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
|
Wang B, Wu Y, Lv X, Liu L, Li J, Du G, Chen J, Liu Y. T7 RNA polymerase-guided base editor for accelerated continuous evolution in Bacillus subtilis. Synth Syst Biotechnol 2025; 10:876-886. [PMID: 40386441 PMCID: PMC12083895 DOI: 10.1016/j.synbio.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/29/2025] [Accepted: 04/19/2025] [Indexed: 05/20/2025] Open
Abstract
Targeted in vivo hypermutation mediated by base deaminase-T7 RNA polymerase (T7 RNAP) fusions promotes genetic diversification and accelerates continuous directed evolution. Due to the lack of a T7RNAP expression regulation system and functionally compatible linker for fusion protein expression, T7RNAP-guided continuous evolution has not been established in Bacillus subtilis, which limited long gene fragment continuous evolution targeted on genome. Here, we developed BS-MutaT7 system, which introduced mutations into specific genomic regions by leveraging chimeric fusions of base deaminases with T7RNAP in B. subtilis. We selected seven different sources of adenosine and cytosine deaminases, 14 fusion protein linkers to be fused with T7RNAP, constructing four libraries with the size of 5000, where deaminases were fused at either the N- or C-terminus of T7RNAP. Based on the efficiency of binding to T7 promoter and high mutagenesis activity, two optimal chimeric mutators, BS-MutaT7A (TadA8e-Linker0-T7RNAP) and BS-MutaT7C (PmCDA1-(GGGGS)3-T7RNAP co-expressed with UGI) were identified. The target mutation rates reached 1.2 × 10-5 per base per generation (s.p.b.) and 5.8 × 10-5 s.p.b., representing 7000-fold and 37,000-fold increases over the genomic mutation rate, respectively. Both exhibited high processivity, maintaining mutation rates of 5.8 × 10-6 s.p.b. and 2.9 × 10-5 s.p.b. within a 5 kb DNA region. Notably, BS-MutaT7C exhibited superior mutagenic activity, making it well-suited for applications requiring intensive and sustained genomic diversification. Application of BS-MutaT7 enabled a 16-fold increase in tigecycline resistance and enhanced β-lactoglobulin (β-Lg) expression by evolving the global transcriptional regulator codY, achieving a β-Lg titer of 3.92 g/L. These results highlight BS-MutaT7 as a powerful and versatile tool for genome-scale continuous evolution in B. subtilis.
Collapse
Affiliation(s)
- Bin Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Yaokang Wu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Xueqin Lv
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Long Liu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Jianghua Li
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Guocheng Du
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Jian Chen
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| | - Yanfeng Liu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
3
|
Wu X, Wan X, Yu H, Liu H. Recent advances in CRISPR-Cas system for Saccharomyces cerevisiae engineering. Biotechnol Adv 2025; 81:108557. [PMID: 40081781 DOI: 10.1016/j.biotechadv.2025.108557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Yeast Saccharomyces cerevisiae (S. cerevisiae) is a crucial industrial platform for producing a wide range of chemicals, fuels, pharmaceuticals, and nutraceutical ingredients. It is also commonly used as a model organism for fundamental research. In recent years, the CRISPR-Cas (clustered regularly interspaced short palindromic repeats-CRISPR-associated proteins) system has become the preferred technology for genetic manipulation in S. cerevisiae owing to its high efficiency, precision, and user-friendliness. This system, along with its extensive toolbox, has significantly accelerated the construction of pathways, enzyme optimization, and metabolic engineering in S. cerevisiae. Furthermore, it has allowed researchers to accelerate phenotypic evolution and gain deeper insights into fundamental biological questions, such as genotype-phenotype relationships. In this review, we summarize the latest advancements in the CRISPR-Cas toolbox for S. cerevisiae and highlight its applications in yeast cell factory construction and optimization, enzyme and phenotypic evolution, genome-scale functional interrogation, gene drives, and the advancement of biotechnologies. Finally, we discuss the challenges and potential for further optimization and applications of the CRISPR-Cas system in S. cerevisiae.
Collapse
Affiliation(s)
- Xinxin Wu
- Frontiers Science Center of Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Xiaowen Wan
- State Key Laboratory of Biotherapy and Cancer Centre/Collaborative Innovation Centre for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongbin Yu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huayi Liu
- Frontiers Science Center of Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; State Key Laboratory of Biotherapy and Cancer Centre/Collaborative Innovation Centre for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Hematology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Biber J, Gandor C, Becirovic E, Michalakis S. Retina-directed gene therapy: Achievements and remaining challenges. Pharmacol Ther 2025; 271:108862. [PMID: 40268248 DOI: 10.1016/j.pharmthera.2025.108862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Gene therapy is an innovative medical approach that offers new treatment options for congenital and acquired diseases by transferring, correcting, inactivating or regulating genes to supplement, replace or modify a gene function. The approval of voretigene neparvovec (Luxturna), a gene therapy for RPE65-associated retinopathy, has marked a milestone for the field of retinal gene therapy, but has also helped to accelerate the development of gene therapies for genetic diseases affecting other organs. Voretigene neparvovec is a vector based on adeno-associated virus (AAV) that delivers a functional copy of RPE65 to supplement the missing function of this gene. The AAV-based gene delivery has proven to be versatile and safe for the transfer of genetic material to retinal cells. However, challenges remain in treating additional inherited as well as acquired retinopathies with this technology. Despite the high level of activity in this field, no other AAV gene therapy for retinal diseases has been approved since voretigene neparvovec. Ongoing research focuses on overcoming the current restraints through innovative strategies like AAV capsid engineering, dual-AAV vector systems, or CRISPR/Cas-mediated genome editing. Additionally, AAV gene therapy is being explored for the treatment of complex acquired diseases like age-related macular degeneration (AMD) and diabetic retinopathy (DR) by targeting molecules involved in the pathobiology of the degenerative processes. This review outlines the current state of retinal gene therapy, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Josef Biber
- Department of Ophthalmology, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Catharina Gandor
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Elvir Becirovic
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Stylianos Michalakis
- Department of Ophthalmology, LMU University Hospital, LMU Munich, 80336 Munich, Germany.
| |
Collapse
|
5
|
Li XH, Lu HZ, Yao JB, Zhang C, Shi TQ, Huang H. Recent advances in the application of CRISPR/Cas-based gene editing technology in Filamentous Fungi. Biotechnol Adv 2025; 81:108561. [PMID: 40086675 DOI: 10.1016/j.biotechadv.2025.108561] [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: 12/05/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Filamentous fungi are essential industrial microorganisms that can serve as sources of enzymes, organic acids, terpenoids, and other bioactive compounds with significant applications in food, medicine, and agriculture. However, the underdevelopment of gene editing tools limits the full exploitation of filamentous fungi, which still present numerous untapped potential applications. In recent years, the CRISPR/Cas (clustered regularly interspaced short palindromic repeats) system, a versatile genome-editing tool, has advanced significantly and been widely applied in filamentous fungi, showcasing considerable research potential. This review examines the development and mechanisms of genome-editing tools in filamentous fungi, and contrasts the CRISPR/Cas9 and CRISPR/Cpf1 systems. The transformation and delivery strategies of the CRISPR/Cas system in filamentous fungi are also examined. Additionally, recent applications of CRISPR/Cas systems in filamentous fungi are summarized, such as gene disruption, base editing, and gene regulation. Strategies to enhance editing efficiency and reduce off-target effects are also highlighted, with the aim of providing insights for the future construction and optimization of CRISPR/Cas systems in filamentous fungi.
Collapse
Affiliation(s)
- Xu-Hong Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing 210023, China
| | - Hui-Zhi Lu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing 210023, China
| | - Ji-Bao Yao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing 210023, China
| | - Chi Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing 210023, China.
| | - Tian-Qiong Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing 210023, China.
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing 210023, China
| |
Collapse
|
6
|
Lee SH, Wu J, Im D, Hwang GH, Jeong YK, Jiang H, Lee SJ, Jo DH, Goddard WA, Kim JH, Bae S. Bystander editing by adenine base editors impairs vision restoration in a mouse model of Leber congenital amaurosis. Mol Ther Methods Clin Dev 2025; 33:101461. [PMID: 40290762 PMCID: PMC12032331 DOI: 10.1016/j.omtm.2025.101461] [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: 07/15/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
Base editors (BEs) have emerged as a powerful tool for gene correction with high activity. However, bystander base editing, a byproduct of BEs, presents challenges for precise editing. Here, we investigated the effects of bystander edits on phenotypic restoration in the context of Leber congenital amaurosis (LCA), a hereditary retinal disorder, as a therapeutic model. We observed that in retinal degeneration 12 (rd12) of LCA model mice, the highest editing activity version of an adenine base editors (ABEs), ABE8e, generated substantial bystander editing, resulting in missense mutations despite RPE65 expression, preventing restoration of visual function. Through AlphaFold-based mutational scanning and molecular dynamics simulations, we identified that the ABE8e-driven L43P mutation disrupts RPE65 structure and function. Our findings underscore the need for more stringent requirements in developing precise BEs for future clinical applications.
Collapse
Affiliation(s)
- Seok-Hoon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jun Wu
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Republic of Korea
| | - Dongjoon Im
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
- Division of Chemistry and Chemical Engineering and Materials Process and Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
| | - Gue-ho Hwang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - You Kyeong Jeong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hui Jiang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Republic of Korea
| | - Seok Jae Lee
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03082, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - William A. Goddard
- Division of Chemistry and Chemical Engineering and Materials Process and Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jeong Hun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03082, Republic of Korea
- Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Ophthalmology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Sangsu Bae
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
7
|
Zhou Z, Xiao J, Yin S, Chen Y, Yuan Y, Zhang J, Xiong L, Xie K. Cas9-Rep fusion tethers donor DNA in vivo and boosts the efficiency of HDR-mediated genome editing. PLANT BIOTECHNOLOGY JOURNAL 2025; 23:2006-2017. [PMID: 40043077 PMCID: PMC12120896 DOI: 10.1111/pbi.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/15/2025] [Indexed: 05/31/2025]
Abstract
Genome editing based on the homology-directed repair (HDR) pathway enables scar-free and precise genetic manipulations. However, the low frequency of HDR hinders its application in plant genome editing. In this study, we engineered the fusion of Cas9 and a viral replication protein (Rep) as a molecular bridge to tether donor DNA in vivo, which enhances the efficiency of targeted gene insertion via the HDR pathway. This Rep-bridged knock-in (RBKI) method combines the advantages of rolling cycle replication of viral replicons and in vivo enrichment of donor DNA at the target site for HDR. Chromatin immunoprecipitation indicated that the Cas9-Rep fusion protein bound up to 66-fold more donor DNA than Cas9 did. We exemplified the RBKI method by inserting small- to middle-sized tags (33-519 bp) into 3 rice genes. Compared to Cas9, Cas9-Rep fusion increased the KI frequencies by 4-7.6-fold, and up to 72.2% of stable rice transformants carried in-frame knock-in events in the T0 generation. Whole-genome sequencing of 6 plants segregated from heterozygous KI lines indicated that the knock-in events were faithfully inherited by the progenies with neither off-target editing nor random insertions of the donor DNA fragment. Further analysis suggested that the RBKI method reduced the number of byproducts from nonhomologous end joining; however, HDR-mediated knock-in tended to accompany microhomology-mediated end joining events. Together, these findings show that the in vivo tethering of donor DNAs with Cas9-Rep is an effective strategy to increase the frequency of HDR-mediated genome editing.
