1
|
Moyo B, Brown LBC, Khondaker II, Bao G. Engineering adeno-associated viral vectors for CRISPR/Cas based in vivo therapeutic genome editing. Biomaterials 2025; 321:123314. [PMID: 40203649 DOI: 10.1016/j.biomaterials.2025.123314] [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: 12/23/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
The recent approval of the first gene editing therapy for sickle cell disease and transfusion-dependent beta-thalassemia by the U.S. Food and Drug Administration (FDA) demonstrates the immense potential of CRISPR (clustered regularly interspaced short palindromic repeats) technologies to treat patients with genetic disorders that were previously considered incurable. While significant advancements have been made with ex vivo gene editing approaches, the development of in vivo CRISPR/Cas gene editing therapies has not progressed as rapidly due to significant challenges in achieving highly efficient and specific in vivo delivery. Adeno-associated viral (AAV) vectors have shown great promise in clinical trials as vehicles for delivering therapeutic transgenes and other cargos but currently face multiple limitations for effective delivery of gene editing machineries. This review elucidates these challenges and highlights the latest engineering strategies aimed at improving the efficiency, specificity, and safety profiles of AAV-packaged CRISPR/Cas systems (AAV-CRISPR) to enhance their clinical utility.
Collapse
Affiliation(s)
- Buhle Moyo
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Lucas B C Brown
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA; Graduate Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, 77030, USA
| | - Ishika I Khondaker
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Yu W, Wang Y, Li S, Dai Y, Li Y, Zhang X, Li B, Qian S, Zhang X, Bi C. Optimized dual-AAV base editor delivery system with enhanced editing efficiency and virion production titer. Synth Syst Biotechnol 2025; 10:697-706. [PMID: 40248483 PMCID: PMC12002711 DOI: 10.1016/j.synbio.2025.03.007] [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: 12/10/2024] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Base editors (BEs) are a promising tool for precise base conversion in human cells and animals, while the adeno-associated virus (AAV) is the major vector for human gene therapy. However, the size of the DNA cassette required for BE expression exceeds the 4.7 kb packing capacity of the AAV vector, making dual-AAV approaches based on trans-splicing intein necessary. Even with this approach, current split DNA cassettes are still larger than the AAV packing limit, posing a challenge for cellular production of AAV. Moreover, some split strategies yield variable editing results and target coverage. To address these limitations, 25 different split sets for BE4max and A3A-BE4max were tested at two target sites respectively, with splitting sites ranging from 493rd to 517th amino acids on the Cas9 peptide. Fortunately, the best Cas9 split site was identified between His511 and Ser512 and the arrangement of the AAV expression cassette was further manipulated to create evenly distributed CBE and ABE intein systems within 4.7 kb. These novel dual-AAV systems, designated 4.6AAV-CBE and 4.7AAV-ABE, were found to have base editing efficiencies similar to wild-type BEs, with a narrower editing window than the current 573 split system. Notably, 4.6AAV-CBE yield a higher AAV production titer, up to 2.1-fold in AAV-N and 1.5-fold in AAV-C, compared to the split-573BE system, likely due to the reduction of DNA cassette size within the AAV packaging capacity. Moreover, after packaging and infecting cells with AAV-N and AAV-C at the same volume and number of cells, the multiplicities of infection (MOI) and editing efficiency of 4.6 AAV-CBE were both higher than those of the split-573BE system. This study present advanced dual-AAV systems for ABE and CBE delivery, establishing a basis for safe and efficient BE therapies.
Collapse
Affiliation(s)
- Wenjia Yu
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| | - Yujie Wang
- Binzhou Medical University, Shandong, China
| | - Siwei Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| | - Yingcai Dai
- Shanghai Jiao Tong University, Shanghai, China
| | - Yucheng Li
- Tianjin University of Science and Technology, Tianjin, China
| | - Xinyue Zhang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| | - Bo Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| | - Siriguleng Qian
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
| | - Xueli Zhang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| | - Changhao Bi
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| |
Collapse
|
3
|
Cao X, Gao Z, Yin P, Wang H, Yang L. Crystal structure and inhibition mechanism of AcrIIA11. Biochem Biophys Res Commun 2025; 772:152073. [PMID: 40414010 DOI: 10.1016/j.bbrc.2025.152073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Anti-CRISPR (Acr) proteins are naturally evolved inhibitors that precisely target and suppress CRISPR-Cas systems, representing a sophisticated molecular arms race between bacteriophages and their bacterial hosts. While Class 1 systems dominate among sequenced prokaryotic genomes, Class 2 systems remain primary sources of editing tools. Here, we report the structural and mechanistic characterization of AcrIIA11, an anti-CRISPR protein that simultaneously inhibits Streptococcus pyogenes (SpyCas9) and Staphylococcus aureus Cas9 (SauCas9). The 3.2 Å crystal structure reveals a compact α/β fold with distinct electropositive clefts implicated in DNA binding. While DALI analysis identified structural homology to transcriptional regulators and the RecA inhibitor PsiB (RMSD 3.3 Å), functional studies established that AcrIIA11 forms stable ternary complexes with both Cas9 orthologs and sgRNA. Biochemical assays demonstrated stronger inhibition of SauCas9 compared to SpyCas9, with EMSA revealing a critical dichotomy: AcrIIA11 maintains SauCas9-sgRNA binding to specific target DNA while completely blocking cleavage activity. Computational docking localizes AcrIIA11 at the HNH-RuvC interface without obstructing DNA-binding channels in SauCas9, suggesting allosteric inhibition through HNH domain displacement. This work establishes AcrIIA11 as a dual-purpose Cas9 inhibitor that preserves target recognition while inactivating nuclease function-a mechanism with potential applications in precision CRISPR control.
Collapse
Affiliation(s)
- Xueli Cao
- State Key Laboratory of Green Biomanufacturing, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zirui Gao
- State Key Laboratory of Green Biomanufacturing, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Peipei Yin
- State Key Laboratory of Green Biomanufacturing, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China; College of Chemical and Biological Engineering, Yichun University, Yichun, 336000, China.
| | - Hao Wang
- State Key Laboratory of Green Biomanufacturing, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Lingguang Yang
- State Key Laboratory of Green Biomanufacturing, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China; College of Chemical and Biological Engineering, Yichun University, Yichun, 336000, China.
| |
Collapse
|
4
|
Diversi F, Dabin J, Mazza E, Rinaldin M, de Castro Reis F, Hackett JA, Heppenstall PA. Papillomavirus-like particles as vectors for ex vivo gene therapy of the skin. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102501. [PMID: 40171279 PMCID: PMC11960642 DOI: 10.1016/j.omtn.2025.102501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025]
Abstract
Ex vivo gene delivery to the skin utilizing retroviral vectors has been demonstrated to be a viable clinical option for replacement of defective genes. However, because these vectors integrate their cargo into the genome, safety issues arise when utilizing them to deliver gene-editing nucleases. Here, we explored the use of Papillomavirus, a non-integrating viral vector, for ex vivo skin gene editing, exploiting its natural tropism for basal keratinocytes. We demonstrated that Papillomavirus-like particles (PVLPs) can deliver a variety of DNA constructs encoding fluorophores, Cre recombinase, calcium indicators, Cas9, and short hairpin RNA (shRNA) to keratinocytes, offering advantages over other viral vectors such as adeno-associated virus (AAV) and Lentivirus. We further showed that PVLPs can be used for gene therapy for Olmsted syndrome, a genetic skin disease caused by a gain-of-function mutation in the Trpv3 gene. Specifically, PVLP-delivered SaCas9 and shRNA effectively disrupted the Trpv3 gene or reduced its expression, leading to decreased TRPV3 activity and mitigating the hyperactivity associated with Olmsted syndrome. Skin equivalents generated from PVLP-treated keratinocytes exhibited complete transduction, and PVLP-shRNA treatment significantly reduced hyperkeratosis in skin equivalents from mice bearing the Olmsted syndrome mutation. These findings highlight PVLP as a promising tool for ex vivo skin gene therapy.
Collapse
Affiliation(s)
- Francesco Diversi
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Via Bonomea 265, 34136 Trieste, Italy
| | - Juliette Dabin
- Epigenetics & Neurobiology Unit European Molecular Biology Laboratory (EMBL) Rome, Italy
| | - Elisa Mazza
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Via Bonomea 265, 34136 Trieste, Italy
| | - Mirko Rinaldin
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Via Bonomea 265, 34136 Trieste, Italy
| | - Fernanda de Castro Reis
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Via Bonomea 265, 34136 Trieste, Italy
| | - Jamie A. Hackett
- Epigenetics & Neurobiology Unit European Molecular Biology Laboratory (EMBL) Rome, Italy
| | - Paul A. Heppenstall
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Via Bonomea 265, 34136 Trieste, Italy
| |
Collapse
|
5
|
Khirallah J, Bloomer H, Wich D, Huang C, Workman JN, Li Y, Newby GA, Liu DR, Xu Q. In vivo base editing of Angptl3 via lipid nanoparticles to treat cardiovascular disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102486. [PMID: 40129501 PMCID: PMC11932681 DOI: 10.1016/j.omtn.2025.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 02/12/2025] [Indexed: 03/26/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally and is exacerbated by elevated blood levels of low-density lipoprotein cholesterol (LDL-C) and triglycerides (TGs). Existing approaches for decreasing blood lipid levels rely on daily medications, leading to poor patient adherence. Gene therapy represents a promising "one and done" strategy to durably reduce blood lipid levels. ANGPTL3 has emerged as a potential target for gene therapy, as naturally occurring loss-of-function variants are cardioprotective. Here, we use lipid nanoparticles to package and deliver CRISPR adenine base editors (ABEs), which enable gene knockout without requiring potentially harmful DNA double-strand breaks. We package ABE mRNA and a synthetic guide RNA targeted to disrupt an important splice site in Angptl3, which we administered to mice intravenously. We achieved over 60% base editing in the liver and durable reductions in serum ANGPTL3, LDL-C, and TGs for at least 100 days. Notably, blood lipid levels remained low when mice were challenged with a high-fat high-cholesterol diet up to 191 days after therapy. These results provide a foundation for a potential one-and-done treatment for CVD.
Collapse
Affiliation(s)
- Jennifer Khirallah
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Hanan Bloomer
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Douglas Wich
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Changfeng Huang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - J. Noah Workman
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Gregory A. Newby
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
6
|
Gameiro M, Almeida-Pinto J, Moura BS, Mano JF, Gaspar VM. Designer mammalian living materials through genetic engineering. Bioact Mater 2025; 48:135-148. [PMID: 40034809 PMCID: PMC11872553 DOI: 10.1016/j.bioactmat.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/18/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Emerging genome editing and synthetic biology toolboxes can accurately program mammalian cells behavior from the inside-out. Such engineered living units can be perceived as key building blocks for bioengineering mammalian cell-dense materials, with promising features to be used as living therapeutics for tissue engineering or disease modeling applications. Aiming to reach full control over the code that governs cell behavior, inside-out engineering approaches have potential to fully unlock user-defined living materials encoded with tailored cellular functionalities and spatial arrangements. Dwelling on this, herein, we discuss the most recent advances and opportunities unlocked by genetic engineering strategies, and on their use for the assembly of next-generation cell-rich or cell-based materials, with an unprecedent control over cellular arrangements and customizable therapeutic capabilities. We envision that the continuous synergy between inside-out and outside-in cell engineering approaches will potentiate the future development of increasingly sophisticated cell assemblies that may operate with augmented biofunctionalities.
