improvements. To this end, we also provide detail workflows for engineered VLP (eVLP) selection, production, and purification, along with methods for characterization and validation.
Collapse
Affiliation(s)
- Christopher Lu
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Biological Design Center, Boston University, Boston, MA, United States
| | - Yuanhang Li
- Biological Design Center, Boston University, Boston, MA, United States; Department of Mechanical Engineering, Boston University, Boston, MA, United States
| | - Jacob Ryan Cummings
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Biological Design Center, Boston University, Boston, MA, United States
| | - Samagya Banskota
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Biological Design Center, Boston University, Boston, MA, United States.
| |
Collapse
|
43
|
Fan X, Lei Y, Wang L, Wu X, Li D. Advancing CRISPR base editing technology through innovative strategies and ideas. SCIENCE CHINA. LIFE SCIENCES 2025; 68:610-627. [PMID: 39231901 DOI: 10.1007/s11427-024-2699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
The innovation of CRISPR/Cas gene editing technology has developed rapidly in recent years. It is widely used in the fields of disease animal model construction, biological breeding, disease diagnosis and screening, gene therapy, cell localization, cell lineage tracking, synthetic biology, information storage, etc. However, developing idealized editors in various fields is still a goal for future development. This article focuses on the development and innovation of non-DSB editors BE and PE in the platform-based CRISPR system. It first explains the application of ideas for improvement such as "substitution", "combination", "adaptation", and "adjustment" in BE and PE development and then catalogues the ingenious inversions and leaps of thought reflected in the innovations made to CRISPR technology. It will then elaborate on the efforts currently being made to develop small editors to solve the problem of AAV overload and summarize the current application status of editors for in vivo gene modification using AAV as a delivery system. Finally, it summarizes the inspiration brought by CRISPR/Cas innovation and assesses future prospects for development of an idealized editor.
Collapse
Affiliation(s)
- Xiongwei Fan
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yang Lei
- Shanghai Frontiers Science Research Base 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
| | - Liren Wang
- Shanghai Frontiers Science Research Base 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.
| | - Xiushan Wu
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou, 510100, China.
| | - Dali Li
- Shanghai Frontiers Science Research Base 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
|
44
|
Hamze JG, Cambra JM, Navarro-Serna S, Martinez-Serrano CA. Navigating gene editing in porcine embryos: Methods, challenges, and future perspectives. Genomics 2025; 117:111014. [PMID: 39952413 DOI: 10.1016/j.ygeno.2025.111014] [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: 11/03/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Gene editing technologies, particularly CRISPR/Cas9, have emerged as transformative tools in genetic modification, significantly advancing the use of porcine embryos in biomedical and agricultural research. This review comprehensively examines the various methodologies for gene editing and delivery methods, such as somatic cell nuclear transfer (SCNT), microinjection, electroporation, and lipofection. This review, focuses on the advantages or limitations of using different biological sources (in vivo- vs. in vitro oocytes/embryos). Male germ cell manipulation using sperm-mediated gene transfer (SMGT) and testis-mediated gene transfer (TMGT) represent innovative approaches for producing genetically modified animals. Although these technologies have revolutionized the genetic engineering field, all these strategies face challenges, including high rates of off-target events and mosaicism. This review emphasizes the need to refine these methods, with a focus on reducing mosaicism and improving editing accuracy. Further advancements are essential to unlocking the full potential of gene editing for both agricultural applications and biomedical innovations.
Collapse
Affiliation(s)
- Julieta G Hamze
- Department of Cell Biology and Histology, Faculty of Medicine, University of Murcia, Murcia, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.
| | - Josep M Cambra
- Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany.
| | | | - Cristina A Martinez-Serrano
- Department of Biotechnology, National Institute for Agriculture and Food Research and Technology (INIA-CSIC), Madrid, Spain.
| |
Collapse
|
45
|
Askary A, Chen W, Choi J, Du LY, Elowitz MB, Gagnon JA, Schier AF, Seidel S, Shendure J, Stadler T, Tran M. The lives of cells, recorded. Nat Rev Genet 2025; 26:203-222. [PMID: 39587306 DOI: 10.1038/s41576-024-00788-w] [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] [Accepted: 09/26/2024] [Indexed: 11/27/2024]
Abstract
A paradigm for biology is emerging in which cells can be genetically programmed to write their histories into their own genomes. These records can subsequently be read, and the cellular histories reconstructed, which for each cell could include a record of its lineage relationships, extrinsic influences, internal states and physical locations, over time. DNA recording has the potential to transform the way that we study developmental and disease processes. Recent advances in genome engineering are driving the development of systems for DNA recording, and meanwhile single-cell and spatial omics technologies increasingly enable the recovery of the recorded information. Combined with advances in computational and phylogenetic inference algorithms, the DNA recording paradigm is beginning to bear fruit. In this Perspective, we explore the rationale and technical basis of DNA recording, what aspects of cellular biology might be recorded and how, and the types of discovery that we anticipate this paradigm will enable.