Collapse
Affiliation(s)
- Zhentao Zhou
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
- Hubei Key Laboratory of Plant PathologyHuazhong Agricultural UniversityWuhanChina
| | - Jiahui Xiao
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
- Hubei Key Laboratory of Plant PathologyHuazhong Agricultural UniversityWuhanChina
| | - Shuai Yin
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
- Hubei Key Laboratory of Plant PathologyHuazhong Agricultural UniversityWuhanChina
| | - Yache Chen
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
- Hubei Key Laboratory of Plant PathologyHuazhong Agricultural UniversityWuhanChina
| | - Yang Yuan
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
| | - Jianwei Zhang
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
| | - Lizhong Xiong
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
| | - Kabin Xie
- National Key Laboratory of Crop Genetic ImprovementHubei Hongshan Laboratory, Huazhong Agricultural UniversityWuhanChina
- Hubei Key Laboratory of Plant PathologyHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
8
|
Hoffmann S, Seeger T. Advances in human induced pluripotent stem cell (hiPSC)-based disease modelling in cardiogenetics. MED GENET-BERLIN 2025; 37:137-146. [PMID: 40207041 PMCID: PMC11976404 DOI: 10.1515/medgen-2025-2009] [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] [Indexed: 04/11/2025]
Abstract
Human induced pluripotent stem cell (hiPSC)-based disease modelling has significantly advanced the field of cardiogenetics, providing a precise, patient-specific platform for studying genetic causes of heart diseases. Coupled with genome editing technologies such as CRISPR/Cas, hiPSC-based models not only allow the creation of isogenic lines to study mutation-specific cardiac phenotypes, but also enable the targeted modulation of gene expression to explore the effects of genetic and epigenetic deficits at the cellular and molecular level. hiPSC-based models of heart disease range from two-dimensional cultures of hiPSC-derived cardiovascular cell types, such as various cardiomyocyte subtypes, endothelial cells, pericytes, vascular smooth muscle cells, cardiac fibroblasts, immune cells, etc., to cardiac tissue cultures including organoids, microtissues, engineered heart tissues, and microphysiological systems. These models are further enhanced by multi-omics approaches, integrating genomic, transcriptomic, epigenomic, proteomic, and metabolomic data to provide a comprehensive view of disease mechanisms. In particular, advances in cardiovascular tissue engineering enable the development of more physiologically relevant systems that recapitulate native heart architecture and function, allowing for more accurate modelling of cardiac disease, drug screening, and toxicity testing, with the overall goal of personalised medical approaches, where therapies can be tailored to individual genetic profiles. Despite significant progress, challenges remain in the maturation of hiPSC-derived cardiomyocytes and the complexity of reproducing adult heart conditions. Here, we provide a concise update on the most advanced methods of hiPSC-based disease modelling in cardiogenetics, with a focus on genome editing and cardiac tissue engineering.
Collapse
Affiliation(s)
- Sandra Hoffmann
- University Hospital HeidelbergInstitute of Human GeneticsHeidelbergGermany
| | | |
Collapse
|
9
|
Huang X, Wu W, Qi H, Yan X, Dong L, Yang Y, Zhang Q, Ma G, Zhang G, Lei H. Exploitation of enhanced prime editing for blocking aberrant angiogenesis. J Adv Res 2025; 72:121-133. [PMID: 38996967 DOI: 10.1016/j.jare.2024.07.006] [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: 09/02/2023] [Revised: 01/26/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
INTRODUCTION Aberrant angiogenesis plays an important part in the development of a variety of human diseases including proliferative diabetic retinopathy, with which there are still numerous patients remaining a therapeutically challenging condition. Prime editing (PE) is a versatile gene editing approach, which offers a novel opportunity to genetically correct challenging disorders. OBJECTIVES The goal of this study was to create a dominant-negative (DN) vascular endothelial growth factor receptor (VEGFR) 2 by editing genomic DNA with an advanced PE system to block aberrant retinal angiogenesis in a mouse model of oxygen-induced retinopathy. METHODS An advanced PE system (referred to as PE6x) was established within two lentiviral vectors, with one carrying an enhanced PE guide RNA and a canonical Cas9 nickase fused with an optimized reversal transcriptase, and the other conveying a nicking guide RNA and a DN-MLH1 to improve PE efficiency. Dual non-integrating lentiviruses (NILVs) produced with the two lentiviral PE6x vectors were then employed to create a mutation of VEGFR2 T17967A by editing the Mus musculus VEGFR2 locus in vitro and in vivo, leading to generation of a premature stop codon (TAG, K796stop) to produce DN-VEGFR2, to interfere with the wild type VEGFR2 which is essential for angiogenesis. RESULTS NILVs targeting VEGFR2 delivered into cultured murine vascular endothelial cells led to 51.06 % VEGFR2 T17967A in the genome analyzed by next generation sequencing and the production of DN-VEGFR2, which was found to hamper VEGF-induced VEGFR2 phosphorylation, as demonstrated by Western blot analysis. Intravitreally injection of the dual NILVs into postnatal day 12 mice in a model of oxygen-induced retinopathy, led to production of retinal DN-VEGFR2 in postnatal day 17 mice which blocked retinal VEGFR2 expression and activation as well as abnormal retinal angiogenesis without interfering with retinal structure and function, as assessed by electroretinography, optical coherence tomography, fundus fluorescein angiography and histology. CONCLUSION DN-VEGFR2 resulted from editing genomic VEGFR2 using the PE6x system can be harnessed to treat intraocular pathological angiogenesis.
Collapse
Affiliation(s)
- Xionggao Huang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wenyi Wu
- Department of Ophthalmology, Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Xiaohe Yan
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Lijun Dong
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Yanhui Yang
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, the School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Qing Zhang
- Department of Ophthalmology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Gaoen Ma
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, China; Department of Ophthalmology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
| | - Guoming Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China.
| | - Hetian Lei
- Department of Ophthalmology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China.
| |
Collapse
|
10
|
Shen P, Zheng Y, Zhang C, Li S, Chen Y, Chen Y, Liu Y, Cai Z. DNA storage: The future direction for medical cold data storage. Synth Syst Biotechnol 2025; 10:677-695. [PMID: 40235856 PMCID: PMC11999466 DOI: 10.1016/j.synbio.2025.03.006] [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: 12/11/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025] Open
Abstract
DNA storage, characterized by its durability, data density, and cost-effectiveness, is a promising solution for managing the increasing data volumes in healthcare. This review explores state-of-the-art DNA storage technologies, and provides insights into designing a DNA storage system tailored for medical cold data. We anticipate that a practical approach for medical cold data storage will involve establishing regional, in vitro DNA storage centers that can serve multiple hospitals. The immediacy of DNA storage for medical data hinges on the development of novel, high-density, specialized coding methods. Established commercial techniques, such as DNA chemical synthesis and next-generation sequencing (NGS), along with mixed drying with alkaline salts and refined Polymerase Chain Reaction (PCR), potentially represent the optimal options for data writing, reading, storage, and accessing, respectively. Data security could be promised by the integration of traditional digital encryption and DNA steganography. Although breakthrough developments like artificial nucleotides and DNA nanostructures show potential, they remain in the laboratory research phase. In conclusion, DNA storage is a viable preservation strategy for medical cold data in the near future.
Collapse
Affiliation(s)
- Peilin Shen
- Department of Urology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China
- Shantou University Medical College, Shantou, Guangdong Province, PR China
| | - Yukui Zheng
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China
- Shantou University Medical College, Shantou, Guangdong Province, PR China
| | - CongYu Zhang
- Shantou University Medical College, Shantou, Guangdong Province, PR China
| | - Shuo Li
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, PR China
- BGI-Shenzhen, Shenzhen, Guangdong Province, PR China
- BGI Hospital Groups, Ltd., Shenzhen, Guangdong Province, PR China
| | - Yongru Chen
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China
| | - Yongsong Chen
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China
| | - Yuchen Liu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen, Guangdong Province, PR China
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, PR China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Guangdong Province, PR China
| | - Zhiming Cai
- Shantou University Medical College, Shantou, Guangdong Province, PR China
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, PR China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong Province, PR China
- State Engineering Laboratory of Medical Key Technologies Application of Synthetic Biology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, PR China
- Carson International Cancer Center of Shenzhen University, Shenzhen, Guangdong Province, PR China
| |
Collapse
|
11
|
Sahin GN, Seli E. Gene editing using CRISPR-Cas9 technology: potential implications in assisted reproduction. Curr Opin Obstet Gynecol 2025; 37:141-148. [PMID: 40232991 DOI: 10.1097/gco.0000000000001022] [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: 04/17/2025]
Abstract
PURPOSE OF REVIEW This article reviews the mechanisms, advancements, and potential implications of clustered regularly interspaced short palindromic repeats-associated (CRISPR-Cas) gene editing technology, with a specific focus on its applications in reproductive biology and assisted reproduction. It aims to explore the benefits and challenges of integrating this revolutionary technology into clinical and research settings. RECENT FINDINGS CRISPR-Cas9 is a transformative tool for precise genome editing, enabling targeted modifications through mechanisms like nonhomologous end joining (NHEJ) and homology-directed repair (HDR). Innovations such as Cas9 nickase and dCas9 systems have improved specificity and expanded applications, including gene activation, repression, and epigenetic modifications. In reproductive research, CRISPR has facilitated gene function studies, corrected genetic mutations in animal models, and demonstrated potential in addressing human infertility and hereditary disorders. Emerging applications include mitochondrial genome editing, population control of disease vectors via gene drives, and detailed analyses of epigenetic mechanisms. SUMMARY CRISPR-Cas9 technology has revolutionized genetic engineering by enabling precise genome modifications. This article discusses its mechanisms, focusing on the repair pathways (NHEJ and HDR) and methods to mitigate off-target effects. In reproductive biology, CRISPR has advanced our understanding of fertility genes, allowed corrections of hereditary mutations, and opened avenues for novel therapeutic strategies. While its clinical application in human-assisted reproduction faces ethical and safety challenges, ongoing innovations hold promise for broader biomedical applications.