Collapse
Affiliation(s)
- Mariana Gameiro
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - José Almeida-Pinto
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Beatriz S. Moura
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F. Mano
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Vítor M. Gaspar
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
7
|
Lin T, Wang X, Zhang Y, Li G, Huang X, Shi M. Developing safe and efficient CGBE editor based on Cas-embedding strategy. Synth Syst Biotechnol 2025; 10:504-510. [PMID: 40027845 PMCID: PMC11872432 DOI: 10.1016/j.synbio.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 03/05/2025] Open
Abstract
CGBE (C-to-G base editor) systems, pivotal components within the base editing arsenal, enable the precise conversion of cytosines to guanines. However, conventional cytidine deaminases possess non-specific single-stranded DNA binding properties, leading to off-target effects and safety concerns. The Cas-embedding strategy, which involves embedding functional proteins like deaminases within the Cas9 enzyme's architecture, emerges as a method to mitigate these off-target effects. Our study pioneers the application of the Cas-embedding strategy to CGBE systems, engineering a suite of novel CGBE editors, CE-CGBE. The CE-CGBE that incorporated eA3A, RBMX and Udgx excelled in editing efficiency, editing purity, and indel formation was named HF-CGBE. HF-CGBE showed no significant difference in off-target effects compared to the negative control group for both DNA and RNA. In summary, the novel HF-CGBE editors we propose expand the base editing toolbox and provide therapeutic approaches for related pathogenic mutations.
Collapse
Affiliation(s)
- Tian Lin
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Xin Wang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Zhang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200032 China
| | - Guanglei Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200032 China
| | - Xingxu Huang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200032 China
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| |
Collapse
|
8
|
Merdler-Rabinowicz R, Dadush A, Patiyal S, Rajagopal PS, Daya G, Ben-Aroya S, Schäffer A, Eisenberg E, Ruppin E, Levanon E. A systematic evaluation of the therapeutic potential of endogenous-ADAR editors in cancer prevention and treatment. NAR Cancer 2025; 7:zcaf016. [PMID: 40330550 PMCID: PMC12053386 DOI: 10.1093/narcan/zcaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/10/2025] [Accepted: 05/05/2025] [Indexed: 05/08/2025] Open
Abstract
Adenosine deaminases acting on RNA (ADAR) enzymes constitute a natural cellular mechanism that induces A-to-I(G) editing, introducing genetic changes at the RNA level. Recently, interest in the endogenous-ADAR editor has emerged for correcting genetic mutations, consisting of a programmed oligonucleotide that attracts the native ADAR, thereby offering opportunities for medical therapy. Here, we systematically chart the scope of cancer mutations that endogenous-ADAR can correct. First, analyzing germline single nucleotide variants in cancer predisposition genes, we find that endogenous-ADAR can revert a fifth of them, reducing the risk of cancer development later in life. Second, examining somatic mutations across various cancer types, we find that it has the potential to correct at least one driver mutation in over a third of the samples, suggesting a promising future treatment strategy. We also highlight key driver mutations that are amenable to endogenous-ADAR, and are thus of special clinical interest. As using endogenous-ADAR entails delivering relatively small payloads, the prospects of delivering endogenous-ADAR to various cancers seem promising. We expect that the large scope of correctable mutations that are systematically charted here for the first time will pave the way for a new era of cancer treatment options.
Collapse
Affiliation(s)
- Rona Merdler-Rabinowicz
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, 5290002, Israel
| | - Ariel Dadush
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, 5290002, Israel
| | - Sumeet Patiyal
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Padma Sheila Rajagopal
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Gulzar N Daya
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Shay Ben-Aroya
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Alejandro A Schäffer
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Eli Eisenberg
- Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel Aviv, 6997801, Israel
| | - Eytan Ruppin
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Erez Y Levanon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, 5290002, Israel
| |
Collapse
|
9
|
Matuszek Z, Arbab M, Kesavan M, Hsu A, Roy JCL, Zhao J, Yu T, Weisburd B, Newby GA, Doherty NJ, Wu M, Shibata S, Cristian A, Tao YA, Fearnley LG, Bahlo M, Rehm HL, Xie J, Gao G, Mouro Pinto R, Liu DR. Base editing of trinucleotide repeats that cause Huntington's disease and Friedreich's ataxia reduces somatic repeat expansions in patient cells and in mice. Nat Genet 2025:10.1038/s41588-025-02172-8. [PMID: 40419681 DOI: 10.1038/s41588-025-02172-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/18/2025] [Indexed: 05/28/2025]
Abstract
Trinucleotide repeat (TNR) diseases are neurological disorders caused by expanded genomic TNRs that become unstable in a length-dependent manner. The CAG•CTG sequence is found in approximately one-third of pathogenic TNR loci, including the HTT gene that causes Huntington's disease. Friedreich's ataxia, the most prevalent hereditary ataxia, results from GAA repeat expansion at the FXN gene. Here we used cytosine and adenine base editing to reduce the repetitiveness of TNRs in patient cells and in mice. Base editors introduced G•C>A•T and A•T>G•C interruptions at CAG and GAA repeats, mimicking stable, nonpathogenic alleles that naturally occur in people. AAV9 delivery of optimized base editors in Htt.Q111 Huntington's disease and YG8s Friedreich's ataxia mice resulted in efficient editing in transduced tissues, and significantly reduced repeat expansion in the central nervous system. These findings demonstrate that introducing interruptions in pathogenic TNRs can mitigate a key neurological feature of TNR diseases in vivo.
Collapse
Affiliation(s)
- Zaneta Matuszek
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Mandana Arbab
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Maheswaran Kesavan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Molecular Medicine Program, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| | - Alvin Hsu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jennie C L Roy
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Jing Zhao
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Tian Yu
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ben Weisburd
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gregory A Newby
- Department of Genetic Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Neil J Doherty
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Muzhou Wu
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Shota Shibata
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ana Cristian
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Y Allen Tao
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Liam G Fearnley
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Heidi L Rehm
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ricardo Mouro Pinto
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
10
|
Jiang B, An Z, Niu L, Qin D. Precise genome editing process and its applications in plants driven by AI. Funct Integr Genomics 2025; 25:109. [PMID: 40413357 DOI: 10.1007/s10142-025-01619-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/12/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Genome editing technologies have emerged as the keystone of biotechnological research, enabling precise gene modification. The field has evolved rapidly through revolutionary advancements, transitioning from early explorations to the breakthrough of the CRISPR-Cas system. The emergence of the CRISPR-Cas system represents a huge leap in genome editing, prompting the development of advanced tools such as base and prime editors, thereby enhancing precise genomic engineering capabilities. The rapid integration of AI across disciplines is now driving another transformative phase in genome editing, streamlining workflows and enhancing precision. The application prospects of genome editing technology are extensive, particularly in plant breeding, where it has already presented unparalleled opportunities for improving plant traits. Here, we review early genome editing technologies, including meganucleases, ZFNs, TALENs, and CRISPR-Cas systems. We also provide a detailed introduction to next-generation editing tools-such as base editors and prime editors-and their latest applications in plants. At the same time, we summarize and prospect the cutting-edge developments and future trends of genome editing technologies in combination with the rapidly rising AI technology, including optimizing editing systems, predicting the efficiency of editing sites and designing editing strategies. We are convinced that as these technologies progress and their utilization expands, they will provide pioneering solutions to global challenges, ushering in an era of health, prosperity, and sustainability.
Collapse
Affiliation(s)
- Bo Jiang
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
- Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, MOE, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
| | - Zeyu An
- University of Science and Technology Beijing, Beijing, 100083, China
| | - Linlin Niu
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
- Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, MOE, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
| | - Debin Qin
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China.
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China.
- Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, MOE, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China.
| |
Collapse
|
11
|
Ishiguro S, Ishida K, Sakata RC, Ichiraku M, Takimoto R, Yogo R, Kijima Y, Mori H, Tanaka M, King S, Tarumoto S, Tsujimura T, Bashth O, Masuyama N, Adel A, Toyoshima H, Seki M, Oh JH, Archambault AS, Nishida K, Kondo A, Kuhara S, Aburatani H, Klein Geltink RI, Yamamoto T, Shakiba N, Takashima Y, Yachie N. A multi-kingdom genetic barcoding system for precise clone isolation. Nat Biotechnol 2025:10.1038/s41587-025-02649-1. [PMID: 40399693 DOI: 10.1038/s41587-025-02649-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/20/2025] [Indexed: 05/23/2025]
Abstract
Cell-tagging strategies with DNA barcodes have enabled the analysis of clone size dynamics and clone-restricted transcriptomic landscapes in heterogeneous populations. However, isolating a target clone that displays a specific phenotype from a complex population remains challenging. Here we present a multi-kingdom genetic barcoding system, CloneSelect, which enables a target cell clone to be triggered to express a reporter gene for isolation through barcode-specific CRISPR base editing. In CloneSelect, cells are first stably tagged with DNA barcodes and propagated so that their subpopulation can be subjected to a given experiment. A clone that shows a phenotype or genotype of interest at a given time can then be isolated from the initial or subsequent cell pools stored during the experiment using CRISPR base editing. CloneSelect is scalable and compatible with single-cell RNA sequencing. We demonstrate the versatility of CloneSelect in human embryonic kidney 293T cells, mouse embryonic stem cells, human pluripotent stem cells, yeast cells and bacterial cells.
Collapse
Affiliation(s)
- Soh Ishiguro
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Rina C Sakata
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Minori Ichiraku
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Ren Takimoto
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Rina Yogo
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Yusuke Kijima
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Hideto Mori
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), The University of Osaka, Osaka, Japan
| | - Mamoru Tanaka
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Samuel King
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Shoko Tarumoto
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Taro Tsujimura
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Omar Bashth
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nanami Masuyama
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Japan
| | - Arman Adel
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Hiromi Toyoshima
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Motoaki Seki
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Ju Hee Oh
- BC Children's Hospital Research Institute, Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Anne-Sophie Archambault
- BC Children's Hospital Research Institute, Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Keiji Nishida
- Engineering Biology Research Center, Kobe University, Kobe, Japan
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Akihiko Kondo
- BC Children's Hospital Research Institute, Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- Engineering Biology Research Center, Kobe University, Kobe, Japan
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe, Japan
| | - Satoru Kuhara
- Graduate School of Bioresource and Bioenvironmental Sciences, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Aburatani
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Ramon I Klein Geltink
- BC Children's Hospital Research Institute, Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Nika Shakiba
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), The University of Osaka, Osaka, Japan
| | - Yasuhiro Takashima
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Nozomu Yachie
- School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), The University of Osaka, Osaka, Japan.