Collapse
Affiliation(s)
- Amjad Askary
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Wei Chen
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Junhong Choi
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lucia Y Du
- Biozentrum, University of Basel, Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Michael B Elowitz
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, USA.
| | - James A Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Alexander F Schier
- Biozentrum, University of Basel, Basel, Switzerland.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Sophie Seidel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Martin Tran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
46
|
Murray JB, Harrison PT, Scholefield J. Prime editing: therapeutic advances and mechanistic insights. Gene Ther 2025; 32:83-92. [PMID: 39609594 PMCID: PMC11946880 DOI: 10.1038/s41434-024-00499-1] [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: 04/19/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024]
Abstract
We are often confronted with a simple question, "which gene editing technique is the best?"; the simple answer is "there isn't one". In 2021, a year after prime editing first made its mark, we evaluated the landscape of this potentially transformative advance in genome engineering towards getting treatments to the clinic [1]. Nearly 20% of the papers we cited were still in pre-print at the time which serves to indicate how early-stage the knowledge base was at that time. Now, three years later, we take a look at the landscape and ask what has been learnt to ensure this tech is broadly accessible, highlighting some key advances, especially those that push this towards the clinic. A big part of the appeal of prime editing is its ability to precisely edit DNA without double stranded breaks, and to install any of the 12 possible single-nucleotide conversion events as well as small insertions and/or deletions, or essentially any combination thereof. Over the last few decades, other transformative and Nobel prize-winning technologies that rely on Watson-Crick base-pairing such as PCR, site-directed mutagenesis, RNA interference, and one might say, "classic" CRISPR, were swiftly adopted across labs around the world because of the speed with which mechanistic rules governing their efficiency were determined. Whilst this perspective focuses on the context of gene therapy applications of prime editing, we also further look at the recent studies which have increased our understanding of the mechanism of PEs and simultaneously improved the efficiency and diversity of the PE toolbox.
Collapse
Affiliation(s)
- Joss B Murray
- Department of Physiology, University College Cork, Cork, Ireland
| | - Patrick T Harrison
- Department of Physiology, University College Cork, Cork, Ireland.
- Division of Pulmonary Medicine, Cincinnati Children's Hospital, Cincinnati, OH, USA.
| | - Janine Scholefield
- Bioengineering and Integrated Genomics, NextGen Health, CSIR, Pretoria, South Africa.
| |
Collapse
|
47
|
Oliynyk RT, Church GM. Circular Vectors as an efficient, fully synthetic, cell-free approach for preparing small circular DNA as a plasmid substitute for guide RNA expression in CRISPR-Cas9 genome editing. Nat Protoc 2025:10.1038/s41596-024-01138-0. [PMID: 39994413 DOI: 10.1038/s41596-024-01138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/19/2024] [Indexed: 02/26/2025]
Abstract
Robust expression of guide RNA (gRNA) is essential for successful implementation of CRISPR-Cas9 genome-editing methods. The gRNA components, such as an RNA polymerase promoter followed by the gRNA coding sequence and an RNA polymerase terminator sequence, and the Cas9 protein are expressed either via an all-in-one plasmid or separate dedicated plasmids. The preparation of such plasmids involves a laborious multi-day process of DNA assembly, bacterial cloning, validation, purification and sequencing. Our Circular Vector (CV) protocol introduces an efficient, fully synthetic, cell-free approach for preparing gRNA expression templates suitable for transfection, marking a significant advancement over traditional plasmid-based approaches. This protocol consists of the circularization and purification of linear double-stranded DNA (dsDNA) containing gRNA expression elements into compact, bacterial-backbone-free circular DNA expression vectors in as little as 3 h. We provide a guide to the design of the dsDNA template coding for gRNA elements for CRISPR-Cas9 base and prime editing, along with step-by-step instructions for the efficient preparation of gRNA-expressing CVs. In addition to rapid preparation, CVs created via this protocol offer several key advantages: a compact size, absence of a bacterial backbone, absence of bacterial endotoxins and no contamination by bacterial RNA or DNA fragments. These features make gRNA-expressing CVs a superior choice over plasmid-based gRNA expression templates.
Collapse
Affiliation(s)
- Roman Teo Oliynyk
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Computer Science, University of Auckland, Auckland, New Zealand.