Collapse
Affiliation(s)
- Gizem Nur Sahin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
12
|
Gasparini K, Zsögön A. Priming crops for heat stress with prime editing. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1846-1848. [PMID: 40227456 DOI: 10.1007/s11427-025-2890-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025]
Affiliation(s)
- Karla Gasparini
- National Institute of Science and Technology on Plant Physiology Under Stress Conditions, Departamento de Biologia Vegetal, Universidade Federal de Viçosa, Viçosa, 36570-900, Brazil
| | - Agustin Zsögön
- National Institute of Science and Technology on Plant Physiology Under Stress Conditions, Departamento de Biologia Vegetal, Universidade Federal de Viçosa, Viçosa, 36570-900, Brazil.
| |
Collapse
|
13
|
Tuncel A, Pan C, Clem JS, Liu D, Qi Y. CRISPR-Cas applications in agriculture and plant research. Nat Rev Mol Cell Biol 2025; 26:419-441. [PMID: 40055491 DOI: 10.1038/s41580-025-00834-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 05/31/2025]
Abstract
Growing world population and deteriorating climate conditions necessitate the development of new crops with high yields and resilience. CRISPR-Cas-mediated genome engineering presents unparalleled opportunities to engineer crop varieties cheaper, easier and faster than ever. In this Review, we discuss how the CRISPR-Cas toolbox has rapidly expanded from Cas9 and Cas12 to include different Cas orthologues and engineered variants. We present various CRISPR-Cas-based methods, including base editing and prime editing, which are used for precise genome, epigenome and transcriptome engineering, and methods used to deliver the genome editors into plants, such as bacterial-mediated and viral-mediated transformation. We then discuss how promoter editing and chromosome engineering are used in crop breeding for trait engineering and fixation, and important applications of CRISPR-Cas in crop improvement, such as de novo domestication and enhancing tolerance to abiotic stresses. We conclude with discussing future prospects of plant genome engineering.
Collapse
Affiliation(s)
- Aytug Tuncel
- Department of Plant Science and Landscape Architecture, University of Maryland, College Park, MD, USA
| | - Changtian Pan
- Department of Horticulture, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou, China
| | - Joshua S Clem
- Department of Plant Science and Landscape Architecture, University of Maryland, College Park, MD, USA
| | - Degao Liu
- Institute of Genomics for Crop Abiotic Stress Tolerance, Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, USA
| | - Yiping Qi
- Department of Plant Science and Landscape Architecture, University of Maryland, College Park, MD, USA.
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, USA.
| |
Collapse
|
14
|
Piao X, Li D, Liu H, Guo Q, Yu Y. Advances in gene and cellular therapeutic approaches for Huntington's disease. Protein Cell 2025; 16:307-337. [PMID: 39121016 PMCID: PMC12120246 DOI: 10.1093/procel/pwae042] [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: 06/24/2024] [Indexed: 08/11/2024] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the abnormal expansion of CAG trinucleotide repeats in the Huntingtin gene (HTT) located on chromosome 4. It is transmitted in an autosomal dominant manner and is characterized by motor dysfunction, cognitive decline, and emotional disturbances. To date, there are no curative treatments for HD have been developed; current therapeutic approaches focus on symptom relief and comprehensive care through coordinated pharmacological and nonpharmacological methods to manage the diverse phenotypes of the disease. International clinical guidelines for the treatment of HD are continually being revised in an effort to enhance care within a multidisciplinary framework. Additionally, innovative gene and cell therapy strategies are being actively researched and developed to address the complexities of the disorder and improve treatment outcomes. This review endeavours to elucidate the current and emerging gene and cell therapy strategies for HD, offering a detailed insight into the complexities of the disorder and looking forward to future treatment paradigms. Considering the complexity of the underlying mechanisms driving HD, a synergistic treatment strategy that integrates various factors-such as distinct cell types, epigenetic patterns, genetic components, and methods to improve the cerebral microenvironment-may significantly enhance therapeutic outcomes. In the future, we eagerly anticipate ongoing innovations in interdisciplinary research that will bring profound advancements and refinements in the treatment of HD.
Collapse
Affiliation(s)
- Xuejiao Piao
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Dan Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Qing Guo
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
15
|
Foyt D, Kuang Y, Rehem S, Yserentant K, Huang B. Accessible and accurate cytometry analysis of adherent cells using fluorescence microscopes. Sci Rep 2025; 15:18691. [PMID: 40436920 PMCID: PMC12120079 DOI: 10.1038/s41598-025-01957-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/09/2025] [Indexed: 06/01/2025] Open
Abstract
We have developed a method along with a Python-based analysis tool to capture images and produce flow-cytometry-like data for adherent cell culture utilizing simple accessible microscopes. Leveraging the recently developed generalist algorithms for cell segmentation, our approach efficiently quantifies single-cell fluorescence signals. We demonstrated the utility of this method by screening a set of 88 prime editing conditions using the integration of mNeonGreen211 as a reporter.
Collapse
Affiliation(s)
- Daniel Foyt
- UCSF-UC Berkeley Joint Graduate Program in Bioengineering, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Yiming Kuang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Samma Rehem
- Barnard College of Columbia University, New York City, NY, 10027, USA
| | - Klaus Yserentant
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 94143, USA.
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
16
|
Wu F, Li N, Xiao Y, Palanki R, Yamagata H, Mitchell MJ, Han X. Lipid Nanoparticles for Delivery of CRISPR Gene Editing Components. SMALL METHODS 2025:e2401632. [PMID: 40434188 DOI: 10.1002/smtd.202401632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 05/05/2025] [Indexed: 05/29/2025]
Abstract
Gene editing has emerged as a promising therapeutic option for treating genetic diseases. However, a central challenge in the field is the safe and efficient delivery of these large editing tools, especially in vivo. Lipid nanoparticles (LNPs) are attractive nonviral vectors due to their low immunogenicity and high delivery efficiency. To maximize editing efficiency, LNPs should efficiently protect gene editing components against multiple biological barriers and release them into the cytoplasm of target cells. In this review, the widely used CRISPR gene editing systems are first overviewed. Then, each component of LNPs, as well as their effects on delivery, are systematically discussed. Following this, the current LNP engineering strategies to achieve non-liver targeting are summarized. Finally, preclinical and clinical applications of LNPs for in vivo genome editing are highlighted, and perspectives for the future development of LNPs are provided.
Collapse
Affiliation(s)
- Fan Wu
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Nei Li
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yudian Xiao
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hannah Yamagata
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xuexiang Han
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
17
|
Wang ZC, Stegall H, Miyazawa T, Keatinge-Clay AT. A CRISPR-Cas9 system for knock-out and knock-in of high molecular weight DNA enables module-swapping of the pikromycin synthase in its native host. Microb Cell Fact 2025; 24:125. [PMID: 40426207 PMCID: PMC12117839 DOI: 10.1186/s12934-025-02741-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Engineers seeking to generate natural product analogs through altering modular polyketide synthases (PKSs) face significant challenges when genomically editing large stretches of DNA. RESULTS We describe a CRISPR-Cas9 system that was employed to reprogram the PKS in Streptomyces venezuelae ATCC 15439 that helps biosynthesize the macrolide antibiotic pikromycin. We first demonstrate its precise editing ability by generating strains that lack megasynthase genes pikAI-pikAIV or the entire pikromycin biosynthetic gene cluster but produce pikromycin upon complementation. We then employ it to replace 4.4-kb modules in the pikromycin synthase with those of other synthases to yield two new macrolide antibiotics with activities similar to pikromycin. CONCLUSION Our gene-editing tool has enabled the efficient replacement of extensive and repetitive DNA regions within streptomycetes.
Collapse
Affiliation(s)
- Zhe-Chong Wang
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hayden Stegall
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Takeshi Miyazawa
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Adrian T Keatinge-Clay
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
18
|
Biar CG, Bodkin N, Carvill GL, Calhoun JD. Protocol to perform multiplexed assays of variant effect using curated loci prime editing. STAR Protoc 2025; 6:103851. [PMID: 40418630 DOI: 10.1016/j.xpro.2025.103851] [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: 01/08/2025] [Revised: 03/26/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Multiplexed assays of variant effect (MAVEs) perform simultaneous characterization of many variants. Here, we present a protocol to perform MAVEs using curated loci prime editing (cliPE), an accessible experimental pipeline that enables prime editing of a target gene. We describe steps for designing prime editing reagents, screening for genome editing efficiency, selecting a pool of cells edited to harbor different genetic variants, and sequencing. Lastly, we detail procedures for performing enrichment analysis to identify variants with normal or aberrant activity.
Collapse
Affiliation(s)
- Carina G Biar
- Ken and Ruth Davee Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA; Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Nicholas Bodkin
- Ken and Ruth Davee Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Gemma L Carvill
- Ken and Ruth Davee Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey D Calhoun
- Ken and Ruth Davee Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
19
|
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
|
20
|
Hassan HM, Zubair A, Helal MH, Almagharbeh WT, Elmagzoub RM. New hope and promise with CRISPR-Cas9 technology for the treatment of HIV. Funct Integr Genomics 2025; 25:108. [PMID: 40411669 DOI: 10.1007/s10142-025-01613-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/26/2025]
Abstract
The commencement of Highly Active Antiretroviral Therapy almost completely stopped viral replication, enabling the immune system to restore its full functionality. The rise in life expectancy has resulted in a decrease in the incidence of classical infections and HIV-associated cancers. HAART has raised concerns, including its exorbitant cost (which hinders its implementation in developing nations), the need for strict adherence, and the potential for both immediate and prolonged ill effects. Lipodystrophy is a significant long-term consequence of HIV that may result in central fat accumulation and severe peripheral fat depletion. Current initiatives to tackle these difficulties include the global expansion of access to HAART, the development of novel drugs that mitigate early side effects, and the introduction of once-daily drug combinations that enhance adherence. The CRISPR-Cas9 system has facilitated the creation of a powerful instrument for precise gene editing. This method has lately established itself as the gold standard for efficient HIV-1 genome editing in HIV therapy, owing to progress in related disciplines. CRISPR may be customized to cleave specific sequences by altering Cas9. This article offers a concise overview of promising CRISPR-Cas9 technology. This technique has the potential to halt the transmission of HIV-1 and alleviate its symptoms. CRISPR-Cas9 technology will be significant in the fight against HIV-1 in the future.