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
12
|
Chapdelaine-Trépanier V, Shenoy S, Masud W, Minju-Op A, Bérubé MA, Schönherr S, Forer L, Fradet-Turcotte A, Taliun D, Cuella-Martin R. CRISPR-BEasy: a free web-based service for designing sgRNA tiling libraries for CRISPR-dependent base editing screens. Nucleic Acids Res 2025:gkaf382. [PMID: 40377102 DOI: 10.1093/nar/gkaf382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
CRISPR-dependent base editing (BE) enables the modeling and correction of genetic mutations at single-base resolution. Base editing screens, where point mutations are queried en masse, are powerful tools to systematically draw genotype-phenotype associations and characterise the function of genes and other genomic elements. However, the lack of user-friendly web-based tools for designing base editing screens can hinder broad technology adoption. Here, we introduce CRISPR-BEasy (https://crispr-beasy.cerc-genomic-medicine.ca), a free, automated web-based server that streamlines the creation of single guide (sg)RNA tiling libraries for base editing screens. Researchers can provide their genes or genomic features of interest, their base editors of choice, and target sequences to act as positive and negative controls. The server designs and annotates sgRNA libraries by integrating custom code with publicly available tools such as crisprVerse and Ensembl's Variant Effect Predictor. CRISPR-BEasy provides downloadable results, including sgRNA on/off-target scores, predicted mutational outcomes per base editor, and intuitive interactive visualizations for data quality assessment. CRISPR-BEasy also provides a separate tool that assembles sgRNA libraries into oligonucleotides for cloning following the detailed protocol documented in the searchable web server manual. Together, CRISPR-BEasy ensures the seamless design of cloning-ready sgRNA libraries, seeking to democratise access to base editing screening technologies.
Collapse
Affiliation(s)
- Vincent Chapdelaine-Trépanier
- Department of Human Genetics, McGill University, Montreal, QC,H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC,H3A 0G1, Canada
| | - Shamika Shenoy
- Department of Human Genetics, McGill University, Montreal, QC,H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC,H3A 0G1, Canada
| | - Wardah Masud
- Department of Human Genetics, McGill University, Montreal, QC,H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC,H3A 0G1, Canada
| | - Amisha Minju-Op
- Department of Human Genetics, McGill University, Montreal, QC,H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC,H3A 0G1, Canada
| | - Marie-Anne Bérubé
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec City, QC,G1V 0A6, Canada
- Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval Research Centre, Québec City, QC,G1R 2J6, Canada
- Université Laval Cancer Research Center, Université Laval, Québec City, QC,G1R 3S3, Canada
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck,6020, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck,6020, Austria
| | - Amélie Fradet-Turcotte
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec City, QC,G1V 0A6, Canada
- Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval Research Centre, Québec City, QC,G1R 2J6, Canada
- Université Laval Cancer Research Center, Université Laval, Québec City, QC,G1R 3S3, Canada
| | - Daniel Taliun
- Department of Human Genetics, McGill University, Montreal, QC,H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC,H3A 0G1, Canada
| | - Raquel Cuella-Martin
- Department of Human Genetics, McGill University, Montreal, QC,H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC,H3A 0G1, Canada
| |
Collapse
|
13
|
Sheng H, Gao P, Yang C, Quilichini TD, Kochian LV, Datla R, Xiang D. Advances in Genome Editing Through Haploid Induction Systems. Int J Mol Sci 2025; 26:4779. [PMID: 40429922 PMCID: PMC12112045 DOI: 10.3390/ijms26104779] [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: 02/12/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Groundbreaking advances in gene editing technologies are transforming modern plant breeding by enabling precise genetic modifications that dramatically accelerate crop improvement. Haploid and diploid induction systems have emerged as particularly powerful tools in this landscape, offering both efficient gene editing capabilities and rapid production of homozygous lines while seamlessly integrating with the advanced genome-editing platforms such as CRISPR-Cas systems. This review synthesizes the current state of knowledge regarding the mechanisms, applications, and recent progress in haploid and diploid induction systems for gene editing. We examine their transformative potential for enhancing genetic gains and compressing breeding timelines, with significant implications for global food security. Additionally, we provide a critical analysis of emerging challenges of genome editing in crops and outline promising future directions for research and development.
Collapse
Affiliation(s)
- Huajin Sheng
- Department of Biology, College of Arts and Science, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (H.S.); (T.D.Q.)
| | - Peng Gao
- Agriculture and Agri-Food Canada, Saskatoon Research and Development Centre, 107 Science Place, Saskatoon, SK S7N 0X2, Canada;
| | - Changye Yang
- Aquatic and Crop Resource Development, National Research Council Canada, Saskatoon, SK S7N 0W9, Canada;
| | - Teagen D. Quilichini
- Department of Biology, College of Arts and Science, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (H.S.); (T.D.Q.)
| | - Leon V. Kochian
- Department of Plant Sciences, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada;
| | - Raju Datla
- Global Institute for Food Security, University of Saskatchewan, Saskatoon, SK S7N 4J8, Canada;
| | - Daoquan Xiang
- Aquatic and Crop Resource Development, National Research Council Canada, Saskatoon, SK S7N 0W9, Canada;
| |
Collapse
|
14
|
Gómez-Escribano AP, García-García G, Pérez-Santamarina E, Aller-Mañas E, Vázquez-Manrique RP, Millán-Salvador JM. Innovative therapies for inherited retinal dystrophies: navigating DNA, RNA, and protein approaches. EBioMedicine 2025; 116:105751. [PMID: 40367639 DOI: 10.1016/j.ebiom.2025.105751] [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: 03/30/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
Gene therapy has become a promising treatment for inherited retinal dystrophies (IRDs), but understanding the genetic mechanisms involved is essential for its success. Various approaches, such as gene augmentation and DNA/RNA-based therapies, have shown effectiveness in some clinical trials. However, gene augmentation is not effective for some dominant mutations and genome editing produces off-target effects. Here, we review safer and viable alternatives that are being evaluated in preclinical models and clinical trials to address this challenge. We propose a novel perspective based on protein-targeting therapies, which although promising, remain unexplored. We suggest that frameshift variants, which produce novel epitopes, may allow for the development of mutant protein targeting agents for selective protein degradation. This approach could be useful for dominant variants, where gene replacement is ineffective. By examining these approaches, we aim to guide more targeted and effective gene therapies for IRDs, offering potential treatments where current methods fall short.
Collapse
Affiliation(s)
- Ana Pilar Gómez-Escribano
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain.
| | - Gema García-García
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | | | - Elena Aller-Mañas
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - Rafael Pascual Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - José María Millán-Salvador
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain.
| |
Collapse
|
15
|
Kissling L, Mollaysa A, Janjuha S, Mathis N, Marquart KF, Weber Y, Moon WJ, Lin PJC, Fan SHY, Muramatsu H, Vadovics M, Allam A, Pardi N, Tam YK, Krauthammer M, Schwank G. Predicting adenine base editing efficiencies in different cellular contexts by deep learning. Genome Biol 2025; 26:115. [PMID: 40340964 PMCID: PMC12060317 DOI: 10.1186/s13059-025-03586-7] [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: 08/01/2024] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Adenine base editors (ABEs) enable the conversion of A•T to G•C base pairs. Since the sequence of the target locus influences base editing efficiency, efforts have been made to develop computational models that can predict base editing outcomes based on the targeted sequence. However, these models were trained on base editing datasets generated in cell lines and their predictive power for base editing in primary cells in vivo remains uncertain. RESULTS In this study, we conduct base editing screens using SpRY-ABEmax and SpRY-ABE8e to target 2,195 pathogenic mutations with a total of 12,000 guide RNAs in cell lines and in the murine liver. We observe strong correlations between in vitro datasets generated by ABE-mRNA electroporation into HEK293T cells and in vivo datasets generated by adeno-associated virus (AAV)- or lipid nanoparticle (LNP)-mediated nucleoside-modified mRNA delivery (Spearman R = 0.83-0.92). We subsequently develop BEDICT2.0, a deep learning model that predicts adenine base editing efficiencies with high accuracy in cell lines (R = 0.60-0.94) and in the liver (R = 0.62-0.81). CONCLUSIONS In conclusion, our work confirms that adenine base editing holds considerable potential for correcting a large fraction of pathogenic mutations. We also provide BEDICT2.0 - a robust computational model that helps identify sgRNA-ABE combinations capable of achieving high on-target editing with minimal bystander effects in both in vitro and in vivo settings.
Collapse
Affiliation(s)
- Lucas Kissling
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Amina Mollaysa
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Sharan Janjuha
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Nicolas Mathis
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Kim F Marquart
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Yanik Weber
- 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
| | - Ahmed Allam
- Department of Quantitative Biomedicine, University of 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, BC, Canada
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland.
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
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
|
17
|
Wang M, Krueger JB, Gilkey AK, Stelljes EM, Kluesner MG, Pomeroy EJ, Skeate JG, Slipek NJ, Lahr WS, Claudio Vázquez PN, Zhao Y, Bell JB, Clement K, Eaton EJ, Laoharawee K, Chang JW, Webber BR, Moriarity BS. Precision enhancement of CAR-NK cells through non-viral engineering and highly multiplexed base editing. J Immunother Cancer 2025; 13:e009560. [PMID: 40341025 PMCID: PMC12067936 DOI: 10.1136/jitc-2024-009560] [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: 04/26/2024] [Accepted: 03/20/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Natural killer (NK) cells' unique ability to kill transformed cells expressing stress ligands or lacking major histocompatibility complexes (MHC) has prompted their development for immunotherapy. However, NK cells have demonstrated only moderate responses against cancer in clinical trials. METHODS Advanced genome engineering may thus be used to unlock their full potential. Multiplex genome editing with CRISPR/Cas9 base editors (BEs) has been used to enhance T cell function and has already entered clinical trials but has not been reported in human NK cells. Here, we report the first application of BE in primary NK cells to achieve both loss-of-function and gain-of-function mutations. RESULTS We observed highly efficient single and multiplex base editing, resulting in significantly enhanced NK cell function in vitro and in vivo. Next, we combined multiplex BE with non-viral TcBuster transposon-based integration to generate interleukin-15 armored CD19 chimeric antigen receptor (CAR)-NK cells with significantly improved functionality in a highly suppressive model of Burkitt's lymphoma both in vitro and in vivo. CONCLUSIONS The use of concomitant non-viral transposon engineering with multiplex base editing thus represents a highly versatile and efficient platform to generate CAR-NK products for cell-based immunotherapy and affords the flexibility to tailor multiple gene edits to maximize the effectiveness of the therapy for the cancer type being treated.