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| |
Collapse
|
48
|
Kanno S, Sato K, Nakazawa T. Precise Generation of Human Induced Pluripotent Stem Cell-derived Cell Lines Harboring Disease-relevant Single Nucleotide Variants Using a Prime Editing System. Bio Protoc 2025; 15:e5191. [PMID: 40028019 PMCID: PMC11865833 DOI: 10.21769/bioprotoc.5191] [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: 10/27/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 03/05/2025] Open
Abstract
Human induced pluripotent stem (iPS) cell lines harboring mutations in disease-related genes serve as invaluable in vitro models for unraveling disease mechanisms and accelerating drug discovery efforts. Introducing mutations into iPS cells using traditional gene editing approaches based on the CRISPR-Cas9 endonuclease often encounters challenges such as unintended insertions/deletions (indels) and off-target effects. To address these limitations, we present a streamlined protocol for introducing highly accurate gene mutations into human iPS cells using prime editing, a "search-and-replace" genome-editing technology that combines unwanted indel-minimized CRISPR-Cas9 nickase with reverse transcriptase. This protocol encompasses the design of prime editing guide RNAs (pegRNAs) required for binding and replacement at target loci, construction of prime editor and pegRNA expression vectors, gene transfer into iPS cells, and cell line selection. This protocol allows for the efficient establishment of disease-associated gene variants within 6-8 weeks while preserving critical genomic context. Key features • Dramatic improvement in efficiency of In-Fusion cloning using inserts assembled from the three pegRNA components (spacer, spCas9 scaffold, and 3' extension) via overlap extension PCR. • Cost-effective and time-saving selection of pegRNAs for prime editing via bulk Sanger sequencing. • Straightforward gene transfection using polymer-based reagents, which requires no specialized equipment or techniques and offers high reproducibility and broad applicability across different cell lines. • Precise genome editing based on pegRNA/prime editing minimizes off-target effects, enabling a wide range of applications in the study of disease-associated genetic variants. Graphical overview Key steps of generation of human induced pluripotent stem (iPS) cell lines harboring disease-relevant single nucleotide variants (SNVs) using a prime editing system.
Collapse
Affiliation(s)
- Seiya Kanno
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
49
|
Zhang H, Ji Q. Prime editing in bacteria with BacPE. Methods Enzymol 2025; 712:405-418. [PMID: 40121080 DOI: 10.1016/bs.mie.2025.01.026] [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: 03/25/2025]
Abstract
Programmable genome editing technologies have revolutionized the ability of researchers to alter the genomes of microorganisms in a straightforward and efficient manner, significantly advancing the field of microbiology. To date, several CRISPR-Cas-based genome-editing systems have been developed for use in E. coli, including CRISPR/Cas9, base editing, and prime editing technologies. In this chapter, we describe the design and experimental application of BacPE, a variant of prime editing technology optimized for E. coli. BacPE facilitates the introduction of point mutations, insertions, and deletions without the need for double-strand DNA breaks. We demonstrate that BacPE is a powerful tool for genome editing in E. coli and highlight its potential applicability to other bacterial species.
Collapse
Affiliation(s)
- Hongyuan Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, P.R. China
| | - Quanjiang Ji
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, P.R. China; Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, P.R. China; Shanghai Clinical Research and Trial Center, Shanghai, P.R. China.
| |
Collapse
|
50
|
Chen PR, Wei Y, Li X, Yu HY, Wang SG, Yuan XZ, Xia PF. Precision engineering of the probiotic Escherichia coli Nissle 1917 with prime editing. Appl Environ Microbiol 2025; 91:e0003125. [PMID: 39887239 PMCID: PMC11837520 DOI: 10.1128/aem.00031-25] [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: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
CRISPR-Cas systems are transforming precision medicine with engineered probiotics as next-generation diagnostics and therapeutics. To promote human health and treat disease, engineering probiotic bacteria demands maximal versatility to enable non-natural functionalities while minimizing undesired genomic interferences. Here, we present a streamlined prime editing approach tailored for probiotic Escherichia coli Nissle 1917 utilizing only essential genetic modules, including Cas9 nickase from Streptococcus pyogenes, a codon-optimized reverse transcriptase, and a prime editing guide RNA, and an optimized workflow with longer induction. As a result, we achieved all types of prime editing in every individual round of experiments with efficiencies of 25.0%, 52.0%, and 66.7% for DNA deletion, insertion, and substitution, respectively. A comprehensive evaluation of off-target effects revealed a significant reduction in unintended mutations, particularly in comparison to two different base editing methods. Leveraging the prime editing system, we inserted a unique DNA sequence to barcode the edited strain and established an antibiotic-resistance-gene-free platform to enable non-natural functionalities. Our prime editing strategy presents a CRISPR-Cas system that can be readily implemented in any laboratories with the basic CRISPR setups, paving the way for future innovations in engineered probiotics.IMPORTANCEOne ultimate goal of gene editing is to introduce designed DNA variations at specific loci in living organisms with minimal unintended interferences in the genome. Achieving this goal is especially critical for creating engineered probiotics as living diagnostics and therapeutics to promote human health and treat diseases. In this endeavor, we report a customized prime editing system for precision engineering of probiotic Escherichia coli Nissle 1917. With such a system, we developed a barcoding system for tracking engineered strains, and we built an antibiotic-resistance-gene-free platform to enable non-natural functionalities. We provide not only a powerful gene editing approach for probiotic bacteria but also new insights into the advancement of innovative CRISPR-Cas systems.
Collapse
Affiliation(s)
- Pei-Ru Chen
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| | - Ying Wei
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| | - Xin Li
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| | - Hai-Yan Yu
- State Key Laboratory of Microbial Technology, 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
| | - Xian-Zheng Yuan
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
- Sino-French Research Institute for Ecology and Environment, Shandong University, Qingdao, China
| | - Peng-Fei Xia
- School of Environmental Science and Engineering, Shandong University, Qingdao, China
| |
Collapse
|