Collapse
Affiliation(s)
- Hesham M Hassan
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Akmal Zubair
- Department of Biotechnology, Quaid-I-Azam University, Islamabad, Pakistan.
| | - Mohamed H Helal
- Center for Scientific Research and Entrepreneurship, Northern Border University, 73213, Arar, Saudi Arabia
| | - Wesam Taher Almagharbeh
- Medical and Surgical Nursing Department, Faculty of Nursing, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Ranya Mohammed Elmagzoub
- Faculty of Science and Technology, Department of Biology and Biotechnology, Al-Neelain University, Khartoum, Sudan
| |
Collapse
|
21
|
Huang CW, Zhang WZ, Liao Y, Hu T, Li JM, Wang CL. A targeted approach: Gene and RNA editing for neurodegenerative disease treatment. Life Sci 2025; 376:123756. [PMID: 40412606 DOI: 10.1016/j.lfs.2025.123756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 05/15/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
With the global aging trend, neurodegenerative diseases (NDs) have emerged as a significant public health concern in the 21st century, imposing substantial economic burdens on families and society. NDs are characterized by cognitive and motor decline, resulting from a combination of genetic and environmental factors. Currently, there is no cure for NDs. Gene and RNA editing therapies offer new possibilities for addressing NDs. Gene editing involves modifying mutant genes associated with NDs, while RNA editing can directly modify RNA molecules to regulate the protein translation process, potentially influencing the expression of genes related to NDs. In this review, we examined the historical evolution, mechanisms of action, applications in NDs, advantages and disadvantages, as well as ethical and safety considerations of gene and RNA editing. While gene and RNA editing technologies hold promise for treating NDs, further research and development are needed to address safety, efficacy, and treatment timing issues, ultimately offering improved treatment options for ND patients. Our review provides valuable insights for future gene and RNA editing applications in ND treatment.
Collapse
Affiliation(s)
- Chen-Wei Huang
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai, 200433, China
| | - Wang-Zheqi Zhang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Yan Liao
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Ting Hu
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai, 200433, China
| | - Jia-Mei Li
- Department of Neurology, The 971st Hospital of Navy, Qingdao 266071, China.
| | - Chang-Li Wang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
22
|
Marei HE. Stem cell therapy: a revolutionary cure or a pandora's box. Stem Cell Res Ther 2025; 16:255. [PMID: 40405306 PMCID: PMC12096755 DOI: 10.1186/s13287-025-04334-1] [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: 02/18/2025] [Accepted: 04/10/2025] [Indexed: 05/24/2025] Open
Abstract
This review article examines how stem cell therapies can cure various diseases and injuries while also discussing the difficulties and moral conundrums that come with their application. The article focuses on the revolutionary developments in stem cell research, especially the introduction of gene editing tools like CRISPR-Cas9, which can potentially improve the safety and effectiveness of stem cell-based treatments. To guarantee the responsible use of stem cells in clinical applications, it is also argued that standardizing clinical procedures and fortifying ethical and regulatory frameworks are essential first steps. The assessment also highlights the substantial obstacles that still need to be addressed, such as the moral dilemmas raised by the use of embryonic stem cells, the dangers of unlicensed stem cell clinics, and the difficulties in obtaining and paying for care for patients. The study emphasizes how critical it is to address these problems to stop exploitation, guarantee patient safety, and increase the accessibility of stem cell therapy. The review also addresses the significance of thorough clinical trials, public education, and policy development to guarantee that stem cell research may fulfill its full potential. The review concludes by describing stem cell research as a promising but complicated topic that necessitates a thorough evaluation of both the hazards and the benefits. To overcome the ethical, legal, and accessibility obstacles and eventually guarantee that stem cell treatments may be safely and fairly included in conventional healthcare, it urges cooperation between the scientific community, legislators, and the general public.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| |
Collapse
|
23
|
Kermanshahi AZ, Ebrahimi F, Taherpoor A, Eslami N, Baghi HB. HPV-driven cancers: a looming threat and the potential of CRISPR/Cas9 for targeted therapy. Virol J 2025; 22:156. [PMID: 40400023 PMCID: PMC12096790 DOI: 10.1186/s12985-025-02783-x] [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: 03/20/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025] Open
Abstract
Cervical and other anogenital malignancies are largely caused by E6 and E7 oncogenes of high-risk human papillomaviruses (HPVs), which inhibit important tumor suppressors like p53 and pRb when they are persistently activated. The main goal of traditional treatments is to physically or chemically kill cancer cells, but they frequently only offer temporary relief, have serious side effects, and have a high risk of recurrence. Exploring the efficacy and accuracy of CRISPR-Cas9 gene editing in both inducing death in HPV-infected cancer cells and restoring the activity of tumor suppressors is our main goal. In this study, we propose a novel precision oncology strategy that targets and inhibits the detrimental effects of the E6 and E7 oncogenes using the CRISPR-Cas9 gene editing system. In order to do this, we create unique guide RNAs that target the integrated HPV DNA and reactivate p53 and pRb. Reactivation is meant to halt aberrant cell development and restart the cell's natural dying pathways. This review discusses the potential of CRISPR/Cas9 in targeting HPV oncogenes, with a focus on studies that have demonstrated its promise in cancer treatment. Given the absence of a definitive treatment for papillomavirus infection and its subsequent association with various cancers, future clinical trials and experimental investigations appear essential to establish and evaluate the therapeutic potential of CRISPR-based approaches. This approach provides a less invasive alternative to conventional treatments and opens the door to personalized care that considers the genetic makeup of each patient's tumor.
Collapse
Affiliation(s)
- Atefeh Zamani Kermanshahi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ebrahimi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Taherpoor
- Department of Clinical Bacteriology; Virology, Faculty of Medicine and Anti-microbial Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Eslami
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Vergnes JB, Roger B, Iggo R, Wodrich H. Advanced viral genome in vitro Cas9 editing (AdVICE): an overnight method for traceless and limitless manipulation of adenoviral and vector genomes with large transgenes. J Virol 2025:e0226524. [PMID: 40396759 DOI: 10.1128/jvi.02265-24] [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: 12/19/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025] Open
Abstract
The size and complexity of large viral genomes limit the technical possibilities for genome manipulations in fundamental research and medical or technological applications. State-of-the-art recombineering in bacteria has partially overcome this limit but remains a time-consuming and complex procedure requiring specialist expertise. Here, we describe a simplified and highly efficient in vitro protocol for unlimited and traceless manipulation applicable to large viral genomes from DNA viruses using a combination of CRISPR/Cas9 cleavage and in vitro DNA assembly. We successfully used the protocol to manipulate adenovirus genomes, showing that genome rescue from viruses, insertions, deletions, and mutagenesis can be performed in a simple overnight procedure in a standard laboratory setting without the need for advanced knowledge of molecular biology. Finally, we use our approach to demonstrate the de novo, multi-step construction of an adenovirus vector suitable for delivering very large transgenes for gene editing.IMPORTANCEThe 36 kb size of the adenoviral genome has long been a deterrent to the construction of adenoviral mutants by scientists wishing to study the virus itself or to construct adenoviral vectors for cell biology and gene therapy. Most previous techniques, such as recombineering and yeast gap repair, impress more by their elegance than by their ease. In this paper, we use Cas9 ribonucleoprotein particles (RNPs) to target cleavage to specific sites in an adenoviral plasmid, then repair the break by Gibson assembly. Gibson assembly with synthetic DNA fragments has transformed basic cloning. Combining it with Cas9 RNPs, which act like highly specific restriction enzymes, makes adenoviral mutagenesis as easy as traditional plasmid cloning. We have used the approach to modify multiple sites in the adenoviral genome, but it could be applied to any large DNA virus for which the genome can be cloned in a plasmid.
Collapse
Affiliation(s)
- Jean-Baptiste Vergnes
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR5234, University of Bordeaux, Bordeaux, France
| | - Benoit Roger
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR5234, University of Bordeaux, Bordeaux, France
| | - Richard Iggo
- INSERM U1312, University of Bordeaux, Bordeaux, France
| | - Harald Wodrich
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR5234, University of Bordeaux, Bordeaux, France
| |
Collapse
|
25
|
Lee C, Lee JS, Kwon Y, Shin A, Jeong TY, Yang J, Hwang JW, Kim HI, Choi HJ, Kim YK, Choi M, Kim K, Sun W, Chae JH. Effects of heterozygous SMG1 mutations on nonsense-mediated mRNA decay in human pluripotent stem cell model. Mol Cells 2025:100225. [PMID: 40403878 DOI: 10.1016/j.mocell.2025.100225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 05/09/2025] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Nonsense-mediated mRNA decay (NMD) eliminates transcripts containing premature termination codons (PTCs), thereby preventing errors in protein synthesis. Serine/Threonine-protein kinase SMG1 is a crucial kinase for NMD response, interacting with other regulatory proteins such as SMG8 and SMG9. We identified a de novo heterozygous variant in SMG1 p.Gln2398Glu (c.7192C>G) in a patient with global developmental delay, facial dysmorphism, and oculomotor apraxia. Thus, stem cell models with SMG1 mutations using gene editing technology were established to address the functional consequences of this mutation. While mutations causing the reduction in SMG1 gene dosage by alterations in splicing (c.7192_7194delinsGAA; GAA/+) or frameshift (c.4331_4337del; KO/+) led to a mild but significant reduction of NMD activity, NMD activity was not altered in cells with the SMG1 GAG/+ mutation. Furthermore, cortical organoids from hPSCGAA/+ exhibited size reduction compared to the control (CTL) or GAG/+, suggesting that reduced NMD activity can affect nervous system development. These findings suggest that hypomorphic SMG1 mutations can cause reduced NMD activity and subsequent biological responses, while the mutation found in the patient alone may not be sufficient to induce pathological symptoms.