Collapse
Affiliation(s)
- Minjing Wang
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joshua B Krueger
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexandria K Gilkey
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Erin M Stelljes
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mitchell G Kluesner
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily J Pomeroy
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph G Skeate
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Patricia N Claudio Vázquez
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yueting Zhao
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jason B Bell
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kendell Clement
- Department of Biomedical Informatics, The University of Utah, Salt Lake City, Utah, USA
| | - Ella J Eaton
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jae-Woong Chang
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
18
|
Sharma I, Hall K, Moonah S. CRISPR genome editing using a combined positive and negative selection system. PLoS One 2025; 20:e0321881. [PMID: 40327602 PMCID: PMC12054870 DOI: 10.1371/journal.pone.0321881] [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: 12/17/2024] [Accepted: 03/11/2025] [Indexed: 05/08/2025] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system is a powerful genome editing tool that has revolutionized research. Single nucleotide polymorphisms (SNPs) are the most common form of genetic variation in humans. Only a subset of these SNPs has been shown to be linked to genetic diseases, while the biological relevance of the majority remains unclear. Investigating these variants of unknown significance could provide valuable insights into their roles in biological processes, disease susceptibility, and treatment responses. While CRISPR/Cas has emerged as a transformative technology, its ability to make single nucleotide substitutions remains a significant limitation. Other techniques in single nucleotide editing, such as base editing and prime editing, offer promising possibilities to complement CRISPR/Cas systems, though they also have their own limitations. Hence, alternative approaches are necessary to overcome the limitations of CRISPR. Here, to improve the feasibility of generating single base edits in the genome, we provide a protocol that introduces a multiple expression and dual selection (MEDS) system, which, alongside CRISPR, utilizes the opposing roles of cytosine deaminase/uracil phosphoribosyltransferase (CD/UPRT) for negative selection and neomycin phosphotransferase II (NPT II) for positive selection. As a proof of concept and to demonstrate feasibility of the method, we used MEDS, along with traditional CRISPR-Cas9, to generate sickle hemoglobin by introducing a point mutation (A → T) in the sixth codon of the hemoglobin beta gene.
Collapse
Affiliation(s)
- Ishrya Sharma
- Department of Medicine, University of Florida, GainesvilleFlorida, United States of America
| | - Kerisa Hall
- Department of Medicine, University of Florida, GainesvilleFlorida, United States of America
| | - Shannon Moonah
- Department of Medicine, University of Florida, GainesvilleFlorida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, GainesvilleFlorida, United States of America
| |
Collapse
|
19
|
Amato P, Mikhalchenko A, Mitalipov S. The case for germline gene correction: state of the science. Fertil Steril 2025:S0015-0282(25)00253-5. [PMID: 40334730 DOI: 10.1016/j.fertnstert.2025.04.039] [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: 03/02/2025] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
Germline gene editing refers to altering the DNA in the reproductive cells (gametes or embryos). Germline gene editing experiments in human embryos have primarily focused on correcting genetic mutations linked to inherited diseases. This technology has the potential to prevent genetic disease before birth and in future generations. Advances in CRISPR-Cas9 and other gene-editing tools have accelerated scientific progress, raising both promise and safety and ethical concerns. A translational pathway for human heritable genome editing will require an approach that integrates scientific validation, ethical oversight, regulatory frameworks, and public engagement.
Collapse
Affiliation(s)
- Paula Amato
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon; Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, Portland, Oregon.
| | - Aleksei Mikhalchenko
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon
| | - Shoukhrat Mitalipov
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
20
|
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
|
21
|
Schürmann PJL, van Breda Vriesman SPE, Castro-Alpízar JA, Kooijmans SAA, Nieuwenhuis EES, Schiffelers RM, Fuchs SA. Therapeutic Application of mRNA for Genetic Diseases. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70019. [PMID: 40415711 DOI: 10.1002/wnan.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025]
Abstract
While gene therapy has been at the center of an active research field for decades, messenger RNA (mRNA) has long been considered unsuited for therapeutic application due to challenges with stability, immunogenicity, and delivery. Where gene therapy focuses on providing the desired genetic code, mRNA can directly provide the instructions encoded in the corresponding gene. This review aims to explore recent advances in mRNA therapies, building on the success of mRNA COVID-19 vaccines, and extend these insights to the potential treatment of rare genetic diseases. We follow the "outside-in" trajectory of mRNA therapies from administration to intracellular function, focusing on carrier systems such as lipid nanoparticles and virus-like particles, mRNA modifications, and the potential and challenges for clinical applications. To treat rare diseases, different approaches can be envisioned, including chronic or acute delivery of mRNAs encoding functional enzymes for enzyme deficiencies and delivery of CRISPR/Cas9-based gene-editing tools for gene correction. These different approaches determine safety and immunological considerations. By exploring genetic, technical, and therapeutic aspects, this review highlights the potential and current challenges of mRNA therapies to address the large unmet needs in rare genetic disorders.
Collapse
Affiliation(s)
- Paul J L Schürmann
- Division of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Stijn P E van Breda Vriesman
- Division of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Jose A Castro-Alpízar
- Division of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Sander A A Kooijmans
- Division of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Edward E S Nieuwenhuis
- Erasmus MC Rare Disease Center, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
- Nanocell Therapeutics, Utrecht, the Netherlands
| | - Sabine A Fuchs
- Division of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
22
|
Andreatta F, Hendriks D, Artegiani B. Human Organoids as an Emerging Tool for Genome Screenings. Annu Rev Biomed Eng 2025; 27:157-183. [PMID: 40310889 DOI: 10.1146/annurev-bioeng-103023-122327] [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: 05/03/2025]
Abstract
Over the last decade, a plethora of organoid models have been generated to recapitulate aspects of human development, disease, tissue homeostasis, and repair. Organoids representing multiple tissues have emerged and are typically categorized based on their origin. Tissue-derived organoids are established directly from tissue-resident stem/progenitor cells of either adult or fetal origin. Starting from pluripotent stem cells (PSCs), PSC-derived organoids instead recapitulate the developmental trajectory of a given organ. Gene editing technologies, particularly the CRISPR-Cas toolbox, have greatly facilitated gene manipulation experiments with considerable ease and scalability, revolutionizing organoid-based human biology research. Here, we review the recent adaptation of CRISPR-based screenings in organoids. We examine the strategies adopted to perform CRISPR screenings in organoids, discuss different screening scopes and readouts, and highlight organoid-specific challenges. We then discuss individual organoid-based genome screening studies that have uncovered novel genes involved in a variety of biological processes. We close by providing an outlook on how widespread adaptation of CRISPR screenings across the organoid field may be achieved, to ultimately leverage our understanding of human biology.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; ,
| | | |
Collapse
|
23
|
Boff MO, Xavier FAC, Diz FM, Gonçalves JB, Ferreira LM, Zambeli J, Pazzin DB, Previato TTR, Erwig HS, Gonçalves JIB, Bruzzo FTK, Marinowic D, da Costa JC, Zanirati G. mTORopathies in Epilepsy and Neurodevelopmental Disorders: The Future of Therapeutics and the Role of Gene Editing. Cells 2025; 14:662. [PMID: 40358185 PMCID: PMC12071303 DOI: 10.3390/cells14090662] [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: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 05/15/2025] Open
Abstract
mTORopathies represent a group of neurodevelopmental disorders linked to dysregulated mTOR signaling, resulting in conditions such as tuberous sclerosis complex, focal cortical dysplasia, hemimegalencephaly, and Smith-Kingsmore Syndrome. These disorders often manifest with epilepsy, cognitive impairments, and, in some cases, structural brain anomalies. The mTOR pathway, a central regulator of cell growth and metabolism, plays a crucial role in brain development, where its hyperactivation leads to abnormal neuroplasticity, tumor formation, and heightened neuronal excitability. Current treatments primarily rely on mTOR inhibitors, such as rapamycin, which reduce seizure frequency and tumor size but fail to address underlying genetic causes. Advances in gene editing, particularly via CRISPR/Cas9, offer promising avenues for precision therapies targeting the genetic mutations driving mTORopathies. New delivery systems, including viral and non-viral vectors, aim to enhance the specificity and efficacy of these therapies, potentially transforming the management of these disorders. While gene editing holds curative potential, challenges remain concerning delivery, long-term safety, and ethical considerations. Continued research into mTOR mechanisms and innovative gene therapies may pave the way for transformative, personalized treatments for patients affected by these complex neurodevelopmental conditions.
Collapse
Affiliation(s)
- Marina Ottmann Boff
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Mendonça Diz
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Laura Meireles Ferreira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jean Zambeli
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, University of the Valley of the Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, RS, Brazil
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Helena Scartassini Erwig
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Fernanda Thays Konat Bruzzo
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Daniel Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| |
Collapse
|
24
|
Mitsunobu H, Kita Y, Nambu-Nishida Y, Miyazaki S, Nakajima K, Taoka KI, Kondo A, Nishida K. Development of a highly efficient base editing system for Lactobacilli to improve probiotics and dissect essential functions. Appl Microbiol Biotechnol 2025; 109:96. [PMID: 40261411 PMCID: PMC12014835 DOI: 10.1007/s00253-025-13489-z] [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: 02/21/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
Lactobacilli play essential roles in the food industry and have a significant potential as probiotics and therapeutic agents. Genomic and genetic information has increasingly accumulated and been linked to their various functions, to which transgenic approaches are being performed to verify crucial genes. In order to reasonably develop more useful strains, beneficial traits need to be introduced into any given strains and enhanced or combined based on such genotype characterization. However, for practical use as probiotics or foods, organisms with transgene are hardly acceptable. Here, we have introduced the base editing Target-AID system specifically for Lactobacilli, enabling precise installation of point mutations without donor DNA and at multiple genomic loci simultaneously. Lactiplantibacillus plantarum has been successfully engineered to reduce production of imidazole propionate, which has been reported to be associated with type 2 diabetes by impairing glucose tolerance and insulin signaling. Additionally, this system enabled transient knock-out of an essential gene, such as one involved in cell division, resulting in severe filamentous cell phenotype. This demonstrates Target-AID is a promising genetic tool for Lactobacilli and can accelerate both applied and fundamental research. KEY POINTS: • Efficient and multiplexable cytosine base editing established in Lactobacilli. • Edited Lactobacillus reducing imidazole propionate associated with the risk of type 2 diabetes. • Transient knock-out and dissection of an essential gene function.
Collapse
Affiliation(s)
- Hitoshi Mitsunobu
- Engineering Biology Research Center, Kobe University, Kobe, Hyogo, Japan
| | - Yudai Kita
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan
| | | | | | | | - Ken-Ichiro Taoka
- Engineering Biology Research Center, Kobe University, Kobe, Hyogo, Japan
| | - Akihiko Kondo
- Engineering Biology Research Center, Kobe University, Kobe, Hyogo, Japan
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan
- RIKEN Center for Sustainable Resource Science, Yokohama, Japan
| | - Keiji Nishida
- Engineering Biology Research Center, Kobe University, Kobe, Hyogo, Japan.