Collapse
Affiliation(s)
- Chanyoung Lee
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jin Sook Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul Korea
| | - Yejin Kwon
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Aeri Shin
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Tae Yeong Jeong
- Department of Physiology, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jiyun Yang
- Department of Physiology, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jung Woo Hwang
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyeong-In Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hee-Jung Choi
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon Ki Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Murim Choi
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Kyoungmi Kim
- Department of Physiology, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.
| | - Jong Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Department of Pediatrics, Seoul National University College of Medicine, Seoul Korea.
| |
Collapse
|
26
|
Chen Z, Xue J, Wang Z, Sun J, Cui Y, Zhu T, Yang H, Li M, Wu B. Small RNA Toxin-Assisted Evolution of GC-Preferred ErCas12a for Enhanced Genome Targeting Range. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17105. [PMID: 40391806 DOI: 10.1002/advs.202417105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/26/2025] [Indexed: 05/22/2025]
Abstract
CRISPR/Cas12a, a promising gene editing technology, faces limitations due to its requirement for a thymine (T)-rich protospacer adjacent motif (PAM). Despite the development of Cas12a variants with expanded PAM profiles, many genomic loci, especially those with guanine-cytosine (GC)-rich PAMs, have remained inaccessible. This study develops a small RNA toxin-aided strategy to evolve ErCas12a for targeting GC-rich PAMs, resulting in the creation of enhanced ErCas12a (enErCas12a). EnErCas12a demonstrates the ability to recognize GC-rich PAMs and target five times more PAM sequences than the wild-type ErCas12a. Furthermore, enErCas12a achieves efficient gene editing in both bacterial and mammalian cells at various sites with non-canonical PAMs, including GC-rich PAMs such as GCCC, CGCC, and GGCC, which are inaccessible to previous Cas12a variants. Moreover, enErCas12a effectively targets PAM sequences with a GC content exceeding 75% in mammalian cells, providing a valuable alternative to the existing Cas12a toolkit. Importantly, enErCas12a maintains high specificity at targets with canonical PAMs, while also demonstrating enhanced specificity at targets with non-canonical PAMs. Collectively, this work establishes enErCas12a as a promising tool for gene editing in both eukaryotes and prokaryotes.
Collapse
Affiliation(s)
- Zehua Chen
- AIM center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Junyuan Xue
- State Key Laboratory of Microbial Diversity and Innovative Utilization, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ziying Wang
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100000, China
| | - Jinyuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yinglu Cui
- AIM center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Tong Zhu
- AIM center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Huaiyi Yang
- State Key Laboratory of Microbial Diversity and Innovative Utilization, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ming Li
- State Key Laboratory of Microbial Diversity and Innovative Utilization, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bian Wu
- AIM center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- State Key Laboratory of Green Biomanufacturing, Beijing, 100029, China
| |
Collapse
|
27
|
Wei Y, Gao P, Pan D, Li G, Chen Y, Li S, Jiang H, Yue Y, Wu Z, Liu Z, Zhou M, Chen Y, Xu K, Wu Z, Wang X. Engineering eukaryotic transposon-encoded Fanzor2 system for genome editing in mammals. Nat Chem Biol 2025:10.1038/s41589-025-01902-7. [PMID: 40394336 DOI: 10.1038/s41589-025-01902-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 04/03/2025] [Indexed: 05/22/2025]
Abstract
Eukaryotic transposon-encoded Fanzor proteins hold great promise for genome-engineering applications as a result of their compact size and mechanistic resemblance to TnpB. However, the unmodified Fanzor systems show extremely low activity in mammalian cells. Guided by the predicted structure of a Fanzor2 complex using AlphaFold3, we engineered the NlovFz2 nuclease and its cognate ωRNA to create an evolved enNlovFz2 system, with an expanded target-adjacent motif (TAM) recognition scope (5'-NMYG) and a substantially improved genome-editing efficiency, achieving an 11.1-fold increase over the wild-type NlovFz2, comparable to two previously reported IS200 or IS605 transposon-encoded TnpBs and two CRISPR-Cas12f1 nucleases. Notably, enNlovFz2 efficiently mediated gene disruption in mouse embryos and restored dystrophin expression in a humanized Duchenne muscular dystrophy mouse model with single adeno-associated virus delivery. Our findings underscore the potential of eukaryotic RNA-guided Fanzor2 nucleases as a versatile toolbox for both biological research and therapeutic applications.
Collapse
Affiliation(s)
- Yinghui Wei
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China.
- Hainan Institute, Northwest A&F University, Sanya, China.
| | - Pengfei Gao
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Deng Pan
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guoling Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- HuidaGene Therapeutics Co. Ltd, Shanghai, China
| | - Yufei Chen
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shangpu Li
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Henan Jiang
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yang Yue
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhenmin Wu
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zujiang Liu
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Min Zhou
- Life Science Research Core Services, Northwest A&F University, Yangling, China
| | - Yulin Chen
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China
- Hainan Institute, Northwest A&F University, Sanya, China
| | - Kun Xu
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China.
- Hainan Institute, Northwest A&F University, Sanya, China.
| | - Zhaowei Wu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Xiaolong Wang
- International Joint Agriculture Research Center for Animal Bio-Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Northwest A&F University, Yangling, China.
- Hainan Institute, Northwest A&F University, Sanya, China.
| |
Collapse
|
28
|
Gallala M. Application of CRISPR/Cas gene editing for infectious disease control in poultry. Open Life Sci 2025; 20:20251095. [PMID: 40417002 PMCID: PMC12103187 DOI: 10.1515/biol-2025-1095] [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: 10/26/2024] [Revised: 02/11/2025] [Accepted: 03/11/2025] [Indexed: 05/27/2025] Open
Abstract
The poultry industry faces multifaceted challenges, including escalating demand for poultry products, climate change impacting feed availability, emergence of novel avian pathogens, and antimicrobial resistance. Traditional disease control measures are costly and not always effective, prompting the need for complementary methods. Gene editing (GE, also called genome editing) technologies, particularly CRISPR/Cas9, offer promising solutions. This article summarizes recent advancements in utilizing CRISPR/Cas GE to enhance infectious disease control in poultry. It begins with an overview of modern GE techniques, highlighting CRISPR/Cas9's advantages over other methods. The potential applications of CRISPR/Cas in poultry infectious disease prevention and control are explored, including the engineering of innovative vaccines, the generation of disease-resilient birds, and in vivo pathogen targeting. Additionally, insights are provided regarding regulatory frameworks and future perspectives in this rapidly evolving field.
Collapse
Affiliation(s)
- Mahdi Gallala
- Animal Resources Department, Ministry of Municipality, Doha, State of Qatar
| |
Collapse
|
29
|
Rothgangl T, Tálas A, Ioannidi EI, Weber Y, Böck D, Matsushita M, Villiger EA, Schmidheini L, Moon WJ, Lin PJC, Fan SHY, Marquart KF, Schwerdel C, Rimann N, Faccin E, Villiger L, Muramatsu H, Vadovics M, Cremonesi A, Kulcsár PI, Thöny B, Kopf M, Häberle J, Pardi N, Tam YK, Schwank G. Treatment of a metabolic liver disease in mice with a transient prime editing approach. Nat Biomed Eng 2025:10.1038/s41551-025-01399-4. [PMID: 40394220 DOI: 10.1038/s41551-025-01399-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/11/2025] [Indexed: 05/22/2025]
Abstract
Prime editing is a versatile genome editing technology that circumvents the need for DNA double-strand break formation and homology-directed repair, making it particularly suitable for in vivo correction of pathogenic mutations. Here we developed liver-specific prime editing approaches with temporally restricted prime editor (PE) expression. We first established a dual-delivery approach where the prime editor guide RNA is continuously expressed from adeno-associated viral vectors and only the PE is transiently delivered as nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNP). This strategy achieved 26.2% editing with PEmax and 47.4% editing with PE7 at the Dnmt1 locus using a single 2 mg kg-1 dose of mRNA-LNP. When targeting the pathogenic Pahenu2 mutation in a phenylketonuria mouse model, gene correction rates reached 4.3% with PEmax and 20.7% with PE7 after three doses of 2 mg kg-1 mRNA-LNP, effectively reducing blood L-phenylalanine levels from over 1,500 µmol l-1 to below the therapeutic threshold of 360 µmol l-1. Encouraged by the high efficiency of PE7, we next explored a simplified approach where PE7 mRNA was co-delivered with synthetic prime editor guide RNAs encapsulated in LNP. This strategy yielded 35.9% editing after two doses of RNA-LNP at the Dnmt1 locus and 8.0% editing after three doses of RNA-LNP at the Pahenu2 locus, again reducing L-phenylalanine levels below 360 µmol l-1. These findings highlight the therapeutic potential of mRNA-LNP-based prime editing for treating phenylketonuria and other genetic liver diseases, offering a scalable and efficient platform for future clinical translation.
Collapse
Affiliation(s)
- Tanja Rothgangl
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - András Tálas
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Eleonora I Ioannidi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Yanik Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Desirée Böck
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Mai Matsushita
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Lukas Schmidheini
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Woohyun J Moon
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Paulo J C Lin
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Steven H Y Fan
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Kim F Marquart
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Schwerdel
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Nicole Rimann
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Erica Faccin
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Lukas Villiger
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Máté Vadovics
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Péter István Kulcsár
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Beat Thöny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying K Tam
- Acuitas Therapeutics Inc., Vancouver, British Columbia, Canada
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Tenjo-Castaño F, Rout SS, Dey S, Montoya G. Unlocking the potential of CRISPR-associated transposons: from structural to functional insights. Trends Genet 2025:S0168-9525(25)00080-0. [PMID: 40393858 DOI: 10.1016/j.tig.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 05/22/2025]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated transposons (CASTs) are emerging genome-editing tools that enable RNA-guided DNA integration without inducing double-strand breaks (DSBs). Unlike CRISPR-associated (Cas) nucleases, CASTs use transposon machinery to insert large DNA segments with high precision, potentially reducing off-target effects and bypassing DNA damage responses. CASTs are categorized into classes 1 and 2, each employing distinct mechanisms for DNA targeting and integration. Recent structural insights have elucidated how CASTs recognize target sites, recruit transposases, and mediate insertion. These advances position CASTs as promising tools for genome engineering in bacteria and possibly in mammalian cells. Key challenges remain in enhancing efficiency and specificity, particularly for therapeutic use. Ongoing research aims to evolve CAST systems for precise, large-scale genome editing in human cells.