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan.
| |
Collapse
|
25
|
Thornburg CD, Pipe SW, Cantore A, Unzu C, Jones M, Miesbach WA. Clinical perspective: Advancing hemophilia treatment through gene therapy approaches. Mol Ther 2025:S1525-0016(25)00297-7. [PMID: 40263938 DOI: 10.1016/j.ymthe.2025.04.023] [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: 12/23/2024] [Revised: 03/21/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Hemophilia, a congenital bleeding disorder, can cause arthropathy, impaired mobility, pain, and life-threatening hemorrhage events, significantly impacting quality of life for patients and caregivers. Current therapies, although effective, necessitate costly lifelong treatment, often in specialized settings. However, as a monogenic disorder caused by loss-of-function genetic variants, hemophilia is amenable to gene therapy. In this article, three primary gene therapy approaches at the forefront of clinical development are reviewed. Adeno-associated virus-based gene therapy, having secured approval in the EU, UK, and US after promising phase 3 trial results, demonstrates clear superiority over standard-of-care treatment. Lentivirus-based approaches capable of transducing dividing and nondividing cells may improve the durability of treatment and have low susceptibility to pre-existing neutralizing antibodies to viral vectors. Finally, gene editing techniques such as zinc finger nucleases and CRISPR aim to correct genetic defects directly, holding promise as novel, effective, and highly durable therapeutic strategies in adults and children with hemophilia. This review provides a comprehensive summary of the current status of these gene therapy approaches, highlighting advantages, limitations, and potential future developments.
Collapse
Affiliation(s)
- Courtney D Thornburg
- National Institutes of Health, National Heart, Lung, and Blood Institute, Division of Blood Diseases and Resources, Bethesda, MD, USA
| | - Steve W Pipe
- Pediatric Hematology-Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Carmen Unzu
- DNA and RNA Medicine Division, CIMA Universidad de Navarra, Pamplona, Spain
| | | | - Wolfgang A Miesbach
- Department of Haemostaseology University Hospital Frankfurt, Frankfurt, Germany.
| |
Collapse
|
26
|
Kobata K, Unita D, Matsuo K, Waku T, Kobori A. Point Mutation Selective Photo-Cross-Linking Reactions by Diazirine-Derivatized ODNs. J Org Chem 2025; 90:5093-5099. [PMID: 40196989 DOI: 10.1021/acs.joc.4c02493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Point mutations and single base mutations are the dominant mutations found in the KRAS gene. Systematic knockout of the KRAS gene with these point mutations is still challenging. In this study, we developed novel photo-cross-linking oligonucleotides (pU1-ODN and pU2-ODN) that had a diazirine group at the 5-position of the uridine derivatives. Photo-cross-linking studies of the oligonucleotides with wild-type and mutated RNAs revealed that pU2-ODN efficiently and selectively reacts with the mutated RNAs that contain a cytidine, guanosine, or uridine residue at the frontal position of the pU2 nucleoside. These results suggest that pU2-ODN is a promising candidate for use as a photo-cross-linking ODN to selectively inhibit the activity of mRNAs with a point mutation.
Collapse
Affiliation(s)
- Kentaro Kobata
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Daiki Unita
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Kazuya Matsuo
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Tomonori Waku
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Akio Kobori
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
27
|
Navarro C, Díaz MP, Duran P, Castro A, Díaz A, Cano C, Carbonell-Zabaleta AK, Solano-Jimenez DS, Rivera-Porras D, Contreras-Velásquez JC, Bermúdez V. CRISPR-Cas Systems: A Functional Perspective and Innovations. Int J Mol Sci 2025; 26:3645. [PMID: 40332149 PMCID: PMC12026748 DOI: 10.3390/ijms26083645] [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: 01/14/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 05/08/2025] Open
Abstract
Adaptation is a fundamental tenet of evolutionary biology and is essential for the survival of all organisms, including prokaryotes. The evolution of clustered regularity exemplifies this principle of interspaced short palindromic repeats (CRISPR) and associated proteins (Cas), an adaptive immune system that confers resistance to viral infections. By integrating short segments of viral genomes into their own, bacteria and archaea develop a molecular memory that enables them to mount a rapid and targeted response upon subsequent viral challenges. The fortuitous discovery of this immune mechanism prompted many studies and introduced researchers to novel tools that could potentially be developed from CRISPR-Cas and become clinically relevant as biotechnology rapidly advances in this area. Thus, a deeper understanding of the underpinnings of CRISPR-Cas and its possible therapeutic applications is required. This review analyses the mechanism of action of the CRISPR-Cas systems in detail and summarises the advances in developing biotechnological tools based on CRISPR, opening the field for further research.
Collapse
Affiliation(s)
- Carla Navarro
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 40001, Venezuela; (M.P.D.); (P.D.); (A.C.); (A.D.); (C.C.)
| | - María P. Díaz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 40001, Venezuela; (M.P.D.); (P.D.); (A.C.); (A.D.); (C.C.)
| | - Pablo Duran
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 40001, Venezuela; (M.P.D.); (P.D.); (A.C.); (A.D.); (C.C.)
| | - Ana Castro
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 40001, Venezuela; (M.P.D.); (P.D.); (A.C.); (A.D.); (C.C.)
| | - Andrea Díaz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 40001, Venezuela; (M.P.D.); (P.D.); (A.C.); (A.D.); (C.C.)
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 40001, Venezuela; (M.P.D.); (P.D.); (A.C.); (A.D.); (C.C.)
| | - Ana-Karina Carbonell-Zabaleta
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Programa de Medicina, Barranquilla 080001, Colombia; (A.-K.C.-Z.); (D.-S.S.-J.)
| | - Donny-Sabrith Solano-Jimenez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Programa de Medicina, Barranquilla 080001, Colombia; (A.-K.C.-Z.); (D.-S.S.-J.)
| | - Diego Rivera-Porras
- Universidad de la Costa, Departamento de Productividad e Innovación, Barranquilla 080001, Atlántico, Colombia; (D.R.-P.); (J.C.C.-V.)
| | - Julio César Contreras-Velásquez
- Universidad de la Costa, Departamento de Productividad e Innovación, Barranquilla 080001, Atlántico, Colombia; (D.R.-P.); (J.C.C.-V.)
| | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Centro de Investigaciones en Ciencias de la Vida, Barranquilla 080001, Colombia
| |
Collapse
|
28
|
Menon AV, Song B, Chao L, Sriram D, Chansky P, Bakshi I, Ulianova J, Li W. Unraveling the future of genomics: CRISPR, single-cell omics, and the applications in cancer and immunology. Front Genome Ed 2025; 7:1565387. [PMID: 40292231 PMCID: PMC12021818 DOI: 10.3389/fgeed.2025.1565387] [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: 01/23/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
The CRISPR system has transformed many research areas, including cancer and immunology, by providing a simple yet effective genome editing system. Its simplicity has facilitated large-scale experiments to assess gene functionality across diverse biological contexts, generating extensive datasets that boosted the development of computational methods and machine learning/artificial intelligence applications. Integrating CRISPR with single-cell technologies has further advanced our understanding of genome function and its role in many biological processes, providing unprecedented insights into human biology and disease mechanisms. This powerful combination has accelerated AI-driven analyses, enhancing disease diagnostics, risk prediction, and therapeutic innovations. This review provides a comprehensive overview of CRISPR-based genome editing systems, highlighting their advancements, current progress, challenges, and future opportunities, especially in cancer and immunology.
Collapse
Affiliation(s)
- A. Vipin Menon
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Bicna Song
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Lumen Chao
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Diksha Sriram
- The George Washington University, Washington, DC, DC, United States
| | - Pamela Chansky
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Integrated Biomedical Sciences (IBS) Program, The George Washington University, Washington, DC, DC, United States
| | - Ishnoor Bakshi
- The George Washington University, Washington, DC, DC, United States
| | - Jane Ulianova
- Integrated Biomedical Sciences (IBS) Program, The George Washington University, Washington, DC, DC, United States
| | - Wei Li
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| |
Collapse
|
29
|
Yuan K, Xi X, Han S, Han J, Zhao B, Wei Q, Zhou X. Selict-seq profiles genome-wide off-target effects in adenosine base editing. Nucleic Acids Res 2025; 53:gkaf281. [PMID: 40207628 PMCID: PMC11983105 DOI: 10.1093/nar/gkaf281] [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: 10/26/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Adenosine base editors (ABEs) facilitate A·T to G·C base pair conversion with significant therapeutic potential for correcting pathogenic point mutations in human genetic diseases, such as sickle cell anemia and β-thalassemia. Unlike CRISPR-Cas9 systems that induce double-strand breaks, ABEs operate through precise deamination, avoiding chromosomal instability. However, the off-target editing effects of ABEs remain inadequately characterized. In this study, we present a biochemical method Selict-seq, designed to evaluate genome-wide off-target editing by ABEs. Selict-seq specifically captures deoxyinosine-containing single-stranded DNA and precisely identifies deoxyadenosine-to-deoxyinosine (dA-to-dI) mutation sites, elucidating the off-target effects induced by ABEs. Through investigations involving three single-guide RNAs, we identified numerous unexpected off-target edits both within and outside the protospacer regions. Notably, ABE8e(V106W) exhibited distinct off-target characteristics, including high editing rates (>10%) at previously unreported sites (e.g. RNF2 and EMX1) and out-of-protospacer mutations. These findings significantly advance our understanding of the off-target landscape associated with ABEs. In summary, our approach enables an unbiased analysis of the ABE editome and provides a widely applicable tool for specificity evaluation of various emerging genome editing technologies that produce intermediate products as deoxyinosine.
Collapse
Affiliation(s)
- Kexin Yuan
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Xin Xi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Shaoqing Han
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Jingyu Han
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Bin Zhao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Qi Wei
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Xiang Zhou
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, PR China
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, PR China
| |
Collapse
|
30
|
Tachida Y, Manian KV, Butcher R, Levy JM, Pendse N, Hennessey E, Liu DR, Pierce EA, Liu Q, Comander J. Systematic empirical evaluation of individual base editing targets: Validating therapeutic targets in USH2A and comparison of methods. Mol Ther 2025; 33:1466-1484. [PMID: 39881543 PMCID: PMC11997516 DOI: 10.1016/j.ymthe.2025.01.042] [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: 08/23/2024] [Revised: 11/22/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Base editing shows promise for the correction of human mutations at a higher efficiency than other repair methods and is especially attractive for mutations in large genes that are not amenable to gene augmentation therapy. Here, we demonstrate a comprehensive workflow for in vitro screening of potential therapeutic base editing targets for the USH2A gene and empirically validate the efficiency of adenine and cytosine base editor/guide combinations for correcting 35 USH2A mutations. Editing efficiency and bystander edits are compared between different target templates (plasmids vs. transgenes) and assays (next-generation sequencing vs. Sanger), as well as comparisons between unbiased empirical results and computational predictions. Based on these observations, practical assay recommendations are discussed. Finally, a humanized knockin mouse model was created with the best-performing target, the nonsense mutation c.11864G>A p.(Trp3955∗). Split-intein AAV9 delivery of editing reagents resulted in the restoration of USH2A protein and a correction rate of 65% ± 3% at the mutant base pair and of 52% ± 3% excluding bystander amino acid changes. This efficiency is higher than that seen in a retinal gene editing program testing in a clinical trial. These results demonstrate the effectiveness of this overall strategy to identify and test base editing reagents with the potential for human therapeutic applications.
Collapse
Affiliation(s)
- Yuki Tachida
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kannan V Manian
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Rossano Butcher
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Jonathan M Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02142, USA
| | - Nachiket Pendse
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Erin Hennessey
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02142, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Qin Liu
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Jason Comander
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
31
|
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
|
32
|
Hardouin G, Miccio A, Brusson M. Gene therapy for β-thalassemia: current and future options. Trends Mol Med 2025; 31:344-358. [PMID: 39794177 DOI: 10.1016/j.molmed.2024.12.001] [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: 10/08/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 01/13/2025]
Abstract
Beta-thalassemia is a severe, hereditary blood disorder characterized by anemia, transfusion dependence, reduced life expectancy, and poor quality of life. Allogeneic transplantation of hematopoietic stem cells (HSCs) is the only curative treatment for transfusion-dependent β-thalassemia, but a lack of compatible donors prevents the use of this approach for most patients. Over the past 20 years, the rise of gene therapy and the development of lentiviral vectors and genome-editing tools has extended curative options to a broader range of patients. Here, we review breakthroughs in gene addition- and genome-editing-based therapies for β-thalassemia, the clinical outcomes enabling approval by regulatory agencies, and perspectives for further development.
Collapse
Affiliation(s)
- Giulia Hardouin
- Université Paris Cité, Imagine Institute, Laboratory of chromatin and gene regulation during development, INSERM UMR 1163, 75015, Paris, France.
| | - Annarita Miccio
- Université Paris Cité, Imagine Institute, Laboratory of chromatin and gene regulation during development, INSERM UMR 1163, 75015, Paris, France.
| | - Megane Brusson
- Université Paris Cité, Imagine Institute, Laboratory of chromatin and gene regulation during development, INSERM UMR 1163, 75015, Paris, France
| |
Collapse
|
33
|
Reautschnig P, Fruhner C, Wahn N, Wiegand CP, Kragness S, Yung JF, Hofacker DT, Fisk J, Eidelman M, Waffenschmidt N, Feige M, Pfeiffer LS, Schulz AE, Füll Y, Levanon EY, Mandel G, Stafforst T. Precise in vivo RNA base editing with a wobble-enhanced circular CLUSTER guide RNA. Nat Biotechnol 2025; 43:545-557. [PMID: 38997581 PMCID: PMC11994451 DOI: 10.1038/s41587-024-02313-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 05/17/2024] [Indexed: 07/14/2024]
Abstract
Recruiting the endogenous editing enzyme adenosine deaminase acting on RNA (ADAR) with tailored guide RNAs for adenosine-to-inosine (A-to-I) RNA base editing is promising for safely manipulating genetic information at the RNA level. However, the precision and efficiency of editing are often compromised by bystander off-target editing. Here, we find that in 5'-UAN triplets, which dominate bystander editing, G•U wobble base pairs effectively mitigate off-target events while maintaining high on-target efficiency. This strategy is universally applicable to existing A-to-I RNA base-editing systems and complements other suppression methods such as G•A mismatches and uridine (U) depletion. Combining wobble base pairing with a circularized format of the CLUSTER approach achieves highly precise and efficient editing (up to 87%) of a disease-relevant mutation in the Mecp2 transcript in cell culture. Virus-mediated delivery of the guide RNA alone realizes functional MeCP2 protein restoration in the central nervous system of a murine Rett syndrome model with editing yields of up to 19% and excellent bystander control in vivo.
Collapse
Affiliation(s)
- Philipp Reautschnig
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Carolin Fruhner
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Nicolai Wahn
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Charlotte P Wiegand
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Sabrina Kragness
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - John F Yung
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Daniel T Hofacker
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Jenna Fisk
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Michelle Eidelman
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Nils Waffenschmidt
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Maximilian Feige
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Laura S Pfeiffer
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Annika E Schulz
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Yvonne Füll
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany
| | - Erez Y Levanon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Gail Mandel
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Thorsten Stafforst
- Interfaculty Institute of Biochemistry, Faculty of Science, University of Tübingen, Tübingen, Germany.
- Gene and RNA Therapy Center (GRTC), Faculty of Medicine, University of Tübingen, Tübingen, Germany.
- iFIT Cluster of Excellence (EXC2180) Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
34
|
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
|
35
|
Mariam J, Porathoor S, Anand R. Mechanism of Conformational Selection of tRNA Arg2 by Bacterial Deaminase TadA. Biochemistry 2025; 64:1530-1540. [PMID: 40074666 DOI: 10.1021/acs.biochem.4c00649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Base editing is a common mechanism by which organisms expand their genetic repertoire to access new functions. Here, we explore the mechanism of tRNA recognition in the bacterial deaminase TadA, which exclusively recognizes tRNAArg2 and converts the wobble base adenosine (A34) to inosine. We quantitatively evaluate the dynamics of tRNA binding by incorporating the fluorescent adenine analogue 2-aminopurine (2-AP) at position 34 in the wobble base of the anticodon loop. Time-resolved fluorescence and anisotropy studies revealed that the recognition process is finely tuned. Mutations in residues directly involved in facilitating deamination, such as E55A and N42A, showed a minimal impact on binding dynamics. In contrast, mutations in the "capping residues", notably R149, unique to prokaryotic TadAs and located 12-15 Å away from the catalytic center, significantly disrupted binding and consequently catalytic activity. The capping residues play a critical role in enabling tRNA recognition, thereby underscoring their importance in enzyme function. Moreover, for effective catalysis, peripheral positively charged residues (R70, R94) that are part of the adjacent subunit in the dimeric assembly are important to splay out the tRNA, assisting in A34 attaining a flipped-out conformation. Perturbations in these extended regions, although 15-20 Å away from the active site, disrupt the binding dynamics and consequently the function, emphasizing the fine regulation of the tRNA recognition process. Analysis reveals that the C-terminal end of the extended helix where R149 is positioned, acts as a selectivity filter imparting exclusivity toward the deamination of tRNAArg2 by TadA.
Collapse
Affiliation(s)
- Jessy Mariam
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sini Porathoor
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Ruchi Anand
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
36
|
Mustfa SA, Dimitrievska M, Wang C, Gu C, Sun N, Romańczuk K, Karpinski P, Łaczmański Ł, McGrath JA, Jacków‐Malinowska J, Chiappini C. Porous Silicon Nanoneedles Efficiently Deliver Adenine Base Editor to Correct a Recurrent Pathogenic COL7A1 Variant in Recessive Dystrophic Epidermolysis Bullosa. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414728. [PMID: 40072288 PMCID: PMC12038538 DOI: 10.1002/adma.202414728] [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: 09/28/2024] [Revised: 02/26/2025] [Indexed: 04/30/2025]
Abstract
Base editing, a CRISPR-based genome editing technology, enables precise correction of single-nucleotide variants, promising resolutive treatment for monogenic genetic disorders like recessive dystrophic epidermolysis bullosa (RDEB). However, the application of base editors in cell manufacturing is hindered by inconsistent efficiency and high costs, contributed by suboptimal delivery methods. Nanoneedles have emerged as an effective delivery approach, enabling highly efficient, non-perturbing gene therapies both in vitro and in vivo. Here we demonstrate that nanoneedle delivery of an adenine base editor corrects a heterozygous single-nucleotide pathogenic variant in COL7A1 in primary RDEB fibroblasts in vitro with 96.5% efficiency, without inducing off-target variants. The nanoneedle delivery maintains cell viability and displays modest phenotypical alterations unlike conventional cationic lipid transfection. The nanoneedle-mediated editing significantly increases the production and secretion of full-length type VII collagen protein, contributing to restore functional fibroblasts phenotype by improving cell adhesion. These findings underscore the suitability and safety of nanoneedles for gene editing in a clinically relevant context of cell manufacturing, establishing a foundation for their use in cell therapies.
Collapse
Affiliation(s)
- Salman Ahmad Mustfa
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 1ULUK
| | - Marija Dimitrievska
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 1ULUK
- St John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonLondonSE1 1ULUK
| | - Cong Wang
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 1ULUK
- London Centre for NanotechnologyKing's College LondonLondonSE1 1ULUK
| | - Chenlei Gu
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 1ULUK
- London Centre for NanotechnologyKing's College LondonLondonSE1 1ULUK
| | - Ningjia Sun
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 1ULUK
| | - Katarzyna Romańczuk
- Hirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclaw53‐114Poland
| | - Pawel Karpinski
- Department of GeneticsWroclaw Medical UniversityWroclaw50‐367Poland
| | - Łukasz Łaczmański
- Hirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclaw53‐114Poland
| | - John A. McGrath
- St John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonLondonSE1 1ULUK
| | - Joanna Jacków‐Malinowska
- St John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonLondonSE1 1ULUK
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 1ULUK
- London Centre for NanotechnologyKing's College LondonLondonSE1 1ULUK
| |
Collapse
|
37
|
Chen Q, Sun Y, Yao J, Lu Y, Qiu R, Zhou F, Deng Z, Sun Y. Engineering of Peptide-Inserted Base Editors with Enhanced Accuracy and Security. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411583. [PMID: 39995348 PMCID: PMC11983243 DOI: 10.1002/smll.202411583] [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: 12/11/2024] [Revised: 02/08/2025] [Indexed: 02/26/2025]
Abstract
Base editors are effective tools for introducing base conversions without double-strand breaks, showing broad applications in biotechnological and clinical areas. However, their non-negligible bystander mutations and off-target effects have raised extensive safety concerns. To address these issues, a novel method is developed by inserting specific peptide fragments into the substrate binding pocket of deaminases in base editors to modify these outcomes. It is validated that the composition and position of the inserted peptide can significantly impact the performance of A3A-based cytosine base editor and TadA-8e-based adenine base editor, leading to improved editing activity and precision in human HEK293T cells. Importantly, the TadA-8e variant with DPLVLRRRQ peptide inserted behind S116 residue showed a strong motif preference of Y4A5N6, which can accurately edit the A5 base in targeted protospacer with minimized bystander and off-target effects in DNA and RNA-level. By summarizing the regularity during engineering, a set of systematic procedures is established, which can potentially be used to modify other types of base editors and make them more accurate and secure. In addition, the peptide insertion strategy is also proven to be compatible with traditional amino acid changes which have been reported, exhibiting excellent compatibility.