Collapse
Affiliation(s)
- Francisco Tenjo-Castaño
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Sweta Suman Rout
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Sanjay Dey
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Guillermo Montoya
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| |
Collapse
|
31
|
Liu Y, Liu Y, Wu S, Cao R, Pan Y, Zhou F. Engineered Cas12a-based one-tube detection of DNMT3A R882 H/C mutation in acute myeloid leukemia. Biosens Bioelectron 2025; 286:117609. [PMID: 40413994 DOI: 10.1016/j.bios.2025.117609] [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: 03/13/2025] [Revised: 05/09/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Advances in sequencing technologies have identified numerous genetic alterations associated with acute myeloid leukemia (AML), many of which play critical roles in diagnosis, classification, and prognosis. Among these, mutations in the DNA methyltransferase 3 alpha (DNMT3A) gene are particularly prevalent, with the R882H and R882C variants being the most common. Accurate and sensitive detection of DNMT3A mutations is crucial for prognosis, treatment guidance, and early intervention in AML. However, existing detection methods often fail to achieve an optimal balance among sensitivity, turnaround time, and operational simplicity. To address this limitation, we aimed to develop a rapid and highly sensitive method for detecting DNMT3A mutations. The CRISPR/Cas12a system shows promise for genetic detection due to its high sensitivity and single-base specificity. Here, we established a Cas12a-based one-tube assay for the detection of DNMT3A R882 H/C mutations. We utilized the mismatch tolerance of enAsU-R Cas12a to design crRNA for DNMT3A R882 H/C mutation and integrated CRISPR/Cas12a system with ERA. The entire detection process can be completed within 1 h at 37 °C. The optimized detection system demonstrated a sensitivity of 0.1 % when analyzing genomic DNA. To validate its clinical applicability, we tested samples from 49 AML patients and successfully identified all DNMT3A R882H/C-positive cases, including one with a mutation rate as low as 0.24 %. These results highlight the potential of our Cas12a-based one-tube detection system as a rapid, sensitive, and cost-effective method for detecting DNMT3A R882 H/C mutation. This approach could serve as a valuable tool for both diagnostic and therapeutic monitoring.
Collapse
Affiliation(s)
- Yue Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yin Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Sanyun Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Rui Cao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Hubei Engineering Center for Infectious Disease Prevention, Control and Treatment, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
32
|
Koike A, Brindley PJ. CRISPR/Cas genome editing, functional genomics, and diagnostics for parasitic helminths. Int J Parasitol 2025:S0020-7519(25)00092-X. [PMID: 40348052 DOI: 10.1016/j.ijpara.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Functional genomics using CRISPR (Clustered Regulatory Interspaced Short Palindromic Repeats)/Cas (CRISPR-associated endonuclease)-based approaches has revolutionized biomedical sciences. Gene editing is also widespread in parasitology generally and its use is increasing in studies on helminths including flatworm and roundworm parasites. Here, we survey the progress, specifically with experimental CRISPR-facilitated functional genomics to investigate helminth biology and pathogenesis, and also with the burgeoning use of CRISPR-based methods to assist in diagnosis of helminth infections. We also provide an historical timeline of the introduction and uses of CRISPR in helminth species to date.
Collapse
Affiliation(s)
- Akito Koike
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA.
| |
Collapse
|
33
|
Lu X, Sauter B, Keller A, Zhanybekova S, Gillingham D. Exploring the Potential of Homologous Recombination Protein PALB2 in Synthetic Lethal Combinations. ACS Chem Biol 2025; 20:1099-1106. [PMID: 40300769 PMCID: PMC12090178 DOI: 10.1021/acschembio.5c00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025]
Abstract
Cells with defective homologous recombination (HR) are highly sensitive to poly(ADP-ribose) polymerase (PARP) inhibition. Current therapeutic approaches leverage this vulnerability by using PARP inhibitors in cells with genetically compromised HR. However, if HR factors in cancer cells could be inhibited or degraded pharmacologically, it might reveal other opportunities for synergistic combinations. In this study, we developed a model system that recapitulates PARP/HR synthetic lethality by integrating a small-molecule responsive zinc-finger degron into the HR factor Partner and Localizer of BRCA2 (PALB2). We further tested a series of peptide ligands for PALB2 based on its natural binding partners, which led to the discovery of a high affinity peptide that will support future work on PALB2 and HR. Together, our findings validate PALB2 as a promising drug target and provide the tools and starting points for developing molecules with therapeutic applications.
Collapse
Affiliation(s)
- Xinyan Lu
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Basilius Sauter
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Aramis Keller
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Saule Zhanybekova
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Dennis Gillingham
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
34
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 PMCID: PMC12110317 DOI: 10.3390/cancers17101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| |
Collapse
|
35
|
Geilenkeuser J, Armbrust N, Steinmaßl E, Du SW, Schmidt S, Binder EMH, Li Y, Warsing NW, Wendel SV, von der Linde F, Schiele EM, Niu X, Stroppel L, Berezin O, Santl TH, Orschmann T, Nelson K, Gruber C, Palczewska G, Menezes CR, Risaliti E, Engfer ZJ, Koleci N, Schmidts A, Geerlof A, Palczewski K, Westmeyer GG, Truong DJJ. Engineered nucleocytosolic vehicles for loading of programmable editors. Cell 2025; 188:2637-2655.e31. [PMID: 40209705 DOI: 10.1016/j.cell.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 01/03/2025] [Accepted: 03/07/2025] [Indexed: 04/12/2025]
Abstract
Advanced gene editing methods have accelerated biomedical discovery and hold great therapeutic promise, but safe and efficient delivery of gene editors remains challenging. In this study, we present a virus-like particle (VLP) system featuring nucleocytosolic shuttling vehicles that retrieve pre-assembled Cas-effectors via aptamer-tagged guide RNAs. This approach ensures preferential loading of fully assembled editor ribonucleoproteins (RNPs) and enhances the efficacy of prime editing, base editing, trans-activators, and nuclease activity coupled to homology-directed repair in multiple immortalized, primary, stem cell, and stem-cell-derived cell types. We also achieve additional protection of inherently unstable prime editing guide RNAs (pegRNAs) by shielding the 3'-exposed end with Csy4/Cas6f, further enhancing editing performance. Furthermore, we identify a minimal set of packaging and budding modules that can serve as a platform for bottom-up engineering of enveloped delivery vehicles. Notably, our system demonstrates superior per-VLP editing efficiency in primary T lymphocytes and two mouse models of inherited retinal disease, highlighting its therapeutic potential.
Collapse
Affiliation(s)
- Julian Geilenkeuser
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Niklas Armbrust
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Emily Steinmaßl
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Samuel W Du
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, CA, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Sebastian Schmidt
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany; Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany
| | - Eva Maria Hildegard Binder
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Yuchun Li
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Niklas Wilhelm Warsing
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Stephanie Victoria Wendel
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Florian von der Linde
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Elisa Marie Schiele
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Xiya Niu
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany
| | - Luisa Stroppel
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Oleksandr Berezin
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Tobias Heinrich Santl
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Tanja Orschmann
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany
| | - Keith Nelson
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany
| | - Christoph Gruber
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany
| | - Grazyna Palczewska
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, CA, USA
| | - Carolline Rodrigues Menezes
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, CA, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Eleonora Risaliti
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, CA, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Zachary J Engfer
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, CA, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Naile Koleci
- Department of Medicine III: Hematology/Oncology, Klinikum rechts der Isar of the Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Andrea Schmidts
- Department of Medicine III: Hematology/Oncology, Klinikum rechts der Isar of the Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Helmholtz Munich, Neuherberg, Germany
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, CA, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Department of Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Gil Gregor Westmeyer
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany.
| | - Dong-Jiunn Jeffery Truong
- Institute for Synthetic Biomedicine, Helmholtz Munich, Neuherberg, Germany; Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Munich, Germany.
| |
Collapse
|
36
|
Musunuru K, Grandinette SA, Wang X, Hudson TR, Briseno K, Berry AM, Hacker JL, Hsu A, Silverstein RA, Hille LT, Ogul AN, Robinson-Garvin NA, Small JC, McCague S, Burke SM, Wright CM, Bick S, Indurthi V, Sharma S, Jepperson M, Vakulskas CA, Collingwood M, Keogh K, Jacobi A, Sturgeon M, Brommel C, Schmaljohn E, Kurgan G, Osborne T, Zhang H, Kinney K, Rettig G, Barbosa CJ, Semple SC, Tam YK, Lutz C, George LA, Kleinstiver BP, Liu DR, Ng K, Kassim SH, Giannikopoulos P, Alameh MG, Urnov FD, Ahrens-Nicklas RC. Patient-Specific In Vivo Gene Editing to Treat a Rare Genetic Disease. N Engl J Med 2025. [PMID: 40373211 DOI: 10.1056/nejmoa2504747] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Base editors can correct disease-causing genetic variants. After a neonate had received a diagnosis of severe carbamoyl-phosphate synthetase 1 deficiency, a disease with an estimated 50% mortality in early infancy, we immediately began to develop a customized lipid nanoparticle-delivered base-editing therapy. After regulatory approval had been obtained for the therapy, the patient received two infusions at approximately 7 and 8 months of age. In the 7 weeks after the initial infusion, the patient was able to receive an increased amount of dietary protein and a reduced dose of a nitrogen-scavenger medication to half the starting dose, without unacceptable adverse events and despite viral illnesses. No serious adverse events occurred. Longer follow-up is warranted to assess safety and efficacy. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Kiran Musunuru
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | | | - Xiao Wang
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Taylor R Hudson
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | - Kevin Briseno
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | - Anne Marie Berry
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Julia L Hacker
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Alvin Hsu
- Broad Institute of MIT and Harvard, Harvard University, Howard Hughes Medical Institute, Cambridge, MA
| | | | - Logan T Hille
- Massachusetts General Hospital-Harvard Medical School, Boston
| | - Aysel N Ogul
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | | | | | | | | | | | - Sarah Bick
- Children's Hospital of Philadelphia, Philadelphia
| | | | | | | | | | | | | | | | | | | | | | | | | | - He Zhang
- Integrated DNA Technologies, Coralville, IA
| | | | | | | | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | | | - Lindsey A George
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | | | - David R Liu
- Broad Institute of MIT and Harvard, Harvard University, Howard Hughes Medical Institute, Cambridge, MA
| | - Kim Ng
- Children's Hospital of Philadelphia, Philadelphia
| | | | - Petros Giannikopoulos
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
- University of California, San Francisco, San Francisco
| | - Mohamad-Gabriel Alameh
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Fyodor D Urnov
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | - Rebecca C Ahrens-Nicklas
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
37
|
Xiang W, Lin X, Yang Y, Huang L, Chen Y, Chen J, Liu L. Cas12h is a crRNA-guided DNA nickase that can be utilized for precise gene editing. Cell Rep 2025; 44:115718. [PMID: 40372912 DOI: 10.1016/j.celrep.2025.115718] [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: 01/14/2025] [Revised: 03/20/2025] [Accepted: 04/28/2025] [Indexed: 05/17/2025] Open
Abstract
Type V-H CRISPR-Cas system, an important subtype of type V CRISPR-Cas systems, has remained enigmatic in terms of its structure and function despite being discovered several years ago. Here, we comprehensively characterize the type V-H CRISPR-Cas system and elucidate its role as a DNA nicking system. The unique CRISPR RNA (crRNA) employed by Cas12h effector protein enables specific targeting of double-stranded DNA (dsDNA), while its RuvC domain is responsible for cleaving the non-target strand (NTS) of dsDNA. We present the structure of Cas12h bound to crRNA and target DNA. Our structural analysis reveals that the RuvC domain possesses a narrow active pocket that facilitates recognition of NTS but potentially hinders access to the target strand. Furthermore, we demonstrate that Cas12h confers adaptive immunity against invading mobile genetic elements through transcriptional gene inhibition. We have engineered an adenine base editor by fusing Cas12h with an adenine deaminase, achieving effective A-to-G substitution.