Collapse
Affiliation(s)
- Qi Chen
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Yangning Sun
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Jia Yao
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Yingfan Lu
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Ruikang Qiu
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Fuling Zhou
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Zixin Deng
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| | - Yuhui Sun
- Department of HematologyZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- School of PharmacyHuazhong University of Science and TechnologyWuhan430030China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education)Wuhan UniversityWuhan430071China
| |
Collapse
|
38
|
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
|
39
|
Nicosia L, Pranke I, Latorre RV, Murray JB, Lonetti L, Cavusoglu-Doran K, Dreano E, Costello JP, Carroll M, Melotti P, Sorio C, Sermet-Gaudelus I, Scallan MF, Harrison PT. Adenine base editing with engineered virus-like particles rescues the CFTR mutation G542X in patient-derived intestinal organoids. iScience 2025; 28:111979. [PMID: 40144632 PMCID: PMC11938077 DOI: 10.1016/j.isci.2025.111979] [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: 06/11/2024] [Revised: 11/05/2024] [Accepted: 02/05/2025] [Indexed: 03/28/2025] Open
Abstract
Cystic fibrosis (CF) is a life-shortening autosomal recessive disease, caused by loss-of-function mutations that affect the CF transmembrane conductance regulator (CFTR) anion channel. G542X is the second-most common CF-causing variant, and it does not respond to current CFTR modulator drugs. Our study explores the use of adenine base editing to edit G542X to a non-CF-causing variant, G542R, and recover CFTR function. Using base editor engineered virus-like particles (BE-eVLPs) in patient-derived intestinal organoids, we achieved ∼2% G542X-to-G542R editing efficiency and restored CFTR-mediated chloride transport to ∼6.4% of wild-type levels, independent of modulator treatment, and with no bystander edits. This proof-of-principle study demonstrates the potential of base editing to rescue G542X and provides a foundation for future in - vivo applications.
Collapse
Affiliation(s)
- Lucia Nicosia
- Department of Physiology, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Iwona Pranke
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
| | | | - Joss B. Murray
- Department of Physiology, University College Cork, Cork, Ireland
| | - Lisa Lonetti
- Department of Physiology, University College Cork, Cork, Ireland
| | | | - Elise Dreano
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
| | | | - Michael Carroll
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paola Melotti
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Claudio Sorio
- Department of Medicine, University of Verona, Verona, Italy
| | - Isabelle Sermet-Gaudelus
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
- Cystic Fibrosis National Pediatric Reference Center, Pneumo-Allergologie Pédiatrique, Hôpital Necker Enfants Malades, , Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
- European Reference Network, ERN-Lung CF, Frankfurt am Mein, Germany
| | | | - Patrick T. Harrison
- Department of Physiology, University College Cork, Cork, Ireland
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| |
Collapse
|
40
|
Chia BS, Seah YFS, Wang B, Shen K, Srivastava D, Chew WL. Engineering a New Generation of Gene Editors: Integrating Synthetic Biology and AI Innovations. ACS Synth Biol 2025; 14:636-647. [PMID: 39999982 PMCID: PMC11934138 DOI: 10.1021/acssynbio.4c00686] [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: 10/04/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025]
Abstract
CRISPR-Cas technology has revolutionized biology by enabling precise DNA and RNA edits with ease. However, significant challenges remain for translating this technology into clinical applications. Traditional protein engineering methods, such as rational design, mutagenesis screens, and directed evolution, have been used to address issues like low efficacy, specificity, and high immunogenicity. These methods are labor-intensive, time-consuming, and resource-intensive and often require detailed structural knowledge. Recently, computational strategies have emerged as powerful solutions to these limitations. Using artificial intelligence (AI) and machine learning (ML), the discovery and design of novel gene-editing enzymes can be streamlined. AI/ML models predict activity, specificity, and immunogenicity while also enhancing mutagenesis screens and directed evolution. These approaches not only accelerate rational design but also create new opportunities for developing safer and more efficient genome-editing tools, which could eventually be translated into the clinic.
Collapse
Affiliation(s)
- Bing Shao Chia
- Genome
Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore 138672, Singapore
| | - Yu Fen Samantha Seah
- Genome
Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore 138672, Singapore
| | - Bolun Wang
- Genome
Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore 138672, Singapore
| | - Kimberle Shen
- Genome
Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore 138672, Singapore
| | - Diya Srivastava
- Genome
Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore 138672, Singapore
| | - Wei Leong Chew
- Genome
Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore 138672, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| |
Collapse
|
41
|
He X, Yan T, Song Z, Xiang L, Xiang J, Yang Y, Ren K, Bu J, Xu X, Li Z, Guo X, Lin B, Zhou Q, Lin G, Gu F. Correcting a patient-specific Rhodopsin mutation with adenine base editor in a mouse model. Mol Ther 2025:S1525-0016(25)00195-9. [PMID: 40119518 DOI: 10.1016/j.ymthe.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/05/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Genome editing offers a great promise to treating human genetic diseases. To assess genome-editing-mediated therapeutic effects in vivo, an animal model is indispensable. The genomic disparities between mice and humans often impede the direct clinical application of genome-editing-mediated treatments using conventional mouse models. Thus, the generation of a mouse model with a humanized genomic segment containing a patient-specific mutation is highly sought after for translational research. In this study, we successfully developed a knockin mouse model for autosomal-dominant retinitis pigmentosa (adRP), designated as hT17M knockin, which incorporates a 75-nucleotide DNA segment with the T17M mutation (Rhodopsin-c.C50T; p.T17M). This model demonstrated significant reductions in electroretinogram amplitudes and exhibited disruptions in retinal structure. Subsequently, we administered an adeno-associated virus vectors carrying an adenine base editor (ABE) and a single-guide RNA specifically targeting the T17M mutation, achieving a peak correction rate of 39.7% at the RNA level and significantly improving retinal function in ABE-injected mice. These findings underscore that the hT17M knockin mouse model recapitulates the clinical features of adRP patients and exhibits therapeutic effects with ABE-mediated treatments. It offers a promising avenue for the development of gene-editing therapies for RP.
Collapse
Affiliation(s)
- Xiaoxue He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China; School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Tong Yan
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Zongming Song
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Lue Xiang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Jiayang Xiang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Yeqin Yang
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Kaiqun Ren
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China
| | - Jicheng Bu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Xilin Xu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Zhuo Li
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Xiaowei Guo
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China
| | - Bin Lin
- School of Optometry, Hong Kong Polytechnic University, Hong Kong HJ502, China
| | - Qinghua Zhou
- Department of Anesthesiology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Feng Gu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China; School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China; Guangxiu Hospital Affiliated with Hunan Normal University (Hunan Guangxiu Hospital), Changsha, Hunan 410119, China.
| |
Collapse
|
42
|
McDonnell KJ. Operationalizing Team Science at the Academic Cancer Center Network to Unveil the Structure and Function of the Gut Microbiome. J Clin Med 2025; 14:2040. [PMID: 40142848 PMCID: PMC11943358 DOI: 10.3390/jcm14062040] [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: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Oncologists increasingly recognize the microbiome as an important facilitator of health as well as a contributor to disease, including, specifically, cancer. Our knowledge of the etiologies, mechanisms, and modulation of microbiome states that ameliorate or promote cancer continues to evolve. The progressive refinement and adoption of "omic" technologies (genomics, transcriptomics, proteomics, and metabolomics) and utilization of advanced computational methods accelerate this evolution. The academic cancer center network, with its immediate access to extensive, multidisciplinary expertise and scientific resources, has the potential to catalyze microbiome research. Here, we review our current understanding of the role of the gut microbiome in cancer prevention, predisposition, and response to therapy. We underscore the promise of operationalizing the academic cancer center network to uncover the structure and function of the gut microbiome; we highlight the unique microbiome-related expert resources available at the City of Hope of Comprehensive Cancer Center as an example of the potential of team science to achieve novel scientific and clinical discovery.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
43
|
Siles L, Pomares E. Rescue of the disease-associated phenotype in CRISPR-corrected hiPSCs as a therapeutic approach for inherited retinal dystrophies. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102482. [PMID: 40083649 PMCID: PMC11903799 DOI: 10.1016/j.omtn.2025.102482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Inherited retinal dystrophies (IRDs), such as retinitis pigmentosa and Stargardt disease, are a group of rare diseases caused by mutations in more than 300 genes that currently have no treatment in most cases. They commonly trigger blindness and other ocular affectations due to retinal cell degeneration. Gene editing has emerged as a promising and powerful strategy for the development of IRD therapies, allowing the permanent correction of pathogenic variants. Using clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 and transcription activator-like effector nucleases (TALEN) gene-editing tools, we precisely corrected seven hiPS cell lines derived from IRD patients carrying mutations in ABCA4, BEST1, PDE6A, PDE6C, RHO, or USH2A. Homozygous mutations and point insertions/deletions resulted in the highest homology-directed repair efficiencies, with at least half of the clones repaired properly without off-target effects. Strikingly, correction of a heterozygous pathogenic variant was achieved using the wild-type allele of the patient as the template for DNA repair. These results suggest the unexpected potential application of CRISPR as a donor template-free strategy for single-nucleotide modifications. Additionally, the corrected clones exhibited a reversion of the disease-associated phenotype in retinal cellular models. These data strengthen the study and application of gene editing-based approaches for IRD treatment.
Collapse
Affiliation(s)
- Laura Siles
- Departament de Genètica, Institut de Microcirurgia Ocular, IMO Grupo Miranza, 08035 Barcelona, Spain
| | - Esther Pomares
- Departament de Genètica, Institut de Microcirurgia Ocular, IMO Grupo Miranza, 08035 Barcelona, Spain
| |
Collapse
|
44
|
Ukaji T, Arai D, Tsutsumi H, Nakagawa R, Matsumoto F, Ikeda K, Nureki O, Kamiya K. AAV-mediated base editing restores cochlear gap junction in GJB2 dominant-negative mutation-associated syndromic hearing loss model. JCI Insight 2025; 10:e185193. [PMID: 40059830 PMCID: PMC11949026 DOI: 10.1172/jci.insight.185193] [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: 07/23/2024] [Accepted: 01/24/2025] [Indexed: 03/29/2025] Open
Abstract
Mutations in the gap junction β2 (GJB2) gene, which encodes connexin 26, are the leading cause of genetic deafness. These mutations are characterized by the degeneration and fragmentation of gap junctions and gap junction plaques (GJPs) composed of connexin 26. Dominant-negative mutations of GJB2, such as R75W, cause syndromic hearing loss and palmoplantar keratoderma. We previously reported that the R75W mutation, a single-base substitution where C is replaced by T, causes fragmentation of GJPs. Therefore, an adenine base editor (ABE), which enables A-to-G base conversions, can potentially be useful for the treatment of this genetic disease. Here, we report that an all-in-one adeno-associated virus (AAV) vector, which includes a compact ABE (SaCas9-NNG-ABE8e) with broad targeting range, and a sgRNA targeting the R75W mutation in GJB2 corrected this pathogenic mutation and facilitated the recovery of the gap junction intercellular communication network of GJPs. In a transgenic mouse model with the GJB2 R75W mutation, AAV-mediated base editing also restored the fragmented GJPs to orderly outlines in cochlear supporting cells. Our findings suggest that an ABE-based base-editing strategy could be an optimal treatment for the dominant form of GJB2-related hearing loss, GJB2-related skin diseases, and other deafness-related mutations, especially single-base substitutions.