Collapse
Affiliation(s)
- Wenwen Xiang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiaofeng Lin
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yunqian Yang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Linglong Huang
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jiyun Chen
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| | - Liang Liu
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
38
|
Metz S, Belanich JR, Claussnitzer M, Kilpeläinen TO. Variant-to-function approaches for adipose tissue: Insights into cardiometabolic disorders. CELL GENOMICS 2025; 5:100844. [PMID: 40185091 DOI: 10.1016/j.xgen.2025.100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic disorders. However, the functional interpretation of these loci remains a daunting challenge. This is particularly true for adipose tissue, a critical organ in systemic metabolism and the pathogenesis of various cardiometabolic diseases. We discuss how variant-to-function (V2F) approaches are used to elucidate the mechanisms by which GWAS loci increase the risk of cardiometabolic disorders by directly influencing adipose tissue. We outline GWAS traits most likely to harbor adipose-related variants and summarize tools to pinpoint the putative causal variants, genes, and cell types for the associated loci. We explain how large-scale perturbation experiments, coupled with imaging and multi-omics, can be used to screen variants' effects on cellular phenotypes and how these phenotypes can be tied to physiological mechanisms. Lastly, we discuss the challenges and opportunities that lie ahead for V2F research and propose a roadmap for future studies.
Collapse
Affiliation(s)
- Sophia Metz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jonathan Robert Belanich
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melina Claussnitzer
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine, Endocrine Division, Massachusetts General Hospital, Harvard Medical School, Cambridge, MA 02142, USA
| | - Tuomas Oskari Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
39
|
Zang SS, Zhang R, Zhang JR, Zhang X, Li J. Progress, Applications and Prospects of CRISPR-Based Genome Editing Technology in Gene Therapy for Cancer and Sickle Cell Disease. Hum Gene Ther 2025. [PMID: 40351170 DOI: 10.1089/hum.2024.262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
The advent of genome-editing technologies, particularly the RNA-guided the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system (Cas) 9, which originates from prokaryotic CRISPR/Cas adaptive immune mechanisms, has revolutionized molecular biology. Renowned for its simplicity, cost-effectiveness, and capacity for multiplexed gene editing, CRISPR/Cas9 has emerged as the most versatile and widely adopted genome-editing platform. Its applications span fundamental research, biotechnology, medicine, and therapeutics. This review highlights recent advancements in CRISPR-based technologies, focusing on CRISPR/Cas9, CRISPR/Cas12a, and CRISPR/Cas12f. It emphasizes precision editing methods like base editing and prime editing, which enable targeted nucleotide changes without double-strand breaks. The specificity of these tools, including on-target accuracy and off-target risks, is critically evaluated. Additionally, recent preclinical and clinical efforts to treat diseases such as cancer and sickle cell disease using CRISPR are summarized. Finally, the challenges and future directions of CRISPR-mediated gene therapy are discussed, emphasizing its potential to integrate with other molecular approaches to address unmet medical needs.
Collapse
Affiliation(s)
- Sha-Sha Zang
- Department of Geriatric Medicine, Affiliated Hospital of Hebei University, Baoding, China
| | - Ruirui Zhang
- Department of Employee Health Care, West China Hospital, Sichuan University, Chengdu, China
| | - Jia-Run Zhang
- Putian University School of Basic Medicine, Putian, China
| | - Xi Zhang
- Department of Comprehensive Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Li
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
40
|
Ding N, Jiang Y, Lee S, Cheng Z, Ran X, Ding Y, Ge R, Zhang Y, Yang ZJ. Enzyme miniaturization: Revolutionizing future biocatalysts. Biotechnol Adv 2025; 82:108598. [PMID: 40354901 DOI: 10.1016/j.biotechadv.2025.108598] [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: 01/24/2025] [Revised: 04/05/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Enzyme miniaturization offers a transformative approach to overcome limitations posed by the large size of conventional enzymes in industrial, therapeutic, and diagnostic applications. However, the evolutionary optimization of enzymes for activity has not inherently favored compact structures, creating challenges for modern applications requiring smaller catalysts. In this review, we surveyed the advantages of miniature enzymes, including enhanced expressivity, folding efficiency, thermostability, and resistance to proteolysis. We described the applications of miniature enzymes as industrial catalysts, therapeutic agents, and diagnostic elements. We highlighted strategies such as genome mining, rational design, random deletion, and de novo design for achieving enzyme miniaturization, integrating both computational and experimental techniques. By investigating these approaches, we aim to provide a framework for advancing enzyme engineering, emphasizing the unique potential of miniature enzymes to revolutionize biocatalysis, gene therapy, and biosensing technologies.
Collapse
Affiliation(s)
- Ning Ding
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States; Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, United States.
| | - Yaoyukun Jiang
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States; Department of Chemistry and California Institute for Quantitative Biosciences, University of California-Berkeley, Berkeley, CA 94720, United States
| | - Sangsin Lee
- Department of Genetics, Stanford University, Stanford, CA 94305, United States
| | - Zihao Cheng
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States
| | - Xinchun Ran
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States
| | - Yujing Ding
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Robbie Ge
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States
| | - Yifei Zhang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Zhongyue J Yang
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States; Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, United States.
| |
Collapse
|
41
|
Milheiro C, Moura ML, Amendola M, Barbosa MA, Caldeira J. Harnessing CRISPR potential for intervertebral disc regeneration strategies. Front Bioeng Biotechnol 2025; 13:1562412. [PMID: 40406584 PMCID: PMC12095242 DOI: 10.3389/fbioe.2025.1562412] [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: 01/17/2025] [Accepted: 04/15/2025] [Indexed: 05/26/2025] Open
Abstract
Genome editing technologies, particularly CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats), have broadened the possibilities of genetic research and molecular biology by enabling precise modifications of the genome, offering novel therapeutic potential for various disorders. Herein, we present an overview of traditional genome editing techniques and delve deeper into the CRISPR toolbox, with particular attention given to epigenetic and transcriptional regulation. In the context of the intervertebral disc (IVD), CRISPR offers an unprecedented approach to address the mechanisms underlying tissue degeneration, advancing the development of revolutionary therapies for Low Back Pain (LBP). As so, we showcase how to leverage CRISPR systems for IVD. This cutting-edge technology has been successfully used to improve our understanding of IVD biology through functional studies and disease modeling. Most relevant research prioritizes new targets associated with the extracellular matrix (ECM), pain sensing or inflammatory pathways. Promising CRISPR applications encompass IVD regeneration by recapitulation of a regenerative environment or by targeting important degenerative catalysts. In the future, priority should be given to fetal gene reactivation, multiple healthy gene expression enhancement and disease-associated polymorphisms' correction. Despite several challenges such as effective delivery, off-target effects, as well as ethical and safety concerns, exciting clinical trials are anticipated in the years to come, providing more effective and long-lasting solutions for IVD degeneration.
Collapse
Affiliation(s)
- Catarina Milheiro
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Maria L. Moura
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Mario Amendola
- Généthon, Évry, France
- Integrare Research Unit UMR_S951, Université Paris-Saclay, Université Evry, Inserm, Généthon, Évry, France
| | - Mário A. Barbosa
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Joana Caldeira
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| |
Collapse
|
42
|
Dacquay LC, Antoniou P, Mentani A, Selfjord N, Mårtensson H, Hsieh PP, Mustfa S, Thom G, Wimberger S, Firth M, Akrap N, Maresca M, Peterka M. Dual inhibition of DNA-PK and Polϴ boosts precision of diverse prime editing systems. Nat Commun 2025; 16:4290. [PMID: 40341582 PMCID: PMC12062269 DOI: 10.1038/s41467-025-59708-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 05/01/2025] [Indexed: 05/10/2025] Open
Abstract
Prime editing is a genome engineering tool that allows installation of various small edits with high precision. However, prime editing efficiency and purity can vary widely across different edits, genomic targets, and cell types. Prime editing typically offers more precise editing outcomes compared to other genome editing methods such as homology-directed repair. However, it can still result in significant rates of unintended editing outcomes, such as indels or imprecise prime edits. This issue is particularly notable in systems utilizing a second nicking gRNA, such as PE3 and PE5, as well as in dual pegRNA systems and fully active nuclease systems such as PEn, which increase efficiency but compromise precision. In this work, we show that pharmacological inhibition of DNA-PK and Polϴ, two major mediators of mutagenic DNA repair pathways, improves precision of PEn, PE3, PE5, PE7, and dual pegRNA editing systems, including TwinPE, HOPE, and Bi-PE, across multiple genomic loci and edit types. We show that co-inhibition of DNA-PK and Polϴ mitigates both prime editing-unrelated indels and prime editing by-products such as template duplications. Moreover, in the case of PEn, this strategy also substantially improved the off-target editing profile. Together, our data establish small molecule modulation of DNA repair pathways as a general strategy to maximize the precision of diverse prime editing systems.
Collapse
Affiliation(s)
- Louis C Dacquay
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Promega Corporation, Madison, WI, USA
| | - Panagiotis Antoniou
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Astrid Mentani
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Niklas Selfjord
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Hanna Mårtensson
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Pei-Pei Hsieh
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Salman Mustfa
- RNA Therapy, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - George Thom
- RNA Therapy, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sandra Wimberger
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Nina Akrap
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Marcello Maresca
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Martin Peterka
- Genome Engineering, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
43
|
Ball J, Bradley A, Le A, Tisdale JF, Uchida N. Current and future treatments for sickle cell disease: From hematopoietic stem cell transplantation to in vivo gene therapy. Mol Ther 2025; 33:2172-2191. [PMID: 40083162 DOI: 10.1016/j.ymthe.2025.03.016] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Sickle cell disease (SCD) is a single-gene disorder caused by a point mutation of the β-globin gene, resulting in hemolytic anemia, acute pain, multiorgan damage, and early mortality. Hydroxyurea is a first-line drug therapy that switches sickle-globin to non-pathogenic γ-globin; however, it requires lifelong oral administration. Allogeneic hematopoietic stem cell (HSC) transplantation allows for a one-time cure for SCD, albeit with histocompatibility limitations. Therefore, autologous HSC gene therapy was developed to cure SCD in a single treatment, without HSC donors. Current HSC gene therapy is based on the ex vivo culture of patients' HSCs with lentiviral gene addition and gene editing, followed by autologous transplantation back to the patient. However, the complexity of the treatment process and high costs hinder the universal application of ex vivo gene therapy. Therefore, the development of in vivo HSC gene therapy, where gene therapy tools are directly administered to patients, is desirable to provide a more accessible, cost-effective solution that can cure SCD worldwide. In this review, we discuss current treatments, including drug therapies, HSC transplantation, and ex vivo gene therapy; the development of gene therapy tools; and progress toward curative in vivo gene therapy in SCD.
Collapse
Affiliation(s)
- Julia Ball
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Avery Bradley
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anh Le
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
44
|
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
|
45
|
Bengtsson NE, Tasfaout H, Chamberlain JS. The road toward AAV-mediated gene therapy of Duchenne muscular dystrophy. Mol Ther 2025; 33:2035-2051. [PMID: 40181545 DOI: 10.1016/j.ymthe.2025.03.065] [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: 03/10/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025] Open
Abstract
Forty years after the dystrophin gene was cloned, significant progress has been made in developing gene therapy approaches for Duchenne muscular dystrophy (DMD). The disorder has presented numerous challenges, including the enormous size of the gene (2.2 MB), the need to target muscles body wide, and immunogenic issues against both vectors and dystrophin. Among human genetic disorders, DMD is relatively common, and the genetics are complicated since one-third of all cases arise from a spontaneous new mutation, resulting in thousands of independent lesions throughout the locus. Many approaches have been pursued in the goal of finding an effective therapy, including exon skipping, nonsense codon suppression, upregulation of surrogate genes, gene replacement, and gene editing. Here, we focus specifically on methods using AAV vectors, as these approaches have been tested in numerous clinical trials and are able to target muscles systemically. We discuss early advances to understand the structure of dystrophin, which are crucial for the design of effective DMD gene therapies. Included is a summary of efforts to deliver micro-, mini-, and full-length dystrophins to muscles. Finally, we review current approaches to adapt gene editing to the enormous DMD gene with prospects for improved therapies using all these methods.
Collapse
Affiliation(s)
- Niclas E Bengtsson
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98109, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA 98109, USA.
| | - Hichem Tasfaout
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98109, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA 98109, USA.
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98109, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Biochemistry, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
46
|
Wu B, Li J, Jin X. Every cell every gene all at once: Systems genetic approaches toward corticogenesis. Curr Opin Neurobiol 2025; 92:103034. [PMID: 40339387 DOI: 10.1016/j.conb.2025.103034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/24/2024] [Accepted: 04/09/2025] [Indexed: 05/10/2025]
Abstract
The development of the cerebral cortex is a stepwise process that involves numerous cell types and signaling pathways to achieve the functional assembly of neural circuits. Our understanding of this process is primarily rooted in findings from studying transgenic knockout models, which reveal coordinated molecular actions, particularly transcription factor cascades critical for cell type acquisition and maintenance in a context-dependent manner. Further resolving their cell type specificity necessitates the use of high-throughput, high-content methodologies. Over the past decade, the emerging single-cell genomics and in vivo CRISPR tools have provided new approaches to study neurodevelopment with elevated scale and resolution. In this review, we discussed efforts to study mouse cortical cell fate determination using single-cell genomics methods. Additionally, we explored recent studies combining programmable gene editing with single-cell phenotypic assays to investigate the function of transcription factors in perinatal cortical development, delineating cell-type specific, functional cytoarchitecture of the developing brain.
Collapse
Affiliation(s)
- Boli Wu
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiwen Li
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xin Jin
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
47
|
Osborn MJ, Panda S, Reineke TM, Tolar J, Nyström A. Progress in skin gene therapy: From the inside and out. Mol Ther 2025; 33:2065-2081. [PMID: 40077969 DOI: 10.1016/j.ymthe.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
The skin is the largest organ of the body and forms and serves as the barrier for preventing external material from accessing and damaging internal organs. As the outward interface to the environment, it is accessible for the application of therapeutic agents and cellular and gene therapy represent attractive and promising options to treat severe genetic conditions for which palliation has long been the main stay. However, because of its barrier function, transit across and to the subdermal compartment can be challenging. This commentary examines the current approaches of cell and gene therapies for genetic skin disorders. We write this from a local and systemic "outside and inside." perspective. Delivery from the outside encompasses topical, intradermal, and transdermal strategies for cell and vector delivery and ex vivo cell expansion and grafting. The inside approach details systemic delivery via infusion of cells or agents toward providing benefit to the skin. We use recessive dystrophic epidermolysis bullosa (RDEB) as a representative and paradigmatic disease to showcase these approaches as a means to highlight potential broader applicability to other conditions.
Collapse
Affiliation(s)
- Mark J Osborn
- Medical School, Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular and Gene Therapy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Sidharth Panda
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jakub Tolar
- Medical School, Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular and Gene Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
48
|
Wang D, Stevens G, Flotte TR. Gene therapy then and now: A look back at changes in the field over the past 25 years. Mol Ther 2025; 33:1889-1902. [PMID: 40022444 DOI: 10.1016/j.ymthe.2025.02.040] [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: 01/15/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025] Open
Abstract
Since the inception of Molecular Therapy in 2000, the field of gene therapy has made remarkable progress, evolving from no approved clinical products to 23 clinical gene therapy products today. In this review, we aim to capture the transformative changes in the field by surveying the literature over this period, with a particular focus on advancements in gene delivery vector technology, disease and tissue targeting, and the revolutionary molecular tools that have become central to the field. We also discuss the current challenges facing gene therapy and the need for greater collaboration to ensure its accessibility worldwide.
Collapse
Affiliation(s)
- Dan Wang
- Department of Genetic and Cellular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Gregg Stevens
- Lamar Soutter Library, UMass Chan Medical School, Worcester, MA, USA
| | - Terence R Flotte
- Department of Genetic and Cellular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
49
|
Butt H, Sathish S, London E, Lee Johnson T, Essawi K, Leonard A, Tisdale JF, Demirci S. Genome editing strategies for targeted correction of β-globin mutation in sickle cell disease: From bench to bedside. Mol Ther 2025; 33:2154-2171. [PMID: 40165374 DOI: 10.1016/j.ymthe.2025.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025] Open
Abstract
Sickle cell disease (SCD) includes a range of genotypes that result in a clinical syndrome, where abnormal red blood cell (RBC) physiology leads to widespread complications affecting nearly every organ system. Treatment strategies for SCD can be broadly categorized into disease-modifying therapies and those aimed toward a cure. Although several disease-modifying drugs have been approved, they do not fully address the complexity and severity of SCD. Recent advances in allogeneic transplantation and autologous gene therapy show promising outcomes in terms of efficacy and safety. While these approaches have improved the lives of many patients, achieving a durable and comprehensive cure for all remains challenging. To address this, gene-editing technologies, including zinc-finger nucleases, TALENs, CRISPR-Cas, base editing, and prime editing, have been explored both ex vivo and in vivo for targeted correction of the β-globin gene (HBB) in SCD. However, direct correction of HBB and its translation from the laboratory to the clinic presents ongoing limitations, with challenges involved in achieving robust mutation-correction efficiency, off-target effects, and high costs of therapies. The optimal strategy for curing SCD remains uncertain, but several promising approaches are emerging. This review touches on past, present, and future developments in HBB correction.
Collapse
Affiliation(s)
- Henna Butt
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Shruti Sathish
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Evan London
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Taylor Lee Johnson
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Khaled Essawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan 45142, Saudi Arabia
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| |
Collapse
|
50
|
Chen W, Choi J. Molecular circuits for genomic recording of cellular events. Trends Genet 2025:S0168-9525(25)00079-4. [PMID: 40335327 DOI: 10.1016/j.tig.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Advances in precise genome editing are enabling genomic recordings of cellular events. Since the initial demonstration of CRISPR-based genome editing, the field of genomic recording has witnessed key strides in lineage recording, where clonal lineage relationships among cells are indirectly recorded as synthetic mutations. However, methods for directly recording and reconstructing past cellular events are still limited, and their potential for revealing new insights into cell fate decisions has yet to be realized. The field needs new sensing modules and genetic circuit architectures that faithfully encode past cellular states into genomic DNA recordings to achieve such goals. Here we review recently developed strategies to construct diverse sensors and explore how emerging synthetic biology tools may help to build molecular circuits for genomic recording of diverse cellular events.
Collapse
Affiliation(s)
- Wei Chen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| | - Junhong Choi
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, NY 10065, USA.
| |
Collapse
|