Collapse
Affiliation(s)
- Takao Ukaji
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Daisuke Arai
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Harumi Tsutsumi
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ryoya Nakagawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Fumihiko Matsumoto
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Katsuhisa Ikeda
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Mach RQ, Miller SM. Bacterial directed evolution of CRISPR base editors. Methods Enzymol 2025; 712:317-350. [PMID: 40121078 DOI: 10.1016/bs.mie.2025.01.003] [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
Base editing and other precision editing agents have transformed the utility and therapeutic potential of CRISPR-based genome editing. While some native enzymes edit efficiently with their nature-derived function, many enzymes require rational engineering or directed evolution to enhance the compatibility with mammalian cell genome editing. While many methods of engineering and directed evolution exist, plate-based discrete evolution offers an ideal balance between ease of use and engineering power. Here, we describe a detailed method for the bacterial directed evolution of CRISPR base editors that compounds technical ease with flexibility of application.
Collapse
|
46
|
Song B. Efforts to Downsize Base Editors for Clinical Applications. Int J Mol Sci 2025; 26:2357. [PMID: 40076976 PMCID: PMC11900391 DOI: 10.3390/ijms26052357] [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: 12/27/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Since the advent of the clustered regularly interspaced short palindromic repeats (CRISPR) system in the gene editing field, diverse CRISPR-based gene editing tools have been developed for treating genetic diseases. Of these, base editors (BEs) are promising because they can carry out precise gene editing at single-nucleotide resolution without inducing DNA double-strand breaks (DSBs), which pose significant risks of genomic instability. Despite their outstanding advantages, the clinical application of BEs remains challenging due to their large size, which limits their efficient delivery, particularly in adeno-associated virus (AAV)-based systems. To address this issue, various strategies have been explored to reduce the size of BEs. These approaches include truncating the nonessential domains and replacing the bulky components with smaller substitutes without compromising the editing efficiency. In this review, we highlight the importance of downsizing BEs for therapeutic applications and introduce recent advances in size-reduction strategies. Additionally, we introduce the ongoing efforts to overcome other limitations of BEs, providing insights into their potential for improving in vivo gene editing.
Collapse
Affiliation(s)
- Beomjong Song
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| |
Collapse
|
47
|
Carrozzo I, Maule G, Gentile C, Umbach A, Ciciani M, Guidone D, De Santis M, Petris G, Vicente Galietta LJ, Arosio D, Cereseto A. Functional rescue of F508del-CFTR through revertant mutations introduced by CRISPR base editing. Mol Ther 2025; 33:970-985. [PMID: 39797401 PMCID: PMC11897810 DOI: 10.1016/j.ymthe.2025.01.011] [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: 09/06/2024] [Revised: 12/11/2024] [Accepted: 01/08/2025] [Indexed: 01/13/2025] Open
Abstract
Cystic fibrosis (CF) is a life-shortening autosomal recessive disease caused by mutations in the CFTR gene, resulting in functional impairment of the encoded ion channel. F508del mutation, a trinucleotide deletion, is the most frequent cause of CF, affecting approximately 80% of persons with CF (pwCFs). Even though current pharmacological treatments alleviate the F508del-CF disease symptoms, there is no definitive cure. Here, we leveraged revertant mutations (RMs) in cis with F508del to rescue CFTR protein folding and restore its function. We developed CRISPR base editing strategies to efficiently and precisely introduce the desired mutations in the F508del locus. Both editing and CFTR function recovery were verified in CF cellular models, including primary epithelial cells derived from pwCFs. The efficacy of the CFTR recovery strategy was validated in cultures of pseudostratified epithelia from pwCF cells showing full recovery of ion transport. Additionally, we observed an additive effect by combining our strategy with small molecules that enhance F508del activity, thus paving the way to combinatorial therapies.
Collapse
Affiliation(s)
- Irene Carrozzo
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Giulia Maule
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Carmelo Gentile
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Alessandro Umbach
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Matteo Ciciani
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | | | - Gianluca Petris
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Luis Juan Vicente Galietta
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; Department of Translational Medical Sciences, University of Napoli "Federico II", 80138 Napoli, Italy
| | - Daniele Arosio
- Institute of Biophysics, CNR, Via alla Cascata 56/C, 38123 Trento, Italy.
| | - Anna Cereseto
- Department CIBIO, University of Trento, Via delle Regole 101, 38123 Trento, Italy.
| |
Collapse
|
48
|
Chen Q, Chang Y, He X, Ding Y, Wang R, Luo R, Yuan J, Chen J, Zhong G, Yang H, Chen J, Li J. Targeted Delivery of mRNA with Polymer-Lipid Nanoparticles for In Vivo Base Editing. ACS NANO 2025; 19:7835-7850. [PMID: 39962883 DOI: 10.1021/acsnano.4c14041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Messenger RNA (mRNA) encoding base editors, along with single guide RNAs (sgRNAs), have emerged as a promising therapeutic approach for various disorders. However, there is still insufficient exploration in achieving targeted and efficient delivery of mRNA and sgRNA to multiple organs while ensuring high biocompatibility and stability in vivo. To address this challenge, we synthesized a library of 108 poly(β-amino) esters (PBAEs) by incorporating 100% hydrophobic side chains and end-caps with varying amines. These PBAEs were further formulated with other excipients, including helper lipids, cholesterol, and PEGylated lipids, to form polymer-lipid nanoparticles (PLNPs). Structure-function analysis revealed that eLog P of PBAEs could serve as a predictive parameter for determining the liver or lung tropism of PLNPs. The biocompatibility of PBAEs end-capped with monoamines was significantly higher compared to those end-capped with diamines. Leveraging these findings, we expanded the PBAE library and identified a leading PBAE (7C8C8) with mRNA delivery efficiency outperforming current FDA-approved ionizable lipids (ALC-0315, SM-102, and Dlin-MC3-DMA). The LD50 of the empty PLNPs (7C8C8) was determined to be 403.8 ± 49.46 mg/kg, indicating a significantly high safety profile. Additionally, PLNPs (7C8C8) demonstrated sustained transfection activity for at least 2 months when stored at -20 °C after freezing or at 4 °C following lyophilization. Subsequently, in vivo base editing using PLNPs (7C8C8) achieved an impressive editing efficiency of approximately 70% along with a significant reduction in protein levels exceeding 90%. Notably, synergistic effects were observed through simultaneous disruption of proprotein convertase subtilisin/kexin type 9 and angiopoietin-like protein 3 genes, resulting in a sustained low-density lipoprotein cholesterol reduction of over 60% for several months. These compelling findings provide strong support for the further development of PLNPs as promising platforms for mRNA-based therapies.
Collapse
Affiliation(s)
- Qimingxing Chen
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yan Chang
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoyan He
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yan Ding
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Runyuan Wang
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Ran Luo
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Jialu Yuan
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Jiabei Chen
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Guisheng Zhong
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Huiying Yang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jia Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Li
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
49
|
Zhang Y, Liu Z, Hirschi M, Brodsky O, Johnson E, Won SJ, Nagata A, Bezwada D, Petroski MD, Majmudar JD, Niessen S, VanArsdale T, Gilbert AM, Hayward MM, Stewart AE, Nager AR, Melillo B, Cravatt BF. An allosteric cyclin E-CDK2 site mapped by paralog hopping with covalent probes. Nat Chem Biol 2025; 21:420-431. [PMID: 39294320 PMCID: PMC11867888 DOI: 10.1038/s41589-024-01738-7] [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: 01/21/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024]
Abstract
More than half of the ~20,000 protein-encoding human genes have paralogs. Chemical proteomics has uncovered many electrophile-sensitive cysteines that are exclusive to subsets of paralogous proteins. Here we explore whether such covalent compound-cysteine interactions can be used to discover ligandable pockets in paralogs lacking the cysteine. Leveraging the covalent ligandability of C109 in the cyclin CCNE2, we substituted the corresponding residue in paralog CCNE1 to cysteine (N112C) and found through activity-based protein profiling that this mutant reacts stereoselectively and site-specifically with tryptoline acrylamides. We then converted the tryptoline acrylamide-CCNE1-N112C interaction into in vitro NanoBRET (bioluminescence resonance energy transfer) and in cellulo activity-based protein profiling assays capable of identifying compounds that reversibly inhibit both the N112C mutant and wild-type CCNE1:CDK2 (cyclin-dependent kinase 2) complexes. X-ray crystallography revealed a cryptic allosteric pocket at the CCNE1:CDK2 interface adjacent to N112 that binds the reversible inhibitors. Our findings, thus, show how electrophile-cysteine interactions mapped by chemical proteomics can extend the understanding of protein ligandability beyond covalent chemistry.
Collapse
Affiliation(s)
- Yuanjin Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Zhonglin Liu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Marscha Hirschi
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Oleg Brodsky
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Eric Johnson
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Sang Joon Won
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Asako Nagata
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Divya Bezwada
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Jaimeen D Majmudar
- Discovery Sciences, Pfizer Research and Development, Pfizer, Inc., Cambridge, MA, USA
| | - Sherry Niessen
- Oncology Research and Development, Pfizer, Inc., La Jolla, CA, USA
- Belharra Therapeutics, San Diego, CA, USA
| | - Todd VanArsdale
- Oncology Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Adam M Gilbert
- Discovery Sciences, Pfizer Research and Development, Pfizer, Inc., Groton, CT, USA
| | - Matthew M Hayward
- Discovery Sciences, Pfizer Research and Development, Pfizer, Inc., Groton, CT, USA
- Magnet Biomedicine, Boston, MA, USA
| | - Al E Stewart
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Andrew R Nager
- Oncology Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
50
|
Perez AR, Mavrothalassitis O, Chen JS, Hellman J, Gropper MA. CRISPR: fundamental principles and implications for anaesthesia. Br J Anaesth 2025; 134:839-852. [PMID: 39855935 PMCID: PMC11867086 DOI: 10.1016/j.bja.2024.11.040] [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] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 01/27/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-based medical therapies are increasingly gaining regulatory approval worldwide. Consequently, patients receiving CRISPR therapy will come under the care of anaesthesiologists. An understanding of CRISPR, its technological implementations, and the characteristics of patients likely to receive this therapy will be essential to caring for this patient population. However, the role of CRISPR in anaesthesiology extends beyond simply caring for patients with prior CRISPR therapy. CRISPR has multiple direct potential applications in anaesthesia, particularly for managing chronic pain and critical illness. Additionally, given the unique skills anaesthesiologists possess, CRISPR potentially allows new roles for anaesthesiologists in the field of oncology. Consequently, CRISPR technology could enable new domains of anaesthetic practice. This review provides a primer on CRISPR for anaesthesiologists and an overview on how the technology could impact the field.
Collapse
Affiliation(s)
- Alexendar R Perez
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Silico Therapeutics, Inc., San Jose, CA, USA.
| | - Orestes Mavrothalassitis
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | | | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Gropper
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|